CELL101210

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 212

Volume 143

Number 6

December 10, 2010

www.cell.com

Computing Cancer Drivers


Lipids Step into the Spotlight
.
E W Intelligent design

N +
inCuSaFe™ copper enriched
stainless steel interior

+ Single-beam, dual capture


infrared CO2 sensor

+ SafeCell UV
protection in situ

+ Hydrogen peroxide
vapor sterilization in situ

1
Good laboratory technique

Sterisonic™ GxP
Performance and Productivity
Delivers Best Efficiency Value:

2 Hours 14 Hours

SANYO Brand X
Sterisonic™

H2O2 sterilization vs. high heat sterilization


= Uptime (Hours) = Downtime (Hours)

The rapid in situ H2O2 sequence returns the


fully sterilized Sterisonic™ GxP to normal use quicker

Spot on results.
than any competitive incubator in the world.

The industry’s first in situ H2O2 sterilization with the fastest turnaround.
For maximum productivity in clinical, general purpose or the most highly compliant GMP applications,
the new SANYO Sterisonic™ GxP CO2 incubator offers an impressive return on investment. With multiple
contamination control safeguards, exclusive on-board H2O2 sterilization, FDA-21CFR data capture and
graphical LCD display, the Sterisonic™ GxP rewards good laboratory technique with performance
you can trust. Learn more, visit www.sterisonic.com or call 800-858-8442.

pictured above: Sterisonic™ GxP MCO-19AICUVH with rapid H2O2 vapor sterilization system.

FREEER
OFF
[ D E TA IL
S
!
O N L IN E ]
FREE!
FREE!
H2O2 sterilization system acessory, plus BD labware consumables kit.
Limited time upgrade offer with purchase of the Sterisonic™ GxP MCO 19AICUVH. ($900 Value)
BD stem cell starter kit with Sterisonic™ GxP quote.
No purchase necessary! Act now. Supplies are limited. ($150 Value)
www.sterisonic.com
©2009 Sanyo Biomedical OWS 1015 05/09
Do A 180

Turn to a world of true


CO2 incubator sterilization.
Binder CO2 incubators offer:
A 180°C hot-air sterilization cycle that
4
eliminates contamination.
A hot-air jacket that delivers consistent,
4

uniform temperatures.
Drift-free real-time CO2 measurement that
4

ensures stable pH values.
Condensation-free humidification that
4

maintains dry interior walls.
A one-piece weld-free interior with rounded
4

corners and integrated shelf supports.

Get directions at www.true-sterilization.com


or 866-885-9794.

BINDER Inc. | Toll Free 866 885 9794


sales@binder-world.us | www.binder-world.us
BE THE FIRSTto read the latest issue of any Cell Press journal.

Register for Cell Press Email Alerts and get the complete table of contents as soon as
the issue publishes online — FREE!

Cell Press Email Alerts deliver the news, research, and commentaries featured in each
journal’s latest issue, including the full title of every article, direct links to the articles,
and the complete author list. Plus, to save you time, each research article has a brief
summary highlighting its significant findings.

You don’t have to be a subscriber to sign up for Cell Press Email Alerts. While subscribers have
instant access to the full text of all articles listed in the Email Alerts, non-subscribers can read
the abstracts of all articles as well as the full text of the issue’s Featured Article.

www.cellpress.com
017.A1.0115.A © 2010 Eppendorf AG
 Joystick provides
intuitive control

microinjection

search level

B
A
 Patented axial
injection level
C
injection movement
of the capillary
carrier

 Semi-Automatic
microinjection into
adherent cells

 Pre-pull capillaries for


reproducible injection
of adherent cells
InjectMan
NI 2

Microinjection simplified!
Microinjection is one of the core methods to introduce Eppendorf InjectMan NI 2 microinjector has it all:
foreign DNA and other non-permeable molecules into  Motorized X-Y-Z movements provide precise movement
cells. Nuclear injection of plasmid DNA enables rapid  Pre-setting and storage of up to 2 locations in X-Y-Z,
expression of proteins in specific cells within saves time in returning to pre-set work locations
a population.  Automated Home function for rapid capillary exchange
 Joystick-controlled provides overall ergonomic manipulator
The menu-controlled, programmable micromanipulator  Fine adjustment of work speed made easy with
InjectMan NI 2 is ideally suited for microinjection of adherent positioning wheel
cells. Connection with the FemtoJet and the axial mounting  Can be adapted to all common microscopes
allows injections at 45˚ angle reducing cell damage during
injection and increases cell viability. This guarantees a very For more information visit www.eppendorf.com
rapid, safe and reproducible microinjection process.

www.eppendorf.com • Email: info@eppendorf.com


In the U.S.: Eppendorf North America, Inc. 800-645-3050 • In Canada: Eppendorf Canada Ltd. 800-263-8715
bioreprogramming

Biopotential.
Unlock extraordinary potential with stem cell technologies from Sigma . ®

Stem cell biology offers astonishing


research potential; Sigma® Life Science has
the innovations you need to discover the
promise it holds. Access a world of RNAi with
the MISSION® RNAi Library, efficiently edit
genes of interest using advanced CompoZr®
ZFN technology, and characterize your stem
cells with our Prestige Antibodies®, powered
by Atlas Antibodies. The applications are
endless—and so are the possibilities.

wherebiobegins.com/bioreprogramming

Sigma, MISSION, CompoZr and Prestige Antibodies are registered trademarks of


Sigma-Aldrich and its affiliate Sigma-Aldrich Biotechnology, L.P.
Editor Editorial Board Douglas Green Dana Pe’er
Emilie Marcus Abul Abbas Leonard Guarente Kathrin Plath
C. David Allis Taekjip Ha Carol Prives
Senior Deputy Editor
Geneviève Almouzni Daniel Haber Klaus Rajewsky
Elena Porro
Uri Alon Ulrike Heberlein Venki Ramakrishnan
Deputy Editors Angelika Amon Nobutaka Hirokawa Rama Ranganathan
Robert Kruger Johan Auwerx Mark Hochstrasser Anne Ridley
Connie M. Lee Richard Axel Arthur Horwich James Roberts
Cori Bargmann Tony Hunter Alexander Rudensky
Scientific Editors Konrad Basler James Hurley Helen Saibil
Karen Carniol
Bonnie Bassler Richard Hynes Joshua Sanes
Kara Cerveny David Baulcombe Thomas Jessell Randy Schekman
Michaeleen Doucleff Jeffrey Benovic Narry Kim Ueli Schibler
Fabiola Rivas Carolyn Bertozzi Mary-Claire King Joseph Schlessinger
Niki Scaplehorn
Wendy Bickmore David Kingsley Hans Schöler
Lara Szewczak Elizabeth Blackburn Frank Kirchhoff Trina Schroer
Senior Managing Editor Joan Brugge Richard Kolodner Geraldine Seydoux
Meredith Adinolfi Lewis Cantley John Kuriyan Kevan Shokat
Joanne Chory Robert Lamb Pamela Sklar
Deputy Managing Editor David Clapham Mark Lemmon Nahum Sonenberg
Andy Smith Andrew Clark Beth Levine James Spudich
Lead Illustrator Hans Clevers Wendell Lim Paul Sternberg
Andrew A. Tang Stephen Cohen Jennifer Lippincott-Schwartz Bruce Stillman
Pascale Cossart Dan Littman Azim Surani
Illustrators Jeff Dangl Richard Losick Keiji Tanaka
Yvonne Blanco Ted Dawson Scott Lowe Craig Thompson
Kate Mahan Pier Paolo di Fiore Tom Maniatis Robert Tjian
Marileen Dogterom Matthias Mann Jürg Tschopp
Production Staff
Julian Downward Kelsey Martin Ulrich von Andrian
Reyna Clancy
Bruce Edgar Joan Massagué Gerhard Wagner
Editorial Assistant Steve Elledge Iain Mattaj Detlef Weigel
Mary Beth O’Leary Anne Ephrussi Satyajit Mayor Alan Weiner
Ronald Evans Ruslan Medzhitov Jonathan Weissman
Witold Filipowicz Craig Mello Matthew Welch
Marco Foiani Tom Misteli Tian Xu
Elaine Fuchs Tim Mitchison Shinya Yamanaka
Yukiko Goda Alex Mogilner Marino Zerial
Stephen Goff Paul Nurse Xiaowei Zhuang
Joe Goldstein Roy Parker Huda Zoghbi

Cell Office
Cell, Cell Press, 600 Technology Square, 5th Floor, Cambridge, Massachusetts 02139
Phone: (+1) 617 661 7057, Fax: (+1) 617 661 7061, E-mail: celleditor@cell.com
Online Publication: http://www.cell.com

Cell (ISSN 0092-8674) is published biweekly by Cell Press, 600 Technology Square, 5th Floor, Cambridge, Massachusetts 02139. The institutional subscription rate for
2010 is $1,360 (US and Canada) or $1,532 (elsewhere). The individual subscription rate is $202 (US and Canada) or $305 (elsewhere). The individual copy price is $50.
Periodicals postage paid at Boston, Massachusetts and additional mailing offices. Postmaster: send address changes to Elsevier Customer Service Americas,
Cell Press Journals, 11830 Westline Industrial Drive, St. Louis, MO 63146, USA.
The paper used in this publication meets the requirments of ANSI/NISO Z39.48-1992 (Permanence of Paper). Printed by Dartmouth Printing Company, Hanover, NH.
Cell Press Display Advertising
Northeast/Mid-Atlantic: Victoria Macomber, ph: 508 928
President & CEO 1255; fax: 508 928 1256; e-mail: v.macomber@elsevier.com
Lynne Herndon Midwest/Southeast/Eastern Canada: Inez Herrero-Redman,
ph: 585 678 4395; fax: 585 678 4722; e-mail: i.herrero@elsevier.
Editor in Chief, Vice President of Content Development
com
Emilie Marcus
Northwest/Southwest/Western Canada: Lori Young, ph: 646
Vice President of Marketing and Publishing 370 6312; fax: 212 462 1915; e-mail: l.young@elsevier.com
Els Bosma California: Elizabeth Loennborn, ph: 714 655 1877; fax: 214 452
Vice President of Web Development and Operations 9627; e-mail: e.loennborn@elsevier.com
Keith Wollman UK/Europe: James Kenney, ph: +44 20 7424 4216; fax: +44 18
6585 3136; e-mail: j.kenney@elsevier.com
Director of Marketing
Jonathan Atkinson Asia: Wendy Xie, ph: +86 10 8520 8827; e-mail: w.xie@
elsevier.com
Production Manager
Meredith Adinolfi Classified Advertising
United States and Canada:
Press Officer Gordon Sheffield, Key Account Manager, ph: 617 386 2189; fax:
Cathleen Genova 617 397 2805; e-mail: g.sheffield@elsevier.com
UK, Europe, and Asia:
Sabrina Dodge, Key Account Manager, ph: +44 20 7424 4997;
fax: +44 18 6585 3136; e-mail: s.dodge@elsevier.com

ª2010 Elsevier Inc. All rights reserved. advances in the medical sciences, in particular, independent verification
This journal and the individual contributions contained in it are protected of diagnoses and drug dosages should be made. Although all advertis-
under copyright by Elsevier Inc., and the following terms and conditions ing material is expected to conform to ethical (medical) standards, inclu-
apply to their use: sion in this publication does not constitute a guarantee or endorsement
Photocopying: of the quality or value of such product or of the claims made of it by its
Single photocopies of single articles may be made for personal use as al- manufacturer.
lowed by national copyright laws. Permission of the Publisher and payment Reprints:
of a fee are required for all other photocopying, including multiple or system- Article reprints are available through Cell’s reprint service; for informa-
atic copying, copying for advertising or promotional purposes, resale, and tion, contact Nicholas Pavlow (e-mail: n.pavlow@elsevier.com; ph: (+1)
all forms of document delivery. Special rates are available for educational 212 633 3960).
institutions that wish to make photocopies for nonprofit educational class- Subscription Orders and Inquiries:
room use. For information on how to seek permission, visit www.elsevier. Mail, fax, or e-mail address changes to Elsevier Customer Service Amer-
com/permissions or call (+44) 1865 843830 (UK) / (+1) 215 239 3804 (US). icas, allowing 4–6 weeks for processing. Lost or damaged issues will be
Permissions: replaced, subject to availability, if Cell Press is notified within the claim
For information on how to seek permission, visit www.elsevier.com/ period (US and airmail delivery: 3 months from issue date; surface deliv-
permissions or call (+44) 1865 843830 (UK) / (+1) 215 239 3804 (US). ery: 4 months from issue date). Periodical delivery in the US can take up
Derivative Works: to 3 weeks. Airmail delivery can take 2–4 weeks.
Subscribers may reproduce tables of contents or prepare lists of articles The price of a single copy of Cell is $50 (excluding special issues).
including summaries for internal circulation within their institutions. All orders must be prepaid and in writing. Please include the volume
Permission of the Publisher is required for resale or distribution outside and issue number, payment (check or credit card, MasterCard, Visa,
the institution. Permission of the Publisher is required for all other deriv- or American Express only), and a delivery address. Allow 4–6 weeks
ative works, including compilations and translations (please consult for delivery.
www.elsevier.com/permissions). Mailing address: Elsevier Customer Service Americas, Cell Press
Journals, 11830 Westline Industrial Drive, St. Louis, MO 63146,
Electronic Storage or Usage:
USA. Toll-free phone within USA/Canada: 866 314 2355; phone for
Permission of the Publisher is required to store or use electronically any
outside US/Canada: (+1) 314 453 7038; fax: (+1) 314 523 5170; e-mail:
material contained in this journal, including any article or part of an article
subs@cell.com; internet: www.cellpress.com or <www.cell.com>.
(please consult www.elsevier.com/permissions). Except as outlined
above, no part of this publication may be reproduced, stored in a retrieval Funding Body Agreements and Policies:
system, or transmitted in any form or by any means, electronic, mechan- Elsevier has established agreements and developed policies to allow au-
ical, photocopying, recording, or otherwise, without prior written permis- thors whose articles appear in journals published by Elsevier to comply
sion of the Publisher. with potential manuscript archiving requirements as specified as condi-
Notice: tions of their grant awards. To learn more about existing agreements and
No responsibility is assumed by the Publisher for any injury and/or dam- policies, visit http://www.cell.com/cellpress/FundingBodyAgreements.
age to persons or property as a matter of products liability, negligence, Guide for Authors:
or otherwise, or from any use or operation of any methods, products, For a full and complete guide for authors, please go to www.cell.com/
instructions, or ideas contained in the material herein. Because of rapid authors.
MASTERING CHANGE

Breakthrough in 5-hmC quantitation for epigenetics


Interested in simplifying the study of DNA methylation, particularly 5-hydroxymethylcytosine? Try the EpiMark™
5-hmC and 5-mC Analysis Kit, a robust enzymatic method for the locus-specific detection of methylated (5-mC) and
hydroxymethylated (5-hmC) cytosine. As the first commercially available PCR-based kit to reproducibly identify and
quantitate the presence of 5-hmC, this simple 3-step protocol will expand your potential for epigenetics research and
biomarker discovery.

Identify and quantitate methylation states with the Advantages:


EpiMark™ 5-hmC and 5-mC Analysis Kit
• Reproducible quantitation of 5-hmC and 5-mC
% 5-hmC
• Easy-to-use protocols
% 5-mC
% Unmethylated C • Compatible with existing techniques (PCR)
100
• Amenable to high throughput
80

60

40
Visit neb.com/epigenetics to learn more, and
explore the complete listing of EpiMark validated
20 products from NEB.
0
Brain Liver Heart Spleen

Analysis of the different methylation states in Balb/C mouse tissue samples


shows a variation in the amount of 5-hmC present at locus 12.

DNA AMPLIFICATION PROTEIN EXPRESSION GENE EXPRESSION


CLONING & MAPPING RNA ANALYSIS
& PCR & ANALYSIS & CELLULAR ANALYSIS
www.neb.com
Leading Edge
Cell Volume 143 Number 6, December 10, 2010

IN THIS ISSUE

SELECT
853 Lipids Out Loud

PREVIEWS

861 Insulin Signaling: B.D. Manning


Inositol Phosphates Get into the Akt

863 Consequences of mRNA C.J. Wilusz and J. Wilusz


Wardrobe Malfunctions
865 Kinases Charging to the Membrane M.A. Hadders and R.L. Williams

867 Exposing Contingency Plans A.M. Klein, E.M. Dioum, and M.H. Cobb
for Kinase Networks

ESSAY

870 Lipid Trafficking sans Vesicles: W.A. Prinz


Where, Why, How?

REVIEW

875 Membrane Budding J.H. Hurley, E. Boura, L.-A. Carlson,


and B. Roz_ ycki

PRIMER
888 Lipidomics: New Tools and Applications M.R. Wenk

SNAPSHOT

1030 Inositol Phosphates A.J. Hatch and J.D. York


Antibodies and Related Reagents for Signal Transduction Research

Announcing XP ™
Monoclonal Antibodies,
eXceptional Monoclonal Technology™
exclusively from Cell Signaling Technology ®
Above: Confocal IF analysis of rat colon using Phospho-β-Catenin (Ser675) (D2F1) XP™ Rabbit mAb #4176. Red = DY-554 phalloidin. Blue = DRAQ5® #4084.

Signaling Technology, Inc. / DRAQ5® is a registered trademark of Biostatus Limited


sensitivity™, eXceptional stability and reproducibility™, eXceptional Performance™ and Cell Signaling Technology® are trademarks of Cell
© 2010 Cell Signaling Technology, Inc.
XMT™, an eXceptional Monoclonal Technology™ eXceptional specificity™
from Cell Signaling Technology®, enables the + eXceptional sensitivity™
development and production of antibodies with
eXceptional Performance™ (XP™). This revolutionary + eXceptional stability and reproducibility™
and proprietary technology provides us with access to = eXceptional Performance™

XMT™, XP™ , eXceptional Monoclonal Technology™, eXceptional specificity™, eXceptional


a broad range of antibody-producing B cells unattainable
with traditional monoclonal technologies, allowing XMT coupled with our extensive antibody validation and
more comprehensive screening and the identification stringent quality control delivers XP monoclonal antibodies with
of XP monoclonal antibodies. eXceptional Performance in the widest range of applications.

untreated EGF-treated (100 ng/ml, 15 min.)


3
31

PC

La
4

kDa
He
Bx
A-

200
Events

EGFR Receptor
140
100
80

60
EGF Receptor
human lung carcinoma Jurkat cells (red) and Kyse70 cells (blue) A-549 cells

EGF Receptor (D38B1) XP™ Rabbit mAb #4267 demonstrates eXceptional Performance in a wide range of applications.
See www.cellsignal.com for full validation details.

for more information on XMT™ visit...


www.cellsignal.com
China www.cst-c.com.cn | Europe www.cellsignal.eu | Japan www.cstj.co.jp Tel: 978-867-2300 | E-mail: info@cellsignal.com
Articles
Cell Volume 143 Number 6, December 10, 2010

897 Inositol Pyrophosphates Inhibit A. Chakraborty, M.A. Koldobskiy, N.T. Bello, M. Maxwell,
Akt Signaling, Thereby Regulating J.J. Potter, K.R. Juluri, D. Maag, S. Kim, A.S. Huang,
Insulin Sensitivity and Weight Gain M.J. Dailey, M. Saleh, A.M. Snowman, T.H. Moran,
E. Mezey, and S.H. Snyder

911 Loss of Anion Transport without Increased J.-H. Chen, D.A. Stoltz, P.H. Karp, S.E. Ernst,
Sodium Absorption Characterizes Newborn A.A. Pezzulo, T.O. Moninger, M.V. Rector,
Porcine Cystic Fibrosis Airway Epithelia L.R. Reznikov, J.L. Launspach, K.Chaloner,
J. Zabner, and M.J. Welsh

924 Sister Cohesion and Structural Axis K.P. Kim, B.M. Weiner, L. Zhang, A. Jordan, J. Dekker,
Components Mediate Homolog Bias and N. Kleckner
of Meiotic Recombination

938 Upf1 ATPase-Dependent mRNP Disassembly T.M. Franks, G. Singh, and J. Lykke-Andersen
Is Required for Completion of Nonsense-
Mediated mRNA Decay

951 Dynamics of Cullin-RING Ubiquitin E.J. Bennett, J. Rush, S.P. Gygi, and J.W. Harper
Ligase Network Revealed
by Systematic Quantitative Proteomics

966 Kinase Associated-1 Domains Drive K. Moravcevic, J.M. Mendrola, K.R. Schmitz, Y.-H. Wang,
MARK/PAR1 Kinases to Membrane Targets D. Slochower, P.A. Janmey, and M.A. Lemmon
by Binding Acidic Phospholipids

978 The Fused/Smurf Complex Controls the L. Xia, S. Jia, S. Huang, H. Wang, Y. Zhu, Y. Mu,
Fate of Drosophila Germline Stem Cells L. Kan, W. Zheng, D. Wu, X. Li, Q. Sun, A. Meng,
by Generating a Gradient BMP Response and D. Chen

991 Functional Overlap and Regulatory Links S. van Wageningen, P. Kemmeren, P. Lijnzaad,
Shape Genetic Interactions T. Margaritis, J.J. Benschop, I.J. de Castro,
between Signaling Pathways D. van Leenen, M.J.A. G. Koerkamp, C.W. Ko, A.J. Miles,
N. Brabers, M.O. Brok, T.L. Lenstra, D. Fiedler,
L. Fokkens, R. Aldecoa, E. Apweiler, V. Taliadouros,
K. Sameith, L.A.L. van de Pasch, S.R. van Hooff,
L.V. Bakker, N.J. Krogan, B. Snel, and F.C.P. Holstege

(continued)
the difference between discover and do over

Gibco. Because every little thing matters.


Fact: your research will only be as accurate, as efficient, as groundbreaking as the cell culture
you’re working with. That’s why you’re settling for nothing less than Gibco®. Considering it could
be the difference between publishing your results and reading someone else’s, why experiment
with your experiment?

invitrogen.com/discovergibco
THEORY
1005 An Integrated Approach U.D. Akavia, O. Litvin, J. Kim, F. Sanchez-Garcia,
to Uncover Drivers of Cancer D. Kotliar, H.C. Causton, P. Pochanard, E. Mozes,
L.A. Garraway, and D. Pe’er

RESOURCE

1018 Comprehensive Polyadenylation Site F. Ozsolak, P. Kapranov, S. Foissac, S.W. Kim,


Maps in Yeast and Human Reveal E. Fishilevich, A.P. Monaghan, B. John,
Pervasive Alternative Polyadenylation and P.M. Milos

ANNOUNCEMENTS

POSITIONS AVAILABLE

On the cover: In cancer, heterogeneous genetic aberrations frequently result in similar


phenotypic outcomes. In this issue, Akavia et al. (pp. 1005–1017) report a computational
algorithm that identifies driving mutations and links them with their downstream
transcriptional effects. The approach thus allows the authors to begin disentangling the
complex mechanisms by which genetic aberrations drive transformation. The cover shows
a driver with many DNA-mediated paths to a tumor.
R&D Systems Tools for Cell Biology Research™

Proteins
R&D Systems Quantikine® ELISAs
Antibodies

ELISAs
The Most Referenced Immunoassays
A direct measure of product quality is the frequency of citations in the scientific literature. R&D Systems has more
Assay Services
than 20 years of experience designing, testing, and optimizing the most cited ELISA kits in the world. Find out why
MultiAnalyte Profiling scientists trust R&D Systems ELISAs more than any other brand.

Activity Assays R&D Systems is the Most Referenced ELISA Manufacturer NEW Quantikine ELISA Kits
 α1-Acid Glycoprotein
Stem Cells  Angiopoietin-like 3
 Cathepsin V
ELISpot Kits  Clusterin
17.3%  Dkk-1
R&D Systems  EGF R/ErbB1
Flow Cytometry 41.8% 7.9%  EG-VEGF/PK1
 Fetuin A
Cell Selection 3.7%  FGF-21
 Galectin-3
2.5%  Gas 6
2.1%  GDF-15
1.6%  IL-17A/F Heterodimer
1.4%
 IL-19
 Lipocalin-2/NGAL
 MBL
 Proprotein Convertase 9/PCSK9
Approximately 42% of Referenced Immunoassays are Developed and Manufactured by  Periostin/OSF-2
R&D Systems. A survey of 860 manuscripts from 44 journals was conducted to compare the  Progranulin
number of citations specifying the use of R&D Systems ELISAs to the number citing ELISAs from  ST2/IL-1 R4
other commercial sources. A total of 433 ELISA citations referencing immunoassays from 66  Thrombomodulin/CD141
different vendors were identified in the survey.  Tie-1
 TIM-1/KIM-1

For more information visit our website at www.RnDSystems.com/go/ELISA

For research use only. Not for use in diagnostic procedures.

R&D Systems, Inc. www.RnDSystems.com

R&D Systems Europe, Ltd. www.RnDSystems.co.uk

R&D Systems China Co., Ltd. www.RnDSystemsChina.com.cn


Leading Edge

In This Issue

New Akt-Ins Diet


PAGE 897
Chakraborty et al. show that inositol pyrophosphate IP7 plays a key role in
high-fat diet-induced insulin resistance and weight gain. Mechanistically,
IP7 inhibits Akt kinase activation by blocking PH domain-mediated phos-
phorylation and membrane recruitment. Mice deficient for IP7 synthesis
exhibit resistance to obesity induced by aging or high-fat diet. The results
thus suggest that inhibitors of the kinase that generates IP7 may be benefi-
cial for treatment of obesity and diabetes.

Cystic Fibrosis Revisited


PAGE 911
How does mutation of the CFTR chloride ion channel cause cystic fibrosis
(CF)? Using a new porcine model of CF, Chen et al. see reduced chloride
and bicarbonate flow across CF airway epithelia, as expected. However, in contrast to a widely held hypothesis,
lack of CFTR does not increase sodium or liquid absorption. The data explain how loss of CFTR alters cellular
electrical properties that had been previously interpreted as sodium hyperabsorption and clarify the initiating events
in CF.

Ditching Your Sister, Finding Your Mate


PAGE 924
During meiosis, recombination occurs between homologous maternal and paternal chromosomes. Kim et al.
investigate how interhomolog crossover is favored over crossovers between the two sister, or duplicate, chromatids
that are also present. The authors find that, whereas the proteins that promote sister chromatid cohesion
inhibit homolog recombination, the meiotic proteins Red1 and Mek1 counteract this effect to promote homolog
recombination.

Adaptors Drive Ubiquitination Dynamics


PAGE 951
Cullin-RING ubiquitin ligases (CRLs) are modular ubiquitin ligases that rely on substrate adaptors to regulate degrada-
tion of specific proteins. In this issue, Bennett et al. analyze CRL complex dynamics in the cell with a novel quantitative
proteomics platform. The authors find that the cellular abundance of
substrate adaptors drives CRL network organization. These findings chal-
lenge the prevailing view that CRL complexes are principally regulated by
cycles of deneddylation and complex disassembly.

mRNP Striptease
PAGE 938
The nonsense-mediated mRNA decay (NMD) pathway rids the cell
of aberrant mRNAs with premature translation termination codons. Franks
et al. demonstrate that disassembly of protein complexes from mRNAs
targeted for NMD is required for complete mRNA degradation. This disas-
sembly requires the ATPase activity of the Upf1 helicase and is critical for
the recycling and reuse of NMD factors. These findings identify active
disassembly of mRNPs as a critical step in mRNA decay.

Cell 143, December 10, 2010 ª2010 Elsevier Inc. 849


2nd Edition
Bacterial StreSS reSponSeS
Editors: Gisela Storz and Regine Hengge

T he second edition of the highly


acclaimed Bacterial Stress Responses

Stay Connected
incorporates and reviews the vast num-
ber of new findings that have greatly
advanced the understanding of bacterial
stress responses in the decade since the
Join ASHG Today! publication of the first edition. Readers will
discover how this improved understand-
ing not only enhances our knowledge of
all cellular regulation at the molecular level, but also provides new
ammunition in the fight against pathogens and helps optimize the use
Benefits of Membership: of bacteria in biotechnology.
• Subscription to AJHG, now published All chapters have been contributed by leaders and pioneers in their
by Cell Press respective fields and then carefully edited to ensure conciseness
and clarity. With its coverage of a broad range of model organisms
• Reduced registration fees for the as well as biotechnologically, medically, and environmentally relevant
Annual Meeting bacteria, this new edition fully encapsulates our understanding of
bacterial stress responses. Moreover, it serves as a springboard for
• Discounted subscriptions to Cell new investigations and new applications.
Press journals November 2010. Hardcover.
ISBN: 978-1-55581-621-6, 540 pages est., illustrations, index.
• Access to educational materials List price: $169.95; ASM member price: $159.95
• Influence on public policy process Shop early and qualify for introductory pricing.

Web: http://estore.asm.org • CALL: 800-546-2416


www.ashg.org or 703-661-1593 • Write to: ASM Press, P. O. Box
605, Herndon, VA 20172, USA • FAX: 703-661-1501
Cell_Scopus_192X123.qxd:Layout 3 2/12/08 3:10 PM Page 1


Scopus is the largest abstract and citation
database of peer-reviewed literature and
quality web sources with smart tools to
track, analyze and visualize research.

enrich
your

experience

www.scopus.com

New Tickets to the Membrane
PAGE 966
Spatial organization of cellular signaling relies on protein modules that
interact with membrane surfaces. Moravcevic et al. now identify a new
phospholipid-binding domain. A crystal structure reveals it to be a KA1
domain, seen in human MARK/PAR1 kinases implicated in disease. The
results show that KA1 domains bind acidic phospholipids and, by cooper-
ating with other binding modules, detect a coincidence of signals on
membranes to target the kinases to specific subcellular locations.

Unusual Suspects in Redundancy


PAGE 991
Signaling networks include redundant components, such as homologous
kinases, that compensate for each other when one component is lost. Could nonhomologous proteins, or even proteins
of opposite function like kinases and phosphatases, compensate for each other, too? van Wageningen et al. develop
a systems approach that not only identifies network redundancies involving nonhomologous proteins but also
uncovers the molecular mechanisms generating such relationships. These findings explain how signaling pathways
integrate to coordinate responses to stimuli.

Nearest Neighbors Are Miles apart in


Signaling
PAGE 978
In the Drosophila ovary, germline stem cells (GSCs) divide asymmetri-
cally to self-renew and produce daughter cytoblasts (CBs). GSCs are
maintained by BMPs produced by niche cells. Xia et al. report a
pathway for regulated proteolysis of the BMP receptor in CBs that
generates a steep gradient of BMP activity between GSCs and the
immediately adjacent CBs. This pathway confers divergent responses
to secreted ligands to daughters just one cell diameter apart and allows
CB differentiation.

Computing through Cancer’s Complexity


PAGE 1005
Cancer genomes are extremely diverse from patient to patient,
rendering identification of the genetic aberrations key for cancer initiation and progression challenging. Akavia et al.
report a computational method that leverages DNA copy number and gene expression information to identify cancer
drivers. Applying their method to a melanoma dataset, the authors revealed two genes involved in protein trafficking as
drivers required for tumor cell proliferation.

Poly(A)-OK in Human RNAs


PAGE 1018
30 untranslated regions and poly(A) tails orchestrate mRNA localization, stability, and translation. In this issue, Ozsolak
et al. map genome-wide human and yeast polyadenylation states. From this analysis, they identify new sequence
motifs correlated with human polyadenylation patterns and suggest that these position-specific sequences may be
associated with polyadenylation of noncoding RNAs.

Cell 143, December 10, 2010 ª2010 Elsevier Inc. 851


.%7

Reach Your Ideal Candidate!


så0OSTåONEåORåMULTIPLEåå
JOBåOPENINGSå

så0ROMOTEåANåUPCOMINGåå
CONFERENCEåORåEVENTå

så!NNOUNCEåAåGRANTAWARDå

så%LEVATEåYOURåå
ORGANIZATIONSåPROlLE

careers.cell.com
Leading Edge

Select: Lipids Out Loud

If RNA ruled the last decade and DNA dominated the previous one, could the next decade be the one for
lipids? The ultimate energy storage units, lipids are well known for encasing the cell in a watertight
membrane, but lipids are oh-so much more. In particular, lipids are emerging as key signaling molecules
in eukaryotes, transmitting messages both within and between cells. In this Select, we explore recent studies
highlighting the diverse physiological roles of these ‘‘lipid messengers’’—from modulating pain perception
and bone formation to reining in renegade inflammatory responses.

Lipid pH Meter Senses Nutrient Status


The diversity of lipids in eukaryotic cells—estimated to be > 1000—is daunting. But, in
terms of signaling roles, phosphatidic acid is quickly rising to the top. Ubiquitous in
cellular membranes, phosphatidic acid is known to recruit cytosolic proteins to their
appropriate location at membrane surfaces. Now, Young et al. demonstrate that
phosphatidic acid also serves as a pH sensor in Saccharomyces cerevisiae, coupling
the nutrient status of the cell to membrane biogenesis.
In yeast, the transcriptional repressor Opi1 regulates membrane production by
blocking transcription of genes required for the synthesis of lipid precursors. During
periods of growth, Opi1 is retained outside of the nucleus by its interaction with phos-
phatidic acids in the endoplasmic reticulum (ER) membrane. To identify new path-
Phosphatidic acid (PA) serves as a pH ways that regulate Opi1’s localization, Young et al. screen a library of yeast mutants
sensor in yeast, linking glucose avail- for their ability to make lipid precursors. Surprisingly, many genes that govern intra-
ability to membrane biogenesis through cellular pH are also required for keeping Opi1 bound to the ER membrane. Indeed,
its interaction with the transcriptional a fluorescent version of Opi1 demonstrates that, when the pH of the cell drops,
repressor Opi1. Image courtesy of Opi1 dissociates from the phosphatidic acid and travels to the nucleus to repress lipid
C. Loewen. synthesis genes.
How does phosphatidic acid sense pH, and what physiological role does it serve?
Glucose starvation triggers a rapid drop in intracellular pH from 7 to 6.0. The phosphate group of phosphatidic acid is unique
among phospholipids in that it is negatively charged at pH 7 but neutral at pH < 6.6. Young and colleagues demonstrate that
neutralizing phosphatidic acid dramatically weakens its affinity for Opi1, leading to the release of the metabolic suppressor
when nutrients are low. Given the universality of pH regulation in the cell and the ubiquity of phosphatidic acid in cellular
membranes, the authors speculate that this type of pH biosensor is probably a common signaling mechanism for coupling
a physiological state to membrane biogenesis.
Young et al. (2010). Science 329, 1085–1088.

A New High for Pain Treatment


Whereas phosphatidic acid is a key signaling lipid inside of cells, many lipids can also
transmit information between cells. One potent class of these ‘‘lipid messengers’’ is
endocannabinoids, such anandamide. Neurons secrete anandamide, which then
blocks pain perception by activating cannabinoid receptors in both the central and
peripheral nervous systems. Now, Clapper et al. (2010) develop a small molecule
that boosts anandamide levels in the peripheral nervous system, but not in the brain
or spinal cord. Despite its restricted range of action, this molecule exhibits surprisingly
powerful analgesic effects in mice models of acute and chronic pain, opening a new
avenue for treating pain without unwanted psychotropic effects.
Anandamide is degraded by a membrane protein called fatty acid amide hydrolase,
or FAAH. Current inhibitors of FAAH raise anandamide concentrations but are quite
hydrophobic and thus easily cross the blood-brain barrier. To create FAAH inhibitors
that act only in the periphery, Clapper et al. add hydrophilic groups at sites unlikely to
alter interactions with FAAH. One molecule, called URB937, increases anandamide Fatty acid amide hydrolase (FAAH)
levels in peripheral tissues, but not in the forebrain or hypothalamus. Most impor- degrades the endocannabinoid ananda-
tantly, administration of this compound near damaged tissue reduces pain responses mide; ‘‘ananda’’ is Sanskrit for ‘‘bliss.’’
in mice with efficacies comparable to centrally acting FAAH inhibitors and a common Image courtesy of D. Piomelli.
nonsteroidal anti-inflammatory compound. These results suggest that amplifying
endocannabinoid levels in the peripheral nervous system alters the processing of
pain signals in the spinal cord, highlighting the potency of these lipid-based neuromo-
dulators throughout the body.
Clapper et al. (2010). Nature Neuroscience 13, 1265–1270.

Cell 143, December 10, 2010 ª2010 Elsevier Inc. 853


60
years of leadership in
human genetics research,
education and service.
1948–2008
www.ashg.org
C’est Bone La Vie
Although endocannabinoids are well known for regulating pain perception and appe-
tite, cannabinoid receptors are present on almost every cell type in the human body,
including osteoclasts and osteoblasts in bones. Mammalian bones undergo
a constant remodeling process whereby osteoclasts break up the mineralized matrix
while osteoblasts replace it with new tissue. Endocannabinoids are known to regulate
this process, but the details are still unclear. Now, Smoum et al. identify an endocan-
nabinoid-related lipid called oleoyl serine, which tilts bone remodeling in favor of the
osteoblasts. Remarkably, treatment with this lipid rescues more than half of the bone
loss observed in a mice model of osteoporosis.
Smoum et al. first extract lipids from the femur and tibia of mice. They then use
a combination of mass spectrometry and chromatography to confirm the presence
of known lipids such as anandamide and uncover new ones such as oleoyl serine,
The lipid oleoyl serine increases bone a lipid synthesized from a major ingredient in olive oil (i.e., oleic acid). Smoum et al.
mass, which is measured by microcom- find that oleoyl serine stimulates the growth of cultured osteoblasts at extremely
puted tomography. Image courtesy of low concentrations (10 picomolar), making oleoyl serine the most potent of the
I. Bab and R. Mechoulam. bone lipids in this in vitro cell proliferation assay. In addition, oleoyl serine limits the
life span of osteoclasts by triggering apoptosis. Despite the orthogonal effects of
oleoyl serine in osteoclasts and osteoblasts, this lipid signals through a Gi protein-coupled receptor and the Erk1/2 (extracel-
lular regulated kinases 1/2) kinase pathway in both cells.
Finally, Smoum and colleagues demonstrate that, unlike current treatments for osteoporosis, oleoyl serine displays
a double-pronged attack against bone loss in a mouse model for osteoporosis. It not only boosts the rate of bone formation,
but also slows down bone resorption, making oleoyl serine an attractive new lead for more potent therapeutics against this
widespread disease.
Smoum et al. (2010). Proceedings of the National Academy of Sciences 107, 17710–17715.

OMGega-3s! COX-2 Makes Anti-Inflammatories


Endocannabinoids and oleoyl serine are lipid messengers synthesized within cells,
but dietary fats can also serve signaling roles. For example, clinical studies suggest
that omega-3 fatty acids in fish oil, such as docosahexaenoic acid (DHA), help to
prevent diseases associated with chronic inflammation, but how these lipids mediate
an anti-inflammatory effect is still largely unknown. Now, Groeger et al. demonstrate
that, during an inflammatory response, human macrophages convert DHA into
oxidized fatty acids that directly activate anti-inflammatory transcription factors,
such as the nuclear receptor peroxisome proliferator-activated g (PPARg). Moreover,
these anti-inflammatory lipids are generated by cyclooxygenase-2 (COX-2), the
enzyme best known as the target of ibuprofen and for catalyzing the synthesis of
pro-inflammatory lipids prostaglandins.
Groeger et al. first develop a clever mass spectrometry approach that detects
reactive lipids even at extremely low concentrations. Using this technique, they
then identify derivatives of DHA generated in cultured monocytes and macrophages Cyclooxygenase-2 (COX-2) converts do-
after these cells are activated by various immune triggers, including interferon g and cosahexaenoic acid (DHA) from fish oil
lipopolysaccharide. Next, the authors show that the production of these DHA metab- into ‘‘electrophilic’’ fatty acids that acti-
olites requires COX-2, and purified COX-2 can synthesize these lipids in vitro. Finally, vate and inhibit pro- and anti-inflamma-
Groeger and colleagues demonstrate that two of the DHA metabolites suppress the tory processes, respectively. Image
expression of pro-inflammatory cytokines (e.g., IL-6 and IL-10) in a dose-dependent courtesy of F. Schopfer.
manner and boost nuclear localization of Nrf2, the master transcription factor for the
antioxidant response in immune cells.
Interestingly, COX-2 starts to generate these anti-inflammatory lipids 8–10 hours after the initiation of an immune
response. Together with the clinical studies on fish oil supplementation, these results suggest that the production of oxidized
DHA derivatives by COX-2 may be a key mechanism for preventing an acute inflammatory response from turning into a chronic
and damaging one.
Groeger et al. (2010). Nature Chemical Biology 6, 433–441.

Michaeleen Doucleff

Cell 143, December 10, 2010 ª2010 Elsevier Inc. 855


Workflow
Oriented
Easy-to-Find
Tools

Clean
Desktop

Experiment
History

MetaMorph® NX Microscopy Automation


and Image Analysis Software
Complexity made simple!
Easy-to-Find Tools
• Simplify training with the right tool to complete each task.
• Vital devices for set-up and review are close at hand, without obscuring images.

Workflow Oriented
• Consistent workflow-oriented interface guides you through even the most complicated tasks.
• Fluently track data when moving between parameters of interest - time, position, and wavelength.

Clean Desktop*
• Desktop is always clean and organized for a clear view of your data.
• Clearly visualize and compare multi-parametric datasets side-by-side across different data views.

Experiment History
Dwww.moleculardevices.com
etect. Decode. Discover.
• Dynamic dataset linking ensures analyzed results remain linked to the image source.
1311 Orleans Drive, Sunnyvale, CA 94089-1136 USA
Phone: 1 408 747 1700 | Fax: 1 408 747 3601
Toll Free: 1 800 635 5577 * Patent pending

Email Contacts Sales & Support China


+86 10 6410 8669 (Beijing)
Info@moldev.com United States & Canada +86 21 6887 8820 (Shanghai)
Sales@moldev.com 1 800 635 5577
Support@moldev.com Japan
Service@moldev.com Brazil: +55 11 3616 6607 +81 6 6399 8211 (Osaka)
United Kingdom: +44 118 944 8000 +81 3 5282 5261 (Tokyo)
Germany: +49 89/96 05 88 0 South Korea: +82 2 3471 9531
For Research Use Only. Not for use in diagnostic procedures.
©2010 Molecular Devices, Inc. All Rights Reserved. Molecular Devices, the Molecular Devices logo, and all other trademarks are the property of Molecular Devices, Inc.
Announcing an innovative new textbook from Academic Cell
Primer to The Immune Response, Academic Cell Update Edition
By Tak W. Mak and Mary Saunders

Primer to The Immune Response, Academic Cell Update


Edition, is an invaluable resource for students who
need a concise but complete and understandable
introduction to immunology.

Academic Cell textbooks contain premium journal


content from Cell Press and are part of a new
cutting-edge textbook/journal collaboration
designed to help today’s instructors teach students to
“think like a scientist.”

academiccell.com

Primer to The Immune Response


Academic Cell Update Edition
Academic Cell is a dynamic textbook publishing partnership between
Tak W. Mak Academic Press and Cell Press, two market-leading publishers bringing
The Campbell Family Institute for Breast scientific advances from the world of life science research into the classroom.
Cancer Research, Ontario, Canada
Mary Saunders Order online now from: elsevierdirect.com/9780123847430
The Campbell Family Institute for Breast Request and examination copy from textbooks.elsevier.com
Cancer Research, Ontario, Canada

Paperback/456 pages
ISBN: 9780123847430 Twitter.com/academiccell Facebook.com/academiccell
$79.95/£54.99/€64.95
“Not to brag but
I automatically hit the
bullseye every time.”

SCEPTER™ : ON TARGET EVERY TIME.

Scepter, the first handheld automated cell counter, uses the unequalled accuracy of the Coulter Principle to
count cells and measure their dimensions automatically. No more clumsy benchtop work stations or counts
limited by vision-based counting techniques. Just handheld freedom combined with pinpoint accuracy.
A bold new statement in cell counting.

Get accurate results with Scepter


www.millipore.com/scepter

Millipore and Advancing Life Science Together are registered trademarks of Millipore Corporation. The M mark is and Scepter are trademarks of Millipore Corporation
©2010 Millipore Corporation. All rights reserved.
Expand your stem cell library and save
today on the latest books on stem cells
and regenerative medicine

Stem Cells Stem Cell Anthology


Scientific Facts and Fiction From Stem Cell Biology, Tissue Engineering,
Christine Mummery, Ian Wilmut, Anja Van de Stolpe Regenerative Medicine, Cloning and Stem
and Bernard Roelen
Cell Methods
November 2010 | 400 pages | Paperback | Bruce M. Carlson
$79.95 | €57.95 | £48.99 | October 2009 | 450 pp. | Hardback | $150.00 |
ISBN: 9780123815354 €100.00 | £95.00 |AU$222.00 |
ISBN: 9780123756824

Principles of Regenerative
Medicine, 2nd Edition Essential Stem Cell Methods
Anthony Atala and Robert Lanza A Volume in the Reliable Lab Solutions Series
November 2010 | 1400 pages | Hardback | Robert Lanza and Irina Klimanskaya
$199.95 | €143.00 | £125.00 | April 2009 | 628 pp. | Paperback | $75.00 | €50.95
ISBN: 9780123814227 | £45.99 |AU$111.00 | ISBN: 9780123750617

Heart Development and Tissue Engineering


Regeneration, 2-Volume Set Clemens van Blitterswijk, Peter Thomsen, Jeffrey Hubbell,
Ranieri Cancedda, Anders Lindahl Sahlgrenska,
Nadia Rosenthal and Richard P. Harvey
Jerome Sohier and David F. Williams
June 2010 | 1072 pp. | Hardback
| $199.95 | €143.00 | £125.00 | March 2008 | 760 pp. | Hardback | $115.00 |
AU$296.00 | ISBN: 9780123813329 €76.95 | £69.99 |AU$170.00 |
ISBN: 9780123708694

Essentials of Stem Cell


Biology, 2nd Edition Human Stem Cell Manual
Robert Lanza, Roger Pedersen, John Gearhart, E. A Laboratory Guide
Donnall Thomas, Brigid Hogan, James Thomson, Jeanne F. Loring, Robin L. Wesselschmidt and Philip H. Schwartz
Douglas Melton and Sir Ian Wilmut June 2007 | 488 pp. | Spiral bound | $88.95 |
June 2009 | 600 pp. | Hardback | $199.95 €59.95 | £53.99 |AU$132.00 |
| €134.00 | £125.00 | AU$302.00 | ISBN: ISBN: 9780123704658
9780123747297

Handbook of Stem Cells


Foundations of 2-Volume Set with CD-ROM Vol. 1–2
Regenerative Medicine Vol. 1 – Embryonic Stem Cells
Clinical and Therapeutic Applications Vol. 2 – Adult & Fetal Stem Cells
Anthony Atala, Robert Lanza, James Thomson and Robert Lanza, Roger Pedersen, Helen Blau, E. Donnall Thomas,
Robert Nerem John Gearhart, James Thomson, Brigid Hogan, Catherine
Verfaillie, Douglas Melton, Irving Weissman, Malcolm Moore
September 2009 | 750 pp. | Hardback |
and Michael West
$99.95 | €66.95 | £60.99|AU$148.00 |
ISBN: 9780123750853 September 2004 | 1,760 pp. | Hardback | $566.00
| €380.00 | £345.00 | AU$817.00 |
ISBN: 9780124366435

Cell Stem Cell subscribers save 25% on their book order


Secure ordering online at elsevierdirect.com
Enter promo code 28024 at check out
Prices and publication dates subject to change without notice.
Leading Edge
Previews

Insulin Signaling:
Inositol Phosphates Get into the Akt
Brendan D. Manning1,*
1Department of Genetics and Complex Diseases, Harvard School of Public Heath, Boston, MA 02115, USA

*Correspondence: bmanning@hsph.harvard.edu
DOI 10.1016/j.cell.2010.11.040

An acute but transient response to insulin is essential for glucose homeostasis in mammals. Chak-
raborty et al. (2010) uncover a new feedback mechanism regulating insulin signaling. They show
that the inositol pyrophosphate IP7, which is produced in response to insulin, inhibits the Akt
kinase, a primary effector of insulin signaling.

Pancreatic b cells produce insulin in stimulating the production of the key lipid insulin-stimulated pathway leads to atten-
response to the rise in circulating glucose second messenger phosphatidylinositol- uation of insulin signaling.
levels after a meal. Insulin restores basal 3,4,5-trisphosphate (PIP3). PIP3 then Inositol phosphates are a diverse group
blood glucose levels by eliciting distinct binds the pleckstrin homology (PH) of signaling molecules in which hydroxyl
metabolic responses in target tissues, domain of the serine/threonine kinase Akt, groups positioned around an inositol ring
including the stimulation of glucose allowing two other kinases—the phos- are phosphorylated in different combina-
uptake into skeletal muscle and adipose phoinositide-dependent kinase (PDK1) tions by an array of inositol phosphate
tissue and the inhibition of glucose output and the mammalian target of rapamycin kinases. One such kinase, inositol hexa-
in the liver. The homeostatic response to (mTOR) complex 2 (mTORC2) —to phos- kisphosphate (IP6) kinase 1 (IP6K1), pro-
insulin must occur rapidly but transiently phorylate and activate Akt. Akt is a major duces a pyrophosphate group at the 5
following a spike in blood glucose. Thus, effector of the insulin response, and its position of IP6 to generate 5-diphospho-
proper control over both stimulatory downstream substrates directly mediate inositolpentakisphosphate (5-PP-IP5, or
and inhibitory signals affecting the re- many of the metabolic effects of insulin IP7; Figure 1B). Studies on IP6K demon-
sponse to insulin is important for prevent- (Manning and Cantley, 2007). Insulin strate a role for the IP7 product in
ing metabolic imbalance and common resistance is a hallmark of type-2 diabetes promoting insulin production by pancre-
metabolic diseases such as type-2 dia- and is characterized by an inability of atic b cells (Illies et al., 2007). Of interest,
betes. Chakraborty et al. (2010) now insulin to signal to Akt (Whiteman et al., despite low blood insulin levels in the
identify a new feedback mechanism that 2002). Ip6k1 knockout mice due to defects in
attenuates insulin signaling. They show Insulin signaling can be inhibited at insulin secretion, the levels of blood
that the production of a specific inositol multiple steps between the insulin glucose in these mice are normal, sug-
pyrophosphate, which is stimulated receptor and Akt activation. The best- gesting that these mice have enhanced
by insulin, inhibits canonical insulin sig- characterized inhibitors include lipid peripheral insulin sensitivity (Bhandari
naling by preventing activation of the phosphatases such as PTEN and SHIP2, et al., 2008).
kinase Akt. which dephosphorylate lipids produced Chakraborty et al. examine the molec-
Whereas the response to insulin varies by PI3K. In addition, insulin induces ular mechanism and physiological conse-
among tissues, the signal transduction signaling pathways that can promote quences of the increased responsiveness
pathway triggered by insulin is conserved inhibitory phosphorylation of the IRS to insulin suggested by the IP6K1
(Taniguchi et al., 2006; Figure 1A). Insulin proteins, preventing the activation of knockout mouse phenotype. Using insulin
binds to and activates cell surface PI3K and Akt. For instance, Akt signaling and insulin-like growth factor 1 (IGF-1) to
insulin receptors, and these receptor tyro- activates mTOR complex 1 (mTORC1) stimulate hepatocytes and mouse
sine kinases phosphorylate the insulin and its downstream target S6K1, and embryo fibroblasts, the authors demon-
receptor substrate (IRS) proteins on these ser/thr kinases can directly phos- strate enhanced Akt activation in Ip6k1
specific tyrosine residues. Phosphory- phorylate serine residues on IRS1, leading knockout cells relative to wild-type. Of
lated IRS proteins serve as scaffolding to its inhibition (Harrington et al., 2005). In interest, the authors also find that insulin
adaptors for signaling proteins, the most this manner, the stimulation of mTORC1 and IGF-1 stimulate a gradual increase
important of which is the class IA phos- activity in response to insulin creates an in the levels of the IP6K1 product IP7 in
phatidylinositol 3-kinase (PI3K). Engage- inhibitory feedback mechanism that wild-type cells, and this inositol pyro-
ment of PI3K by the IRS protein activates decreases insulin signaling. Chakraborty phosphate inhibits Akt translocation to
this lipid kinase at the plasma membrane, et al. now report that production of a the plasma membrane and its subsequent
where its substrate phosphatidylinositol- specific inositol pyrophosphate repre- phosphorylation by PDK1. Taken together
4,5-bisphosphate (PIP2) is abundant, sents another mechanism by which an with a previous study by this group

Cell 143, December 10, 2010 ª2010 Elsevier Inc. 861


demonstrating that IP7 can in metabolic tissues following
bind directly to the PH feeding and whether the rela-
domain of Akt (Luo et al., tive levels of these opposing
2003), the data suggest that molecules change under
IP7 competes with PIP3 for different conditions of insulin
binding to Akt, thereby block- resistance. It will also be
ing Akt activation (Figure 1A). important to understand the
Thus, insulin and IGF-1 stimu- mechanism by which insulin
late the production of two and IGF-1, and perhaps other
phosphoinositol species, growth factors, stimulate the
PIP3 through PI3K and IP7 production of IP7 by IP6K1.
through IP6K1 (Figure 1B), It remains possible that this
which have reciprocal effects stimulation is downstream of
on Akt activation. Akt, making this a classic
These cell-intrinsic effects negative-feedback mecha-
of IP6K1 and its product IP7 nism analogous to that medi-
suggest a mechanistic basis ated by mTORC1 and S6K1
for the enhanced insulin sen- (Harrington et al., 2005). Of
sitivity implied from previous interest, the major metabolic
studies on the Ip6k1 knock- features of the Ip6k1 knock-
out mice (Bhandari et al., out phenotype—defects in
2008). Measuring systemic b cell insulin production,
responses to insulin, Chakra- resistance to obesity, and
borty et al. (2010) find that improved peripheral insulin
Figure 1. The Insulin Signaling Pathway and Inositol Phosphates
Ip6k1 knockout mice display sensitivity—are the same as
(A) The figure shows the canonical insulin signaling pathway leading to
enhanced activation of Akt in activation of the serine/threonine kinase Akt. Chakraborty et al. (2010) show those reported for the S6k1
response to insulin in both that insulin also stimulates the inositol phosphate kinase IP6K1 to produce knockout mice (Um et al.,
skeletal muscle and adipose IP7 (5-diphosphoinositolpentakisphosphate), which in turn inhibits Akt. 2004), perhaps suggesting
The authors’ results suggest a model for the inhibition of Akt by IP7. In this
tissue, accompanied by in- model, IP7 binding to the PH domain of Akt prevents the translocation of Akt a mechanistic link between
creased glucose uptake into to the membrane by preventing the binding of PIP3 (phosphatidylinositol- the IP6K and mTORC1 path-
these tissues. Importantly, 3,4,5-trisphosphate) to the same domain, thus blocking insulin signaling ways. Finally, IP6K1 could
to Akt.
the Ip6k1 knockout mice are (B) Inositol derivatives serve as signaling molecules when phosphorylated on
represent a new therapeutic
lean and resistant to both distinct hydroxyl groups on the inositol ring. The figure shows the reactions target to improve insulin
age- and diet-induced obe- catalyzed by phosphatidylinositol 3-kinase (PI3K) and IP6K1. PI3K phosphor- sensitivity in type-2 diabetics.
sity, showing greatly dimin- ylates the 3 position of PIP2 (phosphatidylinositol-4,5-bisphosphate) to make However, a major consider-
PIP3. IP6K1 phosphorylates the phosphate group at the 5 position of IP6
ished white adipose depots. (inositol hexakisphosphate) to generate IP7. ation in the development of
As it is well known that such inhibitors is the involve-
increased adiposity is closely ment of IP6K1 in pancreatic
associated with the develop- insulin output (Illies et al.,
ment of systemic insulin resistance tant role in promoting adipocyte differen- 2007; Bhandari et al., 2008). Though it is
(Guilherme et al., 2008), the lean pheno- tiation. clear that there are many new avenues
type of the Ip6k1 knockout mice This study raises some interesting to explore, the findings reported by Chak-
confounds the interpretation of their questions regarding control of the insulin raborty et al. add another key element
enhanced insulin sensitivity. Indeed, the response at both the cellular and organ- to the complex regulation of the insulin
improved insulin sensitivity of the ismal levels. The findings by Chakraborty response.
knockout mice is more pronounced on et al. that the same signals that increase
a high fat-diet, on which the control mice the levels of PIP3 also increase the levels
develop obesity and insulin resistance. of IP7, which appears to compete with REFERENCES
Therefore, the beneficial effects of PIP3 for binding the Akt-PH domain,
Bhandari, R., Juluri, K.R., Resnick, A.C., and
IP6K1 loss on global insulin action reflect suggest a rheostat-like control over Akt
Snyder, S.H. (2008). Proc. Natl. Acad. Sci. USA
both increased cellular insulin signaling activation. Although these inositol deriva- 105, 2349–2353.
and the systemic effects of decreased tives bind with different affinities to the
Chakraborty, A., Koldobskiy, M.A., Bello, N.T.,
adiposity. The lean nature of the Ip6k1 Akt-PH domain, this model suggests Maxwell, M., Potter, J.J., Juluri, K.R., Maag, D.,
knockout mice appears to be due to that the relative localized concentrations Kim, S., Huang, A.S., Dailey, M.J., et al. (2010).
an increase in lean muscle mass and of PIP3 and IP7 directly influence the Cell 143, this issue, 897–910.
in the breakdown of fatty acids by spatial and temporal status of Akt activa- Guilherme, A., Virbasius, J.V., Puri, V., and
b oxidation. However, the authors also tion. Further studies are needed to deter- Czech, M.P. (2008). Nat. Rev. Mol. Cell Biol. 9,
demonstrate that Ip6k1 plays an impor- mine how the ratios of PIP3 to IP7 change 367–377.

862 Cell 143, December 10, 2010 ª2010 Elsevier Inc.


Harrington, L.S., Findlay, G.M., and Lamb, R.F. Luo, H.R., Huang, Y.E., Chen, J.C., Saiardi, A., Um, S.H., Frigerio, F., Watanabe, M., Picard, F.,
(2005). Trends Biochem. Sci. 30, 35–42. Iijima, M., Ye, K., Huang, Y., Nagata, E., Devreotes, Joaquin, M., Sticker, M., Fumagalli, S., Allegrini,
P., and Snyder, S.H. (2003). Cell 114, 559–572. P.R., Kozma, S.C., Auwerx, J., and Thomas, G.
Manning, B.D., and Cantley, L.C. (2007). Cell 129, (2004). Nature 431, 200–205.
Illies, C., Gromada, J., Fiume, R., Leibiger, B.,
Yu, J., Juhl, K., Yang, S.N., Barma, D.K., Falck, 1261–1274.
J.R., Saiardi, A., et al. (2007). Science 318, 1299– Taniguchi, C.M., Emanuelli, B., and Kahn, C.R. Whiteman, E.L., Cho, H., and Birnbaum, M.J.
1302. (2006). Nat. Rev. Mol. Cell Biol. 7, 85–96. (2002). Trends Endocrinol. Metab. 13, 444–451.

Consequences of mRNA
Wardrobe Malfunctions
Carol J. Wilusz1 and Jeffrey Wilusz1,*
1Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA

*Correspondence: jeffrey.wilusz@colostate.edu
DOI 10.1016/j.cell.2010.11.041

As mRNAs are generated, they are clothed with proteins to form messenger ribonucleoprotein
particles (mRNPs), which are then actively remodeled during various steps of gene expression.
Franks et al. (2010) now show that mRNP remodeling is required even for the death of an mRNA.

Although we tend to sketch mRNAs as end of its useful life, the mRNP must be 50 -30 pathway, is not as robust as once
naked molecules, they are rapidly assem- completely disassembled to allow recy- presumed. In fact, they show that XRN1
bled into messenger ribonucleoprotein cling of its components. Several recent requires that UPF1 hydrolyze ATP in order
particles (mRNPs) during transcription. studies have suggested that many of to dissociate other RNA binding factors
Proteins and protein complexes such as these dramatic changes in the mRNP before it can act on the 30 fragment.
the cap-binding complex, exon junction can be modulated through posttransla- When UPF1 ATPase activity is impaired,
complex, and nuclear poly(A)-binding tional modification and RNA chaperone XRN1 fails to efficiently degrade the
protein are specifically deposited on the activity. However, the mechanism by mRNA and the fragment accumulates
nascent transcript (Figure 1). Each of which mRNPs are finally undressed to along with its associated proteins, which
these factors has the capacity to influence allow degradation of the mRNA has until are then no longer available to bind other
downstream events such as mRNA now remained a mystery. transcripts. The authors further show
export and translation, and failure to In this issue, Franks et al. (2010) that granular structures known as pro-
assemble an appropriate mRNP may uncover a role for the ATPase activity of cessing bodies (P bodies) may be the
result in its decay through nuclear surveil- the nonsense-mediated decay (NMD) location where improperly dressed
lance pathways. Despite the ordered and factor hUPF1 in remodeling the mRNP to mRNPs are held. In addition, undegraded
precise assembly of nuclear mRNPs, allow 50 -30 exonucleolytic decay of an RNA fragments could become substrates
these complexes are rather transient, as mRNA fragment. NMD is a well-charac- for the rather mysterious process of cyto-
by the time the transcript is being actively terized mechanism that recognizes plasmic recapping (Otsuka et al., 2009) in
translated in the cytoplasm, it has a very mRNAs bearing premature termination which the 50 monophosphate of the RNA
different array of proteins associated codons and can trigger an endonucleo- fragment is replaced with a methylated
with it. The nuclear cap-binding complex lytic cleavage close to the site of prema- cap structure. This may allow translation
has been replaced by the translation ture translation termination. For this and of novel downstream open reading
initiation factor eIF4E and its associated many other decay events, it had been frames or may result in sequestration of
proteins, the poly(A) tail is now bound assumed that the 50 -30 exonuclease translation initiation factors that could
exclusively to the cytoplasmic poly(A)- XRN1 and 30 -50 exosome activity simply dramatically impact the expression of
binding protein, and, at least for normal displace any associated proteins as they many other genes.
mRNAs, exon junction complexes have plough through the transcript. The work Although the hUPF1 protein has been
dissociated and returned to the nucleus. from the Lykke-Andersen lab suggests known to be essential for NMD for a long
Moreover, as an mRNA comes to the that exonucleolytic decay, at least the time, the precise role of its ATPase activity

Cell 143, December 10, 2010 ª2010 Elsevier Inc. 863


was not clear. These new find- exon junction complex and
ings put hUPF1 in the com- induces its dissociation
pany of other ATPases such through an uncharacterized
as DBP5 and RCK/p54/ mechanism (Gehring et al.,
DHH1, which have also been 2009).
implicated in mRNP remodel- One interesting conclusion
ing events. In Saccharomyces that can be drawn from the
cerevisiae, Dbp5p is required findings of Franks et al. is
to displace the Nab2p RNA- that XRN1 does not aggres-
binding protein from the sively attack every 50 mono-
mRNA as it exits the nuclear phosphorylated RNA but, at
pore (Tran et al., 2007). Of least in some cases, must be
interest, in this case, ATPase licensed or assisted. This is
activity is not required; the supported by the existence
ADP-bound form of Dbp5p is of intermediates generated
able to displace Nab2p. The by the failure of XRN1 activity
role of DHH1, another RNA to degrade other potential
helicase, is comparatively substrates, including poly(G)
poorly understood, but it tracts or the 30 untranslated
appears to be essential for region of flavivirus transcripts
allowing an mRNA to cease (Silva et al., 2010). Why then
translation and either become would the processive XRN1
translationally silent or under- exonuclease need additional
go decay. This likely involves factors in order to degrade
a significant amount of a substrate? In the case of
mRNP remodeling, but the poly(G) tracts and the flavivi-
factors and mechanisms in- rus transcripts, it seems that
volved are yet to be strong secondary structure
characterized. blocks the enzyme, as it is
There are a number of other unable to proceed through
ways in which mRNPs can be these regions even in recon-
Figure 1. A Model for the Generic Remodeling that Takes Place
undressed to make way for during the Life Span of an mRNA
stituted reactions containing
subsequent RNA processing Upon synthesis and nuclear RNA processing, a variety of proteins are loaded just RNA and XRN1. In this
events, including competitive onto an mRNA, including the nuclear cap-binding complex (CBC) at the 50 end, case, cofactors could act to
displacement and posttrans- the exon junction complex (EJC), and the poly(A)-binding proteins NAB2, destabilize the structure and
PABPN1, and PABPC1. During passage through the nuclear pore, proteins
lational modification. For ex- such as DBP5 and TOM1 remove specific proteins from the mRNA, including allow XRN1 to process the
ample, in yeast, the export NAB2. Prior to translation, additional remodeling of the mRNP occurs, transcript. In other instances,
licensing factor Yra1 must be including exchange of the CBC on the cap with eiF4E. The assembly of trans- proteins associated with the
lation factors and movement of the ribosome on the transcript during protein
dislodged from the mRNA synthesis cause extensive remodeling of the mRNP. Finally, mRNAs targeted
RNA could sterically block
and recycled prior to translo- for decay become associated with a variety of regulatory decay factors and the enzyme, perhaps by con-
cation at the nuclear pore. often lose proteins from their 50 cap and 30 poly(A) tail prior to and during degra- cealing the free 50 end. RNA
dation by decapping factors (DCP1/2), deadenylases, and exonucleases
Yra1 is ubiquitinated by the chaperones like hUPF1 may
(XRN1 and the exosome).
nuclear pore-associated li- dissociate these inhibitory
gase Tom1, causing it to factors, allowing decay to
dissociate from the mRNP proceed. Finally, it is possible
(Iglesias et al., 2010). Soon after export, severely reduces its affinity for the mRNA that XRN1 associates with the target, but
nuclear cap and poly(A)-binding proteins cap and results in its dissociation from RNA refolding is required to allow it to
are replaced with their cytoplasmic coun- the mRNA, allowing eIF4E to replace it access the free 50 end. This type of regula-
terparts, and though translation is known (Dias et al., 2009). Finally, during the first tion occurs during processing of the yeast
to be required for this exchange, it is not round of translation, the exon junction 18S ribosomal RNA, whereby the Nob1
clear whether specific cofactors are complex must be stripped from the endonuclease associates with the tran-
required (Hosoda et al., 2006). In contrast, mRNA in order to allow passage of the script but cannot cleave until subsequent
the nuclear cap-binding complex is dis- ribosome. Even though the ribosome has structural rearrangements are complete
placed from the mRNA cap in a transla- a huge size advantage as it traverses the (Granneman et al., 2010). Whichever of
tion-independent manner once the mRNA, it appears that a specific protein, these mechanisms turns out to be correct,
mRNA enters the cytoplasm. This occurs PYM, is still necessary for effective re- one thing remains clear: undressing an
through interaction of the CBP20 subunit moval of the complex. PYM binds to both mRNA molecule is not as simple as we
of the complex with importin-b, which the ribosome and components of the once thought.

864 Cell 143, December 10, 2010 ª2010 Elsevier Inc.


REFERENCES Granneman, S., Petfalski, E., Swiatkowska, A., and Otsuka, Y., Kedersha, N.L., and Schoenberg, D.R.
Tollervey, D. (2010). EMBO J. 29, 2026–2036. (2009). Mol. Cell. Biol. 29, 2155–2167.
Dias, S.M., Wilson, K.F., Rojas, K.S., Ambrosio,
A.L., and Cerione, R.A. (2009). Nat. Struct. Mol. Hosoda, N., Lejeune, F., and Maquat, L.E. (2006). Silva, P.A., Pereira, C.F., Dalebout, T.J., Spaan,
Biol. 16, 930–937. Mol. Cell. Biol. 26, 3085–3097. W.J., and Bredenbeek, P.J. (2010). J. Virol. 84,
Franks, T.M., Singh, G., and Lykke-Andersen, J. 11395–11406.
(2010). Cell 143, this issue, 938–950. Iglesias, N., Tutucci, E., Gwizdek, C., Vinciguerra, P.,
Gehring, N.H., Lamprinaki, S., Kulozik, A.E., and Von Dach, E., Corbett, A.H., Dargemont, C., and Tran, E.J., Zhou, Y., Corbett, A.H., and Wente, S.R.
Hentze, M.W. (2009). Cell 137, 536–548. Stutz, F. (2010). Genes Dev. 24, 1927–1938. (2007). Mol. Cell 28, 850–859.

Kinases Charging to the Membrane


Michael A. Hadders1,* and Roger L. Williams1,*
1MRC Laboratory of Molecular Biology, Hills Road, Cambridge CB2 0QH, UK

*Correspondence: mhadders@mrc-lmb.cam.ac.uk (M.A.H.), rlw@mrc-lmb.cam.ac.uk (R.L.W.)


DOI 10.1016/j.cell.2010.11.044

Being at the right place and time is as fundamental to biology as it is to academic careers. In this
issue, Moravcevic and colleagues (2010) survey membrane-interacting proteins in yeast and
discover a new membrane-targeting module, the kinase associated-1 domain KA1, which ensures
that proteins are active at the correct place and time.

Proteins and their associated activities of KA1 domains in direct membrane tar- To identify new membrane-binding
must be tightly regulated in cells, both geting was not fully appreciated until now. motifs, Moravcevic and colleagues
spatially and temporally. Binding interac- Lipid-binding modules target proteins examine 62 of the 128 proteins that were
tions are a common mechanism for local- and their associated activities to previously shown to bind phosphoinositi-
izing proteins to their target sites, usually membranes. To date, more than a dozen des in yeast (Zhu et al., 2001). Using both
through protein-protein or protein-lipid membrane-interacting domains have cellular and in vitro assays, they find that
interactions. Despite the absolute impor- been identified, and several common 21 of these proteins bind membranes.
tance of protein-lipid contacts, the molec- themes for lipid interactions are becoming For five of these proteins, truncation
ular basis of these regulatory interactions apparent (Lemmon, 2008). In general, mutants pinpoint a specific region in the
remains largely obscure, as underscored membrane-binding domains can either protein involved in membrane targeting,
by a study that used yeast proteome chips recognize specific structural features of suggesting the presence of new mem-
to identify over 100 membrane-binding headgroups on lipids, as illustrated by brane-binding modules. The authors focus
proteins, none of which contained a known the binding of FYVE, PH, and PX domains on one of these proteins, the septin-asso-
lipid-interacting domain (Zhu et al., 2001). to phosphoinositides, or recognize more ciated kinase Kcc4p.
In this issue of Cell, Moravcevic and general physical properties of the mem- Septin-associated kinases are re-
colleagues analyze these membrane- brane, such as its charge and/or shape, quired for bud formation in the dividing
binding proteins and identify a new as is the case for annexins and BAR and yeast cell. The kinases localize to the bud
membrane-interacting domain in septin- C2 domains (Lemmon, 2008). These neck where they regulate the degradation
associated kinases. They demonstrate stereospecific and electrostatic interac- of the mitotic inhibitor Swe1 (Saccharo-
that this domain cooperates with protein- tions frequently cooperate with hydro- myces Wee1), thereby allowing the cells
protein interactions to target septin-asso- phobic penetration into the membrane to to proceed through mitosis (Lew, 2003).
ciated kinases to their site of action in stabilize binding by a single domain. Aside from a protein kinase domain, no
yeast. Unexpectedly, structural analysis Nevertheless, the presence of other other domain was apparent in these
of the domain shows a kinase associ- protein- or lipid-binding elements in multi- proteins. Moravcevic et al. now show
ated-1 (KA1) fold, which is also present in domain proteins can further modulate that septin-associated kinases have a
MARK/PAR1 kinases (microtubule-asso- targeting, and this cooperativity is often C-terminal membrane-binding domain
ciated protein affinity-regulating/partition- required for proper membrane localiza- and that membrane binding is required
ing-defective 1 kinases). However, the role tion of a protein. for localization to the bud neck.

Cell 143, December 10, 2010 ª2010 Elsevier Inc. 865


KA1 domain. A structural comparison,
however, highlights several differences
between the KA1 domains of Kcc4p and
AMPK proteins (Figure 1). The Kcc4p
KA1 domain has two anionic binding sites
on the surface, separated by a hydro-
phobic loop that penetrates into the
membrane. This combination of two
membrane-recognizing properties in a
single domain (i.e., hydrophobic penetra-
tion and electrostatic interaction) is
a feature of many membrane-interacting
domains such as some C2, PX, PH, and
FYVE domains (Lemmon, 2008). Interest-
ingly, in the MARK1 KA1 domain, only
one of the two positively charged anionic
binding sites is conserved, and there is
no hydrophobic loop (Tochio et al., 2006).
Instead, a single patch of positively
charged residues extends down along
one side of the molecule, suggesting
different membrane-binding orientations
for the KA1 domains of Kcc4p and
MARK1 (Figure 1). Future studies are
needed to determine whether these differ-
ences in orientation have functional conse-
Figure 1. Putative Membrane Interactions of KA1 Domains
The yeast septin-associated kinases, including Kccp4, contain a membrane-interacting domain that
quences or whether the two types of
recognizes negatively charged phospholipids, such as phosphatidylserine (Moravcevic et al., 2010). domains are functionally interchangeable.
(A) Structural analysis of this domain shows that it is a kinase associated-1 (KA1) domain, which is also The diversity of KA1 domains may
found in MARK/PAR1 kinases (microtubule-associated protein affinity-regulating/partitioning-defective reflect the different roles that they play in
1 kinases). Unlike Kccp4, MARK/PAR1 kinases also possess a ubiquitin-associated (UBA) domain.
(B) Residues involved in membrane binding are labeled and depicted as ball and stick, as is the their respective proteins. In Kcc4p, the
hydrophobic loop that is proposed to insert into the membrane. KA1 domain alone is insufficient for proper
localization, and it is only in the context of
Unlike well-characterized membrane- a five-stranded antiparallel b sheet. They an intact Kcc4p protein that membrane
binding domains that rely on stereospe- were first described as the C-terminal and septin interactions cooperate to target
cific interactions, the membrane-binding domains of MARK/PAR1 kinases, which Kcc4p activity to the bud neck. This type of
domains of septin-associated kinases are related to the AMP-activated protein cooperation in which two input signals
appear to recognize a broad range of kinase (AMPK) family of Ser/Thr protein (e.g., membrane and septin binding) are
anionic phospholipids; they interact kinases. MARK/PAR1 kinases play simultaneously required for output (e.g.,
in vitro with phosphoinositides, phospha- diverse cellular roles and have been kinase activity at the bud neck) is called
tidic acid, and phosphatidylserine. implicated in cancer and Alzheimer’s ‘‘coincidence detection.’’
However, given the abundance of phos- disease (Marx et al., 2010). Until now, In contrast, the KA1 domain of the
phatidylserine in the cytoplasmic mem- the precise role of KA1 domains was MARK kinases does not appear to work
brane, this phospholipid is the likely target unclear, although it was proposed that as a coincidence detector. For several
for this new membrane-binding domain in this domain might play an autoinhibitory MARK isoforms, Göransson et al.
cells. Indeed, Moravcevic and colleagues role in regulating kinase activity through demonstrated that the cellular localiza-
elegantly demonstrate that, in a mutant intramolecular interactions (Marx et al., tion depends on binding to 14-3-3
yeast strain unable to produce phosphati- 2010). proteins (Göransson et al., 2006). These
dylserine, the septin-associated kinase Moravcevic and colleagues have now adaptor proteins modulate the activity of
fails to localize to the bud neck. re-examined the role of the KA1 domain their target proteins by binding to phos-
To further understand the mechanism in several human AMPK proteins. In all phorylated serine or threonine residues.
of membrane binding by Kcc4p, the cases, they show that the KA1 domain In the case of MARK kinases, association
authors solve the structure of the Kcc4p binds anionic phospholipids in vitro and with a 14-3-3 protein sequesters the
membrane-binding domain by X-ray mediates membrane localization in cells, kinases to the cytoplasm. Disassociation
crystallography. Surprisingly, this domain establishing the KA1 domain as a bona fide then results in relocalization of the kinase
adopts a KA1 fold (Figure 1). KA1 conserved membrane-binding module. to the plasma membrane, which depends
domains have 100 residues and consist The authors have also determined on its KA1 domain. The data presented
of two helices packed on one side of the crystal structure of the MARK1 by Moravcevic and colleagues now

866 Cell 143, December 10, 2010 ª2010 Elsevier Inc.


show that this membrane relocalization is In summary, the KA1 domain joins the Komander, D., Fairservice, A., Deak, M., Kular,
due to the binding of the KA1 domain to growing list of membrane-targeting G.S., Prescott, A.R., Peter Downes, C., Safrany,
S.T., Alessi, D.R., and van Aalten, D.M. (2004).
phosphatidylserine. Although it is still domains with broad specificity for anionic
EMBO J. 23, 3918–3928.
unclear how the 14-3-3 protein prevents phospholipids and the growing list of
Lemmon, M.A. (2008). Nat. Rev. Mol. Cell Biol. 9,
membrane binding, taken together the coincidence detectors involved in lipid
99–111.
data suggest that for MARK kinases, the recognition. The fact that this module
Lew, D.J. (2003). Curr. Opin. Cell Biol. 15, 648–653.
14-3-3 protein functions as part of now turns out to be present in several
Marx, A., Nugoor, C., Panneerselvam, S., and
a switch that regulates the shuttling of membrane-interacting proteins that were Mandelkow, E. (2010). FASEB J. 24, 1637–1648.
MARK kinases between a membrane- previously overlooked in a large screen Moravcevic, K., Mendrola, J.M., Schmitz, K.R.,
bound and a cytoplasmic state. Future for lipid interactors (Zhu et al., 2001) Wang, Y.-H., Slochower, D., Janmey, P.A., and
studies are needed to determine the suggests the exciting possibility that Lemmon, M.A. (2010). Cell 143, this issue, 966–
functional relevance of this relocalization many unidentified membrane-interacting 977.
and whether it targets the kinase to domains await discovery. Tochio, N., Koshiba, S., Kobayashi, N., Inoue, M.,
specific substrates at the plasma Yabuki, T., Aoki, M., Seki, E., Matsuda, T., Tomo,
Y., Motoda, Y., et al. (2006). Protein Sci. 15,
membrane. Like 3-phosphoinositide-
REFERENCES 2534–2543.
dependent kinase (PDK1) (Komander
Zhu, H., Bilgin, M., Bangham, R., Hall, D., Casa-
et al., 2004), the MARK kinases may Göransson, O., Deak, M., Wullschleger, S., mayor, A., Bertone, P., Lan, N., Jansen, R., Bidling-
have roles both at the membrane and in Morrice, N.A., Prescott, A.R., and Alessi, D.R. maier, S., Houfek, T., et al. (2001). Science 293,
the cytosol. (2006). J. Cell Sci. 119, 4059–4070. 2101–2105.

Exposing Contingency Plans


for Kinase Networks
Aileen M. Klein,1 Elhadji M. Dioum,1 and Melanie H. Cobb1,*
1Department of Pharmacology, UT Southwestern Medical Center at Dallas, Dallas, TX 75390, USA

*Correspondence: melanie.cobb@utsouthwestern.edu
DOI 10.1016/j.cell.2010.11.046

Understanding how signaling pathways are interconnected is vital for characterizing mechanisms
of normal development and disease pathogenesis. In this issue, Van Wageningen et al. (2010)
examine phosphorylation networks in Sacharromyces cerevisiae with genome-wide expression
profiling to identify recurring themes in signaling redundancy.

Reversible posttranslational modifica- modifications are more realistically in the budding yeast Saccharomyces
tions, such as phosphorylation, provide viewed as a network in which individual cerevisiae and, in the process, uncover a
practical mechanisms to transmit infor- signaling cascades are interconnected recurrent regulatory motif that links phos-
mation from the extracellular milieu to by common substrates and interdepen- phorylation pathways together to ensure
regulatory centers inside of the cell. dent regulation. Indeed, understanding robust responses.
Phosphorylation pathways, comprised of the biological significance of a regulated Two genes ‘‘interact’’ when disrupting
kinases, phosphatases, and their sub- event in the life of a multicellular organism, both genes simultaneously increases or
strates, are frequently studied as linear such as a response to inflammation, or the decreases the growth of the organism
entities in isolation from their surrounding etiology of a complex human disease, compared to that predicted for the combi-
cellular context (Chen and Thorner, 2007; such as cancer, demands detailed knowl- nation of the single mutants (Figure 1A)
Fiedler et al., 2009). Although this edge of network properties of signaling (Dixon et al., 2009). Such interactions illu-
simplistic treatment has identified thou- cascades. In this issue of Cell, van Wage- minate features of a signaling network,
sands of kinase and phosphatase sub- ningen and colleagues use global gene including redundancies. Redundancy
strates, many of which display tissue expression analysis to characterize the occurs when the functions of two compo-
specificity (Old et al., 2009), regulatory network properties of kinase pathways nents in a pathway overlap significantly

Cell 143, December 10, 2010 ª2010 Elsevier Inc. 867


and thus can compensate for wall integrity and osmotic
each other when one is lost response pathways. Two
(Costanzo et al., 2010). In other cases (i.e., PTC1-PTC2
general, redundancies are and PPH3-PTC1) exhibited
predicted only when two what the authors call ‘‘quanti-
proteins share significant tative redundancy’’ (Fig-
sequence homology. ure 1B). In these cases, the
Most large-scale screens expression profile of one
that identify genetic interac- single mutant resembles that
tions perform combinatorial of wild-type, whereas disrupt-
deletion (or knockdown) of ing the second factor signifi-
gene pairs and then compare cantly alters the expression
the growth of the ‘‘double of a limited number of genes.
mutants’’ to that of the single Then, deleting both genes
mutants (Costanzo et al., simultaneously exacerbates
2010; Whitehurst et al., 2007). the altered gene expression
These ‘‘synthetic lethal’’ of the single mutant (Fig-
screens provide insights into ure 1B). To explore the mech-
the network’s landscape but anism underlying ‘‘quantita-
often do not illuminate the tive redundancy,’’ van
underlying molecular mecha- Wageningen et al. demon-
nisms vital to decode the logic strate that PTC1 and PTC2
of signaling networks. inactivate a common sub-
S. cerevisiae has 141 genes strate (i.e., the mitogen-acti-
encoding protein kinases vated protein kinase [MAPK]
and 38 genes encoding phos- HOG1) but with different effi-
phoprotein phosphatases. ciencies.
Remarkably, 150 of these The remaining cases of in-
genes are dispensable for teracting genes display more
growth because yeast strains complex and unexpected
with mutations in these genes behavior, which the authors
are still viable (Fiedler et al., call ‘‘mixed epistasis.’’ In
2009). A previous synthetic these cases, changes in
lethal screen identified ge- gene expression patterns are
netic interactions between different for the single and
24 pairs of these signaling double mutants and, as a
components. To identify result, are not readily predict-
principles underlying these able. In double mutants, both
genetic interactions, van Figure 1. Three Categories of Genetic Interactions in Phosphoryla- full and quantitative redun-
Wageningen et al. use global tion Networks dancy are observed often in
gene expression as the A genetic interaction occurs when two single-mutant phenotypes are insuffi- conjunction with opposite
cient to predict the phenotype of the double mutant.
readout of the cell’s response effects on the expression
(A) When two genes are ‘‘completely redundant,’’ disrupting either gene alone
to mutating specific interact- has no effect on growth and gene expression, but disrupting both genes of some genes. Remarkably,
ing pairs: two kinases, two severely alters both properties. mixed epistasis, which in-
phosphatases, or a kinase- (B) Two genes can also exhibit ‘‘quantitative redundancy’’ (Van Wageningen cludes kinase-phosphatase
et al., 2010) in which the phenotype of a single mutant is greatly exacerbated
phosphatase pair. First, they in the double mutant. pairs, is the most prevalent
generate gene expression (C) The kinases Fus3 and Kss1 in Saccharomyces cerevisiae display a third genetic interaction found.
profiles for each of the 150 type of genetic interaction called ‘‘mixed epistasis.’’ Fus3 and Kss1 can func- To identify mechanisms
tion redundantly in the mating response, but Fus3 normally represses the fila-
strains with one gene dis- mentous growth pathway, leading to different gene expression profiles in the
that could lead to mixed epis-
rupted. They then compare single and double knockouts of these genes. tasis, van Wageningen and
these profiles to those of colleagues then use mathe-
strains with two genes dis- matical modeling to search
rupted (i.e., double mutants). In total, redundancy (Figure 1A). For example, for network topologies consistent with
they query more than 20 negatively inter- deleting either protein-tyrosine phospha- the observed expression phenotypes.
acting kinases and/or phosphatases by tase PTP2 or PTP3 has no effect on gene Their findings suggest that pairs of genes
DNA microarray analysis. expression, but disrupting both phospha- showing mixed epistasis have two proper-
Sixteen of the double-mutant strains are tases simultaneously significantly alters ties. First, the functions of the two genes
viable, and of those, four display simple the expression of genes regulating cell partially overlap; second, one gene

868 Cell 143, December 10, 2010 ª2010 Elsevier Inc.


represses or inhibits the other. The clear- epistasis utilizes nonhomologous pro- ningen and colleagues? First, functional
est validation for these characteristics teins to achieve the same outcome redundancy is not limited to proteins
comes from the two MAPKs Fus3 and accomplished with homologous proteins, with primary sequence similarity; in fact,
Kss1 (Figure 1C). Fus3 and Kss1 are providing an additional mechanism for functional redundancy is even common
both activated by the same MAPK kinase ensuring robust signaling. Of interest, it among nonhomologous proteins. Sec-
kinase Ste11 in a scaffold-restricted is interdependencies such as these that ond, the wiring of signaling pathways in
manner. Fus3 mediates pheromone- make it difficult to predict relative contri- the cell can easily facilitate a broad
induced mating of yeast, whereas Kss1 butions of different regulators when spectrum of redundancies from complete
regulates filamentous growth. However, studying individual pathways in isolation. compensation to mixed epistasis.
in the absence of Fus3, Kss1 can also A continuing debate in the field is
support mating at an extremely low rate. whether or not findings from single-celled
REFERENCES
Thus, these two largely independent path- organisms, such as S. cerevisiae, will
ways have partially overlapping functions. be relevant to signaling networks in more Breitkreutz, A., Choi, H., Sharom, J.R., Boucher, L.,
Fus3 phosphorylates and promotes the complex metazoans. Although recent Neduva, V., Larsen, B., Lin, Z.Y., Breitkreutz, B.J.,
degradation of a factor necessary for studies suggest that information gained Stark, C., Liu, G., et al. (2010). Science 328,
filamentous growth, inhibiting the function from experiments with S. cerevisiae may 1043–1046.
of Kss1. Furthermore, Fus3 apparently not provide a good platform for homology Chen, R.E., and Thorner, J. (2007). Biochim.
induces a phosphatase that selectively mapping to multicellular organisms (Dixon Biophys. Acta. 1773, 1311–1340.
dephosphorylates and inactivates Kss1 et al., 2009), a reductionist approach may Costanzo, M., Baryshnikova, A., Bellay, J., Kim, Y.,
(Figure 1C) (Chen and Thorner, 2007). still have predictive power for dissecting Spear, E.D., Sevier, C.S., Ding, H.M., Koh, J.L.Y.,
Therefore, Fus3 inhibits the function and pathway interactions in metazoans. For Toufighi, K., Mostafavi, S., et al. (2010). Science
327, 425–431.
activity of Kss1. example, previous studies in S. cerevisiae
Other regulatory pairs displaying identified negative interactions between Dixon, S.J., Costanzo, M., Baryshnikova, A.,
Andrews, B., and Boone, C. (2009). Annu. Rev.
‘‘mixed epistasis’’ are from signaling Fus3 and the MAPK Hog1 (Hall et al.,
Genet. 43, 601–625.
pathways that are known to act on 1996). Of interest, this interaction and the
Fiedler, D., Braberg, H., Mehta, M., Chechik, G.,
different cellular events. Although the new one observed for Fus3 and Kss1 are
Cagney, G., Mukherjee, P., Silva, A.C., Shales,
mechanisms conferring mixed epistasis reminiscent of the relationship between M., Collins, S.R., van Wageningen, S., et al.
to these other pairs are not immediately two distinct MAPK pathways in the (2009). Cell 136, 952–963.
obvious or already validated by the litera- mammalian myogenic program, the p38 Hall, J.P., Cherkasova, V., Elion, E., Gustin, M.C.,
ture, several of these interactions pinpoint (a mammalian homolog of Hog1) and and Winter, E. (1996). Mol. Cell. Biol. 16,
well-known communications between ERK1/2 pathways. In mouse muscle 6715–6723.
environmental sensing and regulatory progenitor cells (i.e., myoblasts), reduced Old, W.M., Shabb, J.B., Houel, S., Wang, H.,
processes. For example, connections growth factor stimulation from serum Couts, K.L., Yen, C.Y., Litman, E.S., Croy, C.H.,
between energy sensing and the cell- activates p38, which then triggers tran- Meyer-Arendt, K., Miranda, J.G., et al. (2009).
cycle machinery are well known (Breitk- scription of early regulators of differentia- Mol. Cell 34, 115–131.
reutz et al., 2010). Now, van Wageningen tion. ERK1/2 are indirectly inactivated Van Wageningen, S., Kemmeren, P., Lijnzaad, P.,
and colleagues find that ELM1 (or HSL1), in a p38-dependent manner, similar to Benschop, J.J., de Castro, I.J., van Leenen, D.,
Groot Koerkamp, M.J., Ko, C., Miles, A.J., Brabers,
a kinase that phosphorylates and how Fus3 inhibits the activity of Kss1.
N.A.C., et al. (2010). Cell 143, this issue, 991–1004.
increases the activity of the AMP-acti- However, later in differentiation, ERK1/2
Whitehurst, A.W., Bodemann, B.O., Cardenas, J.,
vated protein kinase (AMPK), displays stimulation promotes the differentiated
Ferguson, D., Girard, L., Peyton, M., Minna, J.D.,
mixed epistasis with MIH1, the budding state (Wu et al., 2000). Undoubtedly,
Michnoff, C., Hao, W.H., Roth, M.G., et al. (2007).
yeast homolog of the cell-cycle phospha- many more examples of this regulatory Nature 446, 815–819.
tase Cdc25. The interaction between motif have and will be identified.
Wu, Z.G., Woodring, P.J., Bhakta, K.S., Tamura,
these genes reveals the direct link What general conclusions can we K., Wen, F., Feramisco, J.R., Karin, M., Wang,
between energy sensing by AMPK and infer from the global perspective of J.Y.J., and Puri, P.L. (2000). Mol. Cell. Biol. 20,
cell-cycle control. This example of mixed kinase signaling provided by van Wage- 3951–3964.

Cell 143, December 10, 2010 ª2010 Elsevier Inc. 869


Leading Edge
Essay

Lipid Trafficking sans Vesicles:


Where, Why, How?
William A. Prinz1,*
1Laboratory of Cell Biochemistry and Biology, National Institute of Diabetes and Digestive and Kidney Diseases,

National Institutes of Health, Bethesda, MD 20892, USA


*Correspondence: williamp@intra.niddk.nih.gov
DOI 10.1016/j.cell.2010.11.031

Eukaryotic cells possess a remarkable diversity of lipids, which distribute among cellular membranes
by well-characterized vesicle trafficking pathways. However, transport of lipids by alternate, or
‘‘nonvesicular,’’ routes is also critical for lipid synthesis, metabolism, and proper membrane partition-
ing. In the past few years, considerable progress has been made in characterizing the mechanisms of
nonvesicular lipid transport and how it may go awry in particular diseases, but many fundamental
questions remain for this rising field.
A typical higher eukaryotic cell contains obscure and difficult to characterize. In ulum (ER) to the Golgi (Kok et al., 1998; Fu-
more than 1000 different lipid species. the past few years, researchers have nato and Riezman, 2001; Hanada et al.,
These lipids are not homogenously distrib- made significant progress toward under- 2003), GlcCer transfer from the Golgi
uted among intracellular membranes, but standing how and why nonvesicular lipid complex to the ER and plasma membrane
instead each organelle has a characteristic trafficking occurs. This Essay summarizes (Halter et al., 2007; D’Angelo et al., 2007),
lipid composition that is required for its the current state of the field and the major and sterols from the plasma membrane to
proper function. For example, cholesterol challenges for its future. endocytic recycling compartment (Mesmin
and sphingolipids are highly enriched in and Maxfield, 2009). For example, studies
the plasma membrane and endosomes, How Much Nonvesicular Lipid using dehydroergosterol, a fluorescent
and indeed, many diseases, such as Trafficking Occurs in Cells? analog of cholesterol, found that, when
atherosclerosis, type II diabetes, and lyso- The first studies suggesting the existence this sterol is added to cells, it initially
somal storage disorders, are associated of nonvesicular lipid exchange pathways incorporates into the plasma membrane
with defects in maintaining the correct in the cell examined the movement of but then moves to the endocytic
distribution of intracellular lipids. How do newly synthesized lipids from the endo- recycling compartment by a nonvesicular,
these hydrophobic molecules shuttle plasmic reticulum (ER), where they are energy-independent pathway. Dehydroer-
between intracellular membranes inside made, to the plasma membrane. Drugs gosterol equilibrates between the
the aqueous milieu of the cell? that halt vesicular trafficking do not stop plasma membrane and endocytic recy-
Although trafficking largely determines lipid transfer from the ER to the plasma cling compartment quite quickly—within
the intracellular distribution of most lipids, membrane, indicating that some lipids, 2–3 min—and astonishingly, an estimated
we currently understand less about lipid including phosphatidylcholine (PC), one million dehydroergosterol molecules
trafficking than we do about protein traf- phosphoatidylethanolamine (PE), choles- exchange between these compart-
ficking. Nevertheless, proteins and lipids terol, and glucosylceramide (GlcCer), can ments each second (Maxfield and Mondal,
do share similar properties. Both lipids move between the ER and plasma 2006).
and integral membrane proteins move membrane by nonvesicular pathways. Collectively, these and many other
between organelles in membrane-en- Moreover, these pathways have substan- studies indicate that the cell possesses
closed sacs called transport vesicles, tial capacity because the rate of lipid numerous pathways of nonvesicular lipid
and there is growing evidence that lipids, transfer does not decrease when vesic- transport, and more pathways will prob-
like proteins, are sorted during the forma- ular trafficking is blocked (Sleight and ably be discovered in the future. However,
tion of transport vesicles. Pagano, 1983; Kaplan and Simoni, in most cases, we still are uncertain about
However, unlike proteins, lipids can 1985a, 1985b; Warnock et al., 1994). of how much nonvesicular pathways
rapidly and efficiently move between Nevertheless, it remains unclear what contribute to the total lipid exchange
cellular membranes by routes independent fraction of the lipid exchange between inside of a cell. Are the nonvesicular path-
of transport vesicle, or ‘‘nonvesicular the ER and plasma membrane is nonve- ways needed for exchanging a large
transport’’ pathways. This important differ- sicular when vesicular trafficking is not proportion of lipids between organelles,
ence between protein and lipid trafficking blocked. or do only a small fraction of lipids
is not widely appreciated, in part, because More recently, studies have reported move by nonvesicular mechanisms? In
the roles and mechanisms of nonvesicular strong evidence for nonvesicular transfer addition, some classes of lipids, such as
lipid exchange have, in many cases, been of ceramides from the endoplasmic retic- complex glycolipids, gangliosides, and

870 Cell 143, December 10, 2010 ª2010 Elsevier Inc.


sphingolipids, may transfer by only vesic- synthesized, to the Golgi complex, where mammalian cells. Sphingomyelin is
ular routes (Wattenberg, 1990; Hecht- they are converted into glycolipids and synthesized in the Golgi complex, but ce-
berger and Daum, 1995). sphingolipids, may regulate the produc- ramide is made in the ER. Therefore, to
tion of these lipids. Finally, it is possible produce sphingomyelin, ceramide must
Roles of Nonvesicular Lipid that nonvesicular lipid transfer is required be transported from the ER to the Golgi
Trafficking in Cells for the transmission of a lipid as part of complex, and this is accomplished by
Nonvesicular lipid trafficking serves at a signaling or regulatory pathway. For CERT, the ceramide transport protein
least four important functions in cells. example, diacylglycerol activates protein (Hanada et al., 2009).
First, it provides lipids that are needed kinase C and ceramides serve as signal- CERT is expressed ubiquitously in
for membrane biogenesis in organelles ing molecules to regulate differentiation, higher eukaryotes, but it is not present in
that cannot obtain sufficient lipids from proliferation, programmed cell death, yeast. CERT was identified from a mutant
vesicular trafficking. Mitochondria, chlo- and apoptosis. cell line of Chinese hamster ovary cells,
roplasts, and lipid droplets lack most of called LY-A, which has low levels of sphin-
the enzymes needed to make certain Mechanisms of Nonvesicular Lipid gomyelin (Hanada et al., 2003). Studies
lipids required for their biogenesis. These Trafficking found that, although LY-A mutant cells
organelles are not connected to the rest Lipid monomers can exchange spontane- make sphingomyelin at a reduced rate,
of the cell by vesicular trafficking path- ously between membranes by simply these cells produce normal amounts of
ways and thus rely on nonvesicular traf- diffusing through the aqueous phase enzymes that synthesize sphingomyelin
ficking pathways to obtain these lipids. (Figure 1A). However, for most classes of (i.e., sphingomyelin synthase) and the
Indeed, many studies show that lipids lipids, this process occurs too slowly to sphingomyelin precursors, ceramide and
exchange between the ER and mitochon- be physiologically relevant; for example, PC. These results suggested that LY-A
dria or chloroplasts by nonvesicular routes most glycerolipids and sphingolipids spon- cells have a defect in the nonvesicular
(Voelker, 2009; Benning, 2009). Less is taneously exchange between membranes transfer of ceramide from the ER to the
known about lipid transfer among lipid with half-times > 40 hr. The rate-limiting Golgi complex. The gene that comple-
droplets or between lipid droplets and step in this process is lipid desorption mented the cell’s defect was isolated
other organelles, but these pathways are from a membrane, and thus proteins that and named CERT. Disruption of the
almost certainly nonvesicular as well. accelerate lipid transfer may increase the CERT gene in mice results in death at
There is also evidence for nonvesicular rate of lipid egress from the membrane. approximately embryonic day 11.5
lipid exchange between the ER and perox- Lipid transfer between membranes (Wang et al., 2009), and flies lacking
isomes (Raychaudhuri and Prinz, 2008). may also occur when two membranes CERT have a dramatic decrease in ceram-
Nonvesicular transport also helps to collide (Figure 1B). Although the mecha- ide phosphoethanolamine, the fly analog
maintain the proper level of a lipid in an nism of lipid exchange during collision is of sphingomyelin (Rao et al., 2007).
organelle or domain of an organelle. not well understood, one model is that CERT encodes a 68 kDa protein that
Compared to vesicular routes, one a lipid must be ‘‘activated,’’ or partially has three domains, an N-terminal PH
obvious advantage of nonvesicular traf- extended from the bilayer, prior to colli- (pleckstrin homology) domain, a FFAT
ficking is that it can rapidly move lipids sion (Steck et al., 2002). This activation (two phenylalanines in an acidic tract)
between specific compartments in cells increases the probability of transfer to motif, and a C-terminal START (steroido-
without having to also transfer integral a second membrane during collision. genic acute regulatory protein [StAR]-
membrane proteins. This may be particu- Activation could be stochastic, resulting related) domain. The PH domain binds to
larly important for lipids, such as choles- from the thermal motion that causes lipids phosphoinositides (PIPs), whereas the
terol, which can be toxic to cells. Cells to bounce or bob in a bilayer, or it could be FFAT motif associates with proteins on
use a number of mechanisms to rapidly mediated by a protein. the ER called VAPs (vesicle-associated
decrease cholesterol levels when they Proteins clearly facilitate the lipid non- membrane protein-associated proteins).
are too high, such as effluxing cholesterol vesicular transport between membranes. The START domain is the portion of the
out of cells to external lipoproteins and Although this process has been well char- protein that transports lipids, and it binds
producing cholesteryl esters (i.e., ester acterized in vitro, studies are only begin- a single molecule of ceramide in a hydro-
linkages between the hydroxyl group of ning to unravel the mechanisms for these phobic cavity (Kudo et al., 2008).
cholesterol and the carboxylate group of pathways inside the cell (Voelker, 2009; CERT facilitates the movement of ce-
a fatty acid), which are stored in lipid drop- Benning, 2009). Nevertheless, in the three ramide between liposomes in vitro
lets. Nonvesicular transport of cholesterol cases described below, specific details (Hanada et al., 2003). The PH domain
probably provides a route to move choles- have emerged, including how defects in and FFAT motif in CERT target it to the
terol quickly and efficiently to the enzymes these lipid trafficking pathways cause ER and Golgi complex, respectively.
that perform these reactions without dis- disease. Thus, in vivo CERT probably extracts ce-
rupting vesicular trafficking. CERT, a Typical Lipid Transport ramide from the ER, shuttles it through
Third, nonvesicular lipid trafficking may Protein? the cytoplasm, and delivers it to the Golgi
also regulate lipid metabolism. For Ceramide is the precursor of sphingoli- complex. In general, proteins that
example, the nonvesicular transfer of ce- pids, including sphingomyelin, an abun- mediate lipid transfer by this mechanism
ramides from the ER, where they are dant lipid in the plasma membrane of all are called lipid transfer proteins (LTPs)

Cell 143, December 10, 2010 ª2010 Elsevier Inc. 871


complex, some LTPs are enriched at
these membrane junctions, including the
oxysterol-binding protein (OSBP) ORP1L
in mammals and most of the OSBP-
related proteins in yeast (the Osh proteins)
(Levine and Munro, 2001; Loewen et al.,
2003; Rocha et al., 2009; Schulz et al.,
2009).
CERT is part of a large family of proteins
that contain START domains, and many
members of this family can facilitate lipid
transfer between membranes in vitro. In
addition, there are approximately four
other large families of LTPs, and most cells
express numerous LTPs (D’Angelo et al.,
2008; Lev, 2010). Some LTPs have high
specificity and bind only a few lipids,
whereas others can associate with a broad
range of lipids. The different families of
LTPs are quite diverse, with few similarities
in sequence or structure. However, all
LTPs share the ability to bind lipid mono-
mers with a stoichiometry of one lipid for
each protein. In addition, all LTPs bind the
lipid monomer in a pocket covered with
a flexible ‘‘lid’’ domain that shields the
associated lipid from the aqueous phase
(Figure 1C). As with CERT, lipid exchange
by LTPs does not require energy.
A major controversy in the field is
whether the primary function of many
Figure 1. Possible Mechanisms of Nonvesicular Lipid Exchange between Membranes LTPs in cells is to transfer lipids between
(A and B) Lipids can spontaneously exchange between two membranes without the assistance of membranes, as they do in vitro, or
proteins. (A) Monomers can diffuse through the aqueous phase or (B) during the collision of two-
membrane collision after the lipid is ‘‘activated.’’ whether they serve another main purpose
(C–G) (C) Lipid transport proteins (LTPs) can also exchange lipids between membranes and organelles. in cells. Aside from CERT, there is indeed
LTPs have a lipid-binding domain (blue) and, many times, targeting domains (purple) that may direct lipid compelling evidence that other LTPs,
transfer to particular membranes by binding to lipids or proteins. Lipids may exchange at membrane
contact sites where two membranes come together in close proximity. Protein complexes may facilitate
such as FAPP2 (Golgi-associated four-
this process (D) by forming a tunnel that allows lipids to diffuse between the membranes, (E) by promoting phosphate adaptor protein 2), NPC2
lipid desorption from one membrane, (F) by activating lipids prior to membrane collision, or (G) by (Niemann-Pick disease, type C2), and
promoting transient membrane hemifusion.
some oxysterol-binding proteins in yeast,
transfer lipids in cells (Yamaji et al., 2008;
(Figure 1C). The consumption of ceramide mostly at regions where membranes are D’Angelo et al., 2008; Prinz, 2007). That
in the Golgi complex to produce sphingo- closely apposed and come within said, many LTPs do not appear to trans-
myelin probably drives the directionality 20 nm of each other. Called membrane port lipids in cells but rather serve as lipid
of the ceramide transport. contact sites or MCSs, these junctions sensors or regulate lipid metabolism and
Ceramide transfer by CERT in vitro does are present ubiquitously in all cells and signaling by presenting lipids to metabolic
not require energy. Surprisingly, however, are frequently found between the ER and enzymes. For example, the Sec14 super-
ATP depletion blocks ceramide transport a second organelle (Levine and Loewen, family of LTPs has been proposed to
by CERT in cells (Hanada et al., 2003), 2006). present phosphoinositol to kinases that
and the role that energy plays in CERT At membrane contact sites between produce PIPs, and thus these LTPs regu-
function in vivo remains an interesting, the ER and Golgi complex, CERT would late many membrane trafficking and
unsolved mystery. The rate-limiting step have to diffuse only a small distance, or signaling events that require PIPs (Bank-
for ceramide transport by CERT is likely it may even bind both membranes simul- aitis et al., 2010).
diffusion through the cytosol. This is prob- taneously using its two targeting domains, Lipid Exchange between the ER
ably true of other LTPs as well. PH and FFAT (Hanada et al., 2009). and Mitochondria
Nevertheless, it is unlikely that CERT or Although it is still unknown for certain Nonvesicular lipid trafficking that occurs
other LTPs diffuse long distances through whether CERT localizes to membrane at membrane contact sites does not
the cytosol. Rather, they probably operate contact sites between the ER and Golgi always require soluble LTPs. Indeed, lipid

872 Cell 143, December 10, 2010 ª2010 Elsevier Inc.


exchange between the ER and mitochon- and mitochondria may occur at stand and begin developing treatments for
dria probably occurs independently of membrane contact sites. First, protein many diseases caused by defects in lipid
LTPs. Lipid transport between these complexes in the two organelles could transport.
organelles is critical for the synthesis of interact to form a type of hydrophobic Cholesterol Transfer by NPC1
phosphatidylcholine (PC) and phosphoa- tunnel or conduit that allows lipids to and NPC2
tidylethanolamine (PE), two of the most passively diffuse between the two Low-density lipoproteins (LDLs) transport
abundant lipids in the membranes of membranes with little or no contact with cholesterol and other lipids through the
eukaryotes. In one of the two major path- the aqueous phase (Figure 1D). Second, bloodstream, and receptor-mediated
ways for producing PC, the first step is the a membrane protein complex at a contact endocytosis of LDLs serves as a major
synthesis of phosphatidylserine (PS), site could use energy to facilitate lipid source of cholesterol in mammalian cells.
which occurs at the ER. PS is then trans- desorption from one of the membranes. When endocytosed LDL reaches late en-
ferred to the inner mitochondrial mem- The probability that the lipid then diffuses dosome/lysosome compartments, cho-
brane, where it is decarboxylated to into the adjacent membrane is compa- lesteryl esters in these particles are
form PE, the precursor of PC. However, rable to that of it diffusing back into original hydrolyzed and the resulting cholesterol
the enzymes that convert PE to PC reside membrane (Figure 1E), leading to a net is subsequently trafficked to the rest of
back in the ER, and thus to make PC, the transfer of lipid from one membrane to the cell. Nonvesicular mechanisms trans-
PE must be returned to the ER from the the other. Third, if lipid transfer occurs by port cholesterol from internal membranes
mitochondrial inner membrane. Conse- an activated collision mechanism, then to the outer membrane of the late endo-
quently, producing PE and PC by this a protein complex could also promote lipid some/lysosome and then eventually out
pathway requires multiple nonvesicular activation and increase the chance of lipid of the organelle.
lipid transfer steps. Remarkably, yeast exchange during membrane collision Two proteins required for this type of
mutants that can make PE and PC solely (Figure 1F). Membranes at contact sites cholesterol transport are NPC1 and
by this pathway grow as well as wild- may not be held a fixed distance and NPC2. These proteins were identified by
type cells and have similar levels of PE may frequently collide. A fourth possibility studies on patients with Niemann-Pick
and PC (Trotter et al., 1995). These results is that transmembrane proteins on two type C, a rare autosomal recessive lyso-
indicate that nonvesicular lipid transfer different organelles bring two membranes somal storage disease in which cholesterol
between ER and mitochondria must be in close apposition so that they undergo and other lipids accumulate in late endo-
highly efficient. transient hemifusion (Figure 1G). Lipids somes/lysosomes. NPC1 is an integral
Surprisingly, phospholipid exchange could then easily diffuse between the membrane protein with 13 putative trans-
between the ER and mitochondria hemifused membranes without contact- membrane domains that reside in the
requires neither cytosolic factors nor ing the aqueous phase. outer membrane of late endosomes/
energy. It is thought to occur at special- Defects in lipid transport to mitochon- lysosomes. In contrast, NPC2 is a small
ized regions of the ER called mitochon- dria cause multiple diseases. For soluble protein in the lumen of these organ-
dria-associated membranes (MAMs), example, some forms of congenital elles. NPC2 is an LTP that facilitates
which are closely apposed to mitochon- adrenal hyperplasia, which is character- cholesterol transport between membranes
dria (Choi et al., 2006). An important ques- ized by an impaired ability to produce the in vitro (Cheruku et al., 2006). In cells, it
tion in the field is how these membrane steroid cortisol, are caused by defects in probably transfers cholesterol between
contact sites form. In mammals, a number cholesterol transport to the inner mito- internal membranes in the late endosome/
of proteins, such as mitofusins, GRP75 chondrial membranes. Steroids are lysosome and then hands it off to NPC1 in
(glucose-regulated protein 75), and synthesized from cholesterol, and the first the outer membrane (Infante et al., 2008;
PACS2 (phosphofurin acidic cluster sort- step in this process occurs in the inner Kwon et al., 2009; Wang et al., 2010).
ing protein 2), have been proposed to mitochondrial membranes. Transporting NPC1 may then facilitate the egress of
mediate contacts between the MAM and cholesterol to the inner mitochondrial cholesterol from the late endosome/
mitochondria, but whether any of these membranes requires the LTP StAR lysosome to other cellular compartments.
proteins are needed for efficient lipid (steroidogenic acute regulatory protein). However, future studies are needed to
exchange between these organelles is Although StAR binds cholesterol and can confirm this hypothesis and to characterize
not known (Lev, 2010). In yeast, studies transfer it between membranes in vitro exactly how NPC1 facilitates cholesterol
recently found that lipid transfer between (Kallen et al., 1998), its role in cholesterol transfer to other cellular membranes.
the ER and mitochondria slows down in transport in cells remains controversial. It
mutants missing a complex of four is not clear whether StAR moves choles- Future
proteins called the ERMES complex, terol from the outer to the inner mitochon- Many details of nonvesicular lipid traf-
which bridges the ER and mitochondria drial membrane, moves cholesterol from ficking remain open questions and are
(Kornmann et al., 2009). Thus, maintaining another organelle to the outer mitochon- currently the focus of intense research.
close contacts between the ER and mito- drial membrane, or regulates the proteins However, a few concepts are clear. For
chondria is required for efficient lipid that are actually responsible for choles- one, most nonvesicular lipid transfer prob-
exchange between these organelles. terol transport to the inner mitochondrial ably occurs at membrane contact sites,
There are a number of ways in which membrane. Such fundamental questions and undoubtedly, new techniques are
lipid transport exchange between the ER need to be resolved before we can under- needed to study these junctions and

Cell 143, December 10, 2010 ª2010 Elsevier Inc. 873


identify proteins that function at these key Funato, K., and Riezman, H. (2001). J. Cell Biol. Loewen, C.J., Roy, A., and Levine, T.P. (2003).
locations in the cell. In addition, a significant 155, 949–959. EMBO J. 22, 2025–2035.
portion of lipid trafficking at membrane Halter, D., Neumann, S., van Dijk, S.M., Wolthoorn, Maxfield, F.R., and Mondal, M. (2006). Biochem.
contact sites probably does not require J., de Mazière, A.M., Vieira, O.V., Mattjus, P., Soc. Trans. 34, 335–339.
Klumperman, J., van Meer, G., and Sprong, H.
soluble LTPs, but the mechanistic details Mesmin, B., and Maxfield, F.R. (2009). Biochim. Bi-
(2007). J. Cell Biol. 179, 101–115.
for how transfer occurs remain an impor- ophys. Acta 1791, 636–645.
Hanada, K., Kumagai, K., Yasuda, S., Miura, Y.,
tant question. Other fundamental issues Prinz, W.A. (2007). Prog. Lipid Res. 46, 297–314.
Kawano, M., Fukasawa, M., and Nishijima, M.
in this field include the energetics of nonve- (2003). Nature 426, 803–809. Rao, R.P., Yuan, C., Allegood, J.C., Rawat, S.S.,
sicular lipid trafficking and its regulatory Hanada, K., Kumagai, K., Tomishige, N., and Ya- Edwards, M.B., Wang, X., Merrill, A.H.J., Jr.,
mechanisms, including if and how its direc- maji, T. (2009). Biochim. Biophys. Acta 1791, Acharya, U., and Acharya, J.K. (2007). Proc. Natl.
tionality is determined. Answers to these 684–691. Acad. Sci. USA 104, 11364–11369.
questions are imperative for understanding Hechtberger, P., and Daum, G. (1995). FEBS Lett. Raychaudhuri, S., and Prinz, W.A. (2008). Proc.
how defects in nonvesicular lipid trafficking 367, 201–204. Natl. Acad. Sci. USA 105, 15785–15790.
cause disease, but they are also critical for Infante, R.E., Wang, M.L., Radhakrishnan, A., Rocha, N., Kuijl, C., van der Kant, R., Janssen, L.,
deciphering fundamental processes in eu- Kwon, H.J., Brown, M.S., and Goldstein, J.L. Houben, D., Janssen, H., Zwart, W., and Neefjes,
karyotic cells, including lipid metabolism, (2008). Proc. Natl. Acad. Sci. USA 105, 15287– J. (2009). J. Cell Biol. 185, 1209–1225.
signaling, and intracellular distribution. 15292. Schulz, T.A., Choi, M.-G., Raychaudhuri, S.,
Kallen, C.B., Billheimer, J.T., Summers, S.A., Mears, J.A., Ghirlando, R., Hinshaw, J.E., and
Stayrook, S.E., Lewis, M., and Strauss, J.F., III. Prinz, W.A. (2009). J. Cell Biol. 187, 889–903.
ACKNOWLEDGMENTS
(1998). J. Biol. Chem. 273, 26285–26288.
Sleight, R.G., and Pagano, R.E. (1983). J. Biol.
I thank Ted Steck, Jim Hurley, and Tim Schulz for Kaplan, M.R., and Simoni, R.D. (1985a). J. Cell Chem. 258, 9050–9058.
reading the manuscript. This work was supported Biol. 101, 446–453.
Steck, T.L., Ye, J., and Lange, Y. (2002). Biophys.
by the Intramural Research Program of the Kaplan, M.R., and Simoni, R.D. (1985b). J. Cell J. 83, 2118–2125.
National Institute of Diabetes and Digestive and Biol. 101, 441–445.
Kidney Diseases. Trotter, P.J., Pedretti, J., Yates, R., and Voelker,
Kok, J.W., Babia, T., Klappe, K., Egea, G., and D.R. (1995). J. Biol. Chem. 270, 6071–6080.
Hoekstra, D. (1998). Biochem. J. 333, 779–786.
Voelker, D.R. (2009). Annu. Rev. Biochem. 78,
REFERENCES Kornmann, B., Currie, E., Collins, S.R., Schuldiner,
827–856.
M., Nunnari, J., Weissman, J.S., and Walter, P.
Bankaitis, V.A., Mousley, C.J., and Schaaf, G. (2009). Science 325, 477–481. Wang, X., Rao, R.P., Kosakowska-Cholody, T.,
(2010). Trends Biochem. Sci. 35, 150–160. Masood, M.A., Southon, E., Zhang, H., Berthet,
Kudo, N., Kumagai, K., Tomishige, N., Yamaji, T.,
C., Nagashim, K., Veenstra, T.K., Tessarollo, L.,
Benning, C. (2009). Annu. Rev. Cell Dev. Biol. 25, Wakatsuki, S., Nishijima, M., Hanada, K., and
et al. (2009). J. Cell Biol. 184, 143–158.
71–91. Kato, R. (2008). Proc. Natl. Acad. Sci. USA 105,
Cheruku, S.R., Xu, Z., Dutia, R., Lobel, P., and 488–493. Wang, M.L., Motamed, M., Infante, R.E., Abi-Mo-
Storch, J. (2006). J. Biol. Chem. 281, 31594–31604. Kwon, H.J., Abi-Mosleh, L., Wang, M.L., Dei- sleh, L., Kwon, H.J., Brown, M.S., and Goldstein,
senhofer, J., Goldstein, J.L., Brown, M.S., and J.L. (2010). Cell Metab. 12, 166–173.
Choi, J.Y., Riekhof, W.R., Wu, W.I., and Voelker,
D.R. (2006). Biochem. Soc. Trans. 34, 404–408. Infante, R.E. (2009). Cell 137, 1213–1224. Warnock, D.E., Lutz, M.S., Blackburn, W.A.,
Lev, S. (2010). Nat. Rev. Mol. Cell Biol. 11, Young, W.W.J., Jr., and Baenziger, J.U. (1994).
D’Angelo, G., Polishchuk, E., Di Tullio, G., Santoro,
739–750. Proc. Natl. Acad. Sci. USA 91, 2708–2712.
M., Di Campli, A., Godi, A., West, G., Bielawski, J.,
Chuang, C.C., van der Spoel, A.C., et al. (2007). Levine, T.P., and Munro, S. (2001). Mol. Biol. Cell Wattenberg, B.W. (1990). J. Cell Biol. 111,
Nature 449, 62–67. 12, 1633–1644. 421–428.
D’Angelo, G., Vicinanza, M., and De Matteis, M.A. Levine, T., and Loewen, C. (2006). Curr. Opin. Cell Yamaji, T., Kumagai, K., Tomishige, N., and
(2008). Curr. Opin. Cell Biol. 20, 360–370. Biol. 18, 371–378. Hanada, K. (2008). IUBMB Life 60, 511–518.

874 Cell 143, December 10, 2010 ª2010 Elsevier Inc.


Leading Edge
Review

Membrane Budding
_
James H. Hurley,1,* Evzen Boura,1 Lars-Anders Carlson,1 and Bartosz Rózycki 2
1Laboratory of Molecular Biology
2Laboratory of Chemical Physics
National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892-0580, USA
*Correspondence: hurley@helix.nih.gov
DOI 10.1016/j.cell.2010.11.030

Membrane budding is a key step in vesicular transport, multivesicular body biogenesis, and envel-
oped virus release. These events range from those that are primarily protein driven, such as the
formation of coated vesicles, to those that are primarily lipid driven, such as microdomain-
dependent biogenesis of multivesicular bodies. Other types of budding reside in the middle of
this spectrum, including caveolae biogenesis, HIV-1 budding, and ESCRT-catalyzed multivesicular
body formation. Some of these latter events involve budding away from cytosol, and this unusual
topology involves unique mechanisms. This Review discusses progress toward understanding
the structural and energetic bases of these different membrane-budding paradigms.

Eukaryotic cells are defined by their compartmentalization into thermal energy (Bloom et al., 1991).This is important for biology
membrane-delimited structures. The protein and lipid content because events that require thermal energy of this magnitude
of these membranes is maintained and regulated by a constant (that is, of 100 kBT or greater) do not occur spontaneously.
flux of vesicular trafficking. Each vesicular trafficking event Biophysical studies of membrane budding, which offer the
involves the budding of a membrane vesicle from a donor promise of accounting for energetics, are typically carried out
membrane, typically followed by its regulated transport to, dock- in vesicles that are much larger than their counterparts in biolog-
ing to, and fusion with an acceptor membrane. Many viruses also ical systems. Fortunately, the energetic cost of bud formation is
have membrane envelopes and escape from host cells by to a first approximation independent of the size of the bud.
membrane-budding events. In pure lipid mixtures used in biophysical studies, vesicles are
Our laboratory has been characterizing the unusual microns in size, spreading the energetic cost over 106 or
membrane-budding reaction promoted by the ESCRTs, which more lipid molecules. In cells, however, membrane buds have
has led us to take a fresh look at how membrane lipid properties a diameter of 20–100 nm, thus involving as few as 103–104 lipid
might make protein-dependent, energetically expensive reac- molecules. This poses the question, how do a modest number of
tions easier. Several excellent reviews have covered the way protein-lipid interactions create the free energy that is needed for
proteins induce curvature in biological membranes (Farsad and budding, or alternatively, how do lipids themselves contribute to
De Camilli, 2003; McMahon and Gallop, 2005; Voeltz and Prinz, lowering the energy barrier?
2007) and the physical principles of membrane curvature
(Zimmerberg and Kozlov, 2006). This Review will take a different Coated Vesicle Budding
viewpoint and consider the comparative roles of proteins and Clathrin
lipids in select examples of vesicular budding events (Figure 1) The dominant mechanism of membrane budding into the cytosol
to discuss similarities and differences in budding events in and the paradigm for protein-directed budding is the formation
synthetic versus cellular contexts, the potential roles of proteins of coated vesicles (Figures 1F and 1G and Figure 2). Clathrin-
in orchestrating lipid phase changes, and the roles of lipids in coated vesicles (CCVs) are typically 60–100 nm in diameter
recruiting and regulating proteins. We also examine the implica- (Bonifacino and Lippincott-Schwartz, 2003; Brodsky et al.,
tions of the above for cell physiology. This article is not intended 2001). Clathrin can form baskets in vitro that resemble the
as a comprehensive review of all cellular budding events. Rather, CCVs in the absence of membranes, and the basket structure
we consider emerging mechanistic thinking in multivesicular has been characterized in molecular detail (Fotin et al., 2004).
body formation and virus budding, placing these in the context Clathrin itself binds neither membranes nor cargo but relies on
of the classical mechanisms underlying budding of coated adaptors for this function. Among the most comprehensively
vesicles. studied is adaptor protein complex 2 (AP-2 complex) (Robinson
and Bonifacino, 2001), which functions in clathrin-mediated
Energetics of Vesicle Budding endocytosis at the plasma membrane. The AP-2 adaptor
The formation of spherical vesicles from a flat membrane of complex opens up in the presence of cargo and the lipid phos-
typical biological composition and no intrinsic propensity to phatidylinositol (4,5)-bisphosphate (PI(4,5)P2) to form a flat plat-
curve entails a membrane-bending free energy (Helfrich, 1973), form capable of binding multiple PI(4,5)P2 and cargo molecules
DG = 8pk 250–600 kBT, given k 10–25 kBT, where kBT is (Jackson et al., 2010). The established role for PI(4,5)P2 in this

Cell 143, December 10, 2010 ª2010 Elsevier Inc. 875


Figure 1. Proteins and Lipid Microdomains in Membrane Budding
(A) Budding of phase-separated lipid microdomains from GUVs (giant unilamellar vesicles) composed of synthetic lipids is an example of membrane budding in
the absence of any proteins. Reproduced by permission from Baumgart et al. (2003).
(B) Shiga toxin (black dots) acts from outside the plasma membrane to induce membrane buds and is an example of a protein triggering budding events that are
primarily driven by lipid microdomains. Image reproduced by permission from Macmillan Publishers Ltd: Nature, Römer et al. (2007), copyright 2007.
(C) Budding by caveolae represents a hybrid between a membrane microdomain and protein coat-driven mechanisms. Reproduced by permission from Mac-
millan Publishers Ltd: Nat. Rev. Mol. Cell. Biol., Parton and Simons (2007), copyright 2007.
(D) ESCRT-I and -II induce buds in synthetic GUVs. Reproduced by permission from Wollert and Hurley (2010). Proteins organize these structures but do not form
a coat, suggesting a possible role for microdomains.
(E) HIV-1 buds visualized by electron tomography (Carlson et al., 2008). The bud is organized by the HIV-1 capsid protein, heavily enriched in raft lipids, and
cleaved by ESCRT proteins.
(F) Deep etch visualization of clathrin-coated pits (image courtesy of J. Heuser). Clathrin assembles into baskets in the absence of membranes but is thought to be
too flexible to deform membranes on its own. For this, clathrin needs help from other membrane-deforming proteins and possibly from lipids.
(G) The COP II cage is an example of a protein structure that can form in the absence of lipids and can impose its shape on any simple bilayer-forming lipid mixture.
Reproduced by permission from Russell and Stagg (2010).

pathway is to recruit AP-2 and other proteins to the site of domain proteins, and PI(4,5)P2 constitute the minimum require-
budding. A role for PI(4,5)P2 clustering into microdomains has ments for membrane bud formation in this pathway.
been suggested on theoretical grounds (Liu et al., 2006) but The scission of the clathrin-coated bud to form a detached
has yet to be directly visualized. vesicle is a complex process in its own right, and the reader is
Clathrin is absolutely required for the budding of AP-2- and referred to recent reviews (Pucadyil and Schmid, 2009). Finally,
cargo-rich plasma membrane domains, which remain flat in its following scission, the clathrin coat is removed by the ATP-
absence (Hinrichsen et al., 2006). However, clathrin monomers dependent action of the molecular chaperone Hsc70 and its
are flexible, which gives clathrin the ability to form different types cofactor auxillin (Eisenberg and Greene, 2007). It is only following
of lattices and to adapt to various cargoes (Ehrlich et al., 2004). nucleotide hydrolysis that the energetic cost of clathrin-induced
Given the flexibility of clathrin monomers, the energy of clathrin membrane deformation is finally paid, making the full reaction
polymerization has been proposed on theoretical grounds to cycle—from flat membrane to uncoated vesicle—thermody-
be insufficient on its own to bend the membrane into a bud (Nos- namically irreversible.
sal, 2001). However, this concept has yet to be confirmed exper- COP I and COP II
imentally and is not universally accepted. Vesicles carrying cargo from the endoplasmic reticulum (ER) to
Cholesterol is important for clathrin-mediated endocytosis by the Golgi are coated by the COP II complex, which, like clathrin,
many (though not all) accounts (Rodal et al., 1999; Subtil et al., can form membrane-free baskets in vitro with vesicle-like dimen-
1999), although it is less sensitive to cholesterol depletion than sions (Stagg et al., 2006). COP II vesicles have a preferred size,
most coat-independent budding pathways (Sandvig et al., but as with clathrin, the flexibility of the COP II subunits allows
2008). Clathrin, cargo adaptors, and PI(4,5)P2 are necessary formation of expanded lattices that can accommodate large
but not sufficient on their own to induce membrane curvature. cargoes such as procollagen and large lipoprotein particles
The essential early endocytic factor epsin wedges its amphi- known as chylomicrons (Stagg et al., 2008).
pathic helix a0 into the membrane upon PI(4,5)P2 binding, COP II vesicle budding has been reconstituted in vitro from
promoting positive curvature (Ford et al., 2002). The cargo- purified proteins and synthetic lipids (Lee et al., 2005; Matsuoka
binding muniscin proteins FCHo1/2 (Syp1 in yeast) contain et al., 1998). A membrane consisting only of synthetic unsatu-
BAR domains that promote positive curvature very early in endo- rated phospholipids was capable of supporting budding
cytosis (Henne et al., 2010; Reider et al., 2009; Stimpson et al., (Matsuoka et al., 1998). COP II consists of the Sec23/24 sub-
2009; Traub and Wendland, 2010). In principle, the reagents complex, which binds lipids and cargo via a gently curved face
and concepts would appear to be in place to reconstitute (Bi et al., 2002), the Sec13/31 subcomplex, which forms an outer
clathrin-dependent membrane budding. Reconstitution of cage around the vesicle, and the membrane-bending GTPase
clathrin-mediated endocytosis using synthetic lipids and purified Sar1. The Sec23/24 and Sec13/31 subcomplexes in combina-
proteins would be an important step in determining whether tion are sufficient to form buds, with Sar1 strictly required only
clathrin, AP-2, one or more amphipathic helix and/or BAR for the scission of the buds. GTP hydrolysis by Sar1 provides

876 Cell 143, December 10, 2010 ª2010 Elsevier Inc.


Figure 2. Coated Vesicle Budding
(A) Structure of a clathrin basket from cytoelectron microscopy; reproduced by
permission from Macmillan Publishers Ltd: Nature, Fotin et al. (2004), copy-
right 2004.
(B) COP II vesicles produced from purified components; reproduced by
permission from Lee et al. (2005).
(C) Structural parallels between clathrin, COP I, and COP II. Adapted from Lee
and Goldberg (2010).

energy input into the system, making the overall process (which
Figure 3. Membrane Microdomains and Budding
culminates in the uncoating of cargo-loaded vesicles) thermody-
(A) Coexistence of phases in model membranes visualized by atomic force
namically irreversible. microscopy in a supported bilayer (a membrane bilayer adsorbed onto a solid
COP I-coated vesicles are responsible for retrograde traffic support, usually glass). Reproduced with permission from Chiantia et al.
from the Golgi to the ER, and this reaction has also been recon- (2006).
(B) Phase transitions in a single-lipid membrane analyzed by molecular
stituted from purified proteins and synthetic lipids. The budding dynamics simulations. Reproduced with permission from Heller et al. (1993).
reaction requires the coatomer complex, GTP-bound Arf1, and Copyright 1993 American Chemical Society.
protein cargo tails tethered to the membrane but has no special (C) Schematic model of a raft-type membrane microdomain, including a model
of a myristoylated ESCRT-III subunit Vps20 as an example of protein that
lipid requirements (Bremser et al., 1999). Budding occurs even might anchor to rafts.
from vesicles composed of the pure synthetic phospholipid
DOPC doped with small amounts of a lipopeptide cargo.
Recently, a composite crystallographic structure of cage-form- phase, with the translational and conformational order of the lipid
ing components of coatomer consisting of the a, b0 , and 3 chains depending on their composition and the temperature. The
subunits has been determined and shown to resemble the cla- liquid phase is the more relevant to biology and can be subdi-
thrin triskelion (Lee and Goldberg, 2010). In sum, COP I and vided into liquid disordered (Ld) and liquid ordered (Lo) phases.
COP II provide some of the purest examples of protein-directed Lipids in the Ld phase have higher conformational freedom and
membrane budding, in which the protein coat imposes its shape diffusion coefficients than in the Lo phase. At biological temper-
upon the membrane with minimal dependence on its lipid atures, the Ld and Lo phases can coexist in membranes of mixed
composition. composition (Elson et al., 2010; Garcı́a-Sáez and Schwille,
2010).
Membrane Microdomains and Budding In general, phospholipids with unsaturated chains prefer the
Lipid Phase Separation as a Budding Mechanism Ld phase, whereas cholesterol, sphingolipids, and phospholipids
In contrast to the protein-dominated paradigm of coated vesicle with saturated chains prefer the Lo phase (Lingwood and
budding, phase separation in simple lipid mixtures can drive Simons, 2010). Typically, the energetic cost for contact between
budding on a micron scale in synthetic model membranes, in dissimilar lipids is small, 0.5 kBT (Garcı́a-Sáez and Schwille,
the absence of proteins (Baumgart et al., 2003) (Figure 1A and 2010), but becomes significant when summed over many lipids.
Figure 3). Membrane bilayers can adopt either a solid or a liquid The higher acyl chain order in the Lo phase results in their

Cell 143, December 10, 2010 ª2010 Elsevier Inc. 877


Figure 4. Protein Structures that Cluster Raft Lipids
(A) Simian virus 40 VP1 pentamer bound to the membrane via the headgroup of the ganglioside GM1 (Neu et al., 2008).
(B) Cholera toxin B subunit pentamer bound to GM1 (Merritt et al., 1994).
(C) Composite model of the myristoylated HIV-1 matrix domain trimer bound to PI(4,5)P2 (Hill et al., 1996; Saad et al., 2006, 2008). In each case, lipid tails are
modeled. Images were generated with VMD 1.8.6.

elongation to their maximum extent, hence Lo membrane domains known as ‘‘rafts.’’ Why don’t rafts and other microdo-
domains are thicker than Ld domains. The height mismatch at mains coalesce on the micron scale in living cells, as they do
the phase boundary is energetically unfavorable because it in model membranes? The answer is not known, but the action
forces the polar headgroup region of the Ld domain into contact of the cytoskeleton and membrane traffic, and the large fraction
with the hydrophobic portion of the Lo domain. The free energy of protein in cellular membranes, are usually invoked. Indeed, it
cost per unit length is known as the line tension and has units is to be expected that cells would have mechanisms to
of force. In order to minimize the free energy associated with block the unchecked growth of microdomains, as the ensuing
line tension, membrane domains will coalesce with one another spontaneous vesiculation of cell membranes would be disas-
into circular zones. When circular domains reach a critical size at trous.
which the line tension energy term exceeds the Helfrich (curva- Soluble and lumenally anchored cargoes, viruses, and toxins
ture-dependent) energy of membrane deformation, the are selectively transported in vesicular carriers even though
membrane will deform out of plane in order to minimize the they have no direct communication with the cytosol to signal
zone of contact (Lipowsky, 1992). If the line tension is high their packaging and sorting. In some cases, transmembrane-
enough, the neck connecting the membrane bud can be sorting receptors serve as adaptors to link cargo to conventional
severed, leading to the formation of detached vesicles. In addi- cytosolic coat complexes. In other cases, membrane rafts make
tion to line tension effects, membrane microdomain formation the link. Simian virus 40 (SV40) and cholera toxin enter cells by
can bend membranes by concentrating lipids with distinct binding to multiple molecules of the ganglioside GM1 (Damm
intrinsic curvatures, and the contents of such microdomains et al., 2005; Kirkham et al., 2005), a raft-favoring lipid. The
can not only drive budding but dictate its direction (Bacia cholera toxin B subunit (Merritt et al., 1994) and the SV40 VP1
et al., 2005). protein (Neu et al., 2008) both bind to GM1 as pentamers.
The complex lipid mixture of the plasma membrane supports Cholera toxin pentamer binds GM1 (Figure 4) and thus induces
phase separation in micron-sized domains when reconstituted formation of an Lo microdomain in model membranes
in giant unilamellar vesicles (Baumgart et al., 2007). However, (Hammond et al., 2005) and in turn leads to budding (Bacia
in living cells, membrane microdomains are heterogeneous, et al., 2005; Ewers et al., 2010). Shiga toxin B subunit binds
highly dynamic nanoscale structures (Hancock, 2006; Lingwood the glycolipid Gb3 and appears to operate by a similar paradigm.
and Simons, 2010; Pike, 2006). In the most up-to-date biophys- In this case tubular vesicles are formed, and lipid compression
ical view, these nanoscale structures likely correspond to critical favoring negative curvature is thought to be the driving force
fluctuations (Veatch et al., 2007). Although the concepts of the (Römer et al., 2007). In each of these examples, it is clear that
Lo and Ld phases are oversimplifications of the variety of clustering of lipids leads to important changes in membrane
dynamic membrane substructures that exist in cells (Lingwood structure that contribute to budding. The proposed physical
and Simons, 2010), they will be used in this Review because mechanisms remain speculative, however. Revealing these
they are useful intuitive handles, deeply ingrained in the mechanisms remains a profound challenge to experimen-
literature, and helpful in relating model membrane studies to talists and thus is an area that will benefit from increasing
biology. Most, but not all, of the membrane microdomains impli- sophisticated computer simulations of membrane dynamics on
cated in cellular budding are the sterol- and sphingolipid-rich realistic timescales.

878 Cell 143, December 10, 2010 ª2010 Elsevier Inc.


Caveolae brane-spanning a helices (Hemler, 2005). Tetraspanins have two
Caveolae (‘‘little caves’’) are flask-shaped 60–80 nm invagina- extracellular domains; the second, EC2, is the larger of the two.
tions of the plasma membrane that consist of raft lipids, caveo- The structure of the EC2 region of CD81 has been determined,
lins 1–3, and the caveolin-associated cavins 1–4 (Hansen and revealing an extensive dimerization interface (Kitadokoro et al.,
Nichols, 2010). Caveolins are structurally analogous to the retic- 2001). The minimal functional tetraspanin oligomer is probably
ulons and DP1/Yop1 proteins that maintain the curvature of ER a homodimer. These proteins are multiply palmitoylated on their
membrane tubules (Hu et al., 2008; Shibata et al., 2009) and to short intracellular loop and N- and C-terminal extensions, and
another plasma membrane raft protein, flotillin (Bauer and Pelk- these palmitoylations are central to their ability to form TEMs.
mans, 2006). Caveolins are pentahelical proteins, with two of the Tetraspanins bind to a wide range of potential cargo proteins
helices inserting deeply into the membrane, almost but not (Hemler, 2005), potentially coupling them to TEMs and thereby
completely spanning the bilayer. The other three helices are to microdomain-mediated budding. More extensive mechanistic
amphipathic and are thought to wedge themselves into the inter- analysis of the budding mechanism responsible for TEM traffic
facial region of the membrane (Parton et al., 2006). Caveolae will be eagerly awaited.
contain a consistent number of caveolin molecules, 144, which
suggests the formation of a highly organized coat (Pelkmans and Multivesicular Bodies
Zerial, 2005). The sorting of unneeded, damaged, or dangerous plasma
Posttranslational modification of caveolins is important to their membrane proteins to the lysosome for degradation is carried
function. Palmitoylation at multiple residues promotes their out by endosomes (Sorkin and von Zastrow, 2009). This pathway
constitutive association with cholesterol and other raft lipids. also is central to the biogenesis of the lysosome (or yeast
Caveolins also undergo phosphoregulation by multiple protein vacuole), as it carries newly synthesized lysosomal enzymes
kinases (Pelkmans and Zerial, 2005). For instance, when caveo- from the trans-Golgi to their destination. In the metazoa, the
lin-1 is phosphorylated at serine 80, which adjoins one of the endosomal pathways have many additional roles, with the
predicted interfacial a helices, its ability to induce curvature is most pertinent to this Review being the biogenesis of lyso-
turned off. Although the energetic book-keeping of caveolin- some-related organelles (Raposo and Marks, 2007) and
induced curvature has not been worked out, it is likely to differ exosomes. Multivesicular bodies (MVBs, also known as multive-
greatly from that of conventional coated vesicles. Insertion of sicular endosomes) are key intermediates in endolysosomal
caveolin into the membrane presumably shifts the intrinsic transport (Figure 5; Gruenberg and Stenmark, 2004; Piper and
curvature of the membrane such that the positively curved bud Katzmann, 2007). MVBs are formed by the invagination and
is the low-energy state and the flat caveolin microdomain is scission of buds from the limiting membrane of the endosome
the high-energy state. Thus, once the caveolin microdomain is into the lumen. MVB biogenesis is the main physiological
formed, energy input is probably needed to flatten the example of membrane budding away from the cytosol.
membrane rather than to curve it. ATP hydrolysis by protein ESCRTs and Multivesicular Bodies
kinases that phosphorylate caveolin might provide the thermo- Yeast (Saccharomyces cerevisiae) has a single MVB pathway
dynamic driving force for membrane flattening. Dephosphoryla- that drives the internalization of ubiquitinated transmembrane
tion by protein phosphatases would, in this speculative scheme, proteins into the lumens of early endosomes (Piper and Katz-
allow the membrane to spring back to its low-energy state. mann, 2007). The pathway is initiated by the presence of the lipid
Cavins are soluble proteins rich in predicted coiled-coil struc- phosphatidylinositol 3-phosphate (PI(3)P) and membrane-teth-
ture and basic residues but otherwise structurally uncharacter- ered ubiquitin moieties on the endosome surface. PI(3)P is
ized. They seem to be important for caveolar structure, but the synthesized by the class III PI 3-kinase Vps34, an enzyme essen-
precise role of these recently discovered factors in structuring tial for the progression of the endolysosomal pathway. PI(3)P is
the caveolar coat is not clear. Given that caveolae have a consis- the defining marker of early endosomes, autophagosomes,
tent amount of caveolin protomers, they could be viewed as highly and, in mammalian cells, phagosomes. PI(3)P signals are recog-
organized assemblies whose specialized structure and distinct nized by FYVE and PX domain-containing proteins (Misra et al.,
curvature are caveolin driven but lipid stabilized. Alternatively, if 2001). In the MVB pathway, the key FYVE domain protein is
viewed from the standpoint of their lipid content, caveolae could a subunit of the ESCRT-0 complex. ESCRT-0 contains five
be viewed as specialized, morphologically distinct membrane ubiquitin-binding domains (UBDs) (Ren and Hurley, 2010) and
microdomains, whose formation is driven by lipids but stabilized clusters ubiquitinated cargo in vitro (Wollert and Hurley, 2010).
by caveolin (Parton and Simons, 2007). The hybrid nature of Recruitment of ESCRT-0 to the early endosomal membrane
caveolae, seemingly at once both coated vesicle and membrane initiates the recruitment of the ESCRT-I, -II, and –III complexes
microdomain, makes them a particularly fascinating example of (Saksena et al., 2007; Williams and Urbé, 2007). Based on
the interplay between proteins and lipids in membrane budding. in vitro reconstitution, ESCRT-I and -II drive membrane budding,
Tetraspanin-Enriched Microdomains whereas ESCRT-III cleaves the bud necks to form intralumenal
Tetraspanin-enriched microdomains (TEMs), which are abun- vesicles (Hurley and Hanson, 2010; Wollert and Hurley, 2010;
dant in exosomes and in the intralumenal vesicles of immune Wollert et al., 2009). In vitro ESCRT budding reactions have
cell multivesicular bodies, are another potential example of been carried out with a mixture of saturated and unsaturated
a membrane microdomain involved in budding (Pols and Klum- phospholipids and cholesterol (Wollert and Hurley, 2010), but
perman, 2009). Tetraspanins are a family of at least 32 proteins the precise lipid requirements for the reaction have yet to be
in mammals and are defined by the presence of four transmem- analyzed in detail.

Cell 143, December 10, 2010 ª2010 Elsevier Inc. 879


Figure 5. Multivesicular Bodies Bud via
Diverse Mechanisms
(A) Multivesicular bodies (MVBs) form from late en-
dosomes in animal cells. Their formation is depen-
dent on both ESCRT complexes and the unusual
lipid lysobisphosphatidic acid (LBPA).
(B) The conserved ESCRT-dependent MVB
biogenesis pathway from early endosomes in
yeast and animal cells. PI(3)P has been directly
visualized in these MVBs. Cholesterol has been
visualized in MVBs from animal cells, but it has
not been directly confirmed whether these are
ESCRT dependent or not.
(C) Specialized formation of MVBs containing
polymerized Pmel17.
(D) Ceramide-dependent MVBs bud from raft-like
and tetraspanin-enriched microdomains in animal
cells.

complexes in solution (Im and Hurley,


2008; Kostelansky et al., 2007). These
structures show that multiple membrane
and ESCRT-III attachment sites are sepa-
rated by rigid spacers of up to 18 nm
across, suggesting a mechanism to
Strikingly, ESCRT-I, -II, and -III all localize to the bud neck induce or at least stabilize formation of a membrane neck of
(Wollert and Hurley, 2010). ESCRT-III subunits assemble into roughly those dimensions. Subsequent recruitment and poly-
tubular structures in vitro and when overexpressed (Bajorek merization of ESCRT-III into spiral domes (Fabrikant et al.,
et al., 2009; Hanson et al., 2008; Lata et al., 2008). The 2009) would then narrow and sever the neck in the current model
ESCRT-III proteins coat the interior of lipid tubes created (Hurley and Hanson, 2010).
in vitro (Lata et al., 2008) and have diameters of 40–50 nm for The observation that the ESCRT complexes localize to the bud
lipid-free tubes, and 100 nm for lipid-coated tubes. These neck explains how they bud membranes away from the cytosol
tubes exceed the narrowest dimensions of bud necks in cells, without themselves being consumed in the bud. This mechanism
based on just a few observations that suggest a size closer to stands in sharp contrast to the familiar budding of coated vesi-
20 nm (Murk et al., 2003). However, the tubes taper to cles toward cytosol, described above. The thermodynamic
a dome at their ends (Fabrikant et al., 2009), which may repre- driving force for the pathway is the coupling of ESCRT-III solubi-
sent the tubes’ most important functional feature. Lipid tube lization and recycling to ATP hydrolysis by the dodecameric AAA
extrusion by ESCRT-III seems to have no special lipid require- ATPase Vps4 (Babst et al., 1998; Wollert et al., 2009). Although
ments, as it can be supported in vitro by a simple mixture of the overall thermodynamic driving force is clear, the energetic
the unsaturated phospholipids SOPC and DOPS (Lata et al., trajectory of neck-directed bud formation is currently unknown.
2008). Indeed, whereas most ESCRTs are unique to the eukarya, Theoretical analysis of the membrane mechanics of this process
ESCRT-III is conserved in a subset of Archaea, where it functions is urgently needed, as is a better understanding of the roles of
in the membrane abscission step of cell division (Lindås et al., lipids.
2008; Samson et al., 2008). Thus the Archaeal ESCRT-III ortho- All four ESCRT complexes are conserved between yeast and
logs can presumably function in membrane scission with metazoa. In its broad outlines, the ESCRT-dependent conver-
Archaeal lipids, which are radically different from eukaryotic sion of early endosomes into MVBs is the same in yeast and
lipids and rich in rigid, bilayer-spanning tetraether linkages metazoa (Raiborg and Stenmark, 2009). Intralumenal vesicles
(Koga and Morii, 2005). It is thought on theoretical grounds in mammalian cells are highly enriched in cholesterol and tetra-
that membrane tubes are induced by the binding of the curved spanins (Möbius et al., 2003; van der Goot and Gruenberg,
ESCRT-III polymer to the membrane (Lenz et al., 2009). 2006). However, at least some of the cholesterol- and tetraspa-
ESCRT-III polymerization governs the late stage of neck devel- nin-rich intralumenal vesicles in mammalian cells are part of
opment leading to scission, but it is not likely to be the main process that is distinct from the ESCRT pathway (Simons and
factor in the initial budding event. The initial formation of the Raposo, 2009). Raft markers such as long-chain sphingomyelins
bud is driven by the assembly of ESCRT-I and -II with one transit through MVBs (Koivusalo et al., 2007). Consistent with
another and with the endosome membrane (Wollert and Hurley, a possible ESCRT-sterol connection, defects in ESCRT function
2010). The structure of this assembly is unknown, and the nature block endosomal cholesterol transport in mammalian cells
of the assembly is a pressing question in the field. Composite (Bishop and Woodman, 2000; Peck et al., 2004). In yeast, ergos-
structures of the ESCRT-I and -II complexes have been devel- terol and, more speculatively, Sna3 (Piper and Katzmann, 2007)
oped on the basis of crystal structures of the separate compo- might replace the roles of cholesterol and tetraspanins in micro-
nents together with hydrodynamic information of the complete domain formation. Given that ESCRTs bud membranes without

880 Cell 143, December 10, 2010 ª2010 Elsevier Inc.


a coat, and that most other coatless budding mechanisms rely
on membrane microdomains of some sort, it is tempting to
speculate that ESCRT-mediated budding could involve tetra-
spanin and cholesterol-rich domains. Very little is known about
how ESCRTs might couple to such microdomains.
Lipids modifications might also play a role. The ESCRT-III
subunit Vps20 must be myristoylated for full function (Babst
et al., 2002; Yorikawa et al., 2005). Yet even unmyristoylated
Vps20 has an affinity for membranes in the tens of nM, dropping
to low single digit nM range when bound to ESCRT-II (Im et al.,
2009), suggesting that myristoylation is required for another
reason than membrane targeting alone. The myristoyl moiety is
saturated and favors association with Lo phase microdomains
(Resh, 2006). Ubiquitination of tetraspanins (Lineberry et al.,
2008) and, in yeast, Sna3 (Stawiecka-Mirota et al., 2007) has
also been reported.
Another important question surrounds the nature of the PI(3)P
lipid that binds to ESCRT complexes through its headgroup. Figure 6. Lipids and ESCRTs in HIV-1 Assembly
Substantial levels of PI(3)P are found in the MVB lumen (Gillooly Apart from viral proteins, the release of HIV-1 requires both specific cellular
et al., 2000). A critical gap in understanding the formation of in- lipids and proteins, which are recruited to the budding site by the viral Gag
protein. Gag assembles into an imperfect hexagonal lattice on the plasma
tralumenal vesicles is the lack of data on the tail compositions
membrane (Briggs et al., 2009). It binds the plasma membrane marker PI
of the total endosomal and intralumenal vesicle pools of PI(3)P. (4,5)P2 through a specific binding site in its N terminus. PI(4,5)P2, cholesterol,
The concept of an ESCRT-microdomain link is speculative. In and certain other raft lipids are enriched in the viral membrane compared to the
the absence of other explanations for the unusual coatless plasma membrane. Through its C terminus, Gag recruits the ESCRT proteins
to the budding site. Gag can bind both ESCRT-I and ALIX, which both recruit
budding by the ESCRTs, these issues call for further investiga- ESCRT-III to the budding site.
tion.
Animal Cells Have More than One Kind of MVB
Animal cells have additional pathways of MVB formation not cles) with pre-existing phase separation (Trajkovic et al., 2008).
found in yeast. The mammalian late endosomal and lysosomal Sphingomyelinase cleavage of the phosphodiester bond
lipidome contains up to 20% of the unusual lipid lysobisphos- between ceramide and the SM headgroup provides a potential
phatidic acid (LBPA), which is not found in other organelles or mechanism to put energy into this budding pathway and make
in yeast. Mammalian cells have a late endosomal MVB pathway it thermodynamically irreversible. Ceramide-induced intralume-
that seems to depend on LBPA microdomains that are probably nal vesicles bud exclusively from the Lo phase (Trajkovic et al.,
induced on the lumenal leaflet by acidic pH (Matsuo et al., 2004). 2008). Ceramide has several special properties, including a small
The ultimate thermodynamic basis for membrane curvature in headgroup that would favor its presence in the inner leaflet of the
the LBPA pathway would presumably come from the energy intralumenal vesicle and an ability to self-associate through
expended in the pumping of protons into the lumen of the endo- headgroup hydrogen bonding. It is not clear which properties
some. This late endosomal pathway also involves ESCRT of ceramide are most important for the formation of intralumenal
proteins (Falguières et al., 2008). The late endosomal MVB vesicles. Exosomes produced by the sphingomyelinase
pathway should not, however, be confused with the canonical pathway are highly enriched in the tetraspanin CD63, suggestive
early endosomal ESCRT pathway described above, which of a coupling between TEMs and ceramide domains. Of the three
does not involve LBPA. MVB formation is involved in the biogen- pathways described above, the latter two are, based on current
esis of lysosome-related organelles, of which melanosomes are knowledge, ESCRT independent. It will be interesting to see if
the most intensively studied (Raposo and Marks, 2007). In there are ever circumstances under which the ESCRTs coop-
melanosome biogenesis, the glycoprotein Pmel17 is sorted erate with the melanosome or ceramide pathways.
into intralumenal vesicles in an ESCRT-independent reaction
(Theos et al., 2006). Pmel17 is a special cargo in that its lumenal Viral Budding
domain forms fibers and may be an example of the lumenal Enveloped Virus Budding: With ESCRTs and without
assembly of a cargo helping to drive its own inward budding Membrane budding is an essential part of the life cycle of envel-
into the endosome. oped viruses. Most, but not all, enveloped viruses bud from cells
Exosomes are 50–100 nm vesicles released from cells by the by co-opting the host ESCRT machinery (Bieniasz, 2006; Morita
fusion of MVBs with the plasma membrane (Simons and Raposo, and Sundquist, 2004; Welsch et al., 2007), whose role in budding
2009). At least one population of exosomes is produced by an of vesicles in MVBs is described above (Figure 6). Virus budding,
ESCRT-independent pathway in which neutral sphingomyeli- like MVB formation, involves budding away from cytosol. In the
nase, acting from the cytosolic face of the membrane, hydro- well-studied example of HIV-1, formation of the initial plasma-
lyzes sphingomyelin to ceramide (Trajkovic et al., 2008). The membrane attached bud is driven by the energetically favorable
formation of intralumenal vesicles by sphingomyelinase has self-assembly of the capsid (CA) domain of Gag into hexamers
been reconstituted in vitro using GUVs (giant unilamellar vesi- (Briggs et al., 2009; Wright et al., 2007). CA does not bind directly

Cell 143, December 10, 2010 ª2010 Elsevier Inc. 881


to membranes, so the energy of CA self-assembly is transduced The in vitro buds are not cleaved by the matrix protein, indicating
to the membrane through the membrane-binding matrix (MA) the requirement for additional scission factors. Indeed, VSV
domain, part of the same polypeptide chain at this stage in budding from cells requires an ESCRT-I-binding late domain
HIV-1 assembly (Hill et al., 1996). Recombinant HIV-1 Gag (Irie et al., 2004). Why does the matrix protein of one putatively
constructs lacking a part of the membrane-binding MA domain ESCRT-dependent virus, NDV, support both budding and
and all of the ESCRT-binding p6 domain are able to assemble scission on its own, whereas that of another, VSV, supports
with RNA to form spherical shells in vitro, in the absence of only formation of attached buds? It is too soon to say whether
membranes (Campbell et al., 2001). These lipid-free shells are these are intrinsic differences between these viruses or relate
slightly smaller than authentic immature HIV-1 virions, with the merely to experimental differences.
differences accounted for by the absence of membrane and Even for HIV-1, the archetypal ESCRT-dependent virus, there
the MA domain (Briggs et al., 2009). The shells assemble via seem to be circumstances in which ESCRT dependence can be
CA domain hexamers, which cannot pack into a sphere, and circumvented. The effect of mutating its two ESCRT-interacting
therefore a few gaps remain in an otherwise almost complete late domains depends on the cell type, with primary monocyte-
lattice (Briggs et al., 2009). However, in authentic released HIV-1 derived macrophages and the Jurkat T cell line retaining >20%
particles, the Gag shells are only 60% complete on average particle release even when both domains were inactivated (Fujii
(Carlson et al., 2008). Could such an incomplete shell scaffold et al., 2009). Further, replacing the C-terminal part of Gag,
bud formation? Below, we describe the role of membrane micro- including the RNA-binding nucleocapsid domain and the late
domains as another key contributor to HIV-1 bud formation. domain-containing p6 domain, with a leucine zipper motif
In contrast to the self-encoded ability of HIV-1 to form preserves efficient particle release despite absence of ESCRT-
attached buds, the release of these buds from the host cell interacting motifs (Zhang et al., 1998). Deleting part of the
requires the co-option of the host cell ESCRT machinery. nucleocapsid domain and the flanking p1 sequence has the
ESCRT-recruiting motifs known as ‘‘late domains’’ for their func- same effect of making HIV-1 release independent of a functional
tion in late stages of virus assembly and release have been ESCRT machinery (Popova et al., 2010). All in all, these findings
identified in most genera of enveloped viruses (Bieniasz, 2006; show that there is a baseline level of ESCRT-independent HIV-1
Chen and Lamb, 2008; Freed, 2002; Morita and Sundquist, release, which can be elevated by subtle alterations in the Gag
2004). The prototypical example of an ESCRT-dependent virus protein.
is HIV-1, which engages the ESCRT-I complex though a PTAP The studies mentioned above quantified the amount of virus
motif in the p6 region of its Gag protein (Huang et al., 1995), released on a timescale of 16–72 hr, and it is still possible that
and interference with this interaction dramatically reduces the microscopic kinetics of the budding process, which takes
HIV-1 release (Demirov et al., 2002a, 2002b; Garrus et al., place on the timescale of 5–25 min (Ivanchenko et al., 2009;
2001; Martin-Serrano et al., 2001; VerPlank et al., 2001). To Jouvenet et al., 2008), may have been more severely compro-
make matters more complicated, efficient release can be mised. The ESCRT-independent scission observed for NDV
rescued by overexpressing the ESCRT-associated protein in vitro and for certain HIV-1 variants in vivo suggests that in
ALIX, which binds to another motif in Gag p6, YPXnL (Fisher some cases the role of ESCRTs is merely to speed up the final
et al., 2007; Usami et al., 2007). Defects in both of these interac- stage of release. In other cases, such as wild-type HIV-1, the
tions can be rescued by overexpression of HECT domain ubiqui- ESCRTs appear to have a deeper role in viral morphogenesis
tin ligases (Chung et al., 2008; Jadwin et al., 2010; Usami et al., (Carlson et al., 2008).
2008). All of these interactions serve the same ultimate purpose Membrane Microdomains and Influenza Budding
of recruiting ESCRT-III to the nascent viral bud for scission, The influenza virus is the best characterized example of an envel-
which is thought to be carried out by the same process as for oped virus that buds without an ESCRT. Influenza does not have
cleavage of intralumenal vesicles in MVBs (Hurley and Hanson, a typical late domain sequence, nor is its budding inhibited by
2010). overexpressing a dominant-negative Vps4 (Bruce et al., 2009;
If HIV-1 is the archetype of a virus dependent on the host cell Chen et al., 2007).
machinery for membrane scission, other viruses appear to carry Influenza virus associates with lipid rafts via the transmem-
out both budding and scission entirely with virally encoded brane domains of hemagglutinin and neuraminidase (Barman
proteins. The membrane-associated matrix protein of Newcastle et al., 2004), and the membrane of released influenza virions
disease virus (NDV, a paramyxovirus) induces both bud forma- has a pronounced raft character with higher order than the
tion and scission when assembled on model membranes (Shnyr- membranes of non-raft-associated enveloped virus (Polozov
ova et al., 2007). The release of virus-like particles is stimulated et al., 2008; Scheiffele et al., 1999). This raft association serves
by negatively charged lipids and cholesterol. NDV contains a late to cluster hemagglutinin on the plasma membrane, thus
domain motif identical to that of the closely related ESCRT- increasing its concentration on the released particles (Barman
dependent paramyxovirus SV5 (Schmitt et al., 2005). The func- et al., 2004; Takeda et al., 2003), and it is further involved in
tion, if any, of ESCRTs in NDV release might be to accelerate the sorting of hemagglutinin and neuraminidase to the apical
vesiculation, which the virus already is capable of performing. face of polarized cells (Barman et al., 2004).
The matrix protein of vesicular stomatitis virus (VSV) is capable Influenza’s M2 ion channel has recently been implicated in its
of inducing membrane buds in vitro (Solon et al., 2005). In vitro budding mechanism, reconciling its ESCRT independence and
VSV budding occurs in a simple mixture of acidic phospholipids raft association (Rossman et al., 2010). M2 has a conserved
and appears to be driven by self-assembly of the matrix protein. amphipathic helix that is sufficient for vesicle scission in

882 Cell 143, December 10, 2010 ª2010 Elsevier Inc.


a minimal in vitro system, where it predominantly acts at the The binding of PI(4,5)P2 to Gag triggers the myristoyl switch,
border between Ld and cholesterol-enriched Lo domains. M2 leading to exposure of the buried myristoyl group (Saad et al.,
was further localized to the necks of budding influenza particles 2006). In the solution structure of the myristoylated matrix
by immunoelectron microscopy, and mutations disrupting its domain complex bound to a short-chain PI(4,5)P2, the myristoyl
amphipathic helix appear to increase the number of virus buds and the 10 fatty acid tail of PI(4,5)P2 extend into the lipid bilayer,
that remain associated with the cell. This is the first detailed whereas the 20 fatty acid tail of PI(4,5)P2 becomes buried in
description of an ESCRT-independent viral budding mechanism, a pocket in the matrix domain vacated by ejection of the myris-
and it will be interesting to see if it is paralleled in other systems. tate (Saad et al., 2006). In the current view of this mechanism,
How HIV-1 Uses Raft and Non-Raft Lipids to Bud the 10 tail is preferentially saturated and the 20 preferentially
from Cells unsaturated. Thus the matrix domain-PI(4,5)P2 complex would
The HIV-1 membrane is highly ordered (Aloia et al., 1993; in this scheme expose two saturated chains, transforming it
Lorizate et al., 2009), with elevated levels of cholesterol and into a raftophile. It will be interesting to see whether any cellular
certain other raft lipids (GM3 and ceramide) compared to the budding proteins—perhaps including the myristoylated ESCRT-
plasma membrane from which they bud (Brügger et al., 2006; III protein Vps20—use similar mechanisms to bridge raft and
Chan et al., 2008). Cholesterol depletion blocks HIV-1 particle non-raft lipids. HIV-1 release, with its exploitation of so many
release by inhibiting membrane binding and multimerization of of the known physiological budding paradigms in a single event,
Gag (Ono et al., 2007). Thus, the lipid segregation at HIV-1 is one of the most remarkable illustrations of how the dance
budding sites clearly has a functional role in the formation and between proteins and lipids leads to membrane buds.
release of HIV-1 particles. What, precisely, is this role? It is
tempting to speculate that microdomain formation not only Concluding Remarks
contributes to the normal HIV-1 budding pathway but facilitates We hope to have provided a few examples of how the geometry,
the ESCRT-independent budding noted above for unusual HIV-1 topology, and energetics of some selected membrane-budding
Gag constructs (for instance lacking the PTAP motif). However, events in cells are adapted to their biological functions. Trans-
note that cholesterol depletion actually promotes HIV-1 budding port through cytosolic vesicular carriers of membrane proteins
in the case of the PTAP-defective virus that buds independent of that have cytosolic tails is carried out most often through vesicles
the ESCRTs (Ono and Freed, 2001). This suggests that as with coated by the clathrin, COP I, and COP II complexes, which we
the ESCRT-independent budding of influenza, cholesterol has now know to have structural similarities to one another (Lee and
multiple roles. Goldberg, 2010). The cytosolic tails provide the signal for
HIV-1 and other retroviruses use protein-lipid interactions to assembly, coat proteins scaffold the membrane, amphipathic
target their assembly to the plasma membrane. The N-terminal helix and BAR domain factors help bend the membrane, and
matrix domain of HIV-1 Gag has a basic surface (Hill et al., uncoating-coupled hydrolysis of ATP or GTP provides the ther-
1996) and a covalently bound myristoyl fatty acid chain that is modynamic driving force. In the evolution of coats, the tradeoff
necessary for virus release (Ono and Freed, 1999). The ‘‘myristoyl has been between the benefits of flexibility and scaffolding
switch’’ model describes how this myristoyl moiety is in a buried power, with clathrin apparently optimized for flexibility, whereas
conformation in the monomeric cytosolic protein and becomes COP II is optimized as a more potent membrane-curving
exposed upon Gag oligomerization (Saad et al., 2006, 2008; scaffold.
Tang et al., 2004). Thus, the membrane binding of the Gag protein Viruses and toxins often enter cells by engaging with host
is linked to its multimerization and assembly into a lattice. The transmembrane proteins and co-opting coat-dependent
weak membrane affinity of the MA myristate and nonspecific budding mechanisms, but the defensive evolution of host organ-
interactions between the basic face of the matrix domain and isms combats this. Lipid-based entry through the induction of
bulk acidic phospholipids are not sufficient for efficient HIV-1 membrane microdomains, as exemplified by SV40, Shiga toxin,
particle release. For release to occur, the particle assembly and cholera toxin, illustrates one way that pathogens avoid
must be targeted either to the plasma membrane or to membra- having to rely on mutable surface proteins of the host. The phys-
nous compartments that can fuse with the plasma membrane, ical basis of this entry mechanism uses completely different prin-
leading to virion release. PI(4,5)P2, described above as a key ciples to the same functional end. Caveolae present a fascinating
factor in the formation of clathrin-coated vesicles, is the defining hybrid of protein scaffolding and membrane microdomain mech-
lipid marker of the plasma membrane (McLaughlin et al., 2002). anisms. The real cellular function(s) of caveolae are enigmatic,
The matrix domain of HIV-1 Gag targets specifically to the plasma leaving us for now in the dark as to the evolutionary drive for
membrane by binding tightly to the phosphoinositide PI(4,5)P2, such unusual structures.
and this interaction is required for Gag assembly and HIV-1 The ESCRT system, the main interest of our laboratory, is
budding (Ono et al., 2004). adapted for budding away from the cytosol in the opposite
How can the raft dependence of HIV-1 Gag assembly be topology of conventional coated vesicles. The ESCRT system
reconciled with its dependence on PI(4,5)P2? PI(4,5)P2 is gener- evolved to avoid the use of a protein coat because of this unusual
ally considered a non-raft lipid, although the microscopic anal- topology. The unique mechanism by which ESCRTs stabilize
ysis of the tail composition of different pools of PI(4,5)P2 is not and sever membrane buds has become much clearer over the
elaborated to the point where this can be said with certainty past year. However, the pathway of early bud formation, before
for all PI(4,5)P2. The apparent answer to this question highlights the bud neck has contracted enough for the ESCRT proteins to
the frightening ingenuity of HIV-1 in co-opting cellular systems. bridge across it, is still obscure. This led us to ask whether

Cell 143, December 10, 2010 ª2010 Elsevier Inc. 883


membrane microdomains might have a role in ESCRT-mediated Barman, S., Adhikary, L., Chakrabarti, A.K., Bernas, C., Kawaoka, Y., and
bud formation. If this were the case, membrane microdomains Nayak, D.P. (2004). Role of transmembrane domain and cytoplasmic tail amino
acid sequences of influenza a virus neuraminidase in raft association and virus
might serve as a unifying principle connecting the diverse types
budding. J. Virol. 78, 5258–5269.
of ESCRT-dependent and microdomain-dependent MVBs in
Bassereau, P. (2010). Division of labour in ESCRT complexes. Nat. Cell Biol.
animal cells. The various ESCRT- and microdomain-dependent
12, 422–423.
flavors of enveloped virus budding mirror the distinct varieties
of animal cell MVBs. This is not surprising given that these two Bauer, M., and Pelkmans, L. (2006). A new paradigm for membrane-organizing
and -shaping scaffolds. FEBS Lett. 580, 5559–5564.
processes share the same unusual property of budding away
from cytosol. Microdomains and ESCRTs have the same advan- Baumgart, T., Hess, S.T., and Webb, W.W. (2003). Imaging coexisting fluid
domains in biomembrane models coupling curvature and line tension. Nature
tage for budding away from cytosol, in that cytosolic coat
425, 821–824.
proteins need not be irreversibly consumed in the process.
Membrane budding and the related topic of membrane tubu- Baumgart, T., Hammond, A.T., Sengupta, P., Hess, S.T., Holowka, D.A., Baird,
B.A., and Webb, W.W. (2007). Large-scale fluid/fluid phase separation of
lation have become exceptionally vibrant fields, driven by
proteins and lipids in giant plasma membrane vesicles. Proc. Natl. Acad.
advances in technology. Computational resources now allow Sci. USA 104, 3165–3170.
sophisticated simulations of budding (Reynwar et al., 2007).
Bi, X., Corpina, R.A., and Goldberg, J. (2002). Structure of the Sec23/24-Sar1
Reconstitution of budding events from completely defined pre-budding complex of the COPII vesicle coat. Nature 419, 271–277.
systems (Bremser et al., 1999; Matsuoka et al., 1998; Römer
Bieniasz, P.D. (2006). Late budding domains and host proteins in enveloped
et al., 2007; Wollert and Hurley, 2010) has established molecular
virus release. Virology 344, 55–63.
mechanisms in several cases and opened the door to more
Bishop, N., and Woodman, P. (2000). ATPase-defective mammalian VPS4
sophisticated biophysical analysis (Bassereau, 2010). Electron
localizes to aberrant endosomes and impairs cholesterol trafficking. Mol.
microscopy has been the foundation of our understanding of Biol. Cell 11, 227–239.
membrane budding in cells since the beginning. Looking
Bloom, M., Evans, E., and Mouritsen, O.G. (1991). Physical properties of the
forward, advanced electron tomography will undoubtedly shape fluid lipid-bilayer component of cell membranes: a perspective. Q. Rev. Bio-
our future views of how membranes bud, as classical electron phys. 24, 293–397.
microscopy has in the past and present. As in other areas of
Bonifacino, J.S., and Lippincott-Schwartz, J. (2003). Coat proteins: shaping
cell biology, rapid advances in live-cell imaging are making membrane transport. Nat. Rev. Mol. Cell Biol. 4, 409–414.
powerful and ever-increasing contributions. Membrane budding
Bremser, M., Nickel, W., Schweikert, M., Ravazzola, M., Amherdt, M., Hughes,
is a required part of the life cycle of two of the most dangerous C.A., Söllner, T.H., Rothman, J.E., and Wieland, F.T. (1999). Coupling of coat
human pathogens, HIV and influenza, and insight into the funda- assembly and vesicle budding to packaging of putative cargo receptors.
mental nature of these budding events is perhaps the most Cell 96, 495–506.
urgently needed of all. Briggs, J.A.G., Riches, J.D., Glass, B., Bartonova, V., Zanetti, G., and
Kräusslich, H.G. (2009). Structure and assembly of immature HIV. Proc. Natl.
ACKNOWLEDGMENTS Acad. Sci. USA 106, 11090–11095.
Brodsky, F.M., Chen, C.Y., Knuehl, C., Towler, M.C., and Wakeham, D.E.
We thank E. Freed, G. Raposo, W. Prinz, M. Marks, J. Gruenberg, and (2001). Biological basket weaving: formation and function of clathrin-coated
J. Bonifacino for comments on drafts of the manuscript, J. Heuser for providing vesicles. Annu. Rev. Cell Dev. Biol. 17, 517–568.
the image used in Figure 1F, and many colleagues for stimulating discussions.
Bruce, E.A., Medcalf, L., Crump, C.M., Noton, S.L., Stuart, A.D., Wise, H.M.,
Research in the Hurley laboratory is supported the Intramural program of the
Elton, D., Bowers, K., and Digard, P. (2009). Budding of filamentous and
National Institutes of Health, NIDDK, and IATAP. B.R. was supported by
non-filamentous influenza A virus occurs via a VPS4 and VPS28-independent
a Marie Curie International Outgoing Fellowship within the 7th European
pathway. Virology 390, 268–278.
Community Framework Programme.
Brügger, B., Glass, B., Haberkant, P., Leibrecht, I., Wieland, F.T., and Kräus-
slich, H.G. (2006). The HIV lipidome: a raft with an unusual composition.
REFERENCES Proc. Natl. Acad. Sci. USA 103, 2641–2646.

Aloia, R.C., Tian, H.R., and Jensen, F.C. (1993). Lipid composition and fluidity Campbell, S., Fisher, R.J., Towler, E.M., Fox, S., Issaq, H.J., Wolfe, T., Phillips,
of the human immunodeficiency virus envelope and host cell plasma L.R., and Rein, A. (2001). Modulation of HIV-like particle assembly in vitro by
membranes. Proc. Natl. Acad. Sci. USA 90, 5181–5185. inositol phosphates. Proc. Natl. Acad. Sci. USA 98, 10875–10879.

Babst, M., Wendland, B., Estepa, E.J., and Emr, S.D. (1998). The Vps4p AAA Carlson, L.A., Briggs, J.A., Glass, B., Riches, J.D., Simon, M.N., Johnson,
ATPase regulates membrane association of a Vps protein complex required M.C., Müller, B., Grünewald, K., and Kräusslich, H.G. (2008). Three-dimen-
for normal endosome function. EMBO J. 17, 2982–2993. sional analysis of budding sites and released virus suggests a revised model
for HIV-1 morphogenesis. Cell Host Microbe 4, 592–599.
Babst, M., Katzmann, D.J., Estepa-Sabal, E.J., Meerloo, T., and Emr, S.D.
(2002). Escrt-III: An endosome-associated heterooligomeric protein complex Chan, R., Uchil, P.D., Jin, J., Shui, G.H., Ott, D.E., Mothes, W., and Wenk, M.R.
required for mvb sorting. Dev. Cell 3, 271–282. (2008). Retroviruses human immunodeficiency virus and murine leukemia virus
are enriched in phosphoinositides. J. Virol. 82, 11228–11238.
Bacia, K., Schwille, P., and Kurzchalia, T. (2005). Sterol structure determines
the separation of phases and the curvature of the liquid-ordered phase in Chen, B.J., and Lamb, R.A. (2008). Mechanisms for enveloped virus budding:
model membranes. Proc. Natl. Acad. Sci. USA 102, 3272–3277. can some viruses do without an ESCRT? Virology 372, 221–232.
Bajorek, M., Schubert, H.L., McCullough, J., Langelier, C., Eckert, D.M., Chen, B.J., Leser, G.P., Morita, E., and Lamb, R.A. (2007). Influenza virus
Stubblefield, W.M.B., Uter, N.T., Myszka, D.G., Hill, C.P., and Sundquist, hemagglutinin and neuraminidase, but not the matrix protein, are required
W.I. (2009). Structural basis for ESCRT-III protein autoinhibition. Nat. Struct. for assembly and budding of plasmid-derived virus-like particles. J. Virol. 81,
Mol. Biol. 16, 754–762. 7111–7123.

884 Cell 143, December 10, 2010 ª2010 Elsevier Inc.


Chiantia, S., Kahya, N., Ries, J., and Schwille, P. (2006). Effects of ceramide ordered-liquid disordered phase separation in model plasma membranes.
on liquid-ordered domains investigated by simultaneous AFM and FCS. Proc. Natl. Acad. Sci. USA 102, 6320–6325.
Biophys. J. 90, 4500–4508. Hancock, J.F. (2006). Lipid rafts: contentious only from simplistic standpoints.
Chung, H.Y., Morita, E., von Schwedler, U., Müller, B., Kräusslich, H.G., and Nat. Rev. Mol. Cell Biol. 7, 456–462.
Sundquist, W.I. (2008). NEDD4L overexpression rescues the release and infec- Hansen, C.G., and Nichols, B.J. (2010). Exploring the caves: cavins, caveolins
tivity of human immunodeficiency virus type 1 constructs lacking PTAP and and caveolae. Trends Cell Biol. 20, 177–186.
YPXL late domains. J. Virol. 82, 4884–4897.
Hanson, P.I., Roth, R., Lin, Y., and Heuser, J.E. (2008). Plasma membrane
Damm, E.M., Pelkmans, L., Kartenbeck, J., Mezzacasa, A., Kurzchalia, T., and
deformation by circular arrays of ESCRT-III protein filaments. J. Cell Biol.
Helenius, A. (2005). Clathrin- and caveolin-1-independent endocytosis: entry
180, 389–402.
of simian virus 40 into cells devoid of caveolae. J. Cell Biol. 168, 477–488.
Helfrich, W. (1973). Elastic properties of lipid bilayers: theory and possible
Demirov, D.G., Ono, A., Orenstein, J.M., and Freed, E.O. (2002a). Overexpres-
experiments. Z. Naturforsch. C 28, 693–703.
sion of the N-terminal domain of TSG101 inhibits HIV-1 budding by blocking
late domain function. Proc. Natl. Acad. Sci. USA 99, 955–960. Heller, H., Schaefer, M., and Schulten, K. (1993). Molecular dynamics simula-
tion of a bilayer of 200 lipids in the gel and in the liquid crystal phases. J. Phys.
Demirov, D.G., Orenstein, J.M., and Freed, E.O. (2002b). The late domain of
Chem. 97, 8343–8360.
human immunodeficiency virus type 1 p6 promotes virus release in a cell
type-dependent manner. J. Virol. 76, 105–117. Hemler, M.E. (2005). Tetraspanin functions and associated microdomains.
Nat. Rev. Mol. Cell Biol. 6, 801–811.
Ehrlich, M., Boll, W., Van Oijen, A., Hariharan, R., Chandran, K., Nibert, M.L.,
and Kirchhausen, T. (2004). Endocytosis by random initiation and stabilization Henne, W.M., Boucrot, E., Meinecke, M., Evergren, E., Vallis, Y., Mittal, R., and
of clathrin-coated pits. Cell 118, 591–605. McMahon, H.T. (2010). FCHo proteins are nucleators of clathrin-mediated
endocytosis. Science 328, 1281–1284.
Eisenberg, E., and Greene, L.E. (2007). Multiple roles of auxilin and hsc70 in
clathrin-mediated endocytosis. Traffic 8, 640–646. Hill, C.P., Worthylake, D., Bancroft, D.P., Christensen, A.M., and Sundquist,
W.I. (1996). Crystal structures of the trimeric human immunodeficiency virus
Elson, E.L., Fried, E., Dolbow, J.E., and Genin, G.M. (2010). Phase separation
type 1 matrix protein: implications for membrane association and assembly.
in biological membranes: Integration of theory and experiment. Annu. Rev.
Proc. Natl. Acad. Sci. USA 93, 3099–3104.
Biophys. 39, 207–226.
Ewers, H., Römer, W., Smith, A.E., Bacia, K., Dmitrieff, S., Chai, W.G., Mancini, Hinrichsen, L., Meyerholz, A., Groos, S., and Ungewickell, E.J. (2006). Bending
R., Kartenbeck, J., Chambon, V., Berland, L., et al. (2010). GM1 structure a membrane: how clathrin affects budding. Proc. Natl. Acad. Sci. USA 103,
determines SV40-induced membrane invagination and infection. Nat. Cell 8715–8720.
Biol. 12, 11–18, 1–12. Hu, J.J., Shibata, Y., Voss, C., Shemesh, T., Li, Z.L., Coughlin, M., Kozlov,
Fabrikant, G., Lata, S., Riches, J.D., Briggs, J.A.G., Weissenhorn, W., and M.M., Rapoport, T.A., and Prinz, W.A. (2008). Membrane proteins of the endo-
Kozlov, M.M. (2009). Computational model of membrane fission catalyzed plasmic reticulum induce high-curvature tubules. Science 319, 1247–1250.
by ESCRT-III. PLoS Comput. Biol. 5, e1000575. Huang, M.J., Orenstein, J.M., Martin, M.A., and Freed, E.O. (1995). p6Gag is
Falguières, T., Luyet, P.P., Bissig, C., Scott, C.C., Velluz, M.C., and required for particle production from full-length human immunodeficiency
Gruenberg, J. (2008). In vitro budding of intralumenal vesicles into late endo- virus type 1 molecular clones expressing protease. J. Virol. 69, 6810–6818.
somes is regulated by Alix and Tsg101. Mol. Biol. Cell 19, 4942–4955. Hurley, J.H., and Hanson, P.I. (2010). Membrane budding and scission by the
Farsad, K., and De Camilli, P. (2003). Mechanisms of membrane deformation. ESCRT machinery: it’s all in the neck. Nat. Rev. Mol. Cell. Biol. 8, 556–566.
Curr. Opin. Cell Biol. 15, 372–381. Im, Y.J., and Hurley, J.H. (2008). Integrated structural model and membrane
Fisher, R.D., Chung, H.Y., Zhai, Q.T., Robinson, H., Sundquist, W.I., and targeting mechanism of the human ESCRT-II complex. Dev. Cell 14, 902–913.
Hill, C.P. (2007). Structural and biochemical studies of ALIX/AIP1 and its role Im, Y.J., Wollert, T., Boura, E., and Hurley, J.H. (2009). Structure and function
in retrovirus budding. Cell 128, 841–852. of the ESCRT-II-III interface in multivesicular body biogenesis. Dev. Cell 17,
Ford, M.G.J., Mills, I.G., Peter, B.J., Vallis, Y., Praefcke, G.J.K., Evans, P.R., 234–243.
and McMahon, H.T. (2002). Curvature of clathrin-coated pits driven by epsin. Irie, T., Licata, J.M., Jayakar, H.R., Whitt, M.A., Bell, P., and Harty, R.N. (2004).
Nature 419, 361–366. Functional analysis of late-budding domain activity associated with the PSAP
Fotin, A., Cheng, Y.F., Sliz, P., Grigorieff, N., Harrison, S.C., Kirchhausen, T., motif within the vesicular stomatitis virus M protein. J. Virol. 78, 7823–7827.
and Walz, T. (2004). Molecular model for a complete clathrin lattice from elec- Ivanchenko, S., Godinez, W.J., Lampe, M., Kräusslich, H.G., Eils, R., Rohr, K.,
tron cryomicroscopy. Nature 432, 573–579. Bräuchle, C., Müller, B., and Lamb, D.C. (2009). Dynamics of HIV-1 assembly
Freed, E.O. (2002). Viral late domains. J. Virol. 76, 4679–4687. and release. PLoS Pathog. 5, e1000652.
Fujii, K., Munshi, U.M., Ablan, S.D., Demirov, D.G., Soheilian, F., Nagashima, Jackson, L.P., Kelly, B.T., McCoy, A.J., Gaffry, T., James, L.C., Collins, B.M.,
K., Stephen, A.G., Fisher, R.J., and Freed, E.O. (2009). Functional role of Alix Höning, S., Evans, P.R., and Owen, D.J. (2010). A large-scale conformational
in HIV-1 replication. Virology 391, 284–292. change couples membrane recruitment to cargo binding in the AP2 clathrin
Garcı́a-Sáez, A.J., and Schwille, P. (2010). Stability of lipid domains. FEBS adaptor complex. Cell 141, 1220–1229.
Lett. 584, 1653–1658. Jadwin, J.A., Rudd, V., Sette, P., Challa, S., and Bouamr, F. (2010). Late
Garrus, J.E., von Schwedler, U.K., Pornillos, O.W., Morham, S.G., Zavitz, K.H., domain-independent rescue of a release-deficient Moloney murine leukemia
Wang, H.E., Wettstein, D.A., Stray, K.M., Côté, M., Rich, R.L., et al. (2001). virus by the ubiquitin ligase itch. J. Virol. 84, 704–715.
Tsg101 and the vacuolar protein sorting pathway are essential for HIV-1 Jouvenet, N., Bieniasz, P.D., and Simon, S.M. (2008). Imaging the biogenesis
budding. Cell 107, 55–65. of individual HIV-1 virions in live cells. Nature 454, 236–240.
Gillooly, D.J., Morrow, I.C., Lindsay, M., Gould, R., Bryant, N.J., Gaullier, J.M., Kirkham, M., Fujita, A., Chadda, R., Nixon, S.J., Kurzchalia, T.V., Sharma, D.K.,
Parton, R.G., and Stenmark, H. (2000). Localization of phosphatidylinositol Pagano, R.E., Hancock, J.F., Mayor, S., and Parton, R.G. (2005). Ultrastruc-
3-phosphate in yeast and mammalian cells. EMBO J. 19, 4577–4588. tural identification of uncoated caveolin-independent early endocytic vehicles.
Gruenberg, J., and Stenmark, H. (2004). The biogenesis of multivesicular en- J. Cell Biol. 168, 465–476.
dosomes. Nat. Rev. Mol. Cell Biol. 5, 317–323. Kitadokoro, K., Bordo, D., Galli, G., Petracca, R., Falugi, F., Abrignani, S.,
Hammond, A.T., Heberle, F.A., Baumgart, T., Holowka, D., Baird, B., and Fei- Grandi, G., and Bolognesi, M. (2001). CD81 extracellular domain 3D structure:
genson, G.W. (2005). Crosslinking a lipid raft component triggers liquid insight into the tetraspanin superfamily structural motifs. EMBO J. 20, 12–18.

Cell 143, December 10, 2010 ª2010 Elsevier Inc. 885


Koga, Y., and Morii, H. (2005). Recent advances in structural research on ether Morita, E., and Sundquist, W.I. (2004). Retrovirus budding. Annu. Rev. Cell
lipids from archaea including comparative and physiological aspects. Biosci. Dev. Biol. 20, 395–425.
Biotechnol. Biochem. 69, 2019–2034. Murk, J.L.A.N., Humbel, B.M., Ziese, U., Griffith, J.M., Posthuma, G.,
Koivusalo, M., Jansen, M., Somerharju, P., and Ikonen, E. (2007). Endocytic Slot, J.W., Koster, A.J., Verkleij, A.J., Geuze, H.J., and Kleijmeer, M.J.
trafficking of sphingomyelin depends on its acyl chain length. Mol. Biol. Cell (2003). Endosomal compartmentalization in three dimensions: implications
18, 5113–5123. for membrane fusion. Proc. Natl. Acad. Sci. USA 100, 13332–13337.
Kostelansky, M.S., Schluter, C., Tam, Y.Y.C., Lee, S., Ghirlando, R., Beach, B., Neu, U., Woellner, K., Gauglitz, G., and Stehle, T. (2008). Structural basis of
Conibear, E., and Hurley, J.H. (2007). Molecular architecture and functional GM1 ganglioside recognition by simian virus 40. Proc. Natl. Acad. Sci. USA
model of the complete yeast ESCRT-I heterotetramer. Cell 129, 485–498. 105, 5219–5224.
Lata, S., Schoehn, G., Jain, A., Pires, R., Piehler, J., Gottlinger, H.G., and Weis- Nossal, R. (2001). Energetics of clathrin basket assembly. Traffic 2, 138–147.
senhorn, W. (2008). Helical structures of ESCRT-III are disassembled by VPS4. Ono, A., and Freed, E.O. (1999). Binding of human immunodeficiency virus
Science 321, 1354–1357. type 1 Gag to membrane: role of the matrix amino terminus. J. Virol. 73,
Lee, C., and Goldberg, J. (2010). Structure of coatomer cage proteins and the 4136–4144.
relationship among COPI, COPII, and clathrin vesicle coats. Cell 142, 123–132. Ono, A., and Freed, E.O. (2001). Plasma membrane rafts play a critical role in
Lee, M.C.S., Orci, L., Hamamoto, S., Futai, E., Ravazzola, M., and HIV-1 assembly and release. Proc. Natl. Acad. Sci. USA 98, 13925–13930.
Schekman, R. (2005). Sar1p N-terminal helix initiates membrane curvature Ono, A., Ablan, S.D., Lockett, S.J., Nagashima, K., and Freed, E.O. (2004).
and completes the fission of a COPII vesicle. Cell 122, 605–617. Phosphatidylinositol (4,5) bisphosphate regulates HIV-1 Gag targeting to the
Lenz, M., Crow, D.J.G., and Joanny, J.F. (2009). Membrane buckling induced plasma membrane. Proc. Natl. Acad. Sci. USA 101, 14889–14894.
by curved filaments. Phys. Rev. Lett. 103, 038101. Ono, A., Waheed, A.A., and Freed, E.O. (2007). Depletion of cellular cholesterol
Lindås, A.C., Karlsson, E.A., Lindgren, M.T., Ettema, T.J.G., and Bernander, R. inhibits membrane binding and higher-order multimerization of human immu-
(2008). A unique cell division machinery in the Archaea. Proc. Natl. Acad. Sci. nodeficiency virus type 1 Gag. Virology 360, 27–35.
USA 105, 18942–18946. Parton, R.G., and Simons, K. (2007). The multiple faces of caveolae. Nat. Rev.
Lineberry, N., Su, L., Soares, L., and Fathman, C.G. (2008). The single subunit Mol. Cell Biol. 8, 185–194.
transmembrane E3 ligase gene related to anergy in lymphocytes (GRAIL) Parton, R.G., Hanzal-Bayer, M., and Hancock, J.F. (2006). Biogenesis of cav-
captures and then ubiquitinates transmembrane proteins across the cell eolae: a structural model for caveolin-induced domain formation. J. Cell Sci.
membrane. J. Biol. Chem. 283, 28497–28505. 119, 787–796.
Lingwood, D., and Simons, K. (2010). Lipid rafts as a membrane-organizing Peck, J.W., Bowden, E.T., and Burbelo, P.D. (2004). Structure and function of
principle. Science 327, 46–50. human Vps20 and Snf7 proteins. Biochem. J. 377, 693–700.
Lipowsky, R. (1992). Budding of membranes induced by intramembrane Pelkmans, L., and Zerial, M. (2005). Kinase-regulated quantal assemblies and
domains. J. Phys. II France 2, 1825–1840. kiss-and-run recycling of caveolae. Nature 436, 128–133.
Liu, J., Kaksonen, M., Drubin, D.G., and Oster, G. (2006). Endocytic vesicle Pike, L.J. (2006). Rafts defined: a report on the Keystone Symposium on Lipid
scission by lipid phase boundary forces. Proc. Natl. Acad. Sci. USA 103, Rafts and Cell Function. J. Lipid Res. 47, 1597–1598.
10277–10282. Piper, R.C., and Katzmann, D.J. (2007). Biogenesis and function of multivesic-
Lorizate, M., Brügger, B., Akiyama, H., Glass, B., Müller, B., Anderluh, G., Wie- ular bodies. Annu. Rev. Cell Dev. Biol. 23, 519–547.
land, F.T., and Kräusslich, H.G. (2009). Probing HIV-1 membrane liquid order Polozov, I.V., Bezrukov, L., Gawrisch, K., and Zimmerberg, J. (2008). Progres-
by Laurdan staining reveals producer cell-dependent differences. J. Biol. sive ordering with decreasing temperature of the phospholipids of influenza
Chem. 284, 22238–22247. virus. Nat. Chem. Biol. 4, 248–255.
Martin-Serrano, J., Zang, T., and Bieniasz, P.D. (2001). HIV-1 and Ebola virus Pols, M.S., and Klumperman, J. (2009). Trafficking and function of the tetra-
encode small peptide motifs that recruit Tsg101 to sites of particle assembly to spanin CD63. Exp. Cell Res. 315, 1584–1592.
facilitate egress. Nat. Med. 7, 1313–1319. Popova, E., Popov, S., and Göttlinger, H.G. (2010). Human immunodeficiency
Matsuo, H., Chevallier, J., Mayran, N., Le Blanc, I., Ferguson, C., Fauré, J., virus type 1 nucleocapsid p1 confers ESCRT pathway dependence. J. Virol.
Blanc, N.S., Matile, S., Dubochet, J., Sadoul, R., et al. (2004). Role of LBPA 84, 6590–6597.
and Alix in multivesicular liposome formation and endosome organization. Pucadyil, T.J., and Schmid, S.L. (2009). Conserved functions of membrane
Science 303, 531–534. active GTPases in coated vesicle formation. Science 325, 1217–1220.
Matsuoka, K., Orci, L., Amherdt, M., Bednarek, S.Y., Hamamoto, S., Schek- Raiborg, C., and Stenmark, H. (2009). The ESCRT machinery in endosomal
man, R., and Yeung, T. (1998). COPII-coated vesicle formation reconstituted sorting of ubiquitylated membrane proteins. Nature 458, 445–452.
with purified coat proteins and chemically defined liposomes. Cell 93, Raposo, G., and Marks, M.S. (2007). Melanosomes—dark organelles
263–275. enlighten endosomal membrane transport. Nat. Rev. Mol. Cell Biol. 8,
McLaughlin, S., Wang, J.Y., Gambhir, A., and Murray, D. (2002). PIP(2) and 786–797.
proteins: interactions, organization, and information flow. Annu. Rev. Biophys. Reider, A., Barker, S.L., Mishra, S.K., Im, Y.J., Maldonado-Báez, L., Hurley,
Biomol. Struct. 31, 151–175. J.H., Traub, L.M., and Wendland, B. (2009). Syp1 is a conserved endocytic
McMahon, H.T., and Gallop, J.L. (2005). Membrane curvature and mecha- adaptor that contains domains involved in cargo selection and membrane
nisms of dynamic cell membrane remodelling. Nature 438, 590–596. tubulation. EMBO J. 28, 3103–3116.
Merritt, E.A., Sarfaty, S., van den Akker, F., L’Hoir, C., Martial, J.A., and Hol, Ren, X.F., and Hurley, J.H. (2010). VHS domains of ESCRT-0 cooperate in
W.G.J. (1994). Crystal structure of cholera toxin B-pentamer bound to receptor high-avidity binding to polyubiquitinated cargo. EMBO J. 29, 1045–1054.
GM1 pentasaccharide. Protein Sci. 3, 166–175. Resh, M.D. (2006). Trafficking and signaling by fatty-acylated and prenylated
Misra, S., Miller, G.J., and Hurley, J.H. (2001). Recognizing phosphatidylinosi- proteins. Nat. Chem. Biol. 2, 584–590.
tol 3-phosphate. Cell 107, 559–562. Reynwar, B.J., Illya, G., Harmandaris, V.A., Müller, M.M., Kremer, K., and
Möbius, W., van Donselaar, E., Ohno-Iwashita, Y., Shimada, Y., Heijnen, Deserno, M. (2007). Aggregation and vesiculation of membrane proteins by
H.F.G., Slot, J.W., and Geuze, H.J. (2003). Recycling compartments and the curvature-mediated interactions. Nature 447, 461–464.
internal vesicles of multivesicular bodies harbor most of the cholesterol found Robinson, M.S., and Bonifacino, J.S. (2001). Adaptor-related proteins. Curr.
in the endocytic pathway. Traffic 4, 222–231. Opin. Cell Biol. 13, 444–453.

886 Cell 143, December 10, 2010 ª2010 Elsevier Inc.


Rodal, S.K., Skretting, G., Garred, O., Vilhardt, F., van Deurs, B., and Subtil, A., Gaidarov, I., Kobylarz, K., Lampson, M.A., Keen, J.H., and McGraw,
Sandvig, K. (1999). Extraction of cholesterol with methyl-beta-cyclodextrin T.E. (1999). Acute cholesterol depletion inhibits clathrin-coated pit budding.
perturbs formation of clathrin-coated endocytic vesicles. Mol. Biol. Cell 10, Proc. Natl. Acad. Sci. USA 96, 6775–6780.
961–974. Takeda, M., Leser, G.P., Russell, C.J., and Lamb, R.A. (2003). Influenza virus
Römer, W., Berland, L., Chambon, V., Gaus, K., Windschiegl, B., Tenza, D., hemagglutinin concentrates in lipid raft microdomains for efficient viral fusion.
Aly, M.R.E., Fraisier, V., Florent, J.C., Perrais, D., et al. (2007). Shiga toxin Proc. Natl. Acad. Sci. USA 100, 14610–14617.
induces tubular membrane invaginations for its uptake into cells. Nature
Tang, C., Loeliger, E., Luncsford, P., Kinde, I., Beckett, D., and Summers, M.F.
450, 670–675.
(2004). Entropic switch regulates myristate exposure in the HIV-1 matrix
Rossman, J.S., Jing, X., Leser, G.P., and Lamb, R.A. (2010). Influenza virus M2 protein. Proc. Natl. Acad. Sci. USA 101, 517–522.
protein mediates ESCRT-independent membrane scission. Cell 142, 902–913.
Theos, A.C., Truschel, S.T., Tenza, D., Hurbain, I., Harper, D.C., Berson, J.F.,
Russell, C., and Stagg, S.M. (2010). New insights into the structural mecha- Thomas, P.C., Raposo, G., and Marks, M.S. (2006). A lumenal domain-depen-
nisms of the COPII coat. Traffic 11, 303–310. dent pathway for sorting to intralumenal vesicles of multivesicular endosomes
Saad, J.S., Miller, J., Tai, J., Kim, A., Ghanam, R.H., and Summers, M.F. involved in organelle morphogenesis. Dev. Cell 10, 343–354.
(2006). Structural basis for targeting HIV-1 Gag proteins to the plasma
Trajkovic, K., Hsu, C., Chiantia, S., Rajendran, L., Wenzel, D., Wieland, F.,
membrane for virus assembly. Proc. Natl. Acad. Sci. USA 103, 11364–11369.
Schwille, P., Brügger, B., and Simons, M. (2008). Ceramide triggers budding
Saad, J.S., Ablan, S.D., Ghanam, R.H., Kim, A., Andrews, K., Nagashima, K., of exosome vesicles into multivesicular endosomes. Science 319, 1244–1247.
Soheilian, F., Freed, E.O., and Summers, M.F. (2008). Structure of the myristy-
Traub, L.M., and Wendland, B. (2010). Cell biology: How to don a coat. Nature
lated human immunodeficiency virus type 2 matrix protein and the role of
465, 556–557.
phosphatidylinositol-(4,5)-bisphosphate in membrane targeting. J. Mol. Biol.
382, 434–447. Usami, Y., Popov, S., and Göttlinger, H.G. (2007). Potent rescue of human
immunodeficiency virus type 1 late domain mutants by ALIX/AIP1 depends
Saksena, S., Sun, J., Chu, T., and Emr, S.D. (2007). ESCRTing proteins in the
on its CHMP4 binding site. J. Virol. 81, 6614–6622.
endocytic pathway. Trends Biochem. Sci. 32, 561–573.
Samson, R.Y., Obita, T., Freund, S.M., Williams, R.L., and Bell, S.D. (2008). A Usami, Y., Popov, S., Popova, E., and Göttlinger, H.G. (2008). Efficient and
role for the ESCRT system in cell division in archaea. Science 322, 1710–1713. specific rescue of human immunodeficiency virus type 1 budding defects by
a Nedd4-like ubiquitin ligase. J. Virol. 82, 4898–4907.
Sandvig, K., Torgersen, M.L., Raa, H.A., and van Deurs, B. (2008). Clathrin-
independent endocytosis: from nonexisting to an extreme degree of van der Goot, F.G., and Gruenberg, J. (2006). Intra-endosomal membrane
complexity. Histochem. Cell Biol. 129, 267–276. traffic. Trends Cell Biol. 16, 514–521.
Scheiffele, P., Rietveld, A., Wilk, T., and Simons, K. (1999). Influenza viruses Veatch, S.L., Soubias, O., Keller, S.L., and Gawrisch, K. (2007). Critical fluctu-
select ordered lipid domains during budding from the plasma membrane. ations in domain-forming lipid mixtures. Proc. Natl. Acad. Sci. USA 104,
J. Biol. Chem. 274, 2038–2044. 17650–17655.
Schmitt, A.P., Leser, G.P., Morita, E., Sundquist, W.I., and Lamb, R.A. (2005). VerPlank, L., Bouamr, F., LaGrassa, T.J., Agresta, B., Kikonyogo, A., Leis, J.,
Evidence for a new viral late-domain core sequence, FPIV, necessary for and Carter, C.A. (2001). Tsg101, a homologue of ubiquitin-conjugating (E2)
budding of a paramyxovirus. J. Virol. 79, 2988–2997. enzymes, binds the L domain in HIV type 1 Pr55(Gag). Proc. Natl. Acad. Sci.
Shibata, Y., Hu, J.J., Kozlov, M.M., and Rapoport, T.A. (2009). Mechanisms USA 98, 7724–7729.
shaping the membranes of cellular organelles. Annu. Rev. Cell Dev. Biol. 25, Voeltz, G.K., and Prinz, W.A. (2007). Sheets, ribbons and tubules - how organ-
329–354. elles get their shape. Nat. Rev. Mol. Cell Biol. 8, 258–264.
Shnyrova, A.V., Ayllon, J., Mikhalyov, I.I., Villar, E., Zimmerberg, J., and Welsch, S., Müller, B., and Kräusslich, H.G. (2007). More than one door -
Frolov, V.A. (2007). Vesicle formation by self-assembly of membrane-bound Budding of enveloped viruses through cellular membranes. FEBS Lett. 581,
matrix proteins into a fluidlike budding domain. J. Cell Biol. 179, 627–633. 2089–2097.
Simons, M., and Raposo, G. (2009). Exosomes—vesicular carriers for intercel- Williams, R.L., and Urbé, S. (2007). The emerging shape of the ESCRT
lular communication. Curr. Opin. Cell Biol. 21, 575–581. machinery. Nat. Rev. Mol. Cell Biol. 8, 355–368.
Solon, J., Gareil, O., Bassereau, P., and Gaudin, Y. (2005). Membrane defor-
Wollert, T., and Hurley, J.H. (2010). Molecular mechanism of multivesicular
mations induced by the matrix protein of vesicular stomatitis virus in a minimal
body biogenesis by ESCRT complexes. Nature 464, 864–869.
system. J. Gen. Virol. 86, 3357–3363.
Wollert, T., Wunder, C., Lippincott-Schwartz, J., and Hurley, J.H. (2009).
Sorkin, A., and von Zastrow, M. (2009). Endocytosis and signalling: intertwin-
Membrane scission by the ESCRT-III complex. Nature 458, 172–177.
ing molecular networks. Nat. Rev. Mol. Cell Biol. 10, 609–622.
Stagg, S.M., Gürkan, C., Fowler, D.M., LaPointe, P., Foss, T.R., Potter, C.S., Wright, E.R., Schooler, J.B., Ding, H.J., Kieffer, C., Fillmore, C., Sundquist,
Carragher, B., and Balch, W.E. (2006). Structure of the Sec13/31 COPII coat W.I., and Jensen, G.J. (2007). Electron cryotomography of immature HIV-1
cage. Nature 439, 234–238. virions reveals the structure of the CA and SP1 Gag shells. EMBO J. 26,
2218–2226.
Stagg, S.M., LaPointe, P., Razvi, A., Gürkan, C., Potter, C.S., Carragher, B.,
and Balch, W.E. (2008). Structural basis for cargo regulation of COPII coat Yorikawa, C., Shibata, H., Waguri, S., Hatta, K., Horii, M., Katoh, K., Kobaya-
assembly. Cell 134, 474–484. shi, T., Uchiyama, Y., and Maki, M. (2005). Human CHMP6, a myristoylated
ESCRT-III protein, interacts directly with an ESCRT-II component EAP20
Stawiecka-Mirota, M., Pokrzywa, W., Morvan, J., Zoladek, T., Haguenauer-
and regulates endosomal cargo sorting. Biochem. J. 387, 17–26.
Tsapis, R., Urban-Grimal, D., and Morsomme, P. (2007). Targeting of Sna3p
to the endosomal pathway depends on its interaction with Rsp5p and multive- Zhang, Y.Q., Qian, H.Y., Love, Z., and Barklis, E. (1998). Analysis of the
sicular body sorting on its ubiquitylation. Traffic 8, 1280–1296. assembly function of the human immunodeficiency virus type 1 gag protein
Stimpson, H.E.M., Toret, C.P., Cheng, A.T., Pauly, B.S., and Drubin, D.G. nucleocapsid domain. J. Virol. 72, 1782–1789.
(2009). Early-arriving Syp1p and Ede1p function in endocytic site placement Zimmerberg, J., and Kozlov, M.M. (2006). How proteins produce cellular
and formation in budding yeast. Mol. Biol. Cell 20, 4640–4651. membrane curvature. Nat. Rev. Mol. Cell Biol. 7, 9–19.

Cell 143, December 10, 2010 ª2010 Elsevier Inc. 887


Leading Edge
Primer

Lipidomics: New Tools and Applications


Markus R Wenk1,2,*
1National University of Singapore, Yong Loo Lin School of Medicine, Department of Biochemistry and Faculty of Science,

Department of Biological Sciences, Centre for Life Sciences (CeLS), 28 Medical Drive, Singapore 117456
2Swiss Tropical and Public Health Institute and the University of Basel, Switzerland

*Correspondence: markus_wenk@nuhs.edu.sg
DOI 10.1016/j.cell.2010.11.033

Once viewed simply as a reservoir for carbon storage, lipids are no longer cast as bystanders in the
drama of biological systems. The emerging field of lipidomics is driven by technology, most notably
mass spectrometry, but also by complementary approaches for the detection and characterization
of lipids and their biosynthetic enzymes in living cells. The development of these integrated tools
promises to greatly advance our understanding of the diverse biological roles of lipids.

Lipids are not genetically encoded. Instead, like other small Mass Spectrometry-Based Lipidomics
molecules they are generated and metabolized by enzymes The first reports of mass spectrometric (MS) analysis of complex
that are influenced by the environment of a given biological lipid mixtures via soft ionization techniques (matrix-assisted
system, for instance by diet and temperature. Although still laser desorption ionization, MALDI, and electrospray ionization,
poorly defined, some estimates have placed the number of ESI) date back to the 1990s (Han and Gross, 1994; Kim et al.,
distinct chemical entities within the lipid sphere between 1994). A large number of methods have been developed since
10,000 and 100,000. Although it is unclear how and why nature then, and many biologically important lipids can now be
generates this staggering diversity, there is an increasing aware- analyzed on a fairly routine basis. However, unlike genomics
ness across many disciplines of the critical importance of lipids and proteomics, which are well represented in various forms at
in all aspects of life. leading research institutions worldwide, this is not yet the case
First, coordinated lipid anabolism and catabolism is a key for lipidomics (Figure 2).
molecular integrator of energy homeostasis, membrane struc- A major difference in mass spectrometry of lipids (as opposed
ture and dynamics, and signaling (Figure 1) with imbalances in to proteins) is the large chemical diversity found in these
lipid metabolism contributing to diverse phenotypes and disease molecules (Figure 1 and Figure 3A) (Fahy et al., 2005). As a conse-
states. Second, there is an expanding number of drugs that quence, it is currently not possible to comprehensively measure
target lipid metabolic and signaling pathways, including the the lipidome of a cell or tissue in a single experiment. Further-
well-known and profitable cholesterol-lowering agents (statins) more, one often does not know what precise alteration in lipids
and cyclooxygenase inhibitors. For therapeutic intervention in to expect in any given case. Thus, first surveys are often
diseases ranging from inflammation and cancer to metabolic exploratory, which is to say they often have ‘‘untargeted’’ read-
diseases, lipid researchers are seeking specific regulators of outs (Figure 3B). Such methods should have high mass accuracy
numerous targets, including phosphatidylinositol (PI) 3-kinases, and resolution, a characteristic of time of flight and Orbitrap mass
nuclear hormone receptors (for instance, liver X receptor, LXR; spectrometry. Fragmentation of an ion of interest is then used for
peroxisome proliferator-activated receptors, PPARs), sphingo- identification (Figure 3C). Analysis of fragmentation pathways
sine, and ceramide kinases. A recent example is FTY720, has led to a detailed understanding of ‘‘bonding’’ between the
approved for the treatment of multiple sclerosis in October different building blocks found in lipids (such as fatty acids,
2010, an immunosuppressant that targets sphingosine-1-phos- sphingoid bases, and head groups). It has also formed a basis
phate receptors (but interestingly does not inhibit serine palmi- for ‘‘shotgun’’ lipidomics in which precursor lipids are determined
toyl transferase, unlike its mother compound myriocin, a natural based on characteristic fragment ions. Other targeted ap-
product). proaches based on tandem mass spectrometry are now available
The scarcity of pertinent tools has led to investments in for analysis of many different classes of lipids and in complex
programs to develop new approaches for lipid research. Collec- mixtures (Wenk, 2005; Blanksby and Mitchell, 2010).
tively, these efforts have added momentum to the field (reflected The coordinated efforts of LIPID MAPS (http://www.lipidmaps.
in part by the increasing number of publications and conferences org) have laid the groundwork for standardization (for example, in
dedicated to lipids), which promises to address fundamental protocols and in the nomenclature relevant to databases) in the
questions of lipid function and to meet practical demands in field and to foster the commercial availability of many pure and
the applied sciences. The aim of this Primer is to introduce the synthetic lipid standards. These standards are deuterated
basic concepts behind biochemical (mass spectrometry-based) versions or close chemical analogs of naturally occurring lipids
lipidomics, to discuss how these approaches are being inte- that are used to quantify ion responses. They are used in a rapidly
grated with complementary techniques, and to offer a view on increasing number of lipidomic programs around the world
the future of the field. (LIPID MAPS, Kansas Lipidomics Research Center, COBRE,

888 Cell 143, December 10, 2010 ª2010 Elsevier Inc.


Figure 1. The Cellular Compartments of Common Biological Lipids
Lipids are small molecules of enormous chemical diversity. Unlike other major biomolecules (i.e., nucleic acids, polysaccharides, and proteins), they are not
polymers of relatively small numbers of chemically distinct building blocks. Instead, they are the result of anabolic and catabolic reaction pathways that are under
complex dietary and physiological control. It is thus difficult to define, name, and categorize lipids in a coherent and comprehensive fashion. Lipids of different
chemical structures are highly organized within a typical eukaryotic cell. The lipid portion of biological membranes is to a large extent made up of glycerophos-
pholipids, sterols, and sphingolipids (blue box, structures of three representative lipids from the different classes are shown). These are all examples of
amphiphilic lipids, which have both hydrophilic and hydrophobic portions. The membrane-associated lipids are not evenly distributed. Some organelles are
enriched with certain lipids (for instance, cardiolipin in mitochondria and lysobisphosphatidic acid/bis(monoacylglycero)phosphate in endosomes), and lateral
distribution within membranes leads to functional domains. Metabolism of membrane lipids generates highly active signaling molecules (red box). These lipids,
often much more soluble and diffusible than their membrane-associated parent, control organismal physiology. Very nonpolar lipids, such as sterol-esters and
triglycerides, are assembled in the endoplasmic reticulum and stored in lipid bodies within cells.

WUSTL, Southampton Lipidomics Research Group, Lipidomics. characterized by mass spectrometric methods and in complex
Net, LipidX, Lipidomics Research Center Graz, LipidProfiles). In lipid extracts, a task that would otherwise require laborious
addition to these centers that harbor substantial analytical (and often indirect) techniques for detection. It should however
capabilities, individual laboratories are increasingly engaging in be noted that, even with the major advances made by MS
the analysis of specific metabolites and lipid pathways. The latter approaches, the detection of lipid species of very low abundance
development can be explained, at least in part, by lowered costs is still a major challenge (discussed below).
and easier handling of modern mass spectrometers. High-resolution mass spectrometry aids in identification of
Two technical characteristics, high sensitivity and high previously uncharacterized lipids and discrimination between
specificity (mass resolution), account for the success of mass lipids with similar mass and chemical structures. It has also
spectrometry in lipid analysis. For example, mass spectrometry provided evidence for the presence of isomeric species (which
has provided a detailed knowledge of the chemical (lipid) compo- have the same chemical formula but different structures) and
sition of highly purified vesicles or viruses, preparations in which isobaric species (ions with the same mass) in cellular lipidomes.
sample amounts are limited. These ‘‘organelles’’ stay largely For example, ether phospholipids are often isomeric with other
intact during preparation and are thus biochemically more acces- abundant cellular phospholipids (Yang et al., 2007).
sible than other membrane fractions. Studies such as these There are several analytical challenges that cannot be
provide evidence for segregation of specific sterol and sphingo- addressed satisfactorily by mass spectrometry alone. These
lipid species during formation of secretory vesicles at the trans- include unequivocal assignment of structures: double bond
Golgi (Klemm et al., 2009) or enrichment of certain membrane configurations are difficult to determine and cannot be readily
lipids during formation of viruses at donor membranes of the assigned based on tandem mass spectrometry (Thomas et al.,
host cell (Brügger et al., 2006; Chan et al., 2008). Sensitivity is 2009); chemical derivatization and/or nuclear magnetic reso-
also required for lipid metabolites that occur at low and transient nance might be required for structure determination of complex
levels. Phosphoinositides or fatty acyl derivatives have all been glycolipids.

Cell 143, December 10, 2010 ª2010 Elsevier Inc. 889


Figure 2. Lipidomics Is an Emerging Field
The sequencing of the human genome in the year 2000 sparked interest and
investment in technologies and programs for the systematic analysis of
genetic variation. As a result, the study of genomes and proteomes has Figure 3. New Research Tools for Lipidomics
produced large numbers of findings reported in the scientific literature The precise size and dynamics of a cellular lipidome remains poorly under-
(measured here as the cumulative numbers of citations in PubMed over stood both on theoretical as well as on experimental grounds. Hundreds to
time). Complete genomes can now be sequenced (and annotated) in a matter thousands of different chemical entities are recovered in an organic extract
of days or weeks, and current development is primarily focused on lowering from a biological specimen where lipids are assembled in a coordinated
the cost per sequenced base. Many commercial products are available for fashion.
sample preparation, analysis, and interpretation. This is also true for protein (A) An assembly of fatty acyl-containing membrane lipids with different head
analysis, though it is still challenging to determine whole proteomes. Proteo- group decorations, for example phosphorylated (1) or glycosylated (2) forms,
mics has gained tremendously from mass spectrometry for peptide detection is depicted. Lipases hydrolyze lipids at various positions. Phospholipases
and quantification. The boundaries for experimental measurements (such as A2 generate lysolipids (3), which have profound structural effects on lipid
number of proteins) are reasonably well established based on genetic informa- assemblies as well as signaling functions via G protein-coupled receptors.
tion. None of the above is the case for lipidomic analysis. Currently, most of the Less well understood are other modifications such as hydroxylations or
mass spectrometric measurements are conducted by a few consortia and methylations and oxidations or nitrosylation introduced via enzymatic and
laboratories. The community is growing very rapidly, however, and these chemical reactions, respectively (4).
activities have led to interest in many disciplines. The first studies combining (B) Single stage and tandem mass spectrometry (C) have yielded tremendous
genomics and lipidomics have just been published. Given the central role of insight into chemical details of cellular lipids. An ion with a mass/charge (m/z)
lipids as key metabolites with remarkably diverse biological roles, the field of ratio corresponding to the expected structure shown in red (structure 1 in
lipidomics may follow a trajectory comparable to the developments seen in panel A) can be fragmented and characterized based on product ions, which
genomics and proteomics over the past decade. in the case of glycerophospholipids and negative mode ionization are fatty
acyls, head group, and backbone-derived moieties.
(D) Complementary technologies that are currently being developed include
Current Challenges chemical-biological approaches to probe lipid-protein interactions. For
example, lipid-binding domains are used to visualize lipids in living cells or
Integration
to locally interfere with lipid metabolism.
Lipids and their metabolites serve as integrators of many cellular (E) Analogues of lipids can be introduced into cells to interfere with protein-lipid
functions. Energy homeostasis is tightly coupled to fatty acid interactions, to inhibit enzymes, or for biochemical isolation of lipid-binding
metabolism, and fatty acids are key building blocks of many factors.
(F) Finally, bioinformatic tools will need to be further developed to support
cellular lipids (Figure 1). Thus, it seems evident that lipid metab- these experimental technologies (panels B–E) to facilitate combinations of
olism must follow a very coordinated program during the cell genomics and lipidomics, compare between biological species, and identify
cycle and proliferation. Given that cancer cells are dependent clinically relevant biomarkers.
on fatty acids for the synthesis of membranes and signaling
lipids, fatty acid synthase (FAS) is considered a potential thera-
peutic target. Recent work using cell biological approaches phenotypes other than growth (Patwardhan et al., 2010). The
(Kurat et al., 2009) and functional proteomics (Nomura et al., known lipid ‘‘signaling’’ network is thus poised for a great
2010) discovered that breakdown of glycerolipids via lipases is expansion, in particular in the context of human disease
a key mechanism for the generation of free fatty acids during (Wymann and Schneiter, 2008).
cell proliferation, thus metabolically coupling lipid bodies with These are recent examples of integrated experimental
membrane synthesis (Figure 1) (Singh et al., 2009). Similar approaches involving experiments that combine lipid biochem-
metabolic coupling, for instance, between membrane lipids istry (via mass spectrometry or other means) and functional
and soluble lipid mediators, is likely to be discovered for specific readouts. The first challenge in such endeavors is defining the

890 Cell 143, December 10, 2010 ª2010 Elsevier Inc.


sample. Whole-cell extracts are often used, and as a conse- in lipid levels will be relevant not only for population-based
quence all information on spatial distribution is lost (van Meer, studies but also at the level of individuals.
2005). In the future, ensembles of protein markers will allow for Data Analysis and Interpretation
better identification of subcellular organelles (Andreyev et al., Arguably, proteomics was transformed by the development
2010) and thus aid in preparations of lipid fractions related to of search algorithms that enabled assignment of protein
specific cellular functions. The generation of lipid extracts prior sequences by comparisons of experimental and theoretical MS
to analysis is a critical aspect that currently attracts only fragmentation patterns of tryptic peptides. In the case of lipids,
moderate attention. Biochemical fractionation has inherent the bioinformatic needs are different and to a substantial extent
limitations in terms of the resulting purity and integrity of the remain unmet. Biological lipids are small, nonpolymeric mole-
samples. Furthermore, recovery rates during partitioning in cules (with molecular weights less than 2000 Da). Typical analyt-
organic solvents strongly depend on the lipid class, and ical readouts in ‘‘untargeted’’ approaches include retention time
nonquantitative recovery during extraction introduces variability. (in the case of LC separation), mass-to-charge ratio, (m/z, ideally
Natural Variation with high mass accuracy), and information on fragment ions (in
Metabolites can vary substantially between individuals and on the case of tandem MS). ‘‘Targeted’’ analysis delivers a matrix
a day-to-day basis (Assfalg et al., 2008), which complicates of lipid identities (including precursors to fragment ions) and their
comparative studies. Often, the degree of natural variation of intensities. Typical informatic frameworks include data process-
a metabolite/lipid in an individual or population is not known. In ing (peak integration, identification, and normalization), statistics
mice, some lysolipids display remarkable circadian patterns (univariate or multivariate), and integration into pathways (e.g.,
with up to 2-fold differences in their levels (Minami et al., 2009). the Kyoto Encyclopedia of Genes and Genomes, KEGG) or other
Whereas modern mass spectrometers provide linear outputs datasets (see above). Open source and commercial software
over several orders of magnitude (linear dynamic range), the packages are now becoming available to support some of these
biologically relevant dynamic range is lipid specific, varying functions (Wheelock et al., 2009; Blanksby and Mitchell, 2010).
from 2- to 3-fold for abundant membrane lipids to 10- to 100- Building databases for lipids follows closely related efforts for
fold in extreme cases (such as mediator lipids). Importantly, other small molecule metabolites (Fahy et al., 2007; Kind et al.,
lipids are also found at very different basal concentrations and 2009). Appropriate data processing and validation will be a
have distinct temporal dependencies. Cellular fractionation, particularly critical element in biomarker discovery where many
liquid chromatographic separation prior to MS (LC-MS), time hundreds of different lipids are measured in human body fluids
course experiments, as well as selective capture of lipids will such as blood plasma (Quehenberger et al., 2010).
be required to overcome analytical challenges to resolving lipid These examples illustrate the benefits of data integration at all
species of interest when they are of low abundance. In cellular levels and across scientific disciplines. Biochemical analysis of
studies, metabolic labeling with chemical isotopes of lipid lipids by mass spectrometry is only one element in such interdis-
precursors followed by mass spectrometry is an elegant and ciplinary projects but will be a key tool in many fields including
powerful way to study kinetics of incorporation and turnover of cell and developmental biology, molecular medicine, and
some classes of lipids (Postle and Hunt, 2009). nutrition (Shevchenko and Simons, 2010).
In population studies, efforts to combine mass spectrometry-
based lipidomics with genomics have been guided by the Future Developments and Prospects
technical feasibility of measuring lipids on a large scale, the New features and functions will undoubtedly be introduced to
popularity of genome-wide association studies (GWAS), and augment those currently used in the MS analysis of lipids. For
human diseases associated with aberrant lipid metabolism. instance, ion mobility mass spectrometry (IM-MS), which com-
Strong associations are found between the levels of some poly- bines information of molecular shape (the collisional cross-
unsaturated fatty acids (measured as fatty acid methyl esters by section) with the mass/charge, has not yet been extensively
gas chromatography-MS) and lipid desaturases (Tanaka et al., applied to the analysis of lipids. Biophysical studies have shown
2009). GWAS with larger numbers of metabolic traits, measured that the double bond configuration of fatty acyls determines the
via MS methods introduced above, have been conducted and conformation of lipids in bilayers, and this structural character-
published recently. In one study following 33 metabolic traits, istic might also affect the collisional cross-section. It is also
several circulating sphingolipids were found to be under strong conceivable that ion mobility is affected by head group geometry
genetic control (Hicks et al., 2009). In another study with 163 (which is impacted by phosphorylation and glycosylation). Thus,
metabolites (including major glycerophospholipids as well as it is likely that IM-MS will provide valuable information that is
acyl-CoAs and amino acids), ratios of substrate-product otherwise difficult to obtain. IM-MS has been successfully
concentrations, rather than single metabolite levels, reduced used for detection of lipids directly from tissue sections via
variance and improved statistical significance (Illig et al., 2010). MALDI (Ridenour et al., 2010). The resulting ‘‘image’’ containing
Sequencing of candidate genes in individuals at the extremes mass spectral data yields spatial information on lipid distribution
of the population distribution with respect to lipoprotein levels (Murphy et al., 2009).
led to the discovery of nonsynonymous sequence variants in Many lipids bind to cations, such as Ca2+ and Mg2+, via their
enzymes involved in cholesterol metabolism (Fahmi et al., charged head groups. These reactions regulate assembly of
2008). Targeted genomics of lipid metabolic pathways in lipids in biological as well as cell-free systems. Lipid oxidation
combination with biochemical lipid analysis is an area of great on the other hand is in part coupled to free radical chemistry.
future potential. The link between genetic variation and changes Thus, elemental composition of lipid preparations (metal ions in

Cell 143, December 10, 2010 ª2010 Elsevier Inc. 891


particular) could yield important additional information related to for identifying new lipid-binding factors and off-targets (Yang
biomarker discovery. Such information can be determined by et al., 2010).
inductively coupled plasma (ICP) mass spectrometry (Becker Enzymatic versus Chemical Modification of Lipids
and Jakubowski, 2009), a method that is amenable to imaging. Unlike the generation of ‘‘lipid mediators’’ (Serhan, 2009), oxida-
An interesting new technique for imaging of lipids is coherent tion of intact glycerophospholipids can be mediated by reactive
anti-stokes Raman scattering (CARS) microscopy. Images are oxygen species in addition to enzymes such as lipoxygenases.
generated based on the vibrational states of molecules, such Typically, oxidation of polyunsaturated fatty acyls (PUFAs) in
as the CH2 bonds found in fatty acyls. Thus, CARS does not glycerophospholipids by reactive oxygen species leads to a
require external labels. It is rapid (1 s/frame) and can be used variety of different products including hydroxyls, hemiacetals,
for live imaging. Currently, CARS works well in applications and furans. Oxidized forms of membrane phospholipids are
with high signal-to-noise ratios, for example lipid bodies that short-lived, reactive species that undergo fatty acyl chain short-
harbor many CH2 segments (Volkmer, 2005; Müller and Zum- ening or covalent adduct formation with nearby proteins.
busch, 2007). Future developments might lead to CARS Furthermore, such ‘‘damaged’’ lipids occur in very low abun-
spectroscopy, moving the technique beyond the monitoring of dance compared to their parent lipid thus complicating analytical
a single frequency such that C-C double bonds (lipid unsatura- capture (Zemski Berry et al., 2010). These lipids might exert their
tion), or ester bonds could also be imaged. Such refinements effects via receptor activation (for instance via G protein-coupled
should also help overcome background problems. receptors, nuclear receptors, and/or innate immune receptors;
Greenberg et al., 2006) and other mechanisms due to their reac-
Molecular Recognition of Lipids tivity and biophysical properties (Deigner and Hermetter, 2008).
Substrate Specificities The proportion of fatty acyls differs dramatically between
Progress has been made toward ascertaining the determinants organs. The brain, for example, is very rich in polyunsaturated
of specificity for lipid enzymes and protein effectors. For fatty acyls (such as arachidonic acid, C20:4, and docosahexae-
instance, mammalian FAS generates mainly palmitic acid noic acid, C22:6), whereas the liver contains primarily saturated
(C16:0,16 carbons and no double bonds between them) and to and monounsaturated fatty acyls. It is thus conceivable that
a lesser extent produces C14:0 and C18:0. This specificity is oxidative stress might produce different lipid reaction products
determined, at least in part, by the thioesterase domain of FAS depending on the precise organ and/or cell type affected. This
and the geometry of its catalytic cavity (Pemble et al., 2007). would influence downstream reactions such as activation of
Phospholipases and lipid kinases are other well-studied exam- cell surface or nuclear lipid receptors and elevation of antibodies
ples. Both require interfacial targeting and specific recognition directed against lipids (discussed below). This characteristic is
of their substrates for catalysis (Manford et al., 2010). Phosphoi- also relevant for biomarker development, which would require
nositides, an important class of cellular signaling lipids, are careful inspection and understanding of chemical versus enzy-
recognized by a large number of protein effectors that have matic oxidations as well as an appreciation of the potential for
vastly different folds. Sophisticated technology based on induc- selective transport as in the case of oxidized sterols.
ible formation of protein-protein complexes (Suh et al., 2006) or Antibodies Directed against Lipids
peptide sensors has helped to monitor (using optical imaging) With the important exception of glycolipids, relatively few
the distributions of phosphoinositides and associated protein antibodies that recognize specific lipids have been described.
factors within cells (Fairn et al., 2009). This cannot be ascribed solely to an inherent lack of antigenicity
Despite these advances, it is clear that recognition of lipids at on the part of lipids. Certain glycosphingolipids, which are
the atomic level remains poorly understood (Manford et al., present in normal cells, are more abundant in tumor cells and
2010; Ernst et al., 2010). It is becoming increasingly evident elicit an antibody response (Hakomori and Zhang, 1997). In
that highly specific lipid-lipid and lipid-protein interactions many cases, the precise chemical nature of the antigens remains
regulate cell physiology (Guan et al., 2009; Shevchenko and unclear and is dependent on cell type and experimental condi-
Simons, 2010). It will therefore be a challenge to understand tions. Heteromeric glycolipid complexes, rather than an indi-
and therapeutically target such interactions. Lipid enzymes are vidual glycolipid, modulate (auto)antibody responses (Rinaldi
an interesting case to consider given that they produce media- et al., 2010), meaning that the antigenic determinant consists
tors that have closely related structures but opposing functions of a combination of two (or more) glycans. One explanation for
(Figure 1). Cyclooxygenase 2 (COX-2) is involved in the genera- this might be the different surface arrangement and presentation
tion of both inflammatory compounds (e.g., prostaglandins) as of glycosphingolipids on tumor cells. Indeed, it is becoming
well as anti-inflammatory compounds from similar, albeit chem- increasingly accepted that ‘‘local topography’’ influences
ically distinct, substrates (glycerophospholipids with omega-6 antigenicity and immunogenicity of glycosphingolipids. Another
and omega-3 fatty acyl, respectively) (Groeger et al., 2010). explanation is that anti-lipid antibodies (of a limited range of iso-
Acetyl-salicylic acid (aspirin), a natural compound that targets types) against cardiolipin and other phospholipids might be
COX-2, decreases production of proinflammatory mediators present at considerable frequencies but in hidden forms, for
and increases production of anti-inflammatory compounds. example, as circulating immune complexes, and therefore
This shift in COX activity is not achieved by synthetic and selec- unable to engage normal tissues or cells (Alving, 2006). Lipids
tive inhibitors of COX that are designed based on active site from external sources are likely to produce immune responses.
catalysis. Chemically synthesized derivatives of natural products Such lipids come from the diet or pathogens or are derivatives
are therefore promising tools for probing enzyme cavities and of endogenous lipids, such as oxidized lipids and their adducts.

892 Cell 143, December 10, 2010 ª2010 Elsevier Inc.


Indeed, there is increasing evidence for the presence of anti-lipid lines have been used to target phospholipases in protein
antibodies, for example in individuals with HIV infections extracts with the proteins then identified via alkyne-azide-based
and autoinflammatory conditions such as multiple sclerosis click chemistry (Tully and Cravatt, 2010).
(Kanter et al., 2006). Synthetic forms of lipid A have been used Lipidomics across Biological Species
to raise monoclonal antibodies that can be utilized in vivo to Many lipid metabolic pathways are conserved in function from
target gram-negative bacteria (Syed et al., 1992). Antibodies yeast to man. However, it is not trivial to search for lipid enzymes,
against lipid components of mycobacteria have been in modulators of enzymes, or even lipid effectors based on protein
development for a number of years as a way of controlling sequence information alone. Phosphatidylinositol transfer
M. tuberculosis and M. leprae infections. These include anti- proteins (PITPs), for example, share some functional redundancy
bodies specific for lipoteichoic acid and lipoarabinomannan but almost no sequence similarity between yeast (Sec14p-like)
(Hamasur et al., 2004). and metazoans. They also adopt very different structural folds.
Relatively little is known about the precise molecular require- Certain lipid classes differ substantially between biological
ments for successful generation of antibodies against lipids species. The sphingolipids in yeast, mammals, and insects
either in terms of their presentation during immunization in vivo have very different head group decorations, hydroxylation
or their selection in vitro. In an interesting example, liposomes patterns, and lengths of fatty acids and long chain bases.
with very high content of cholesterol (71%) were used to Thus, in addition to experimental methods (Guan et al., 2009;
generate monoclonal antibodies that recognized membranes Ejsing et al., 2009), new in silico approaches (Fahy et al., 2007;
with high cholesterol (as well as crystalline cholesterol in vitro) Baker et al., 2008) are needed to tap the information stored in
but not liposomes with 40% cholesterol (Swartz et al., 1988). existing databases, such as gene ontologies and protein-protein
Thus, there is reason to the hope that it will be possible to interaction maps of model organisms.
generate new and specific lipid antibodies with improved Our appreciation of lipid heterogeneity, biosynthetic routes,
technologies for presentation and selection. Production of and process engineering has been substantially bolstered by
pure, synthetic, and stable lipids is one prerequisite. A second, work coming out of the environmental and plant sciences. These
more complicated issue is the selection of the lipid species developments are supported by the belief that whatever can be
that acts as the antigen. Such antibodies, if successful, would derived from fossil fuels can also be made from vegetable oils
be entirely new tools for basic research in membrane trafficking and the fact that the cost differential between these two sources
with applications in immunohistochemistry, cytochemistry, and of lipids has decreased over the past 20 years. Currently, 90% of
biochemistry. If proven highly specific, such antibodies could fossil oil is converted to fuel and 10% is used by the petrochem-
be used for clinical applications, including for diagnostics or ical industry for production of plastics, detergents, etc. This
potentially for therapeutic purposes. presents numerous opportunities for lipidomic research and
Chemical Biology of Lipids development, in addition to the obvious desire to generate
Small-molecule chemical probes (so-called activity-based or biofuels via food crops or other feedstock.
affinity-based probes) have in recent years become increasingly Take for example spermaceti oil (cetyl-palmitate, a wax),
popular for the study of kinases, phosphatases, and hydrolytic which was harvested from the heads of sperm whales and
enzymes (hydrolases and proteases). To date relatively little used in lubricants until whale hunting bans mandated the search
has been done to engineer lipid-based probes capable of for alternative sources. It is indeed difficult to find a petroleum-
detecting or capturing lipid-interacting proteins. ‘‘Click chem- based replacement. Likewise, a wax derived from the seed of
istry’’ is a recently developed approach in which small molecules the Jojoba plant is used in cosmetics and would also be a useful
can be joined selectively and has been used for selective industrial lubricant were it not for its current cost of production.
chemical remodeling of cell-surface glycoproteins (Mahal Several large-scale programs are currently addressing this
et al., 1997). The technique builds on the assumption that need. These efforts will likely tap into lipidomic technologies at
biosynthetic enzymes are promiscuous enough to allow incorpo- various levels. Ultra high-resolution mass spectrometry can be
ration of precursors that have a chemically reactive ‘‘molecular used to provide detailed chemical information of petroleum
handle’’ (a bio-orthogonal reporter) that subsequently can be crude oils from different sources (Marshall and Rodgers, 2008).
used to form a covalent bond with a fluorophore for visualization This molecular information can then be used to correlate and
or a solid resin for biochemical isolation. Such approaches predict, using theoretical chemistry, their properties during the
should in principle be applicable to lipids. Indeed, palmitoylation refining process (chemical cracking). Mathematical modeling is
(Martin and Cravatt, 2009; Yount et al., 2010) and myristoylation also applicable to enzymatic lipid metabolism (Miskovic and
(Martin et al., 2008) of proteins can be successfully studied using Hatzimanikatis, 2010). Identification of lipid enzymes and their
such approaches. Alkyne-derivatized fatty acid incorporation cell biological and biochemical characterization will require
into cells, followed by solid-phase sequestration and release, additional tools, some of which can be taken from the current
is a promising new method for unequivocally monitoring indi- set that have proven successful in life sciences. New tools in
vidual glycerophospholipids (Milne et al., 2010). Bio-orthogonal bioinformatics are needed to address plant-specific pathways.
chemistry is not limited to the use of one reporter at a time. For For example, comparative deep sequencing of transcripts from
example, it can be combined with photoaffinity labeling. Such multiple plant tissues aided in the identification of an acyltrans-
strategies open new avenues for investigation of lipid-protein ferase that produces an unusual triacylglycerol in which one of
interactions (Gubbens and de Kroon, 2010) or asymmetry across the fatty acyls is an acetyl residue, rather than a fatty acid of
a lipid bilayer. Fluorophosphonate derivates of phosphatidylcho- C16 or C18 (Durrett et al., 2010). This particular lipid has

Cell 143, December 10, 2010 ª2010 Elsevier Inc. 893


desirable cold temperature properties, and thus this finding synthesizes unusual, reduced-viscosity oils in Euonymus and transgenic
might be readily translatable. seeds. Proc. Natl. Acad. Sci. USA 107, 9464–9469.
Ejsing, C.S., Sampaio, J.L., Surendranath, V., Duchoslav, E., Ekroos, K.,
Klemm, R.W., Simons, K., and Shevchenko, A. (2009). Global analysis of the
Concluding Remarks yeast lipidome by quantitative shotgun mass spectrometry. Proc. Natl.
Methods based on mass spectrometry are now available for Acad. Sci. USA 106, 2136–2141.
qualitative and quantitative analysis of many major lipids in Ernst, A.M., Contreras, F.X., Brügger, B., and Wieland, F. (2010). Determinants
complex samples (such as tissue and cell extracts) and from of specificity at the protein-lipid interface in membranes. FEBS Lett. 584,
several biological species (including yeast and mammals). The 1713–1720.
near future promises technical improvements stemming from Fahmi, S., Yang, C., Esmail, S., Hobbs, H.H., and Cohen, J.C. (2008).
cell isolation, sample fractionation and preparation, standardiza- Functional characterization of genetic variants in NPC1L1 supports the
sequencing extremes strategy to identify complex trait genes. Hum. Mol.
tion and cross-validation, and automation as well as wider
Genet. 17, 2101–2107.
coverage of biochemical lipidomics from integration with
Fahy, E., Subramaniam, S., Brown, H.A., Glass, C.K., Merrill, A.H., Jr., Murphy,
imaging, databases, and inclusion of additional biological
R.C., Raetz, C.R., Russell, D.W., Seyama, Y., Shaw, W., et al. (2005).
species. In parallel to these trends it can be anticipated that inter- A comprehensive classification system for lipids. J. Lipid Res. 46, 839–861.
disciplinary programs will continue to integrate biochemical Fahy, E., Cotter, D., Byrnes, R., Sud, M., Maer, A., Li, J., Nadeau, D., Zhau, Y.,
lipidomics with chemical biology, proteomics, and genomics to and Subramaniam, S. (2007). Bioinformatics for lipidomics. Methods Enzymol.
span the entire flow of information encoded in biological 432, 247–273.
systems. These efforts will provide us with a better under- Fairn, G.D., Ogata, K., Botelho, R.J., Stahl, P.D., Anderson, R.A., De Camilli, P.,
standing of natural variation found within lipids and will likely Meyer, T., Wodak, S., and Grinstein, S. (2009). An electrostatic switch
lead to customized applications in life sciences, industrial displaces phosphatidylinositol phosphate kinases from the membrane during
settings, and medicine. phagocytosis. J. Cell Biol. 187, 701–714.
Greenberg, M.E., Sun, M., Zhang, R., Febbraio, M., Silverstein, R., and Hazen,
S.L. (2006). Oxidized phosphatidylserine-CD36 interactions play an essential
ACKNOWLEDGMENTS
role in macrophage-dependent phagocytosis of apoptotic cells. J. Exp.
Med. 203, 2613–2625.
Work in our laboratories (http://www.lipidprofiles.com) is supported by
the National University of Singapore and by grants from the Singapore Groeger, A.L., Cipollina, C., Cole, M.P., Woodcock, S.R., Bonacci, G.,
National Research Foundation under CRP Award No. 2007-04, the Biomedical Rudolph, T.K., Rudolph, V., Freeman, B.A., and Schopfer, F.J. (2010).
Research Council of Singapore (R-183-000-211-305), the National Medical Cyclooxygenase-2 generates anti-inflammatory mediators from omega-3 fatty
Research Council (R-183-000-224-213), as well as the SystemsX.ch RTD acids. Nat. Chem. Biol. 6, 433–441.
project LipidX. Guan, X.L., Souza, C.M., Pichler, H., Dewhurst, G., Schaad, O., Kajiwara, K.,
Wakabayashi, H., Ivanova, T., Castillon, G.A., Piccolis, M., et al. (2009).
Functional interactions between sphingolipids and sterols in biological
REFERENCES
membranes regulating cell physiology. Mol. Biol. Cell 20, 2083–2095.

Alving, C.R. (2006). Antibodies to lipids and liposomes: immunology and Gubbens, J., and de Kroon, A.I. (2010). Proteome-wide detection of
safety. J. Liposome Res. 16, 157–166. phospholipid-protein interactions in mitochondria by photocrosslinking and
click chemistry. Mol. Biosyst. 6, 1751–1759.
Andreyev, A.Y., Shen, Z., Guan, Z., Ryan, A., Fahy, E., Subramaniam, S.,
Hakomori, S., and Zhang, Y. (1997). Glycosphingolipid antigens and cancer
Raetz, C.R., Briggs, S., and Dennis, E.A. (2010). Application of proteomic
therapy. Chem. Biol. 4, 97–104.
marker ensembles to subcellular organelle identification. Mol. Cell.
Proteomics 9, 388–402. Hamasur, B., Haile, M., Pawlowski, A., Schroder, U., Kallenius, G., and
Svenson, S.B. (2004). A mycobacterial lipoarabinomannan specific mono-
Assfalg, M., Bertini, I., Colangiuli, D., Luchinat, C., Schäfer, H., Schütz, B., and
clonal antibody and its F(ab’) fragment prolong survival of mice infected with
Spraul, M. (2008). Evidence of different metabolic phenotypes in humans.
Mycobacterium tuberculosis. Clin. Exp. Immunol. 138, 30–38.
Proc. Natl. Acad. Sci. USA 105, 1420–1424.
Han, X., and Gross, R.W. (1994). Electrospray ionization mass spectroscopic
Baker, C.J., Kanagasabai, R., Ang, W.T., Veeramani, A., Low, H.S., and Wenk,
analysis of human erythrocyte plasma membrane phospholipids. Proc. Natl.
M.R. (2008). Towards ontology-driven navigation of the lipid bibliosphere.
Acad. Sci. USA 91, 10635–10639.
BMC Bioinformatics 9 (Suppl 1), S5.
Hicks, A.A., Pramstaller, P.P., Johansson, A., Vitart, V., Rudan, I., Ugocsai, P.,
Becker, J.S., and Jakubowski, N. (2009). The synergy of elemental and
Aulchenko, Y., Franklin, C.S., Liebisch, G., Erdmann, J., et al. (2009). Genetic
biomolecular mass spectrometry: new analytical strategies in life sciences.
determinants of circulating sphingolipid concentrations in European popula-
Chem. Soc. Rev. 38, 1969–1983.
tions. PLoS Genet. 5, e1000672.
Blanksby, S.J., and Mitchell, T.W. (2010). Advances in mass spectrometry for
Illig, T., Gieger, C., Zhai, G., Römisch-Margl, W., Wang-Sattler, R., Prehn, C.,
lipidomics. Annu Rev Anal Chem (Palo Alto Calif) 3, 433–465.
Altmaier, E., Kastenmüller, G., Kato, B.S., Mewes, H.W., et al. (2010).
Brügger, B., Glass, B., Haberkant, P., Leibrecht, I., Wieland, F.T., and A genome-wide perspective of genetic variation in human metabolism. Nat.
Kräusslich, H.G. (2006). The HIV lipidome: a raft with an unusual composition. Genet. 42, 137–141.
Proc. Natl. Acad. Sci. USA 103, 2641–2646.
Kanter, J.L., Narayana, S., Ho, P.P., Catz, I., Warren, K.G., Sobel, R.A.,
Chan, R., Uchil, P.D., Jin, J., Shui, G., Ott, D.E., Mothes, W., and Wenk, M.R. Steinman, L., and Robinson, W.H. (2006). Lipid microarrays identify key
(2008). Retroviruses human immunodeficiency virus and murine leukemia virus mediators of autoimmune brain inflammation. Nat. Med. 12, 138–143.
are enriched in phosphoinositides. J. Virol. 82, 11228–11238. Kim, H.Y., Wang, T.C., and Ma, Y.C. (1994). Liquid chromatography/mass
Deigner, H.P., and Hermetter, A. (2008). Oxidized phospholipids: emerging spectrometry of phospholipids using electrospray ionization. Anal. Chem.
lipid mediators in pathophysiology. Curr. Opin. Lipidol. 19, 289–294. 66, 3977–3982.
Durrett, T.P., McClosky, D.D., Tumaney, A.W., Elzinga, D.A., Ohlrogge, J., and Kind, T., Wohlgemuth, G., Lee, Y., Lu, Y., Palazoglu, M., Shahbaz, S., and
Pollard, M. (2010). A distinct DGAT with sn-3 acetyltransferase activity that Fiehn, O. (2009). FiehnLib: mass spectral and retention index libraries for

894 Cell 143, December 10, 2010 ª2010 Elsevier Inc.


metabolomics based on quadrupole and time-of-flight gas chromatography/ by MALDI traveling-wave ion mobility-mass spectrometry. Anal. Chem. 82,
mass spectrometry. Anal. Chem. 81, 10038–10048. 1881–1889.
Klemm, R.W., Ejsing, C.S., Surma, M.A., Kaiser, H.J., Gerl, M.J., Sampaio, Rinaldi, S., Brennan, K.M., and Willison, H.J. (2010). Heteromeric glycolipid
J.L., de Robillard, Q., Ferguson, C., Proszynski, T.J., Shevchenko, A., and complexes as modulators of autoantibody and lectin binding. Prog. Lipid
Simons, K. (2009). Segregation of sphingolipids and sterols during formation Res. 49, 87–95.
of secretory vesicles at the trans-Golgi network. J. Cell Biol. 185, 601–612. Serhan, C.N. (2009). Systems approach to inflammation resolution: identifica-
Kurat, C.F., Wolinski, H., Petschnigg, J., Kaluarachchi, S., Andrews, B., Natter, tion of novel anti-inflammatory and pro-resolving mediators. J. Thromb.
K., and Kohlwein, S.D. (2009). Cdk1/Cdc28-dependent activation of the major Haemost. 7 (Suppl 1), 44–48.
triacylglycerol lipase Tgl4 in yeast links lipolysis to cell-cycle progression. Shevchenko, A., and Simons, K. (2010). Lipidomics: coming to grips with lipid
Mol. Cell 33, 53–63. diversity. Nat. Rev. Mol. Cell Biol. 11, 593–598.
Mahal, L.K., Yarema, K.J., and Bertozzi, C.R. (1997). Engineering chemical Singh, R., Kaushik, S., Wang, Y., Xiang, Y., Novak, I., Komatsu, M., Tanaka, K.,
reactivity on cell surfaces through oligosaccharide biosynthesis. Science Cuervo, A.M., and Czaja, M.J. (2009). Autophagy regulates lipid metabolism.
276, 1125–1128. Nature 458, 1131–1135.
Manford, A., Xia, T., Saxena, A.K., Stefan, C., Hu, F., Emr, S.D., and Mao, Y. Suh, B.C., Inoue, T., Meyer, T., and Hille, B. (2006). Rapid chemically induced
(2010). Crystal structure of the yeast Sac1: implications for its phosphoinosi- changes of PtdIns(4,5)P2 gate KCNQ ion channels. Science 314, 1454–1457.
tide phosphatase function. EMBO J. 29, 1489–1498.
Swartz, G.M., Jr., Gentry, M.K., Amende, L.M., Blanchette-Mackie, E.J., and
Marshall, A.G., and Rodgers, R.P. (2008). Petroleomics: chemistry of the Alving, C.R. (1988). Antibodies to cholesterol. Proc. Natl. Acad. Sci. USA 85,
underworld. Proc. Natl. Acad. Sci. USA 105, 18090–18095. 1902–1906.
Martin, B.R., and Cravatt, B.F. (2009). Large-scale profiling of protein palmi- Syed, S.A., Taylor, R.H., Crean, P.M., and Stewart, R.J. (1992). Successful use
toylation in mammalian cells. Nat. Methods 6, 135–138. of monoclonal anti-lipid-A IgM in infant with meningococcal sepsis. Lancet
Martin, D.D., Vilas, G.L., Prescher, J.A., Rajaiah, G., Falck, J.R., Bertozzi, C.R., 339, 496.
and Berthiaume, L.G. (2008). Rapid detection, discovery, and identification of Tanaka, T., Shen, J., Abecasis, G.R., Kisialiou, A., Ordovas, J.M., Guralnik,
post-translationally myristoylated proteins during apoptosis using a bio- J.M., Singleton, A., Bandinelli, S., Cherubini, A., Arnett, D., et al. (2009).
orthogonal azidomyristate analog. FASEB J. 22, 797–806. Genome-wide association study of plasma polyunsaturated fatty acids in
Milne, S.B., Tallman, K.A., Serwa, R., Rouzer, C.A., Armstrong, M.D., Marnett, the InCHIANTI Study. PLoS Genet. 5, e1000338.
L.J., Lukehart, C.M., Porter, N.A., and Brown, H.A. (2010). Capture and release Thomas, M.C., Mitchell, T.W., and Blanksby, S.J. (2009). OnLine ozonolysis
of alkyne-derivatized glycerophospholipids using cobalt chemistry. Nat. methods for the determination of double bond position in unsaturated lipids.
Chem. Biol. 6, 205–207. Methods Mol. Biol. 579, 413–441.
Minami, Y., Kasukawa, T., Kakazu, Y., Iigo, M., Sugimoto, M., Ikeda, S., Yasui, Tully, S.E., and Cravatt, B.F. (2010). Activity-based probes that target
A., van der Horst, G.T., Soga, T., and Ueda, H.R. (2009). Measurement of functional subclasses of phospholipases in proteomes. J. Am. Chem. Soc.
internal body time by blood metabolomics. Proc. Natl. Acad. Sci. USA 106, 132, 3264–3265.
9890–9895.
van Meer, G. (2005). Cellular lipidomics. EMBO J. 24, 3159–3165.
Miskovic, L., and Hatzimanikatis, V. (2010). Production of biofuels and Volkmer, A. (2005). Vibrational imaging and microspectroscopies based on
biochemicals: in need of an ORACLE. Trends Biotechnol. 28, 391–397. coherent anti-Stokes Raman scattering microscopy. J. Phys. D Appl. Phys.
Müller, M., and Zumbusch, A. (2007). Coherent anti-stokes raman scattering 38, R59–R81.
microscopy. ChemPhysChem 8, 2156–2170. Wenk, M.R. (2005). The emerging field of lipidomics. Nat. Rev. Drug Discov. 4,
Murphy, R.C., Hankin, J.A., and Barkley, R.M. (2009). Imaging of lipid species 594–610.
by MALDI mass spectrometry. J. Lipid Res. Suppl. 50, S317–S322. Wheelock, C.E., Goto, S., Yetukuri, L., D’Alexandri, F.L., Klukas, C., Schreiber,
Nomura, D.K., Long, J.Z., Niessen, S., Hoover, H.S., Ng, S.W., and Cravatt, F., and Oresic, M. (2009). Bioinformatics strategies for the analysis of lipids.
B.F. (2010). Monoacylglycerol lipase regulates a fatty acid network that Methods Mol. Biol. 580, 339–368.
promotes cancer pathogenesis. Cell 140, 49–61. Wymann, M.P., and Schneiter, R. (2008). Lipid signalling in disease. Nat. Rev.
Patwardhan, A.M., Akopian, A.N., Ruparel, N.B., Diogenes, A., Weintraub, Mol. Cell Biol. 9, 162–176.
S.T., Uhlson, C., Murphy, R.C., and Hargreaves, K.M. (2010). Heat generates Yang, K., Zhao, Z., Gross, R.W., and Han, X. (2007). Shotgun lipidomics
oxidized linoleic acid metabolites that activate TRPV1 and produce pain in identifies a paired rule for the presence of isomeric ether phospholipid
rodents. J. Clin. Invest. 120, 1617–1626. molecular species. PLoS ONE 2, e1368.
Pemble, C.W., 4th, Johnson, L.C., Kridel, S.J., and Lowther, W.T. (2007). Yang, P.Y., Liu, K., Ngai, M.H., Lear, M.J., Wenk, M.R., and Yao, S.Q. (2010).
Crystal structure of the thioesterase domain of human fatty acid synthase Activity-based proteome profiling of potential cellular targets of Orlistat—
inhibited by Orlistat. Nat. Struct. Mol. Biol. 14, 704–709. an FDA-approved drug with anti-tumor activities. J. Am. Chem. Soc. 132,
Postle, A.D., and Hunt, A.N. (2009). Dynamic lipidomics with stable isotope 656–666.
labelling. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 877, 2716–2721. Yount, J.S., Moltedo, B., Yang, Y.Y., Charron, G., Moran, T.M., López, C.B.,
Quehenberger, O., Armando, A.M., Brown, A.H., Milne, S.B., Myers, D.S., and Hang, H.C. (2010). Palmitoylome profiling reveals S-palmitoylation-
Merrill, A.H., Bandyopadhyay, S., Jones, K.N., Kelly, S., Shaner, R.L., et al. dependent antiviral activity of IFITM3. Nat. Chem. Biol. 6, 610–614.
(2010). Lipidomics reveals a remarkable diversity of lipids in human plasma. Zemski Berry, K.A., Turner, W.W., VanNieuwenhze, M.S., and Murphy, R.C.
J. Lipid Res. 51, 3299–3305. (2010). Characterization of oxidized phosphatidylethanolamine derived
Ridenour, W.B., Kliman, M., McLean, J.A., and Caprioli, R.M. (2010). Structural from RAW 264.7 cells using 4-(dimethylamino)benzoic acid derivatives.
characterization of phospholipids and peptides directly from tissue sections Eur. J. Mass Spectrom. (Chichester, Eng.) 16, 463–470.

Cell 143, December 10, 2010 ª2010 Elsevier Inc. 895


Inositol Pyrophosphates Inhibit
Akt Signaling, Thereby Regulating
Insulin Sensitivity and Weight Gain
Anutosh Chakraborty,1 Michael A. Koldobskiy,1 Nicholas T. Bello,2 Micah Maxwell,1 James J. Potter,3
Krishna R. Juluri,1 David Maag,1 Seyun Kim,1 Alex S. Huang,1 Megan J. Dailey,2 Masoumeh Saleh,1
Adele M. Snowman,1 Timothy H. Moran,2 Esteban Mezey,3 and Solomon H. Snyder1,2,4,*
1The Solomon H. Snyder Department of Neuroscience
2Department of Psychiatry and Behavioral Sciences
3Department of Medicine
4Department of Pharmacology and Molecular Sciences

Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA


*Correspondence: ssnyder@jhmi.edu
DOI 10.1016/j.cell.2010.11.032

SUMMARY and telomere length (Saiardi et al., 2005; York et al., 2005)
maintenance. Another isoform of IP7, identified as 1/3-PP-IP5,
The inositol pyrophosphate IP7 (5-diphosphoinosi- is formed by the Vip1 enzyme (Lin et al., 2009; Mulugu et al.,
tolpentakisphosphate), formed by a family of three 2007) and in yeast influences cell shape, growth, and phosphate
inositol hexakisphosphate kinases (IP6Ks), modu- disposition (Lee et al., 2007).
lates diverse cellular activities. We now report that IP6K1 depletion by RNA interference impairs insulin secretion
IP7 is a physiologic inhibitor of Akt, a serine/threo- by pancreatic b cells (Illies et al., 2007), and IP6K1 KO mice
manifest reduced circulating insulin levels (Bhandari et al.,
nine kinase that regulates glucose homeostasis and
2008). Despite low serum insulin, IP6K1-deleted (IP6K1 KO)
protein translation, respectively, via the GSK3b and
mice display normal blood glucose levels and tolerance,
mTOR pathways. Thus, Akt and mTOR signaling implying insulin hypersensitivity (Bhandari et al., 2008).
are dramatically augmented and GSK3b signaling IP7 can signal by physiologically pyrophosphorylating protein
reduced in skeletal muscle, white adipose tissue, targets (Bhandari et al., 2007; Saiardi et al., 2004). In yeast,
and liver of mice with targeted deletion of IP6K1. 1/3-PP-IP5 binds the cyclin-cdk complex to regulate phosphate
IP7 affects this pathway by potently inhibiting the metabolism (Lee et al., 2007).
PDK1 phosphorylation of Akt, preventing its activa- Pleckstrin homology domains (PH domains) (Lemmon, 2008)
tion and thereby affecting insulin signaling. IP6K1 bind phospholipids such as phosphatidylinositol(3,4,5)-trisphos-
knockout mice manifest insulin sensitivity and are phate (PIP3) and phosphatidylinositol (4,5)-bisphosphate (PIP2)
resistant to obesity elicited by high-fat diet or aging. (Di Paolo and De Camilli, 2006; Fruman et al., 1999), thereby
recruiting signaling proteins to membranes. IP7 interferes with
Inhibition of IP6K1 may afford a therapeutic ap-
the binding of PIP3 to the PH domain of the Dictyostelium-
proach to obesity and diabetes.
specific cytosolic regulator of adenylyl cyclase (CRAC) to inhibit
chemotaxis (Luo et al., 2003).
INTRODUCTION Akt (PKB), a PH domain containing serine/threonine kinase,
regulates growth factor signaling (Chan et al., 1999; Cho et al.,
Inositol phosphates are widely distributed in animal and plant 2001; Taniguchi et al., 2006) to stimulate glucose uptake (Welsh
tissues. Most studied is inositol 1,4,5-trisphosphate (IP3), which et al., 2005), glycogen synthesis (Cross et al., 1995), and protein
releases calcium from intracellular stores (Berridge et al., 2000; synthesis (Memmott and Dennis, 2009; Ruggero and Sonen-
Irvine and Schell, 2001). More recently, higher inositol phos- berg, 2005) by influencing glucose transporter 4 (GLUT4),
phates with energetic pyrophosphate bonds have been de- glycogen synthase kinase 3 (GSK3)a/b, and tuberous sclerosis
scribed (Shears, 2007), which are synthesized by a family of complex 2 (TSC2)-mTOR signaling pathways.
three IP6 kinases (IP6Ks) (Saiardi et al., 1999; Saiardi et al., Increased protein translation following Akt activation elicits
2001). Best characterized is diphosphoinositol pentakisphos- skeletal muscle hypertrophy (Rommel et al., 2001) and augments
phate (5-PP-[1,2,3,4,6]IP5), here designated IP7 (Barker et al., hepatic fatty acid oxidation with reduced fat accumulation (Izu-
2009). In mammals, IP7 modulates numerous physiologic func- miya et al., 2008). GSK3b, which influences insulin resistance,
tions, including apoptosis (Chakraborty et al., 2008; Koldobskiy is phosphorylated and inhibited by Akt (Cross et al., 1995). Akt
et al., 2010) and insulin secretion (Illies et al., 2007), whereas, and GSK3b activity are reciprocally regulated in insulin resis-
in budding yeast, it influences endocytosis (Saiardi et al., 2002) tance and obesity. Akt/mTOR activity is decreased (Funai

Cell 143, 897–910, December 10, 2010 ª2010 Elsevier Inc. 897
Figure 1. Growth Factor-Induced IP7 Regulates Akt Activity
(A) IGF-1 treatment enhances intracellular IP7 levels in WT, but not in IP6K1 KO, MEFs.
(B) IP6K1 KO MEFs exhibit increased phosphorylation of Akt and Akt/mTOR downstream targets GSK3b, TSC2 S6K1, and S6 after 15 min IGF-1 treatment.
Tyrosine phosphorylation of IGF-1-induced upstream PI3 kinase activator IRS1 and PDK1 target PKCz is unchanged.
(C) Densitometric analysis displays 3-fold and 1.75-fold enhancement, respectively, in T308 and S473 Akt phosphorylation of IP6K1 KO MEFs following IGF-1
treatment.

898 Cell 143, 897–910, December 10, 2010 ª2010 Elsevier Inc.
et al., 2006; Shao et al., 2000) and GSK3b increased (Kaidano- (Alessi et al., 1997). We evaluated the formation of PIP3 in WT
vich and Eldar-Finkelman, 2002) in insulin-resistant tissues of and IP6K1 KO MEFs (Figure 1D). Serum deprivation of WT
aging and obese mice. MEFs markedly decreases PIP3 formation, which is reversed
The apparent insulin sensitivity of the IP6K1 KO mice promp- by treatment with IGF-1. The effects of serum deprivation
ted our interest in IP7 regulation of Akt and insulin signaling. and IGF-1 treatment are the same in IP6K1-deleted as in WT
We now show that IP7 is a physiologic inhibitor of Akt signaling, MEFs. We also measured PI3 kinase catalytic activity and tyro-
acting at the enzyme’s PH domain to block phosphorylation and sine phosphorylation status of its p85 subunit, which are
activation by PDK1. Thus, IP6K1 KO mice display a very marked unaltered in IP6K1 KO MEFs following IGF-1 treatment (Figures
enhancement of Akt activity accompanied by augmented insulin S1E and S1F). Basal PI3 kinase activity in WT and IP6K1
sensitivity and resistance to weight gain. KO MEFs is also unaltered (data not shown). Thus, IP6K1 regu-
lation of Akt is not due to alteration of PI3 kinase activity or
RESULTS PIP3 levels.
To examine insulin signaling in IP6K1 KO liver, we isolated
Growth Factor-Induced IP7 Formation Inhibits Akt primary hepatocytes, which display 60% reduction in IP7,
Signaling with unaltered levels of IP6 relative to WT hepatocytes (Figure 1E
We monitored IP7 formation of serum-starved MEFs in response and Figures S1G and S1H). IP6K1 KO hepatocytes manifest
to IGF-1 (Figure 1A and Figure S1A available online). In WT elevated phosphorylation of Akt, GSK3b, and S6 in response
MEFs, serum starvation decreases IP7 formation more than to insulin, with no alteration in p-PKCz/p-PKCD, other targets
90%, whereas IGF-1 rapidly restores IP7 levels with complete of PDK1 (Figures 1F and 1G).
restoration to WT values by 60 min. The stimulation of IP7 forma- Complementation of IP6K1-WT, but not IP6K1-K/A (kinase
tion by IGF-1 is abolished in IP6K1-deleted MEFs. In WT MEFs, dead), restores physiological IP7 levels in IP6K1 KO MEFs (Fig-
serum deprivation reduces levels of IP6 much less than IP7, and ure 1H and Figures S1I and S1J). Levels of p-Akt (T308/S473)
IGF-1 enhances formation of IP6 much less than IP7 (Figure S1B). and p-GSK3b are diminished in IGF-1-stimulated MEFs
In hepatocellular carcinoma cell line HEPG2, insulin or IGF-1 expressing IP6K1-WT, but not in IP6K1-K/A clone (Figures 1I
treatment similarly stimulates IP7 formation (Figure S1C). and 1J). Growth factor signaling is inhibited by S6K1 via phos-
Akt is activated by phosphorylation at T308 by PDK1 and at phorylation of IRS1 at S636/639 residues (Um et al., 2004). We
S473 by mTOR (Alessi et al., 1997; Sarbassov et al., 2005). do not observe any change in phosphorylation status of IRS1
IP6K1 KO MEFs display markedly augmented IGF-1-stimulated at S636/639 or at tyrosine residues (Figure 1I). We observe
phosphorylation of Akt (T308/S473) (Figures 1B and 1C) without similar effects in complemented MEFs induced with serum (Fig-
any alteration in tyrosine phosphorylation of insulin receptor ure S1K). IP6K1-WT overexpression lowers Akt and GSK3b
substrate 1 (IRS-1), an upstream activator of PI3 kinase. We phosphorylation levels in IGF-1-stimulated HEK293 cells (Fig-
also observe increased phosphorylation of Akt downstream ures 1K and 1L).
effectors GSK3b (S9), TSC2 (T1462), S6K1 (T389), and S6 The enhancement in Akt/mTOR signaling is accompanied by
(S235/S236) in response to IGF-1 (Figure 1B). We detect similarly parallel changes in protein synthesis. Thus, IP6K1 KO MEFs
increased growth factor-mediated signaling in a separate clone manifest a 15% increase in protein translation (Figure S1L).
of IP6K1 KO MEFs (Figure S1D). To assess specificity, we moni- Wortmannin and rapamycin each reduce wild-type protein trans-
tored an atypical PKC, PKCz, which is a PH domain-deficient lation about 20%–25%, consistent with the Akt-mTOR pathway
PDK1 target (Figure 1B). PKCz phosphorylation levels are the accounting for only about 20%–25% of total protein synthesis
same in IP6K1-deleted and WT MEFs in the absence or presence (Holz et al., 2005). The increased protein translation of IP6K1
of IGF-1. Phosphorylation of the growth factor-stimulated kinase KO MEFs is reduced by about 25% following overexpression
ERK and the PDK1 target PKCD are also unchanged (Fig- of IP6K1-WT, but not IP6K1-K/A (Figure S1M). To ascertain
ure S1D). Akt can be activated via a variety of mechanisms, whether IP6K1 regulates Akt/mTOR activation in intact organ-
especially those involving PI3 kinase and its generation of PIP3 isms, we monitored phosphorylation of ribosomal protein S6 in

(D) Increased activation in IP6K1 KO MEFs is not due to elevated PI3 kinase signaling. Intracellular PIP3 levels are similar in WT and IP6K1 KO MEFs under basal
and after 15 min IGF-1 treatment.
(E) IP6K1 is a primary source of IP7 synthesis in the liver. Primary hepatocytes isolated from 10-month-old IP6K1 KO mice display 60% reduction in the IP7
levels.
(F) Primary hepatocytes of 10-month-old IP6K1 KO mice after insulin treatment manifest enhanced phosphorylation of Akt, GSK3b, and S6, with unaltered
phosphorylation status of PDK1 targets PKCz and PKCD.
(G) Densitometry reveals 5-fold and 2-fold enhancement, respectively, in T308 and S473 phosphorylation levels of Akt in IP6K1 KO hepatocytes following
insulin treatment for 30 min.
(H) Complementation of IP6K1-WT, but not IP6K1-K/A, restores physiological levels of IP7 in the IP6K1 KO MEFs.
(I) Complementation of IP6K1 KO MEFs with IP6K1-WT reduces phosphorylation of Akt and GSK3b, with IP6K1-K/A having no effect. IGF-1-dependent tyrosine
and S636/639 phosphorylation of upstream PI3 kinase activator IRS1 are unaltered.
(J) IP6K1-WT complementation elicits 3-fold reduction in IGF-1-induced T308 and S473 Akt phosphorylation. IP6K1-K/A does not have any effect.
(K) Transient Myc-IP6K1 overexpression elicits decrease in IGF-1-dependent Akt and GSK3b phosphorylation in HEK293 cells.
(L) Overexpression of IP6K1-WT reduces IGF-1-induced phosphorylation of T308 and S473 Akt to 3-fold, whereas IP6K1-K/A has much less effect.
Each experiment was repeated at least three times. ***p < 0.001; **p < 0.01; and *p < 0.05). See also Figure S1.

Cell 143, 897–910, December 10, 2010 ª2010 Elsevier Inc. 899
A B C
WT KO IP6K1
Total p-T308 Merged 0 5 15 30 0 5 15 30 IGF-1
WT - IGF1 Akt
p-T308 Akt

Caveolin

Membrane
KO - IGF1
Akt
p-T308 Akt

LDH

WT + IGF1 Cytosol
MEF

F
KO + IGF1
0 0 0.01 0.25 0.5 1 5 IP6/IP7 (μM)
- + + + + + + PIP3 (1 μM)
+ + + + + + + PDK1

Endogenous Akt MEF IP7

IP6
D E WB: p-T308 Akt ab

PDK1 activity on recombinant


- WT K/A - WT K/A - WT K/A IP6K1
purified Akt in vitro
- - - 5 5 5 15 15 15 IGF-1
Akt G
Cadherin
Membrane
Akt
Myc-IP6K1

βTubulin
Total extract
IP6K1 KO MEF

H I J - + + + PDK1
0 0 0.01 0.05 0.1 0.25 0.5 1 IP7 (μM) - - IP4 IP7 (1 μM)
p-T308 Akt - - - - Serum

PDK1 activity on purified p-T308 Akt


Akt in vitro
V5 Akt

IP: Akt HEK 293

Figure 2. IP7 Inhibits Akt T308 Phosphorylation and Membrane Translocation


(A) Immunofluorescence analysis of IGF-1-induced T308 phosphorylation and membrane translocation of Akt in absence of IP6K1. IGF-1-treated IP6K1 KO MEFs
display enhanced T308 phosphorylation of Akt and augmented membrane translocation. Green and red represent total and p-T308 Akt, respectively, whereas
yellow is the merged color for total and p-T308 Akt.
(B–D) Western blot analysis demonstrates increased T308 phosphorylation and membrane localization of Akt in IP6K1 KO MEFs after IGF-1 treatment. We also
observe an increase in cytosolic p-T308 Akt levels in the IP6K1 KO MEFs.
(E) Complementation of IP6K1 KO MEFs with IP6K1-WT causes a delay in Akt translocation to the plasma membrane, whereas IP6K1-K/A does not show this effect.

900 Cell 143, 897–910, December 10, 2010 ª2010 Elsevier Inc.
the gastrocnemius muscle and liver of IP6K1 mutant mice and In the absence of added PIP3, IP7 is substantially more potent,
observed a pronounced enhancement (Figure S1N). IP6K1 dele- inhibiting PDK phosphorylation of Akt with an IC50 of about 20 nM
tion leads to decreased 4EBP1 binding to eIF4E (Holz et al., (Figures 2H and 2I). Phosphorylation of overexpressed Akt
2005) at the mRNA cap in insulin-treated mice liver and gastroc- immunoprecipitated from serum-starved HEK293 cells by PDK1
nemius muscle (Figure S1O). in vitro is abolished by 1 mM IP7, with IP4 having no effect (Fig-
In summary, growth factor stimulation enhances IP7 forma- ure 2J). The inhibitory action of IP7 is selective, with IP5 and IP6
tion, which in turn inhibits Akt signaling. Accordingly, marked exerting much less inhibition and IP3 and IP4 inactive (Figure S2D).
augmentation of Akt signaling is seen in IP6K1-deleted tissues. Because of the competition between IP7 and PIP3 for PH
domain binding (Luo et al., 2003), we presume that the inhibitory
IP7 Inhibits Akt T308 Phosphorylation effect of IP7 on Akt phosphorylation is primarily exerted via the
and Membrane Translocation PH domain. IP7 fails to inhibit PDK phosphorylation of Akt lack-
In response to growth factors, PIP3 stimulates Akt at the ing its PH domain (Figure S2E). IP7 at 1 mM concentration does
membrane by facilitating its phosphorylation by PDK1 (Alessi not inhibit S6K1 catalytic activity on peptide substrates in vitro
et al., 1997). We monitored IGF-1-dependent membrane translo- (data not shown). IP7 binds to PDK1 (data not shown) but does
cation of Akt in MEFs of WT and IP6K1 KO mice (Figures 2A and not affect its catalytic activity on artificial peptide substrates,
2B). We observe increased membrane localization of total Akt indicating that IP7 does not inhibit PDK1 activity in general (Fig-
and p-T308 Akt following IGF-1 treatment in IP6K1-deleted ure S2F), consistent with an earlier report (Komander et al.,
MEFs (Figure 2A). Membrane levels of Akt protein are markedly 2004). The PH domain of PDK1 occurs in the enzyme’s C
enhanced by IGF-1 in WT preparations, with the enhancement terminus and does not influence its catalytic activity.
increased in IP6K1 KO cells (Figures 2B and 2C). Membrane- We presume that IP7 regulates Akt by binding directly to its PH
associated p-T308 Akt is also strikingly increased in IP6K1 KO domain. Previously, we demonstrated that IP7 potently and
preparation, with some cytosolic increase as well, presumably selectively competes with PIP3 for binding to the PH domain of
reflecting movement of phosphorylated Akt from membrane to Akt, as IP6 failed to inhibit binding except at very high concentra-
cytosol (Figure 2B). Complementation of IP6K1-WT markedly tions (Luo et al., 2003). In the present study, [3H]IP7 binds to full-
reduces IGF-1-elicited membrane translocation of Akt. Vector length Akt, with binding drastically reduced for Akt lacking the
alone or kinase dead IP6K1 (IP6K1-K/A) does not reduce PH domain (Figure S2G). IP7 does not affect mTORC2 activity
membrane Akt (Figure 2E). toward Akt-S473 in vitro (Figures S2H and S2I).
The IP6K inhibitor TNP (10 mM) (Padmanabhan et al., 2009)
increases the IGF-1-elicited stimulation of T308 phosphorylation IP6K1-Deleted Mice Display Sustained Insulin
of Akt without influencing p-S473. (Figure S2A). The increased Sensitivity
Akt signaling elicited by TNP is not evident in IP6K1 null cells Six-week-old IP6K1 KO mice displayed reduced blood levels of
(Figure S2B). TNP increases T308 Akt phosphorylation in both insulin, with normal plasma glucose implying insulin hypersensi-
membrane and cytosol fractions (Figure S2C). tivity (Bhandari et al., 2008). Age-induced insulin resistance is
PDK1-mediated phosphorylation of Akt is dramatically associated with decreased Akt activity (Funai et al., 2006; Shay
increased by PIP3 binding to Akt’s PH domain via presumed and Hagen, 2009). Accordingly, we explored insulin sensitivity
conformational alterations (Calleja et al., 2007). We examined in terms of blood glucose levels in 10-month-old mice (Figure 3A
the influence of IP7 or IP6 upon PDK1-elicited phosphorylation and 3B). These mice display significantly improved glucose
of Akt in the presence of PIP3 in vitro (Figures 2F and 2G). IP7 tolerance following glucose infusion (Figure 3A). Following insulin
inhibits phosphorylation of Akt at T308 about 50% at 1 mM, administration, the IP6K1 KO mice display significantly lower
whereas IP6 does not. Of interest, the IC50 for IP7 inhibition blood levels of glucose than do WT mice (Figure 3B), establishing
resembles the PIP3 concentration required for maximal activa- that older IP6K1 knockouts are indeed hypersensitive to insulin.
tion. We observe the inhibitory effect only when IP7 and Akt Increased insulin sensitivity should be associated with im-
are preincubated together at the same time. When PIP3 is prein- proved glucose uptake from plasma. To evaluate glucose utiliza-
cubated with Akt prior to the addition of IP7, IP7’s IC50 increases tion, we employed hyperinsulinemic-euglycemic clamp studies
to 50 mM (data not shown), beyond its physiological range. This (Figure 3C). The insulin sensitivity of the IP6K1 KO is more than
observation also fits with the prior reports that IP7 failed to double that of WT mice. We monitored the uptake of glucose
release Akt prebound to PIP3 (Downes et al., 2005). Myristoyla- into muscle and fat tissue in the clamp experiments (Figure 3D).
tion anchors Akt to the plasma membrane and irreversibly In gastrocnemius muscle and epididymal white adipose tissues
 et al., 1997). Thus, IP6K1-WT overex-
activates it (Andjelkovic (EWAT), glucose uptake is approximately tripled in the mutant
pression in HEK293 cells reduces T308 phosphorylation of mice. We do not observe any significant change in liver glucose
WT-Akt, but not of myristoylated Akt, upon growth factor stimu- uptake (data not shown), presumably because uptake is largely
lation (data not shown). mediated by GLUT4 in muscle and adipose tissue.

(F and G) PIP3-induced (1 mM) Akt-T308 phosphorylation is inhibited by IP7, with an IC50 of 1 mM in vitro.
(H and I) IP7 inhibits PDK1-dependent Akt phosphorylation at T308 in vitro, with an IC50 value of 20 nM.
(J) IP7 inhibition of PDK1-dependent phosphorylation of overexpressed Akt immunoprecipitated from serum-starved HEK293 cells. PDK1 increases Akt phos-
phorylation in vitro, which is abolished by IP7. IP4 does not have any significant effect.
Each experiment was repeated at least three times. ***p < 0.001; **p < 0.01; and *p < 0.05. See also Figure S2.

Cell 143, 897–910, December 10, 2010 ª2010 Elsevier Inc. 901
Figure 3. IP6K1 KO Mice Manifest Sustained Insulin Sensitivity
(A) Glucose tolerance test (GTT). IP6K1 KO mice display improved glucose tolerance than WT (male, n = 5, each set).
(B) Insulin tolerance test (ITT). In response to insulin, IP6K1 KO mice display a greater glucose removal rate than WT littermates (male, n = 5, each set).
(C) Hyperinsulinemic-euglycemic clamp studies. Glucose infusion rates (GIR) display 3-fold increase in IP6K1 KO mice than WT littermates (male, n = 4,
each set).
(D) Glucose uptake in gastrocnemius muscle and in epididymal white adipose tissue (WAT) is significantly enhanced in IP6K1 KO mice (male, n = 4, each set).
(E) Acute insulin sensitivity in IP6K1 KO mice. Insulin treatment causes enhanced p-Akt and p-GSK3b levels downstream of IRS-1 phosphorylation in the
gastrocnemius muscles of IP6K1 KO mice.
(F) Acute insulin treatment leads to 2-, 2.5-, and 4-fold increase in phosphorylation status of T308, S473 of Akt, and S9 of GSKb, respectively.
(G) Increased glycogen content in gastrocnemius muscle of IP6K1 KO mice after 30 min insulin treatment of 16 hr fasted mice (n = 3, each set).
(H) IP7 levels in young and old hepatocytes. IP7 levels increase significantly with age in the WT mice (n = 3, each set).
***p < 0.001; **p < 0.01; and *p < 0.05. See also Figure S3.

We monitored Akt signaling in response to acute insulin treat- epididymal white adipose tissue (EWAT) of IP6K1 KO mice (Fig-
ment (Figures 3E and 3F). In gastrocnemius muscle, levels of ure S3A). We detect enhancement in insulin-mediated glycogen
p-Akt (T308/S473) are markedly increased in IP6K1 KO mice, formation in the gastrocnemius muscle of IP6K1 KO mice
as are levels of the Akt downstream target p-GSK3b. On the (Figure 3G).
other hand, insulin receptor substrate (IRS1) phosphorylation is To explore relationships between age-dependent Akt activity
similar in KO and WT mice, indicating that the insulin sensitivity and IP7 levels, we measured inositol phosphates in 2- and
is due to regulation of Akt/GSK3b downstream of IRS1. We do 10-month-old mice (Figure 3H and Figures S3B and S3C).
not observe any alteration in S6K1-mediated inhibitory phos- Both IP6 and IP7 levels are elevated in the older mice, with
phorylation of S636/S639 IRS1 under these conditions (data greater augmentation in IP7, resulting in increased IP7/IP6 ratios.
not shown). Increased insulin sensitivity is also observed in The knockout hepatocyte preparations display an enhancement

902 Cell 143, 897–910, December 10, 2010 ª2010 Elsevier Inc.
in age-dependent increase in p-T308 Akt, suggesting that impaired glucose tolerance. Insulin tolerance tests (ITT) reveal
increases in IP7 levels with age interfere with Akt activation greater insulin-induced reductions of blood glucose in KO mice
(Figures S3D and S3E). on HFD, with no difference on regular diet (Figure 5D). Serum
In summary, in WT animals, age-dependent increases in IP7 insulin levels are significantly lower in IP6K1 KOs on regular
formation accompany decreased insulin sensitivity, which may diet (Bhandari et al., 2008), which is even more striking after
explain the increased insulin sensitivity in aged IP6K1 KO mice. high-fat exposure when the WT insulin levels reach pathologic
levels (Figure 5E). Under the same experimental conditions
IP6K1 KO Mice Are Resistant to Obesity described in Figure 5E, we measured Akt signaling in 4 hr fasted
IP6K1 knockout mice exhibit reduced body weight (Bhandari mice (Figure 5F). HFD elicits higher levels of phosphorylated Akt,
et al., 2008), which is more prominent with age (Figure 4A). The GSK3b, and S6 in IP6K1 KO mice than in WT. The mutant mice
reduced body weight primarily reflects reduced fat accumulation display similar insulin levels as WT mice on CD. Despite high
with decreased weight of epididymal adipose tissue (EWAT) insulin levels, WT mice on HFD do not exhibit increased Akt
(Figure 4B) as well as diminished weights of other visceral and phosphorylation, consistent with insulin resistance. IP6K1 KO
subcutaneous fat (data not shown). Despite lower body weight, mice are protected from HFD-induced insulin resistance. Thus,
IP6K1 KO mice display increased gastrocnemius muscle mass IP6K1 KO mice do not display the HFD-induced insulin resis-
(Figure S4A). These findings may be consistent with earlier tance associated with reductions in Akt signaling.
observations (Izumiya et al., 2008) that increased Akt signaling
leads to muscle hypertrophy, enhanced insulin sensitivity, and IP7 Reduces Fat Breakdown and Enhances
resistance to HFD-induced weight gain. Adipogenesis
We examined body weight of IP6K1 KO mice under high-fat Besides altering insulin sensitivity, Akt and its downstream effec-
diet (HFD) conditions. Six-week-old IP6K1 KO mice on control tors can reduce fat accumulation by: (1) diminishing food intake
diet (CD) are slightly smaller than WT littermates (Figures 4C, via mTOR (Cota et al., 2006), (2) increasing fat utilization or oxida-
4E, and 4F, orange and brown circles). However, when exposed tion via Akt (Izumiya et al., 2008), and (3) reducing adipogenesis
to HFD, they display striking resistance to body weight gain via GSK3b (Ross et al., 2000).
(Figures 4D–4F, light and dark green triangles), with less than Food intake of IP6K1 KOs does not differ from WT on control
one-third of WT weight gain. WT mice on HFD display a 300% diet (Bhandari et al., 2008) or HFD (Figure 6A). WT mice on HFD
greater increase in body fat than IP6K1 KO mice (Figures 4G exhibit reduced oxygen consumption (VO2) and carbon dioxide
and 4H and Figure S4B), as assessed by Echo-MRI analysis. release (VCO2) (Figures 6B and 6C). We assessed energy expen-
With or without HFD, IP6K1 KO mice display a markedly lower diture (EE) based on both fat and lean body mass, as fat mass
weight of diverse fat pads with unchanged brown fat (BAT) also alters energy expenditure (Kaiyala et al., 2010). WT on
weight on control diet (Figure 4I). HFD display reduced EE, presumably reflecting locomotor hypo-
Serum leptin levels are markedly lower in KO mice on CD or activity, similar to adipose tissue-specific PPARg knockout mice
HFD (Figure 4J), consistent with their reduced fat mass and (Jones et al., 2005; Tou and Wade, 2002) (Figure 6D). IP6K1 KO
indicating increased leptin sensitivity (Myers et al., 2008). mice on HFD are protected from reductions in VO2, VCO2, and
The liver is the major organ responsible for metabolizing fat to energy expenditure, resulting in an increase in energy expendi-
generate energy. Aberrations in the process lead to fatty liver ture in the knockouts (Figure 6D). Respiratory quotient (RQ),
disease or hepatic steatosis (Reddy and Rao, 2006). IP6K1 KO a reflection of carbohydrate and fat consumption, is decreased
mice display resistance to high-fat diet-induced weight gain in to a similar extent in WT and IP6K1 KO mice (Figure 6E).
the liver (Figure 4K). Lipid droplets visible in the WT liver on Increased fat oxidation in IP6K1 KO mice is confirmed by
control or high-fat diet are absent in IP6K1 KO mice (Figure 4L switching mice from high-fat to control diet. The change in diet
and Figure S4C). Thus, in the IP6K1 KO mice, resistance to elicits decreased body weight to a much greater extent in
weight gain is due to reduced fat accumulation. High-fat diets IP6K1 mutants than in WT mice (Figures 6F and 6G). Plasma
cause increases in serum triglycerides, cholesterols, aspartate ketone concentrations, which reflect hepatic fat oxidation, are
aminotransferase (AST), and lactate dehydrogenase (LDH) significantly increased in IP6K1 KO mice on both control and
(Hoffler et al., 2009; Ito et al., 2008). These substances are signif- high-fat diet (data not shown).
icantly lower in IP6K1 KO than WT mice (Figures S4D–S4G). During adipogenic differentiation of NIH 3T3-L1 cells, IP7
levels rise and are substantially reduced by the IP6K inhibitor
IP6K1 Deletion Improves Glucose Homeostasis TNP (Figure 6H and Figure S6A). IP6 levels are increased much
in High-Fat Diet-Fed Mice Associated less and are unaffected by TNP (Figure S6B). GSK3b, inhibited
with Increased Akt Signaling by Akt, inhibits adipogenesis (Ross et al., 2000). The GSK3b
HFD-induced weight gain impairs insulin sensitivity and glucose inhibitor SB21676 inhibits differentiation of NIH 3T3-L1 cells
homeostasis (Kahn et al., 2006), whereas mice with insulin (Tang et al., 2005). We monitored differentiation of 3T3-L1
hypersensitivity resist the sequelae of HFD (Elchebly et al., preadipocytes in the presence of IP6K and GSK3b inhibitors
1999; Izumiya et al., 2008). After 8 weeks on HFD, IP6K1 KO (Figures 6I and 6J). SB216763 completely blocks 3T3-L1
mice do not display the hyperglycemia evident in WT mice differentiation at 10 mM, whereas 1 mM drug elicits minimal
(Figures 5A and 5B). HFD in WT mice leads to prolonged eleva- effects. TNP (10 mM) inhibits differentiation 20%–25%. The
tions in blood glucose levels following a glucose injection (Fig- combination of TNP (10 mM) and SB216763 (1 mM) virtually
ure 5C and Figure S5). IP6K1 KO mice are protected from the abolishes adipogenesis (Figures 6I and 6J). GSK3b facilitates

Cell 143, 897–910, December 10, 2010 ª2010 Elsevier Inc. 903
Figure 4. IP6K1 KO Mice Are Resistant to Obesity
(A) IP6K1 KO mice display significant reduction in body weight compared to WT littermates at the age of 10 months (male, n = 5, each set).
(B) Reduced body weight in IP6K1 KO mice reflects less fat accumulation. Epididymal white adipose tissue (EWAT) weight is significantly less in 10-month-old
IP6K1 KO mice than WT littermates (male, n = 5, each set).
(C) Six-week-old WT and IP6K1 KO mice under control diet (CD) conditions.
(D) IP6K1 KO mice are resistant to weight gain following high-fat diet (HFD) exposure. Six-week-old IP6K1 KO and their WT littermates (males and females) were
exposed to HFD for 15 weeks.
(E and F) Time-dependent increase in body weight of IP6K1 KO and WT littermate males (E) and females (F) upon exposure to control and high-fat diet
(***p < 0.001, n = 8, each set).
(G and H) Echo-MRI analysis for body fat quantification in IP6K1 KO mice after 8 weeks of HFD exposure (male, n = 5, each set). IP6K1 KO mice display signif-
icantly less deposition of total fat (G) and percent fat/lean mass (H).
(I) Weights of epididymal (E), retroperitoneal (R), dorso-subcutaneous (D), inguinal (I) white adipose tissues (WAT), and brown adipose tissue (BAT) isolated from
WT and IP6K1 KO mice on CD and on HFD for 8 weeks (male, n = 3, each set). IP6K1 KO display reduced WAT mass under both diet conditions. BAT mass is
similar in mice on CD but is increased at a lower rate in the IP6K1 KO on HFD.
(J) IP6K1 KO mice display low serum leptin levels and are resistant to HFD-induced hyperleptinemia (male, n = 6, each set).
(K) IP6K1 KO mice are protected from high-fat diet-induced enhancement in liver weight (male, n = 3, each set). Mice were exposed to CD or HFD for 8 weeks.
(L) Oil red O staining of lipid droplets in the livers of WT and IP6K1 KO mice on CD or HFD. Magnification, 203; scale bar, 30 mM.
***p < 0.001; **p < 0.01; and *p < 0.05. See also Figure S4.

904 Cell 143, 897–910, December 10, 2010 ª2010 Elsevier Inc.
Figure 5. IP6K1 Deletion Improves Glucose Homeo-
stasis under High-Fat Conditions
(A and B) IP6K1 KO mice are significantly resistant to hypergly-
cemia induced by 8 weeks exposure to HFD (male, n = 8,
each set).
(C) Glucose tolerance test (GTT) in mice after CD and HFD
exposure for 8 weeks (male, n = 5, each set). IP6K1 KO mice
on HFD display more efficient glucose removal from serum
than WT. Same aged IP6K1 KO and WT mice have similar
glucose tolerance on CD.
(D) Insulin tolerance test (ITT) at 8 weeks of CD or HFD expo-
sure in mice (male, n = 5, each set). In response to insulin,
IP6K1 KO mice display a greater glucose disposal rate than
WT littermates on HFD, with no difference on control diet.
(E) IP6K1 KO mice display reduced serum insulin under control
diet conditions and do not display the hyperinsulinemia of WT
mice at 8 weeks of HFD exposure (male, n = 6, each set).
(F) Representative western blot of 4 hr fasted IP6K1 KO mice
(as described in Figure 5E) do not display insulin resistance of
WT mice. Knockouts on HFD exhibit increased Akt signaling in
skeletal muscle.
***p < 0.001; **p < 0.01; and *p < 0.05. See also Figure S5.

ing insulin sensitivity and protein translation via the


GSK3b and mTOR signaling pathways, which are
associated with insulin resistance and weight gain
(Figure 7). Insulin activation of Akt stimulates
protein translation as well as glucose uptake and
glycogen formation (Figure 7A). Aging or high-fat
diet increases IP7 levels, which interfere with Akt
activation, leading to insulin resistance and weight
gain (Figure 7B).
IP7 inhibits Akt by acting at the PH domain of Akt
to prevent its phosphorylation and activation by
PDK1 both in vitro and in vivo. IP7’s regulation of
Akt phosphorylation by PDK1 is selective, as the
catalytic activity of PDK1 toward artificial
substrates is not affected by IP7. IP7 exerts this
action with marked potency, with its IC50 of 20
nM being several orders of magnitude lower than
the IC50 values for other reported actions of inositol
pyrophosphates, such as inhibition of cyclin-CDK
activity by 1/3-IP7 (Lee et al., 2007), and similar to
the Kd (35 nM) for PIP3 binding to the PH domain
of Akt (Currie et al., 1999). Even in the presence
of 1 mM PIP3, the physiologic activator of Akt, IP7
inhibits PDK1’s influences on Akt at equimolar
adipogenesis through enhanced expression of the adipogenic concentration, comparable to endogenous levels of IP7 (Bennett
transcription factor PPARg (Farmer, 2005). PPARg protein levels et al., 2006). Effects of IP7 are highly selective, with other inositol
decline with cotreatment of IP6K and GSK3b inhibitors and in phosphates being substantially less potent. The diphosphate in
IP6K1 KO mice white adipose tissues (Figures S5C and S5D). IP7 differentiates it from IP6 and has been shown to alter the
These observations indicate that reduced fat accumulation in protonation state of the molecule (Hand and Honek, 2007).
the IP6K1 KO mice is a result of sustained insulin sensitivity, Thus, IP7 binds the clathrin assembly protein AP3 with 5- to
increased fatty acid oxidation, and reduced adipogenesis. 10-fold greater affinity than IP6 (Ye et al., 1995).
The physiologic relevance of these findings is buttressed by
DISCUSSION the increased Akt signaling, decreased GSK3b phosphorylation,
and augmented protein translation in IP6K1 knockouts. Phos-
In summary, IP7 generation by IP6K1 is enhanced by insulin. phorylation of GSK3b inhibits its catalytic activity, leading
Moreover, IP7 is a physiologic inhibitor of Akt signaling, diminish- to increased glycogen levels and reduced adipogenesis

Cell 143, 897–910, December 10, 2010 ª2010 Elsevier Inc. 905
Figure 6. IP7 Reduces Fat Breakdown and Enhances Adipogenesis
(A) IP6K1 KO mice and WT littermates consume high-fat diets similarly (male, n = 4, each set).
(B–E) Whole-body oxygen consumption (VO2), carbon dioxide release (VCO2), energy expenditure (EE), and respiratory exchange ratio (RER) in IP6K1 KO mice on
control and high-fat diet (male, n = 4, each set). IP6K1 KO mice do not display high-fat diet-induced hypoactivity elicited by WT littermates, resulting in increased
VO2 and EE in the knockouts.
(F and G) Increased fat breakdown in IP6K1 KO mice. Mice on HFD for 25 weeks were switched to regular diet for the indicated time periods. IP6K1 KO mice
display significantly greater decreases in body weight compared to WT littermates (male, n = 3, each set).
(H) Enhancement in IP7 levels during differentiation of NIH 3T3-L1 cells. Inositol phosphate levels were detected in undifferentiated and 3 days postdifferentiated
cells. TNP reduces IP7 levels under both the conditions (n = 3).
(I and J) IP7 regulates adipogenesis through GSK3b pathway. In conjunction, TNP (10 mM) and SB216763 (1 mM) completely block differentiation of NIH 3T3-L1
cells, with minimal effect when treated alone (n = 3).
Each experiment was repeated at least three times. ***p < 0.001; **p < 0.01; *p < 0.05. See also Figure S6.

906 Cell 143, 897–910, December 10, 2010 ª2010 Elsevier Inc.
A and insulin resistance in mice, at least in part, by inhibiting Akt
Normal
and increasing GSK3b activity.
Glucose
Genetic models of insulin hypersensitivity, such as murine
Insulin
mutants of protein phosphatase 1B, PPARg, S6K1, and JNK
mutants, are resistant to HFD-induced obesity (Elchebly et al.,
PIP2 PIP3
1999; Hirosumi et al., 2002; Izumiya et al., 2008; Jones et al.,
IRS1 K
PI
3- T4 2005; Um et al., 2004). Akt activation is a common feature of
LU
G
these diverse models of increased insulin sensitivity. These
models support the notion that the sustained insulin sensitivity
Akt
IP6K1 IP7
of IP6K1 KO mice conveys resistance to weight gain. Both
reduced obesity and increased Akt signaling may elicit the
improved glucose tolerance and insulin sensitivity of the IP6K1
mTOR GSK3β Glycogen mutants.
Akt has lipogenic effects. Akt 1 and Akt 2 double-knockout
Adipogenesis mice display reduced adipose mass and skeletal muscle atrophy
Glucose homeostasis (Peng et al., 2003). Akt 2 deletion in ob/ob mice reduces fat accu-
Protein translation
mulation with insulin resistance and hyperglycemia (Leavens
et al., 2009). On the other hand, high-fat diet-induced hepatic
steatosis is correlated with decreased Akt phosphorylation
upon insulin treatment (Pinto Lde et al., 2010). Skeletal muscle-
B specific overexpression of Akt 1 reduces fat accumulation while
Insulin resistance increasing fatty acid oxidation in the liver with less steatosis (Izu-
miya et al., 2008). Akt/mTOR-mediated skeletal muscle hyper-
Insulin Glucose
trophy (Rommel et al., 2001) leading to increased insulin sensi-
tivity (Izumiya et al., 2008) may be physiologically associated
PIP2 PIP3 with the alterations in insulin sensitivity of IP6K1-deleted mice.
IRS1
3-
K
T4 Moreover, GSK3b is adipogenic so that its inhibition in IP6K1
PI LU
G mutants may contribute to their leanness (Ross et al., 2000).
Thus, the role of Akt in lipogenesis is complex and may reflect
IP6K1 IP7 Akt
isoform- and tissue-specific effects.
Overexpression of Akt can be tumorigenic (Manning and
Cantley, 2007). IP6K1 knockouts do not display spontaneous
mTOR GSK3β Glycogen tumors in their lifetime (data not shown), though we have not
exhaustively explored possible tumorigenicity.
Adipogenesis
We observe increased IP6K activity in the skeletal muscle of
HFD mice and older mice. Moreover, leptin receptor-deficient
Protein translation Glucose homeostasis
obese ‘‘pound mice’’ display increased IP6K protein levels
(A.C. and S.H.S., unpublished data). These findings are consis-
tent with age-dependent increases in IP7 levels leading to insulin
resistance and obesity.
Our findings imply that selective inhibitors of IP6K1 will have
Figure 7. Model Depicting Insulin and IP6K1 Regulation of Akt therapeutic potential in treating type-2 diabetes associated
and Sequelae
with obesity and insulin resistance. The risk of adverse effects
(A) Basal signaling. Insulin stimulates IP7 formation. IP7 inhibits Akt activity and
its downstream targets. Akt physiologically stimulates mTOR while inhibiting from such treatment can be inferred from the phenotype of
GSK3b. IP6K1 knockouts. IP6K1 mutants weigh about 15% less than
(B) Signaling in insulin resistant tissues. In aging tissues that manifest insulin controls due to less fat deposition but otherwise appear normal.
resistance, insulin stimulation of IP7 formation is augmented, leading to Males manifest decreased sperm formation, but potential infer-
pronounced inhibition of Akt, with associated lessening of mTOR activation tility of males may not represent a major problem in typical
and GSK3b inhibition.
elderly type-2 diabetics.
Arrows: green, activation; red, inhibition; bold, increased; regular, decreased;
dotted, unknown mechanism. Boxes: large, active; small, less active.
EXPERIMENTAL PROCEDURES

(Kaidanovich and Eldar-Finkelman, 2002), predicting that dele- Detection of Intracellular Inositol Phosphates
tion of IP6K1 should lead to insulin hypersensitivity, as observed The cells were plated at 60% density and incubated with 100 mCi [3H]myoino-
sitol for 3 days. For IGF-1 treatment, on the third day, cells were incubated
in IP6K1 KO mice. Insulin hypersensitivity of IP6K1 KO mice
overnight with serum-free media containing 100 mCi [3H]myoinositol. The
protects them from the impaired glucose tolerance and hyperin- next morning, cells were harvested after indicated IGF-1 treatment and were
sulinemia associated with age or high-fat diet consumption. processed for inositol phosphate detection by HPLC. For details, please see
Thus, IP7 synthesized by IP6K1 appears to mediate obesity Extended Experimental Procedures.

Cell 143, 897–910, December 10, 2010 ª2010 Elsevier Inc. 907
IGF-1, Insulin, and Serum Treatment of Mouse Embryonic body mass (fat mass + lean mass) (Kaiyala et al., 2010). Details are in Extended
Fibroblasts, Primary Hepatocytes, and HEK293 Cells Experimental Procedures.
Unless otherwise stated, cells were starved overnight and then treated with
media containing one of the following: (1) 10 nM IGF-1, (2) 10% FBS, or (3) Adipocyte Differentiation Studies
10–20 ng/ml insulin for indicated time periods. NIH 3T3-L1 preadipocyte cells were cultured and differentiated following
standard protocol (ZenBio). In brief, preadipocytes were maintained in preadi-
Membrane Isolation pocyte media (PM-1-L1) differentiated for 3 days with differentiation media
Membrane isolation employed a standard protocol using a Biovision cell (DM-2-L1). After 3 days of differentiation, cells were maintained for another 7
fractionation kit. Caveolin1 or cadherin and lactate dehydrogenase were days in adipocyte maintenance media (AM-1-L1). See details in Extended
used as membrane and cytosolic markers, respectively. Cytosolic contamina- Experimental Procedures.
tion of the membrane preparations were checked by blotting with cytosolic
markers, which showed negative results. Statistical Analysis
Membrane isolation of TNP-treated HEK293 cells employed the above All results are presented as the mean and standard error of at least three
protocol after 10 mM TNP treatment of serum-starved cells for indicated time independent experiments. Statistical significance was calculated by Student’s
periods. Cells were fractionated 15 min after IGF-1 treatment. t test using the Sigmaplot software (***p < 0.001; **p < 0.01; *p < 0.05).

Enzymatic Synthesis of Radiolabeled IP7 by IP6K1


SUPPLEMENTAL INFORMATION
Purified recombinant 6 3 His-IP6K1 was used in the reaction containing
500 mM cold IP6, 85 nCi of [3H]IP6 (total 8 3 104 cpm). IP7 was purified based
Supplemental Information includes Extended Experimental Procedures and
on standard procedures (Saiardi et al., 2004).
six figures and can be found with this article online at doi:10.1016/j.cell.
2010.11.032.
PDK1 Activity Assay on Akt T308 Site In Vitro
Purified recombinant, inactive unphosphorylated Akt at 20 nM final concentra-
tion (unless otherwise stated) was incubated with 100 mM ATP and indicated ACKNOWLEDGMENTS
concentrations of inositol polyphosphates for 10 min in a reaction buffer
containing 50 mM Tris, 100 mM NaCl, and 1 mM DTT. PDK1, final concentra- We thank Robert Luo for providing the pCDNA-TOPO-V5/His full-length and
tion 20 nM, was added, and the mixture incubated at 30 C for 30 min. Samples DPH Akt constructs; Susan Aja for the Oxymax experiments; Cory Brayton
were then boiled with LDS buffer, run on SDS-PAGE, and detected with for histological analysis; Molee Chakraborty, Nadine Forbes, Kent Werner,
a-p-T308 antibody. Bands were quantified using ImageJ software. Data and Gary Ho for technical support; and Asif Mustafa for helpful discussions.
from three independent experiments were plotted using Sigmaplot software. This work was supported by U.S. Public Health Service Grants MH18501
Details are in Extended Experimental Procedures. and DA-000266 and Research Scientist Award DA00074 (to S.H.S.).

Received: November 20, 2009


Metabolic Measurements
Revised: August 17, 2010
Metabolic parameters were measured in 10-month-old mice ad libitum fed or
Accepted: November 1, 2010
4 hr/16 hr fasted mice. Blood glucose levels were measured from tail vein
Published: December 9, 2010
bleedings of mice using an Ascensia Contour blood glucose meter and test
strips (Bayer). Ultrasensitive mouse insulin ELISA kit (Alpco Diagnostics) and
REFERENCES
mouse leptin ELISA kit (Millipore) were used to measure insulin and leptin,
respectively.
Alessi, D.R., James, S.R., Downes, C.P., Holmes, A.B., Gaffney, P.R., Reese,
Glucose tolerance test (GTT) was performed on 16 hr fasted mice injected
C.B., and Cohen, P. (1997). Characterization of a 3-phosphoinositide-
i.p. with D-glucose (2 g/kg body weight). Blood glucose level was monitored
dependent protein kinase which phosphorylates and activates protein kinase
by tail bleeding immediately before and at indicated time points after injection
Balpha. Curr. Biol. 7, 261–269.
(Bhandari et al., 2008). For insulin tolerance tests, mice were fasted 4 hr and
were given 0.75 units/kg body weight human recombinant insulin (Invitrogen) Andjelkovic, M., Alessi, D.R., Meier, R., Fernandez, A., Lamb, N.J., Frech, M.,
i.p. Blood glucose measurements were obtained from tail veins at indicated Cron, P., Cohen, P., Lucocq, J.M., and Hemmings, B.A. (1997). Role of
time points postinjection (Bhandari et al., 2008). translocation in the activation and function of protein kinase B. J. Biol.
Chem. 272, 31515–31524.
Hyperinsulinemic-Euglycemic Clamp Study and Tissue Glucose Barker, C.J., Illies, C., Gaboardi, G.C., and Berggren, P.O. (2009). Inositol
Uptake Analysis pyrophosphates: structure, enzymology and function. Cell. Mol. Life Sci. 66,
Ten-month-old mice were used in the study. Details are in Extended Experi- 3851–3871.
mental Procedures. Bennett, M., Onnebo, S.M., Azevedo, C., and Saiardi, A. (2006). Inositol
pyrophosphates: metabolism and signaling. Cell. Mol. Life Sci. 63, 552–564.
Acute Insulin Treatment in Mice
Berridge, M.J., Lipp, P., and Bootman, M.D. (2000). The versatility and univer-
Ten-month-old mice, after 4 hr fast, were anaesthetized, and 25 mU/kg insulin
sality of calcium signalling. Nat. Rev. Mol. Cell Biol. 1, 11–21.
(Invitrogen) or equal volumes of vehicle were administered through the portal
Bhandari, R., Saiardi, A., Ahmadibeni, Y., Snowman, A.M., Resnick, A.C.,
vein. Gastrocnemius muscle, epididymal white adipose tissue (EWAT), and
Kristiansen, T.Z., Molina, H., Pandey, A., Werner, J.K., Jr., Juluri, K.R., et al.
liver were collected 120 s after the injection and immediately stored in liquid
(2007). Protein pyrophosphorylation by inositol pyrophosphates is a posttrans-
nitrogen. Protein extracts from the tissue samples were prepared and run on
lational event. Proc. Natl. Acad. Sci. USA 104, 15305–15310.
SDS-PAGE. For detection of tyrosine phosphorylation on IRS1, IRS1 was
immunoprecipitated from 1 mg total cell lysate and was blotted with a-p-tyro- Bhandari, R., Juluri, K.R., Resnick, A.C., and Snyder, S.H. (2008). Gene
sine and a-IRS1 antibody. deletion of inositol hexakisphosphate kinase 1 reveals inositol pyrophosphate
regulation of insulin secretion, growth, and spermiogenesis. Proc. Natl. Acad.
Indirect Calorimetry Sci. USA 105, 2349–2353.
Indirect calorimetry was conducted in an open-flow indirect calorimeter Calleja, V., Alcor, D., Laguerre, M., Park, J., Vojnovic, B., Hemmings, B.A.,
(Oxymax Equal Flow System; Columbus Instruments, Columbus, OH) at the Downward, J., Parker, P.J., and Larijani, B. (2007). Intramolecular and
Center for Metabolism and Obesity Research (Johns Hopkins University intermolecular interactions of protein kinase B define its activation in vivo.
School of Medicine). Energy expenditure (EE) was calculated based on total PLoS Biol. 5, e95.

908 Cell 143, 897–910, December 10, 2010 ª2010 Elsevier Inc.
Chakraborty, A., Koldobskiy, M.A., Sixt, K.M., Juluri, K.R., Mustafa, A.K., Izumiya, Y., Hopkins, T., Morris, C., Sato, K., Zeng, L., Viereck, J., Hamilton,
Snowman, A.M., van Rossum, D.B., Patterson, R.L., and Snyder, S.H. J.A., Ouchi, N., LeBrasseur, N.K., and Walsh, K. (2008). Fast/Glycolytic muscle
(2008). HSP90 regulates cell survival via inositol hexakisphosphate kinase-2. fiber growth reduces fat mass and improves metabolic parameters in obese
Proc. Natl. Acad. Sci. USA 105, 1134–1139. mice. Cell Metab. 7, 159–172.
Chan, T.O., Rittenhouse, S.E., and Tsichlis, P.N. (1999). AKT/PKB and other Jones, J.R., Barrick, C., Kim, K.A., Lindner, J., Blondeau, B., Fujimoto, Y.,
D3 phosphoinositide-regulated kinases: kinase activation by phosphoinosi- Shiota, M., Kesterson, R.A., Kahn, B.B., and Magnuson, M.A. (2005).
tide-dependent phosphorylation. Annu. Rev. Biochem. 68, 965–1014. Deletion of PPARgamma in adipose tissues of mice protects against high fat
Cho, H., Mu, J., Kim, J.K., Thorvaldsen, J.L., Chu, Q., Crenshaw, E.B., III, diet-induced obesity and insulin resistance. Proc. Natl. Acad. Sci. USA 102,
Kaestner, K.H., Bartolomei, M.S., Shulman, G.I., and Birnbaum, M.J. (2001). 6207–6212.
Insulin resistance and a diabetes mellitus-like syndrome in mice lacking the Kahn, S.E., Hull, R.L., and Utzschneider, K.M. (2006). Mechanisms linking
protein kinase Akt2 (PKB beta). Science 292, 1728–1731. obesity to insulin resistance and type 2 diabetes. Nature 444, 840–846.
Cota, D., Proulx, K., Smith, K.A., Kozma, S.C., Thomas, G., Woods, S.C., and Kaidanovich, O., and Eldar-Finkelman, H. (2002). The role of glycogen
Seeley, R.J. (2006). Hypothalamic mTOR signaling regulates food intake. synthase kinase-3 in insulin resistance and type 2 diabetes. Expert Opin.
Science 312, 927–930. Ther. Targets 6, 555–561.
Cross, D.A., Alessi, D.R., Cohen, P., Andjelkovich, M., and Hemmings, B.A. Kaiyala, K.J., Morton, G.J., Leroux, B.G., Ogimoto, K., Wisse, B., and
(1995). Inhibition of glycogen synthase kinase-3 by insulin mediated by protein Schwartz, M.W. (2010). Identification of body fat mass as a major determinant
kinase B. Nature 378, 785–789. of metabolic rate in mice. Diabetes 59, 1657–1666.
Currie, R.A., Walker, K.S., Gray, A., Deak, M., Casamayor, A., Downes, C.P., Koldobskiy, M.A., Chakraborty, A., Werner, J.K., Jr., Snowman, A.M., Juluri,
Cohen, P., Alessi, D.R., and Lucocq, J. (1999). Role of phosphatidylinositol K.R., Vandiver, M.S., Kim, S., Heletz, S., and Snyder, S.H. (2010). p53-medi-
3,4,5-trisphosphate in regulating the activity and localization of 3-phosphoino- ated apoptosis requires inositol hexakisphosphate kinase-2. Proc. Natl.
sitide-dependent protein kinase-1. Biochem. J. 337, 575–583. Acad. Sci. USA, in press. Published online November 15, 2010. 10.1073/
Di Paolo, G., and De Camilli, P. (2006). Phosphoinositides in cell regulation and pnas.1015671107.
membrane dynamics. Nature 443, 651–657.
Komander, D., Fairservice, A., Deak, M., Kular, G.S., Prescott, A.R., Peter
Downes, C.P., Gray, A., Fairservice, A., Safrany, S.T., Batty, I.H., and Fleming, Downes, C., Safrany, S.T., Alessi, D.R., and van Aalten, D.M. (2004).
I. (2005). The regulation of membrane to cytosol partitioning of signalling Structural insights into the regulation of PDK1 by phosphoinositides and
proteins by phosphoinositides and their soluble headgroups. Biochem. Soc. inositol phosphates. EMBO J. 23, 3918–3928.
Trans. 33, 1303–1307.
Leavens, K.F., Easton, R.M., Shulman, G.I., Previs, S.F., and Birnbaum, M.J.
Elchebly, M., Payette, P., Michaliszyn, E., Cromlish, W., Collins, S., Loy, A.L., (2009). Akt2 is required for hepatic lipid accumulation in models of insulin
Normandin, D., Cheng, A., Himms-Hagen, J., Chan, C.C., et al. (1999). resistance. Cell Metab. 10, 405–418.
Increased insulin sensitivity and obesity resistance in mice lacking the protein
Lee, Y.S., Mulugu, S., York, J.D., and O’Shea, E.K. (2007). Regulation of a
tyrosine phosphatase-1B gene. Science 283, 1544–1548.
cyclin-CDK-CDK inhibitor complex by inositol pyrophosphates. Science 316,
Farmer, S.R. (2005). Regulation of PPARgamma activity during adipogenesis. 109–112.
Int J Obes (Lond) 29 (Suppl 1), S13–S16.
Lemmon, M.A. (2008). Membrane recognition by phospholipid-binding
Fruman, D.A., Snapper, S.B., Yballe, C.M., Alt, F.W., and Cantley, L.C. (1999).
domains. Nat. Rev. Mol. Cell Biol. 9, 99–111.
Phosphoinositide 3-kinase knockout mice: role of p85alpha in B cell develop-
ment and proliferation. Biochem. Soc. Trans. 27, 624–629. Lin, H., Fridy, P.C., Ribeiro, A.A., Choi, J.H., Barma, D.K., Vogel, G., Falck,
J.R., Shears, S.B., York, J.D., and Mayr, G.W. (2009). Structural analysis and
Funai, K., Parkington, J.D., Carambula, S., and Fielding, R.A. (2006). Age-
detection of biological inositol pyrophosphates reveal that the family of VIP/
associated decrease in contraction-induced activation of downstream targets
diphosphoinositol pentakisphosphate kinases are 1/3-kinases. J. Biol. Chem.
of Akt/mTor signaling in skeletal muscle. Am. J. Physiol. Regul. Integr. Comp.
284, 1863–1872.
Physiol. 290, R1080–R1086.
Luo, H.R., Huang, Y.E., Chen, J.C., Saiardi, A., Iijima, M., Ye, K., Huang, Y.,
Hand, C.E., and Honek, J.F. (2007). Phosphate transfer from inositol pyro-
Nagata, E., Devreotes, P., and Snyder, S.H. (2003). Inositol pyrophosphates
phosphates InsP5PP and InsP4(PP)2: a semi-empirical investigation. Bioorg.
mediate chemotaxis in Dictyostelium via pleckstrin homology domain-PtdIns
Med. Chem. Lett. 17, 183–188.
(3,4,5)P3 interactions. Cell 114, 559–572.
Hirosumi, J., Tuncman, G., Chang, L., Görgün, C.Z., Uysal, K.T., Maeda, K.,
Karin, M., and Hotamisligil, G.S. (2002). A central role for JNK in obesity and Manning, B.D., and Cantley, L.C. (2007). AKT/PKB signaling: navigating
insulin resistance. Nature 420, 333–336. downstream. Cell 129, 1261–1274.

Hoffler, U., Hobbie, K., Wilson, R., Bai, R., Rahman, A., Malarkey, D., Travlos, Memmott, R.M., and Dennis, P.A. (2009). Akt-dependent and -independent
G., and Ghanayem, B.I. (2009). Diet-induced obesity is associated with hyper- mechanisms of mTOR regulation in cancer. Cell. Signal. 21, 656–664.
leptinemia, hyperinsulinemia, hepatic steatosis, and glomerulopathy in C57Bl/ Mulugu, S., Bai, W., Fridy, P.C., Bastidas, R.J., Otto, J.C., Dollins, D.E.,
6J mice. Endocrine 36, 311–325. Haystead, T.A., Ribeiro, A.A., and York, J.D. (2007). A conserved family
Holz, M.K., Ballif, B.A., Gygi, S.P., and Blenis, J. (2005). mTOR and S6K1 of enzymes that phosphorylate inositol hexakisphosphate. Science 316,
mediate assembly of the translation preinitiation complex through dynamic 106–109.
protein interchange and ordered phosphorylation events. Cell 123, 569–580. Myers, M.G., Cowley, M.A., and Münzberg, H. (2008). Mechanisms of leptin
Illies, C., Gromada, J., Fiume, R., Leibiger, B., Yu, J., Juhl, K., Yang, S.N., action and leptin resistance. Annu. Rev. Physiol. 70, 537–556.
Barma, D.K., Falck, J.R., Saiardi, A., et al. (2007). Requirement of inositol Padmanabhan, U., Dollins, D.E., Fridy, P.C., York, J.D., and Downes, C.P.
pyrophosphates for full exocytotic capacity in pancreatic beta cells. Science (2009). Characterization of a selective inhibitor of inositol hexakisphosphate
318, 1299–1302. kinases: use in defining biological roles and metabolic relationships of inositol
Irvine, R.F., and Schell, M.J. (2001). Back in the water: the return of the inositol pyrophosphates. J. Biol. Chem. 284, 10571–10582.
phosphates. Nat. Rev. Mol. Cell Biol. 2, 327–338. Peng, X.D., Xu, P.Z., Chen, M.L., Hahn-Windgassen, A., Skeen, J., Jacobs, J.,
Ito, M., Gomori, A., Suzuki, J., Tsujioka, S., Sasaki, M., Matsuda, M., Sundararajan, D., Chen, W.S., Crawford, S.E., Coleman, K.G., and Hay, N.
Bednarek, M.A., Ito, M., Ishihara, A., Iwaasa, H., et al. (2008). Antagonism of (2003). Dwarfism, impaired skin development, skeletal muscle atrophy,
central melanin-concentrating hormone 1 receptor alleviates steatohepatitis delayed bone development, and impeded adipogenesis in mice lacking Akt1
in mice. J. Endocrinol. 198, 309–315. and Akt2. Genes Dev. 17, 1352–1365.

Cell 143, 897–910, December 10, 2010 ª2010 Elsevier Inc. 909
Pinto Lde, F., Compri, C.M., Fornari, J.V., Bartchewsky, W., Cintra, D.E., Sarbassov, D.D., Guertin, D.A., Ali, S.M., and Sabatini, D.M. (2005).
Trevisan, M., Carvalho, Pde.O., Ribeiro, M.L., Velloso, L.A., Saad, M.J., et al. Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex.
(2010). The immunosuppressant drug, thalidomide, improves hepatic Science 307, 1098–1101.
alterations induced by a high-fat diet in mice. Liver Int. 30, 603–610. Shao, J., Yamashita, H., Qiao, L., and Friedman, J.E. (2000). Decreased
Reddy, J.K., and Rao, M.S. (2006). Lipid metabolism and liver inflammation. II. Akt kinase activity and insulin resistance in C57BL/KsJ-Leprdb/db mice.
Fatty liver disease and fatty acid oxidation. Am. J. Physiol. Gastrointest. Liver J. Endocrinol. 167, 107–115.
Physiol. 290, G852–G858. Shay, K.P., and Hagen, T.M. (2009). Age-associated impairment of Akt
Rommel, C., Bodine, S.C., Clarke, B.A., Rossman, R., Nunez, L., Stitt, T.N., phosphorylation in primary rat hepatocytes is remediated by alpha-lipoic
Yancopoulos, G.D., and Glass, D.J. (2001). Mediation of IGF-1-induced acid through PI3 kinase, PTEN, and PP2A. Biogerontology 10, 443–456.
skeletal myotube hypertrophy by PI(3)K/Akt/mTOR and PI(3)K/Akt/GSK3 Shears, S.B. (2007). Understanding the biological significance of diphosphoi-
pathways. Nat. Cell Biol. 3, 1009–1013. nositol polyphosphates (‘inositol pyrophosphates’). Biochem. Soc. Symp. 74,
211–221.
Ross, S.E., Hemati, N., Longo, K.A., Bennett, C.N., Lucas, P.C., Erickson, R.L.,
and MacDougald, O.A. (2000). Inhibition of adipogenesis by Wnt signaling. Tang, Q.Q., Grønborg, M., Huang, H., Kim, J.W., Otto, T.C., Pandey, A., and
Science 289, 950–953. Lane, M.D. (2005). Sequential phosphorylation of CCAAT enhancer-binding
protein beta by MAPK and glycogen synthase kinase 3beta is required for
Ruggero, D., and Sonenberg, N. (2005). The Akt of translational control.
adipogenesis. Proc. Natl. Acad. Sci. USA 102, 9766–9771.
Oncogene 24, 7426–7434.
Taniguchi, C.M., Emanuelli, B., and Kahn, C.R. (2006). Critical nodes in signal-
Saiardi, A., Erdjument-Bromage, H., Snowman, A.M., Tempst, P., and Snyder, ling pathways: insights into insulin action. Nat. Rev. Mol. Cell Biol. 7, 85–96.
S.H. (1999). Synthesis of diphosphoinositol pentakisphosphate by a newly
Tou, J.C., and Wade, C.E. (2002). Determinants affecting physical activity
identified family of higher inositol polyphosphate kinases. Curr. Biol. 9,
levels in animal models. Exp. Biol. Med. (Maywood) 227, 587–600.
1323–1326.
Um, S.H., Frigerio, F., Watanabe, M., Picard, F., Joaquin, M., Sticker, M.,
Saiardi, A., Nagata, E., Luo, H.R., Snowman, A.M., and Snyder, S.H. (2001).
Fumagalli, S., Allegrini, P.R., Kozma, S.C., Auwerx, J., and Thomas, G.
Identification and characterization of a novel inositol hexakisphosphate
(2004). Absence of S6K1 protects against age- and diet-induced obesity while
kinase. J. Biol. Chem. 276, 39179–39185.
enhancing insulin sensitivity. Nature 431, 200–205.
Saiardi, A., Sciambi, C., McCaffery, J.M., Wendland, B., and Snyder, S.H. Welsh, G.I., Hers, I., Berwick, D.C., Dell, G., Wherlock, M., Birkin, R., Leney, S.,
(2002). Inositol pyrophosphates regulate endocytic trafficking. Proc. Natl. and Tavaré, J.M. (2005). Role of protein kinase B in insulin-regulated glucose
Acad. Sci. USA 99, 14206–14211. uptake. Biochem. Soc. Trans. 33, 346–349.
Saiardi, A., Bhandari, R., Resnick, A.C., Snowman, A.M., and Snyder, S.H. Ye, W., Ali, N., Bembenek, M.E., Shears, S.B., and Lafer, E.M. (1995). Inhibition
(2004). Phosphorylation of proteins by inositol pyrophosphates. Science of clathrin assembly by high affinity binding of specific inositol polyphosphates
306, 2101–2105. to the synapse-specific clathrin assembly protein AP-3. J. Biol. Chem. 270,
Saiardi, A., Resnick, A.C., Snowman, A.M., Wendland, B., and Snyder, S.H. 1564–1568.
(2005). Inositol pyrophosphates regulate cell death and telomere length York, S.J., Armbruster, B.N., Greenwell, P., Petes, T.D., and York, J.D. (2005).
through phosphoinositide 3-kinase-related protein kinases. Proc. Natl. Acad. Inositol diphosphate signaling regulates telomere length. J. Biol. Chem. 280,
Sci. USA 102, 1911–1914. 4264–4269.

910 Cell 143, 897–910, December 10, 2010 ª2010 Elsevier Inc.
Loss of Anion Transport without Increased
Sodium Absorption Characterizes Newborn
Porcine Cystic Fibrosis Airway Epithelia
Jeng-Haur Chen,1,3 David A. Stoltz,1 Philip H. Karp,1,3 Sarah E. Ernst,1 Alejandro A. Pezzulo,1 Thomas O. Moninger,1
Michael V. Rector,1 Leah R. Reznikov,1,3 Janice L. Launspach,1 Kathryn Chaloner,2 Joseph Zabner,1
and Michael J. Welsh1,3,*
1Department of Internal Medicine
2Department of Biostatistics
3Howard Hughes Medical Institute

Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
*Correspondence: michael-welsh@uiowa.edu
DOI 10.1016/j.cell.2010.11.029

SUMMARY cates features of the disease; mice with mutated CFTR genes
do not develop gastrointestinal or lung disease typical of
Defective transepithelial electrolyte transport is human CF (Grubb and Boucher, 1999). Therefore, we recently
thought to initiate cystic fibrosis (CF) lung disease. developed CFTR / pigs (hereafter referred to as CF pigs)
Yet, how loss of CFTR affects electrolyte transport (Rogers et al., 2008b). At birth, they manifest features typically
remains uncertain. CFTR / pigs spontaneously observed in patients with CF, including pancreatic destruction,
develop lung disease resembling human CF. At birth, meconium ileus, early focal biliary cirrhosis, and microgallblad-
der (Meyerholz et al., 2010b). Within a few months of birth, CF
their airways exhibit a bacterial host defense defect,
pigs spontaneously develop lung disease with the hallmark
but are not inflamed. Therefore, we studied ion trans-
features of CF including inflammation, infection, mucus accu-
port in newborn nasal and tracheal/bronchial mulation, tissue remodeling, and airway obstruction (Stoltz
epithelia in tissues, cultures, and in vivo. CFTR / et al., 2010).
epithelia showed markedly reduced Cl- and HCO3- Finding that CF pigs develop airway disease like that in
transport. However, in contrast to a widely held humans provided an opportunity to explore very early events in
view, lack of CFTR did not increase transepithelial the disease. We previously showed that within hours of birth,
Na+ or liquid absorption or reduce periciliary liquid CF pigs have a reduced ability to eliminate bacteria that either
depth. Like human CF, CFTR / pigs showed enter the lung spontaneously or that are introduced experimen-
increased amiloride-sensitive voltage and current, tally (Stoltz et al., 2010). However, like newborn human babies
but lack of apical Cl- conductance caused the with CF, CF pigs lack airway inflammation at birth. Those data
indicate that impaired bacterial elimination is the pathogenic
change, not increased Na+ transport. These results
event that begins a cascade of inflammation, remodeling and
indicate that CFTR provides the predominant trans-
pathology in CF lungs. Thus, these newborn animals provide
cellular pathway for Cl- and HCO3- in porcine airway an ideal model in which to evaluate ion transport processes
epithelia, and reduced anion permeability may because they possess the host defense defect, but they do not
initiate CF airway disease. yet exhibit inflammation, tissue remodeling or other features of
progressive CF. Hence, electrolyte transport defects can be
INTRODUCTION attributed to loss of CFTR rather than to secondary manifesta-
tions of the disease.
Loss of cystic fibrosis transmembrane conductance regulator Abnormal electrolyte transport across airway epithelia has
(CFTR) function causes CF (Davis, 2006; Quinton, 1999; frequently been hypothesized to cause the initial CF host
Rowe et al., 2005; Welsh et al., 2001). Disease manifestations defense defect (Boucher, 2007; Davis, 2006; Quinton, 1999;
appear in many organs, but most morbidity and mortality Rowe et al., 2005; Verkman et al., 2003; Welsh et al., 2001;
currently arise from airway disease, where inflammation and Wine, 1999). In CF epithelia, loss of CFTR decreases airway
infection destroy the lung. Understanding the pathogenesis of Cl- and HCO3- transport. This result is consistent with the anion
lung disease has been difficult, and there are many theories channel activity of CFTR (Sheppard and Welsh, 1999). Some
to explain how deficient CFTR function causes airway disease have also concluded that CFTR negatively regulates epithelial
(Boucher, 2007; Davis, 2006; Quinton, 1999; Rowe et al., 2005; Na+ channels (ENaC); hence CFTR mutations are proposed to
Verkman et al., 2003; Welsh et al., 2001; Wine, 1999). One eliminate that ENaC inhibition, increase Na+ permeability, and
factor impeding progress in identifying the events that initiate cause Na+ hyperabsorption, which is widely viewed as the initial
airway disease has been lack of an animal model that repli- event in CF lung disease pathogenesis (Boucher, 2007).

Cell 143, 911–923, December 10, 2010 ª2010 Elsevier Inc. 911
To understand how CF affects airway epithelial ion transport, There is evidence that changes in Na+ transport can affect the
we asked if loss of CFTR would disrupt transepithelial Cl-, lung. For example, transgenic mice overexpressing the b subunit
HCO3-, and Na+ transport in CF pigs. We studied newborn of the epithelial Na+ channel (bENaC) had lung disease that
animals to identify defects prior to the onset of inflammation. shared some features with CF (Mall et al., 2004). Mutations
Knowledge of the extent to which these processes are disrupted have also been reported in human ENaC genes, and they may
is key to understanding CF airway disease and is important for contribute to lung disease with some CF-like features. However,
developing mechanism-based treatments and preventions. the ENaC mutations are associated with both decreases and
increases in ENaC activity (Azad et al., 2009; Baker et al.,
RESULTS 1998; Huber et al., 2010; Kerem et al., 1999; Schaedel et al.,
1999; Sheridan et al., 2005). Thus, while alterations in Na+
CF Pig Airways Lack cAMP-Stimulated permeability can contribute to lung disease, those results do
Cl and HCO3 Transport not indicate whether Na+ absorption is increased, reduced, or
We measured the nasal and tracheal transepithelial voltage (Vt) unchanged in CF.
in vivo in newborn pigs. Perfusion of the apical surface of Therefore, we measured Vt and the response to amiloride
epithelia with a Cl -free solution and isoproterenol (to increase in vivo in newborn pigs. In the nose, Vt and DVtamiloride were
cellular cAMP levels) hyperpolarized Vt in non-CF pigs (Figures greater in CF than non-CF pigs (Figures 2A and 2C) (Rogers
1A and 1B) (Rogers et al., 2008b). In contrast, Vt failed to hyper- et al., 2008b). Remarkably, this was not the case in tracheal
polarize in CF pigs. These data suggest a lack of cAMP-stimu- epithelia; Vt and DVtamiloride were similar in non-CF and CF pigs
lated Cl permeability in CF. (Figures 2B and 2C).
When non-CF nasal, tracheal, and bronchial epithelia were Earlier studies showed that Vt and DVtamiloride are more nega-
excised or cultured as differentiated airway epithelia and studied tive in nasal and tracheal epithelia of CF patients than in non-CF
in Ussing chambers, adding forskolin and isobutylmethylxan- controls (Davies et al., 2005; Knowles et al., 1981; Standaert
thine (IBMX) to elevate cellular cAMP levels increased absolute et al., 2004). Our data in porcine nasal epithleia parallel those
values of Vt (Figures 1C and 1D), short-circuit current (Isc) results. However, interestingly, when measurements were
(Figures 1E and 1G), and transepithelial electrical conductance made in main bronchi and distal airways of children, Vt values
(Gt) (Figures 1F and 1H). Adding GlyH-101, which inhibits were similar in CF and non-CF (Davies et al., 2005). Those
CFTR (Figure S1 available online) (Muanprasat et al., 2004), results are like the data in porcine trachea. It seems that airway
had the opposite effects (Figure 1I–1L). In contrast, CF epithelia region and age, and perhaps inflammation and infection influ-
failed to respond to either forskolin and IBMX or GlyH-101 (Fig- ence the activity of epithelial ion channels and thereby whether
ure 1C–1L). CFTR has a significant HCO3 conductance, and a Vt difference exists between CF and non-CF epithelia. It will
human non-CF airway epithelia transport HCO3- (Poulsen also be important to study electrolyte transport in vivo, in
et al., 1994; Smith and Welsh, 1992). When we studied non-CF excised tissue, and in cultures from older CF pigs as the disease
tracheal epithelia in Cl--free bathing solution containing 25 mM progresses.
HCO3 , forskolin and IBMX stimulated and then GlyH-101 in-
hibited Isc and Gt (Figures 1M and 1N), revealing electrically Absorptive Na+ Fluxes Are Not Increased in Porcine CF
conductive HCO3- transport. CF epithelia lacked these Airway Epithelia
responses. The difference in Vt between CF and non-CF nasal epithelia
These data indicate that porcine CF airway epithelia extending could relate to differences in Na+ transport. Therefore, we
from nose to bronchi lack cAMP-stimulated Cl and HCO3 directly examined Na+ transport by measuring transepithelial
22
permeability. Our findings agree with studies of human airway Na+ fluxes. We studied primary cultures of differentiated
epithelia, which have consistently demonstrated a loss of Cl airway epithelia and used open-circuit conditions to mimic the
and HCO3 permeability in CF airway epithelia (Knowles et al., in vivo situation. There were three main observations. First, in
1983; Smith and Welsh, 1992; Standaert et al., 2004; Widdi- tracheal epithelia, unidirectional and net Na+ fluxes did not differ
combe et al., 1985). Moreover, our results indicate that in wild- between CF and non-CF epithelia (Figure 3A, Table S1). Adding
type porcine airway epithelia, CFTR provides an important trans- amiloride decreased the unidirectional absorptive (apical to ba-
epithelial pathway for Cl and HCO3 . solateral) and net Na+ fluxes, indicating the importance of apical
Na+ channels for Na+ absorption. Second, in nasal epithelia, Na+
Vt Is Abnormal in CF Nasal, but Not Tracheal Epithelia fluxes and the response to amiloride were also similar in CF and
In Vivo non-CF epithelia (Figure 3B). Third, nasal epithelia had greater
The first indication of abnormal electrolyte transport in CF unidirectional absorptive fluxes and net Na+ absorption than
airways was the finding that nasal Vt was more electrically nega- tracheal epithelia (compare Figures 3A and 3B).
tive in CF than non-CF subjects and that amiloride produced Like our data in pigs, in human nasal epithelia, 22Na+ fluxes
a greater reduction in Vt (DVtamiloride) in CF (Knowles et al., measured under open-circuit conditions revealed no difference
1981). Those and additional observations led the authors to between non-CF and CF (Willumsen and Boucher, 1991a,
conclude that CF epithelia have increased Na+ absorption that 1991b). Under short-circuited conditions, which differ from the
depletes periciliary liquid, which in turn impairs mucociliary in vivo situation, net 22Na+ fluxes were reported to be either the
clearance and initiates lung disease (Boucher, 2007; Donaldson same or increased in CF versus non-CF (Boucher et al., 1986;
and Boucher, 2007). Knowles et al., 1983).

912 Cell 143, 911–923, December 10, 2010 ª2010 Elsevier Inc.
Non-CF CF

A Nasal in vivo B Tracheal in vivo C Excised tissue D Culture


# -0.8 -16 (61)
# (21)
-20 -20

ΔVtF&I (mV)

ΔVtF&I (mV)
(8)
Vt (mV)

Vt (mV)
# #
(10)
-10 -10 -0.4 (25) -8 (34)
(6) (27) (22)
(5) (20) (26)
0 0 0 * * 0
* *
Amil 0Cl Iso Amil 0Cl Iso Nasal T/B Nasal T/B

E Excised tissue F Excised tissue G Culture H Culture

ΔIscF&I (μA/cm )
ΔGtF&I (mS/cm )

ΔGtF&I (mS/cm )
ΔIscF&I (μA/cm )

24 4 50 6

2
2

2
2

(61)

(21)
12 (25) 2 25 (34)
3
(26) (27) (22)
(20)
0 * * 0 * * 0 * * 0 * *
Nasal T/B Nasal T/B Nasal T/B Nasal T/B

I Excised tissue J Excised tissue K Culture L Culture


ΔIscGlyH (μA/cm )
ΔGtGlyH (mS/cm )

ΔGtGlyH (mS/cm )
ΔIscGlyH (μA/cm )

*
2

* 0
2

2
2

0 * 0
(20)
* (27) * *
(26)* 0 (34)
*
(22)
-12 -25 -3
(25)
(61)

-24 (21) -4 -50 -6


Nasal T/B Nasal T/B Nasal T/B Nasal T/B

Culture Culture
M - - N - -
(Cl -free/HCO3 ) (Cl -free/HCO3 )
12 (9) 1
ΔGt (mS/cm )
ΔIsc (μA/cm )

2
2

(7)

0 * 0 * *
*
-12 -1
F&I GlyH F&I GlyH

Figure 1. Loss of CFTR Decreases Anion Transport in CF Airway Epithelia


Data are means ± SE from newborn CFTR+/+ (open symbols and bars) and CFTR / (closed symbols and bars) pigs. Amiloride (100 mM) was present on the apical
surface in all cases. Numbers in parentheses indicate n, asterisk indicates p < 0.05 between CF and non-CF, and T/B indicates tracheal/bronchial.
(A and B) Vt measured in vivo in nasal and tracheal epithelia in the presence of amiloride (100 mM), during perfusion with a Cl -free solution (0Cl) containing ami-
loride, and during perfusion with a Cl -free solution containing isoproterenol (10 mM) and amiloride. Nasal epithelia include data from four non-CF and four CF pigs
that were previously reported (Rogers et al., 2008b). #p < 0.05 compared to initial value.
(C–H) Change in Vt, Isc, and Gt induced by adding 10 mM forskolin and 100 mM IBMX (DVtF&I, DIscF&I, and DGtF&I) to excised and cultured nasal and tracheal/
bronchial epithelia.
(I–L) Change in Isc (DIscGlyH) and Gt (DGtGlyH) following addition of GlyH-101 (100 mM) to excised and cultured nasal and tracheal/bronchial epithelia.
(M and N) CFTR-mediated HCO3- transport in cultured tracheal epithelia. Solution was Cl -free and contained 25 mM HCO3 . Data are DIsc and DGt following
addition of forskolin and IBMX and GlyH-101.
See also Figure S1.

Liquid Absorption Is Not Increased in Porcine CF a greater rate than non-CF epithelia. In fact, in nasal epithelia,
Epithelia the absorption rate was less in CF than non-CF epithelia.
We also measured rates of transepithelial liquid absorption, which In studies of cultured human airway epithelia, the initial rate of
is driven by Na+ absorption. Liquid absorption rates were greater liquid absorption has been reported to be increased (Matsui
in nasal than tracheal epithelia, consistent with the 22Na+ fluxes et al., 1998), similar (Van Goor et al., 2009), or reduced (Zabner
(Figure 3C). However, CF epithelia did not absorb liquid at et al., 1998) in CF compared to non-CF. The reason for the

Cell 143, 911–923, December 10, 2010 ª2010 Elsevier Inc. 913
Non-CF CF

A B C
Nasal Tracheal (8) (5) (10) (6)
-30 * -30 0

ΔVtAmil (mV)
Vt (mV)

Vt (mV)
*
0 0
30 *
Basal Amil Basal Amil

l
sa

ea
Na

ch
Tra
Figure 2. Vt In Vivo Is Abnormal in CF Nasal Epithelia, but Not Tracheal Epithelia
Data are mean ± SE from CFTR+/+ (open symbols and bars) and CFTR / (closed symbols and bars) pigs.
(A and B) Effects of amiloride (100 mM) on nasal and tracheal Vt in vivo.
(C) Amiloride-sensitive change in Vt (DVtamiloride) in vivo. *p < 0.05 compared to non-CF.

differences is uncertain, but might relate to variations in basal images also revealed both erect and bent cilia and heterogeneity
CFTR activity (Zabner et al., 1998). in the depth of periciliary liquid covering airways of both geno-
types (Figure 3E). The periciliary liquid depth was not statistically
The Depth of Periciliary Liquid Is Not Altered by Lack different between non-CF (4.0 ± 0.3 mm, n = 5 pigs) and CF (4.7 ±
of CFTR 0.3 mm, n = 5 pigs) epithelia (Figure 3G).
Transepithelial ion and H2O movement contribute to the depth of Compared to earlier studies, our measurements of periciliary
liquid covering the airway surface. Twenty-four hours after adding liquid depth have the advantages that the epithelia were in vivo
liquid to the apical surface of cultured epithelia, the depth of peri- rather than cultured, they were immediately prepared without
ciliary liquid has been reported to be less in CF compared to non- other manipulations, the epithelia did not demonstrate inflamma-
CF epithelia (Matsui et al., 1998; Van Goor et al., 2009). A study that tion from chronic infection, and the experiments were performed
obtained bronchoscopic biopsies from patients with CF reported at a time point when bacterial eradication was impaired. Our data
that although not statistically significant, there was a trend toward also agree with an earlier study of maximal cilia length in formalin
reduced periciliary liquid height in CF (Griesenbach et al., 2010). fixed/paraffin embedded newborn porcine airway epithelia,
However, the authors noted that inflammation (most patients which showed no difference between CF and non-CF (Meyerholz
were experiencing a respiratory exacerbation) and the methods et al., 2010a). Potential differences with a study of broncho-
used (periciliary liquid height could not be measured in half the scopic biopsies in patients with acute and chronic disease (Grie-
patients or over the majority of cells) limit the interpretation. senbach et al., 2010) raise interesting questions of whether
To test the hypothesis that loss of CFTR alters periciliary liquid inflammation with its associated effects on surface epithelium
depth in the absence of infection, inflammation, and tissue re- and submucosal glands might change ion transport or periciliary
modeling, we studied pigs 8–15 hr after birth. In <1 min following liquid height. Although our data show no difference in periciliary
euthanasia, we removed and placed tracheal segments in a non- liquid depth between CF and non-CF newborn pigs, it is possible
aqueous fixative containing osmium tetroxide to rapidly preserve that with time and progression of disease, the depth of periciliary
the morphology of the airway surface (Matsui et al., 1998; Satir, liquid might differ between the genotypes. In addition, although
1963; Sims and Horne, 1997). The depth of periciliary liquid we measured periciliary liquid depth in trachea because of the
showed substantial variability in both non-CF and CF epithelia, speed with which we could remove and prepare the tissue, it
with areas of deeper liquid and outstretched cilia and shallower will also be important to study its depth in distal airways.
areas with cilia that appeared bent over (Figure 3D). Therefore, All these measurements indicated that Na+ absorption by CF
we examined multiple portions of trachea, prepared multiple tracheal/bronchial epithelia did not exceed that in non-CF. Strik-
sections from each portion, and made many measurements ingly, this was also true in nasal epithelia. So why in nasal
from each section. Observers unaware of genotype measured epithelia are Vt and DVtamiloride increased in CF? To answer this
periciliary liquid depth. A histogram of periciliary liquid depth is question, we first studied cultured and excised epithelia and
shown in Figure 3F; the mean depths of non-CF (4.5 ± 0.3 mm, examined electrophysiological properties (Vt, Isc, and Gt) that
n = 8 pigs) and CF (4.4 ± 0.2 mm, n = 5 pigs) periciliary liquid are influenced by apical Na+ conductance. Those results,
did not differ statistically. In addition, we prepared thin sections considered together with an equivalent circuit model of the
from the same blocks and examined them with transmission epithelium suggested an explanation for why electrophysiolog-
electron microscopy. The transmission electron microscopic ical properties differ between CF and non-CF nasal epithelia
images provided a smaller area for observation than light micro- even though Na+ absorption is not increased. We then tested
scopic images and the number of samples was lower. These predictions of that analysis.

914 Cell 143, 911–923, December 10, 2010 ª2010 Elsevier Inc.
Non-CF CF Figure 3. Porcine CF Epithelia Do Not
Hyperabsorb Na+
A Tracheal + + + C Tracheal Data are means ± SE from newborn CFTR+/+ (open
JJNa
ap-bl JJNa
bl-ap JJNa
Net bars) and CFTR / (closed bars) pigs. Numbers in
(μmol cm hr )

Jv (μl cm hr )
1.5 12
-1

-1
(6) (6) (12) parentheses indicate n; *p < 0.05.
Na flux

(12) (A and B) Isotopic 22Na+ unidirectional and net Na+


-2

-2
flux rates under basal conditions and after adding
+

+
100 mM amiloride apically. JNa +
ap bl indicates Na flux
0 from the apical (ap) to the basolateral (bl) surface,
0 +
-4 JNa
bl ap indicates flux in the opposite direction,
+
Basal Amil Basal Amil Basal Amil Basal Amil and JNanet indicates net flux. # indicates that value
B Nasal + + + Nasal in nasal epithelia differed from that in tracheal
JJNa
ap-bl JJNa
bl-ap JJNa
Net
(6)
# epithelia, p < 0.05.
(μmol cm hr )

Jv (μl cm hr )
3 12
-1

-1
(9) (C) Rate of liquid absorption (Jv) in differentiated
(10) (6)
#
#
*
primary cultures of nasal and tracheal epithelia
-2

-2
Na flux

# under basal conditions and after adding 100 mM


# amiloride apically. # indicates that value in nasal
+

# #
# 0 epithelia differed from that in tracheal epithelia,
0 -4 p < 0.05. In panels (A)–(C), the basal electrophysi-
Basal Amil Basal Amil Basal Amil Basal Amil ological properties of matched epithelia are shown
in Table S1.
D Non-CF CF
(D) Examples of light microscopic images of
tracheal epithelia. Note heterogeneity in depth of
periciliary liquid in both non-CF and CF epithelia.
(E) Examples of transmission electron microscopic
images of tracheal epithelia showing periciliary
Light
liquid.
(F) Histogram of periciliary liquid depth over
20 μm 20 μm tracheal epithelia obtained from light microscopic
images. n = 9140 non-CF and 6260 CF measure-
E ments. Multiple images were made from each of
four segments of trachea obtained from eight
non-CF and 5 CF animals. See Experimental
Procedures for additional details. Three observers
EM unaware of genotype then measured periciliary
liquid depth using a standardized protocol. A linear
mixed model and maximum likelihood estimation
5 μm 5 μm were used to calculate means and standard errors
allowing for variability between observers,
F Non-CF CF G measurements, images, segments and pigs. There
16 16 was no significant difference between periciliary
measurements

measurements
Percentage of

Percentage of

Light EM liquid depth in non-CF and CF epithelia


(p = 0.96), and the difference was 0.71 mm or less
8 8 with 95% confidence. The residual variability on
the same image had an estimated standard devia-
tion of 1.29 mm and between images was 0.60 mm.
0 0 For comparison, non-CF trachea was air-exposed
0 4 8 12 0 4 8 12 and showed a reduced height of periciliary liquid
Periciliary liquid height (μm) Periciliary liquid height (μm) (2.81 mm).
(G) Histogram of periciliary liquid depth measured
from transmission electron microscopic images.
n = 600 measurements for each genotype and 5 animals per genotype. There was no significant difference in periciliary liquid depth between non-CF and
CF, p = 0.12. For comparison the standard deviations of measurements on an image and between images were both 0.95 mm.

Vt, Isc, Gt and the Response to Amiloride under Basal smaller or no differences between CF and non-CF (Figures S2A
Conditions in Nasal Epithelia and S2D).
Transepithelial Voltage Thus, like in vivo measurements, airway location influenced
In nasal epithelia, basal Vt was greater in CF than non-CF, both in whether Vt differed between CF and non-CF epithelia.
excised and cultured epithelia (Figures 4A and 4D). DVtamiloride This similarity suggests that cultured and excised epithelia
was also greater in CF than non-CF nasal epithelia. Absolute reflect in vivo transport. However, Vt does not measure rates
values of Vt were less in excised than in vivo and cultured of ion transport.
epithelia, because of damage caused by clamping epithelia in Short-Circuit Current
Ussing chambers, i.e., ‘‘edge damage’’ (Helman and Miller, In nasal epithelia, basal Isc and the amiloride-induced reduc-
1973). Excised and cultured tracheal/bronchial epithelia showed tion in Isc (DIscamiloride) were greater in CF than non-CF

Cell 143, 911–923, December 10, 2010 ª2010 Elsevier Inc. 915
Excised tissue Non-CF CF

A B C
(25) (20)
-6 0 0 40 0
60 *

ΔGtAmil (mS/cm )
ΔIscAmil (μA/cm )
*

2
2

Gt (mS/cm )
Isc (μA/cm )
ΔVtAmil (mV)

2
2
Vt (mV)

-3
3 30 -40 20 -0.7

* *
0
* * 6
0
-80 0 -1.4
l

l
il

il
l
il
sa

sa
sa
Am

Am
Am
Ba

Ba
Ba
Culture

D E F
(34) (27)
-50 0 100 0 8 0
*

ΔGtAmil (mS/cm )
ΔIscAmil (μA/cm )
*

2
2

Gt (mS/cm )
Isc (μA/cm )
ΔVtAmil (mV)

2
2
Vt (mV)

-25 25 50 -60 4 * -3
*
0 * 50
0
* * -120 0
* -6
l
il

l
il
l
il
sa

sa
sa
Am

Am
Am
Ba

Ba
Ba

Figure 4. Amiloride Alters Electrical Properties in Non-CF and CF Nasal Epithelia


Data are means ± SE from CFTR+/+ (open symbols and bars) and CFTR / (closed symbols and bars) pigs. Numbers in parentheses indicate n; and *p < 0.05.
(A–F) Effects of adding amiloride (100 mM) to the apical solution on Vt, Isc, and Gt of freshly excised (A–C) and differentiated primary cultures (D–F) of nasal
epithelia. DVtamil, DIscamil, and DGtamil indicate changes induced by amiloride. See also Figure S2.

(Figures 4B and 4E). This was the case for both excised and the Na+ conductance. Gt is also a more direct function of
cultured epithelia. In tracheal/bronchial epithelia, basal Isc permeability than Vt or Isc, both of which are much more
did not significantly differ between CF and non-CF; DIscamiloride strongly determined by ion concentration gradients and
was greater in excised but not cultured CF epithelia (Figures membrane voltages. If CF epithelia had a greater apical Na+
S2B and S2E). conductance than non-CF epithelia, and other conductances
These results largely parallel the Vt measurements, indicating (except for the CFTR Cl- conductance) were equal, then
a strong effect of airway region on these electrical measure- DGtamiloride should have been greater in CF. That was not the
ments. Studies of excised human epithelia reported that CF case (Figures 4C and 4F). Indeed, even if apical Na+ conduc-
nasal epithelia had either higher or the same Isc values as non- tance were equal in CF and non-CF epithelia, then DGtamiloride
CF epithelia (Boucher et al., 1986; Knowles et al., 1983; Mall should have been greater in CF than non-CF epithelia
et al., 1998). (Extended Results, Note S1). Thus, finding that DGtamiloride
Transepithelial Conductance was not greater in CF epithelia suggests that Na+ conductance
In excised nasal epithelia, there was little difference between CF might be less in CF than non-CF epithelia.
and non-CF basal Gt, perhaps because the large Gt values asso- The lack of a greater DGtamiloride in CF than non-CF nasal
ciated with edge damage obscured small differences (Figure 4C). epithelia is consistent with the lack of greater Na+ absorption
However in cultured epithelia, basal Gt was greater in non-CF measured with Na+ fluxes and volume absorption. However,
epithelia (Figure 4F); this can be explained by the presence of basal Vt, DVtamiloride, basal Isc, and DIscamiloride were greater in
CFTR anion channels. Most importantly for assessing Na+ CF than non-CF nasal epithelia. Because those differences are
permeability, the amiloride-induced decrease in Gt (DGtamiloride) commonly interpreted to demonstrate that CF epithelia have
in CF did not exceed that in non-CF epithelia (Figures 4C and 4F). an increased Na+ permeability and hyperabsorb Na+ (Boucher,
Likewise, DGtamiloride of CF tracheal/bronchial epithelia did not 2007; Boucher et al., 1988; Donaldson and Boucher, 2007;
exceed that in non-CF (Figures S2C and S2F). Knowles et al., 1981), it was important to understand what
Electrical conductance is directly related to the ion perme- causes the CF/non-CF difference in electrical properties in nasal
ability of channels, and DGtamiloride is directly influenced by epithelia.

916 Cell 143, 911–923, December 10, 2010 ª2010 Elsevier Inc.
Equivalent Electrical Circuit Analyses Indicate A B
that Apical Cl Conductance Can Alter Vt, Isc, (22) (11)
2 0

ΔGtGlyH (mS/cm )
and the Response to Amiloride without a Change

2
(nonCF - CF)
(34,27)

Gt (mS/cm )
in Na+ Permeability *

2
Could loss of CFTR increase Vt, DVtamiloride, Isc, and DIscamiloride
without increasing apical Na+ permeability? A simple explana- 1 -1
tion for how this could occur arises from the fact that placing
a second ion channel (i.e., a CFTR anion conductance) in the (61,22)

apical membrane in parallel with a Na+ conductance changes 0 -2


apical membrane voltage in three ways. First, it introduces

l
l
l

sa

ea
ea
sa
another electromotive force generated by transmembrane ion

Na
Na

ch
ch

Tra
Tra
concentration gradients. Second, it introduces a conductance
that can shunt the voltage generated by other apical membrane
channels, transporters, and pumps. Third, it alters the effect on C D
apical voltage of current that is generated at the basolateral
(9)
membrane. The resulting changes in apical voltage (as well as 80
(6)
basolateral voltage) can then alter transepithelial Vt and Isc. 1

ΔI (μA/cm )
CFTR mRNA

2
(6)
Horisberger (Horisberger, 2003) developed an equivalent elec-

Relative
trical circuit model to simulate the effect of an apical membrane
* (6)

Cl conductance (CFTR) on electrical properties that are influ- 0.5


40 *
enced by ENaC-mediated Na+ conductance. He showed that
activating CFTR reduced DVtamiloride and DIscamiloride even
when Na+ conductance was held constant. He concluded that 0 0

l
l
l

sa

ea
sa

ea
a decrease in DIscamiloride or DVtamiloride upon CFTR activation

Na
Na

ch
ch
could not be interpreted to indicate a regulatory interaction

Tra
between CFTR and ENaC. Another mathematical model also Tra
showed that increasing apical Cl permeability could reduce
Na+ transport under short-circuited conditions even though Figure 5. Non-CF Nasal Epithelia Have a Larger Cl Conductance
apical Na+ permeability remained unchanged (Duszyk and Than Tracheal/Bronchial Epithelia
Data are means ± SE from nasal (cross-hatched bars) and tracheal/bronchial
French, 1991). Thus, increasing apical Cl conductance can
(shaded bars) epithelia. Amiloride (100 mM) was present on the apical surface
reduce Vt and Isc without a change in Na+ conductance. in panels (A), (B), and (D). Numbers in parentheses indicate n; *p < 0.05.
Conversely, eliminating an apical Cl conductance, as in CF, (A) Difference between Gt in cultured non-CF and CF epithelia.
can increase Vt, DVtamiloride, Isc and DIscamiloride even without (B) Change in Gt (DGtGlyH) following addition of 100 mM GlyH-101 to cultured
changing Na+ conductance. Of course, in these models, non-CF epithelia.
changes in electrophysiological properties will depend on the (C) Relative CFTR mRNA by q-RT-PCR in primary cultures of non-CF epithelia.
(D) Apical Cl- currents measured in nasal and tracheal epithelia from non-CF
absolute values of the Cl and Na+ conductances and electro-
cultured epithelia. Apical solution was Cl--free with 100 mM amiloride, 100
motive forces relative to that of all the other channels and mM DIDS, 10 mM forskolin, and 100 mM IBMX, and basolateral solution con-
transporters. tained 139.8 mM Cl . Data are current following permeabilization of basolat-
eral membrane with nystatin (0.36 mg.ml-1).
See also Figure S3.
CFTR-Mediated Cl Conductance Is Greater in Nasal
versus Tracheal/Bronchial Epithelia
Based on the equivalent circuit analysis, we reasoned that if more CFTR transcripts in cultured nasal than tracheal epithelia
nasal epithelia had a greater basal Cl conductance than (Figure 5C).
tracheal/bronchial epithelia, it might explain the CF/non-CF The data suggested that apical Cl conductance under stimu-
difference in Vt, DVtamiloride, Isc and DIscamiloride even though lated conditions was also greater in nasal than tracheal/bronchial
nasal epithelia did not hyperabsorb Na+. To further test this epithelia. First, forskolin and IBMX increased Gt approximately
possibility, we added amiloride to eliminate the Na+ conduc- twice as much in nasal as in tracheal/bronchial epithelia from
tance and then compared Gt in non-CF and CF epithelia. The normal pigs (Figures 1F and 1H). Second, adding GlyH-101 (after
difference between non-CF and CF Gt was much greater in nasal forskolin and IBMX) caused a greater Gt reduction in nasal
than tracheal epithelia (Figure 5A), indicating that nasal epithelia epithelia (Figures 1J and 1L). The Gt response to cAMP-depen-
have a greater Cl conductance under basal conditions. As an- dent stimulation and GlyH-101 inhibition showed similar trends
other test, we added amiloride to inhibit Na+ channels and DIDS in excised and cultured epithelia. Third, as an additional test of
to inhibit other Cl channels, and we then examined the apical Cl conductance, we imposed a transepithelial Cl
response to GlyH-101 (Figure 5B). GlyH-101 reduced Gt more concentration gradient, added forskolin and IBMX, permeabi-
in nasal than tracheal epithelia, indicating that nasal epithelia lized the basolateral membrane with nystatin, and measured
have a greater CFTR Cl conductance under basal conditions. Cl current (Figure 5D). Cl current was greater in nasal than
In addition, quantitative RT-PCR (q-RT-PCR) revealed relatively tracheal epithelia, indicating a greater Cl conductance. We

Cell 143, 911–923, December 10, 2010 ª2010 Elsevier Inc. 917
A Nasal Figure 6. Increased Cl Conductance Is

Basal Isc (μA/cm )


2
Associated with Reduced Basal and

ΔIscAmil (μA/cm )
-30 20 80 -80

2
Basal Vt (mV)

(22)

ΔVtAmil (mV)
Amiloride-Sensitive Vt and Isc
(A) Relationship between basal Vt, DVtamil, basal
-15 10 40 -40 Isc, and DIscamil and the change in Gt produced
by adding apical 100 mM GlyH-101 (DGtGlyH) in
0 0 0 0 the presence of amiloride. Epithelia were cultured
0 -2 -4 non-CF nasal epithelia. Each data point represents
0 -2 -4 0 -2 -4 0 -2 -4
ΔGtGlyH (mS/cm2) ΔGtGlyH (mS/cm2) ΔGtGlyH (mS/cm2) ΔGtGlyH (mS/cm2) a different epithelium. Blue lines indicate linear
regression fits to data. Correlation coefficients
and p values were: basal Vt, R = 0.831,
B Nasal Vehicle Amil F&I Amil p < 0.001; DVtamil, R = 0.592, p < 0.005; basal
Isc, R = 0.495, p < 0.02; and DIscamil, R =
100 100
0.450, p < 0.05. Spearman rank order correlation
Isc (μA/cm )

Isc (μA/cm )
2

2
was used to test statistical significance.
50 50 (B and C) Effect of 10 mM forskolin and 100 mM
IBMX (F&I) or vehicle control on basal Vt and Isc
0 0 and on changes induced by 100 mM amiloride in
cultured nasal epithelia. Panel (B) shows represen-
5 min 5 min tative experiments, and panel (C) shows means ±
SE. B, basal; *p < 0.05 versus vehicle controls.
C Nasal Vehicle F&I (D) Same as panel (C), except tracheal epithelia.
(7) (8)

ΔIscAmil (μA/cm )
-20 0 0

2
60
Isc (μA/cm )
ΔVtAmil (mV)

2
Vt (mV)

-10 * 10 30 -30
*
between these values and basal Cl
0 20 0 -60
conductance. To test this prediction, we
B F&I Amil B F&I Amil
measured Vt and Isc in nasal epithelia
before and after adding amiloride. Then,
D Tracheal Vehicle F&I to obtain an approximation of Cl
(6) (6) conductance, we measured the decrease
ΔIscAmil (μA/cm )

-30 0 0
2

60 in Gt following GlyH-101 addition


Isc (μA/cm )
ΔVtAmil (mV)

2
Vt (mV)

(DGtGlyH). We plotted these values, which


-15 10 30 -30 varied spontaneously from epithelium to
*
epithelium, and found an inverse relation-
-60
0 20 0 ship (Figure 6A).
B F&I Amil B F&I Amil
Second, further increasing apical Cl-
conductance in nasal epithelia should
reduce Vt, Isc, DVtamiloride, and DIscamiloride.
To test this prediction, we added forskolin
noticed that although nasal epithelia have a greater Cl conduc- and IBMX and found that compared to vehicle control, it
tance than tracheal/bronchial epithelia, tracheal/bronchial decreased these electrophysiological properties in non-CF nasal
epithelia have a greater Isc response to forskolin and IBMX epithelia (Figures 6B and 6C). The cAMP-induced reductions in
when studied in the presence of amiloride; this difference Isc and Vt were opposite to the increases observed when forsko-
appears to result from a greater driving force for Cl secretion lin and IBMX were added after first blocking Na+ channels with
that is generated by a greater basolateral K+ conductance amiloride (Figures 1C–1E and 1G). Because cAMP can increase
(Extended Results, Note S2, and Figure S3). Na+ transport with a slow time course (Boucher et al., 1988;
Thus, basal CFTR Cl- conductance was greater in nasal than Cullen and Welsh, 1987), we tested this possibility by adding for-
tracheal/bronchial epithelia. That result plus equivalent circuit skolin and IBMX to CF epithelia, where Cl channel activity would
analyses may explain why in nasal epithelia, Vt, DVtamiloride, Isc not confound interpretation; with this protocol, forskolin and
and DIscamiloride are greater in CF than non-CF epithelia. IBMX did not alter DIscamiloride ( 104 ± 19 mA.cm 2 after forskolin
and IBMX versus 106 ± 11 mA.cm 2 with vehicle control, n = 6
Altering Apical Cl Conductance Changes Vt, Isc, each).
DVtamiloride, and DIscamiloride Although tracheal epithelia showed little difference in electro-
Our conclusion that Cl conductance affects these electrical physiological properties between CF and non-CF, we also tested
parameters in nasal epithelia together with the equivalent circuit the effect of forskolin and IBMX in non-CF tracheal epithelia. As
analysis make four testable predictions. in nasal epithelia, increasing cAMP reduced DVtamiloride (Fig-
First, if Vt, DVtamiloride, Isc and DIscamiloride were increased in ure 6D), due largely to an increase in Gt (change in Gt with
CF compared to non-CF nasal epithelia because of a lack of vehicle +0.26 ± 0.04 mS.cm 2 and with forskolin and
Cl conductance, then there should be an inverse relationship IBMX +2.40 ± 0.25 mS.cm 2, n = 6, p < 0.001). However,

918 Cell 143, 911–923, December 10, 2010 ª2010 Elsevier Inc.
A Figure 7. A Decreased Cl Conductance
Vehicle Amil GlyH Amil Reduces the Difference between CF and
100 100 Non-CF Vt and Isc
Isc (μA/cm )

Isc (μA/cm )
2

2
Epithelia were cultured non-CF nasal epithelia.
50 50 (A and B) Effect of GlyH-101 (100 mM) on Vt and Isc
and the response to 100 mM amiloride. Panel (A)
shows representative experiments, and panel (B)
0 0 shows the mean ± SE. B, basal; *p < 0.05 versus
5 min 5 min vehicle controls.
(C and D) Effect of Cl--free apical (ap) and basolat-
eral (bl) solutions on the response to amiloride
B Vehicle GlyH
in non-CF and CF epithelia. Panel (C) shows repre-
(7) (13)

ΔIscAmil (μA/cm )
-30 0 100 0 sentative experiments in non-CF (left) and CF (right)

2
Isc (μA/cm )
ΔVtAmil (mV)

2 epithelia, and panel (D) shows means ± SE.


Vt (mV)

The two arrows for the change to Cl--free solution


-15 15 50 -50 in panel (C) indicate two exchanges of bathing
* solution.
*
0 30 0 -100
B GlyH Amil B GlyH Amil

C Non-CF CF (Figures 7C and 7D). However, in a Cl -


0Cl (ap+bl) Amil 0Cl (ap+bl) Amil and HCO3 -free solution, those values
150 150 and DVtamiloride and DIscamiloride did not
Isc (μA/cm )

Isc (μA/cm )
2

100 100 differ between genotypes.


These data further clarify how electro-
50 50 physiological measurements (increased
0 0 Vt, DVtamiloride, Isc and DIscamiloride) that
5 min 5 min are often interpreted to demonstrate
increased CF Na+ absorption may simply
D Non-CF CF reflect the lack of a Cl- conductance.
(6) (6)
ΔIscAmil (μA/cm )

0 0
2

-60 100
DISCUSSION
Isc (μA/cm )
ΔVtAmil (mV)

2
Vt (mV)

-30 30 50 -50
Advantages, Limitations, and
Considerations of This Study
0 60 0 -100
Our work has the advantage that we
B 0Cl Amil B 0Cl Amil
studied airway epithelia in vivo, in freshly
excised tissue, and in primary cultures
of differentiated airway epithelia, and we
obtained similar results. In this regard,
most studies of CF ion transport have
DIscamiloride did not change, perhaps because tracheal epithelia relied either on in vivo nasal Vt or on cultured airway epithelia
may have greater membrane driving forces for Cl- secretion or cell lines. However, the relationship between the quantitative
under short-circuit conditions (Extended Results, Note S2). and qualitative aspects of ion transport in vivo and those
These results further indicate that the electrophysiological prop- measured in cultured airway epithelia have been uncertain. In
erties are affected by factors other than just Na+ permeability. addition, chronic infection and inflammation may influence
These results may also explain two earlier studies that reported measures of ion transport in nasal Vt, in excised tissue, and
that increasing cAMP increased Isc or calculated current in perhaps in epithelia cultured from patients (Fu et al., 2007;
human nasal epithelia (Boucher et al., 1988, 1986). Gray et al., 2004; Kunzelmann et al., 2006). Thus, it is encour-
Third, decreasing apical Cl- conductance in nasal epithelia aging that our data from newborn pigs indicate that primary
should increase Vt, Isc, DVtamiloride, and DIscamiloride. Adding airway cultures retain many of the properties of in vivo and
GlyH-101 to reduce the Cl- conductance of non-CF epithelia excised airways. For example, like excised nasal epithelia,
acutely increased these properties (Figures 7A and 7B). Note cultured epithelia derived from nasal tissue had a greater CFTR
that the increase in Isc and Vt are opposite to what occurs Cl conductance than tracheal/bronchial epithelia. In addition,
when we added GlyH-101 in the presence of amiloride, which the response to interventions was also similar in vivo, in excised
eliminates the Na+ conductance (Figures 1I and 1K). tissue, and in differentiated cultures. These data suggest that
Fourth, non-CF and CF epithelia should show similar proper- cultured epithelia provide a valuable model for studying electro-
ties when Cl- conductance is eliminated by replacing Cl with lyte transport by porcine airway.
gluconate, an impermeant anion. In CF nasal epithelia, Vt and In this study, we primarily investigated CFTR-mediated anion
Isc were approximately double the values of non-CF epithelia conductance and amiloride-sensitive Na+ conductance.

Cell 143, 911–923, December 10, 2010 ª2010 Elsevier Inc. 919
However, numerous other channels and transporters may CFTR negatively regulates ENaC, and that the loss of this regu-
contribute to electrolyte transport across airway epithelia, lation in CF causes airway epithelia to hyperabsorb Na+
including SLC26 transporters, other HCO3 transporters, electri- (Boucher, 2007; Donaldson and Boucher, 2007). Although we
cally neutral Na+ transporters, K+ channels and the Na+/K+- studied electrolyte transport by airway epithelia of pigs shortly
ATPase. Ca2+-activated Cl channels are also of interest after birth, data from that time-point is germane to the issue
because it has been speculated that they might compensate because newborn CF pigs have an impaired ability to eliminate
for the loss of CFTR anion channels in CFTR / mice, thereby bacteria (Stoltz et al., 2010). Nevertheless, assaying these prop-
accounting for lack of a typical CF phenotype (Clarke et al., erties in older animals as the disease progresses will also be
1994). Although our data do not indicate whether or not these important.
other transport processes are altered by loss of CFTR, their func- Elucidating the first steps leading to CF lung disease is key if
tion remains an important area for investigation. we are to understand pathogenesis and develop mechanism-
Our conclusions also have limitations. In comparing how loss based treatments and preventions. CF pigs provide a unique
of CFTR function affects Na+ absorption in pigs and humans, we opportunity to investigate those initiating steps, because they
acknowledge that regulation of Na+ absorption might differ spontaneously develop lung disease like humans, and at birth
between the two species, i.e., human CF airway epithelia might they already manifest a bacterial host defense defect, but they
hyperabsorb Na+, whereas porcine airway epithelia do not. In do not have the secondary consequences of infection. Our
addition, although we studied newborn pigs that exhibit a bacte- studies using this model identify loss of CFTR anion permeability
rial host defense defect, it is possible that epithelial transport as the predominant transport defect at birth. In this regard,
properties differ in older animals and adults. We also studied porcine CF airway epithelia are similar to two other tissues that
CFTR / pigs, whereas most patients have at least one DF508 express both CFTR and ENaC channels, sweat gland ducts
allele (Welsh et al., 2001). We previously showed that human, and submucosal glands, where loss of anion transport and not
porcine, and murine CFTR-DF508 show some differences in pro- Na+ hyperabsorption is the CF defect (Joo et al., 2006; Quinton,
cessing (Ostedgaard et al., 2007), and thus, it should be inter- 1999, 2007). Thus, our data emphasize the role that loss of Cl
esting to learn how CFTRDF508/DF508 pigs compare to CFTR / and HCO3 permeability may play in impairing bacterial eradica-
pigs. tion and the subsequent development of airway disease.
There are additional considerations from our studies. First,
although we found that CF epithelia do not hyperabsorb Na+, EXPERIMENTAL PROCEDURES
in vivo measures of basal Vt and DVtamiloride can be valuable
For a detailed description of all the methods, please see the Extended Exper-
assays in the diagnosis of CF and for assessing the response
imental Procedures.
to interventions designed to increase CFTR activity in patients
with CF (Standaert et al., 2004). Second, our conclusions do CFTR / and CFTR+/+ Pigs
not mean that increased Na+ absorption could not occur at a later We previously reported generation of CFTR / pigs (Rogers et al., 2008a,
time-point as disease progresses or under some conditions 2008b; Stoltz et al., 2010). The University of Iowa Animal Care and Use
(Myerburg et al., 2006). Third, improving airway surface liquid Committee approved the animal studies. Animals were produced by mating
CFTR+/ male and female pigs. Newborn littermates were obtained from
hydration may benefit patients with CF (Elkins et al., 2006;
Exemplar Genetics. Animals were studied and/or euthanized 8–15 hr after birth
Donaldson et al., 2006; Robinson et al., 1997); our study did
(Euthasol, Virbac).
not address that issue. Fourth, it may seem paradoxical that
CF nasal epithelia have a greater DVtamiloride and DIscamiloride Measurement of Transepithelial Voltage In Vivo
than non-CF epithelia, and yet Na+ absorption is not increased Transepithelial voltage (Vt) was measured in the nose and trachea of newborn
in CF. As one example, consider that in non-CF epithelia studied pigs using a standard protocol as described previously (Rogers et al., 2008b;
under short-circuit conditions, adding amiloride will hyperpo- Standaert et al., 2004).
larize apical membrane voltage, thereby increasing the driving
Preparation of Differentiated Primary Cultures of Airway Epithelia
force for Cl secretion, whereas lack of CFTR precludes Cl Epithelial cells were isolated from the various tissues by enzymatic digestion,
secretion in CF epithelia. Thus, adding amiloride under short- seeded onto permeable filter supports, and grown at the air-liquid interface as
circuit conditions will inhibit Na+ absorption and increase Cl previously described (Karp et al., 2002). Differentiated epithelia were used at
secretion in non-CF epithelia, and therefore DIscamiloride will be least 14 days after seeding.
greater in CF than non-CF epithelia when apical Na+ conduc-
Electrophysiological Measurements in Freshly Excised and
tance is the same. While this is not the only factor involved in
Cultured Epithelia
determining the response to amiloride (see above), it provides Epithelial tissues were excised from the nasal turbinate and septum, and from
an example of the complexity of interpreting electrical properties trachea through 2nd-generation bronchi immediately after animals were eutha-
in assessing epithelial ion transport. nized. Tissues and cultured epithelia were studied in modified Ussing cham-
bers. Epithelia were bathed on both surfaces with solution containing (mM):
Implications for CF Pathogenesis and Treatments 135 NaCl, 2.4 K2HPO4, 0.6 KH2PO4, 1.2 CaCl2, 1.2 MgCl2, 10 dextrose,
Our data indicate that Na+ absorption is not increased in airway 5 HEPES (pH = 7.4) at 37 C and gassed with compressed air. For Cl -free
solution, Cl was replaced with gluconate and Ca2+ was increased to 5 mM.
epithelia from newborn CF compared to non-CF pigs. We also
For the high K+ and Na+-free solution, Na+ was replaced with K+. To study
explain how loss of CFTR can alter electrophysiological proper- HCO3 transport, we used Cl -free Kreb’s solution containing (mM): 118.9 Na-
ties that have been construed to indicate enhanced Na+ absorp- Gluconate, 25 NaHCO3, 2.4 K2HPO4, 0.6 KH2PO4, 5 CaGluconate, 1 MgGluc-
tion in CF. These results conflict with the widely held view that onate, and 5 dextrose and gassed with 5% CO2.

920 Cell 143, 911–923, December 10, 2010 ª2010 Elsevier Inc.
Vt was maintained at 0 mV to measure short-circuit current (Isc). Transepi- SUPPLEMENTAL INFORMATION
thelial electrical conductance (Gt) was measured by intermittently clamping Vt
to +5 and/or 5 mV. Spontaneous values of Vt were measured by transiently Supplemental Information includes Extended Results, Extended Experimental
removing the voltage clamp. At the beginning of these experiments, we used Procedures, three figures, and two tables and can be found with this article on-
cultured, non-CF tracheal epithelia to test the dose-response relationship for line at doi:10.1016/j.cell.2010.11.029.
the agents used in this study (Figure S1).
ACKNOWLEDGMENTS
Measurement of Na+ Flux and Fluid Transport
Transepithelial Na+ flux and liquid absorption were measured using methods We thank Lisaurie Lopez Rivera, Paula Ludwig, Theresa Mayhew, Peter Taft,
similar to those we previously reported (Flynn et al., 2009; Zabner et al., Jingyang Zhang, and Yuping Zhang for excellent assistance. We thank Drs.
1998). The supplemental methods describe the detailed methods. John B Stokes and Peter M Snyder for helpful discussions. GlyH-101 was
a generous gift from the Cystic Fibrosis Foundation Therapeutics and
R. Bridges. This work was supported by the National Heart Lung and Blood
Measurement of Periciliary Liquid Depth Institute (grants HL51670, HL091842, and HL097622), the National Institute
Newborn pigs (8–15 hr old) were sedated with ketamine and xylazine (15– of Diabetes and Digestive and Kidney Diseases (grant DK54759), and the
20 mg/kg and 1.5 mg/kg, IM, respectively) and immediately euthanized with Cystic Fibrosis Foundation. D.A.S. is a Parker B. Francis Fellow and was sup-
intravenous Euthasol. A 1–2 cm portion of the trachea was immediately ported by the National Institute of Allergy and Infectious Diseases (grant
removed, immersed in 2% osmium tetroxide dissolved in FC-72 perfluorocar- AI076671). M.J.W. is an Investigator of the HHMI. M.J.W. was a cofounder
bon (3M, St Paul, MN), and fixed for 90–120 min. The trachea was then rinsed of Exemplar Genetics, a company that is licensing materials and technology
in FC-72 and dehydrated in three changes of 100% ethanol, one hr each. related to this work.
During the second ethanol step, the samples were hand-trimmed into four
pieces with a scalpel to 1 mm slices. Both open ends of the tracheas were Received: July 3, 2010
removed and discarded to avoid areas possibly disturbed during removal Revised: August 31, 2010
from the animal. Tissue near the trachealis muscle was avoided. After dehydra- Accepted: November 2, 2010
tion, samples were placed in 2:1 100% ethanol:Eponate 12 resin (Ted Pella, Published: December 9, 2010
Inc., Redding, CA) followed by 1:2 100% ethanol:Eponate 12 for one hr
each. Tracheal segments were then infiltrated in 3 changes of 100% Eponate REFERENCES
12 for at least 2 hr each and polymerized for 24 hr at 60 C.
Following processing, four tissue blocks from each trachea were trimmed Azad, A.K., Rauh, R., Vermeulen, F., Jaspers, M., Korbmacher, J., Boissier, B.,
and thick-sectioned for light-level PCL thickness determination after staining Bassinet, L., Fichou, Y., des Georges, M., Stanke, F., et al. (2009). Mutations in
with Toluidine Blue. Imaging was performed on an Olympus BX-51 equipped the amiloride-sensitive epithelial sodium channel in patients with cystic
with a DP-72 CCD camera (Olympus America Inc., Center Valley, PA) using fibrosis-like disease. Hum. Mutat. 30, 1093–1103.
a 1003 NA 1.35 PlanApo lens. Five random images were taken from each
Baker, E., Jeunemaitre, X., Portal, A.J., Grimbert, P., Markandu, N., Persu, A.,
block and PCL measured using ImageJ (NIH, Bethesda, MD). PCL height
Corvol, P., and MacGregor, G. (1998). Abnormalities of nasal potential differ-
was determined by drawing a line perpendicular to the apical membrane of
ence measurement in Liddle’s syndrome. J. Clin. Invest. 102, 10–14.
the epithelial cell surface. On each image, PCL height measurements were
performed at 20 random locations. Three observers who were unaware of Bates, D., and Maechler, M. (2009). Ime4: Linear mixed-effects models using
the CFTR genotype made independent measurements on every image S4 classes. R package version. R package version 0999375-32 http://CRAN.
(number of approximate measurements per trachea: four tracheal blocks/ R-project.org/package=lme4.
animal 3 5 images/block 3 20 measurements/image 400 PCL measure- Boucher, R.C. (2007). Airway surface dehydration in cystic fibrosis: pathogen-
ments/piglet trachea/observer). Measurements were made by three indepen- esis and therapy. Annu. Rev. Med. 58, 157–170.
dent observers; therefore 1200 PCL measurements/piglet trachea were Boucher, R.C., Cotton, C.U., Gatzy, J.T., Knowles, M.R., and Yankaskas, J.R.
obtained. A linear mixed model and maximum likelihood estimation were (1988). Evidence for reduced Cl- and increased Na+ permeability in cystic
used to estimate means and standard errors (Bates and Maechler, 2009; fibrosis human primary cell cultures. J. Physiol. 405, 77–103.
R Development Core Team, 2009). The model included fixed effects for
Boucher, R.C., Stutts, M.J., Knowles, M.R., Cantley, L.C., and Gatzy, J.T.
observers and genotype and random effects for pigs, segments of the trachea
(1986). Na+ transport in cystic fibrosis respiratory epithelia. Abnormal basal
within a pig, images within a segment, and inter-observer variability of
rate and response to adenylate cyclase activation. J. Clin. Invest. 78, 1245–
measurements on the same image. Tissue blocks used for light microscopy
1252.
were also trimmed and sectioned at 80 nm for transmission electron micros-
copy. Non poststained grids were imaged in a JEOL 1230 TEM (JEOL USA Clarke, L.L., Grubb, B.R., Yankaskas, J.R., Cotton, C.U., McKenzie, A., and
Inc., Peabody, MA) equipped with a Gatan 2k 3 2k camera (Gatan Inc., Pleas- Boucher, R.C. (1994). Relationship of a non-cystic fibrosis transmembrane
anton, CA). The transmission electron microscope data were analyzed simi- conductance regulator-mediated chloride conductance to organ-level disease
larly to the light microscopic images. in Cftr(-/-) mice. Proc. Natl. Acad. Sci. USA 91, 479–483.
Cullen, J.J., and Welsh, M.J. (1987). Regulation of sodium absorption by
canine tracheal epithelium. J. Clin. Invest. 79, 73–79.
Quantitative Real-Time RT-PCR
Total RNA from excised tissue and cultures was isolated and prepared using Davies, J.C., Davies, M., McShane, D., Smith, S., Chadwick, S., Jaffe, A.,
standard techniques. Table S2 shows the PCR primers. Real-time RT-PCR Farley, R., Collins, L., Bush, A., Scallon, M., et al. (2005). Potential difference
was performed using standard methodology and analysis. measurements in the lower airway of children with and without cystic fibrosis.
Am. J. Respir. Crit. Care Med. 171, 1015–1019.
Davis, P.B. (2006). Cystic fibrosis since 1938. Am. J. Respir. Crit. Care Med.
Statistical Analysis
173, 475–482.
Data are presented as means ± standard error (SE). Spearman rank order
correlation was used to test statistical significance of relationships shown in Donaldson, S.H., Bennett, W.D., Zeman, K.L., Knowles, M.R., Tarran, R., and
Figure 6A. The methods for statistical evaluation of periciliary liquid depth Boucher, R.C. (2006). Mucus clearance and lung function in cystic fibrosis with
are described in that section of the methods. All other statistical analysis hypertonic saline. N. Engl. J. Med. 19, 241–250.
used an unpaired t test. Differences were considered statistically significant Donaldson, S.H., and Boucher, R.C. (2007). Sodium channels and cystic
at p < 0.05. fibrosis. Chest 132, 1631–1636.

Cell 143, 911–923, December 10, 2010 ª2010 Elsevier Inc. 921
Duszyk, M., and French, A.S. (1991). An analytical model of ionic movements in Matsui, H., Grubb, B.R., Tarran, R., Randell, S.H., Gatzy, J.T., Davis, C.W., and
airway epithelial cells. J. Theor. Biol. 151, 231–247. Boucher, R.C. (1998). Evidence for periciliary liquid layer depletion, not
Elkins, M.R., Robinson, M., Rose, B.R., Harbour, C., Moriarty, C.P., Marks, abnormal ion composition, in the pathogenesis of cystic fibrosis airways
G.B., Belousova, E.G., Xuan, W., and Bye, P.T.; National Hypertonic Saline disease. Cell 95, 1005–1015.
in Cystic Fibrosis (NHSCF) Study Group. (2006). A controlled trial of long- Meyerholz, D.K., Stoltz, D.A., Namati, E., Ramachandran, S., Pezzulo, A.A.,
term inhaled hypertonic saline in patients with cystic fibrosis. N. Engl. J. Smith, A.R., Rector, M.V., Suter, M.J., Kao, S., McLennan, G., et al. (2010a).
Med. 354, 229–240. Loss of CFTR function produces abnormalities in tracheal development in
Flynn, A.N., Itani, O.A., Moninger, T.O., and Welsh, M.J. (2009). Acute regula- neonatal pigs and young children. Am. J. Respir. Crit. Care Med., in press.
tion of tight junction ion selectivity in human airway epithelia. Proc. Natl. Acad. Meyerholz, D.K., Stoltz, D.A., Pezzulo, A.A., and Welsh, M.J. (2010b).
Sci. USA 106, 3591–3596. Pathology of gastrointestinal organs in a porcine model of cystic fibrosis.
Fu, Z., Bettega, K., Carroll, S., Buchholz, K.R., and Machen, T.E. (2007). Role of Am J Pathol 176 10.2353/ajpath.2010.090849.
Ca2+ in responses of airway epithelia to Pseudomonas aeruginosa, flagellin, Muanprasat, C., Sonawane, N.D., Salinas, D., Taddei, A., Galietta, L.J., and
ATP, and thapsigargin. Am. J. Physiol. Lung Cell. Mol. Physiol. 292, L353– Verkman, A.S. (2004). Discovery of glycine hydrazide pore-occluding CFTR
L364. inhibitors: mechanism, structure-activity analysis, and in vivo efficacy. J.
Gray, T., Coakley, R., Hirsh, A., Thornton, D., Kirkham, S., Koo, J.S., Burch, L., Gen. Physiol. 124, 125–137.
Boucher, R., and Nettesheim, P. (2004). Regulation of MUC5AC mucin secre- Myerburg, M.M., Butterworth, M.B., McKenna, E.E., Peters, K.W., Frizzell,
tion and airway surface liquid metabolism by IL-1beta in human bronchial R.A., Kleyman, T.R., and Pilewski, J.M. (2006). Airway surface liquid volume
epithelia. Am. J. Physiol. Lung Cell. Mol. Physiol. 286, L320–L330. regulates ENaC by altering the serine protease-protease inhibitor balance:
Griesenbach, U., Soussi, S., Larsen, M.B., Casamayor, I., Dewar, A., Rega- a mechanism for sodium hyperabsorption in cystic fibrosis. J. Biol. Chem.
mey, N., Bush, A., Shah, P.L., Davies, J.C., and Alton, E.W. (2010). Quantifica- 281, 27942–27949.
tion of periciliary fluid (PCL) height in human airway biopsies is feasible, but not Ostedgaard, L.S., Rogers, C.S., Dong, Q., Randak, C.O., Vermeer, D.W.,
suitable as a biomarker. Am. J. Respir. Cell Mol. Biol. 10.1165/rcmb.2009- Rokhlina, T., Karp, P.H., and Welsh, M.J. (2007). Processing and function of
0265OC. CFTR-DeltaF508 are species-dependent. Proc. Natl. Acad. Sci. USA 104,
Grubb, B.R., and Boucher, R.C. (1999). Pathophysiology of gene-targeted 15370–15375.
mouse models for cystic fibrosis. Physiol. Rev. 79(1, Suppl), S193–S214. Poulsen, J.H., Fischer, H., Illek, B., and Machen, T.E. (1994). Bicarbonate
Helman, S.I., and Miller, D.A. (1973). Edge damage effect on electrical conductance and pH regulatory capability of cystic fibrosis transmembrane
measurements of frog skin. Am. J. Physiol. Cell Physiol. 225, 972–977. conductance regulator. Proc. Natl. Acad. Sci. USA 91, 5340–5344.
Horisberger, J.D. (2003). ENaC-CFTR interactions: the role of electrical Quinton, P.M. (1999). Physiological basis of cystic fibrosis: a historical
coupling of ion fluxes explored in an epithelial cell model. Pflugers Arch. perspective. Physiol. Rev. 79(1, Suppl), S3–S22.
445, 522–528. Quinton, P.M. (2007). Cystic fibrosis: lessons from the sweat gland. Physiology
Huber, R., Krueger, B., Diakov, A., Korbmacher, J., Haerteis, S., Einsiedel, J., (Bethesda) 22, 212–225.
Gmeiner, P., Azad, A.K., Cuppens, H., Cassiman, J.J., et al. (2010). Functional
R Development Core Team. (2009). A language and environment for statistical
characterization of a partial loss-of-function mutation of the epithelial sodium
computing, Vol http://www.R-project.org (Vienna Austria, R Foundation for
channel (ENaC) associated with atypical cystic fibrosis. Cell. Physiol. Bio-
Statistical Computing).
chem. 25, 145–158.
Robinson, M., Hemming, A.L., Regnis, J.A., Wong, A.G., Bailey, D.L., Bauto-
Joo, N.S., Irokawa, T., Robbins, R.C., and Wine, J.J. (2006). Hyposecretion,
vich, G.J., King, M., and Bye, P.T. (1997). Effect of increasing doses of hyper-
not hyperabsorption, is the basic defect of cystic fibrosis airway glands. J.
tonic saline on mucociliary clearance in patients with cystic fibrosis. Thorax 52,
Biol. Chem. 281, 7392–7398.
900–903.
Karp, P.H., Moninger, T., Weber, S.P., Nesselhauf, T.S., Launspach, J.,
Rogers, C.S., Hao, Y., Rokhlina, T., Samuel, M., Stoltz, D.A., Li, Y., Petroff, E.,
Zabner, J., and Welsh, M.J. (2002). An in vitro model of differentiated human
Vermeer, D.W., Kabel, A.C., Yan, Z., et al. (2008a). Production of CFTR null and
airway epithelia: methods and evaluation of primary cultures. In Epithelial
DF508 heterozygous pigs by AAV-mediated gene targeting and somatic cell
Cell Culture Protocols, C. Wise, ed. (Totowa, NJ: Humana Press, Inc.),
nuclear transfer. J. Clin. Invest. 118, 1571–1577.
pp. 115–137.
Rogers, C.S., Stoltz, D.A., Meyerholz, D.K., Ostedgaard, L.S., Rokhlina, T.,
Kerem, E., Bistritzer, T., Hanukoglu, A., Hofmann, T., Zhou, Z., Bennett, W.D.,
Taft, P.J., Rogan, M.P., Pezzulo, A.A., Karp, P.H., Itani, O.A., et al. (2008b).
MacLaughlin, E., Barker, P., Nash, M., Quittell, L., et al. (1999). Pulmonary
Disruption of the CFTR gene produces a model of cystic fibrosis in newborn
epithelial sodium-channel dysfunction and excess airway liquid in pseudohy-
pigs. Science 321, 1837–1841.
poaldosteronism. N. Engl. J. Med. 341, 156–162.
Rowe, S.M., Miller, S., and Sorscher, E.J. (2005). Cystic fibrosis. N. Engl. J.
Knowles, M., Gatzy, J.T., and Boucher, R.C. (1981). Increased bioelectric
Med. 352, 1992–2001.
potential difference across respiratory epithelia in cystic fibrosis. N. Engl. J.
Med. 305, 1489–1495. Satir, P. (1963). Studies on Cilia. The Fixation of the Metachronal Wave. J. Cell
Biol. 18, 345–365.
Knowles, M.R., Stutts, M.J., Spock, A., Fischer, N., Gatzy, J.T., and Boucher,
R.C. (1983). Abnormal ion permeation through cystic fibrosis respiratory Schaedel, C., Marthinsen, L., Kristoffersson, A.C., Kornfält, R., Nilsson, K.O.,
epithelium. Science 221, 1067–1070. Orlenius, B., and Holmberg, L. (1999). Lung symptoms in pseudohypoaldos-
Kunzelmann, K., Scheidt, K., Scharf, B., Ousingsawat, J., Schreiber, R., Wain- teronism type 1 are associated with deficiency of the alpha-subunit of the
wright, B., and McMorran, B. (2006). Flagellin of Pseudomonas aeruginosa epithelial sodium channel. J. Pediatr. 135, 739–745.
inhibits Na+ transport in airway epithelia. FASEB J. 20, 545–546. Sheppard, D.N., and Welsh, M.J. (1999). Structure and function of the CFTR
Mall, M., Bleich, M., Greger, R., Schreiber, R., and Kunzelmann, K. (1998). The chloride channel. Physiol. Rev. 79(1, Suppl), S23–S45.
amiloride-inhibitable Na+ conductance is reduced by the cystic fibrosis trans- Sheridan, M.B., Fong, P., Groman, J.D., Conrad, C., Flume, P., Diaz, R., Harris,
membrane conductance regulator in normal but not in cystic fibrosis airways. C., Knowles, M., and Cutting, G.R. (2005). Mutations in the beta-subunit of the
J. Clin. Invest. 102, 15–21. epithelial Na+ channel in patients with a cystic fibrosis-like syndrome. Hum.
Mall, M., Grubb, B.R., Harkema, J.R., O’Neal, W.K., and Boucher, R.C. (2004). Mol. Genet. 14, 3493–3498.
Increased airway epithelial Na+ absorption produces cystic fibrosis-like lung Sims, D.E., and Horne, M.M. (1997). Heterogeneity of the composition and
disease in mice. Nat. Med. 10, 487–493. thickness of tracheal mucus in rats. Am. J. Physiol. 273, L1036–L1041.

922 Cell 143, 911–923, December 10, 2010 ª2010 Elsevier Inc.
Smith, J.J., and Welsh, M.J. (1992). cAMP stimulates bicarbonate secretion Welsh, M.J., Ramsey, B.W., Accurso, F., and Cutting, G.R. (2001). Cystic
across normal, but not cystic fibrosis airway epithelia. J. Clin. Invest. 89, Fibrosis. In The Metabolic and Molecular Basis of Inherited Disease, C.R.
1148–1153. Scriver, A.L. Beaudet, W.S. Sly, D. Valle, B. Childs, and B. Vogelstein, eds.
Standaert, T.A., Boitano, L., Emerson, J., Milgram, L.J., Konstan, M.W., (New York: McGraw-Hill), pp. 5121–5189.
Hunter, J., Berclaz, P.Y., Brass, L., Zeitlin, P.L., Hammond, K., et al. (2004). Widdicombe, J.H., Welsh, M.J., and Finkbeiner, W.E. (1985). Cystic fibrosis
Standardized procedure for measurement of nasal potential difference: an decreases the apical membrane chloride permeability of monolayers cultured
outcome measure in multicenter cystic fibrosis clinical trials. Pediatr. Pulmo- from cells of tracheal epithelium. Proc. Natl. Acad. Sci. USA 82, 6167–6171.
nol. 37, 385–392.
Willumsen, N.J., and Boucher, R.C. (1991a). Sodium transport and intracellular
Stoltz, D.A., Meyerholz, D.K., Pezzulo, A.A., Ramachandran, S., Rogan, M.P., sodium activity in cultured human nasal epithelium. Am. J. Physiol. 261, C319–
Davis, G.J., Hanfland, R.A., Wohlford-Lenane, C.L., Dohrn, C.L., Bartlett, J.A., C331.
et al. (2010). Cystic fibrosis pigs develop lung disease and exhibit defective
bacterial eradication at birth. Science Translational Medicine 2, 29ra31. Willumsen, N.J., and Boucher, R.C. (1991b). Transcellular sodium transport in
cultured cystic fibrosis human nasal epithelium. Am. J. Physiol. 261, C332–
Van Goor, F., Hadida, S., Grootenhuis, P.D., Burton, B., Cao, D., Neuberger,
C341.
T., Turnbull, A., Singh, A., Joubran, J., Hazlewood, A., et al. (2009). Rescue
of CF airway epithelial cell function in vitro by a CFTR potentiator, VX-770. Wine, J.J. (1999). The genesis of cystic fibrosis lung disease. J. Clin. Invest.
Proc. Natl. Acad. Sci. USA 106, 18825–18830. 103, 309–312.
Verkman, A.S., Song, Y., and Thiagarajah, J.R. (2003). Role of airway surface Zabner, J., Smith, J.J., Karp, P.H., Widdicombe, J.H., and Welsh, M.J. (1998).
liquid and submucosal glands in cystic fibrosis lung disease. Am. J. Physiol. Loss of CFTR chloride channels alters salt absorption by cystic fibrosis airway
Cell Physiol. 284, C2–C15. epithelia in vitro. Mol. Cell 2, 397–403.

Cell 143, 911–923, December 10, 2010 ª2010 Elsevier Inc. 923
Sister Cohesion and Structural Axis
Components Mediate Homolog Bias
of Meiotic Recombination
Keun P. Kim,1 Beth M. Weiner,1 Liangran Zhang,1 Amy Jordan,1 Job Dekker,1,2 and Nancy Kleckner1,*
1Department of Molecular and Cellular Biology, Harvard University, 52 Oxford Street, Cambridge, MA 02138, USA
2Program in Gene Function and Expression and Department of Biochemistry and Molecular Pharmacology, University of Massachusetts
Medical School, 364 Plantation Street, Worcester, MA 01655, USA
*Correspondence: kleckner@fas.harvard.edu
DOI 10.1016/j.cell.2010.11.015

SUMMARY arms, create connections that direct homolog segregation at


Meiosis I (MI) (Figure 1B).
Meiotic double-strand break (DSB)-initiated recom- Meiotic recombination initiates after DNA replication. Thus,
bination must occur between homologous maternal sister chromatids are present throughout. Nonetheless, in accord
and paternal chromosomes (‘‘homolog bias’’), even with its roles for IH interactions, this recombination usually occurs
though sister chromatids are present. Through phys- between two homolog chromatids rather than between sisters
ical recombination analyses, we show that sister (homolog bias; Figure 1C; Zickler and Kleckner, 1999; Hunter,
2006). In contrast, recombinational repair of DNA damage in the
cohesion, normally mediated by meiotic cohesin
mitotic cycle occurs preferentially between sister chromatids
Rec8, promotes ‘‘sister bias’’; that meiosis-specific
(sister bias), thus minimizing collateral damage (Bzymek et al.,
axis components Red1/Mek1kinase counteract 2010).
this effect, thereby satisfying an essential precondi- In both situations, partner bias is specifically programmed,
tion for homolog bias; and that other components, with chromosome structure components playing central roles.
probably recombinosome-related, directly ensure During mitotic repair, the sister may be favored partly because
homolog partner selection. Later, Rec8 acts posi- it is nearby; however, this intrinsic tendency is reinforced by
tively to ensure maintenance of bias. These complex- sister chromatid cohesins (e.g., Covo et al., 2010; Heidinger-
ities mirror opposing dictates for global sister Pauli et al., 2010). During meiosis, recombination occurs in the
cohesion versus local separation and differentiation context of tightly conjoined sister chromatid structural axes,
of sisters at recombination sites. Our findings support which are implicated in many effects, including partner choice.
These axes comprise co-oriented linear arrays of loops whose
DSB formation within axis-tethered recombinosomes
bases are AT-rich ‘‘axis association sites’’ that preferentially
containing both sisters and ensuing programmed bind specific proteins (Figure 1D; Blat et al., 2002; Kleckner,
sequential release of ‘‘first’’ and ‘‘second’’ DSB 2006). Recombinosomes bind directly to regions between these
ends. First-end release would create a homology- sites and are associated with axes via tethered-loop axis
searching ‘‘tentacle.’’ Rec8 and Red1/Mek1 also complexes (Figure 1E; Blat et al., 2002). In budding yeast, and
independently license recombinational progression similarly in other organisms, homolog bias requires two interact-
and abundantly localize to different domains. These ing meiosis-specific axis components, Red1 and Hop1, plus
domains could comprise complementary environ- their associated Rad53-related kinase Mek1 (Figure 1D; Schwa-
ments that integrate inputs from DSB repair and cha and Kleckner, 1994, 1997; Niu et al., 2005, 2007; Latypov
mitotic chromosome morphogenesis into the et al., 2010; Terentyev et al., 2010; Goldfarb and Lichten, 2010;
complete meiotic program. Martinez-Perez and Villeneuve, 2005; Sanchez-Moran et al.,
2007; Wu et al., 2010; Lao and Hunter, 2010).
Meiotic homolog bias is established very early (Hunter, 2006).
Recombination initiates via programmed DSBs whose 50 termini
INTRODUCTION are rapidly resected, giving 30 single-stranded (ss) DNA tails. A
‘‘first’’ DSB end then contacts a homolog partner chromatid,
Meiosis involves a complex program of interhomolog (IH) e.g., via a nascent D-loop (Figure 1C). The ‘‘second’’ DSB end
interactions mediated by DNA recombination. Recombination probably remains associated with its donor chromosome via
directs homolog pairing, promoting both homology recognition interaction with its sister, yielding an ‘‘ends-apart’’ configuration,
and physical juxtaposition of whole chromosomes in space also seen cytologically (Figure 1A). Homolog bias persists
(Figure 1A; Storlazzi et al., 2010). Later, recombination-gener- thereafter. A few nascent D-loop interactions are designated
ated crossovers (COs), plus cohesion along sister chromatid for maturation into IH crossover (IH-CO) products. COs arise

924 Cell 143, 924–937, December 10, 2010 ª2010 Elsevier Inc.
Thus, at all sites, sister cohesion must be locally compromised.
(2) CO at the DNA level is accompanied by exchange at the struc-
tural (axis) level (‘‘axis exchange’’; Kleckner, 2006; Figure 1B).
Thus, at CO sites, but not NCO sites, sisters must be locally differ-
entiated and separated at both the DNA and axis levels (Blat et al.,
2002). In fact, Rec8 is specifically absent at chiasmata (Eijpe
et al., 2003), and local separation is seen at CO sites while recom-
bination is in progress during prophase (Storlazzi et al., 2008).
However, despite these local modulations, sister cohesion
must concomitantly be maintained globally along chromosome
arms to enable regular homolog pairing at prophase and regular
segregation at MI (Figure 1B). Thus, meiotic chromosomes face
conflicting demands for global cohesion maintenance versus
local weakening of cohesion at recombination sites.
Results presented below define distinct, but integrated, roles
for Rec8/cohesion and Red1/Mek1kinase in homolog bias, sister
cohesion, and recombination timing and/or kinetics; present
evidence for association of recombinosomes with developing
chromosome axes before DSB formation; and show that Red1
and Rec8 localize to different chromosomal domains on a per-
cell basis. Multiple general implications emerge.
Figure 1. Meiotic Interhomolog Interactions
(A) Top: Presynaptic alignment of homolog axes (Sordaria image by D. Zickler).
RESULTS
Bottom: Coaligned axes exhibit matched pairs of DSB-associated Mer3
complexes in an ends-apart configuration (Storlazzi et al., 2010).
(B) Homologs are connected by COs between homologs plus global sister Physical Analysis of Recombination
connections along chromosome arms (chiasmata from Jones and Franklin, Recombination intermediates and products were analyzed at the
2006). Note local sister separation at chiasmata. HIS4LEU2 hot spot (Figures 2A–2D; Hunter and Kleckner, 2001;
(C) Meiotic recombination between one sister of each homolog (Hunter, 2006). Oh et al., 2007). In cultures undergoing synchronous meiosis,
Purple and green bars indicate proposed sister cohesion near DSBs.
samples were taken at desired time points and subjected to
(D) Co-oriented sister linear loop array.
(E) Recombining DNAs in chromatin loops are tethered to axes via axis/recom-
DNA extraction, restriction digestion, and 1D and 2D gel electro-
binosome (purple ball) contacts in ‘‘tethered-loop axis complexes’’ (Blat et al., phoresis. Species of interest were detected by Southern blotting
2002). (Probe 4; except as noted). DSBs, SEIs, and dHJs are detected in
2D gels, which separate species first by molecular weight (MW)
via single-end invasions (IH-SEIs) and double Holliday junctions and then by shape. IH-COs and -NCOs are detected via diag-
(IH-dHJs). Remaining interactions are mostly resolved as IH nostic fragments in 1D gels. In wild-type (WT) meiosis, intermedi-
noncrossover products (IH-NCOs) via other intermediates. ates appear and disappear and products emerge (Figure 2E).
Here, we further define roles of meiotic chromosome structure Recombination in the absence of Rec8 and/or Red1 or, anal-
components for homolog bias, other recombination aspects, ogously, Rec8 and/or Mek1kinase was examined in two isogenic
and chromosome morphogenesis. Of special interest is Rec8, sets of WT, single- and double-mutant strains. Alleles were
a meiosis-specific homolog of general kleisin cohesin Mcd1/ complete deletion mutations (rec8D, red1D) or mek1as, which
Scc1/Rad21 (hereafter Mcd1). Rec8 occurs abundantly along encodes a mutant protein whose kinase activity can be abol-
conjoined sister axes (Klein et al., 1999) and, in yeast, is the ished by a chemical inhibitor (Niu et al., 2005). mek1as(IN)
only other known meiosis-specific axis component besides and mek1as(+IN) denote absence or presence of inhibitor added
Red1/Hop1/Mek1. Sister cohesion, thus Rec8, is expected a pri- at t = 0, respectively. Time courses were performed for all strains
ori to play a role in homolog-versus-sister partner discrimination. at both 33 C and 30 C with samples taken at t = 0, 2, 3, 4, 5, 6, 7,
Two opposite models could be envisioned. (Model 1) Tight 8, 10, and 24 hr after initiation of meiosis. The same patterns
conjunction of sister axes might block a DSB from interacting occur at both temperatures; 33 C data are shown to permit
with its sister, thus forcing use of a homolog partner by default; optimal comparison with zmm mutants (Börner et al., 2004;
Red1/Hop1/Mek1 would exert their effects by promoting such below). Each strain, at each temperature, was examined in
sister axis conjunction (Niu et al., 2005, 2007; Thompson and multiple independent time courses (n = 53) with highly consistent
Stahl, 1999; Bailis and Roeder, 1998). (Model 2) Rec8-mediated results (Figure S1A available online).
reinforcement of sister cohesion might favor intersister (IS) All mutants have reduced DSB levels (below) and thus
recombination, as during mitotic repair, thereby inhibiting use reduced total recombinational interactions. To permit direct
of the homolog. Cohesion would then be locally modulated for comparisons among all strains with regard to post-DSB effects,
use of the homolog to predominate during meiosis. we normalized levels of all species shown in graphs such that
In support of the second possibility, two features of recombina- they are presented on a per DSB basis. Specifically, for all
tion intrinsically require local loosening of sister relationships. (1) mutants, levels of all species are increased to those predicted
Recombination occurs between one chromatid of each homolog. if DSB levels would be the same as in WT.

Cell 143, 924–937, December 10, 2010 ª2010 Elsevier Inc. 925
Figure 2. Physical Analysis of Meiotic
Recombination
(A) HIS4LEU2 locus (Martini et al., 2006) and
Southern blot probes.
(B) DNA species generated by indicated digests.
(C) Fragments diagnostic of IH-COs and IH-NCOs,
each representing a subset of total products
(Storlazzi et al., 1995).
(D) Top: Two-dimensional gel displaying parental
and intermediate species (B, plus MCJMs [Oh
et al., 2007]). Bottom: Illustration. IH/IS species in
blue and pink, respectively (B, and species
described in text).
(E) Recombination in WT meiosis (S = IH+IS). See
also Figure S1.

Homolog Bias in WT
CO-fated interactions yield IH-dHJs plus
two types of IS-dHJs as seen in 2D gels
(Schwacha and Kleckner, 1994, 1997;
Figure 2D). The ratio of IH-dHJs to
IS-dHJs (summed from both parents) is
5:1 in WT and mek1as(IN) (Figure 4B,
Figure S1B, Figure S2, Figure S3, and
Figure S4), reflecting homolog bias for
CO recombination. Homolog bias is also
robust for NCOs: at HIS4LEU2, total IH
events (COs plus NCOs), account for
90% of total DSBs (Martini et al., 2006;
N. Hunter, personal communication).

In the Absence of Red1/


Mek1kinase, Homolog Bias Is
Converted to Sister Bias
In red1D and mek1as(+IN), total dHJ
levels (IH+IS) are the same as in WT/
mek1as(IN). However, in both mutants,
DSB Formation and Resection IH-dHJs are strongly reduced while IS-dHJs are compensatorily
DSBs were assayed in rec8D and/or red1D with a background increased, yielding an IH:IS dHJ ratio of 1:10 (versus 5:1 in WT)
where DSBs accumulate rather than turning over (rad50S; (Figures 4A–4D). Absolute IH-CO levels are also strongly
Figures 3A and 3B). At HIS4LEU2, each single mutant exhibits reduced in both mutants, as are IH-NCO levels (Figure 4D).
modestly reduced DSB levels. The double mutant exhibits These findings, plus prior findings (Introduction), point to
approximately the product of the two individual defects. a general defect in homolog bias at an early step in recombina-
Thus, Rec8 is required for DSB formation, similar to, but largely tion, prior to CO/NCO differentiation, with consequences for
independent of, Red1. DSB deficits occur in rec8D at three both branches. This constellation of mutant phenotypes is
other DSB hot spots (A.J., unpublished data), as for red1D at defined as ‘‘Type I’’ (Figure 4C). It is interpreted as reflecting roles
the same sites (Blat et al., 2002), and for rec8D genome- for Red1 and Mek1kinase in ‘‘establishment’’ of homolog bias.
wide (Kugou et al., 2009). mek1as(+IN) confers the same Thus, in WT meiosis, Red1/Mek1kinase converts sister bias
reduction in HIS4LEU2 DSBs as red1D (K.P.K., unpublished into homolog bias at an early step.
data).
WT and mek1as(IN) DSBs exhibit 500 nt 30 single-stranded Homolog Bias Is Detectable at the SEI Stage
(ss) DNA tails (Hunter, 2006), sensitively revealed by 2D gels Previous studies identified IH-SEIs (Hunter and Kleckner, 2001).
(Figure 3C). rec8D and rec8D mek1as(IN) exhibit modest IS-SEI signals were not identified. In red1D and mek1as(+IN),
hyperresection; red1D and mek1as(+IN) exhibit dramatic hyper- where IH interactions are strongly reduced and IS interactions
resection; double mutants exhibit more hyperresection than are strongly increased, IH-SEI signals are not visible; however,
either component single mutant (Figure 3C). Thus, Rec8 and in the ‘‘SEI’’ region of the gel (Figure 2D), two arc signals are
Red1/Mek1kinase each contribute to control of DSB end resec- prominent (Figure 4A and Figure 5A). These signals correspond
tion via distinct effects. to Mom-Mom and Dad-Dad IS-SEI species. (1) The centers of

926 Cell 143, 924–937, December 10, 2010 ª2010 Elsevier Inc.
Figure S5). (2) IS-SEIs appear and disappear with the same
kinetics as IH-SEIs, qualitatively and quantitatively (red1D/
mek1as(+IN) versus WT/mek1as(IN) in Figure 4D and Figure S3;
WT/mek1as(IN) gels in Figure S2 and Figure S4). (3) In Red1/
Mek1kinase strains, where IS-dHJs occur at the same high
levels as IH-dHJs in WT meiosis, there are no other detectable
species in the MW region of a 2D gel where SEIs should appear;
moreover, the IS-SEI levels in these mutants are the same as for
IH-SEIs in WT. Thus, the arc morphology of IS-SEIs suggests
that the 30 end status of CO-fated IS-SEIs is intrinsically less
stringently controlled than that of CO-fated IH-SEIs.

In the Absence of Rec8, Homolog Bias Is Established,


Then Lost, during CO Formation at the SEI-to-dHJ
Transition
In rec8D and mek1as(IN) rec8D, DSBs, SEIs and dHJs appear
and disappear, and IH-CO and IH-NCO products appear, all at
Figure 3. DSB Levels and Resection
substantial levels (Figure 4D and Figure S3 legend). IH-NCO
(A) One-dimensional gel showing rad50S DSBs.
(B) Quantification of DSB levels in (A). levels are very similar to those in WT/mek1as(IN) strains, sug-
(C) Two-dimensional gel detection of DSB resection: illustration plus WT and gesting that homolog bias is established normally for NCO
mutant data from time point of maximum abundance. recombination (Figure 4D and Figure S3). Further, just as in
WT/mek1as(IN), IH-SEIs are more abundant than IS-SEIs
(Figures 5B and 5C). Thus, homolog bias is established efficiently
mass of the two signals occur at the expected MW positions, also for CO recombination.
9.2 and 7.3 kb (Figure 5A). (2) Hybridization with homolog- However, the ratio of IH:IS dHJs in both Rec8 strains is 1:1
specific probes shows that each signal contains only material (versus 5:1 in WT), and the IH-CO level, while high, is modestly
from the appropriate parent (Figure 5A). (3) The two signals reduced (Figures 4A–4D). Such effects could be explained in
appear and disappear, coordinately, with the same kinetics as two ways. (1) IH-SEIs might be lost to unknown fates, thus
IH-SEIs in WT strains (Figure 4D). (4) The two arc species are specifically reducing the level of IH-dHJs and IH-COs. (2)
not DNA replication intermediates: they appear 2 hr after Homolog bias might be lost at the SEI-to-dHJ transition, with
completion of replication (e.g., below); further, replication inter- all SEIs progressing, but with each SEI having an equivalent
mediates are not recovered in the DNA extraction procedure probability of giving rise to either an IH-dHJ or an IS-dHJ (IH:IS
used (Hunter and Kleckner, 2001). dHJ = 1:1) and a commensurate reduction in IH-COs. We favor
The same IS-SEI arcs are also detectable in WT and mek1as the second scenario. In rec8D mek1as(IN), total dHJ levels
(IN) (Figures 5B and 5C). IH-SEIs form prominent bar signals are very similar to those in REC8 mek1as(IN); however, the
that hybridize to both Mom- and Dad-specific probes. IH-SEIs level of IH-dHJs is reduced while the level of IS-dHJs is compen-
are detectable by the presence of weak signal in flanking regions satorily increased (Figure 4D). Thus, SEIs progress efficiently to
corresponding, respectively, to the higher MW portion of Mom- dHJs but are concomitantly redistributed between IH and IS
Mom IS-SEIs and the lower MW portion of Dad-Dad IS-SEIs species.
(Figures 5B and 5C, arrows within circles). Each signal migrates In scenario (1), differential loss of IH-SEIs to the same level as
with appropriate mobility, is detected only with the appropriate IS-SEIs predicts that IH-COs will be reduced to 20% the WT
homolog-specific probe, and is rarer than IH-SEIs as expected level; in scenario (2) equi-partitioning of SEIs to IH- and IS-dHJs
from homolog bias. Other portions of IS-SEI arcs overlap IH- predicts that IH-CO levels will be reduced to 60% the WT level
SEI bars. These patterns are confirmed in Rec8 strains (Figures (Figure S3). In rec8D mek1as(IN), IH-COs occur at 60% the
5B and 5C). WT level (Figure 4D).
The unique arc shape of IS-SEI signals is seen in WT, as well as The IH:IS dHJ ratio in Rec8 mutants is exactly 1:1 (Figure 4B;
Red1/Mek1kinase. Thus it is not mutant-specific but is char- 1.04 ± 0.14; range = 0.831.25; n = 12). It seems improbable that
acteristic of IS (versus IH) interactions per se. Each arc spans equivalency would arise by chance as in (1) and probable that it
MWs both higher and lower than expected (Figures 5A and reflects an intrinsic feature of recombination as in (2) (Discus-
5B). Lower MW material is explained by DSB hyperresection, sion). Also, random interaction of a DSB with available partners
prominent in the mutants but discernible at a low level in WT/ would give a 2:1 IH:IS dHJ ratio; thus, it is not the case that
mek1as(IN) (Figure 3C). Higher MW material implies occur- a DSB has access to all possible partner chromatids (two sisters
rence of DNA synthesis, presumably to extend 30 strand termini. and one homolog) at the SEI-to-dHJ transition.
Despite their unusual morphology, these species clearly The Rec8 partner choice phenotype is defined as ‘‘Type II’’
represent CO-designated IS-SEIs. (1) In a strain specifically (Figure 4C). It is interpreted to mean that homolog bias is: (1) effi-
defective for CO recombination versus NCO recombination, ciently established; (2) efficiently maintained both throughout
IS-SEI levels are coordinately reduced, with the same altered NCO formation (giving normal IH-NCO levels) and during CO
variation over time, as all known CO-specific species (zip3D; formation through the SEI stage (giving normal IH bias for

Cell 143, 924–937, December 10, 2010 ª2010 Elsevier Inc. 927
Figure 4. Partner Choice in Chromosome
Structure Mutants
(A) Gels of SEIs/dHJs at time point of maximum
level in (B). Blue indicates IH; Pink indicates IS.
** indicates SEI levels too low for accurate IH/IS
discrimination.
(B) IH/IS dHJ levels over time plotted as percent-
age maximum level of most abundant species.
(C) Summary of data in (A, B, and D) and thus-
defined Type I and Type II phenotypes.
(D) Time course analysis of mek1as strain set
displayed as pair-wise comparisons between
featured strain (solid line) and appropriate refer-
ence strain (dashed line). All species levels in
mutants are normalized for DSB reductions to
permit per DSB comparisons (Results). Gels are
presented without such adjustment with parental
signals at the same intensities in all panels to indi-
cate absolute levels. Corresponding full gels are
shown in Figure S2. Analogous data for MEK1 ±
red1D strains in Figure S3 and Figure S4. Note,
in rec8D, as well as in rec8D mek1as(IN), nearly
all DSBs progress to products, albeit with a signif-
icant delay (Figure S3 legend). See also Figure S2,
Figure S3, Figure S4, and Figure S5.

Rec8 Promotes Sister Bias and


Red1/Mek1 Antagonizes that
Effect, Thus Making Homolog
Bias Possible
rec8D mek1as(+IN) and rec8D red1D
double mutants exhibit the same pheno-
type as rec8D mek1as(IN) and rec8D
RED1: IH:IS dHJ = 1:1; WT levels of IH-
NCOs; and IH-COs reduced to 60%
the WT level (Figure 4). IH/IS SEI status
cannot be assessed because levels
are too low, reflecting reduced total
DSBs (above) and rapid turnover of inter-
mediates (below). Nonetheless, since all
other predicted phenotypes are observed,
we conclude that in Rec8 Red1/
Mek1kinase double mutants, as in
SEIs); but (3) lost at the SEI-to-dHJ transition, with all SEIs Rec8 single mutants, homolog bias is established normally, but
(IH and IS) progressing efficiently but with either type of SEI is not maintained during CO recombination (Type II; Figure 4C).
having an equal probability of giving either an IH- or IS-dHJ This correspondence is confirmed by inactivating Mek1kinase in
(IH:IS dHJ = 1:1) and corresponding products, giving a 40% rec8D mek1as strain at various times in meiosis: a 1:1 IH:IS dHJ
reduction in IH-COs to 60% the WT level. ratio is seen regardless of whether inhibitor is added at t = 0
This interpretation is supported by comparison of rec8D with (Rec8 Mek1kinase condition), t = 7h (Rec8 Mek1kinase +
zip3D (Figure S5). Zip3 represents a prominent group of CO- condition), or any point in between (K.P.K., unpublished data).
specific functions (ZMMs; Börner et al., 2004). Differently from These results were unexpected. Absent further complexities,
rec8D, zip3D: (1) shows defective progression of DSBs to CO- a double mutant should have exhibited the earlier establishment
specific intermediates and a severe reduction in IH-COs; (2) defect of Red1/Mek1kinase (Type I), not the later ‘‘mainte-
exhibits this defect at the DSB-to-SEI transition; and (3) does nance’’ defect of Rec8 (Type II). Several features are thus
not eliminate homolog bias among residual SEIs and dHJs revealed:
(IH:IS dHJ = 3:1). rec8D zip3D exhibits the sum of both single-
mutant defects: severe reductions in SEIs, dHJs, and IH-COs (1) Homolog bias is established even when both Red1/
(zip3D); robust homolog bias at the SEI stage and for NCO Mek1kinase and Rec8 are absent (in double mutants);
recombination (both mutants); and IH:IS dHJ = 1:1 (rec8D). thus, other components directly mediate this process.

928 Cell 143, 924–937, December 10, 2010 ª2010 Elsevier Inc.
(2) Red1/Mek1kinase is important for establishment of
homolog bias when Rec8 is present (Red1/
Mek1kinase single mutants) but not when Rec8 is
absent (double mutants). Thus, formally, Rec8 specifies
an inhibitor of bias and Red1/Mek1kinase is required to
remove that inhibitor. In Rec8 strains, there is no inhib-
itor of homolog bias; thus, homolog bias is established,
regardless of whether the inhibitor of the inhibitor is
present (Rec8 Red1+/Mek1kinase+) or absent (Rec8
Red1/Mek1kinase).
(3) When Rec8 is present and Red1/Mek1kinase is absent,
sister bias is observed (above). Thus, in its inhibitory
role, Rec8 mediates sister bias, concomitantly precluding
establishment of homolog bias. Red1/Mek1kinase coun-
teracts these effects, converting sister bias back to
homolog bias.
(4) Maintenance of bias during CO recombination is defec-
tive in both Rec8 and Rec8 Red1/Mek1kinase.
Red1/Mek1kinase might be irrelevant for bias mainte-
nance. Alternatively, Red1/Mek1kinase may also be
required for maintenance of bias, in addition to Rec8,
with both functions being essential for the same step. If
so, a bias maintenance defect would be observed also
in Red1/Mek1kinase single mutants. Supporting this
model: residual IH products arising in those mutants
exhibit the same differential reduction of COs versus
NCOs, by 60%, as Rec8.

Meiotically Expressed Mcd1 Fully Substitutes for Rec8


during Establishment of Homolog Bias
The general kleisin ortholog of Rec8, Mcd1, is not prominent in
meiosis but can be expressed meiotically from the REC8
promoter (pREC8-MCD1) (Lee and Amon, 2003). Expression of
Mcd1 in Rec8 Red1/Mek1kinase double mutants fully
restores a Rec8+ Red1/Mek1kinase phenotype. That is,
expression of Mcd1 converts the double-mutant Type II pheno-
type back to the Type I phenotype of the single mutant (Figure 4).
Thus, Mcd1 fully substitutes for Rec8 as an inhibitor of homolog
bias establishment and concomitant promoter of sister bias.
Also, expression of Mcd1 in Rec8 Red1+/Mek1kinase+ single
mutants has no effect on establishment of bias: IH-NCOs still
occur at WT-like levels and substantial levels of IH-COs also
occur (K.P.K., unpublished data). Thus, the inhibitory effects of
Mcd1 are efficiently counteracted by Red1/Mek1kinase, just as
for Rec8.
Expression of Mcd1 in Rec8 Red1+/Mek1kinase+ single
Figure 5. Identification of IS-SEIs
mutants increases the IH:IS dHJ ratio from 1:1 to 2:1, but not
(A) dHJs/SEIs from mek1as(+IN) visualized with general and Mom- and Dad-
specific probes (green, orange, and brown; Figure 2A); predicted species sizes to the 5:1 observed in WT (K.P.K., unpublished data). This prob-
from Figure 2B are indicated. * marks IS-SEI. ably implies that Mcd1 can substitute only partially for Rec8
(B) dHJs/SEIs from WT and mutants visualized with Mom- and Dad-specific during maintenance of bias during CO recombination.
probes. Gel regions (bottom); (top) subset of illustration including regions
expanded in (C). Arrows indicate regions of IS-SEI signals visible in WT/rec8D.
(C) Enlarged views of gel areas indicated in (B) subset of illustration; circles
denote regions of differential Mom/Dad hybridization. parameters are denoted, for DSBs, SEIs, and dHJs, by the length, beginning,
(D) Timing and kinetics of recombination in indicated strains. For any interme- and end, respectively, of a corresponding line. Times at which IH-CO and
diate species of interest, integration of the primary data (e.g., Figure 4D) yields IH-NCO products have appeared in 50% of cells (i.e., at half their final level)
three parameters: average life span; time of appearance in 50% of cells; and shown by corresponding flags. Analogous data for MEK1± red1D strain set
time of disappearance (one life span later) (Hunter and Kleckner, 2001). These in Figure S6. See also Figure S2, Figure S4, and Figure S5.

Cell 143, 924–937, December 10, 2010 ª2010 Elsevier Inc. 929
Figure 6. Sister Cohesion and Axis Morphogenesis
(A) Strains carrying lacO and/or tetO array(s) and expressing a cognate fluorescently-tagged Lac and/or Tet repressor were analyzed for sister association in fixed
whole cells. One focus indicates unreplicated, or replicated but unseparated, sisters (upper left). Two foci indicate replicated and visibly distinct sisters (other
panels). The scale bar represents 1mm.
(B) Percentages of cells in representative cultures showing 4C DNA content (black), visibly distinct sisters at a single locus as in (A) (red), or first or both meiotic
divisions (grey).

930 Cell 143, 924–937, December 10, 2010 ª2010 Elsevier Inc.
Red1/Mek1kinase and Rec8 Regulate Progression Rec8 and Red1 Are Both Required for Normal Sister
of Recombination Cohesion
In a given strain, the time at which a given species appears in Sister relationships were examined in intact cells with fluores-
50% of cells, its duration (life span), and the time at which it cent repressor-operator arrays at two loci, each located in the
disappears in 50% of cells (one life span after it appears) can middle of a long chromosome arm and present on one homolog
all be defined (Figure 5D and Figure S6). All mutants exhibit of a diploid (Figure 6A and Figure S7). In WT, cohesion is main-
altered timing and/or kinetics of recombination. tained throughout prophase: separated sister loci (two-focus
Lines 1 versus 2 cells) appear at MI (Figure 6B). The same is true in red1D (Fig-
Absence of Rec8 delays DSB formation by 2 hr (asterisk). Since ure 6B). However, some premature sister separation was seen
replication is only modestly perturbed (Cha et al., 2000), this for Red1/Mek1 mutants in spread preparations (Bailis and
delay arises after S phase. Absence of Rec8 also significantly Roeder, 1998), e.g., because of increased spatial resolution.
prolongs DSB, SEI, and dHJ life spans. However, nearly all In rec8D and red1D rec8D, nuclei with separated sisters
DSBs do finally emerge as products (Figure 4D, Figure S2, and appear early and their level rises to a final value of 50%–60%
Figure S4). (Figure 6B; Klein et al., 1999). Residual sister association is prob-
Lines 2 versus 3 ably not mediated by Mcd1: (1) 50% residual association is
All delays in Rec8 strains are absent in Rec8 Red1/ observed in mnd2D, where premature activation of separase
Mek1kinase strains and the mek1as(IN) allele is hypomorphic should eliminate Mcd1 as well as Rec8 (Penkner et al., 2005);
for this effect (Figure 5D versus Figure S3 and Figure S6). Thus, and (2) 50% residual association is seen in Mcd1-deficient
Red1/Mek1kinase mediates all rec8D timing delays. Importantly, mitotic cells where Rec8 is absent (Dı́az-Martı́nez et al., 2008).
since Rec8 Red1+/Mek1kinase+ and Rec8 Red1/ Sister association might be absent in Rec8 strains via 50%
Mek1kinase strains both exhibit a Type II phenotype (above), loss at each individual locus in every cell. Alternatively, 50% of
Red1/Mek1kinase affects the rate of recombination progression cells might exhibit full association at all loci while 50% exhibit
in rec8D but not its outcome. Red1/Mek1/Hop1 also mediates complete absence at all loci. The first situation pertains: if sister
timing delays in WT meiosis (Malone et al., 2004). In both relationships are analyzed simultaneously at two arm loci, the
Rec8 and in WT, Red1/Mek1/Hop1 may sense local recombi- frequencies of nuclei exhibiting two foci at both loci, or at neither
nation status and block progression to the next stage until prior locus, match the predictions of the binomial distribution for inde-
steps are properly completed (Discussion). pendent absence of association at each locus (Figure 6C).
Lines 1 versus 4 Sister association is established during S phase. Multiple inde-
Red1/Mek1kinase single mutants exhibit reduced DSB life pendent cultures were evaluated for both DNA replication and
spans relative to WT. However, SEI/dHJ life spans and the sister association over time (Figure 6D). The percentage of cells
time of appearance of products are unaltered. Thus, reduced that have completed S phase is the percentage exhibiting a 4C
DSB life span could reflect promiscuous DSB end processing DNA content. For a given locus, the percentage of cells lacking
(resection and/or extension) of IS-fated events (above). Rec8-mediated sister association is the fraction of two-focus
Lines 3 versus 1 or 4 cells at that time point divided by the fraction of two-focus cells
Rec8 Red1/Mek1kinase strains exhibit dramatically shorter at late times when Rec8-mediated association is absent in all
SEI and dHJ life spans than either Rec8+ Red1/Mek1kinase cells (above). In both rec8D and red1D rec8D, two-focus cells
or WT. Rec8 may act as a regulatory ‘‘brake’’ for recombinational appear after completion of S phase. Thus, Rec8 is not required
progression, independent of limitations conferred by Red1/ for establishment of sister association but is required for its
Mek1kinase; when both factors are absent, interactions race maintenance after S phase, as known for all previously studied
through biochemical steps (Discussion). organisms (discussion in Storlazzi et al., 2008). Also, two-focus

(C) For a strain carrying lac and tet arrays at different loci, percentages of cells exhibiting separation at each locus considered individually, at neither locus, or at
both loci (solid lines) and corresponding percentages predicted for independent loss of cohesion at the two loci (dashed lines). Predicted percentages at each
time point given by the binomial distribution, assuming that 5% of cells fail to enter meiosis (Padmore et al., 1991).
(D) Averages of multiple experiments for rec8D and rec8D red1D strains. Values at each time point were normalized to the time when 50% of cells exhibited
4C DNA content (new ‘‘t = 0’’), thus correcting for culture-to-culture variation in timing of meiosis initiation. Left: absolute percentages of cells that have completed
DNA replication (4C; grey; n = 12, including WT and mutant cultures) and of two-focus cells in rec8D (green; n = 5) or rec8D red1D (orange; n = 3). Values =
average ± standard deviation (SD). Note: SDs for the two mutant curves do not overlap; thus, differences in their average values are meaningful. Right: curves
at left were normalized to their final values, which represent completion of the corresponding events in 100% of meiotically active cells, thus permitting compar-
isons with one another and with appearance of DSBs (from [E]). Arrows indicate times when 50% of cells have completed each event.
(E) Chromosome spreads of WT cells immunostained for Rec8-myc or Zip1. Rec8 patterns were assigned to Categories I–IV (Results). Boxed region from (III)
enlarged at right. Zip1 pachytene pattern also shown. The scale bar represents 2mm.
(F) Top: appearance and disappearance of nuclei for each category in (E) over time in meiosis (n > 100 for each time point). Bottom: timing of other events in the
same culture.
(G) Fraction of cells that have progressed up to, or beyond, each indicated stage, given by cumulative curves derived from noncumulative curves in (F) (Hunter and
Kleckner, 2001).
(H) Coimmunostaining of Rec8-myc and Red1 at leptotene-zygotene (left) and pachytene (right) in spread chromosomes.
(I) Enlargements of regions boxed in (H). See also Figure S7.

Cell 143, 924–937, December 10, 2010 ª2010 Elsevier Inc. 931
cells appear about an hour earlier in red1D rec8D than in rec8D tively as it does that of Rec8. Mcd1 also substitutes effectively
(Figure 6D). Thus, Red1 promotes sister association in the for Rec8 for sister chromatid arm cohesion. Thus, Rec8-medi-
absence of Rec8 as well as WT. ated sister bias is probably promoted by cohesion per se. This
meiotic role of Rec8 is analogous to recently-described Mcd1
Rec8 and Red1 Localize to Distinct Domains along roles in promoting sister bias for recombinational repair of
Organized Chromosomes Prior to DSB Formation DSBs in non-meiotic cells (Introduction).
Do pre-DSB recombinosomes interact with chromosome struc- Meiosis requires that cohesion be robust globally, to ensure
ture components even prior to DSB formation and homolog bias regular homolog pairing during prophase and homolog segrega-
establishment? In budding yeast, a challenge to this idea is the tion at MI (Introduction). We infer that meiotic components Red1/
fact that silver-staining axial elements (AEs) and defined lines Mek1kinase are required to counteract this cohesion locally, in
of immunostaining for chromosome structure components the vicinity of recombinational interactions, thereby opening up
become apparent 90 min after DSB formation, concomitant the possibility for actual implementation of homolog bias via
with SEI formation at zygotene (Padmore et al., 1991; Hunter other meiosis-specific features. In this role, Red1/Mek1 probably
and Kleckner, 2001). To further characterize axis morphogen- works together with Hop1, the third yeast meiotic axis compo-
esis, we sorted nuclei exhibiting detectable Rec8 signals into nent. Hop1 interacts closely with Red1/Mek1 physically, cyto-
four categories: Category I, no staining; Category II, modest logically, and functionally with respect to several activities,
numbers of foci with no indication of organization; Category III, including homolog bias: in a hop1D mutant, at HIS4LEU2, only
larger numbers of foci with a clear tendency for linear arrays; IS-dHJs are observed, to the exclusion of IH-dHJs (Schwacha
Category IV, strongly staining lines or rows of prominent foci (Fig- and Kleckner, 1994), exactly as in red1D (above). This role of
ure 6E). Nuclei of the four categories disappear (I) and appear (II– Hop1/Red1/Mek1kinase is the only role for these proteins in
IV) progressively. As expected, Category IV appears contempo- homolog bias establishment because corresponding mutations
raneously with SC formation (lines of SC component Zip1), well in have no effect on establishment if Rec8/cohesion is absent.
advance of COs and MI (Figures 6F and 6G). Identification of The effect of Red1/Mek1kinase on Rec8-mediated cohesion
Category III reveals that longitudinal chromosome organization could occur prior to, concomitant with, or after DSB formation,
is present much earlier: Category III appears after completion by any of several possible mechanisms. An early effect is sup-
of S phase but an hour prior to DSB formation, assayed in the ported by our finding that Rec8 and Red1/Mek1 play multiple
same culture (Figure 6G). The same patterns are seen for Red1 roles, sometimes interactively, prior to and/or concomitant with
(B.M.W., unpublished data). DSB formation, i.e., for sister cohesion, for the levels and timing
Costaining for Red1 and Rec8 further reveals that the two of DSBs, and in early formation of distinct spatial domains.
types of axis components exhibit distinct patterns of loading Homolog bias is probably implemented by components of pre/
along chromosomes, both early and late (Figure 6H). Both post-DSB recombinosomes, including Dmc1 (Sheridan and
components occur broadly throughout the chromosomes; Bishop, 2006). Thus, precondition effects (Figure 7A) probably
however, regions of abundance for Red1 are often depleted for reflect a layer of structural control that is superimposed upon
Rec8, and vice versa. Red1-rich and Rec8-rich domains are recombinosome-mediated events.
seen to alternate along a chromosome (e.g., Figure 6I). Our findings exclude several previous models for establish-
ment of homolog bias. (1) With respect to Model 1, cohesion-
DISCUSSION mediated sister cohesion does not promote bias; rather, it inhibits
bias. Also, Red1/Mek1kinase does not promote sister cohesion;
The present study suggests that Rec8 promotes sister bias, prob- rather it counteracts cohesion (see also Terentyev et al., 2010). (2)
ably via its cohesin function, thereby inhibiting establishment of It was proposed that Mek1-mediated phosphorylation of Rad54
homolog bias. The role of Red1/Mek1kinase is to counteract this plays a role in homolog bias (Niu et al., 2009). The present study
effect (Figure 7A). Despite this interplay, when Red1 and Red1/ suggests that the only role of Red1/Mek1kinase is to counteract
Mek1kinase are both absent, homolog bias is still established Rec8-mediated cohesion. Mek1 phosphorylation of Rad54 may
efficiently. Thus, these structural components satisfy precondi- be important primarily for DNA damage checkpoint responses,
tions for homolog bias, which is then directly implemented by other e.g., in dmc1D where Mek1/Rad54 interactions were examined;
components (Figure 7A). During CO recombination, but not NCO indeed, a nonphosphorylatable rad54 mutant has no phenotype
recombination, bias also must be actively maintained, at the in WT meiosis (Niu et al., 2009). (3) A recent report asserts that
SEI-to-dHJ transition. Rec8 is required positively for this effect Mek1 mediates homolog bias independent of Rec8 (Callender
(Figure 7A). Red1/Mek1kinase might be similarly involved. All roles and Hollingsworth, 2010). However, that study examined only
of Rec8 and Red1 for partner choice mirror the competing dictates progression of DSBs (which we show here is not correlated
of meiosis for maintenance of cohesion globally versus disruption with partner choice), and did not examine whether progressing
locally at sites of recombination. Taken together with other results, DSBs ended up in IH or IS interactions.
our findings have additional implications.
Maintenance of Bias during CO Recombination
Interplay of Rec8-Mediated Cohesion and Red1/ For homolog bias maintenance, Rec8 is required and Mcd1 does
Mek1kinase for Establishment of Homolog Bias not effectively substitute. Thus, meiosis-specific Rec8 functions
Mcd1 substitutes efficiently for Rec8 in promoting sister bias; are involved. Such roles might still be cohesion-related or not.
further, Red1/Mek1kinase can overcome this effect as effec- Intriguingly, Red1/Mek1kinase may work together with Rec8

932 Cell 143, 924–937, December 10, 2010 ª2010 Elsevier Inc.
Figure 7. Roles of Structural Components for Meiotic Recombination
(A) Formal logic for establishment and maintenance of homolog bias as defined by mutant phenotypes.
(B) Quiescence and release of the first DSB end from its sister in relation to establishment of homolog bias and of the second DSB end from its sister in relation to
maintenance of homolog bias.
(C) Initiation of pre-dHJ formation at a homolog-associated first end or a sister-associated second end yields an IH-dHJ or an IS-dHJ, respectively.
(D) Release of the first DSB end from its tethered-loop axis complex yields a nucleus-scaled homology-searching tentacle.

for maintenance of bias (despite working in opposition to Rec8 Maintenance of homolog bias is required specifically during
during bias establishment). Similarly, Red1/Mek1kinase is impli- CO recombination. Perhaps this is because CO recombination,
cated in promoting sister cohesion (despite also counteracting but not NCO recombination, involves accompanying local
its inhibitory effects). Perhaps Red1/Mek1 and Rec8 roles for exchange of individual chromatid axes (Introduction), and thus
bias maintenance both reflect meiotic cohesion-favoring effects. is more dependent on sister stabilization factors to maintain

Cell 143, 924–937, December 10, 2010 ª2010 Elsevier Inc. 933
overall chromosome integrity during disruptive recombinational the SC (Storlazzi et al., 2010), may ensure that initiation of pre-
transitions (Storlazzi et al., 2008). dHJ formation (i.e., initiation 30 extension synthesis) can initiate
on only one of the two ends of any given DSB. In WT, Rec8
Establishment and Maintenance of Homolog Bias via acts to favor initiation at the homolog-associated end; in
Programmed Quiescence and Release of First- and Rec8, this bias is lost. Also, the Rec8 phenotype is probably
Second-DSB Ends not explained by a failure to resolve MCJMs because resolution-
During CO recombination, the two ends of each DSB interact defective mutants still exhibit reasonable homolog bias (IH:IS
with a partner duplex in ordered sequence (Introduction; Fig- dHJ = 3:1; e.g., Oh et al., 2007).
ure 7B). A first DSB end engages the partner in stable strand (4) Modulation of Rec8-mediated sister association would be
invasion (SEI formation), then primes DNA extension synthesis required for second-end release (Figure 7B).
and resultant formation of pre-dHJs. After pre-dHJ formation, Programmed quiescence and release of the second DSB end
this end is captured into the developing recombination complex also explains other findings (Figure 7B). (1) Yeast encodes both
by single-strand annealing. Apparently, during the intervening Dmc1, a meiosis-specific RecA homolog implicated specifically
period, the second end remains associated with its sister at in IH interactions, and Rad51, the general RecA homolog;
both the DNA and axis levels (Introduction). This ends-apart meiosis also specifies a direct inhibitor of Rad51, Hed1, and it
scenario has further implications. (1) At the time of DSB forma- is proposed that Dmc1 binds to the first DSB end while Rad51
tion, both DSB ends would be sister-associated. (2) The first binds to the second DSB end (Hunter, 2006; Sheridan and
DSB end would be released from this association to permit inter- Bishop, 2006). Thus, a key role of Rad51/Hed1 could be to
action with a homolog chromatid. (3) The second DSB end must promote second-end quiescence. Accordingly, a rad52 allele
remain biochemically quiescent while the first DSB end prog- specifically defective in abundant loading of Rad51 confers the
resses. (4) The second DSB end must also eventually be same 1:1 IH:IS dHJ ratio as a Rec8 mutant (Lao et al., 2008).
released from its sister to permit its capture into the recombina- (2) Components of preDSB recombinosomes, e.g., Rec102 in
tion complex during the SEI-to-dHJ transition, which occurs at yeast and Spo11 transesterase in several organisms, remain on
early/midpachytene when SC is fully formed (Hunter and Kleck- the chromosomes after DSB formation and into pachytene;
ner, 2001). Since early/mid-pachytene is an important global further Rec102 is released abruptly, specifically at early/mid-
transition point for meiosis (Kleckner et al., 2004), release of pachytene, i.e., at the time of second-end release (Kee et al.,
quiescence could be a regulated event, which in turn would 2004; Romanienko and Camerini-Otero, 2000). PreDSB recom-
imply that quiescence itself is specifically programmed. binosome components may remain bound (at the second DSB
In correspondence to these implications (Figure 7B): (1) Sister end) in order to mediate second-end quiescence.
association of DSB ends is supported by our finding that cohesin (3) Retention of a Rad51-mediated second end/sister interac-
Rec8 is relevant to events prior to and during DSB formation as tion leaves open the possibility for return to a mitotic-like
well as immediately ensuing homolog bias. intersister DSB repair reaction if meiotic IH recombination goes
(2) Rec8/cohesion concomitantly promotes sister bias and awry with IS events triggered by activation of second-end
inhibits use of the homolog. Perhaps it inhibits release of the first release. Accordingly, (i) in mouse, DSBs that lack an homologous
DSB end from its sister. Red1/Mek1kinase would then coun- partner sequence remain unresolved until early/mid-pachytene,
teract this inhibition, making first-end release possible, thereby and (ii) in allohexaploid wheat, recombinational interactions
satisfying preconditions for meiotic homolog bias. Recombino- between homeologous sequences are specifically lost, pre-
some components would then ensure that the released end sumptively to IS repair, at this same stage (Mahadevaiah et al.,
selects a homolog partner rather than its sister. 2001; Zickler and Kleckner, 1999).
(3) Rec8 could mediate maintenance of bias at the SEI-to-dHJ
transition by mediating second-end quiescence. The events that Establishment of DSB/Homolog Connections via
normally give rise to in IH-dHJ are initiated at the first (homolog- a Nucleus-Scaled Homology-Searching Tentacle
associated) DSB end (above). If these same events initiated, Tethered-loop axis complexes are clearly present shortly after
instead, at the second, sister-associated DSB end, the conse- DSB formation by both molecular and cytological criteria (Blat
quence would be formation of an IS-dHJ rather than an IH-dHJ et al., 2002; Zickler and Kleckner, 1999). It is less clear whether
(Figure 7C). The rec8D phenotype of loss of bias at the SEI stage this association is created prior to DSB formation, concomitant
can be explained, and in such a way as to give a 1:1 IH:IS dHJ with development of axial structure, or after DSB formation,
ratio, if Rec8-mediated second-end quiescence would be defec- with post-DSB complexes associating with already-developed
tive such that pre-dHJ formation can be initiated with equal prob- structure. One prior finding points to pre-DSB recombino-
ability on either end. Red1/Mek1kinase might also contribute to some/axis association: DSBs and DSB-associated Dmc1
second-end quiescence (above). complexes occur, preferentially, half way between flanking axis
Initiation of pre-dHJ formation at both ends of the same DSB association sites, rather than randomly with respect to those
seems to be quite rare. Such events would yield multichromatid sites (Blat et al., 2002; Kugou et al., 2009; F. Klein, personal
joint molecules, (MCJMs) (Oh et al., 2007). While somewhat communication). Thus, developing recombination complexes
elevated in Rec8 strains, MCJMs are not dramatically promi- and axis association sites must communicate prior to DSB
nent (K.P.K., unpublished data). To explain this and other formation. Here we provide additional evidence to this effect.
features of the data, we suggest that communication between (1) All known meiotic axis components are required for maximal
the two DSB ends, via a recombination intermediate that spans levels of DSBs including Rec8, as shown here and elsewhere.

934 Cell 143, 924–937, December 10, 2010 ª2010 Elsevier Inc.
(2) Red1/Rec8 interplay is important for the timing of DSB forma- progression. However, in a mutant lacking both Rec8 and
tion. (3) Red1 and Rec8 localize in abundant domains that exhibit Red1, recombination is still executed normally: initiation, estab-
longitudinal linearity before DSBs form. lishment of homolog bias, and CO/NCO differentiation occur;
Together, these results support a picture in which DSBs occur CO recombination proceeds via SEIs and dHJs; and CO
in tethered-loop axis complexes that contain both sisters with and NCO products are both formed efficiently. Thus, these
DSBs occurring preferentially midway between flanking axis structural components only modulate basic biochemical events,
association sites (Figure 1E and Figure 7D). If so, release of a first which are directly executed by other (i.e., recombinosome)
DSB end (above) will release a tentacle whose length is approx- components.
imately half the length of a chromatin loop (Figure 7D). Budding Red1 and Rec8 tend to be enriched in spatially distinct
yeast loops are 10–15 kb in length (Blat et al., 2002). A released domains along chromosomes on a per-cell basis. We propose
tentacle would thus be 7 kb, i.e., 0.3 or 2 mm of nucleosomal that Red1 and Rec8 carry out their distinct but coordinated roles
filament or naked DNA respectively. These lengths are similar to (for cohesion, homolog bias, and recombinational progression)
the diameter of the meiotic yeast nucleus, 2 mm. Release of via corresponding spatially distinct domains. We proposed
a tentacle would thus permit a DSB to search for a homologous previously that meiotic chromosomes might comprise two func-
partner without the dramatic stirring forces that would otherwise tionally and structurally different types of regions, interaction
be required to bring DSB ends in contact with homologous part- domains and stabilization domains, which would occur alter-
ners. Recent findings support long-distance homology recogni- nately along chromosomes (Zickler and Kleckner, 1999; Storlazzi
tion (Storlazzi et al., 2010). Importantly, chromatin loop size et al., 2008). Interaction domains would encourage structural
scales with genome size (Zickler and Kleckner, 1999; Kleckner, destabilizations needed for pairing and recombination; stabiliza-
2006), which in turn scales with nucleus size. Thus, DSB forma- tion domains would provide structural snaps that counteract
tion should universally release a nucleus-scaled homology- such destabilization, thereby maintaining chromosome integrity.
searching tentacle (Figure 7D). Red1-rich regions (which are also Hop1-rich regions; Börner
et al., 2008) and Rec8-rich regions could be these two types of
Structure-Mediated Control of Recombinational domains. In support of this idea: (1) CO sites are associated
Progression primarily with Red1/Hop1 domains (Joshi et al., 2009); and (2)
Previous considerations suggest that meiotic chromosome Red1 is more strongly required for DSB formation and, sepa-
structure plays a central role in controlling the timing of recombi- rately, to ensure that a DSB gives an IH product (i.e., homolog
nation progression in WT meiosis (e.g., Börner et al., 2008). Our bias) in domains where it is more abundant than in domains
results suggest that Red1/Mek1 and Rec8 are involved in where it is less abundant (Blat et al., 2002). Domainal recombino-
‘‘putting the brakes’’ on recombination progression and that some/axis organization could arise easily if each emerging pre-
they act via distinct effects. As a result, when both types of DSB recombination complex tends to nucleate development of
components are absent, biochemical events proceed extremely a surrounding Red1 domain, concomitantly constraining posi-
rapidly. tions of Rec8 domains.
Red1/Mek1 impedes recombination in both WT and Rec8 In the context of domainal control, a specific idea regarding
strains. Further, Mek1 is Rad53-related, and Rad53 is the homolog bias emerges. Red1 domains might comprise zones
primary downstream target of ATR, the replication and DSB in which, because of the way they developed, Rec8-mediated
repair regulatory surveillance kinase. Thus, Red1/Mek1 might cohesion is relatively depleted and where, additionally, Red1/
monitor local developments within individual recombinational Mek1 mediates another type of sister association. This alterna-
interactions, ensuring that each biochemical step is completed tive mode would compensate for the deficit of Rec8 but, unlike
and new components properly loaded before the next biochem- cohesin-mediated cohesion, would be susceptible to recombi-
ical step can occur (Schwacha and Kleckner, 1997). These nation-directed destabilization. Rec8 domains, in contrast,
effects probably also involve Pch2 (Börner et al., 2008). How would comprise zones of cohesin-mediated cohesion that is
might Rec8 participate in progression timing? Perhaps Rec8 robust and insensitive to recombinosome-directed effects.
responds to global regulatory signals derived from the cell This model can explain how Red1 could act both positively
cycle, licensing major transitions nucleus-wide. Such effects and negatively for sister cohesion. Further, when Red1 is absent,
would link recombination progression to overall cell status recombinosome-nucleated formation of Red1 domains would
and periodically reinforce nucleus-wide synchrony. Together, not occur and unconstrained loading of Rec8 would confer
Red1/Hop1/Mek1 and Rec8 would integrate local surveillance sister bias.
signals and global cell-cycle-related signals to control progres- We previously proposed that meiosis evolved by integration of
sion at both levels. elements from mitotic DSB repair and elements of late-stage
mitotic (G2-anaphase) chromosome morphogenesis, with func-
Domainal Differentiation and Evolution of the Meiotic tional linkage achieved via tethering of recombinosomes to
Interhomolog Interaction Program structural axes (Kleckner et al., 2004; Kleckner, 1996). These
Red1 and Rec8 play functionally distinct roles in every process two sets of evolutionary inputs could be implemented via spatial
examined here: sister association and several aspects of and functional domainal organization along the chromosomes.
recombination, including (1) opposing effects for homolog bias Red1/Hop1/Mek1kinase domains would mediate effects
establishment; (2) cooperative roles for maintenance of homolog evolved from mitotic DSB repair, modulating execution of
bias; and (3) distinct roles for regulation of recombination recombination and controlling local progression (above), while

Cell 143, 924–937, December 10, 2010 ª2010 Elsevier Inc. 935
Rec8 domains would mediate effects evolved from modulation REFERENCES
of cohesion status that normally occur during the latter stages
Bailis, J.M., and Roeder, G.S. (1998). Synaptonemal complex morphogenesis
of the mitotic cell cycle.
and sister-chromatid cohesion require Mek1-dependent phosphorylation of
a meiotic chromosomal protein. Genes Dev. 12, 3551–3563.
EXPERIMENTAL PROCEDURES
Blat, Y., Protacio, R.U., Hunter, N., and Kleckner, N. (2002). Physical and func-
tional interactions among basic chromosome organizational features govern
Time Courses
early steps of meiotic chiasma formation. Cell 111, 791–802.
All strains are isogenic heterothallic SK1 derivatives (Extended Experimental
Procedures). Proper synchronization of a meiotic culture is critical for these Börner, G.V., Barot, A., and Kleckner, N. (2008). Yeast Pch2 promotes
studies. Thus far, only sporulation in liquid medium allows optimal synchrony domainal axis organization, timely recombination progression, and arrest of
of the population. For 33 C analysis, cells were kept at 30 C through t = 2.5 hr defective recombinosomes during meiosis. Proc. Natl. Acad. Sci. USA 105,
with shift to 33 C occurring thereafter (for rationale, see Börner et al., 2004). 3327–3332.
For analysis of mutants containing mek1as, a single culture was synchronized Börner, G.V., Kleckner, N., and Hunter, N. (2004). Crossover/noncrossover
and divided into two identical sporulation cultures; then, in one of the two differentiation, synaptonemal complex formation, and regulatory surveillance
cultures, Mek1 kinase activity was inhibited by addition of fresh 1 mM 1-NA- at the leptotene/zygotene transition of meiosis. Cell 117, 29–45.
PP1 (USBiological) (Niu et al., 2005).
Bzymek, M., Thayer, N.H., Oh, S.D., Kleckner, N., and Hunter, N. (2010).
Double Holliday junctions are intermediates of DNA break repair. Nature
DNA Physical Analysis 464, 937–941.
Strains for recombination analyses are homozygous for leu2::hisG, ura3
Callender, T.L., and Hollingsworth, N.M. (2010). Mek1 suppression of meiotic
(DPst1-Sma1), ho::hisG and nuc1::HPHMX4 with MATa/MATa HIS4::LEU2-
double-strand break repair is specific to sister chromatids, chromosome
(BamHI)/his4X::LEU2-(NgoMIV)-URA3. Chromosomal DNA preparation and
autonomous and independent of Rec8 cohesin complexes. Genetics 185,
physical analysis were performed as described previously (Schwacha and
771–782.
Kleckner, 1994; Hunter and Kleckner, 2001). For DNA physical analysis in 2D
gels, genomic DNA was digested with XhoI and loaded onto an agarose gel Cha, R.S., Weiner, B.M., Keeney, S., Dekker, J., and Kleckner, N. (2000).
lacking ethidium bromide in TBE. Gels were stained in TBE containing ethidium Progression of meiotic DNA replication is modulated by interchromosomal
bromide, and portions of lanes containing DNA species of interest were cut out interaction proteins, negatively by Spo11p and positively by Rec8p. Genes
and placed across a 2D apparatus gel tray at 90 degree to the direction of Dev. 14, 493–503.
electrophoresis. Agarose containing ethidium bromide in TBE was poured Covo, S., Westmoreland, J.W., Gordenin, D.A., and Resnick, M.A. (2010).
around the gel slices and allowed to solidify. Electrophoresis in the second Cohesin Is limiting for the suppression of DNA damage-induced recombination
dimensional gel was performed at 4 C in pre-chilled TBE containing ethidium between homologous chromosomes. PLoS Genet. 6, e1001006.
bromide. For CO/NCO assays, DNA digested with both XhoI and NgoMIV was
Dı́az-Martı́nez, L.A., Giménez-Abián, J.F., and Clarke, D.J. (2008). Chromo-
analyzed on 1D gel electrophoresis. For all analyses, DNA species were quan-
some cohesion - rings, knots, orcs and fellowship. J. Cell Sci. 121, 2107–2114.
tified by phosphorimager analysis, with care to avoid saturation of detection
(Extended Experimental Procedures; Hunter and Kleckner, 2001; Oh et al., Eijpe, M., Offenberg, H., Jessberger, R., Revenkova, E., and Heyting, C.
2007). (2003). Meiotic cohesin REC8 marks the axial elements of rat synaptonemal
complexes before cohesins SMC1beta and SMC3. J. Cell Biol. 160, 657–670.

Microscopy Goldfarb, T., and Lichten, M. (2010). Frequent and efficient use of the sister
Samples for FACS, sister cohesion, and divisions were fixed in 40% ethanol chromatid for DNA double-strand break repair during budding yeast meiosis.
and 0.1 M sorbitol, then stored at 20 C. FACS and divisions were performed PLoS Biol. 8, e1000520.
as described in Cha et al., 2000 except that Sytox Green (Molecular Probes) Heidinger-Pauli, J.M., Mert, O., Davenport, C., Guacci, V., and Koshland, D.
was used to specifically stain DNA rather than propidium iodide. For cohesion (2010). Systematic reduction of cohesin differentially affects chromosome
analysis, cells were spun down, resuspended in 10 mM Tris (pH 8.0), and segregation, condensation, and DNA repair. Curr. Biol. 20, 957–963.
1 mg/ml DAPI and visualized immediately. Immunofluorescence was per-
Hunter, N. (2006). Meiotic Recombination. In Molecular Genetics of Recombi-
formed on chromosome spreads. Primary antibodies were mouse monoclonal
nation, A. Aguilera and R. Rothstein, eds. (Heidelberg: Topics in Current
anti-myc, rabbit anti-Red1, and goat polyclonal anti-Zip1 (Santa Cruz).
Genetics, Springer-Verlag), pp. 381–442.
Additional experimental details are described in the Extended Experimental
Procedures. Hunter, N., and Kleckner, N. (2001). The single-end invasion: an asymmetric
intermediate at the double-strand break to double-holliday junction transition
of meiotic recombination. Cell 106, 59–70.
SUPPLEMENTAL INFORMATION
Jones, G.H., and Franklin, F.C. (2006). Meiotic crossing-over: obligation and
Supplemental Information includes Extended Experimental Procedures and interference. Cell 126, 246–248.
seven figures and can be found with this article online at doi:10.1016/j.cell. Joshi, N., Barot, A., Jamison, C., and Börner, G.V. (2009). Pch2 links chromo-
2010.11.015. some axis remodeling at future crossover sites and crossover distribution
during yeast meiosis. PLoS Genet. 5, e1000557.
ACKNOWLEDGMENTS Kee, K., Protacio, R.U., Arora, C., and Keeney, S. (2004). Spatial organization
and dynamics of the association of Rec102 and Rec104 with meiotic chromo-
We thank Kleckner laboratory members and many other colleagues for helpful somes. EMBO J. 23, 1815–1824.
comments, A. Amon for Tet repressor/operator and pREC8-MCD1 strains, and
Kleckner, N. (1996). Meiosis: how could it work? Proc. Natl. Acad. Sci. USA 93,
N. Hollingsworth for mek1as. Research was supported by National Institutes of
8167–8174.
Health Grant GM-044794 to N.K.
Kleckner, N., Zickler, D., Jones, G.H., Dekker, J., Padmore, R., Henle, J., and
Received: May 1, 2009 Hutchinson, J. (2004). A mechanical basis for chromosome function. Proc.
Revised: October 19, 2010 Natl. Acad. Sci. USA 101, 12592–12597.
Accepted: October 21, 2010 Kleckner, N. (2006). Chiasma formation: chromatin/axis interplay and the
Published: December 9, 2010 role(s) of the synaptonemal complex. Chromosoma 115, 175–194.

936 Cell 143, 924–937, December 10, 2010 ª2010 Elsevier Inc.
Klein, F., Mahr, P., Galova, M., Buonomo, S.B., Michaelis, C., Nairz, K., and Padmore, R., Cao, L., and Kleckner, N. (1991). Temporal comparison of
Nasmyth, K. (1999). A central role for cohesins in sister chromatid cohesion, recombination and synaptonemal complex formation during meiosis in
formation of axial elements, and recombination during yeast meiosis. Cell S. cerevisiae. Cell 66, 1239–1256.
98, 91–103.
Penkner, A.M., Prinz, S., Ferscha, S., and Klein, F. (2005). Mnd2, an essential
Kugou, K., Fukuda, T., Yamada, S., Ito, M., Sasanuma, H., Mori, S., Katou, Y., antagonist of the anaphase-promoting complex during meiotic prophase. Cell
Itoh, T., Matsumoto, K., Shibata, T., et al. (2009). Rec8 guides canonical Spo11 120, 789–801.
distribution along yeast meiotic chromosomes. Mol. Biol. Cell 20, 3064–3076.
Romanienko, P.J., and Camerini-Otero, R.D. (2000). The mouse Spo11 gene is
Lao, J.P., and Hunter, N. (2010). Trying to avoid your sister. PLoS Biol. 8,
required for meiotic chromosome synapsis. Mol. Cell 6, 975–987.
e1000519.
Lao, J.P., Oh, S.D., Shinohara, M., Shinohara, A., and Hunter, N. (2008). Rad52 Sanchez-Moran, E., Santos, J.L., Jones, G.H., and Franklin, F.C. (2007). ASY1
promotes postinvasion steps of meiotic double-strand-break repair. Mol. Cell mediates AtDMC1-dependent interhomolog recombination during meiosis in
29, 517–524. Arabidopsis. Genes Dev. 21, 2220–2233.

Latypov, V., Rothenberg, M., Lorenz, A., Octobre, G., Csutak, O., Lehmann, E., Schwacha, A., and Kleckner, N. (1994). Identification of joint molecules that
Loidl, J., and Kohli, J. (2010). Roles of Hop1 and Mek1 in meiotic chromosome form frequently between homologs but rarely between sister chromatids
pairing and recombination partner choice in Schizosaccharomyces pombe. during yeast meiosis. Cell 76, 51–63.
Mol. Cell. Biol. 30, 1570–1581. Schwacha, A., and Kleckner, N. (1997). Interhomolog bias during meiotic
Lee, B.H., and Amon, A. (2003). Role of Polo-like kinase CDC5 in programming recombination: meiotic functions promote a highly differentiated interhomo-
meiosis I chromosome segregation. Science 300, 482–486. log-only pathway. Cell 90, 1123–1135.
Mahadevaiah, S.K., Turner, J.M., Baudat, F., Rogakou, E.P., de Boer, P.,
Sheridan, S., and Bishop, D.K. (2006). Red-Hed regulation: recombinase
Blanco-Rodrı́guez, J., Jasin, M., Keeney, S., Bonner, W.M., and Burgoyne,
Rad51, though capable of playing the leading role, may be relegated to sup-
P.S. (2001). Recombinational DNA double-strand breaks in mice precede
porting Dmc1 in budding yeast meiosis. Genes Dev. 20, 1685–1691.
synapsis. Nat. Genet. 27, 271–276.
Storlazzi, A., Xu, L., Cao, L., and Kleckner, N. (1995). Crossover and noncross-
Malone, R.E., Haring, S.J., Foreman, K.E., Pansegrau, M.L., Smith, S.M.,
over recombination during meiosis: timing and pathway relationships. Proc.
Houdek, D.R., Carpp, L., Shah, B., and Lee, K.E. (2004). The signal from the
Natl. Acad. Sci. USA 92, 8512–8516.
initiation of meiotic recombination to the first division of meiosis. Eukaryot.
Cell 3, 598–609. Storlazzi, A., Tesse, S., Ruprich-Robert, G., Gargano, S., Pöggeler, S.,
Martinez-Perez, E., and Villeneuve, A.M. (2005). HTP-1-dependent constraints Kleckner, N., and Zickler, D. (2008). Coupling meiotic chromosome axis integ-
coordinate homolog pairing and synapsis and promote chiasma formation rity to recombination. Genes Dev. 22, 796–809.
during C. elegans meiosis. Genes Dev. 19, 2727–2743. Storlazzi, A., Gargano, S., Ruprich-Robert, G., Falque, M., David, M., Kleckner,
Martini, E., Diaz, R.L., Hunter, N., and Keeney, S. (2006). Crossover homeo- N., and Zickler, D. (2010). Recombination proteins mediate meiotic spatial
stasis in yeast meiosis. Cell 126, 285–295. chromosome organization and pairing. Cell 141, 94–106.
Niu, H., Wan, L., Baumgartner, B., Schaefer, D., Loidl, J., and Hollingsworth, Terentyev, Y., Johnson, R., Neale, M.J., Khisroon, M., Bishop-Bailey, A., and
N.M. (2005). Partner choice during meiosis is regulated by Hop1-promoted Goldman, A.S. (2010). Evidence that MEK1 positively promotes interhomo-
dimerization of Mek1. Mol. Biol. Cell 16, 5804–5818. logue double-strand break repair. Nucleic Acids Res. 38, 4349–4360.
Niu, H., Li, X., Job, E., Park, C., Moazed, D., Gygi, S.P., and Hollingsworth,
Thompson, D.A., and Stahl, F.W. (1999). Genetic control of recombination
N.M. (2007). Mek1 kinase is regulated to suppress double-strand break repair
partner preference in yeast meiosis. Isolation and characterization of mutants
between sister chromatids during budding yeast meiosis. Mol. Cell. Biol. 27,
elevated for meiotic unequal sister-chromatid recombination. Genetics 153,
5456–5467.
621–641.
Niu, H., Wan, L., Busygina, V., Kwon, Y.-H., Allen, J.A., Li, X., Kunz, R.C.,
Kubota, K., Wang, B., Sung, P., et al. (2009). Regulation of meiotic recombina- Wu, H.-Y., Ho, H.-C., and Burgess, S.M. (2010). Mek1 kinase governs
tion via Mek1-mediated Rad54 phosphorylation. Mol. Cell 36, 393–404. outcomes of meiotic recombination and the checkpoint response. Curr. Biol.
20, 1707–1716.
Oh, S.D., Lao, J.P., Hwang, P.Y., Taylor, A.F., Smith, G.R., and Hunter, N.
(2007). BLM ortholog, Sgs1, prevents aberrant crossing-over by suppressing Zickler, D., and Kleckner, N. (1999). Meiotic chromosomes: integrating
formation of multichromatid joint molecules. Cell 130, 259–272. structure and function. Annu. Rev. Genet. 33, 603–754.

Cell 143, 924–937, December 10, 2010 ª2010 Elsevier Inc. 937
Upf1 ATPase-Dependent mRNP Disassembly
Is Required for Completion of Nonsense-
Mediated mRNA Decay
Tobias M. Franks,1,2,3 Guramrit Singh,1,4 and Jens Lykke-Andersen1,2,*
1Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
2Divisionof Biology, University of California, San Diego, La Jolla, CA 92093, USA
3Present address: The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
4Present address: Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester,

MA 01605, USA
*Correspondence: jlykkeandersen@ucsd.edu
DOI 10.1016/j.cell.2010.11.043

SUMMARY mRNP. In an analogous manner, early models for transcription


activation focused on the recruitment of RNA polymerase,
Cellular mRNAs exist in messenger ribonucleopro- whereas later studies demonstrated the importance of chro-
tein (mRNP) complexes, which undergo transitions matin modification and remodeling (Campos and Reinberg,
during the lifetime of the mRNAs and direct posttran- 2009). Does the mRNP constitute an obstacle to mRNA turnover
scriptional gene regulation. A final posttranscrip- as chromatin does to transcription?
tional step in gene expression is the turnover of the Evidence primarily from the yeast Saccharomyces cerevisiae
suggests that mRNA degradation generally initiates with removal
mRNP, which involves degradation of the mRNA
of the mRNA poly(A)-tail by deadenylases, which stimulates
and recycling of associated proteins. How tightly
either mRNA decapping and subsequent 50 -to-30 exonucleolytic
associated protein components are released from decay by Xrn1 (Doma and Parker, 2007; Garneau et al., 2007) or
degrading mRNPs is unknown. Here, we demon- degradation in the 30 -to-50 direction by the exosome (Schmid and
strate that the ATPase activity of the RNA helicase Jensen, 2008). In addition, some mRNA decay pathways trigger
Upf1 allows disassembly of mRNPs undergoing endonucleolytic cleavage followed by 30 -to-50 and 50 -to-30 exo-
nonsense-mediated mRNA decay (NMD). In the nucleolytic decay of the mRNA fragments by the exosome and
absence of Upf1 ATPase activity, partially degraded Xrn1, respectively (Wilusz, 2009). However, although much has
NMD mRNA intermediates accumulate in complex been learned about the enzymes that degrade mRNAs, it remains
with NMD factors and concentrate in processing unknown how the mRNA decay enzymes negotiate the mRNP.
bodies. Thus, disassembly and completion of Nonsense-mediated mRNA decay (NMD) is an mRNA turnover
pathway that targets mRNAs with premature translation termina-
turnover of mRNPs undergoing NMD requires ATP
tion codons (PTCs) for rapid degradation, thereby suppressing
hydrolysis by Upf1. This uncovers a previously unap-
protein expression from aberrant mRNAs, as well as a subset
preciated and potentially regulated step in mRNA of normal NMD-regulated mRNAs (Amrani et al., 2006; Behm-
decay and raises the question of how other mRNA Ansmant et al., 2007; Chang et al., 2007; Isken and Maquat,
decay pathways release protein components of 2007; Mühlemann et al., 2008; Rebbapragada and Lykke-
substrate mRNPs. Andersen, 2009). How a termination codon is recognized as
premature remains under investigation, but it appears to occur
INTRODUCTION when a ribosome terminates translation sufficiently upstream
of a normal 30 UTR to prevent 30 UTR-associated proteins,
mRNA decay is a critical step in the regulation of gene expres- including cytoplasmic poly(A)-binding protein (PABPC), from
sion. The stability of mRNAs can vary by orders of magnitude stimulating a proper termination event (Amrani et al., 2006; Müh-
and is dictated by the composition of the messenger ribonucleo- lemann et al., 2008; Rebbapragada and Lykke-Andersen, 2009).
protein (mRNP) (Balagopal and Parker, 2009; Moore, 2005). How This initiates the assembly of an NMD mRNP with the recruitment
decay-promoting mRNP components activate mRNA turnover is of the NMD factor Upf1 and its cofactors Upf2 and Upf3 to the
poorly understood. Several studies have shown evidence that terminating ribosome. In vertebrates, NMD is strongly stimulated
the recruitment of mRNA decay enzymes is a critical step in when an exon junction complex (EJC), which interacts with the
mRNA turnover (Cho et al., 2009; Gherzi et al., 2004; Lykke-An- Upf complex, is positioned downstream of the termination event
dersen and Wagner, 2005; Moraes et al., 2006). Yet, it is (Behm-Ansmant et al., 2007; Isken and Maquat, 2007; Moore
unknown how recruited mRNA decay enzymes access the and Proudfoot, 2009; Mühlemann, 2008; Rebbapragada and
mRNA through stably associated protein components of the Lykke-Andersen, 2009). In metazoans, the NMD mRNP is further

938 Cell 143, 938–950, December 10, 2010 ª2010 Elsevier Inc.
modulated by a phosphorylation-dephosphorylation cycle of to pulse-chase mRNA decay assays in human HeLa Tet-off cells,
Upf1 mediated by the kinase Smg1 and by protein phosphatase in which endogenous hUpf1 was depleted with an siRNA and
2A in association with the NMD factors Smg5 and Smg7 (Fuku- replaced with exogenous siRNA-resistant wild-type hUpf1
hara et al., 2005; Glavan et al., 2006; Ohnishi et al., 2003; Page (hUpf1R), or mutants thereof that fail to hydrolyze (hUpf1 DEAAR)
et al., 1999; Yamashita et al., 2001). The assembled NMD or fail to bind (hUpf1 K498AR) ATP (Bhattacharya et al., 2000;
mRNP subsequently recruits mRNA decay enzymes to initiate Cheng et al., 2007). As expected, the b-39 NMD substrate is
mRNA degradation. Depending on the specific organism, decay significantly more stable in the presence of hUpf1 ATPase
can initiate by decapping, deadenylation, and/or endonucleo- mutants than with wild-type hUpf1 (Figure 1A, top panel; see
lytic cleavage (Mühlemann and Lykke-Andersen, 2010). Recent band labeled b-39). Surprisingly, however, a fast migrating
evidence suggests that in human and Drosophila cells, decay mRNA species (indicated by an arrow in Figure 1A) accumulates
of the NMD substrate is primarily initiated by endonucleolytic when hUpf1 ATPase mutant proteins are expressed, but is not
cleavage by the NMD factor Smg6 followed by 30 -to-50 and observed in the presence of wild-type hUpf1 (Figure 1A, top
50 -to-30 exonucleolytic decay of the mRNA fragments by the panel; quantified in Figure 1B). This product corresponds to
exosome and Xrn1, respectively (Gatfield and Izaurralde, 2004; the 30 fragment of the NMD substrate following endonucleolytic
Glavan et al., 2006; Huntzinger et al., 2008; Eberle et al., 2009). cleavage by Smg6, because it is not observed with a probe
However, it remains an unresolved question how the NMD specific to the 50 end of b-globin mRNA and is strongly reduced
factors are recycled from the degrading NMD mRNP. Are they under Smg6 knockdown conditions (see Figures S1A–S1C
released by the activity of the mRNA decay enzymes or do available online). In contrast to the 30 fragment, no 50 fragment
they require active removal prior to or during mRNA decay? was detectable upon hUpf1 ATPase mutant expression (Fig-
A central component of the NMD pathway, Upf1, belongs to ure 1A and Figure S1D). Thus, ATPase-deficient hUpf1 allows
helicase superfamily 1 and shows RNA-dependent ATPase endonucleolytic cleavage of the NMD substrate, followed by
and 50 -to-30 RNA helicase activities in vitro (Bhattacharya et al., exonucleolytic decay of the resulting 50 product, but impairs
2000; Chamieh et al., 2008; Cheng et al., 2007; Czaplinski the degradation of the 30 product.
et al., 1995). The ATPase activity of Upf1 is critical to the NMD How can the failure of hUpf1 to bind or to hydrolyze ATP
pathway (Kashima et al., 2006; Weng et al., 1996a); however, specifically affect the NMD substrate 30 decay intermediate?
its specific role remains unresolved. Although helicases were One possibility is that the 30 intermediate requires Upf1
first described as ATPases that unwind polynucleotide duplexes, ATPase activity to be accessible to Xrn1, the 50 -to-30 exonu-
several helicases of superfamily 2 have more recently been clease that normally degrades this fragment (Gatfield and Izaur-
shown to function as RNPases that promote ATP-dependent ralde, 2004; Huntzinger et al., 2008; Eberle et al., 2009). If so, the
mRNP remodeling in the absence of double-stranded RNA (Fair- same fragment should accumulate upon depletion of Xrn1 in the
man et al., 2004; Jankowsky et al., 2001). Early studies impli- presence of both wild-type and ATPase-deficient hUpf1. To test
cated the Upf1 ATPase at the translation termination step of this idea, Xrn1 was depleted with siRNAs that modestly (Xrn1 #1)
yeast NMD (Weng et al., 1998), but more recent observations or strongly (Xrn1 #2) reduce Xrn1 levels (Figure 1C), and the
in yeast show that ATPase-deficient mutant Upf1 accumulates effect on the decay of the b-39 mRNA was monitored. As seen
with NMD substrates in cytoplasmic mRNP granules called pro- in Figure 1A (middle panel), when Xrn1 is modestly depleted,
cessing bodies (PBs) (Cheng et al., 2007; Sheth and Parker, the b-39 mRNA 30 fragment accumulates strongly in the pres-
2006). This suggests that the failure of Upf1 to hydrolyze ATP ence of ATPase-deficient hUpf1, but not with wild-type hUpf1.
causes the accumulation of an NMD mRNP in association with Only when Xrn1 is strongly depleted does the 30 b-39 mRNA frag-
mRNA decay factors. Here, we demonstrate that the Upf1 ment accumulate in cells expressing wild-type hUpf1 (Figure 1A,
ATPase stimulates the removal and recycling of NMD factors bottom panel; quantified in Figure 1B). However, even under
from mRNPs targeted for NMD. This is required for the comple- these conditions, the resulting 30 mRNA fragment is rapidly
tion of exonucleolytic decay of the NMD substrate. In the degraded with an apparent half-life 2–4-fold shorter than that
absence of Upf1 ATPase activity, NMD factors become trapped observed when hUpf1 ATPase mutants are expressed. A similar
with partially degraded 30 NMD mRNP intermediates. This pattern of NMD substrate 30 fragment accumulation was
demonstrates the importance of mRNP disassembly in mRNA observed when a different NMD substrate, GPx1-46, was tested
turnover, and raises the questions of whether this is a regulated (Figure 1D). These observations are not a result of globally
step in NMD and to what extent mRNP disassembly is a critical impaired Xrn1 activity, because Xrn1-mediated degradation of
step in other mRNA decay pathways. the 30 fragment of a b-globin reporter mRNA subjected to endo-
nucleolytic cleavage by endogenous let-7 microRNA is not
RESULTS impaired in the presence of ATPase-deficient hUpf1 (Figure S1E).
Thus, although it is well established that the Upf1 protein plays
The 30 Fragment Generated upon Endonucleolytic a key role in the recognition step of NMD (Amrani et al., 2006;
Cleavage of NMD mRNA Substrates Accumulates Kashima et al., 2006; Mühlemann et al., 2008; Ohnishi et al.,
in the Presence of ATPase-Deficient Upf1 2003; Rebbapragada and Lykke-Andersen, 2009), the observa-
To investigate the function of Upf1 ATPase activity in NMD, we tions shown here suggest that the ATPase activity of Upf1 is
tested the effect of impairing human (h)Upf1 ATP binding required at a later step in NMD (Figure 1). Consistent with this,
and hydrolysis on the degradation of NMD substrate mRNAs. when mRNA decay is initiated by tethered hUpf1, thereby by-
A b-globin mRNA with a PTC at position 39 (b-39) was subjected passing the Upf1 recruitment step of NMD (Lykke-Andersen

Cell 143, 938–950, December 10, 2010 ª2010 Elsevier Inc. 939
A B

Figure 1. The 30 Fragment Generated upon Endonucleolytic Cleavage of NMD Substrates Accumulates When hUpf1 Fails to Hydrolyze ATP
(A) Northern blots showing the decay of b-globin mRNA with a PTC at position 39 (b-39) in HeLa Tet-off cells depleted of endogenous hUpf1 using an siRNA and
expressing exogenous siRNA-resistant wild-type hUpf1 (hUpf1R), or hUpf1 ATPase (hUpf1 DEAAR) or ATP-binding (hUpf1 K498AR) mutants. siRNAs targeting
Xrn1 were included in the experiments in the bottom two panels. Time points above each lane represent the elapsed time following transcriptional shut-off of
b-39 mRNA by tetracycline addition. The 30 endonucleolytic cleavage fragment of b-39 (b-39 30 ) is indicated by arrows.
(B) Quantification showing the percentage b-39 30 mRNA fragment of total b-39 mRNA immediately after the transcriptional pulse (t = 0) for each condition
indicated. Percentages and standard deviations are calculated from three experiments.
(C) Western blots showing Xrn1 levels in HeLa Tet-off cells treated with a control siRNA (FLuc) (100%, 50%, or 20% total protein was loaded) or with the two Xrn1
siRNAs used in (A) (Xrn1 #1 or Xrn1 #2). hUpf3b served as a loading control.
(D) Northern blots showing GPx1 mRNA with a PTC at position 46 (GPx1-46) after a 6 hr transcriptional pulse in HeLa Tet-off cells treated as described in (A).
See also Figure S1.

et al., 2000), ATP binding-deficient hUpf1 causes accumulation blotting for associated NMD substrate mRNA under strong
of a 30 fragment that is not observed with tethered wild-type Xrn1 knock-down conditions. As seen in Figure 2A, both wild-
Upf1 unless Xrn1 is efficiently knocked down (Figure S1F). type and mutant hUpf1 proteins associate with full-length b-39
NMD substrate produced by a short transcriptional pulse (lanes
ATPase-Deficient hUpf1 Accumulates on the 30 NMD 6–8). However, the association of the accumulating 30 b-39
Intermediate fragment with ATPase-deficient hUpf1 is strongly enhanced
How does the Upf1 ATPase stimulate degradation of the 30 NMD (4.1-fold relative to full-length b-39) as compared with wild-
fragment by Xrn1? One possibility is that the Upf1 ATPase type hUpf1 (Figure 2A; compare lanes 7 and 8 to lane 6; band
triggers removal of protein from the 30 NMD mRNP intermediate, marked by arrow). These interactions occur in the cell and do
thereby allowing access for Xrn1. If so, it is predicted that wild- not form after cell lysis, because b-39 mRNA does not copurify
type hUpf1 should cycle off the 30 NMD intermediate, whereas with wild-type or mutant hUpf1 when expressed in separate cells
ATPase-deficient hUpf1 should fail to do so. This idea was tested and combined during cell lysis (Figure 2B; compare lanes 6 to 5
using hUpf1 immunoprecipitation (IP) followed by Northern and lanes 12 to 11), and the mRNA substrate does not copurify

940 Cell 143, 938–950, December 10, 2010 ª2010 Elsevier Inc.
A B

C D

Figure 2. The 30 NMD Endonucleolytic Cleavage Fragment Is Stuck with ATPase-Deficient Upf1 and Is Resistant to 50 -to-30 Exonucleolytic
Decay In Vitro
(A) Northern blot for b-39 mRNA from pellet (lanes 5-8) or 5% of total extract (lanes 1–4) fractions from anti-myc IP assays from cells transiently expressing
myc-tagged hUpf1 proteins indicated on the top or no exogenous protein (none). Cells were treated with Xrn1 #2 siRNA to promote the accumulation of the
b-39 mRNA 30 fragment.
(B) Same as (A), but b-39 mRNA was expressed either in the same cells as wild-type (WT) or DEAA mutant (DE) hUpf1 (lanes 2, 5, 8, and 11), or in different cells and
mixed prior to extract preparation (lanes 3, 6, 9, and 12). Lanes 1–3 and 7–9: 5% of total extracts; lanes 4–6 and 10–12: IP pellets. Lanes 1, 4, 7, and 10 are from
cells not expressing Myc-hUpf1. All cells were treated with Xrn1 #2 siRNA to promote the accumulation of the b-39 mRNA 30 fragment.
(C) Northern blots showing in vitro Terminator-mediated decay of b-39 30 mRNA fragment from extracts (left panels) or total RNA (right panels) from HeLa Tet-off
cells depleted of endogenous hUpf1 using an siRNA and expressing exogenous siRNA-resistant wild-type hUpf1 (hUpf1R) or hUpf1 ATPase mutants. An siRNA
targeting Xrn1 (Xrn1 #2) was included in all experiments. Time points above each lane represent the time of Terminator incubation. Bottom panels: incubation in
the absence of Terminator.
(D) Quantification for each of the experiments in (C). Percentages and standard deviations are calculated from three experiments.
See also Figure S2.

with the antibody in the absence of exogenous hUpf1 (Figure 2A, The 30 NMD mRNP Fragment Generated in the Presence
lane 5, and Figure 2B, lanes 4 and 10). These observations of ATPase-Deficient Upf1 Is Resistant to 50 -to-30
are consistent with the idea that ATPase activity is not critical Exonucleolytic Decay In Vitro
for recruitment of hUpf1 to the NMD substrate, but is required If the 30 NMD intermediate that forms in the presence of hUpf1
for the release of hUpf1 from the 30 fragment that forms after ATPase mutants is resistant to Xrn1 because of a failure in
initiation of mRNA decay. mRNP disassembly, it should become sensitive to 50 -to-30

Cell 143, 938–950, December 10, 2010 ª2010 Elsevier Inc. 941
exonucleolytic decay if protein is removed from the mRNP. To not due to different efficiencies of the FISH probes, because,
test this idea in vitro, b-39 mRNA was expressed along with in contrast to the b-39 mRNA, each FISH probe equally detected
exogenous wild-type or ATPase-deficient hUpf1 proteins in in PBs a b-globin mRNA targeted for the ARE-mRNA decay
HeLa Tet-off cells depleted for endogenous Xrn1 and hUpf1. pathway (b-ARE) (compare panels 6–10 with panels 1–5; quanti-
Cells were subsequently permeabilized, and the resulting cell fications below). The observed localization pattern is not unique
extracts were incubated with the Terminator enzyme, a commer- to the b-39 mRNA, as in the presence of ATPase-deficient hUpf1
cially available 50 -to-30 exonuclease. As seen in Figure 2C (left the 30 end of an unrelated NMD substrate, GPx1-46, could also
panels), although the 30 NMD mRNA fragment that accumulates be observed in PBs in contrast to its 50 end (Figure 3C). Thus,
as a result of Xrn1 knock-down in the presence of wild-type the 30 NMD mRNA decay intermediate that accumulates when
hUpf1 is degraded efficiently (t1/2 z 18 min), a large fraction Upf1 fails to hydrolyze ATP forms an mRNP that concentrates
(60%–70%) of the same RNA produced in cells expressing in PBs.
ATPase-deficient Upf1 proteins is highly resistant to 50 -to-30 exo-
nucleolytic decay (quantified in Figure 2D, top panel). In contrast, Multiple NMD Factors Accumulate in PBs in the Absence
when mRNPs were disrupted and protein removed from the cell of Upf1 ATPase Activity
extracts by phenol extraction prior to incubation with the What are the protein components of the accumulating 30 NMD
nuclease, the 30 NMD fragment is degraded efficiently regardless mRNP intermediate? On the basis of the observations above,
of the ability of Upf1 to hydrolyze ATP (Figure 2C, right panels; such proteins are predicted (1) to accumulate in PBs in the pres-
quantified in Figure 2D, bottom panel). As expected, full-length ence of ATPase-deficient Upf1, (2) to copurify more strongly with
b-39 mRNA is resistant to the Terminator enzyme, which is ATPase-deficient Upf1 than with wild-type Upf1 in coimmuno-
specific for 50 monophosphate-containing RNA and thus does precipitation (co-IP) assays, and (3) to copurify the NMD mRNA
not target capped RNA (Figure 2C; upper bands), and the 30 frag- 30 fragment when immunoprecipitated. We tested these predic-
ment does not degrade in the absence of Terminator (bottom tions for multiple NMD factors. Consistent with hUpf1 being part
panels). When the endonuclease RNase A was used in place of of the 30 NMD mRNP and with previous observations in yeast and
Terminator, all RNAs rapidly degrade (Figure S2). Thus, when human cells (Sheth and Parker, 2006; Cheng et al., 2007; Cho
Upf1 fails to hydrolyze ATP, the 30 NMD fragment generated by et al., 2009; Stalder and Muhlemann, 2009), indirect immunoflu-
Smg6-mediated endonucleolytic cleavage becomes trapped in orescence assays revealed that ATP binding- and ATPase-
an mRNP that includes hUpf1 and is resistant to exonucleolytic deficient mutant hUpf1 proteins, but not wild-type hUpf1, accu-
decay from the 50 end. mulate strongly in PBs (Figure 4, compare panels 4, 7, 10, 13 to
panel 1). This is consistent with the observation that ATPase
The 30 NMD Intermediate Accumulates in PBs activity is required for the release of hUpf1 from the degrading
in the Presence of ATPase-Deficient Upf1 NMD mRNP (Figure 2A).
A number of studies have demonstrated that cytoplasmic mRNPs What about other NMD factors? Remarkably, exogenously ex-
that accumulate in association with 50 -to-30 mRNA decay pressed ATPase-deficient hUpf1 (Figure 5A), but not wild-type
complexes concentrate in PBs (Eulalio et al., 2007; Franks and hUpf1 (Figure 5B), induces strong accumulation of endogenous
Lykke-Andersen, 2008; Parker and Sheth, 2007). Thus, if hUpf1 Smg5, Smg6, and Smg7 in PBs (panels 4, 7, and 10; transfected
ATPase activity is critical for NMD mRNP disassembly during cells identified by the coexpression of NLS-DsRed are marked
mRNA decay, it is predicted that the NMD intermediate should by arrowheads) but has no observable effect on the localization
accumulate in PBs when hUpf1 fails to hydrolyze ATP. Indeed, of an unrelated RNA-binding protein, HuR (panel 28). Smg1,
as seen in the fluorescence in situ hybridization (FISH) assays in hUpf2, and the EJC components Y14 and eIF4A3 more modestly
Figure 3A, both b-39 (panels 2 and 3) and GPx1-46 (panels 5 accumulate in PBs (panels 13, 16, 19, and 22), whereas hUpf3a
and 6) mRNAs accumulate strongly in PBs in the presence of and hUpf3b were only rarely observed in PBs (unpublished data).
ATPase-deficient hUpf1, but are rarely detected when wild-type None of the NMD factors localized strongly in PBs in untrans-
hUpf1 is expressed (panels 1 and 4). This finding is consistent fected cells (Figures 5A and 5B; cells not indicated by arrow-
with previous observations in yeast (Sheth and Parker, 2006). In heads), which in all cases looked similar to those expressing
contrast, wild-type b-globin mRNA accumulated only at very exogenous wild-type hUpf1 (Figure 5B). Similarly to NMD
low levels in PBs upon mutant hUpf1 expression (Figure S3). factors, Xrn1 consistently showed enhanced accumulation in
We next used individual probes hybridizing to different regions PBs in cells expressing ATPase-deficient hUpf1 (Figure 5A,
along the b-globin mRNA to ask which part of the NMD substrate panel 25; cell marked by arrowhead) as compared with cells
accumulates in PBs. Remarkably, although the region 30 of the expressing exogenous wild-type hUpf1 (Figure 5B, panel 25) or
PTC of b-39 mRNA was readily detectable in PBs in the presence no exogenous hUpf1 (Figures 5A and 5B, panel 25; unmarked
of ATPase-deficient hUpf1, the 50 end remained completely cells). Thus, multiple NMD factors and Xrn1 coaccumulate with
undetectable in PBs (Figure 3B, compare panels 4 and 5 with NMD intermediates in PBs in the presence of ATPase-deficient
panels 1 and 2; quantifications below), despite the fact that the hUpf1 (Figure 5).
full-length mRNA under these conditions is 6–10-fold more
abundant than the 30 fragment (Figures 1A and 1B). A probe ATPase-Deficient hUpf1 Shows Enhanced
that hybridizes across the mapped Smg6 endonucleolytic Copurification with Multiple NMD Factors
cleavage sites (Eberle et al., 2009) modestly detects the mRNA We next tested the prediction that proteins that require Upf1
in PBs (panel 3). The observed differences in PB detection are ATPase activity for release from the NMD mRNP should copurify

942 Cell 143, 938–950, December 10, 2010 ª2010 Elsevier Inc.
A C

Figure 3. The 30 NMD Intermediate Accumulates in PBs in the Presence of ATPase-Deficient hUpf1
(A–C) FISH assays showing localization of b-39, b-ARE and GPx1-46 mRNAs in HeLa cells in which endogenous hUpf1 was replaced with exogenous hUpf1,
hUpf1 DEAA, or hUpf1 K498A as indicated above the panels. Individual Texas-Red–labeled 50-nt probes targeting various regions of b-globin and GPx-1 mRNAs
were used in (B) and (C) as indicated below images, whereas equimolar amounts of all probes were used in the experiments in (A). GFP-hDcp1a was used as a PB
marker. Merged images are displayed (RNA:red, GFP-hDcp1a:green), whereas selected enlarged regions are shown unmerged below. Percentage of cells
displaying mRNA signal in PBs is shown in the bottom right corner of images (with the number of cells counted from at least three experiments in parentheses),
and graphed for individual probes against b-39 or b-ARE mRNA below cell images, with standard deviation from three experiments, in (B). Note: plasmids that
express b-39, b-ARE, and GPx1-46 mRNAs also express GFP; thus, some nuclear staining can be observed in the green channel.
See also Figure S3.

Cell 143, 938–950, December 10, 2010 ª2010 Elsevier Inc. 943
factors nonspecifically copurified with the IP resin (lane 1).
When the same assays were repeated in the presence of RNase,
Smg5 and Smg7 were the only NMD factors that remained en-
riched in the mutant hUpf1 complexes, suggesting the accumu-
lation of an RNA-independent interaction between these factors
when hUpf1 fails to hydrolyze ATP (Figure 6A, lanes 9–16). In
addition to NMD factors, both Xrn1 and PABPC1 showed
enhanced association with ATPase-deficient hUpf1 proteins
over wild-type hUpf1, although this was more evident in the pres-
ence than in the absence of RNase-treatment (compare lanes 11
and 12 to lane 10 and lanes 3 and 4 to lane 2). Unlike Xrn1 and
NMD factors, PABPC1 was not observed to concentrate in
PBs upon ATPase-deficient hUpf1 expression (unpublished
data), perhaps because of the high cytoplasmic abundance of
PABPC1 overwhelming detection in PBs. Taken together, these
observations are consistent with the idea that the hUpf1 ATPase
stimulates disassembly of the NMD mRNP. However, some
NMD factors show stronger accumulation than others in the
trapped mRNP complexes (Figures 5 and 6).

NMD Factors Are Associated More Strongly with the 30


NMD Fragment in the Presence of ATPase-Deficient Upf1
Finally, to test whether NMD factors can be directly observed in
complex with the NMD 30 intermediate, individual NMD factors
were immunoprecipitated from cells depleted of Xrn1 and ex-
pressing the b-39 NMD substrate as well as exogenous wild-
type or ATPase-deficient hUpf1 in place of endogenous hUpf1.
As seen in the Northern blots in Figure 6B, all tested NMD
factors, EJC components, and PABPC1 are found in complex
Figure 4. Mutant hUpf1 Proteins Deficient in ATP Binding or ATP with the 30 NMD intermediate (upper panels, band marked by
Hydrolysis Accumulate in PBs arrow). For the tested NMD factors, the association with the 30
Indirect immunofluorescence assays showing localization of myc-tagged wild-
intermediate relative to that of full-length b-39 mRNA was
type hUpf1, ATPase mutant (DEAA), or ATP-binding mutants (K498A, G494R,
and G496E) hUpf1 proteins transiently expressed in HeLa cells (left panels). enhanced 2.1–6.2-fold in the presence of ATPase-deficient
Human IC-6 serum, which detects the decapping factor Hedls and the nuclear over wild-type hUpf1 (quantifications shown below blots). In
envelope component Lamin, was used as a PB marker (middle panels). contrast, EJC components and PABPC1 showed little or no
Merged images (hUpf1: green; IC-6: red) are shown in the right panels. difference in their association with the 30 fragment whether or
Enlarged images of the indicated boxed areas are shown in the upper left not hUpf1 can hydrolyze ATP (right panels). These observations
corner for each image.
suggest that NMD factors are released from the 30 fragment by
the action of the Upf1 ATPase, whereas release of EJC compo-
more strongly with ATPase-deficient hUpf1 mutant proteins nents and PABPC1 appear to require Xrn1 activity.
than with wild-type hUpf1 in co-IP assays. In striking correlation
with the immunofluorescence assays above, the co-IP assays DISCUSSION
in Figure 6A (lanes 1–8), in which cell extracts were not treated
with RNase, show strong enrichment of endogenous Smg5 The Upf1 ATPase Allows NMD mRNP Disassembly
and Smg7 and exogenous Smg6 in hUpf1 ATPase mutant Here we have provided several lines of evidence showing that
protein complexes, compared with wild-type hUpf1 complexes ATP hydrolysis by Upf1 is critical for the disassembly and
(compare lanes 3 and 4 with lane 2) (see also Figure S4). Other completed degradation of mRNPs undergoing NMD (Figure 7).
NMD factors, hUpf2, hUpf3b, and eIF4A3 show modestly First, mutant Upf1 proteins unable to bind or to hydrolyze ATP
enhanced accumulation with hUpf1 ATPase mutant complexes cause impaired degradation of NMD substrates and accumula-
(Figure 6A), which correlates well with their moderate accu- tion of a 30 intermediate (Figure 1). Second, the 30 intermediate
mulation in PBs under the same conditions (Figure 5A) (our (Figure 2A) and multiple NMD factors (Figure 6) accumulate in
anti-hSmg6 and anti-hSmg1 antibodies failed to detect the complex with Upf1 when it fails to bind or hydrolyze ATP. Third,
endogenous proteins on western blots). These observations the NMD mRNA intermediate (Figure 3) and multiple NMD
are consistent with previous observations of enhanced associa- factors (Figures 4 and 5) accumulate in PBs in the presence of
tion of ATP binding-deficient hUpf1 with Smg7, hUpf3a, and ATP binding- or ATPase-deficient mutant Upf1. The accumula-
hUpf2 (Kashima et al., 2006). b-actin served as a negative control tion of the 30 intermediate in the presence of ATPase-deficient
and did not copurify with wild-type or mutant hUpf1 proteins Upf1 is likely a result of the inability of Xrn1 to degrade the
(Figure 6A, bottom panel), and none of the endogenous NMD RNA in the absence of mRNP disassembly (Figure 7). Consistent

944 Cell 143, 938–950, December 10, 2010 ª2010 Elsevier Inc.
with this, Xrn1 appears to be trapped with the NMD mRNP that RNA in the absence of ATP and shows ATP-dependent 50 -to-30
accumulates upon expression of ATPase-deficient Upf1, as RNA translocation activity in vitro (Cheng et al., 2007; Weng
evidenced by the enhanced association of Xrn1 with Upf1 et al., 1998) favor the former possibility. However, the observa-
complexes and with PBs under those conditions (Figures 5A tion that the level of NMD intermediate associated with PABPC1
and 6) (Cho et al., 2009; Isken et al., 2008). Moreover, the 30 and EJC components, in contrast to NMD factors, is indepen-
NMD mRNP generated in the presence of ATPase-deficient dent of Upf1 ATPase activity (Figure 6B), suggests that these
Upf1 is resistant to 50 -to-30 exonucleolytic decay in vitro unless factors are not released directly by the Upf1 ATPase but rather
protein is first removed by phenol extraction (Figure 2C). It is at a downstream step, perhaps by the activity of Xrn1 (Figure 7).
unclear where on the accumulating 30 NMD mRNP that Upf1 Either way, our observations demonstrate a previously unappre-
and the NMD complex are positioned. Site-specific RNase H ciated step in mRNA decay by which mRNP disassembly allows
cleavage followed by IP-Northern assays indicated that the completion of exonucleolytic decay and the recycling of
ATPase-deficient Upf1 is associated with both 50 and 30 frag- mRNP components. The specific mRNP components respon-
ments of the b-39 NMD 30 mRNP (unpublished data), perhaps re- sible for blockage of exonucleolytic decay of the NMD substrate
flecting interactions of Upf1 with the EJC and PABPC1 (Fig- in the presence of ATPase-deficient Upf1 remain to be deter-
ure 6A) as well as directly with the RNA. On the basis of our mined. Possible candidates could be the NMD factors them-
observations, a simple hypothesis for why NMD shuts down in selves or, perhaps, unreleased ribosomes or ribosomal subunits.
the presence of ATPase-deficient Upf1 (Figure 1A) (Kashima
et al., 2006; Weng et al., 1996a, 1996b) is that the entrapment Is mRNP Disassembly a Regulated Step in NMD?
of NMD factors on partially degraded NMD mRNPs renders the Taken together, our observations uncover a previously unappre-
pathway noncatalytic as a result of the failure of NMD factor re- ciated ATP-dependent mRNP disassembly step in mRNP
cycling. Alternatively, the Upf1 ATPase could be rate-limiting for turnover. A key question is what controls the timing of mRNP
a more upstream mRNP remodeling step, in which case the disassembly in NMD, because slow disassembly would cause
accumulation of full-length NMD substrate and 30 intermediates accumulation of decay intermediates whereas rapid disas-
in the presence of ATPase-deficient Upf1 reflects a stronger sembly could potentially release the NMD mRNP even before it
defect in 50 -to-30 decay than in endonucleolytic cleavage. The initiates decay. The ATPase activity of human Upf1 is stimulated
effect of the Upf1 ATPase on other mRNA decay activities trig- by the Upf2-Upf3 complex (Chamieh et al., 2008), and the yeast
gered by NMD, such as decapping and deadenylation, remains Upf1 ATPase is repressed by translation release factors eRF3
to be tested. In either case, our studies illustrate the importance and eRF1 (Czaplinski et al., 1998). Thus, a transition in the
of mRNP disassembly in mRNA turnover. NMD mRNP in which Upf1 is released from eRFs and associates
Although most NMD factors accumulate in PBs (Figure 5) and with Upf2-Upf3 may precede activation of the Upf1 ATPase and
in association with Upf1 (Figure 6) when Upf1 fails to hydrolyze subsequent mRNP disassembly. Consistent with this, ATP
ATP, Smg5, Smg6, and Smg7 show stronger accumulation binding-deficient Upf1 has been observed to copurify less effi-
than do Upf2, Upf3, and EJC proteins. These weaker associated ciently than wild-type Upf1 with eRF1 and eRF3 (Czaplinski
NMD proteins may either be more loosely associated with the et al., 1998; Kashima et al., 2006; Isken et al., 2008), suggesting
NMD mRNP intermediate, are found at lower stoichiometry in that it becomes trapped in a complex lacking eRFs. Moreover,
the complex, or are found only on a subset of substrates that analyses of NMD complexes stalled by NMD factor mutation or
require Upf1 ATPase activity for mRNP disassembly. Consistent depletion have indicated a transition in the human NMD mRNP
with the latter idea, Upf1 has been implicated independently of from a complex between Upf1, Smg1, and eRFs (called SURF)
Upf2 and Upf3 in the degradation of mRNAs other than NMD to a complex of NMD factors lacking eRFs (called DECID)
substrates, including histone mRNAs (Kaygun and Marzluff, (Kashima et al., 2006). In addition, the phosphorylation and
2005) and mRNAs associated with Staufen (Kim et al., 2005). dephosphorylation of metazoan Upf1 seems to be coordinated
Moreover, evidence has been presented for Upf2-, Upf3-, and with the Upf1 ATPase, because ATPase-deficient Upf1 accumu-
EJC-independent NMD pathways in human cells (Bühler et al., lates in a hyperphosphorylated form (Isken et al., 2008; Kashima
2006; Chan et al., 2007; Gehring et al., 2005). The relatively et al., 2006; Page et al., 1999), which has been reported to
weak accumulation of EJC components could also be a result prevent translation reinitiation on the NMD mRNP (Isken et al.,
of EJC disassembly by the recently discovered EJC disassembly 2008).
activity of the protein PYM (Gehring et al., 2009). Why would mRNP disassembly be under such tight control
The mechanism by which the Upf1 ATPase leads to NMD during NMD? This could possibly ensure that NMD factors are
mRNP disassembly remains to be determined. Upf1 could act released only after mRNA decay factors have already been
as a processive RNPase that uses ATPase activity to traverse recruited and/or mRNA decay initiated. This also raises the
the mRNA while displacing NMD factors and other RNA-binding possibility that ATPase-mediated mRNP disassembly could
proteins from the NMD substrate (Fairman et al., 2004; Jankow- serve as a previously proposed proofreading step in the NMD
sky and Bowers, 2006). Alternatively, Upf1 could remain pathway (Sheth and Parker, 2006), in which rapid hydrolysis of
stationary and hydrolyze ATP to release itself and other associ- ATP by Upf1 would allow the release of the NMD machinery
ated factors from the mRNA (Ballut et al., 2005). Yet another from the mRNA even before initiation of mRNA decay, thus
possibility is that ATP hydrolysis by Upf1 acts upstream of a chain allowing mRNAs wrongly targeted for NMD to be released prior
of mRNP remodeling events that in the end lead to NMD mRNP to decay (Figure 7). Several lines of evidence suggest that the
disassembly. The observations that Upf1 has highest affinity for composition of the mRNP downstream of the translation

Cell 143, 938–950, December 10, 2010 ª2010 Elsevier Inc. 945
A B

946 Cell 143, 938–950, December 10, 2010 ª2010 Elsevier Inc.
A Figure 6. Multiple NMD Factors Accumulate in
Complex with ATPase-Deficient hUpf1 and the
NMD Substrate 30 Fragment
(A) Western blots for the proteins indicated on the left
from pellet (left panels) or 2% of total extract (right panels)
fractions from anti-myc IP assays from HEK293T cells
transiently expressing proteins shown on the top, or no
exogenous protein (none). Cell extracts in lanes 9–16
were treated with RNase A prior to IP.
(B) Northern blots for b-39 mRNA isolated from pellets (IP;
top panels) or 5% total extract (Total; bottom panels)
fractions from immunoprecipitation reactions for tagged
exogenous, or in the case of hUpf2, endogenous, NMD,
EJC or PABPC1 factors, as shown on the top, in the pres-
ence of coexpressed wild-type (WT) or ATPase-deficient
(DEAA) hUpf1. Endogenous hUpf1 and Xrn1 were knocked
down using siRNAs. (-) indicates a reaction using anti-HA
beads in the absence of HA-tagged protein. Anti-FLAG
and anti-Myc beads looked similar (not shown). Below
each panel is shown the calculated enrichment of the 30
fragment relative to full-length b-39 mRNA in IP pellets in
the presence of mutant hUpf1 (DEAA) over that in the
presence of wild-type hUpf1. Representative of three
independent experiments is shown.
See also Figure S4.

generally assumed, but also in part by regulating


the Upf1 ATPase.

Is mRNP Disassembly Critical for mRNA


Turnover Pathways Other Than NMD?
Another important question for future studies is
whether mRNP disassembly is a critical step in
mRNA decay pathways other than NMD. There
have been several observations of mRNA and
mRNP structures impairing exonucleolytic
decay. For example, in S. cerevisiae, both
50 -to-30 and 30 -to-50 exonucleolytic decay is
impaired by strong RNA secondary structures
(Vreken and Raué, 1992; Decker and Parker,
1993; Muhlrad et al., 1995), and 50 -to-30 exonu-
cleolytic decay is inhibited by ribosomes stalled
by cycloheximide or by rare codons (Beelman
and Parker, 1994; Cereghino et al., 1995;
Hu et al., 2009). In Caenorhabditis elegans,
50 -to-30 decay intermediates of lin-41 mRNA tar-
geted by let-7 microRNA have been observed
with the 50 end mapping immediately upstream
termination event controls NMD (Amrani et al., 2006; Mühle- of the let-7-binding sites (Bagga et al., 2005). Even a heterolo-
mann, 2008; Rebbapragada and Lykke-Andersen, 2009). A key gous RNA-binding protein, the MS2 coat protein, appears
question for future studies is whether the downstream mRNP capable of stalling 50 -to-30 exonucleolytic decay in C. elegans
controls NMD not just by NMD factor recruitment as has been (Liu et al., 2003). In addition to exonucleolytic decay, PABPC

Figure 5. Multiple NMD Factors Accumulate in PBs in the Presence of ATPase-Deficient hUpf1
(A and B) Indirect immunofluorescence assays showing localization in HeLa cells of endogenous NMD factors as indicated on the left, or a protein not involved in
NMD, HuR, in the presence of exogenously expressed hUpf1 DEAA (A) or wild-type hUpf1 (B). Middle panels show human IC-6 serum as a PB marker and DsRed
with a nuclear localization signal to mark transfected cells (indicated by white arrowheads). Merged images (NMD factor: green; IC-6/NLS-DsRed: red) are shown
in right panels. An enlarged cell section representing the boxed area of each image is shown in the upper left corner. The average enrichment of the protein factor
in PBs over the general cytoplasm was quantified in transfected cells and given with standard deviation in each of the panels on the left.

Cell 143, 938–950, December 10, 2010 ª2010 Elsevier Inc. 947
Figure 7. mRNP Disassembly during NMD
How mRNP disassembly, dependent on Upf1 ATPase activity, is required for completion of NMD and recycling of NMD factors. See Discussion for details.

and the cap-binding protein, eIF4E, can inhibit initiation of mRNA subjected to immunoprecipitation against the indicated NMD factors, and
decay by deadenylation and decapping, respectively (Schwartz RNA from immunoprecipitated samples was isolated using Trizol. NMD
substrate levels were analyzed by Northern blotting.
and Parker, 2000; Tucker et al., 2002). Thus, disassembly of the
mRNP is likely to be a critical step in both the initiation and
Indirect Immunofluorescence and Fluorescence
the completion of mRNA turnover. Future studies should reveal In Situ Hybridization Assays
the extent to which helicases are involved in these processes. Human HeLa cells transiently expressing wild-type or mutant myc-tagged
Several helicase proteins have been identified in association hUpf1 proteins were fixed with formaldehyde and permeabilized with Triton
with mRNA decay enzymes, including Rck/p54 of the decapping X-100 (Figures 4 and 5) or ethanol (Figure 3). For indirect immunofluorescence
complex and Ski2 of the exosome (Anderson and Parker, 1998; assays, cells were incubated with antibodies against Myc-tag (Figure 4) or
against endogenous NMD factors, Xrn1 or HuR (Figure 5), as well as with
Coller et al., 2001; Fischer and Weis, 2002; Fenger-Grøn et al.,
human IC-6 serum, which recognizes endogenous Hedls (P body marker)
2005), as well as in association with bacterial and mitochondrial and Lamin, followed by fluorescently labeled secondary antibodies (anti-
exonucleases (Carpousis, 2007). Future studies should reveal mouse or –rabbit, Alexa 488; anti-human, Texas Red). Cells in Figure 5 express
whether such helicases are important for mRNP disassembly nuclear DsRed to mark transfected cells. For fluorescence in situ hybridization
to allow for processivity of their associated mRNA decay (FISH) assays, cells were hybridized with a mixture of (Figures 3A and 3C), or
enzymes, and whether pathway-specific mRNP disassembly individual (Figure 3B), TexasRed-50 -labeled 50-nucleotide NMD substrate
mRNA antisense DNA probes. Cells for FISH assays express GFP-tagged
factors are common in mRNA turnover pathways in addition to
hDcp1a to mark P bodies (see Extended Experimental Procedures for details).
NMD.
Coimmunoprecipitation Assays
EXPERIMENTAL PROCEDURES Lysates from HEK293T cells transiently expressing Myc-tagged wild-type or
mutant (DEAA or K498A) hUpf1 were subjected, in the presence or absence
mRNA Decay and RNA Immunoprecipitation Assays of RNase A, to anti-Myc immunoprecipitation followed by Western blotting
Expression of NMD reporter b-39 or GPx1-46 mRNAs was induced for 6 hr by for endogenous NMD factors, Xrn1, PABPC1, or b-actin, or in the case of
incubation in tetracycline-free medium of HeLa Tet-off cells, depleted of Smg6, coexpressed HA-tagged Smg6.
endogenous hUpf1 and/or Xrn1 using siRNAs, and transiently transfected
with plasmids expressing tetracycline-regulated b-39 or GPx1-46 mRNAs, SUPPLEMENTAL INFORMATION
and constitutively expressed control b-GAP mRNAs (Figure 1 only), as well
as plasmids expressing siRNA-resistant wild-type or mutant (DEAA or Supplemental Information includes Extended Experimental Procedures
K498A) hUpf1 protein, and in Figure 6B, other tagged NMD factors as indi- and four figures and can be found with this article online at doi:10.1016/
cated (see Extended Experimental Procedures for details). In endogenous j.cell.2010.11.043.
mRNA decay assays (Figure 1), total RNA was prepared from cells using Trizol
reagent (Invitrogen), 0, 2, 4, or 6 hr after addition of 1 mg/ml tetracycline to ACKNOWLEDGMENTS
repress NMD reporter mRNA transcription. In in vitro decay assays mediated
by Terminator (Figure 2C), cell extracts prepared in hypotonic gentle lysis We thank Drs. Tom Blumenthal (University of Colorado), Melissa Moore
buffer, or total RNA prepared from extracts using Trizol, were incubated with (University of Massachusetts Medical Center), and Sebastien Durand
Terminator 50 -to-30 exonuclease (Epicenter) for 0, 5, 10, 20 or 40 min followed (UCSD) for comments on the manuscript. Alex Choe and Claire Egan are
by RNA preparation using Trizol. In RNA-immunoprecipitation assays (Figures thanked for technical support and Joachim Weischenfeldt for production of
2A and 2B and Figure 6B), cell extracts prepared in isotonic lysis buffer were the antigen for anti-Smg1 antibodies. Drs. Marv Fritzler, Ed Chan, and Donald

948 Cell 143, 938–950, December 10, 2010 ª2010 Elsevier Inc.
Bloch are thanked for human IC-6 serum. Dr. Oliver Mühlemann is thanked for Cho, H., Kim, K.M., and Kim, Y.K. (2009). Human proline-rich nuclear receptor
the HA-Smg6 construct. Work on P bodies in our laboratory is supported by coregulatory protein 2 mediates an interaction between mRNA surveillance
funding from grant R01 GM077243 from the National Institutes of Health to machinery and decapping complex. Mol. Cell 33, 75–86.
J.L.-A. T.M.F. has been supported by National Institutes of Health NRSA Coller, J.M., Tucker, M., Sheth, U., Valencia-Sanchez, M.A., and Parker, R.
Institutional Training grant number GM-07135 from the National Institute of (2001). The DEAD box helicase, Dhh1p, functions in mRNA decapping and
General Medical Sciences. interacts with both the decapping and deadenylase complexes. RNA 7,
1717–1727.
Received: February 9, 2010
Czaplinski, K., Ruiz-Echevarria, M.J., Paushkin, S.V., Han, X., Weng, Y.,
Revised: July 21, 2010
Perlick, H.A., Dietz, H.C., Ter-Avanesyan, M.D., and Peltz, S.W. (1998). The
Accepted: October 19, 2010
surveillance complex interacts with the translation release factors to enhance
Published: December 9, 2010
termination and degrade aberrant mRNAs. Genes Dev. 12, 1665–1677.
Czaplinski, K., Weng, Y., Hagan, K.W., and Peltz, S.W. (1995). Purification and
REFERENCES
characterization of the Upf1 protein: a factor involved in translation and mRNA
degradation. RNA 1, 610–623.
Amrani, N., Sachs, M.S., and Jacobson, A. (2006). Early nonsense: mRNA
decay solves a translational problem. Nat. Rev. Mol. Cell Biol. 7, 415–425. Decker, C.J., and Parker, R. (1993). A turnover pathway for both stable and
0 0
unstable mRNAs in yeast: evidence for a requirement for deadenylation.
Anderson, J.S., and Parker, R.P. (1998). The 3 to 5 degradation of yeast
Genes Dev. 7, 1632–1643.
mRNAs is a general mechanism for mRNA turnover that requires the SKI2
DEVH box protein and 30 to 50 exonucleases of the exosome complex. Doma, M.K., and Parker, R. (2007). RNA quality control in eukaryotes. Cell 131,
EMBO J. 17, 1497–1506. 660–668.

Bagga, S., Bracht, J., Hunter, S., Massirer, K., Holtz, J., Eachus, R., and Eberle, A.B., Lykke-Andersen, S., Muhlemann, O., and Jensen, T.H. (2009).
Pasquinelli, A.P. (2005). Regulation by let-7 and lin-4 miRNAs results in target SMG6 promotes endonucleolytic cleavage of nonsense mRNA in human cells.
mRNA degradation. Cell 122, 553–563. Nat. Struct. Mol. Biol. 16, 49–55.

Balagopal, V., and Parker, R. (2009). Polysomes, P bodies and stress granules: Eulalio, A., Behm-Ansmant, I., and Izaurralde, E. (2007). P bodies: at the cross-
states and fates of eukaryotic mRNAs. Curr. Opin. Cell Biol. 21, 403–408. roads of post-transcriptional pathways. Nat. Rev. Mol. Cell Biol. 8, 9–22.

Ballut, L., Marchadier, B., Baguet, A., Tomasetto, C., Seraphin, B., and Le Hir, Fairman, M.E., Maroney, P.A., Wang, W., Bowers, H.A., Gollnick, P., Nilsen,
H. (2005). The exon junction core complex is locked onto RNA by inhibition of T.W., and Jankowsky, E. (2004). Protein displacement by DExH/D ‘‘RNA
eIF4AIII ATPase activity. Nat. Struct. Mol. Biol. 12, 861–869. helicases’’ without duplex unwinding. Science 304, 730–734.

Beelman, C.A., and Parker, R. (1994). Differential effects of translational Fenger-Grøn, M., Fillman, C., Norrild, B., and Lykke-Andersen, J. (2005).
inhibition in cis and in trans on the decay of the unstable yeast MFA2 mRNA. Multiple processing body factors and the ARE binding protein TTP activate
J. Biol. Chem. 269, 9687–9692. mRNA decapping. Mol. Cell 20, 905–915.

Behm-Ansmant, I., Kashima, I., Rehwinkel, J., Sauliere, J., Wittkopp, N., and Fischer, N., and Weis, K. (2002). The DEAD box protein Dhh1 stimulates the
Izaurralde, E. (2007). mRNA quality control: an ancient machinery recognizes decapping enzyme Dcp1. EMBO J. 21, 2788–2797.
and degrades mRNAs with nonsense codons. FEBS Lett. 581, 2845–2853. Franks, T.M., and Lykke-Andersen, J. (2008). The control of mRNA decapping
Bhattacharya, A., Czaplinski, K., Trifillis, P., He, F., Jacobson, A., and Peltz, and P-body formation. Mol. Cell 32, 605–615.
S.W. (2000). Characterization of the biochemical properties of the human Fukuhara, N., Ebert, J., Unterholzner, L., Lindner, D., Izaurralde, E., and Conti,
Upf1 gene product that is involved in nonsense-mediated mRNA decay. E. (2005). SMG7 is a 14-3-3-like adaptor in the nonsense-mediated mRNA
RNA 6, 1226–1235. decay pathway. Mol. Cell 17, 537–547.
Bühler, M., Steiner, S., Mohn, F., Paillusson, A., and Mühlemann, O. (2006). Gallouzi, I.E., Brennan, C.M., Stenberg, M.G., Swanson, M.S., Eversole, A.,
EJC-independent degradation of nonsense immunoglobulin-mu mRNA Maizels, N., and Steitz, J.A. (2000). HuR binding to cytoplasmic mRNA is
depends on 30 UTR length. Nat. Struct. Mol. Biol. 13, 462–464. perturbed by heat shock. Proc. Natl. Acad. Sci. USA 97, 3073–3078.
Campos, E.I., and Reinberg, D. (2009). Histones: annotating chromatin. Annu. Garneau, N.L., Wilusz, J., and Wilusz, C.J. (2007). The highways and byways of
Rev. Genet. 43, 559–599. mRNA decay. Nat. Rev. Mol. Cell Biol. 8, 113–126.
Carpousis, A.J. (2007). The RNA degradosome of Escherichia coli: an Gatfield, D., and Izaurralde, E. (2004). Nonsense-mediated messenger RNA
mRNA-degrading machine assembled on RNase E. Annu. Rev. Microbiol. decay is initiated by endonucleolytic cleavage in Drosophila. Nature 429,
61, 71–87. 575–578.
Cereghino, G.P., Atencio, D.P., Saghbini, M., Beiner, J., and Scheffler, I.E. Gehring, N.H., Kunz, J.B., Neu-Yilik, G., Breit, S., Viegas, M.H., Hentze, M.W.,
(1995). Glucose-dependent turnover of the mRNAs encoding succinate and Kulozik, A.E. (2005). Exon-junction complex components specify distinct
dehydrogenase peptides in Saccharomyces cerevisiae: sequence elements routes of nonsense-mediated mRNA decay with differential cofactor require-
in the 50 untranslated region of the Ip mRNA play a dominant role. Mol. Biol. ments. Mol. Cell 20, 65–75.
Cell 6, 1125–1143. Gehring, N.H., Lamprinaki, S., Kulozik, A.E., and Hentze, M.W. (2009).
Chamieh, H., Ballut, L., Bonneau, F., and Le Hir, H. (2008). NMD factors UPF2 Disassembly of exon junction complexes by PYM. Cell 137, 536–548.
and UPF3 bridge UPF1 to the exon junction complex and stimulate its RNA Gherzi, R., Lee, K.Y., Briata, P., Wegmuller, D., Moroni, C., Karin, M., and
helicase activity. Nat. Struct. Mol. Biol. 15, 85–93. Chen, C.Y. (2004). A KH domain RNA binding protein, KSRP, promotes
Chan, W.K., Huang, L., Gudikote, J.P., Chang, Y.F., Imam, J.S., MacLean, ARE-directed mRNA turnover by recruiting the degradation machinery. Mol.
J.A., II, and Wilkinson, M.F. (2007). An alternative branch of the nonsense- Cell 14, 571–583.
mediated decay pathway. EMBO J. 26, 1820–1830. Glavan, F., Behm-Ansmant, I., Izaurralde, E., and Conti, E. (2006). Structures of
Chang, Y.F., Imam, J.S., and Wilkinson, M.F. (2007). The nonsense-mediated the PIN domains of SMG6 and SMG5 reveal a nuclease within the mRNA
decay RNA surveillance pathway. Annu. Rev. Biochem. 76, 51–74. surveillance complex. EMBO J. 25, 5117–5125.
Cheng, Z., Muhlrad, D., Lim, M.K., Parker, R., and Song, H. (2007). Structural Hu, W., Sweet, T.J., Chamnongpol, S., Baker, K.E., and Coller, J. (2009).
and functional insights into the human Upf1 helicase core. EMBO J. 26, Co-translational mRNA decay in Saccharomyces cerevisiae. Nature 461,
253–264. 225–229.

Cell 143, 938–950, December 10, 2010 ª2010 Elsevier Inc. 949
Huntzinger, E., Kashima, I., Fauser, M., Sauliere, J., and Izaurralde, E. (2008). Ohnishi, T., Yamashita, A., Kashima, I., Schell, T., Anders, K.R., Grimson, A.,
SMG6 is the catalytic endonuclease that cleaves mRNAs containing nonsense Hachiya, T., Hentze, M.W., Anderson, P., and Ohno, S. (2003).
codons in metazoan. RNA 14, 2609–2617. Phosphorylation of hUPF1 induces formation of mRNA surveillance
Isken, O., Kim, Y.K., Hosoda, N., Mayeur, G.L., Hershey, J.W., and Maquat, complexes containing hSMG-5 and hSMG-7. Mol. Cell 12, 1187–1200.
L.E. (2008). Upf1 phosphorylation triggers translational repression during Page, M.F., Carr, B., Anders, K.R., Grimson, A., and Anderson, P. (1999).
nonsense-mediated mRNA decay. Cell 133, 314–327. SMG-2 is a phosphorylated protein required for mRNA surveillance in
Isken, O., and Maquat, L.E. (2007). Quality control of eukaryotic mRNA: safe- Caenorhabditis elegans and related to Upf1p of yeast. Mol. Cell. Biol. 19,
guarding cells from abnormal mRNA function. Genes Dev. 21, 1833–1856. 5943–5951.

Jankowsky, E., and Bowers, H. (2006). Remodeling of ribonucleoprotein Parker, R., and Sheth, U. (2007). P bodies and the control of mRNA translation
complexes with DExH/D RNA helicases. Nucleic Acids Res. 34, 4181–4188. and degradation. Mol. Cell 25, 635–646.
Rebbapragada, I., and Lykke-Andersen, J. (2009). Execution of nonsense-
Jankowsky, E., Gross, C.H., Shuman, S., and Pyle, A.M. (2001). Active disrup-
mediated mRNA decay: what defines a substrate? Curr. Opin. Cell Biol. 21,
tion of an RNA-protein interaction by a DExH/D RNA helicase. Science 291,
121–125. 394–402.
Schmid, M., and Jensen, T.H. (2008). The exosome: a multipurpose RNA-
Kashima, I., Yamashita, A., Izumi, N., Kataoka, N., Morishita, R., Hoshino, S.,
decay machine. Trends Biochem. Sci. 33, 501–510.
Ohno, M., Dreyfuss, G., and Ohno, S. (2006). Binding of a novel SMG-1-Upf1-
eRF1-eRF3 complex (SURF) to the exon junction complex triggers Upf1 Schwartz, D.C., and Parker, R. (2000). mRNA decapping in yeast requires
phosphorylation and nonsense-mediated mRNA decay. Genes Dev. 20, dissociation of the cap binding protein, eukaryotic translation initiation factor
355–367. 4E. Mol. Cell. Biol. 20, 7933–7942.
Kaygun, H., and Marzluff, W.F. (2005). Regulated degradation of replication- Sheth, U., and Parker, R. (2006). Targeting of aberrant mRNAs to cytoplasmic
dependent histone mRNAs requires both ATR and Upf1. Nat. Struct. Mol. processing bodies. Cell 125, 1095–1109.
Biol. 12, 794–800. Singh, G., Jakob, S., Kleedehn, M.G., and Lykke-Andersen, J. (2007).
Kim, Y.K., Furic, L., Desgroseillers, L., and Maquat, L.E. (2005). Mammalian Communication with the exon-junction complex and activation of nonsense-
Staufen1 recruits Upf1 to specific mRNA 30 UTRs so as to elicit mRNA decay. mediated decay by human Upf proteins occur in the cytoplasm. Mol. Cell
Cell 120, 195–208. 27, 780–792.

Liu, Y., Kuersten, S., Huang, T., Larsen, A., MacMorris, M., and Blumenthal, T. Stalder, L., and Muhlemann, O. (2009). Processing bodies are not required for
(2003). An uncapped RNA suggests a model for Caenorhabditis elegans mammalian nonsense-mediated mRNA decay. RNA 15, 1265–1273.
polycistronic pre-mRNA processing. RNA 9, 677–687. Tucker, M., Staples, R.R., Valencia-Sanchez, M.A., Muhlrad, D., and Parker, R.
Lykke-Andersen, J., Shu, M.D., and Steitz, J.A. (2000). Human Upf proteins (2002). Ccr4p is the catalytic subunit of a Ccr4p/Pop2p/Notp mRNA deadeny-
target an mRNA for nonsense-mediated decay when bound downstream of lase complex in Saccharomyces cerevisiae. EMBO J. 21, 1427–1436.
a termination codon. Cell 103, 1121–1131. Vreken, P., and Raué, H.A. (1992). The rate-limiting step in yeast PGK1 mRNA
Lykke-Andersen, J., and Wagner, E. (2005). Recruitment and activation of degradation is an endonucleolytic cleavage in the 30 -terminal part of the
mRNA decay enzymes by two ARE-mediated decay activation domains in coding region. Mol. Cell. Biol. 12, 2986–2996.
the proteins TTP and BRF-1. Genes Dev. 19, 351–361. Weng, Y., Czaplinski, K., and Peltz, S.W. (1996a). Genetic and biochemical
Moore, M.J. (2005). From birth to death: the complex lives of eukaryotic characterization of mutations in the ATPase and helicase regions of the Upf1
mRNAs. Science 309, 1514–1518. protein. Mol. Cell. Biol. 16, 5477–5490.

Moore, M.J., and Proudfoot, N.J. (2009). Pre-mRNA processing reaches back Weng, Y., Czaplinski, K., and Peltz, S.W. (1996b). Identification and character-
to transcription and ahead to translation. Cell 136, 688–700. ization of mutations in the UPF1 gene that affect nonsense suppression and
the formation of the Upf protein complex but not mRNA turnover. Mol. Cell.
Moraes, K.C., Wilusz, C.J., and Wilusz, J. (2006). CUG-BP binds to RNA
Biol. 16, 5491–5506.
substrates and recruits PARN deadenylase. RNA 12, 1084–1091.
Weng, Y., Czaplinski, K., and Peltz, S.W. (1998). ATP is a cofactor of the Upf1
Mühlemann, O. (2008). Recognition of nonsense mRNA: towards a unified protein that modulates its translation termination and RNA binding activities.
model. Biochem. Soc. Trans. 36, 497–501. RNA 4, 205–214.
Mühlemann, O., and Lykke-Andersen, J. (2010). How and where are nonsense Wilusz, J. (2009). RNA stability: is it the endo’ the world as we know it? Nat.
mRNAs degraded in mammalian cells? RNA Biol. 7, 28–32. Struct. Mol. Biol. 16, 9–10.
Mühlemann, O., Eberle, A.B., Stalder, L., and Zamudio Orozco, R. (2008). Yamashita, A., Ohnishi, T., Kashima, I., Taya, Y., and Ohno, S. (2001). Human
Recognition and elimination of nonsense mRNA. Biochim. Biophys. Acta SMG-1, a novel phosphatidylinositol 3-kinase-related protein kinase, associ-
1779, 538–549. ates with components of the mRNA surveillance complex and is involved in
Muhlrad, D., Decker, C.J., and Parker, R. (1995). Turnover mechanisms of the the regulation of nonsense-mediated mRNA decay. Genes Dev. 15, 2215–
stable yeast PGK1 mRNA. Mol. Cell. Biol. 15, 2145–2156. 2228.

950 Cell 143, 938–950, December 10, 2010 ª2010 Elsevier Inc.
Dynamics of Cullin-RING Ubiquitin
Ligase Network Revealed
by Systematic Quantitative Proteomics
Eric J. Bennett,1,2 John Rush,3 Steven P. Gygi,2 and J. Wade Harper1,2,*
1Department of Pathology
2Department of Cell Biology
Harvard Medical School, Boston, MA 02115, USA
3Cell Signaling Technologies, Danvers, MA 01923, USA

*Correspondence: wade_harper@hms.harvard.edu
DOI 10.1016/j.cell.2010.11.017

SUMMARY cule inhibitors of protein complex assembly or modification


often alter the dynamic reorganization of signaling networks,
Dynamic reorganization of signaling systems fre- trapping a given signaling complex in a perpetual ON or OFF
quently accompanies pathway perturbations, yet state. For example, the microtubule inhibitor taxol binds to
quantitative studies of network remodeling by path- b-tubulin within assembled microtubules, thereby blocking
way stimuli are lacking. Here, we report the develop- cycles of microtubule disassembly and assembly. A barrier to
ment of a quantitative proteomics platform centered understanding the dynamic nature of signaling networks is the
lack of quantitative approaches for determining the occupancy
on multiplex absolute quantification (AQUA) tech-
of protein complexes and how this changes in response to
nology to elucidate the architecture of the cullin-
perturbation. In this report, we globally characterize the cullin-
RING ubiquitin ligase (CRL) network and to evaluate RING ubiquitin ligase (CRL) network and describe the develop-
current models of dynamic CRL remodeling. Current ment and use of a quantitative proteomic platform to elucidate
models suggest that CRL complexes are controlled CRL dynamics.
by cycles of CRL deneddylation and CAND1 binding. CRLs are modular ubiquitin ligases that control much of the
Contrary to expectations, acute CRL inhibition with regulated protein turnover in eukaryotic cells (Petroski and
MLN4924, an inhibitor of the NEDD8-activating Deshaies, 2005). CRLs contain three major elements: a cullin
enzyme, does not result in a global reorganization scaffold, a RING finger protein (RBX1 or RBX2) that recruits
of the CRL network. Examination of CRL complex a ubiquitin-charged E2 enzyme, and a substrate adaptor that
stoichiometry reveals that, independent of cullin places substrates in proximity to the E2 enzyme to facilitate
ubiquitin transfer. The founding member of the CRLs, the SCF
neddylation, a large fraction of cullins are assembled
(Skp1/Cul1/F-box protein) ubiquitin ligase, recognizes
with adaptor modules, whereas only a small fraction
substrates via an adaptor module composed of Skp1 and one
are associated with CAND1. These studies suggest of 68 F-box proteins in humans (Jin et al., 2004). Six additional
an alternative model of CRL dynamicity where the cullin (2, 3, 4A, 4B, 5, and 7)-RING complexes interact with
abundance of adaptor modules, rather than cycles distinct sets of adaptor modules, forming 200 unique CRL
of neddylation and CAND1 binding, drives CRL complexes in total (Petroski and Deshaies, 2005). Central to
network organization. formation of an active CRL complex is the modification of
a single conserved lysine residue in the cullin subunit with the
ubiquitin-like protein NEDD8 (Petroski and Deshaies, 2005;
INTRODUCTION Wolf et al., 2003), which promotes the structural reorganization
of the C-terminal RING-binding domain of the cullin, thereby
Understanding the mechanisms through which protein networks promoting the processivity of ubiquitin transfer (Duda et al.,
are dynamically reorganized is not only important for a complete 2008; Saha and Deshaies, 2008). Neddylation, or rubylation in
description of cell systems but also has important implications yeast, occurs through an E1-E2-E3 cascade involving NEDD8-
for the identification of pharmacological agents that affect activating enzyme (NAE), NEDD8 E2s, cullin-associated RBX1,
particular pathways (Przytycka et al., 2010). Dynamic changes and the E3-like factor DCUN1D1/Dcn1p (Rabut and Peter,
in networks often are provoked by posttranslational modification 2008).
of proteins in the network, yet even for widely studied pathways, CRLs are thought to represent highly dynamic assemblies
we have little quantitative information concerning the occupancy that are regulated by several mechanisms (Bosu and Kipreos,
of individual modification events and how these modifications 2008; Cope and Deshaies, 2003; Wolf et al., 2003). First, with
are linked with dynamic complex reorganization. Small-mole- dozens of substrate adaptor modules for individual cullins, the

Cell 143, 951–965, December 10, 2010 ª2010 Elsevier Inc. 951
repertoire of adaptors may need to be molded for the particular immunoblot approaches to examine interactions, and the
needs of the cell. This could be accomplished via multiple cellular levels of CRL components remain unknown in any
mechanisms, including new adaptor synthesis, adaptor compe- system. Second, although it is generally thought that the
tition, and adaptor turnover through an autocatalytic mecha- majority of cullins in vivo are maintained in the unneddylated
nism referred to as ‘‘adaptor instability,’’ allowing assembly of state, the actual occupancy of NEDD8 on cullins is unknown.
new CRLs with distinct specificities. The rules that govern the Third, the current models suggest that acute inhibition of cullin
repertoire of CRLs in particular cellular settings are largely neddylation would ultimately result in the global sequestration
unknown, but it has been proposed that adaptor instability of cullin-RING complexes into an inactive complex with
ensues after turnover of substrates for a specific CRL is CAND1, but this model has not been rigorously tested without
complete (Chew and Hagen, 2007; Petroski and Deshaies, prolonged genetic perturbations.
2005; Wee et al., 2005; Wolf et al., 2003; Yang et al., 2002). In order to evaluate existing CRL dynamicity models, we have
Second, cullin neddylation is subject to reversal by an eight- performed a systematic analysis of the human CRL regulatory
subunit deneddylase referred to as the COP9 signalosome network in the presence and absence of the specific NAE
complex (CSN), thereby converting active CRLs to inactive inhibitor MLN4924 (Soucy et al., 2009). This inhibitor makes
forms (Cope and Deshaies, 2003; Wolf et al., 2003). COPS5, a covalent adduct with NEDD8, leading to rapid loss of cullin
a JAMM (JAB1, MPN, MOV34) domain metalloisopeptidase, neddylation in cells, followed by accumulation of CRL substrates
contains the catalytic site for deneddylation within the CSN (Brownell et al., 2010). This was accomplished by merging
(Cope et al., 2002). Third, there is evidence of a sequestration semiquantitative spectral counting methods to rapidly evaluate
pathway that serves to inhibit CRLs. This pathway involves the organization of the CRL network and determine general
the heat-repeat protein CAND1, which binds unneddylated trends in network reorganization upon acute deneddylation
adaptor-free cullin-RING complexes, thereby rendering them with quantitative multiplex AQUA (absolute quantification) tech-
in an inactive form (Goldenberg et al., 2004; Liu et al., 2002; nology to determine the occupancy of individual components
Zheng et al., 2002). and complexes within the CRL network. We found that the distri-
Whereas the CSN clearly functions as a negative regulator of bution of CRL regulatory proteins was not uniform across the
CRLs in vitro through removal of NEDD8, genetic data indicate various cullin complexes, implying that individual cullin assem-
a positive role for the CSN in CRL function in vivo (Bosu et al., blies may employ distinct modes of regulation. Contrary to
2010; Bosu and Kipreos, 2008; Cope and Deshaies, 2003; existing models, we found that acute inhibition of cullin neddyla-
Hotton and Callis, 2008; Wolf et al., 2003). This apparent tion does not result in a global reorganization of the CRL pro-
paradox is unresolved but has been rationalized through the teome, loss of adaptor association, or large-scale sequestration
idea that CRLs must undergo cycles of neddylation and of cullins by CAND1. A large fraction of CUL1 and CUL4B is
deneddylation in order to be fully functional in cells. The prevail- assembled with substrate adaptor modules with only a small
ing notion is that dynamic cycling is important for interchanging fraction associated with CAND1, regardless of cullin neddylation
adaptor modules (Figure S1F available online) (Bosu and status. Unexpectedly, we found that a more accurate snapshot
Kipreos, 2008; Cope and Deshaies, 2003; Wolf et al., 2003). of cellular CRL assemblies and the extent of cullin neddylation
This model is based upon the observation that persistent CRL required inhibition of CSN activity upon cell lysis, implying that
neddylation due to genetic CSN inactivation can promote insta- previous studies may have substantially underestimated the
bility of a subset of adaptors, thereby leading to inhibition of abundance of neddylated cullins. These studies suggest an
relevant signaling pathways (Cope and Deshaies, 2003). The alternative model of CRL control where the abundance of
ability of CAND1 to associate with unneddylated, adaptor-free adaptor modules, rather than cycles of neddylation and
cullins has led to a model wherein the CAND1-cullin-RING CAND1 binding, drive the dynamic organization of the CRL
complex serves as an intermediate in the cullin neddylation network and reveal the multiplex AQUA approach as a powerful
cycle, with release of cullin-RING from CAND1 being necessary tool to determine how the architecture of signaling networks is
for assembly with an alternative adaptor module (Bosu and reorganized by perturbations.
Kipreos, 2008). In plants and C. elegans, CAND1 mutations
display defects consistent with a positive role in the function RESULTS
of a subset of CRLs (Bosu et al., 2010; Hotton and Callis,
2008). Nevertheless, loss of CAND1 orthologs in plants, human A Platform for Systematic Proteomic Analysis of the CRL
cells, or yeast has little effect on the abundance of neddylated Regulatory Network
cullins, suggesting that the neddylation cycle may function In order to systematically explore the architecture of the CRL
independently of CAND1 (Chew and Hagen, 2007; Liu et al., regulatory network, we created cell lines using retroviral
2002; Zhang et al., 2008; Zheng et al., 2002). Moreover, deletion induction that expressed FLAG-HA-(TAP) tagged human
of CAND1 orthologs in yeast has no effect on cell viability CUL1, CUL2, CUL4A, CUL4B, CUL5, DCUN1D1, COPS6,
(Schmidt et al., 2009; Siergiejuk et al., 2009). A resolution of COPS5, NEDD8, and CAND1 in 293T cells at or below their
the cullin neddylation cycle paradox is hampered by several endogenous levels (Figure S1A). TAP-CUL3 lines could not be
factors. First, the steady-state occupancy of adaptors, established and were expressed using a transient lentiviral
NEDD8, CSN, CAND1, and DCN1 on individual cullins is approach. Liquid chromatography-tandem mass spectrometry
unknown, even in the most widely studied systems. This limita- (LC-MS/MS) data derived from anti-HA immune complexes
tion is amplified by the virtually universal use of semiquantitative were processed through CompPASS to identify high-confidence

952 Cell 143, 951–965, December 10, 2010 ª2010 Elsevier Inc.
A B C Figure 1. Systematic Proteomic Analysis of
1 TSCs 50 COP the CRL Network at Steady State
COP S1 COP
S8 CCNA1 CCNA2
S2
1 COP
(A) TSCs for CRL components associated with
1D 8 COPS7 CDC2 COP S1
A B 6 5 1 A/B
COP CKS1B S8
COP

L1 L2 L3 L4 L4 L5 PS PS ND UN DD
S3 S2

C
U
C
U
C
U
C
U
C
U
C
U O
C C
O A
C D
C
N
E COP
S6 COP
COP
S4
CDK3
CDK2 COPS7
A/B
COP
S3
each bait are indicated by the heat map. Associ-
S5
SKP2

Cullins
FBXW7
FBXW9 FBXW11

BTRC
COP
S6 COP
S5
COP
S4
FEM1A
ated proteins are depicted within the heat map if
NEDD8 FBXW5 FBXL18 FEM1B the TSCs for the given protein were in excess
FBXW2 SKP1 FBXL14 ZYG11B
CAND1
FBXO9 FBXL15
TCEB1
FEM1C
of 3. For a complete list of interacting proteins,
NEDD8 APPBP2

CSN FBXO7
CUL1
FBXL17
NEDD8 see Table S1.
FBXL8
CUL2
FBXO44
CAND1 FBXO10 VHL
KLHDC10
(B–F) Schematic representation of proteins asso-
FBXO42 FBXO11

FBXO33 FBXO17 KLHDC2


ciated with CUL1 (B), CUL2 (C), CUL3 (D),
RNF187 TCEB2

FBXO31
FBXO18
KLHDC3
CUL4A or CUL4B (E), and CUL5(F).
FBXO30 FBXO21

FBXO3 FBXO22
PPIL5
LRRC14 See also Figure S1.
CUL1 adaptors

D COP
COP S1 COP
S8 S2

COPS7 COP
A/B S3
ARMC5 SHKBP1
COP KLHL12
COP
S6 COP S4 KCTD6
S5 KLHL13
KCTD7
KLHL15
CUL2 adaptors

KCTD9

KCTD3
BTBD1
(Figure S1B). However, the total spectral
KLHL17
BTBD2
KEAP1

KLHDC5
KCTD18

KCTD5
BTBD7 counts (TSCs) for CAND1 varied widely
KLHL18

KLHL2

KLHL11
KCTD13
NEDD8
BTBD8
depending on the individual cullin (Fig-
KLHL20
BTBD9

RHOBTB3
KCTD12

KCTD17
CUL3
BTBD10
ure 1A), indicating that CAND1 is not
KLHL21

CAND1 KLHL22

RHOBTB2 KCTD10 DCUN1D1 GAN uniformly distributed across cullins. Only


KLHL23
KBTBD2

five of the seven cullins were found within


KBTBD8
RHOBTB1
KLHL24
KBTBD7
CUL3 adaptors

KBTBD4
KBTBD6
KLHL9
NEDD8 immune complexes, whereas six
KLHL26
KLHL25

KLHL8

KLHL7
KLHL36
KLHL28
of the seven cullins were present in
KLHL5
KLHL4

COPS6 complexes (Figures S1C and


E COP
S8
COP
S1 COP
S2 F S1D; Table S1). However, the distribution
COPS7
A/B
COP
S3
COP
COP
S1 COP
of cullins differed, suggesting further
COP COP S8 S2
S6 COP
S5
S4
WDR23
DDB2
DTL
COPS7
A/B
COP
S3
heterogeneity in the CRL regulatory
WDR40A

DDB1
ERCC8

CRBN
COP
S6 COP
S5
COP
S4 network. For example, TSCs for CUL5
FEM1B
WDR40C PPIL5

and its associated adaptor proteins


CUL4 adaptors

AMBRA1 KLHDC2
ASB7
WDR42A NEDD8
TCEB1

PHIP CUL4A
VPRBP
WSB2
were lower than other cullins within ASB1

WDR21A NEDD8 ASB13


CUL4B
WDR68

CAND1 WDR21B WSB1 NEDD8 and COPS6 immune complexes.


CUL5
ASB3
WDTC1

TRPC4AP
DCUN1D1 RFWD2 SOCS7
CAND1 was absent from not only
CAND1

DCUN1D1 ASB6
WDR22
BRWD1
SOCS6
NEDD8-associated complexes, as ex-
TCEB2
CUL5 adaptors

LRRC41
TOR1AIP2
IQWD1 SOCS4 PCMTD1
DDA1
DCAF16 WDR32
pected, but also from CSN complexes,
SOCS2 PCMTD2

suggesting that CAND1 and CSN asso-


ciate with distinct populations of cullin
complexes (Olma et al., 2009). Six cullins
were associated with DCUN1D1 (Fig-
candidate-interacting proteins (Sowa et al., 2009), thereby ure S1E), with the CUL3 and CUL5 CRL complexes being the
providing a snapshot of the steady-state CRL network. As most highly represented within the DCUN1D1 complex.
expected, each cullin associated with specific classes of
substrate adaptor proteins in addition to regulatory proteins CSN Activity within Lysates Alters the Architecture
(Figure 1A; Table S1). We found 26 F-box proteins as well as of the CRL Network
SKP1 and the SKP2-associated cyclin A-CDK-CKS complex The majority of previous studies report that only a small fraction
associated with CUL1 (Figure 1B), 12 BC box-containing and of cullins are modified by NEDD8 (typically <10%). However, the
14 SOCS box-containing proteins in addition to elongins B and finding that a substantial fraction of cullins are associated with
C with CUL2 and CUL5, respectively (Figures 1C and 1F), 53 the CSN deneddylase raised the possibility that CSN activity
BTB-containing proteins with CUL3 (Figure 1D), and 24 DCAFs upon cell lysis reduces the apparent extent of neddylation
along with DDB1 associated with CUL4A or CUL4B (Figure 1E). observed. To test this possibility, we lysed TAP-CUL1-express-
Although this represents the largest number of substrate adap- ing cells in the presence and absence of the zinc chelator and
tors identified in a single experiment, the absence of a subset COPS5 inhibitor 1,10-orthophenathroline (OPT) (Cope et al.,
of known or predicted adaptors suggests that the CRL network 2002). TAP-CUL1 was completely unneddylated in the absence
identified here represents the most abundant or avidly associ- of OPT under the lysis conditions used, whereas 50% of CUL1
ated adaptors in 293T cells. The hypothesis that proline at was neddylated with OPT in the lysis buffer (Figure 2A), suggest-
position 2 in the F-box motif is required for CUL1 association ing that inhibition of CSN upon cell lysis can substantially
(Schmidt et al., 2009) was not confirmed, as FBXL18 and increase the extent of CUL1 neddylation similar to what was
FBXO30 lacking this residue were found in association with observed when antibodies against COPS2 (CSN2) were included
CUL1. CAND1 associated with CUL1, CUL3, CUL4B, and during lysis (Yang et al., 2002). Examination of the extent of
CUL5, as expected (Liu et al., 2002; Zheng et al., 2002) endogenous cullin neddylation revealed that addition of OPT,

Cell 143, 951–965, December 10, 2010 ª2010 Elsevier Inc. 953
A B MLN4924-C - - - - + Figure 2. CSN Activity within Lysates Alters
MLN4924-L - - - + - the Architecture of the CRL Network
TAP-CUL1 1,7-OPT - + - - -
- + OPT 1,10-OPT - - + + - (A) TAP-CUL1 cells were lysed in the presence or
absence of 2 mM 1,10 o-phenanthroline (OPT)
IB:CUL1
100 and analyzed by SDS-PAGE and immunoblotted
75 IB:CUL2 with a-HA antibodies.
(B) 293T cells were either untreated or treated with
IB:HA IB:CUL3
1 mM MLN4924 for 4 hr (MLN4924-C). Untreated
IB:CUL4A cells were then lysed without OPT, with 1-10
OPT, 1-7 OPT, or 1-10 OPT with MLN4924 added
IB:CUL5
to the lysis buffer (MLN4924-L). The extent of cullin
C 1 TSCs 100 neddylation was determined by immunoblotting.
1 Arrows indicate the neddylated species.
A B 6 5 1 1D 8
L1 L2 L3 L4 L4 L5 PS PS D U
N D (C) LC-MS/MS analysis of the indicated immune
U U U U U U O O AN C ED TAP-NEDD8
C C C C C C C C C D N - OPT
- + - + - + - + - + - + - + - + - + - + - + OPT D 45
40 + OPT complexes in the presence or absence of OPT.

Normalized TSCs
CUL1
CUL2
35
TSCs were normalized by bait TSCs. Associated
Cullins
CUL3 30 **
CUL4A
CUL4B
CUL5 25 * proteins are depicted within the heat map if the
CUL7
NEDD8 20
NAE1
UBA3
* TSCs for the given protein were in excess of 3
CAND1 15
DCUN1D1
COPS1
COPS2
COPS3
10
within any of the immune complexes.
5
CSN

COPS4
COPS5
COPS6
COPS7A
0 (D) Comparison of cullin TSCs within TAP-NEDD8

CUL1

CUL2

CUL3

CUL4A

CUL4B

CUL5
COPS7B
interactor
COPS8
immune complexes with (red bars) or without OPT
(blue bars) in the lysis buffer.
E
CUL1 adaptors

80
COPS1
(E–G) Bait-normalized TSCs for COPS1 (E),
70
COPS5 (F), or CAND1 (G) associated with the indi-
Normalized TSCs

60
50 cated TAP-immune complexes with (red bars) and
40
without OPT (blue bars) in the lysis buffer.
30
20 * Error bars: standard deviation (SD) of duplicate
CUL2 adaptors

10 measurements (*,** = p value < 0.05, 0.01, respec-


0
tively, by Student’s t test). See also Figure S2 and
CUL1

CUL2

CUL3

COPS6

COPS5
CUL4A

CUL4B

CUL5

NEDD8
TAP-Cell line
Table S2.
F 120
COPS5
Normalized TSCs

100

80
CUL3 adaptors

60
able to detect TSCs for all seven cullins as
40
** *
well as an increase in the amount of bait- 20 *
0 normalized TSCs for individual cullins
CUL1

CUL2

CUL3
CUL4A

CUL4B

CUL5

COPS6

COPS5

NEDD8

TAP-Cell line
within NEDD8 immune complexes
(Figures 2C and 2D). This effect was
G
120 CAND1
particularly striking with CUL3, where
capture of neddylated CUL3 is almost
Normalized TSCs

100
CUL4 adaptors

80

60
completely dependent on CSN inhibition
*
40 (Figures 2B and 2D). CSN association
20 *
0
with cullins was largely unaffected by
CUL1

CUL2

CUL3

CUL4A

CUL4B

CUL5

DCUN1D1
adaptors

OPT addition, except for CUL1, where


CUL5

TAP-Cell line

CSN inhibition reproducibly increased


the interaction between CSN and CUL1
(Figures 2E and 2F). A reduction in
CAND1 TSCs associated within CUL1,
but not the nonchelating 1,7-orthophenanthroline, resulted in CUL3, CUL4A, and DCUN1D1 was also observed, although
dramatically increased levels of observable CUL1 and CUL3 statistical significance was reached only with CUL4A and
neddylation and smaller increases in the amount of CUL2 and DCUN1D1 (Figure 2G). We conclude that CSN inhibition in vitro
CUL4A neddylation (Figure 2B). Addition of the NAE inhibitor via OPT addition increases the extent of CRL neddylation and
MLN4924 in combination with OPT to the lysis buffer did not alter more closely represents the in vivo status of the CRL network.
the levels of cullin neddylation, indicating that the observed As such, OPT was included in all experiments described here-
increase in cullin neddylation upon lysis in the presence of OPT after unless otherwise noted.
was not due to in vitro NAE activity (Figure 2B). As expected,
addition of MLN4924 to cells 4 hr prior to lysis resulted in MLN4924 Treatment Results in Rapid Deneddylation
complete deneddylation of all cullins (Figure 2B). of CRLs
We therefore examined the impact of OPT on the global CRL Having defined conditions that allow us to approximate the
network by measuring TSCs, which provide a semiquantitative in vivo architecture of the CRL network using proteomics, we
measure of protein abundance in parallel immune complexes next examined the effects of acute inhibition of neddylation on
(Figure 2C; Table S2). Only in the presence of OPT were we CRL network organization. In agreement with previous reports,

954 Cell 143, 951–965, December 10, 2010 ª2010 Elsevier Inc.
A
MLN4924 - + - + - + - + - +
100 100 100 150 100
75 75 75
100
* 75
IB:CUL2 IB:CUL3 IB:CUL5
IB:CUL1 75
IB:CUL4

50 50 50 50
50
IB:tubulin IB:tubulin IB:tubulin IB:tubulin
IB:tubulin

B TAP- TAP- TAP- TAP- TAP- TAP- TAP- TAP- TAP- TAP- C TAP-
Nedd8
TAP- TAP- TAP-
COPS6 DCUN1D1 CAND1
CUL1 CUL2 CUL3 CUL4A CUL4B CUL5 Nedd8 COPS6 DCUN1D1 CAND1
MLN4924 - + - + - + - + - + MLN4924 - + - + - + - + - + MLN4924 - + - + - + - +
100 inputs 100 inputs 150
75 IB:CUL1 75 IB:CUL1
100 IP:HA
IB:CUL5
75
150
100
inputs
100 IB:HA 75 50
75 inputs
50 IB:HA 150
37 IP:HA
100
IB:CUL4
25 75

100 IP:HA
75 IB:CUL1

D MLN4924
TAP-NEDD8-IP
30
+
TSCs
-
1

TRPC4AP
VPRBP
CUL1
CUL2
CUL3

COPS7B
CUL4A
CUL4B
CUL5
CUL7
NEDD8
NAE1
UBA3
UBE2M
COPS1
COPS2
COPS3
COPS4
COPS5
COPS6
COPS7A

COPS8
SKP1
SKP2
FBXL15
FBXL18
FBXO11
FBXO17
FBXO21
FBXO22
FBXO3
FBXO38
FBXO42
FBXO7
FBXO9
FBXW11
TCEB1
TCEB2
APPBP2
FEM1B
KLHDC10
KLHDC3
PPIL5
ZYG11B
BTBD9
KBTBD6
KLHDC5
KLHL18
DDB1
AMBRA1
DCAF14
CRBN
TOR1AIP2

WDR12
WDR21A
WDR23
WDR32
WDR40A
WDR40C
ASB6
LRRC47
E F G H
30 40
70 TAP-NEDD8-IP - MLN4924 ** 45
* + MLN4924 ** 35 40
60 25
Normalized TSCs

Normalized TSCs

Normalized TSCs

* 30 Normalized TSCs 35
50 20
** 30
25
40
15
* 25
* 20
30
** 20
** 15
* 10 ** 15
20 **
** ** 10 * * 10
10 5 * * ** * **
5 * * 5
0 0 0 0
NAE1

UBA3
COPS1
COPS2
COPS3
COPS4
COPS5
COPS6

COPS7B
COPS7A

COPS8

SKP1
SKP2

FBXO3

FBXW11
TCEB1
TCEB2
FBXL18
FBXO21

FBXO42

FEM1B
KLHDC10
ZYG11B
KBTBD6
KLHL18
DDB1
AMBRA1
VPRBP
WDR21A
WDR23
WDR40A
CUL1

CUL2

CUL3

CUL4A

CUL4B

CUL5

CUL7

Figure 3. Rapid Deneddylation of CRLs in Response to NAE Inhibition by MLN4924


(A) 293T cells with or without 1 mM MLN4924 (4 hr) treatment were lysed in the presence of OPT, and the extent of neddylation of endogenous cullins was deter-
mined by immunoblotting. * indicates nonspecific background band.
(B) 293T cells expressing the indicated TAP-tagged proteins with or without 4 hr MLN4924 treatment were lysed in the presence of OPT and immunoblotted with
the indicated antibodies. Bait complexes were immunoprecipitated with a-HA and immunoblotted with the indicated antibodies.
(C) Complexes were immunoprecipitated with a-HA-coupled resin and blotted with antibodies against CUL1, CUL5, and CUL4A.
(D) TAP-NEDD8-expressing cells with or without 4 hr MLN4924 treatment were lysed in the presence of OPT. a-HA complexes were analyzed by LC-MS/MS, and
bait-normalized TSCs for known CRL components are displayed.
(E–H) Normalized TSCs for cullins (E), CSN subunits (F), CRL adaptor proteins (G), and the NEDD8 conjugation machinery (H) associated with TAP-NEDD8 with
(red bars) or without (blue bars) MLN4924 treatment. Error bars: SD of duplicate measurements (*,** = p value < 0.05, 0.01, respectively, by Student’s t test).

treatment of 293T cells with the NAE inhibitor MLN4924 (1 mM) endogenous CUL5, CUL4A, and CUL1 associated with CRL
for 4 hr resulted in complete conversion of endogenous neddy- regulatory proteins (Figure 3C). CUL2 and CUL5 expression
lated cullins to their unneddylated forms (Figure 3A) (Soucy can only be detected after HA immunoprecipitation (data not
et al., 2009). Similarly, treatment of the TAP-tagged CRL and shown). To further validate the use of MLN4924 to examine
regulatory protein-expressing cells resulted in near complete CRL dynamics, we treated TAP-NEDD8-expressing cells with
deneddylation of exogenous cullins (Figure 3B), as well as MLN4924 for 4 hr and examined the associated complexes by

Cell 143, 951–965, December 10, 2010 ª2010 Elsevier Inc. 955
LC-MS/MS (Figure 3D). As expected, MLN4924 treatment Whereas TSCs for CUL1 were 10-fold lower than that found
resulted in a severe reduction in the association of CRL with TAP-CUL1 due to differences in antibody binding efficiency,
complexes with NEDD8 (Figure 3E). Bait-normalized TSCs for we found CSN, SKP1, and ten F-box proteins in association with
the cullins, CSN subunits, and associated cullin adaptor proteins endogenous CUL1. Nine of ten F-box proteins, as well as SKP1
within NEDD8 immune complexes were largely lost upon treat- and CSN components, remained associated in comparable
ment with MLN4924 (Figures 3E–3G). In contrast, NEDD8 main- levels 4 hr after NAE inhibition, pointing to the absence of a global
tained its association with components of the NAE heterodimer reorganization of the endogenous CUL1 complex.
(UBA3 and NAE1) upon MLN4924 treatment (Figure 3H), indi-
cating that the reduction of CRLs associated with NEDD8 was Multiplex AQUA for Quantitative Proteomics of the CRL
due to loss of isopeptide-linked NEDD8. Network
Although we used spectral counting to observe increased cullin-
Acute NAE1 Inhibition Does Not Globally Alter the CRL CAND1 association upon deneddylation, it is not possible to use
Network this technique to determine CAND1-cullin stoichiometry. In order
The prevailing models of CRL dynamics, based primarily on pro- to provide a quantitative picture of CRL architecture upon
longed genetic perturbations, predict that inhibition of cullin ned- deneddylation and to determine the occupancy of individual
dylation would result in CRL complex disassembly, release of subunits within the network, we developed a multiplex AQUA
adaptor protein modules, and sequestration of the cullin-RING platform for the CRL network. We synthesized a library of 38
complex by CAND1. In order to test the dynamic nature of reference tryptic peptides corresponding to peptides previously
CRL complexes on a short timescale, we first evaluated the observed by LC-MS/MS for each of the cullins, SKP1, DDB1,
effect of 4 hr MLN4924 treatment on the TAP-CRL pathway CSN subunits, CAND1, DCUN1D1, NEDD8, and the F-box
cell library (Figure 4A; Table S3). In contrast to expectations, proteins BTRC (b-TRCP1) and FBXW11 (b-TRCP2) (Figure 5A;
the array of adaptor proteins associated with individual cullins Table S6). Each reference peptide contained a single N15C13-
based on TSCs was largely unchanged, and in the case of labeled amino acid, allowing heavy and endogenous (light)
CUL2, several adaptor proteins displayed a statistically signifi- peptides to be distinguished and quantified by MS (Kirkpatrick
cant increase in association (Figure 4E). Consistent with these et al., 2005). For 10 of 23 target proteins, we identified 2 or 3 useful
results, MS analysis of TAP-tagged adaptor proteins demon- peptides, whereas for 12 targets, single reference peptides were
strated that, irrespective of the cullin neddylation status, adaptor available. Trypsin-digested CRL complexes were supplemented
proteins remain stably associated with their target cullins with 100 fmoles of the peptide library prior to LC-MS/MS, and the
(Figures S3A and S3B). relative intensities of extracted ion chromatograms from endog-
In contrast with adaptor proteins, analysis of cullin regulatory enous and reference peptides from duplicate MS runs were
components revealed distinct patterns of changes that were used to calculate the abundance of the endogenous protein
generally cullin specific. Inhibition of neddylation resulted in within each immune complex. For those proteins with multiple
a significant (25%–60%) decrease in CSN-CUL1 and CSN- reference peptides the average ratio among the reference
CUL3 association whether examined using CSN or cullin peptides is reported (Table S5). Reference and endogenous
immune complexes (Figures 4B and 4C), a result that was NEDD8 peptide was readily observed within TAP-CUL1 immune
confirmed by immunoblotting (Figure 4D). Given the loss of asso- complexes in untreated cells, but MLN4924 treatment resulted in
ciation of CSN with cullin seen upon deneddylation, one might complete loss of the endogenous NEDD8 peptide, whereas the
anticipate an increase in CAND1 association. Indeed, the extent intensities of the NEDD8 reference peptide and peptides for
of TAP-CAND1 association with CUL1, CUL4, and CUL5 was CUL1 itself were unchanged (Figure 5B). Using this technique
increased 2- to 8-fold as assessed by TSCs (Figure 4B). we determined the mole fraction of CUL1 associated with each
Increased CAND1 association was also seen with TAP-CUL1, CRL regulatory component.
CUL4A, CUL4B, CUL5, and DCUN1D1 upon inhibition of neddy- Consistent with immunoblots, 45% of CUL1 is neddylated
lation, a result that at face value is consistent with the CAND1 under steady-state conditions, and this fraction is lost, as
sequestration model (Figure 4C). Together, this analysis revealed expected, with MLN4924 treatment (Figure 5C). Interestingly,
that although CAND1 association with cullins does increase multiplex AQUA analysis of CUL1 purified without OPT in the
upon deneddylation, this does not occur at the expense of global lysis buffer revealed only 5% of CUL1 to be neddylated, consis-
CRL complexes as the amount of adaptor containing CRL tent with immunoblotting results here and in other studies
complexes was largely unchanged by NAE inhibition (Figure 4E). (Figure 2B and Figures S4A and S4B). It is possible that OPT-
Of note, interrogation of the effect of NAE inhibition on the same mediated CSN inhibition may not be absolute in cell lysates,
complexes but without inhibition of CSN activity with OPT and thus our measurement of the extent of neddylation may
resulted in either reduction or ablation of the changes observed underestimate that in intact cells. Further, we observed a greater
in regulatory protein binding to CRLs in the presence of OPT, than 3-fold increase in the amount of NEDD8 associated with
underscoring the importance of OPT addition to allow changes CSN immune complexes as well as the amount of cullins associ-
in the CRL network upon deneddylation to be revealed (Fig- ated with TAP-NEDD8 immune complexes upon inclusion of
ure S2; Table S4). OPT in lysis conditions (Figures S4A and S4B). Surprisingly,
In order to examine the effects of acute cullin deneddylation on only a small fraction (6%) of CUL1 was associated with CAND1
endogenous complexes, we immunoprecipitated endogenous in the absence of MLN4924, and this increased to 13% upon de-
CUL1 and subjected the complex to LC-MS/MS (Figure S3C). neddylation (Figure 5C). The CUL1/CSN fraction represented

956 Cell 143, 951–965, December 10, 2010 ª2010 Elsevier Inc.
A TSCs
B
- MLN4924
1 100 140 TAP-COPS6-IP + MLN4924 300 TAP-DCUN1D1-IP
1 120 250

Normalized TSCs

Normalized TSCs
A B 6 5 1 1D 100 *
**
L1 L2 L3 L4 L4 L5 PS PS D U
N 200
U U U U U U O O AN C 80
C C C C C C C C C D 150
- + - + - + - + - + - + - + - + - + - + MLN4924 CUL1
60
100
40
**

Cullins
CUL2
CUL3 50 **
CUL4A 20
CUL4B
CUL5 0 0
CUL7

CUL1

CUL2

CUL3

CUL4A

CUL4B

CUL5

CUL1

CUL2

CUL3

CUL4A

CUL4B

CUL5
NEDD8
CAND1
DCUN1D1
COPS1
COPS2
COPS3 TAP-COPS5-IP TAP-CAND1-IP

CSN
COPS4 80 30
COPS5 *
COPS6
COPS7A
70 ** 25
COPS7B *

Normalized TSCs

Normalized TSCs
COPS8 60 * 20
50
40 15
30 10

CUL1 adaptors
20
5
10
0 0

CUL1

CUL2

CUL3

CUL4A

CUL4B

CUL5

CUL1

CUL2

CUL3

CUL5
CUL4A

CUL4B
C 70 COPS1 120 COPS5 - MLN4924
60 + MLN4924

Normalized TSCs

Normalized TSCs
100
50

CUL2 adaptors
80
40
60
30
**
20 * 40 **
10 20
*
0 0

CUL1

CUL2

CUL3

CUL1

CUL2
CUL4A

CUL4B

CUL3
CUL5

COPS6

COPS5

CUL4A

CUL4B

CUL5

COPS6

COPS5
TAP-IP TAP-IP

120
CAND1
D
CUL3 adaptors

TAP- TAP- TAP- TAP- TAP-


100
** CUL1 CUL2 CUL3 CUL4A CUL4B
**
Normalized TSCs
MLN4924 - + - + - + - + - +
80 * 50
inputs
37 IB:CSN5
60
37 IP:HA
IB:CSN5
40
TAP- TAP- TAP- TAP- TAP-
20 CUL5 Nedd8 COPS6 DCUN1D1 CAND1
* MLN4924 - + - + - + - + - +
0 inputs
37
IB:CSN5
CUL1

CUL2

CUL3

CUL4A

CUL4B

CUL5

DCUN1D1
TAP-IP
37 IP:HA
IB:CSN5
CUL4 adaptors

E
25 TAP-CUL1-IP - MLN4924 TAP-CUL5-IP
50
+ MLN4924
45
Normalized TSCs

20

Normalized TSCs
40
35
15 30
25
CUL5 adaptors

10 20
15
5 10
5
0 0
FBXW2
SKP1
SKP2
FBXL15
FBXL18

FBXO3
FBXO7
FBXO9
FBXW11
BTRC

TCEB1
TCEB2
FBXO11
FBXO17
FBXO21
FBXO22

ASB13
ASB3
ASB6
SOCS6
interactor

TAP-CUL3-IP TAP-CUL2-IP TAP-CUL4A-IP DDB1


40
40 * 90 500
35
**
35 80 - MLN4924 450
Normalized TSCs

Normalized TSCs

Normalized TSCs

Normalized TSCs

30 + MLN4924 400
30 70
* 350
25 60
25
50 300
20 20 * 250
40
15 15 200
30 *
10 10 20
* 150
* 100
5 5 10 50
0 0 0 0
ARMC5
BTBD1
BTBD2
KBTBD2
KBTBD4
KBTBD6
KBTBD7
KCTD10
KLHDC5
KLHL12
KLHL13
KLHL15
KLHL22
KLHL23
KLHL24
KLHL26
KLHL36
KLHL8
KLHL9

TCEB1
TCEB2

FEM1B

KLHDC2
KLHDC3

VHL
APPBP2

KLHDC10

LRRC14
PPIL5

ZYG11B

AMBRA1

VPRBP
DCAF16
CRBN
DDA1
DDB2
DTL
ERCC8
IQWD1
TRPC4AP

WDR22
WDR23

WDTC1
WDR21A

WDR40A
WDR42A

TAP-IP CUL4A CUL4B


interactor
interactor

Figure 4. Acute NAE1 Inhibition Does Not Globally Alter the CRL Network
(A) Extracts from 293T cells expressing the indicated proteins (with or without 4 hr MLN4924 treatment) were immunoprecipitated with a-HA, and associated
proteins were identified by LC-MS/MS. Bait-normalized TSCs for associated CRL components are shown.
(B) The relative abundance of cullins associated with COPS6, DCUN1D1, COPS5, or CAND1 immune complexes with (red bars) or without (blue bars) MLN4924
treatment.
(C) Normalized TSCs for COPS1, COPS5, or CAND1 associated with the indicated immune complexes with (red bars) and without MLN4924 (blue bars) treatment.
(D) Extracts from 293T cells expressing the indicated proteins (with or without 4 hr MLN4924 treatment) were probed with antibodies against COPS5. Bait
complexes were immunoprecipitated with a-HA and immunoblotted for COPS5.
(E) Bait-normalized TSCs for a subset of adaptor proteins associated with their cognate cullin with (red bars) and without MLN4924 (blue bars) treatment.
Error bars: SD of duplicate measurements (*,** = p value < 0.05, 0.01, respectively, by Student’s t test). See also Figure S3 and Table S3.

26% of the total CUL1 in untreated cells, and this decreased to tion calculated from multiplex AQUA analysis of all CSN subunits
10% upon NAE1 inhibition. For simplicity, unless otherwise (15 peptides). Interestingly, the majority (73%) of CUL1 was
noted all CSN measurements represent the average mole frac- associated with SKP1, and this fraction increased slightly after

Cell 143, 951–965, December 10, 2010 ª2010 Elsevier Inc. 957
A
Cullins C 293T
293T/TAP-CRL or regulator 0.90
TAP-CUL1-IP
1 4B

Mole Fraction of total CUL1


+/- MLN4924 2 3 4A 0.80
- MLN4924
+ MLN4924
0.70
CSN 5 7 0.60
Adaptors 0.50 **
DCUN1D1 CUL1 Adaptor
NEDD8 SKP1 SKP1 DDB1 BTRC FBXW11
0.40
0.30 **
Regulators 0.20 **
IP CUL 0.10 ** **
CUL1 DCUN1D1 complexes CAND1 COPS1 COPS4 COPS7A
0.00

NEDD8

CAND1

BTRC

FBXW11
CSN

SKP1
CAND1 DCUN1D1 COPS2 COPS5 COPS7B

NEDD8 COPS3 COPS6 COPS8


D HeLa
0.60 TAP-CUL1-IP
CompPASS/CORE Spike in CRL AQUA

Mole Fraction of total CUL1


LC-MS - MLN4924 *
0.50
Quantitative analysis reference peptides + MLN4924

0.40

B TAP-CUL1 IP TAP-CUL1 IP
0.30
Light endogenous Light endogenous
562.63 596.33
100 peptide 100 peptide 0.20
564.63 90
Relative Abundance

Relative Abundance

90
80 Heavy AQUA 80 ** **
Heavy AQUA 0.10
70 reference peptide 70 reference peptide
60 60 0.00

NEDD8

CAND1

CSN

SKP1
50 50
40 40
untreated 598.67 untreated
30 30
20 NEDD8 20
CUL1
10 10
EIEIDIEPTDKvER LLETHIHNQGlAAIEK
0 0
560 562 564 566 568 570 590 595 600 605
m/z m/z

100 564.63 596.33


100
Relative Abundance

90 90
Relative Abundance

80 80
70 70
60 60
50 50
40 40 598.67
30 MLN4924 30
MLN4924
20 20
NEDD8 CUL1
10 10
EIEIDIEPTDKvER LLETHIHNQGlAAIEK
0 0
560 562 564 566 568 570 590 595 600 605
m/z m/z

Figure 5. Application of Multiplex AQUA for Quantitative Analysis of the CRL Network
(A) Schematic multiplex AQUA-based workflow. TAP-CUL1 was immunoprecipitated, eluted, and digested with trypsin. After peptide desalting, 100 fmoles of
heavy-labeled AQUA reference peptide library targeting the indicated CRL components was added prior to LC-MS analysis. The colored lines under each
CRL component indicate the number of AQUA peptides for that particular protein utilized in this study. See also Table S6.
(B) MS chromatogram showing a heavy reference peptide (black) and its corresponding endogenous light peptide (red) for NEDD8 (left) and CUL1 (right) before
(top) and after (bottom) MLN4924 treatment present within TAP-CUL1 immune complexes. m/z values are shown together with the corresponding peptide
sequence (heavy-labeled amino acid in red).
(C) The concentration of the indicated components within TAP-CUL1 immune complexes from 293T cells was determined using multiplex AQUA. The mole frac-
tion of CUL1 was then calculated by the ratio of abundances of the individual components and CUL1 with (red bars) and without MLN4924 (blue bars) treatment.
CSN represents the average mole fraction calculated from AQUA measurements against each of the CSN subunits.
(D) The mole fraction of TAP-CUL1 expressed in HeLa cells bound to individual CRL components with (red bars) and without MLN4924 (blue bars) treatment.
Error bars: SD of duplicate measurements (*,** = p value < 0.05, 0.01, respectively, by Student’s t test). See also Figure S4.

MLN4924 treatment (Figure 5C). This suggests that the majority binding to CUL1. In contrast, 13% of CUL1 was associated
of CUL1 is potentially occupied with F-box proteins under with CAND1 and this did not appreciably change upon deneddy-
steady-state conditions, and acute deneddylation of the cullin lation (Figure 5D). In both 293T and HeLa cells, we found that
does not decrease this fraction, contrary to the prevailing model. CUL1, CUL3, CUL4A, and CUL4B are the most abundant cullins
Analogous measurements of TAP-CUL1 expressed in HeLa cells associated with TAP-NEDD8 (Figures S4B and S4D). Further, the
(Figure 5D) revealed a smaller fraction of neddylated CUL1 (8%) absolute amounts of SKP1 and CUL1 present within NEDD8
and somewhat reduced levels of CSN and SKP1 (6 and 50%, immune complexes from 293T cells are equivalent, indicating
respectively) when compared to 293T cells. As observed with that the entirety of the neddylated cullin fraction also contains
293T cells, deneddylation led to an 2-fold reduction in CSN SKP1 (Figure S4B).

958 Cell 143, 951–965, December 10, 2010 ª2010 Elsevier Inc.
Neddylation Is Dispensable for CUL1 Complex Assembly Quantitative Assessment of CUL1 Complexes upon
but Is Required for CAND1 Association Depletion of COPS5, CAND1, or SKP1
To investigate the requirement of neddylation on complex Previous reports suggested that reduction of COPS5 or CAND1
assembly by proteomics, we created cells with inducible ex- levels resulted in hyperactivation of CRLs leading to the inappro-
pression of non-neddylatable CUL1K720R or CUL1 dominant priate degradation of unstable adaptor proteins, thereby para-
negative (CUL1DN). CUL1DN binds SKP1-F-box protein com- doxically inactivating CRL function (Hotton and Callis, 2008). It
plexes but does not interact with either CAND1 or CSN and therefore remained possible that reduction of CSN or CAND1
therefore serves as a control for adaptor assembly. Western may have large effects on CRL network architecture not seen
blotting confirmed that CUL1K720R was not neddylated (Fig- after acute NAE1 inhibition. Using siRNA oligos targeting either
ure 6B). We found that CUL1K720R assembled with CSN, the catalytic COPS5 subunit or CAND1, we achieved a 90%
SKP1, and a majority of F-box proteins to the same extent as reduction of COPS5 levels with one of the two siRNA oligos
wild-type CUL1 (Figure 6A). As seen previously (Liu et al., and a similar reduction of CAND1 levels with both siRNA
2002), CUL1K720R displayed a 10-fold reduction in CAND1 duplexes (Figure S5B). Surprisingly, the amount of neddylated
binding compared to wild-type CUL1 (Figure 6C). CUL1DN asso- CUL1 was largely unaffected despite greater than 90% reduc-
ciated with F-box proteins but, as expected, did not bind CSN or tion in either COPS5 or CAND1 levels. This unexpected result
CAND1 (Figures 6C and 6D). Quantitative MS analysis may reflect the lack of OPT in previous experiments, which
confirmed that CUL1K720R was deficient in CAND1 binding, underestimated the amount of neddylated cullins in control
leading to an increase in the mole fraction of total CUL1 associ- treated samples. Quantitative assessment of CUL1 complexes
ated with SKP1 approaching 100% (Figure 6E). Compared to after knockdown revealed that loss of COPS5 did not result in
MLN4924-treated CUL1, CUL1K720R bound 2-fold more CSN a significant loss of association with the larger CSN complex
despite both complexes being completely deneddylated and (Figure S5D) despite a reduction in the amount of the COPS5
suggesting that CSN can interact with CRLs independent of subunit associated with CUL1, which is in agreement with
prior neddylation (Figure 6E). As seen by spectral counting, previous studies (Figures S5B and S5D) (Sharon et al., 2009).
CUL1K720R associated with the F-box proteins BTRC The fraction of CAND1 bound to CUL1 remained at similar levels
(b-TRCP1) and FBXW11 (b-TRCP2), albeit reduced by 2-fold in control knockdown cells compared to knockdown of COPS5.
compared to wild-type CUL1 as measured by AQUA (Figure 6E). As expected, knockdown of CAND1 resulted in a 3-fold reduc-
To confirm that F-box proteins similarly associated with wild- tion in the amount of CAND1 bound to CUL1 and a concomitant
type CUL1 and CUL1K720R, we transiently expressed five increase in the amount of SKP1 bound to CUL1 from 62% in
FLAG-tagged-F-box proteins with either wild-type MYC-CUL1 untreated cells to 75% after CAND1 depletion (Figure S5C).
or MYC-CUL1K720R. Subsequent FLAG immunoblotting of the Knockdown of CAND1 had no effect on the amount of total
MYC-IP revealed no difference in F-box binding between wild- CSN bound to CUL1 (Figure S5C). These results suggest that
type and neddylation-defective CUL1 (Figure S5). Further, genetic reduction of CSN activity does not alter the overall
MLN4924 treatment of cells expressing wild-type MYC-CUL1 CRL stoichiometry and that the fraction of the adaptor-assem-
also showed no decrease in ability to associate with coex- bled ligase versus the inhibited CAND1-bound complex can be
pressed F-box proteins, confirming that acute deneddylation altered by lowering CAND1 levels.
does not affect F-box protein association with CUL1 We also examined the effect of depletion of SKP1 on CSN and
(Figure S5A). CAND1 association with HA-CUL1 (Figures S5C and S5E). With
three of four siRNAs targeting SKP1, there was an 40% reduc-
Absence of Global Reorganization of the CRL Network tion in the mole fraction of CUL1 associated with SKP1 not seen
upon Prolonged Deneddylation with control siRNA or the ineffective SKP1 siRNA oligo 1. This
The neddylation cycle paradox emerges from the finding that was accompanied by an increase in the fraction of CUL1 bound
the CSN functions to positively regulate CRL function in vivo. to CAND1 (from 6% to 50%) (Figure S5E). These data are
As such, we considered the possibility that the absence of consistent with mutually exclusive binding of SKP1 and
global reorganization of the CRL network in the experiments CAND1 to CUL1 and reveal that SKP1 binding predominates
presented thus far reflects the relatively short time period in vivo.
(4 hr) allowed for reorganization after NAE inhibition. However,
the mole fraction of TAP-CUL1 associated with SKP1, CSN, Application of Multiplex AQUA for Assessment of CRL
and CAND1 was essentially static from 2 to 16 hr of Occupancy
MLN4924 treatment (Figures 6F and 6G). Immunoblotting of The modular nature of CRL complexes and the presence of vari-
cell extracts revealed complete loss of neddylation after 2 hr able regulatory proteins allow for the construction of a wide
of MLN4924 treatment with a concomitant increase in the variety of heterogeneous assemblages. For example, when
abundance of the well-characterized CUL3/KEAP1 substrate considering only NEDD8, CAND1, CSN, and SKP1 as possible
NRF2 (Figure 6F). Over 70% of CUL1 was associated with CUL1-interacting proteins, it is possible to envision nine distinct
SKP1 in untreated cells, and this level was maintained 16 hr CRL assemblies (Figure 7A). Although this does not consider the
after NAE inhibition. Thus, even upon prolonged deneddylation, heterogeneity of the different F-box proteins, we assume that
CUL1-based CRL complexes are not globally converted to assemblies containing SKP1 represent complexes that are
a CUL1-CAND1 complex, as would be predicted by the current potentially assembled with F-box proteins. The quantitative
model. nature of AQUA allowed us to determine the contribution of

Cell 143, 951–965, December 10, 2010 ª2010 Elsevier Inc. 959
A 1 TSCs 50 C CAND1

90 ** ** 40 CUL1
** CUL1+MLN
35
LN
80
CUL1K720R

R
20
+M

Normalized TSCs

Normalized TSCs
N
K7
CUL1DN

D
70 30
L1 L1

1
UL

UL
U U 60
C C

C
25
CUL1 50
CUL7 20
CAND1 40
COPS1 15
COPS2 30
COPS3 10
20
COPS4
COPS5 10 5
COPS6
0 0
COPS7A

CUL1
CUL1+MLN
CUL1K720R
CUL1 DN

COPS1

COPS2

COPS3

COPS4

COPS5

COPS6

COPS7B

COPS8
COPS7A
COPS7B
COPS8
SKP1
SKP2
FBXL12 D
FBXL14 40 CUL1
FBXL15 CUL1+MLN
FBXL17 35
Normalized TSCs CUL1K720R
FBXL18
FBXO10
30 CUL1DN
FBXO11 25
FBXO17
FBXO18 20
FBXO21
FBXO22 15
FBXO3
FBXO30 10
FBXO31
5
FBXO33
FBXO42 0
FBXO44
SKP2

BTRC
FBXL12
FBXL14
FBXL15
FBXL18
FBXO10
FBXO17
FBXO18
FBXO21
FBXO22
FBXO3
FBXO30
FBXO31
FBXO33
FBXO42
FBXO6
FBXO7
FBXO9
FBXW11

FBXW2
FBXW5
FBXW9
FBXO6
FBXO7
FBXO9
FBXW11
BTRC
FBXW2
FBXW5
FBXW8 E CUL1
FBXW9 1.20 0.07
Mole Fraction of total CUL1

** ** CUL1+MLN

Mole Fraction of total CUL1


** **
CUL1K720R
B 1.00 0.06

0.05
0.80
MLN4924 + 0.04
CUL1K720R + 0.60
0.03 ** **
TAP-CUL1 + + 0.40
** ** 0.02
100 0.20 ** ** 0.01

75 0.00 0.00
CAND1 CSN SKP1 BTRC FBXW11

IB:HA

G 0.90 TAP-CUL1-IP NEDD8


CAND1
Mole Fraction of total CUL1

F inputs IP:HA
0.80 SKP1
CSN
0.70
0 2 4 8 16 0 2 4 8 16 hrs MLN4924
0.60
100
0.50
IB:HA
0.40
0.30
150
IB:CAND1 0.20
** ** **
0.10 ** ** **
** ** **
37 IB:COPS5 0.00
hrs MLN4924 0 2 4 16
100
IB:NRF2
* 0.07
TAP-CUL1-IP
BTRC
Mole Fraction of total CUL1

FBXW11
37 IB:COPS5 0.06

0.05

0.04 ** ** **

0.03

0.02 ** ** **
0.01

0.00
hrs MLN4924 0 2 4 16

Figure 6. Quantitative Proteomic Analysis of Neddylation-Deficient CUL1 Complexes and Time-Course Analysis of CUL1 Complexes with
MLN4924 Treatment
(A) Bait-normalized TSCs of selected CRL components associated with wild-type TAP-CUL1 (with or without 4 hr MLN4924 treatment), a CUL1K720R mutant, and
dominant-negative CUL1 (CUL1DN).
(B) HA-immunoblot of lysates from cells stably expressing wild-type TAP-CUL1 (with or without 4 hr MLN4924 treatment) or TAP-CUL1K720R.

960 Cell 143, 951–965, December 10, 2010 ª2010 Elsevier Inc.
each of these species to the total occupancy of CUL1. Under likely reflects the presence of a large monomeric pool of
steady-state conditions in 293T cells, 19% of CUL1 is unoccu- COPS5 (Tomoda et al., 2002). Interestingly, CUL4B represents
pied whereas greater than 70% contains SKP1, of which the the largest fraction of cullins bound to CSN with 38% occupancy
majority is neddylated (Figure 7B). Note that we are unable to of COPS6 compared to CUL1 with 9% occupancy (Figure 7D).
identify RBX1 peptides in association with CUL1 and for the This underscores our finding that CRL association with CSN
purposes of this discussion, we expect that what we refer to varies depending upon the individual CRL complex examined.
as unoccupied CUL1 is actually associated with RBX1. In Finally, we also measured the fraction of CAND1 that is in
previous studies (Olma et al., 2009; Wolf et al., 2003) and here, complex with cullins. Consistent with spectral counting
almost half of the CSN-bound fraction of CUL1 does not contain (Figure 4), CUL1, CUL4B, and CUL5 represent 95% of the cullins
NEDD8, suggesting that either CSN remains associated with in complex with CAND1 (Figure 7E). Interestingly, less than half
CUL1 after deneddylation or neddylation is not required for of the total CAND1 was in complex with cullins, and this
CSN binding. As CSN associates with neddylation-deficient percentage increased to only 57% after treatment with
CUL1 (Figure 6C), we favor the latter possibility. MLN4924 treat- MLN4924 (Figure 7E). Thus, unneddylated cullins are not
ment resulted in a complete loss of all neddylated species and converted to cullin-CAND1 complexes despite the presence of
a decrease in the amount of unoccupied CUL1 to 3.8%, reflect- available CAND1, suggesting that additional regulatory events
ing increased SKP1 and CAND1 binding. Analogous measure- may be required to facilitate assembly of CAND1 onto unneddy-
ment of CUL1 occupancy from TAP-CUL1 expressed in HeLa lated adaptor-loaded CRL complexes. CAND1 occupancy
cells revealed an increase in the amount of unoccupied CUL1 increased to 85% when OPT was omitted from the lysis buffer
resulting from the observed reduction of CUL1 neddylation as (Figure S6A), indicating that excess CAND1 is available to bind
compared to 293T cells (Figure S6C). This suggests that CRL to in vitro CSN-mediated deneddylated cullins. Taken together,
occupancy and possibly the mechanisms that govern CRL our data necessitate a redefinition of the dynamic model of
assembly may vary between cell types. CRL regulation, where upon translation CUL1 is assembled
Occupancy determinations for CUL4B expressed in 293T cells with SKP1, which in turn is neddylated and CRL activity is modu-
revealed quantitative differences in CUL4B occupancy as lated by successive cycles of CSN-mediated deneddylation and
compared to CUL1 complexes. CUL4B was neddylated to NAE1-dependent neddylation without intervening sequestration
a similar extent as CUL1 but contained less bound DDB1 and by CAND1 (Figure 7F).
CAND1, 40% and 1%, respectively, but more CSN, 40%,
compared to CUL1 (Figure 7C). As such, we observed an DISCUSSION
adaptor-free CSN-bound CUL4B complex under steady-state
conditions, an assembly not seen in CUL1 complexes (Fig- CRLs and the Neddylation Cycle
ure 7C). Conversion of CUL4B to a completely unneddylated Over a decade of research on CRL function and regulation has
state by MLN4924 addition did not substantially alter the fraction elucidated the molecular identity of each of the individual CRL
of CUL4B bound to CSN, DDB1, or, surprisingly, CAND1. complexes as well as the myriad of cellular pathways that
However, MLN4924 treatment dramatically increased the CRLs impinge upon (Petroski and Deshaies, 2005). However,
amount of completely unoccupied CUL4B at the expense of a quantitative snapshot of the CRL network landscape has yet
the neddylated, but otherwise uncomplexed, CUL4B fraction. to be accomplished. By utilizing a quantitative multiplex AQUA
Examination of CUL4A expressed in HeLa cells revealed approach, we provide a description of CRL occupancy and the
CUL4A occupancy to be nearly identical to CUL4B expressed effect of acute deneddylation on CRL network architecture.
from 293T cells (Figures S4C and S6D). The application of multiplex AQUA was essential in describing
We also determined the fraction of CSN occupied by cullins the molecular architecture of the CRL network. However, we
measured from TAP-COPS6 or TAP-COPS5 complexes. In anticipate that as quantitative mass spectrometry techniques
untreated cells, cullins occupy 60% and 40% of the total continue to improve, the precise determination of CRL
COPS6 or COPS5, respectively (Figure 7D). The total occupancy occupancy determined in this study will likely be further refined.
decreases with MLN4924 treatment but is more apparent in It should be noted that, although validated in many systems, utili-
COPS5. The decrease in COPS5 occupancy relative to COPS6 zation of tryptic peptides as surrogates for proteins may not

(C) Normalized TSCs for CAND1 (left) and CSN subunits (right) present in wild-type untreated and MLN4924-treated TAP-CUL1, TAP-CUL1K720R, and TAP-
CUL1DN immune complexes.
(D) Normalized TSCs for a subset of F-box proteins present in wild-type untreated (blue bars) and MLN4924-treated (red bars) TAP-CUL1, TAP-CUL1K720R (green
bars), and TAP-CUL1DN (purple bars) immune complexes.
(E) Multiplex AQUA analysis showing the mole fraction of the indicated CUL1-associated proteins present in untreated (blue bars) and MLN4924-treated (red
bars) TAP-CUL1 and TAP-CUL1K720R (green bars) HA immune complexes.
(F) Either extracts from 293T cells expressing TAP-CUL1 (with or without 1 mM MLN4924 treatment for 2, 4, 8, or 16 hr) were immunoblotted directly or a-HA
immune complexes were probed with the indicated antibodies. * indicates nonspecific background band.
(G) (Top) Multiplex AQUA analysis of TAP-CUL1 immune complexes from (F) showing the mole fraction of NEDD8 (blue bars), CAND1 (red bars), SKP1 (green
bars), and CSN (purple bars) bound to CUL1 with increasing time of MLN4924 treatment. (Bottom) Multiplex AQUA analysis of TAP-CUL1 immune complexes
from (F) showing the mole fraction of BTRC (blue bars) and FBXW11 (red bars) bound to CUL1.
Error bars: SD of duplicate measurements (*,** = p value < 0.05, 0.01, respectively, by Student’s t test, comparison between untreated and MLN time points). See
also Figure S5.

Cell 143, 951–965, December 10, 2010 ª2010 Elsevier Inc. 961
CUL1 N8 CAND1 CSN SKP1
A Adaptors
B - + MLN4924
Fr. 1 CUL1 Fr. 2

Mole Fraction of total CUL1


1.00 0.1446 0.0019 CUL1-N8-SKP1-CSN
CUL1 CUL1 0.0000 0.0000 CUL1-N8-CSN
Fr. 3 N8 Fr. 4 0.80 0.3018 0.0039 CUL1-N8-SKP1
0.0174 0.0002 CUL1-N8
CUL1 CUL1 CUL1 CUL1 0.60
SKP1 SKP1 0.1157 0.0984 CUL1-SKP1-CSN
N8 N8
Adaptors Adaptors 0.40 0.0000 0.0000 CUL1-CSN
0.1658 0.7266 CUL1-SKP1
0.20 0.0654 0.1212 CUL1-CAND1
CSN CSN CSN
CSN 0.00 0.1893 0.0477 CUL1
CUL1 CUL1 CUL1
N8 SKP1 N8 SKP1 CUL1 MLN4924 - +
Fr. 8 Adaptors Fr. 9 Fr. 13 Adaptors Fr. 14 CUL1
CAND1
CUL1 CUL1 CUL1 CUL1 Fr. 11
N8 SKP1 N8 SKP1 C
Fr. 7
Adaptors
Fr. 10 Fr. 12
Adaptors
Fr. 15 - + MLN4924

Mole Fraction of total CUL4B


0.0387 0.0010 CUL4B-N8-DDB1-CSN
α:[SKP1] in N8 IP / [CUL1] in N8 IP 1.00
0.1058 0.0031 CUL4B-N8-CSN
β:[N8] in CSN6 IP / Σ[Cullins] in N8 IP
γ:[SKP1] in CSN6 IP / [CUL1] in CSN6 IP 0.80 0.0448 0.0011 CUL4B-N8-DDB1
Neddylated fractions Non-Neddylated fractions 0.2743 0.0079 CUL4B-N8
0.60 CUL4B-DDB1-CSN
Fr. 1 : [N8] in CUL1 IP / [CUL1] Fr. 2 : 1 - Fr. 1 0.1314 0.1499
Fr. 3 : α x Fr. 1 Fr. 4 : ([SKP1] in CUL1 IP/[CUL1]) - Fr. 3 0.40 0.1407 0.2021 CUL4B-CSN
Fr. 5 : β x Fr. 1 Fr. 6 : (Mean ([CSN subunit] in CUL1 IP/[CUL1])) - Fr. 5 0.1519 0.1733 CUL4B-DDB1
Fr. 7 : Fr. 3 - Fr. 8 Fr. 11 : [CAND1]/[CUL1]
Fr. 8 : Fr. 5 x γ Fr. 12 : Fr. 4 - Fr. 13 0.20 0.0100 0.0040 CUL4B-CAND1
Fr. 9 : Fr. 5 - Fr. 8 Fr. 13 : Fr. 6 x γ 0.1024 0.4577 CUL4B
Fr. 10 : Fr. 1 - (Fr. 7 + Fr. 8 + Fr. 9) Fr. 14 : Fr. 6 - Fr.13 0.00
Fr. 15 : Fr. 2 - (Fr. 11 + Fr. 12 + Fr. 13 + Fr. 14) MLN4924 - +

D E
0.8 0.7
COPS6 COPS5 TAP-IP
0.7
- + - + MLN4924 0.6
** - + MLN4924
Mole Fraction of CSN

Mole Fraction of CAND1


0.6 0.0967 0.0654 0.0521 0.0171 CUL1 0.0088 0.0025 CUL7
0.0103 0.0070 0.0038 0.0019 CUL2 0.5 0.1092 0.0861 CUL5
0.5
* 0.0456 0.0320 0.0845 0.0246 CUL3 0.1003 0.2412 CUL4B
0.4
0.4 0.0636 0.0441 0.0384 0.0239 CUL4A 0.0000 0.0000 CUL4A
0.3805 0.3360 0.1846 0.1230 CUL4B 0.3 0.0596 0.0245 CUL3
0.3
0.0017 0.0013 0.0023 0.0011 CUL5 0.0118 0.0032 CUL2
0.2 0.0004 0.0001 0.0013 0.0017 CUL7 0.2 0.1671 0.2268 CUL1
0.1 0.1

0.0 0.0
MLN4924 - + - + MLN4924 - +
TAP-IP COPS6 COPS5

R CUL1 SKP1 R CUL1 SKP1


SKP1
Adaptors
Adaptors N8 CSN Adaptors
CSN R CUL1 SKP1
RCUL1 A) Newly Adaptor X
CAND1
synthesized CUL1 CSN CSN ? With or without
SKP1
SKP1 NEDD8 or CSN
Adaptors N8
R CUL1 R CUL1 SKP1 R CUL1 SKP1 Adaptor X
Adaptors Adaptors
N8

B) adaptor specific
cullin sequestration
(small number
of adaptors)
R CUL1 SKP1 R CUL1 SKP1 R CUL1 SKP1
Adaptor Z Adaptor Z Adaptor Z

R CUL1 SKP1 R CUL1 SKP1 R CUL1 SKP1


Adaptor Y Adaptor Y Adaptor Y
C) adaptor independent
sequestration of a small R CUL1 SKP1 R CUL1 SKP1 R CUL1 SKP1
fraction of CUL1
Adaptor X Adaptor X Adaptor X

Figure 7. Application of Multiplex AQUA for Assessment of CRL Occupancy


(A) Schematic diagram using the CUL1 CRL as an example to show how each of the nine different assemblages are calculated using multiplex AQUA measure-
ments. The formulas used to calculate the abundance of each fraction are depicted.
(B) The contribution of each of the assemblages depicted in (A) to the total occupancy of TAP-CUL1 immune complexes with and without MLN4924 treatment.
The colors correspond to the colored assemblages in (A).
(C) The occupancy of TAP-CUL4B complexes calculated as in (A), except that DDB1 replaced SKP1. The ratio of DDB1 to CUL4B in NEDD8 immune complexes
represents the ratio of DDB1 to the combined concentrations of CUL4A and CUL4B. The colors correspond to the colored fractions in (A).
(D) Multiplex AQUA analysis of the mole fraction contribution of each of the seven cullins associated with TAP-COPS6 (left) or TAP-COPS5 (right) with or without
MLN4924 treatment.
(E) Multiplex AQUA analysis of the mole fraction contribution of each of the seven cullins associated with TAP-CAND1 with or without MLN4924 treatment. Error
bars: SD of duplicate measurements (*,** = p value < 0.05, 0.01, respectively, by Student’s t test).
(F) Refined model of CRL dynamicity.

962 Cell 143, 951–965, December 10, 2010 ª2010 Elsevier Inc.
precisely reflect protein abundances (Kirkpatrick et al., 2005) increase in the fraction of CUL1 bound to CAND1. In this case,
(see Extended Experimental Procedures). the cellular concentration of SKP1 dictates the proportion of
Cullin neddylation, and by extension CRL activity, is antago- adaptor-assembled CUL1. Alternatively, CUL1 that has
nized by both CSN-mediated deneddylation and CAND1-medi- previously been assembled with adaptor complexes and neddy-
ated cullin sequestration in vitro, whereas both CSN and lated may be the source of CUL1 found in complexes with
CAND1 are needed for optimal in vivo CRL activity in eukaryotes CAND1. This possibility is suggested by the finding that non-
(Bosu and Kipreos, 2008; Cope and Deshaies, 2003; Wolf et al., neddylatable CUL1K720R does not efficiently bind CAND1
2003). Current models invoke a neddylation-CAND1 cycle in vivo, despite the fact that CAND1 interacts with a large surface
wherein deneddylated and adaptor-free cullin is sequestered area on CUL1 (Goldenberg et al., 2004) (Figure 7F). We envision
by CAND1 and this complex is then used to build new cullin two possible scenarios for CAND1 sequestration of previously
complexes with a different adaptor molecule (Figure S1F). assembled and neddylated CUL1. In one scenario, CUL1 that
A central prediction of the model is that persistent cullin dened- was previously associated with a small subset of specific F-
dylation would result in loss of adaptor proteins from cullins and box proteins (Adaptor Z in Figure 7F, pathway B) might be
concomitant global sequestration of cullins by CAND1. selected for CAND1 sequestration. In principle, this subset could
However, our analysis of CRL network architecture with and represent adaptor proteins that are subject to adaptor instability
without cullin neddylation fails to validate this model in 293T or some other form of regulation that marks that CUL1 scaffold
and HeLa cells and suggests that substrate adaptor levels play for CAND1 sequestration. In the second scenario, CAND1 may
a central role in dictating the architecture of the CRL network target CUL1 independently of the identity of the previously asso-
(Figure 7F). ciated F-box protein, but given the CAND1 occupancy on CUL1,
only a small fraction of the total CUL1 pool would be shunted into
An Alternative Model for CRL Dynamics Revealed this pathway (Figure 7F, pathway C). The finding that a small
by Quantitative Proteomics fraction of CUL1 is associated with CAND1 even in the absence
For simplicity, we describe an alternate model in the context of of neddylation would favor pathway B and would explain why
the SCF (Figure 7F), but we envision that similar mechanisms loss of CAND1 function may result in phenotypes reflecting the
will apply for other CRLs. Newly synthesized CUL1-RING activity of a particular F-box protein without affecting global
assembles with adaptor complexes, which then promote CUL1 CRL architecture. In support of this model, loss of CAND1
neddylation (Bornstein et al., 2006; Chew and Hagen, 2007). function in C. elegans resulted in reduction of specific CRL func-
Once assembled, the SCF complex can associate with the tions while leaving others unaffected (Bosu et al., 2010). Further
CSN complex, and this can occur, in principal, with unneddy- studies are required to identify relevant pools of cullins that are
lated cullin as exemplified by the CUL1K720R mutant. However, assembled into CAND1 complexes and signals that control
given the decrease in CSN association with CUL1 seen after CAND1 sequestration. Moreover, further studies are required
acute deneddylation, we favor a model wherein CSN preferen- to determine whether the ‘‘free’’ pool of CAND1 identified by
tially or initially associates with neddylated forms of CRLs. AQUA and its association with cullins are regulated. Our studies
Association of CSN complexes with both neddylated and unned- examine the CRL network in asynchronous cells. It is also
dylated cullins suggests that binding of the CSN to the CRL is not possible that CAND1 restricts CRL activity upon a specific cell
rate-limiting for deneddylation and implies additional regulatory stimulus, state, or lineage where CRL activity may need to be
steps dictating NEDD8 removal from cullins. A large fraction of inhibited beyond CSN-mediated deneddylation. Indeed, we
CUL1 (70% in 293T cells) is in complex with SKP1 (and have found that the extent of CUL1 neddylation in HeLa cells is
presumably F-box proteins) independent of the neddylation 4-fold lower than that seen in 293T cells (Figures 5C and 5D)
status, suggesting that the assembly and activation pathway is yet only 14% of CUL1 is associated with CAND1 independent
dominant for the SCF. In this model, the formation of SCF of neddylation status. Interestingly, our analysis of CRL compo-
complexes is driven primarily by adaptor binding, and CAND1 nents in 293T cell extracts using multiplex AQUA (Figure S6E)
does not play a direct role in the assembly or reassembly revealed that the concentration of cullins is in excess of
process. NEDD8, suggesting that the extent of CRL neddylation may be
We found that only a small fraction of cullins are associated limited by the available pool of free NEDD8. This finding is in
with CAND1 in 293T cells, and association increases by less agreement with the observation that nearly all NEDD8 exits in
than 2-fold in response to acute deneddylation (Figure S6B), a conjugated form (Brownell et al., 2010). Unlike SKP1, the
indicating a minor role for CAND1 in the bulk steady-state DDB1 concentration in extracts is below that of the combined
dynamic remodeling of CRL complexes. However, it is clear CUL4A and CUL4B concentrations. This may explain why we
that CAND1 function is needed for CRL activity in multicellular observe a larger portion of CUL4B that does not have adaptors
eukaryotes (Bosu and Kipreos, 2008; Hotton and Callis, 2008), bound compared to CUL1 (Figures 7B and 7C). The relative
leading to the obvious question: What is CAND1 doing? An concentrations of SKP1, CUL1, and CAND1 in 293T cells are
answer to this question will likely require the elucidation of the consistent with the model shown in Figure S6E.
forms of cullins that serve as targets for CAND1 binding. The Although this work suggests a major role for substrate adaptor
simplest possibility is that newly synthesized CUL1 that escapes modules in dictating the architecture of the CRL network, several
productive interaction with SKP1 serves as the primary target for major issues are left unresolved. Are adaptor modules in rapid
CAND1 (Figure 7F, pathway A), a scenario that is reinforced by equilibrium with cullins, or once an adaptor is associated with
our finding that depletion of SKP1 leads to a concomitant a cullin, is it essentially irreversibly bound during the lifetime of

Cell 143, 951–965, December 10, 2010 ª2010 Elsevier Inc. 963
the CRL complex? Moreover, given that inhibition of NAE leads Cells were harvested 72 hr after transfection and processed for western blot-
to rapid deneddylation, it would appear that the neddylation ting or mass spectrometry analysis.
and deneddylation systems are poised to dynamically regulate
the extent of CRL neddylation on very short timescales. What SUPPLEMENTAL INFORMATION
then is the biological role of such dynamic control under physio-
Supplemental Information includes Extended Experimental Procedures, seven
logical conditions, given the apparent absence of a role of
figures, and five tables and can be found with this article online at doi:10.1016/
neddylation in assembly of substrate adaptors on cullins?
j.cell.2010.11.017.
Finally, what role does cell lineage play in dictating the abun-
dance of factors that control on and off rates for neddylation?
ACKNOWLEDGMENTS
The answers to these questions will likely require the develop-
ment of in vitro systems that fully recapitulate the dynamics of We thank Woong Kim, Ryan Kunz, and Fiona McAllister from the Gygi labora-
CRL assembly seen in vivo. Finally, this work suggests that multi- tory (Harvard Medical School) for assistance with the AQUA analysis, Harper
plex AQUA provides a powerful approach for elucidating how lab members John Lydeard for reagents, Mat Sowa for bioinformatics assis-
cellular perturbations affect the organization of signaling tance, and Brenda O’Connell for a critical reading of the manuscript. This
work was supported by grants to J.W.H. from Millennium Pharmaceuticals,
networks.
the National Institutes of Health, and the Stewart Trust. E.J.B. is a Damon Run-
yon Fellow supported by the Damon Runyon Cancer Research Foundation
EXPERIMENTAL PROCEDURES (DRG 1974-08). J.W.H. is a consultant for Millennium Pharmaceuticals.
J.R. is an employee of Cell Signaling Technologies.
Plasmids, Cell Lines, and Protein Purification
Details of the retroviral plasmids (Sowa et al., 2009), cell culture procedures, Received: July 23, 2010
and antibodies used can found in the Extended Experimental Procedures. Revised: September 21, 2010
Four 15 cm dishes expressing a given TAP-CRL protein (with or without incu- Accepted: October 29, 2010
bation with MLN4924 [provided by Millennium Pharmaceuticals]) were Published: December 9, 2010
harvested and lysed with 3 ml lysis buffer (50 mM Tris, pH 7.5, 150 mM
NaCl, 0.5% NP-40, and Complete protease inhibitor tablet [Roche]). Where
REFERENCES
indicated, 2 mM 1,10-orthophenathroline or 1,7-orthophenathroline (Sigma)
was added to the lysis buffer. Cleared lysates were filtered through 0.45 mm
Bornstein, G., Ganoth, D., and Hershko, A. (2006). Regulation of neddylation
spin filters (Millipore Ultrafree-CL) and immunoprecipitated with 30 ml a-HA
and deneddylation of cullin1 in SCFSkp2 ubiquitin ligase by F-box protein
resin (Sigma). Endogenous a-CUL1 complexes were washed and digested
and substrate. Proc. Natl. Acad. Sci. USA 103, 11515–11520.
with trypsin on beads.
Bosu, D.R., and Kipreos, E.T. (2008). Cullin-RING ubiquitin ligases: global
regulation and activation cycles. Cell Div. 3, 7.
Mass Spectrometry and Quantitative Analysis
Immunoprecipitated complexes were washed three times with lysis buffer, Bosu, D.R., Feng, H., Min, K., Kim, Y., Wallenfang, M.R., and Kipreos, E.T.
exchanged into PBS, and eluted with 150 ml of 250 mg/ml HA peptide in (2010). C. elegans CAND-1 regulates cullin neddylation, cell proliferation and
PBS. Eluted complexes were precipitated with 10% trichloroacetic acid morphogenesis in specific tissues. Dev. Biol. 346, 113–126.
(TCA, Sigma) and pellets were washed three times with cold acetone. TCA Brownell, J.E., Sintchak, M.D., Gavin, J.M., Liao, H., Bruzzese, F.J., Bump,
precipitated proteins were resuspended in 50 mM ammonium bicarbonate N.J., Soucy, T.A., Milhollen, M.A., Yang, X., Burkhardt, A.L., et al. (2010).
(pH 8.0) with 10% acetonitrile and sequencing grade trypsin (Promega) at Substrate-assisted inhibition of ubiquitin-like protein-activating enzymes: the
a concentration of 12.5 ng/ml. Trypsin reactions were quenched by addition NEDD8 E1 inhibitor MLN4924 forms a NEDD8-AMP mimetic in situ. Mol.
of 5% formic acid and peptides were desalted using the C18 stagetip Cell 37, 102–111.
method. Tandem MS/MS data were searched using Sequest and a concate- Chew, E.H., and Hagen, T. (2007). Substrate-mediated regulation of cullin ned-
nated target-decoy IPI human database with a 2 Da mass window for data dylation. J. Biol. Chem. 282, 17032–17040.
generated using LTQ linear ion trap mass spectrometer (ThermoFinnigan)
Cope, G.A., and Deshaies, R.J. (2003). COP9 signalosome: a multifunctional
or LTQ-Velos and a 50 ppm mass window for data generated using an
regulator of SCF and other cullin-based ubiquitin ligases. Cell 114, 663–671.
LTQ-Orbitrap (ThermoFinnigan) instrument. All data were filtered to a 1%
false discovery rate (peptide level) prior to analysis using CompPASS Cope, G.A., Suh, G.S., Aravind, L., Schwarz, S.E., Zipursky, S.L., Koonin, E.V.,
(Sowa et al., 2009). and Deshaies, R.J. (2002). Role of predicted metalloprotease motif of Jab1/
For multiplex AQUA analysis, samples were resuspended with 100 fmoles of Csn5 in cleavage of Nedd8 from Cul1. Science 298, 608–611.
a library of N15C13-labeled reference peptides (see Table S6; Kirkpatrick et al., Duda, D.M., Borg, L.A., Scott, D.C., Hunt, H.W., Hammel, M., and Schulman,
2005) in 5% acetonitrile, 5% formic acid prior to analysis on an LTQ-Orbitrap. B.A. (2008). Structural insights into NEDD8 activation of cullin-RING ligases:
HPLC-purified AQUA reference peptides (Table S6) were quantified using Conformational control of conjugation. Cell 134, 995–1006.
colorimetric detection of primary amines by 2,4,6-trinitrobenzene sulfonic
Goldenberg, S.J., Cascio, T.C., Shumway, S.D., Garbutt, K.C., Liu, J., Xiong,
acid (TNBSA, Pierce) (see Extended Experimental Procedures). The ratios of
Y., and Zheng, N. (2004). Structure of the Cand1-Cul1-Roc1 complex reveals
extracted ion chromatograms for reference and endogenous peptide
regulatory mechanisms for the assembly of the multisubunit cullin-dependent
precursor ions (mass window = 20 ppm) were obtained using PINPOINT soft-
ubiquitin ligases. Cell 119, 517–528.
ware (Thermo) (see Table S5). Endogenous protein concentrations for the indi-
cated CRL components were determined from LTQ-Orbitrap analysis of 1 mg Hotton, S.K., and Callis, J. (2008). Regulation of cullin RING ligases. Annu. Rev.
of 293T whole-cell extract. Due to the low intensity of some endogenous Plant Biol. 59, 467–489.
peptide ions in whole-cell extract digests, ion chromatogram ratios were Jin, J., Cardozo, T., Lovering, R.C., Elledge, S.J., Pagano, M., and Harper, J.W.
determined by manual inspection of MS chromatograms. (2004). Systematic analysis and nomenclature of mammalian F-box proteins.
Genes Dev. 18, 2573–2580.
RNAi Kirkpatrick, D.S., Gerber, S.A., and Gygi, S.P. (2005). The absolute quantifica-
TAP-CUL1 cells were transfected with 20 nM siRNA duplexes (Dharmacon/ tion strategy: a general procedure for the quantification of proteins and post-
Thermo) using RNAiMAX (Invitrogen) according to manufacturer guidelines. translational modifications. Methods 35, 265–273.

964 Cell 143, 951–965, December 10, 2010 ª2010 Elsevier Inc.
Liu, J., Furukawa, M., Matsumoto, T., and Xiong, Y. (2002). NEDD8 modifica- itor of NEDD8-activating enzyme as a new approach to treat cancer. Nature
tion of CUL1 dissociates p120(CAND1), an inhibitor of CUL1-SKP1 binding 458, 732–736.
and SCF ligases. Mol. Cell 10, 1511–1518.
Sowa, M.E., Bennett, E.J., Gygi, S.P., and Harper, J.W. (2009). Defining the
Olma, M.H., Roy, M., Le Bihan, T., Sumara, I., Maerki, S., Larsen, B., Quadroni, human deubiquitinating enzyme interaction landscape. Cell 138, 389–403.
M., Peter, M., Tyers, M., and Pintard, L. (2009). An interaction network of the
mammalian COP9 signalosome identifies Dda1 as a core subunit of multiple Tomoda, K., Kubota, Y., Arata, Y., Mori, S., Maeda, M., Tanaka, T., Yoshida,
Cul4-based E3 ligases. J. Cell Sci. 122, 1035–1044. M., Yoneda-Kato, N., and Kato, J.Y. (2002). The cytoplasmic shuttling and
Petroski, M.D., and Deshaies, R.J. (2005). Function and regulation of cullin- subsequent degradation of p27Kip1 mediated by Jab1/CSN5 and the COP9
RING ubiquitin ligases. Nat. Rev. Mol. Cell Biol. 6, 9–20. signalosome complex. J. Biol. Chem. 277, 2302–2310.

Przytycka, T.M., Singh, M., and Slonim, D.K. (2010). Toward the dynamic inter- Wee, S., Geyer, R.K., Toda, T., and Wolf, D.A. (2005). CSN facilitates Cullin-
actome: it’s about time. Brief. Bioinform. 11, 15–29. RING ubiquitin ligase function by counteracting autocatalytic adapter insta-
Rabut, G., and Peter, M. (2008). Function and regulation of protein neddylation. bility. Nat. Cell Biol. 7, 387–391.
‘Protein modifications: beyond the usual suspects’ review series. EMBO Rep.
Wolf, D.A., Zhou, C., and Wee, S. (2003). The COP9 signalosome: an assembly
9, 969–976.
and maintenance platform for cullin ubiquitin ligases? Nat. Cell Biol. 5, 1029–
Saha, A., and Deshaies, R.J. (2008). Multimodal activation of the ubiquitin 1033.
ligase SCF by Nedd8 conjugation. Mol. Cell 32, 21–31.
Yang, X., Menon, S., Lykke-Andersen, K., Tsuge, T., Di Xiao, Wang, X., Rodri-
Schmidt, M.W., McQuary, P.R., Wee, S., Hofmann, K., and Wolf, D.A. (2009).
guez-Suarez, R.J., Zhang, H., and Wei, N. (2002). The COP9 signalosome
F-box-directed CRL complex assembly and regulation by the CSN and
inhibits p27(kip1) degradation and impedes G1-S phase progression via de-
CAND1. Mol. Cell 35, 586–597.
neddylation of SCF Cul1. Curr. Biol. 12, 667–672.
Sharon, M., Mao, H., Boeri Erba, E., Stephens, E., Zheng, N., and Robinson,
C.V. (2009). Symmetrical modularity of the COP9 signalosome complex Zhang, W., Ito, H., Quint, M., Huang, H., Noël, L.D., and Gray, W.M. (2008).
suggests its multifunctionality. Structure 17, 31–40. Genetic analysis of CAND1-CUL1 interactions in Arabidopsis supports a role
Siergiejuk, E., Scott, D.C., Schulman, B.A., Hofmann, K., Kurz, T., and Peter, for CAND1-mediated cycling of the SCFTIR1 complex. Proc. Natl. Acad. Sci.
M. (2009). Cullin neddylation and substrate-adaptors counteract SCF inhibi- USA 105, 8470–8475.
tion by the CAND1-like protein Lag2 in Saccharomyces cerevisiae. EMBO J. Zheng, J., Yang, X., Harrell, J.M., Ryzhikov, S., Shim, E.H., Lykke-Andersen,
28, 3845–3856. K., Wei, N., Sun, H., Kobayashi, R., and Zhang, H. (2002). CAND1 binds to un-
Soucy, T.A., Smith, P.G., Milhollen, M.A., Berger, A.J., Gavin, J.M., Adhikari, neddylated CUL1 and regulates the formation of SCF ubiquitin E3 ligase
S., Brownell, J.E., Burke, K.E., Cardin, D.P., Critchley, S., et al. (2009). An inhib- complex. Mol. Cell 10, 1519–1526.

Cell 143, 951–965, December 10, 2010 ª2010 Elsevier Inc. 965
Kinase Associated-1 Domains Drive
MARK/PAR1 Kinases to Membrane Targets
by Binding Acidic Phospholipids
Katarina Moravcevic,1,2 Jeannine M. Mendrola,1 Karl R. Schmitz,2 Yu-Hsiu Wang,4,5 David Slochower,2,5
Paul A. Janmey,2,3,5 and Mark A. Lemmon1,2,*
1Department of Biochemistry and Biophysics
2Graduate Group in Biochemistry and Molecular Biophysics
3Department of Physiology

University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA


4Department of Chemistry
5Institute for Medicine and Engineering

University of Pennsylvania, Philadelphia, PA 19104, USA


*Correspondence: mlemmon@mail.med.upenn.edu
DOI 10.1016/j.cell.2010.11.028

SUMMARY are tightly regulated. Others bind phosphatidylserine (PtdSer),


which is concentrated in the plasma membrane inner leaflet
Phospholipid-binding modules such as PH, C1, and (Yeung et al., 2008) and constitutes approximately 20% of phos-
C2 domains play crucial roles in location-dependent pholipid (Stace and Ktistakis, 2006).
regulation of many protein kinases. Here, we identify Many more cellular functions appear to depend on anionic
the KA1 domain (kinase associated-1 domain), found phospholipids than can be explained by currently understood
at the C terminus of yeast septin-associated kinases phospholipid-binding domains (Audhya et al., 2004; Halstead
et al., 2005; McLaughlin and Murray, 2005; Yu et al., 2004).
(Kcc4p, Gin4p, and Hsl1p) and human MARK/PAR1
Indeed, in a microarray-based analysis of the expressed S. cer-
kinases, as a membrane association domain that
evisiae proteome, over 100 proteins that contain no known lipid-
binds acidic phospholipids. Membrane localization binding domain were found to bind phosphoinositides (Zhu et al.,
of isolated KA1 domains depends on phosphatidyl- 2001). Here, we describe an analysis of the membrane associa-
serine. Using X-ray crystallography, we identified a tion properties of these yeast proteins, from which we have iden-
structurally conserved binding site for anionic phos- tified several additional potential phospholipid-binding domains.
pholipids in KA1 domains from Kcc4p and MARK1. We focus in this report on a membrane-targeting domain found
Mutating this site impairs membrane association of at the C terminus of the S. cerevisiae septin-associated protein
both KA1 domains and intact proteins and reveals kinases Kcc4p, Gin4p, and Hsl1p. These kinases are involved
the importance of phosphatidylserine for bud neck in septin organization or in the yeast morphogenesis checkpoint
localization of yeast Kcc4p. Our data suggest that that coordinates cell-cycle progression with bud formation (Lew,
2003; Longtine and Bi, 2003; Shulewitz et al., 1999). They
KA1 domains contribute to ‘‘coincidence detection,’’
become activated at the bud neck and are involved in septin
allowing kinases to bind other regulators (such as
ring assembly and/or promote Swe1p degradation to allow
septins) only at the membrane surface. These find- entry into mitosis (Barral et al., 1999; Sakchaisri et al., 2004).
ings have important implications for understanding The C-terminal phospholipid-binding domain of the septin-asso-
MARK/PAR1 kinases, which are implicated in Alz- ciated kinases is required for their bud neck localization and
heimer’s disease, cancer, and autism. function and appears to bind phosphatidylserine in vivo. Using
X-ray crystallography, we found that this phospholipid-binding
INTRODUCTION domain has the same fold as the KA1, or kinase associated-1
domain (Pfam accession PF02149), one of the only common
Regulation of cellular processes requires precisely controlled domains in protein kinases to which no function has yet been
intermolecular interactions that alter the location and/or activity ascribed (Manning et al., 2002; Tochio et al., 2006).
of effector proteins (Scott and Pawson, 2009), typically driven KA1 domains are also found at the C termini of mammalian
by protein modules that recognize specific features of proteins, Ser/Thr kinases that phosphorylate microtubule-associating
nucleic acids, or membranes (Seet et al., 2006). Several protein proteins (MAPs) such as tau, promoting their detachment from
modules recognize anionic membrane phospholipids, including microtubules and thus reducing microtubule stability (Drewes
PH, C2, PX, and FYVE domains (Lemmon, 2008). Some recog- et al., 1997). These kinases comprise the MARK/PAR1 family,
nize phosphoinositides (PtdInsPns), levels and locations of which which includes MAP/microtubule affinity-regulating kinase

966 Cell 143, 966–977, December 10, 2010 ª2010 Elsevier Inc.
(MARK) and partitioning-defective 1 or PAR1 (Matenia and Man- membrane association when overexpressed as a GFP fusion
delkow, 2009; Timm et al., 2008), as well as the S. cerevisiae protein in either S. cerevisiae or human HeLa cells (Figure 1B),
Kin1/2 kinases (Tassan and Le Goff, 2004). MARK/PAR1 kinases suggesting recognition of a lipid that is common to yeast and
are related to the AMP-activated protein kinase (AMPK)/Snf1 human cells, rather than association with a less abundant protein
family (Manning et al., 2002; Marx et al., 2010). They are target at the membrane.
frequently found associated with membrane structures and
participate in diverse processes from control of the cell cycle The Kcc4p C-Terminal Domain Binds
and polarity to intracellular signaling and microtubule stability Anionic Phospholipids
(Marx et al., 2010; Tassan and Le Goff, 2004). MARK/PAR1 As shown in Figure 1C, purified protein corresponding to resi-
kinases have been implicated in carcinomas, Alzheimer’s dis- dues 901–1037 from the Kcc4p C terminus (Kcc4p901-1037) binds
ease (through tau hyperphosphorylation), and autism (Gray ‘‘promiscuously’’ to PtdIns(4,5)P2 and other acidic phospho-
et al., 2005; Hurov et al., 2007; Maussion et al., 2008; Timm lipids in surface plasmon resonance (SPR) studies. Overlay
et al., 2008). We establish here that KA1 domains from both yeast studies of intact Kcc4p (Table S1A) showed a similar lack of
and human kinases bind anionic phospholipids, thus ascribing specificity, consistent with the binding to several phosphoinosi-
a function to this poorly understood domain and providing tides reported previously by Zhu et al. (2001). Kcc4p901–1037
important clues as to how activation of these AMPK-related bound with similar affinities to membranes containing 10%
kinases may be directly coordinated with membrane localization. (mole/mole) PtdIns(4,5)P2, 20% (mole/mole) phosphatidic acid
(PA), or 20% (mole/mole) PtdSer—all in a dioleoylphosphatidyl-
RESULTS choline (DOPC) background. The binding data fit well to simple
hyperbolic curves with apparent dissociation constant (KD)
Screen for Unidentified Phospholipid-Binding Domains values from 3–10 mM (Table S2), in the same range reported for
Zhu et al. (2001) reported phosphoinositide binding for 128 of several other phospholipid-interaction domains (Lemmon,
5800 protein products from S. cerevisiae open reading frames 2008). The amount of Kcc4p901–1037 bound at saturation (Bmax)
(ORFs) arrayed on proteome chips—excluding dubious ORFs scaled with anionic phospholipid content for PtdIns(4,5)P2 or
and integral membrane proteins. We selected 62 of these for PtdSer (Figure 1D). Interestingly, in all studies, Bmax was propor-
further analysis (15 of which were protein kinases), including all tional to the anticipated negative charge density on the SPR
‘‘strong binders’’ defined by Zhu et al. (2001) plus potentially sensorchip surfaces (rather than number of lipid molecules),
interesting ‘‘weak binders.’’ We first tested in vivo membrane assuming charge valences of 4, 2, and 1 for PtdIns(4,5)P2,
association of these 62 proteins using an S. cerevisiae Ras PA, and PtdSer, respectively, at pH 7.4 (McLaughlin and Murray,
rescue assay (Isakoff et al., 1998; Yu et al., 2004). Each protein 2005). As shown in Figure 1C, Bmax was approximately 2000
was fused to constitutively active (Q61L), nonfarnesylated, resonance units (RUs) for membranes containing either 10%
Ha-Ras and expressed in cdc25ts yeast cells—which harbor PtdIns(4,5)P2 (charge 4) or 20% PA (charge 2) and approxi-
a temperature-sensitive mutation in the Ras guanine nucleotide mately 1000 RUs for membranes containing 20% PtdSer (charge
exchange factor Cdc25p. If the test protein drives plasma mem- 1). These observations suggest that, rather than forming simple
brane recruitment of this Ha-Ras fusion, it promotes growth 1:1 complexes, binding stoichiometry depends on lipid charge
above the restrictive temperature (complementing the cdc25ts each Kcc4p901–1037 chain binding four times more PtdSer mole-
allele) by overcoming the block in endogenous Ras activation cules (charge 1) than PtdIns(4,5)P2 molecules (charge 4).
(Isakoff et al., 1998). Of the 62 proteins analyzed, 33 promoted We also used a centrifugation-based sedimentation assay to
membrane recruitment of constitutively active Ha-Ras (Fig- analyze Kcc4p901–1037 binding to small unilamellar vesicles (Kav-
ure S1A and Table S1A available online), consistent with them ran et al., 1998). Only background levels of Kcc4p901–1037 sedi-
harboring a phospholipid-binding domain. In qualitative lipid mented with vesicles with no net charge, i.e., those containing
overlays (Kavran et al., 1998), 21 of these 33 membrane-targeted 100% phosphatidylcholine (PC) or 20% (mole/mole) phosphati-
proteins also interacted in vitro with filter-bound anionic phos- dylethanolamine (PE) in a PC background (Figure 1E). By con-
pholipids (Table S1A), displaying a broad range of specificities. trast, vesicles containing 20% (mole/mole) of the anionic phos-
Several of the candidate Ras rescue-positive proteins also pholipids PtdSer or PtdIns sedimented the majority of the
showed punctate or plasma membrane fluorescence when Kcc4p901–1037 when anionic lipid was present at R50 mM. Diva-
expressed as GFP fusion proteins in yeast or HeLa cells lent cations did not significantly alter the affinity or specificity of
(Table S1A). For five of the candidate proteins (Cam1p, Dps1p, phospholipid binding by Kcc4p901–1037. Neither elevating diva-
Kcc4p, Rgd1p, and Stp22p), Ras rescue analysis of deletion lent cation levels (by adding 10 mM CaCl2 and 1 mM MgCl2)
mutants identified regions or domains responsible for membrane nor depleting them (by adding 1 mM EDTA) changed apparent
targeting (Table S1B). We focus here on Kcc4p. KD values by more than 2-fold (Table S2).

A Membrane-Targeting Domain at the C Terminus Related C-Terminal Domains in Gin4p and Hsl1p Septin-
of the Septin-Associated Kinase Kcc4p Associated Kinases Also Bind Anionic Phospholipids
In studies of the septin-associated kinase Kcc4p, Ras rescue The only clearly recognizable protein module in Kcc4p according
analysis identified a C-terminal 160 aa fragment (aa 877–1037) to the SMART, Pfam, and UniProt databases is the N-terminal
that is sufficient to drive Ha-Ras membrane recruitment in yeast kinase domain (Figure 1A). However, BLAST searches (Altschul
cells (Figure 1A). This fragment also displays strong plasma et al., 1990) identify an 130 amino acid region related to

Cell 143, 966–977, December 10, 2010 ª2010 Elsevier Inc. 967
877 1037 Figure 1. A C-Terminal Domain in Kcc4p
A Kcc4p Ras B GFP Kcc4p
Rescue Binds Phospholipids and Associates with
21 285 1037 25˚C 37˚C
Cell Membranes
kinase +

S. cerevisiae
1
(A) A C-terminal 160 aa Kcc4p fragment (residues
21 285
1 kinase 302 - 877–1037) is necessary and sufficient for mem-
brane recruitment of Ha-RasQ61L fusions,
21 285
1 kinase 700 - rescuing 37 C growth of cdc25ts yeast cells. Serial
1037
dilutions of yeast cultures expressing each Kcc4p
+ fragment were spotted in duplicate onto selection

HeLa
702

plates and incubated at 25 C or 37 C.


877 1037

+ (B) The same C-terminal Kcc4p fragment, fused to


GFP, shows plasma membrane localization in
S. cerevisiae and HeLa cells.
C D E (C) SPR studies of Kcc4p901–1037 binding to DOPC
3000 3000 70 membranes containing 10% (mole/mole) PtdIns
60 (4,5)P2 (KD = 10.6 ± 1.1 mM), 20% (mole/mole)
Max Binding (RUs)

% Sedimentation
Binding (RUs)

2000 2000
50 phosphatidic acid (KD = 10.2 ± 0.3 mM), or 20%
40 (mole/mole) PtdSer (KD = 7.8 ± 3.4 mM). Binding
30 curves are representative of at least three indepen-
1000 1000
20 dent experiments, and mean KD values ± standard
10
deviation are quoted (Table S2).
0 0 0
0 10 20 30 40 50 60 10 20 3 10 0 125 250 500 1000 (D) SPR signals at saturation show that maximal
[His6-Kcc4p901-1037] (μM) %PtdSer %PtdIns(4,5)P2 Total Available Lipid (μM)
Kcc4p901–1037 binding scales with the negative
100% PC
Phosphatidic acid (20%) charge density in immobilized membranes. Mean
20% PE
PtdIns(4,5)P2 (10%) Bmax values ± standard deviations (for >3 experi-
20% PtdSer
PtdSer (20%)
20% PtdIns ments) are plotted for membranes containing the
noted percentages (mole/mole) of PtdIns(4,5)P2
(valence 4 at pH 7.4) and PtdSer (valence 1 at
pH 7.4).
(E) In vesicle sedimentation studies, His6-Kcc4p901–1037 (at 50 mM) binds small unilamellar vesicles containing 20% (mole/mole) phosphatidylinositol (PtdIns) or
20% (mole/mole) PtdSer in a brominated PC background, but not to phosphatidylethanolamine (PE). At 500 mM ‘‘total available lipid,’’ 100 mM of PtdIns, PE, or
PtdSer is available for binding on the vesicle outer leaflet. Mean ± standard deviation is plotted for at least three independent experiments.
Figure S1 and Tables S1A and S1B summarize results for other potential phosphoinositide-binding proteins.

Kcc4p901–1037 at the C termini of the functionally related S. cere- 1379–1518) is more distantly related (sharing just 16% identity
visiae kinases Gin4p and Hsl1p (Figure 2A and Figure S2). The with Kcc4p901–1037). As shown in Figure 2B, fusing these
Gin4p C terminus (residues 1007–1142) shares 41% sequence C-terminal regions from Gin4p or Hsl1p to Q61L Ha-Ras allowed
identity with Kcc4p901–1037, and the Hsl1p C terminus (residues complementation of the cdc25ts allele in Ras rescue assays. The

A Figure 2. The Membrane-Targeting Domain


of Kcc4p Is Conserved in Gin4p and Hsl1p
(A) Alignment of C-terminal fragments from the
three S. cerevisiae septin-associated kinases
Kcc4p, Gin4p, and Hsl1p. Acidic residues are
red, basic blue, hydrophobic green, and hydro-
philic plum. Colored blocks or text denote posi-
tions at which two or more residues are identical
or similar, respectively. See also Figure S2.
(B) Ras Rescue studies of Gin4p943–1142 and
B D Hsl1p1358–1518.
(C) GFP/Gin4p1003–1142 and GFP/Hsl1p1358–1518
localize to the plasma membrane in S. cerevisiae
cells.
(D) SPR studies show that GST/Gin4p943–1142
binds DOPC membranes containing 20% (mole/
mole) phosphatidic acid (KD = 5.7 ± 0.5 mM),
20% PtdSer (KD = 8.6 ± 2.6 mM), or 10% PtdIns
C (4,5)P2 (KD = 4.7 ± 0.3 mM). Binding curves are
representative of at least three independent exper-
iments. Note that GST dimerization causes over-
estimation of apparent binding affinity in this assay
(Yu et al., 2004).

968 Cell 143, 966–977, December 10, 2010 ª2010 Elsevier Inc.
WT cho1Δ Figure 3. Phosphatidylserine Depletion Reduces
Membrane Association of Kcc4p877–1037,
Gin4p1003–1142, and Hsl1p1358–1518
Localization of GFP-fused Kcc4p877–1037, Gin4p1003–1142,
Kcc4p
and Hsl1p1358–1518 in wild-type yeast cells (left) and in
cho1D cells, which lack PtdSer. The lactadherin C2
domain was used as a control probe for PtdSer (Yeung
et al., 2008). The five panels shown for each GFP fusion
Gin4p in cho1D cells reflect the range of localization phenotypes
observed, illustrating reduced plasma membrane associa-
tion. Figure S3 shows that reducing phosphoinositide
levels has no such effect.
Hsl1p

Lactadherin 0.95 ± 0.13 (Hsl1p1358–1518)—mirroring the


C2 effect on the PtdSer-specific lactadherin C2
domain (FPM/FCyt = 0.61 ± 0.20).
Previous studies employing fluorescent
surface-potential probes and the lactadherin
C2 domain have shown that the plasma mem-
Gin4p and Hsl1p C termini also showed robust plasma mem- brane inner leaflet is the most negatively charged of cyto-
brane localization when expressed in yeast cells as GFP fusion plasmic-facing membranes, and that PtdSer is the primary
proteins (Figure 2C). Moreover, the Gin4p C-terminal domain determinant of this surface charge (Yeung et al., 2006, 2008).
(expressed in E. coli as a GST fusion protein) bound PA, PtdIns C-terminal domains from the septin-associated kinases appear
(4,5)P2, and PtdSer in SPR studies (Figure 2D), resembling to resemble these nonspecific surface-potential probes. They
the in vitro interactions seen for Kcc4p901–1037 (although with show preferential targeting to the plasma membrane that is
different charge dependence, interpretation of which is compli- dependent on PtdSer, although they do not specifically recog-
cated by dimerization of the fused GST). The Gin4p and Hsl1p nize this lipid. The residual plasma membrane association seen
C termini therefore have broadly similar membrane-binding in cho1D cells for these domains (Figure 3) may reflect their
properties to those seen for Kcc4p901–1037. It is important to point ability to bind either PtdIns (see Figure 1E), levels of which are
out that Gin4p and Hsl1p were not found in the proteome-wide known to be elevated in cho1D cells (Hikiji et al., 1988), or other
screen of yeast phospholipid-binding proteins described by less abundant anionic plasma membrane phospholipids.
Zhu et al. (2001), arguing that additional, as-yet-unidentified,
S. cerevisiae phospholipid-binding proteins may exist. Structure of the Kcc4p C-Terminal Domain Reveals
a KA1 Domain Fold
Loss of Phosphatidylserine Impairs Membrane In an effort to understand anionic phospholipid binding by
Targeting of Kcc4p, Gin4p, and Hsl1p C-Terminal C-terminal domains from the septin-associated kinases, we
Domains determined the X-ray crystal structure of Kcc4p917–1037 to 1.7 Å
To determine which cellular phospholipids are important for resolution (see Table S3). The domain contains two interacting
in vivo membrane association of the C termini from Kcc4p, a helices (a1 and a2) that lie on the concave surface of a five-
Gin4p, and Hsl1p, we assessed their localization (as GFP fusion stranded antiparallel b sheet (Figure 4A and Figure S4). A short
proteins) in S. cerevisiae mutants harboring specific phospho- b strand (b1) precedes helix a1, which is then followed by
lipid synthesis defects. Plasma membrane localization was not a four-stranded b-meander (b2–b5) and a C-terminal a helix
detectably altered when levels of PtdIns(4,5)P2 or PtdIns4P (a2). Remarkably, the structure of Kcc4p917–1037 is very similar
were reduced by manipulation of temperature-sensitive yeast to that of the extended KA1 domain from the MARK3 human
strains (Stefan et al., 2002), arguing that neither of these phos- MAP/microtubule affinity-regulating kinase (Tochio et al.,
phoinositides plays a dominant role (Figure S3). By contrast, in 2006), depicted in Figure 4B (Protein Data Bank [PDB] ID
cho1D cells that lack PtdSer (Hikiji et al., 1988), the degree 1UL7). KA1 domains were initially defined as a Pfam domain
of plasma membrane association of each domain was reduced family of 50 amino acids (PF02149) at the C termini of kinases
significantly (Figure 3). Ratios of plasma membrane to cyto- from the MARK/PAR1/Kin family (Matenia and Mandelkow,
solic fluorescence (FPM/FCyt: see Experimental Procedures) 2009; Tassan and Le Goff, 2004; Timm et al., 2008). NMR
in wild-type cells were 1.4 ± 0.35, 1.5 ± 0.08, and 2.9 ± 1.0, structural studies (Tochio et al., 2006) showed that the stable
respectively, for GFP/Kcc4p877–1037, GFP/Gin4p1003–1142, and KA1 domain in MARK3 actually contains 100 amino acids.
GFP/Hsl1p1358–1518,similar to the FPM/FCyt ratio of 1.5 ± 0.16 The 118 residue phospholipid-binding domain at the Kcc4p
measured for the lactadherin discoidin-type C2 domain previ- C terminus that we have identified here also appears to be
ously characterized as a specific PtdSer probe (Yeung et al., a KA1 domain. It contains all secondary structure elements
2008). Loss of PtdSer in cho1D cells reduced FPM/FCyt ratios to seen in MARK3-KA1, plus a short additional a helix at its amino
0.53 ± 0.15 (Kcc4p877–1037), 0.93 ± 0.20 (Gin4p1003–1142), and terminus (aN). As shown in Figure 4C, the core (100 amino acid)

Cell 143, 966–977, December 10, 2010 ª2010 Elsevier Inc. 969
A Kcc4p917-1037

α2 C
β3
α1 α2
β5 β4 β3 β2

α1

αN αN
β1
90˚ β1
C
C
β5 β4

N
N
N

B MARK3 KA1 C
C

α2
β3
β2 α2
N β3 N
β4
β5
α1

α1 90˚
C C β1
β1
β5 β4

Figure 4. The Kcc4p C Terminus Adopts a KA1 Domain Fold


(A) Cartoon representation of Kcc4p917–1037 structure. Helices aN, a1, and a2 are marked, as are strands b1–b5. Two orthogonal views are shown. See also Fig-
ure S4.
(B) NMR structure (Tochio et al., 2006) of the KA1 domain from mouse MARK3 (PDB ID 1UL7), in the same orientations used in (A) for Kcc4p917–1037.
(C) Ca overlay of MARK3-KA1 (cyan) with Kcc4p917–1037 (magenta). The N-terminal part of Kcc4p917–1037, including helix aN, was removed for clarity.

Kcc4p and MARK3 KA1 domains overlay very well (with Ca posi- and MELK KA1 domains (as GFP fusions) showed robust
tion root-mean-square [rms] deviation of just 2.4 Å), despite plasma membrane localization in S. cerevisiae, with FPM/FCyt
sharing only 10% sequence identity—explaining the failure to ratios ranging from 1.8 to 3.1 (Figure 5C). Again, these values
identify this domain through sequence analysis. A structure- were reduced by 50% in PtdSer-deficient cho1D cells
based sequence alignment of KA1 domains from the MARK/ (Figure 5C) but were not significantly altered in mutant yeast
PAR1/Kin family and the Kcc4p/Gin4p/Hsl1p kinases is shown strains with reduced phosphoinositide levels (Figure S5). The
in Figure S2. subcellular localization properties of KA1 domains from human
MARK1, MARK3, and MELK therefore appear similar to those
KA1 Domains from Human MARK/PAR1 Kinases Bind seen for the Kcc4p, Gin4p, and Hsl1p KA1 domains identified
Acidic Phospholipids here. In addition, purified monomeric MARK1-KA1 showed
Although speculated to participate in autoregulatory intramo- essentially the same in vitro phospholipid-binding characteris-
lecular interactions in MARK/PAR1 kinases (Marx et al., tics as Kcc4p-KA1, binding to vesicles that contain PtdSer,
2010), no clear function has been ascribed to KA1 domains. PA, or PtdIns(4,5)P2 (Figure 5D) with KD values in the 2.3 mM–
Having identified the Kcc4p KA1 domain as a phospholipid- 8.9 mM range (Table S2), and with Bmax values that scale with
binding domain, we next asked whether previously recognized membrane charge density. The KA1 domains from MARK/
KA1 domains from human MARK1, MARK3, and MELK PAR1 family kinases thus appear to be phospholipid-binding
(maternal embryonic leucine zipper kinase) also associate domains that are likely to promote membrane association of
with cell membranes and bind phospholipids. As shown in Fig- their host proteins in cells. Indeed, Alessi and colleagues (Gör-
ure 5A, all of these KA1 domains recruit Q61L Ha-Ras fusions ansson et al., 2006) previously implicated the KA1 domain as
to yeast cell membranes, complementing the cdc25ts mutation an important membrane localization determinant in MARK3
in Ras rescue assays. GFP fusions of the MARK1 and MARK3 mutants that fail to bind 14-3-3 proteins. Our findings suggest
KA1 domains showed substantial plasma membrane localiza- that this observation reflects MARK3-KA1 binding to acidic
tion in HeLa cells (Figure 5B). Moreover, the MARK1, MARK3, phospholipids and argue that the KA1 domain should be

970 Cell 143, 966–977, December 10, 2010 ª2010 Elsevier Inc.
A B
25˚C 37˚C
MARK1 KA1
MARK1 KA1
MARK3 KA1

MELK KA1

MARK3 KA1

C wild-type cho1Δ D HeLa Cells

MARK1 KA1 2000

Binding (RUs)
1.8±0.5 1.0±0.2

1000

MARK3 KA1

3.1±0.3 1.0±0.2
0
0 10 20 30 40 50
[His6-MARK1683-795] (μM)
MELK KA1 Phosphatidic acid (20%)
PtdIns(4,5)P2 (10%)
2.3±0.3 1.2±0.2 PtdSer (20%)
S. cerevisiae

Figure 5. KA1 Domains from Human MARK/PAR1 Kinases Bind Phospholipids


(A) KA1 domains from human MARK1 (aa 648–795), human MARK3 (aa 589–729), and human MELK (aa 500–651) all drive membrane recruitment of Ha-RasQ61L
fusions in Ras rescue studies.
(B) GFP-fused human MARK1 and MARK3 KA1 domains show plasma membrane localization in HeLa cells. Unexplained nuclear localization of the MELK-KA1
fusion made interpretation of its behavior difficult (not shown).
(C) GFP-fused KA1 domains from human MARK1, MARK3, and MELK show robust plasma membrane localization in S. cerevisiae cells, which is diminished in
cho1D cells that lack PtdSer. Mean FPM/FCyt ratios for each experiment (±standard deviation) are quoted in individual panels. Figure S5 shows that manipulating
phosphoinositide levels in yeast cells does not affect membrane targeting of MARK family KA1 domains.
(D) Purified MARK1-KA1 binds membranes containing phosphatidic acid (20%), PtdSer (20%), or PtdIns(4,5)P2 (10%) in SPR studies. Binding curves are repre-
sentative of at least three independent experiments. Mean apparent KD values (±standard deviation) are listed in Table S2.

considered as a bona fide membrane-targeting/anionic phos- K1016 in the amino-terminal part of helix a2 (Figure 6A and
pholipid-binding module. Figure S4A). Adjacent electron density (3 Å away) is best fit
with a glycerol molecule that contacts K1010 in strand b5 plus
serine and threonine side chains (S1014 and T1015) at the
Basic Regions on the KA1 Domain Surface Drive beginning of helix a2 (Figure S4A). Intriguingly, in a second
Membrane Association crystal form (Table S3) density for a tartrate ion replaces both
To understand how KA1 domains interact with negatively SO4#1 and the bound glycerol (Figure S4B), implicating this
charged membranes, we analyzed features common to the region as an important anion binding site in Kcc4p-KA1. The
structure of the yeast Kcc4p KA1 domain and a crystal structure second sulfate in Figure 6A (SO4#2) lies in a basic pocket on
of the human MARK1 KA1 domain that we determined to 1.7 Å the Kcc4p-KA1 surface formed largely by side chains from the
resolution (see Table S3). Both have notable positively charged helix a1 C terminus (K959) and the a1/b2 loop (K964).
patches and/or crevices on their surfaces (Figure 6) that result The locations of bound anions in crystal structures of
from basic side-chain arrangements reminiscent of headgroup- membrane-targeting domains frequently reveal the binding sites
binding sites in other phospholipid-interaction domains (Hurley, for phospholipid headgroups (Hurley, 2006; Lemmon, 2008;
2006; Lemmon, 2008). Wood et al., 2009). We therefore used mutagenesis to investi-
For Kcc4p-KA1, clear electron density could be seen for two gate the importance of the SO4#1 and SO4#2 binding sites for
bound sulfate ions, 27 Å apart, which lie on either side of a posi- in vivo membrane association of Kcc4p-KA1. When pairs of
tively charged region that stretches across the width of the basic residues were mutated (Figure 6A), plasma membrane
domain in the orientation shown in Figure 6A and encircles the localization of GFP/Kcc4p-KA1 was only impaired when one or
b3/b4 loop that projects prominently from its surface. One of both mutated residues contributed to binding of one of these
these sulfates (SO4#1) interacts primarily with lysine side chains sulfates (K932, K1007, K1010, K1016, K1020, K964, and K978
in the aN/b1 loop (K932) and b5 (K1010), and it lies close to were implicated). Importantly, mutations at both sulfate-binding

Cell 143, 966–977, December 10, 2010 ª2010 Elsevier Inc. 971
K1016S/K1020S* K964S/K978S* Figure 6. Potential Phospholipid-Binding
Sites on Kcc4p and MARK1 KA1 Domains
A 3/ 4 loop (A) Kcc4p-KA1 is shown in surface representation
(left: with electrostatic surface potential—blue,
K1007S/K1010S* K959S/K964S* positive; red, negative) and in cartoon form (right:
Glycerol Glycerol same orientation). The two ordered sulfate ions
K964
K1020 (SO4#1 and SO4#2) and the glycerol molecule
SO4#2 K1016 SO4#2
K978
close to SO4#1 are marked, as is the b3/b4 loop.
SO4#1
SO4#1 K959
Figure S4 shows the tartrate ion that replaces
K1010 SO4#1 and the glycerol in another crystal form.
K932
Noted residues were mutated in pairs to serine,
K930
K953 expression confirmed by western blotting (not
K1007 shown), and effects on plasma membrane locali-
K930S/K932S* K953S/K959S
R946
zation of GFP fusions assessed in wild-type yeast
K926 K927 cells (right). Double mutations marked with red
K945
asterisks showed significantly reduced FPM/FCyt
ratios compared with wild-type Kcc4p-KA1
Kcc4p-KA1 (mean FPM/FCyt = 1.7 ± 0.3). FPM/FCyt values for
K926S/K930S K945S/R946S
mutated variants were 0.81 ± 0.09 (K930S/
K932S), 0.74 ± 0.03 (K959S/K964S), 0.80 ± 0.15
K927S/K930S (K964S/K978S), 0.92 ± 0.14 (K1007S/K1010S),
0.96 ± 0.06 (K1016S/K1020S). Residues impli-
cated in membrane binding are colored black,
K783S/K788S
whereas those at which mutations did not influ-
B K773S/R774S* R748S ence targeting are gray.
3/ 4 loop (B) Crystal structure of human MARK1-KA1 (Table
S3), shown in the same orientation as in (A).
Compared with an FPM/FCyt ratio of 2.0 ± 0.4 for
R748
R771A/K773A* K735S/R737S wild-type MARK1-KA1, mutated variants denoted
K783 K788 by red asterisks gave FPM/FCyt values of 0.90 ±
0.20 (R698S/R701S), 0.93 ± 0.19 (R771A/
R774 K773A), and 0.99 ± 0.04 (K773S/R774S). Figure S6
K773 K735 describes effects of these mutations on in vitro
K707 binding.
R722
R771 R719
R701 R698
R698S/R701S* R719S/R722S
R737
K761
R764 K696 connects them. In addition to conserved
positive charge in this region (in b5),
MARK1-KA1 K707S K693S/K696S all KA1 domains have serine and/or
threonine residues at the beginning of
K761S/R764S
helix a2 that contact bound glycerol
in Kcc4p-KA1 (Figure S4A) and may
interact similarly with the glycerol back-
sites diminished membrane recruitment, suggesting that the bone of bound phospholipids. As anticipated from these obser-
KA1 domain makes multiple contacts with the bilayer surface. vations, hMARK1-KA1 mutations in the basic patch correspond-
Engaging both the SO4#1 and SO4#2 sites in binding to a ing to the Kcc4p SO4#1 binding site impaired both plasma
membrane surface is difficult to envision without the b3/b4 membrane association (Figure 6B) and in vitro binding to anionic
loop penetrating the bilayer. This loop contains several hydro- phospholipids (Figure S6B). K773 and R774 in strand b5 of
phobic side chains (with sequence VNDSILFL) and resembles hMARK1-KA1 appear to be important for membrane associa-
‘‘membrane insertion loops’’ reported in C2, PX, and FYVE tion. Moreover, an R698S/R701S double mutation close to the
domains (Cho and Stahelin, 2005; Lemmon, 2008). As shown hMARK1-KA1 N terminus prevented plasma membrane associ-
in Figure S6A, Kcc4p-KA1 can indeed penetrate acidic phospho- ation and vesicle binding, suggesting that the basic patch
lipid-containing monolayers that have packing densities similar extending to the bottom left of hMARK1-KA1 in Figure 6B makes
to those estimated for cell membranes (Demel, 1994; Marsh, additional contributions—perhaps functionally replacing the
1996)—resembling C2, PX, FYVE, and some PH domains in SO4#2 binding site of Kcc4p-KA1. Thus, membrane association
this respect (Stahelin et al., 2007). of both the Kcc4p and the MARK1 KA1 domains appears to
Only the SO4#1/glycerol-binding site of Kcc4p-KA1 is involve cooperation of more than one positively charged binding
conserved in the hMARK1 KA1 domain—in location, charge region—centered on the conserved SO4#1 binding site seen in
characteristics (Figure 6B), and sequence (Figure S2). It lies in Kcc4p-KA1. Similar utilization of multiple binding sites has previ-
the most sequence-conserved region of aligned KA1 domains ously been described for annexins, as well as PKC-type C2, PX,
that encompasses strand b5, helix a2, and the loop that and PH domains (Lemmon, 2008).

972 Cell 143, 966–977, December 10, 2010 ª2010 Elsevier Inc.
A GFP-Kcc4p
KA1 K1007S/K1010S
domain
wild-type K1007S/K1010S K1016S/K1020S
K1016S/K1020S
PtdSer
level
normal normal normal normal
wt cho1Δ wt cho1Δ wt cho1Δ wt cho1Δ

Epi

DIC

gin4Δ gin4Δ gin4Δ


B GFP GFP-Gin4p GFP-Gin4pΔKA1

gin4Δ
DIC

Epi

Figure 7. Role of the KA1 Domain in Kcc4p and Gin4p


(A) Localization of wild-type and KA1 domain-mutated intact GFP/Kcc4p in wild-type yeast cells (normal) and PtdSer-deficient cho1D cells. Additional images
and western blot confirmation of intact protein expression are shown in Figure S7.
(B) Yeast cells lacking Gin4p (gin4D) show an elongated bud phenotype (left). Overexpressed GFP-fused full-length Gin4p in gin4D cells rescues this aberrant
elongated-bud morphology and is found at the bud neck in all cells. By contrast, GFP/Gin4pDKA1 fails to rescue the gin4D phenotype and remains diffuse in the
cytoplasm. Examining at least 200 cells in several experiments, the elongated phenotype was seen in 69% of gin4D cells expressing GFP alone, 78% expressing
GFP/Gin4pDKA1, and just 39% of those expressing GFP/Gin4p.

PtdSer-Dependent Bud Neck Localization of KA1 cytosolic fluorescence was increased)—suggesting that the
Domain-Mutated Kcc4p elevated PtdIns levels found in these cells (Hikiji et al., 1988)
Double mutations (K1007S/K1010S or K1016S/K1020S) that may be sufficient. Western blotting confirmed that all GFP/
abolish membrane localization of isolated Kcc4p-KA1 (shown Kcc4p variants were expressed at or above wild-type levels
in Figure 6A) did not prevent intact Kcc4p from being targeted (Figure S7B). Taken together, these data show that bud neck
to the bud neck when overexpressed in wild-type yeast cells targeting of intact GFP/Kcc4p can be abolished either by
(Figure 7A). However, background cytoplasmic fluorescence mutating basic residues in the KA1 domain’s anionic phospho-
was increased to some extent, and simultaneous introduction lipid-binding site or—importantly—by simultaneously reducing
of all four KA1 domain mutations into intact Kcc4p abolished anionic phospholipid levels in the plasma membrane inner
its targeting to bud necks. leaflet and mutating the KA1 domain.
Hypothesizing that the KA1 domain must cooperate with The lack of a clear phenotype for KCC4 mutations (Longtine
other domains in targeting intact Kcc4p specifically to bud et al., 2000) prevented us from being able to assess functional
necks, we surmised that residual low-affinity PtdSer binding consequences of the KA1 domain mutations described above.
by K1007S/K1010S- or K1016S/K1020S-mutated KA1 domains However, studies of Gin4p demonstrated a functional require-
might be sufficient to drive normal Kcc4p targeting in this ment for the KA1 domain (Figure 7B). Deleting the GIN4 (or
overexpression study. We therefore re-examined localization HSL1, but not KCC4) gene in S. cerevisiae leads to an elongated
of the intact GFP/Kcc4p variants in cells lacking PtdSer. As bud phenotype characteristic of a G2/M delay due to morpho-
suspected, PtdSer loss (in cho1D cells) completely abrogated genesis checkpoint failure (Longtine et al., 1998). In gin4D cells,
bud neck localization of K1007S/K1010S-mutated GFP/Kcc4p this elongated bud phenotype can be rescued by overexpress-
(Figure 7A: see also Figure S7A). In other words, K1007S/ ing a wild-type Gin4p GFP fusion (Figure 7B), and the protein is
K1010S-mutated Kcc4p is dependent on normal plasma found at bud necks. However, when just the KA1 domain (but
membrane PtdSer levels for its targeting to the bud neck, impli- not septin-binding region) is deleted, the GFP/Gin4pDKA1 fusion
cating PtdSer as an important determinant of Kcc4p localiza- fails to rescue gin4D cells and is diffusely localized (Figure 7B) in
tion. Bud neck localization was still seen for wild-type and much the same way as GFP/Kcc4p harboring multiple KA1
K1016S/K1020S-mutated GFP/Kcc4p in cho1D cells (although domain mutations.

Cell 143, 966–977, December 10, 2010 ª2010 Elsevier Inc. 973
DISCUSSION by binding close to the C-terminal region and disrupting autoin-
hibitory intramolecular interactions. One concern raised about
Our search for previously undescribed phosphoinositide/phos- this model (Crutchley et al., 2009; Szkotnicki et al., 2008) is
pholipid-binding domains identified a small C-terminal domain that it cannot explain why Hsl1p is activated only by assembled
in S. cerevisiae septin-associated kinases that binds acidic septins at the bud neck, and not by free septin complexes.
phospholipids. Crystallographic studies revealed that this is Our findings provide an explanation: that the C-terminal region
a KA1 domain, a module previously identified at the C termini of Hsl1p (and other septin-associated kinases) must bind to
of kinases from the mammalian MARK/PAR1 family. We show both septins and anionic membrane phospholipids (via its KA1
that KA1 domains from both yeast and human kinases bind domain) to drive the protein to the bud neck and relieve the
acidic phospholipids including PtdSer. For yeast Kcc4p, we proposed intramolecular autoinhibition.
also present data using KA1 domain mutations that implicate Reversing intramolecular autoinhibitory interactions by engag-
PtdSer as an important determinant for targeting this kinase to ing one or more phospholipid-binding domains is a recurring
its site of action at the bud neck. theme in kinase regulation, with protein kinase C (PKC) and other
Our findings with Kcc4p and Gin4p argue that—in addition to AGC kinases providing well-characterized examples (Newton,
its documented dependence on septin binding (Barral et al., 2009). Our studies suggest that the mechanistic role of the
1999; Longtine et al., 1998)—bud neck localization of septin- KA1 domain in septin-associated kinases may be broadly anal-
associated kinases requires KA1 domain,phospholipid interac- ogous to that of C1 and C2 domains in PKC or the PH domain
tions. On their own, neither the KA1 domain nor the septin- in Akt (Newton, 2009). The KA1 domain lacks the lipid selectivity
binding region of Kcc4p/Gin4p/Hsl1p is sufficient for specific of these other modules but appears to restrict specific recogni-
bud neck targeting—but C-terminal fragments encompassing tion of other targets (such as septins) to a membrane context.
both are efficiently localized to bud necks (Crutchley et al., Extending our observations to the MARK/PAR1 family kinases,
2009; Longtine et al., 1998; Okuzaki and Nojima, 2001). Thus, the KA1 domain was previously implicated as a determinant of
simultaneous engagement of the septin- and phospholipid- membrane localization for MARK3 (Göransson et al., 2006),
binding domains appears to be required for Kcc4p, Gin4p, and and dissociation of hMARK2 from the plasma membrane coin-
Hsl1p recruitment to septin assemblies at the bud neck for cides with reduced activity (Hurov et al., 2004). Thus, phospho-
kinase activation. This combination of septin-binding and phos- lipid engagement of the KA1 domain may also play a role in the
pholipid-binding domains may function as an effective ‘‘coinci- activation of these kinases at particular membrane locations.
dence detector,’’ allowing the kinases to bind septins only at Intriguingly, the KA1 domain fold has recently been seen in addi-
membrane locations. The septins themselves also bind weakly tional kinase contexts that warrant further investigation. A
to anionic phospholipids (Casamayor and Snyder, 2003; Zhang C-terminal domain in the Arabidopsis AtSOS2 kinase has a KA1
et al., 1999), suggesting further that kinase,phospholipid, kina- domain fold (Sánchez-Barrena et al., 2007) and includes a pro-
se,septin, and septin,phospholipid interactions all cooperate tein phosphatase-interacting (PPI) motif (in strand b1 and helix
to organize a well-defined assembly at the bud neck. Coinci- a1). It is not known whether this domain binds phospholipids.
dence detection of this sort, in which multivalent interactions A C-terminal domain in the a subunit of heterotrimeric AMPK or-
involving both protein-binding and lipid-binding domains drive thologs also has a KA1 domain fold and is intimately associated
complex formation, has been suggested for several systems with the C-terminal region of the b subunit (Townley and Shapiro,
(Carlton and Cullen, 2005; Lemmon, 2008). It is particularly inter- 2007). Given that KA1 domain-containing proteins are implicated
esting for Kcc4p that the KA1 domain can promote kinase target- in a wide range of diseases, from Alzheimer’s disease to cancer
ing to a specific location (the bud neck) despite binding nonspe- to diabetes, understanding the regulatory role of this domain is
cifically to anionic phospholipids: it appears to restrict the ability an important goal. Our studies show that at least a subgroup
of Kcc4p to bind septins only in the context of a negatively of KA1 domains bind nonspecifically to acidic phospholipids
charged membrane surface, as a logical ‘‘AND’’ gate. Similar and allow kinase activation to be coordinated with membrane
coincidence detection mechanisms may also be relevant for association, in an unexpected variation of a theme used by other
specific membrane targeting of human MARK/PAR1 family kinases that employ C1, C2, PH, and other domains.
proteins. Indeed, we show here that—like their structural coun-
terparts in the yeast septin-associated kinases—KA1 domains EXPERIMENTAL PROCEDURES
of human MARK/PAR1 family proteins bind acidic phospholipids
Ras Rescue Assay
in cells and in vitro.
Ras rescue assays were performed exactly as described (Yu et al., 2004).
Several reports have suggested that the C-terminal tail of Briefly, DNA-encoding candidate proteins or fragments were PCR amplified
MARK/PAR1 kinases (which includes the KA1 domain) plays from S. cerevisiae (BY4741) genomic DNA or a HeLa cell cDNA library and
a role in reversible autoinhibition of kinase activity (Elbert et al., subcloned into modified p3S0BL2 (Isakoff et al., 1998) to generate plasmids
2005; Marx et al., 2010; Timm et al., 2008). For example, the encoding Ha-Ras Q61L fusions. Plasmids were transformed into cdc25ts yeast
C-terminal KA1 domain-containing region of the S. cerevisiae cells, and rescue of the growth defect at 37 C assessed as described (Yu et al.,
2004).
Kin1 and Kin2 kinases was reported to interact with the
N-terminal catalytic domain (Elbert et al., 2005)—suggesting
Microscopy
direct intramolecular autoinhibitory interactions. A similar model For yeast studies, DNA fragments encoding candidate proteins or domains
was also proposed for S. cerevisiae Hsl1p (Hanrahan and were subcloned into modified pGO-GFP (Cowles et al., 1997) and transformed
Snyder, 2003), and septins were suggested to activate Hsl1p into wild-type (BY4741) or cho1D BY4743 cells as described (Audhya and Emr,

974 Cell 143, 966–977, December 10, 2010 ª2010 Elsevier Inc.
2002). Images were collected at 1003 magnification using a Leica-DMIRBE 1UL7) (Tochio et al., 2006), using Phaser (CCP4, 1994). Model building em-
microscope and processed using Volocity deconvolution software (Improvi- ployed Coot (Emsley and Cowtan, 2004), following each round of refinement
sion). All images of yeast cells are representative of >90% of cells expressing using Refmac (CCP4, 1994) and PHENIX (Adams et al., 2010). Data collection
the relevant GFP fusion protein (from over 100 cells in at least three experi- and refinement statistics are presented in Table S3. Structure figures were
ments). Analysis of full-length (or DKA1) Gin4p was performed in YEF1238 generated using PyMol (DeLano, 2002).
gin4D::TRP1 (YEF473A) cells (Longtine et al., 1998). To quantify plasma
membrane localization, lines were drawn across individual cells using ImageJ
ACCESSION NUMBERS
and mean values for fluorescence in the plasma membrane (FPM) and cytosolic
(FCyt) regions were determined along the length of these lines as described
Coordinates and structure factors have been deposited in the Protein Data
(Szentpetery et al., 2009). The ratio of these means (FPM/FCyt) was used as
Bank (http://www.rcsb.org/pdb) with identification numbers 3OSE (MARK1-
a measure of plasma membrane localization.
KA1), 3OSM (Kcc4p-KA1 with bound tartrate), and 3OST (Kcc4p-KA1 with
For analysis of subcellular localization in mammalian cells, domains of
bound sulfates).
interest were subcloned into pEGFP-C1 (Clontech) and transiently transfected
into HeLa cells using Lipofectamine 2000 (Invitrogen). Cells were imaged at
403, and images processed as above. All microscopy images presented are SUPPLEMENTAL INFORMATION
representative of at least three independent experiments, assessing over
100 cells each. Supplemental Information includes Extended Experimental Procedures, seven
figures, and three tables and can be found with this article online at doi:10.
Surface Plasmon Resonance and Phospholipid Binding 1016/j.cell.2010.11.028.
Phospholipid-binding experiments were performed using surface plasmon
resonance (SPR) exactly as described previously (Yu et al., 2004) or sedimen- ACKNOWLEDGMENTS
tation assays (Kavran et al., 1998). For SPR studies, vesicles contained dio-
leoylphosphatidylcholine (DOPC) alone or the noted percent (mole/mole) of We thank members of the Lemmon, Ferguson, and Bi laboratories and Ben
test lipid in a DOPC background and were immobilized on L1 sensor chip Black, Jim Shorter, and Greg Van Duyne for constructive comments. Erfei
surfaces (BIAcore). Purified test proteins were flowed over these surfaces at Bi, Scott Emr, and Daryll DeWald provided yeast strains used in this study.
a series of concentrations, determined by absorbance at 280 nm using calcu- Crystallographic data were collected in part at the GM/CA Collaborative
lated extinction coefficients. SPR signals for each experiment were corrected Access Team at the Advanced Photon Source (APS), funded by NCI (Y1-
for background (DOPC) binding and plotted against protein concentration to CO-1020) and NIGMS (Y1-GM-1104). Use of APS was supported by the
yield binding curves that were fit to simple hyperbolae. Experiments were per- U.S. Department of Energy, under contract No. DE-AC02-06CH11357. Addi-
formed in 25 mM HEPES, pH 7.4, containing 150 mM NaCl. For sedimentation tional crystallographic data were collected at beamline F2 at the Cornell
assays, brominated PC was used as the background lipid and experiments High Energy Synchrotron Source (CHESS), supported by NIGMS and the
were performed exactly as described (Kavran et al., 1998). NSF (under award DMR-0936384), using the Macromolecular Diffraction at
CHESS (MacCHESS) facility, supported by the NIH (award RR-01646). This
Protein Preparation, Crystallization, and Data Collection work was funded in part by NIH grant R01-GM056846 (to M.A.L.) and a predoc-
DNA encoding the KA1 domains from Kcc4p (residues 917–1037) and MARK1 toral fellowship from the American Heart Association Great Rivers Affiliate
(residues 683–795), plus an N-terminal hexahistidine tag, were subcloned into (K.M.).
pET21a (Novagen) for expression in E. coli BL21 (DE3) cells. For generating
selenomethionine (SeMet)-containing Kcc4p-KA1 protein, a third methionine Received: March 19, 2010
was introduced by substitution at L936, and protein was produced from Revised: August 3, 2010
B834(DE3) methionine auxotrophs in MOPS-based minimal medium supple- Accepted: November 1, 2010
mented with SeMet. Proteins were purified from cell lysates in three steps, Published: December 9, 2010
using Ni-NTA resin (QIAGEN), cation exchange chromatography, and a Super-
dex 75 size exclusion column (GE Healthcare). Crystals were grown at 21 C
REFERENCES
using the hanging drop vapor diffusion method by mixing equal parts of protein
(at 300–400 mM) and reservoir solutions. MARK1-KA1 crystals were obtained
Adams, P.D., Afonine, P.V., Bunkóczi, G., Chen, V.B., Davis, I.W., Echols, N.,
from 0.1 M Na acetate, pH 4.6, containing 0.04 M CaCl2, and 15%–25%
Headd, J.J., Hung, L.W., Kapral, G.J., Grosse-Kunstleve, R.W., et al. (2010).
(w/v) PEG 3350. Kcc4p-KA1 crystals were obtained both from 0.1 M HEPES,
PHENIX: a comprehensive Python-based system for macromolecular struc-
pH 7.4, containing 0.2 M (NH4)2SO4 plus 20% (w/v) PEG3350 (for both native
ture solution. Acta Crystallogr. D Biol. Crystallogr. 66, 213–221.
and SeMet protein) and from 1.0 M K/Na tartrate, 0.1 M Tris, pH 7.0, with 0.2 M
LiSO4. Crystals were cryo-protected by direct transfer into reservoir solution Altschul, S.F., Gish, W., Miller, W., Myers, E.W., and Lipman, D.J. (1990). Basic
containing 15% (w/v) glycerol and were flash frozen in liquid nitrogen. Data local alignment search tool. J. Mol. Biol. 215, 403–410.
were collected at the Advanced Photon Source (Argonne, IL) beamlines Audhya, A., and Emr, S.D. (2002). Stt4 PI 4-kinase localizes to the plasma
23ID-D and 23ID-B or the Cornell High Energy Synchrotron Source (CHESS) membrane and functions in the Pkc1-mediated MAP kinase cascade. Dev.
beamline F2 and were processed using HKL2000 (Otwinowski and Minor, Cell 2, 593–605.
1997). Audhya, A., Loewith, R., Parsons, A.B., Gao, L., Tabuchi, M., Zhou, H., Boone,
C., Hall, M.N., and Emr, S.D. (2004). Genome-wide lethality screen identifies
Structure Determination and Refinement new PI4,5P2 effectors that regulate the actin cytoskeleton. EMBO J. 23,
Experimental phase information was obtained for Kcc4p-KA1 using data 3747–3757.
collected from the SeMet-containing Kcc4p-KA1/L936M crystals, with
Barral, Y., Parra, M., Bidlingmaier, S., and Snyder, M. (1999). Nim1-related
single-wavelength anomalous diffraction (SAD) methods implemented in
kinases coordinate cell cycle progression with the organization of the periph-
SHELX C/D/E (Schneider and Sheldrick, 2002). The resulting experimentally
eral cytoskeleton in yeast. Genes Dev. 13, 176–187.
phased map was excellent and allowed all but the first eight amino acids
(including the His6 tag) to be traced with Coot (Emsley and Cowtan, 2004). Carlton, J.G., and Cullen, P.J. (2005). Coincidence detection in phosphoinosi-
The resulting model was used to identify molecular replacement (MR) solutions tide signaling. Trends Cell Biol. 15, 540–547.
for datasets obtained with native protein using the program Phaser (CCP4, Casamayor, A., and Snyder, M. (2003). Molecular dissection of a yeast septin:
1994). For MARK1-KA1, the structure was solved using MR with a search distinct domains are required for septin interaction, localization, and function.
model based on the mouse MARK3 KA1 domain NMR structure (PDB ID Mol. Cell. Biol. 23, 2762–2777.

Cell 143, 966–977, December 10, 2010 ª2010 Elsevier Inc. 975
Collaborative Computational Project, Number 4 (CCP4). (1994). The CCP4 Longtine, M.S., Fares, H., and Pringle, J.R. (1998). Role of the yeast Gin4p
suite: programs for protein crystallography. Acta Crystallogr. D Biol. Crystal- protein kinase in septin assembly and the relationship between septin
logr. 50, 760–763. assembly and septin function. J. Cell Biol. 143, 719–736.
Cho, W., and Stahelin, R.V. (2005). Membrane-protein interactions in cell Longtine, M.S., Theesfeld, C.L., McMillan, J.N., Weaver, E., Pringle, J.R., and
signaling and membrane trafficking. Annu. Rev. Biophys. Biomol. Struct. 34, Lew, D.J. (2000). Septin-dependent assembly of a cell cycle-regulatory
119–151. module in Saccharomyces cerevisiae. Mol. Cell. Biol. 20, 4049–4061.
Cowles, C.R., Odorizzi, G., Payne, G.S., and Emr, S.D. (1997). The AP-3 Manning, G., Whyte, D.B., Martinez, R., Hunter, T., and Sudarsanam, S. (2002).
adaptor complex is essential for cargo-selective transport to the yeast The protein kinase complement of the human genome. Science 298, 1912–
vacuole. Cell 91, 109–118. 1934.
Crutchley, J., King, K.M., Keaton, M.A., Szkotnicki, L., Orlando, D.A., Zyla, Marsh, D. (1996). Lateral pressure in membranes. Biochim. Biophys. Acta
T.R., Bardes, E.S., and Lew, D.J. (2009). Molecular dissection of the check- 1286, 183–223.
point kinase Hsl1p. Mol. Biol. Cell 20, 1926–1936. Marx, A., Nugoor, C., Panneerselvam, S., and Mandelkow, E. (2010). Structure
DeLano, W.L. (2002). The PyMOL Molecular Graphics System (Palo Alto, CA: and function of polarity-inducing kinase family MARK/Par-1 within the branch
DeLano Scientific). of AMPK/Snf1-related kinases. FASEB J. 24, 1637–1648.
Demel, R.A. (1994). Monomolecular layers in the study of biomembranes. Matenia, D., and Mandelkow, E.M. (2009). The tau of MARK: a polarized view
Subcell. Biochem. 23, 83–120. of the cytoskeleton. Trends Biochem. Sci. 34, 332–342.
Drewes, G., Ebneth, A., Preuss, U., Mandelkow, E.M., and Mandelkow, E. Maussion, G., Carayol, J., Lepagnol-Bestel, A.M., Tores, F., Loe-Mie, Y.,
(1997). MARK, a novel family of protein kinases that phosphorylate microtu- Milbreta, U., Rousseau, F., Fontaine, K., Renaud, J., Moalic, J.M., et al.
bule-associated proteins and trigger microtubule disruption. Cell 89, 297–308. (2008). Convergent evidence identifying MAP/microtubule affinity-regulating
Elbert, M., Rossi, G., and Brennwald, P. (2005). The yeast par-1 homologs kin1 kinase 1 (MARK1) as a susceptibility gene for autism. Hum. Mol. Genet. 17,
and kin2 show genetic and physical interactions with components of the exo- 2541–2551.
cytic machinery. Mol. Biol. Cell 16, 532–549. McLaughlin, S., and Murray, D. (2005). Plasma membrane phosphoinositide
Emsley, P., and Cowtan, K. (2004). Coot: model-building tools for molecular organization by protein electrostatics. Nature 438, 605–611.
graphics. Acta Crystallogr. D Biol. Crystallogr. 60, 2126–2132. Newton, A.C. (2009). Lipid activation of protein kinases. J. Lipid Res. Suppl. 50,
Göransson, O., Deak, M., Wullschleger, S., Morrice, N.A., Prescott, A.R., and S266–S271.
Alessi, D.R. (2006). Regulation of the polarity kinases PAR-1/MARK by 14-3-3 Okuzaki, D., and Nojima, H. (2001). Kcc4 associates with septin proteins of
interaction and phosphorylation. J. Cell Sci. 119, 4059–4070. Saccharomyces cerevisiae. FEBS Lett. 489, 197–201.
Gray, D., Jubb, A.M., Hogue, D., Dowd, P., Kljavin, N., Yi, S., Bai, W., Frantz, Otwinowski, Z., and Minor, W. (1997). Processing of X-ray diffraction data
G., Zhang, Z., Koeppen, H., et al. (2005). Maternal embryonic leucine zipper collected in oscillation mode. Methods Enzymol. 276, 307–326.
kinase/murine protein serine-threonine kinase 38 is a promising therapeutic Sakchaisri, K., Asano, S., Yu, L.R., Shulewitz, M.J., Park, C.J., Park, J.E., Cho,
target for multiple cancers. Cancer Res. 65, 9751–9761. Y.W., Veenstra, T.D., Thorner, J., and Lee, K.S. (2004). Coupling morphogen-
Halstead, J.R., Jalink, K., and Divecha, N. (2005). An emerging role for PtdIns esis to mitotic entry. Proc. Natl. Acad. Sci. USA 101, 4124–4129.
(4,5)P2-mediated signalling in human disease. Trends Pharmacol. Sci. 26, Sánchez-Barrena, M.J., Fujii, H., Angulo, I., Martı́nez-Ripoll, M., Zhu, J.K., and
654–660. Albert, A. (2007). The structure of the C-terminal domain of the protein kinase
Hanrahan, J., and Snyder, M. (2003). Cytoskeletal activation of a checkpoint AtSOS2 bound to the calcium sensor AtSOS3. Mol. Cell 26, 427–435.
kinase. Mol. Cell 12, 663–673. Schneider, T.R., and Sheldrick, G.M. (2002). Substructure solution with
Hikiji, T., Miura, K., Kiyono, K., Shibuya, I., and Ohta, A. (1988). Disruption of SHELXD. Acta Crystallogr. D Biol. Crystallogr. 58, 1772–1779.
the CHO1 gene encoding phosphatidylserine synthase in Saccharomyces cer- Scott, J.D., and Pawson, T. (2009). Cell signaling in space and time: where
evisiae. J. Biochem. 104, 894–900. proteins come together and when they’re apart. Science 326, 1220–1224.
Hurley, J.H. (2006). Membrane binding domains. Biochim. Biophys. Acta 1761, Seet, B.T., Dikic, I., Zhou, M.M., and Pawson, T. (2006). Reading protein modi-
805–811. fications with interaction domains. Nat. Rev. Mol. Cell Biol. 7, 473–483.
Hurov, J.B., Watkins, J.L., and Piwnica-Worms, H. (2004). Atypical PKC phos- Shulewitz, M.J., Inouye, C.J., and Thorner, J. (1999). Hsl7 localizes to a septin
phorylates PAR-1 kinases to regulate localization and activity. Curr. Biol. 14, ring and serves as an adapter in a regulatory pathway that relieves tyrosine
736–741. phosphorylation of Cdc28 protein kinase in Saccharomyces cerevisiae. Mol.
Hurov, J.B., Huang, M., White, L.S., Lennerz, J., Choi, C.S., Cho, Y.R., Kim, Cell. Biol. 19, 7123–7137.
H.J., Prior, J.L., Piwnica-Worms, D., Cantley, L.C., et al. (2007). Loss of the Stace, C.L., and Ktistakis, N.T. (2006). Phosphatidic acid- and phosphatidyl-
Par-1b/MARK2 polarity kinase leads to increased metabolic rate, decreased serine-binding proteins. Biochim. Biophys. Acta 1761, 913–926.
adiposity, and insulin hypersensitivity in vivo. Proc. Natl. Acad. Sci. USA Stahelin, R.V., Karathanassis, D., Murray, D., Williams, R.L., and Cho, W.
104, 5680–5685. (2007). Structural and membrane binding analysis of the Phox homology
Isakoff, S.J., Cardozo, T., Andreev, J., Li, Z., Ferguson, K.M., Abagyan, R., domain of Bem1p: basis of phosphatidylinositol 4-phosphate specificity.
Lemmon, M.A., Aronheim, A., and Skolnik, E.Y. (1998). Identification and anal- J. Biol. Chem. 282, 25737–25747.
ysis of PH domain-containing targets of phosphatidylinositol 3-kinase using Stefan, C.J., Audhya, A., and Emr, S.D. (2002). The yeast synaptojanin-like
a novel in vivo assay in yeast. EMBO J. 17, 5374–5387. proteins control the cellular distribution of phosphatidylinositol (4,5)-bisphos-
Kavran, J.M., Klein, D.E., Lee, A., Falasca, M., Isakoff, S.J., Skolnik, E.Y., and phate. Mol. Biol. Cell 13, 542–557.
Lemmon, M.A. (1998). Specificity and promiscuity in phosphoinositide binding Szentpetery, Z., Balla, A., Kim, Y.J., Lemmon, M.A., and Balla, T. (2009). Live
by pleckstrin homology domains. J. Biol. Chem. 273, 30497–30508. cell imaging with protein domains capable of recognizing phosphatidylinositol
Lemmon, M.A. (2008). Membrane recognition by phospholipid-binding 4,5-bisphosphate; a comparative study. BMC Cell Biol. 10, 67.
domains. Nat. Rev. Mol. Cell Biol. 9, 99–111. Szkotnicki, L., Crutchley, J.M., Zyla, T.R., Bardes, E.S.G., and Lew, D.J.
Lew, D.J. (2003). The morphogenesis checkpoint: how yeast cells watch their (2008). The checkpoint kinase Hsl1p is activated by Elm1p-dependent phos-
figures. Curr. Opin. Cell Biol. 15, 648–653. phorylation. Mol. Biol. Cell 19, 4675–4686.
Longtine, M.S., and Bi, E. (2003). Regulation of septin organization and func- Tassan, J.P., and Le Goff, X. (2004). An overview of the KIN1/PAR-1/MARK
tion in yeast. Trends Cell Biol. 13, 403–409. kinase family. Biol. Cell 96, 193–199.

976 Cell 143, 966–977, December 10, 2010 ª2010 Elsevier Inc.
Timm, T., Marx, A., Panneerselvam, S., Mandelkow, E., and Mandelkow, E.M. Yeung, T., Gilbert, G.E., Shi, J., Silvius, J., Kapus, A., and Grinstein, S. (2008).
(2008). Structure and regulation of MARK, a kinase involved in abnormal phos- Membrane phosphatidylserine regulates surface charge and protein localiza-
phorylation of Tau protein. BMC Neurosci. 9 (Suppl 2), S9. tion. Science 319, 210–213.
Tochio, N., Koshiba, S., Kobayashi, N., Inoue, M., Yabuki, T., Aoki, M., Seki, E.,
Matsuda, T., Tomo, Y., Motoda, Y., et al. (2006). Solution structure of the Yu, J.W., Mendrola, J.M., Audhya, A., Singh, S., Keleti, D., DeWald, D.B.,
kinase-associated domain 1 of mouse microtubule-associated protein/micro- Murray, D., Emr, S.D., and Lemmon, M.A. (2004). Genome-wide analysis of
tubule affinity-regulating kinase 3. Protein Sci. 15, 2534–2543. membrane targeting by S. cerevisiae pleckstrin homology domains. Mol.
Cell 13, 677–688.
Townley, R., and Shapiro, L. (2007). Crystal structures of the adenylate sensor
from fission yeast AMP-activated protein kinase. Science 315, 1726–1729.
Zhang, J., Kong, C., Xie, H., McPherson, P.S., Grinstein, S., and Trimble, W.S.
Wood, C.S., Schmitz, K.R., Bessman, N.J., Setty, T.G., Ferguson, K.M., and (1999). Phosphatidylinositol polyphosphate binding to the mammalian septin
Burd, C.G. (2009). PtdIns4P recognition by Vps74/GOLPH3 links PtdIns H5 is modulated by GTP. Curr. Biol. 9, 1458–1467.
4-kinase signaling to retrograde Golgi trafficking. J. Cell Biol. 187, 967–975.
Yeung, T., Terebiznik, M., Yu, L., Silvius, J., Abidi, W.M., Philips, M., Levine, T., Zhu, H., Bilgin, M., Bangham, R., Hall, D., Casamayor, A., Bertone, P., Lan, N.,
Kapus, A., and Grinstein, S. (2006). Receptor activation alters inner surface Jansen, R., Bidlingmaier, S., Houfek, T., et al. (2001). Global analysis of protein
potential during phagocytosis. Science 313, 347–351. activities using proteome chips. Science 293, 2101–2105.

Cell 143, 966–977, December 10, 2010 ª2010 Elsevier Inc. 977
The Fused/Smurf Complex Controls the
Fate of Drosophila Germline Stem Cells
by Generating a Gradient BMP Response
Laixin Xia,1,2,4 Shunji Jia,3,4 Shoujun Huang,1,4 Hailong Wang,1 Yuanxiang Zhu,1 Yanjun Mu,1 Lijuan Kan,1 Wenjing Zheng,1
Di Wu,3 Xiaoming Li,2 Qinmiao Sun,2 Anming Meng,2,3 and Dahua Chen1,*
1State Key Laboratory of Reproductive Biology
2State Key Laboratory of Biomembrane and Membrane Biotechnology
Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
3College of Life Sciences, Tsinghua University, Beijing 100084, China
4These authors contributed equally to this work

*Correspondence: chendh@ioz.ac.cn
DOI 10.1016/j.cell.2010.11.022

SUMMARY maintain stem cells (Li and Xie, 2005; Ohlstein et al., 2004; Spra-
dling et al., 2001; Yamashita et al., 2005). The asymmetric
In the Drosophila ovary, germline stem cells (GSCs) division of GSCs takes place within a niche made up of a small
are maintained primarily by bone morphogenetic number of stromal cells (terminal filament, cap cells, and inner
protein (BMP) ligands produced by the stromal cells sheath cells) at the tip of the germarium (Figures 1A and 1C) to
of the niche. This signaling represses GSC differenti- produce two daughter cells along the anterior-posterior axis of
ation by blocking the transcription of the differentia- the ovary. The anterior daughter cell retains contact with the
stromal cap cells and becomes a stem cell, whereas the poste-
tion factor Bam. Remarkably, bam transcription
rior daughter cell dissociates from the cap cells but associates
begins only one cell diameter away from the GSC in
with inner sheath cells and becomes a cystoblast (CB), which
the daughter cystoblasts (CBs). How this steep divides four times to produce a cyst of 16 interconnected cells
gradient of response to BMP signaling is formed that can sustain oogenesis. The stromal cells form the niche by
has been unclear. Here, we show that Fused (Fu), secreting signaling ligands that direct the fate of GSCs and their
a serine/threonine kinase that regulates Hedgehog, immediate daughter cells (King et al., 2001; Song et al., 2004).
functions in concert with the E3 ligase Smurf to regu- Bone morphogenetic protein (BMP) ligands, Decapentaplegic
late ubiquitination and proteolysis of the BMP (Dpp) and Glass bottom boat (Gbb), produced from cap cells
receptor Thickveins in CBs. This regulation gener- (Song et al., 2004; Xie and Spradling, 1998), and perhaps other
ates a steep gradient of BMP activity between niche cells, maintain GSCs by suppressing GSC differentiation
GSCs and CBs, allowing for bam expression on (Figure 1B) (Chen and McKearin, 2003a; Song et al., 2004).
In GSCs, BMP signaling activates the Drosophila Smads, Mad
CBs and concomitant differentiation. We observed
(the Drosophila Smad1/5/8 homolog) and Medea (the Drosophila
similar roles for Fu during embryonic development
Smad4 homolog), that bind to both the bag of marbles (bam)
in zebrafish and in human cell culture, implying broad transcriptional silencer element and the nuclear membrane
conservation of this mechanism. protein Otefin, resulting in bam transcriptional silencing (Chen
and McKearin, 2003a; Jiang et al., 2008; Song et al., 2004). Given
INTRODUCTION that bam expression is essential for differentiation of CBs, cells
with active BMP signaling cannot differentiate but remain
In adult tissues, stem cells execute asymmetric cell divisions to GSCs by default. Thus, bam silencing is the hallmark of asymme-
self-renew and produce differentiated daughters for maintaining try in the Drosophila ovarian germline stem cell niche, and its
tissue homeostasis via interaction with their surrounding stromal range is restricted to one cell diameter at the most anterior end
cells, which form a microenvironment commonly termed as of the germarium (Chen and McKearin, 2003b).
a niche (Nishikawa et al., 2008; Spradling et al., 2008). Although How is this very steep gradient of BMP response formed? One
the signaling pathways involved in this interaction have been possible explanation is that Dpp/Gbb ligands are secreted only
identified in many stem cell populations, the mechanisms to from one point source, such as cap cells. Previous studies,
explain how stem cells and their specialized sisters differentially however, have suggested that the Dpp ligands are present in
respond to and interpret the signals from the niche remain poorly both cap cells and inner sheath cells (Casanueva and Ferguson,
understood. 2004; Song et al., 2004), raising the likelihood that Dpp ligands
The germline stem cells (GSCs) in the Drosophila ovary have are not restricted to a single source. An alternative possibility
provided heuristic examples for understanding the niches that (Figure 1B) is that CBs develop a cell-autonomous mechanism

978 Cell 143, 978–990, December 10, 2010 ª2010 Elsevier Inc.
Figure 1. A Dpp Antagonist Is Required for
A B
the Proper Differentiation of CBs
(A) A schematic diagram of the germarium, with
different cell types and organelles indicated as
follows: terminal filament (TF), cap cells (CPC),
inner germarium sheath cells (IGC), germline
stem cells (GSC), cystoblast cells (CB), follicle cells
(FC), somatic stem cells (SSC), cyst (differentiated
germ cells with extended or branched fusomes),
and spectrosome (Sp). Among these, TFs, CPCs,
C D and IGCs produce Dpp ligands.
(B–M) Schematic diagram summarizing that dpp
signal from CPCs silences bam transcription and
is necessary for maintaining the self-renewal of
GSCs. CBs are exposed to the Dpp signal but are
bam active, raising the hypothesis that Dpp antag-
E F onism involves CB differentiation. Ovaries collected
from wild-type w1118 (C), P{nosP-gal4:vp16}/P
{uasp-tkv(ca)} (D), P{bamP-gal4:vp16}/P{uasp-
tkv(ca)} (E), and P{bamP-tkv(ca)} (F) flies were
stained with anti-Vasa (green) and anti-Hts (red)
G H antibodies. Anti-Hts was used to outline the germa-
rium and the morphology of the fusome, and the
staining of anti-Vasa was used to visualize all
germ cells in the germarium and egg chambers.
Ovaries from wild-type w1118 (G) and P{bamP-tkv
I J (ca)} (H) flies were stained with anti-Vasa (green)
and anti-BamC (red) antibodies. Ovaries from
wild-type w1118 (I) and P{bamP-tkv(ca)} (J) flies
were stained with anti-BamC (green) and anti-Hts
(red) antibodies. Ovaries from P{bamP-gfp} (K), P
K L M {bamP-tkv:gfp} (L), and P{bamP-tkv(ca):gfp} (M)
were stained with anti-GFP (green) and anti-Hts
(red) antibodies.
(N–P) Quantitative PCR (N and O) and Western blot
(P) analysis of gfp and bam expression in P{bamP-
gfp}, P{bamP-tkv:gfp}, and P{bamP-tkv(ca):gfp}
ovaries. Scale bar, 10 mm.
N O P The experiments were carried out by duplicates,
and the standard deviations were calculated by
Excel. See also Figure S1.

in a spatiotemporal manner (Itoh and


ten Dijke, 2007; Kitisin et al., 2007). In
Drosophila ovary, it has been shown that
BMP signaling maintains GSCs, whereas
diminished signaling, such as that pro-
duced by the action of Drosophila smurf,
to antagonize BMP/Dpp activity and derepress bam transcrip- promotes CB differentiation (Casanueva and Ferguson, 2004).
tion to promote their differentiation. However, the molecular mechanisms underlying the Smurf-
The transforming growth factor b (TGFb) and BMP signals play mediated regulation of BMP in Drosophila germline cells remain
important roles in controlling diverse normal developmental elusive. In this study, we have identified a mechanism involving
processes as well as tissue homeostasis (Feng and Derynck, Fused (Fu), a serine/threonine kinase, which regulates
2005; Wu and Hill, 2009). Dysregulation of TGFb/BMP signals Hedgehog (Hh) signaling as a core component of Hh-signaling
results in numerous developmental abnormalities and has complexes, functions in concert with Smurf to promote the
been linked to many human diseases, including cancer and proper turnover of Thickveins (Tkv), and generates a steep
degenerative diseases. Therefore the precise activity of TGFb/ gradient of BMP activity between GSCs and CBs. In addition,
BMP must be tightly controlled. TGFb/BMP signaling has been we find that the roles of Fu in regulating the BMP/TGFb signaling
proposed to be balanced through the regulation of Smads pathway are conserved in zebrafish during embryonic develop-
and/or their receptors to trigger distinct target gene expression ment and in human cell cultures.

Cell 143, 978–990, December 10, 2010 ª2010 Elsevier Inc. 979
RESULTS ing factor(s). Mass spectrometry analysis of Flag-Tkv complexes
from S2 cells, which were treated with MG132, revealed that
CB Differentiation Involves Antagonism of BMP Fused (Fu), which has been demonstrated as a positive regulator
Signaling in Hh signaling, was present in the Tkv complex (Figure 2A).
To understand the mechanism underlying the formation of Reciprocal immunoprecipitation experiments showed that Fu
a steep gradient of BMP response between GSCs and differen- and Tkv could be coimmunoprecipitated with each other in
tiated CBs, we used a transgene that expressed the constitu- transfected S2 cells (Figures 2B and 2C), indicating that Fu and
tively active Dpp receptor, Tkv(ca) (Wieser et al., 1995), to Tkv could form a complex together. Domain mapping of Tkv
explore the sensitivity of CBs to BMP signaling. It has been showed that the fragment lacking extracellular and transmem-
shown that driving Tkv(ca) expression in pole cells, primordial brane regions exhibited the strongest binding activity to Fu
germ cells, and adult germ cells with a nanos promoter (Van (Figure 2F), although all of the truncation mutants of Tkv
Doren et al., 1998) blocked bam transcription, prevented GSC (Figure 2D) interacted with Fu. Domain mapping of Fu showed
differentiation, and caused germ cell hyperplasia (Casanueva that both the N and C terminus of Fu could associate with Tkv
and Ferguson, 2004; Figure 1D). We were surprised, however, (Figures 2E and 2G). Further detailed domain mapping analysis
to find that controlling expression of Tkv(ca) with a bam promoter revealed that the STYKc domain is essential for Tkv interaction
(Chen and McKearin, 2003b) permitted normal germline devel- with the N terminus of Fu (Figures S2A–S2D).
opment (Figure 1E). To exclude the possibility that transcriptional
delays accounted for the failure of Tkv(ca) to block bam expres- fu Is Required for CB Differentiation by Antagonizing
sion due to the bipartite strategy, we attempted to transcribe the BMP/Dpp Signaling
Tkv(ca) transgene P{bamP-gal4:vp16}; P{uasp-tkv(ca)}. We To test whether Fu acts in balancing BMP/Dpp signal activity by
therefore repeated the experiment with the new transgenes, regulating Tkv to control the fate of GSCs and CBs, we examined
P{bamP-tkv(ca)} or P{bamP-tkv(ca):gfp}, in which either tkv(ca) the behavior of fu A mutant germ cells at an early stage by
or tkv(ca):gfp was placed directly under the control of the bam measuring the number of germ cells carrying spectrosomes in
promoter. These transgenes produced normal oogenesis and ovaries using a previously described method (Cox et al., 2000).
wild-type expression patterns of Bam and Hts proteins in ovaries We observed that, in contrast to the wild-type control, the fu A
(Figures 1F–1J). Whereas females carrying either the P{bamP-tkv mutant contained multiple types of germaria, with each type
(ca)} or P{bamP-tkv(ca):gfp} transgene were fertile, transgenic carrying different numbers of the spectrosome-containing
males were sterile, and their testes filled with many undifferenti- germ cells. Approximately 10% of germaria (n = 113) contained
ated germ cells lacking Bam expression (Figure S1 available a normal number of the spectrosome-containing germ cells per
online), indicating that these transgenes were indeed active. germarium (Figure 2H), nearly 60% of germaria (n = 113) con-
Thus, our results suggested that, in contrast to GSCs, CBs tained 5–10 GSC-like cells, and 30% of germaria (n = 113)
become insensitive to BMP signaling. were tumorous (Figures 2H–2J and 2L), suggesting that loss of
fu blocks or delays GSC/CB differentiation. Because the defects
Tkv(ca) Protein Is Subject to Degradation in CBs of GSC/CB differentiation associated with the fu mutant can be
To investigate the mechanism underlying the potential antagonism rescued by the transgene P{fuP-fu} (Figures 2K and L), we
of BMP signaling in CBs, we examined Tkv(ca):GFP expression concluded that fu is required for the proper differentiation of
driven by the bam promoter at both the transcriptional and protein GSCs/CBs.
levels. As shown in a quantitative RT-PCR analysis, there was To determine whether fu has a cell-autonomous role in
similar gfp expression in P{bamP-gfp}ovaries and tkv:gfp (a wild- promoting germ cell differentiation, we specifically knocked
type form of tkv tagged with gfp) expression in P{bamP-tkv:gfp} down fu in CBs by constructing P{uasp-shmiR-fu}; P{bamP-
ovaries, with tkv(ca):gfp expression present at normal levels in gal4:vp16} flies according to a method described previously
P{bamP-tkv(ca):gfp} ovaries (Figure 1N). Consistent with this (Haley et al., 2008). As shown in Figures S3A–S3E, knockdown
observation, no difference in the endogenous bam expression of fu by the bam promoter increased the number of GSC-like cells
was detected in ovaries of these transgene flies (Figure 1O), sug- to nearly seven per germarium (n = 72) (Figure S3B). Similarly, in
gesting that the bam promoter had normal transcriptional activity P{uasp-shmiR-fu}; P{nosP-gal4:vp16} ovaries, 90% of germa-
in P{bamP-tkv(ca):gfp} ovaries. We then performed analysis by ria (n = 111) contained 5–10 GSC-like cells (Figure S3C), and
both immunostaining and western blot to examine the expression nearly 5% of germaria were tumorous (Figure S3C0 ). Thus, fu
of Tkv(ca):GFP in P{bamP-tkv(ca):gfp} ovaries. As shown in has a cell-autonomous role in promoting germ cell differentiation.
Figures 1K–1M and 1P, GFP and Tkv:GFP were easily detected We then asked whether the kinase activity was essential for
in control ovaries from P{bamP-gfp} and P{bamP-tkv:gfp} trans- the function of Fu in germ cells by generating a transgene line,
gene flies, respectively. However, no apparent expression of Tkv P{fuP-fuKD}, which expresses a kinase dead form of Fu, FuKD,
(ca):GFP was observed in P{bamP-tkv(ca):gfp} ovaries, revealing by the fu promoter. As shown in Figures S3F and S3G, in contrast
the existence of a mechanism that negatively regulates the acti- to P{fuP-fu}, P{fuP-fuKD} completely failed to rescue germ cell
vated form of Tkv at the protein level in CBs. defects in fu mutant, revealing that fu acts in a kinase-dependent
manner for germ cell differentiation.
Identification of Fu as a Tkv-Interacting Factor Previous studies have shown that CB differentiation was
To explore how Tkv is regulated, we performed immunoprecipi- controlled by either the bam-dependent or bam-independent
tation followed by mass spectrometry to search for Tkv-interact- pathway (Chen and McKearin, 2005; Szakmary et al., 2005).

980 Cell 143, 978–990, December 10, 2010 ª2010 Elsevier Inc.
A B C Figure 2. Identification of Fu as a Tkv-Inter-
acting Protein
(A) Lysates from S2 cells expressing Flag-tagged
Tkv were immunoprecipitated with Flag beads
and then fractionated by electrophoresis through
polyacrylamide gels followed by staining with
silver. Mass spectrometry analysis showed that
the amino acid sequence of two peptides, as indi-
cated, matched the Drosophila Fu protein.
(B and C) S2 cells were transfected with combina-
tions of DNA constructs as indicated. At 48 hr
posttransfection, lysates from transfected S2 cells
were immunoprecipitated with anti-Myc antibody
(B) or anti-Flag M2 affinity gel (C). Western blots
were performed to analyze the presence of Flag-
F or Myc-tagged proteins.
D (D and E) Schematic drawings of Tkv (D) and Fu (E)
and their deletion mutants correspond to (F) and (G).
(F and G) S2 cells were transfected with different
combinations of constructs. Lysates from trans-
fected S2 cells were immunoprecipitated with anti-
Flag M2 affinity gel (F) or with anti-Myc antibody.
Western blots were performed to analyze the pres-
ence of Flag- or Myc-tagged protein as indicated.
E (H–K) Ovaries from wild-type w1118, fu mutant, and fu
mutant flies carrying the P{fuP-fu} transgene were
stained with anti-Vasa (green) and anti-Hts (red) anti-
G bodies.
(L) Quantitative analysis of the percentage of germa-
ria types in wild-type, fu mutants, and fu mutants
H carrying the P{fuP-fu} transgene. The x axis shows
genotypes of tested flies, whereas the y axis shows
the percentage of types of germaria in different
genotypes. Scale bar, 10 mm.
I
See also Figure S2.

J L

negative and lacZ was positive in these


cells (Figures 3D–3G). To test whether
the induction of GSC-like cells through
K the loss of fu depends on the activity
of the dpp signal, we employed the trans-
gene P{uasp-dad} (Jiang et al., 2008) to
overexpress Dad (the Drosophila Smad6/
7 homolog), a BMP/Dpp inhibitor. As
shown in Figures S3J–S3L, ectopic
expression of Dad also completely drove
fu mutant germ cell differentiation, sug-
To define the pathway through which fu acts, we overexpressed gesting that induction of GSC-like cells through the loss of fu
bam on a fu mutant background using the transgene P{hs-bam} depends on Dpp signaling. Taken together, our findings strongly
(Ohlstein and McKearin, 1997). As shown in Figures S3H and argue that fu is intrinsically required for GSC and CB differentiation
S3I, ectopic expression of bam completely drove fu mutant by antagonizing Dpp signaling.
germ cell differentiation, suggesting that fu acts mainly in a bam-
dependent manner for the differentiation of GSCs and CBs, raising Fu Negatively Regulates BMP/Dpp Signaling
the possibility that fu acts as a negative component of the Dpp by Controlling Tkv Stability
pathway. We then tested whether the ectopic GSC-like cells in Given that Fu forms a complex with Tkv, we then asked whether
fu mutants respond to Dpp signaling by introducing the Dpp- fu has a direct role in affecting Dpp signaling through regulating
responsive reporters, bamP-gfp and dad-lacZ, into the fu mutant the expression of Tkv and established a bam transcription-
background. In agreement with previous findings (Narbonne-Re- dependent luciferase reporter assay in S2 cells. As shown in
veau et al., 2006), we found that many of the fu-inducing GSC- Figure 3A, the bam transcription reporter was silenced by the
like cells behaved as GSCs rather than CBs, given that gfp was expression of Tkv(ca) in a dose-dependent manner, which

Cell 143, 978–990, December 10, 2010 ª2010 Elsevier Inc. 981
A B C Figure 3. Fu Negatively Regulates BMP/
Dpp Signaling by Controlling Tkv Stability
(A) The S2 cells were transfected with the bamP-
luciferase reporter with gradient concentrations
of actinP-tkv(ca). At 48 hr posttransfection, cells
were harvested for luciferase analysis.
(B) The S2 cells were transfected with bamP-lucif-
erase and actinP-tkv(ca) and also treated with
dsRNAs of fu or gfp. Knockdown of fu enhanced
the repression of the bam reporter by Tkv(ca).
(C) The S2 cells were transfected with pMT-tkv(ca)
and actinP-lacZ constructs or were also treated
with dsRNAs of fu or gfp. Western blots were per-
formed to analyze the presence of Myc-tagged
D E Tkv(ca).
F
(D and E) Ovaries from P{bamP-gfp} and fu mutant
flies carrying P{bamP-gfp} were stained with anti-
GFP (green) and anti-Hts (red) antibodies.
(F and G) Ovaries from P{dad-lacZ} and fu mutant
flies carrying P{dad-lacZ} were stained with anti-
G H I Vasa (green) and anti-b-gal (red) antibodies.
(H–J) Ovaries from different genotype flies as indi-
cated were stained with anti-Vasa (green) and anti-
Hts (red) antibodies.
(K and L) Ovaries from the indicated flies were
stained with anti-Vasa (green) and anti-BamC
J K L
(red) antibodies.
(M and N) Ovaries from fu and fu mutant flies
carrying P{bamP-tkv(ca)} were stained with anti-
Vasa (green) and anti-Hts (red) antibodies.
(O) Quantitative analysis of the percentage of ger-
maria types as indicated in wild-type, fu mutant,
and fu mutant carrying the P{bamP-tkv(ca)} trans-
M O
gene. Scale bar, 10 mm.
The experiments were carried out by duplicates,
and the standard deviations were calculated by
Excel. See also Figure S3.

N
(Jia et al., 2003; Claret et al., 2007),
partially suppressed the overexpression
of Tkv(ca) driven by the nanos promoter,
as indicated by the presence of
branched fusomes and ectopic Bam
expression, as well as 30% of ovarioles
(n = 50) carrying normal egg chambers,
in P{uasp-tkv(ca)}; P{nosP-gal4:vp16}/
P{uasp-SRC-fu} ovaries (Figures 3H–3L).
Taken together, we argue that Fu nega-
mimics the response of the bam promoter to Dpp signaling in tively regulates Tkv stability to determine the fate of GSCs
the in vivo GSC system. Of interest, we found that knockdown and CBs.
of fu in S2 cells increased stability of the Tkv protein (Figure 3C)
and accordingly enhanced Tkv-mediated bam transcriptional Smurf Interacts Physically and Genetically with Tkv
silencing (Figure 3B), indicating that knockdown of fu influences We noted that the phenotype of the GSC-like cells in the fu mutant
the Dpp signal by stabilizing the Tkv protein. To confirm this ovary resembled that in the Drosophila smurf mutant. It has been
finding, we performed a genetic assay by constructing the strain shown that smurf antagonizes BMP signaling by targeting phos-
fu; P{bamP-tkv(ca):gfp}/+. As shown in Figures 3M–3O, consti- phorylated Mad for degradation in Drosophila somatic cells (Liang
tutive dpp signaling from the transgene P{bamP-tkv(ca):gfp} et al., 2003; Podos et al., 2001). In ovaries, smurf transcript is ubiq-
resulted in a stronger tumorous germarium phenotype in the uitously present in the germarium (Figures S4E and S4F), and loss
fu mutant background than that in fu mutant alone. Consis- of smurf delays the differentiation of CBs (Casanueva and Fergu-
tently, overexpression of an activated form of Fu, in which the son, 2004). However, the molecular mechanism underlying the
Fu protein was tagged with an SRC domain at its N terminus action of smurf in CBs remains unknown. To test whether smurf

982 Cell 143, 978–990, December 10, 2010 ª2010 Elsevier Inc.
A B C Figure 4. Fu Physically and Genetically
Interacts with Smurf
(A and B) S2 cells were transfected with combina-
tions of DNA constructs as indicated. At 48 hr
posttransfection, lysates from transfected S2 cells
were immunoprecipitated with anti-Flag M2
affinity gel. Western blots were performed to
analyze the presence of Myc-tagged (A) or HA-
tagged (B) proteins as indicated.
(C) Ovarian extracts from P{uasp-HA:fu}; P{nosP-
gal4:vp16} and w1118 flies were immunoprecipi-
tated with anti-HA antibody. Western blots were
performed with anti-Smurf and anti-HA antibodies
D E to analyze the presence of Smurf and HA:Fu
proteins, respectively, as indicated.
(D and E) Schematic drawings of Smurf (D) and Fu (E)
and their deletion mutants correspond to (F) and (G).
(F and G) S2 cells were transfected with different
combinations of DNA constructs. Lysates from
transfected S2 cells were immunoprecipitated with
anti-Flag M2 affinity gel (F) or anti-Myc antibody
(G). Western blots were performed to analyze the
G presence of Myc- or Flag-tagged proteins (F) or the
F presence of HA- or Myc-tagged proteins (G).
(H) Quantitative analysis of the percentage of germa-
ria types in different genotypes.
(I) The S2 cells were transfected with bamP-luc-
iferase, actinP-lacZ, and actinP-tkv(ca) and were
also treated with dsRNAs of either fu or smurf, or
both. The gfp dsRNA was used as a control.
The experiments were carried out by duplicates, and
the standard deviations were calculated by Excel.
See also Figure S4.

a tumorous germarium, and 62% of the


ovarioles (n = 84) contained tumorous ger-
H I
maria that were attached to one or several
egg chambers, suggesting that, like in the
fu mutant background, smurf mutant
germ cells were also much more sensitive
to Dpp signaling than were smurf+ cells.

Fu Interacts Physically and


Genetically with Smurf
To explore whether fu acts on a common
pathway with smurf to regulate Tkv and
accordingly control BMP signal activity,
we determined whether Smurf physically
interacts with the Fu protein by performing
reciprocal immunoprecipitation assays in
is involved in regulating Tkv, we performed coimmunoprecipitation S2 cells. As shown in Figures 4A and 4B, Smurf and Fu coimmuno-
and reporter assays as well as ubiquitination analysis of Tkv in S2 precipitated with each other in transfected S2 cells. Consistently,
cells. As shown in Figures S4A and S4B, Smurf and Tkv coimmu- we found that endogenous Smurf physically associated with
noprecipitated with each other. Knockdown of smurf reduced the HA:Fu in P{uasp-HA:fu}; P{nosP-gal4:vp16} ovaries (Figure 4C).
ubiquitination of Tkv (Figure 5F) and accordingly enhanced Tkv- These results suggested that Fu could form a complex with Smurf
mediated bam reporter silencing (Figure 4I). To determine the bio- in both S2 cells and germ cells. To map the essential domain in
logical importance of this interaction in vivo, we examined the Smurf that interacts with Fu, we generated truncated forms of
genetic relationship between smurf and tkv in the ovary. As shown Smurf. As shown in Figures 4D and 4F, the HECT domain is an
in Figures S4C and S4D, overexpression of Tkv(ca) driven by the essential domain for Smurf to interact with Fu. We then determined
bam promoter in the smurf mutant strongly blocked CB differenti- the region of Fu required for interaction with Smurf. As shown in
ation. Nearly 38% of the ovarioles (n = 84) was composed of Figures 4E and 4G, both the N and C terminus of Fu could

Cell 143, 978–990, December 10, 2010 ª2010 Elsevier Inc. 983
A B Figure 5. Fu in Concert with Smurf Targets
Tkv for Ubiquitination
(A and B) S2 cells were transfected with different
combinations of constructs as indicated. Lysates
from transfected S2 cells were used in a two-
step immunoprecipitation method employing
anti-Flag and anti-Myc successively, and western
blots were performed to analyze the presence of
HA-tagged Smurf, Myc-tagged Fu, or Flag-tagged
Tkv as indicated.
(C and D) Ovaries from different genotype flies as
indicated were stained with anti-Vasa (green) and
F G anti-Hts (red) antibodies.
C (E) Ovaries from the indicated flies were stained
with anti-Vasa (green) and anti-BamC (red) anti-
bodies. Scale bar, 10 mm.
(F and G) In vivo assay of Tkv ubiquitination. S2 cells
were transfected with DNA combinations, including
D Myc and His double epitope-tagged Tkv(ca) and HA
epitope-tagged Ubiquitin (Ub) with dsRNAs of gfp
(as a control) or smurf (F) or fu (G) treatment, or
were transfected with FuKD, the kinase dead form
of Fu (G). Western blots were performed to analyze
the ubiquitination product of Tkv.
(H and I) An in vitro ubiquitin reaction was reconsti-
E tuted with components that contained HA-Ub, E1,
E2, Flag-Smurf complexes purified from S2 cells,
and the Myc:TkvC (Figure 2D) produced by in vitro
translation as indicated in lane 2 (lane 1 was a control
lacking Flag-Smurf complexes). In lane 3, the ubiqui-
tin reaction was the same as that in lane 2 except
that Flag-Smurf complexes purified from S2 cells
H I were treated with fu dsRNA. Western blots were per-
formed to analyze ubiquitination products using the
antibodies indicated.

Experimental Procedures). As shown in


Figures 5A and 5B, after the two-step
immunoprecipitations, both Flag-Tkv and
HA-Smurf were present in the Myc-Fu
complex, suggesting that Fu, Smurf, and
Tkv form a trimeric complex rather than
mutually exclusive heterodimers such as
Fu/Smurf, Fu/Tkv, and Smurf/Tkv, raising
the possibility that Fu, like Smurf, is
involved in ubiquitination of Tkv. We then
coimmunoprecipitate with Smurf. To test the genetic relationship evaluated whether Fu was also involved in ubiquitination of Tkv.
between smurf and fu, we constructed smurf and fu double As shown in Figure 5G, knockdown of fu greatly reduced the conju-
mutants and found that the ovaries in these double mutants closely gation of ubiquitin to Tkv, suggesting that, like Smurf, the Fu
resembled those in the fu single-mutant ovaries (Figure 4H). protein is also essential for Tkv ubiquitination. Given that Fu is
Consistently, as shown in Figure 4I, there was no greater effect a serine/threonine protein kinase, we then tested whether Fu
on the bam-luc reporter by knockdown of both smurf and fu supports Tkv ubiquitination in a kinase-dependent manner by
compared with knockdown of smurf or fu alone. Together, these using the kinase dead form of Fu, FuKD. As shown in Figure 5G,
data support that Fu and Smurf are functionally dependent upon the efficiency of Tkv ubiquitination was greatly reduced when
each other and act in a complex by regulating BMP/Dpp activity. FuKD was overexpressed in S2 cells, indicating that the kinase
activity of Fu is important for Fu-mediated ubiquitination of Tkv.
Fu, Smurf, and Tkv Form a Trimeric Complex to Promote To substantiate the model that Fu functions in concert with
Tkv Ubiquitination Smurf to catalyze the ubiquitination of Tkv, we performed
To determine whether Fu, Smurf, and Tkv formed a trimeric biochemical assays to assess the Smurf E3 ligase activity in the
complex, we coexpressed Flag-Tkv, Myc-Fu, and HA-Smurf in Fu/Smurf complexes by reconstituting Tkv ubiquitination
S2 cells and performed two-step immunoprecipitation (Extended in vitro. Smurf complex from S2 cell lysates efficiently supports

984 Cell 143, 978–990, December 10, 2010 ª2010 Elsevier Inc.
A B Figure 6. Identification of the S238 Site,
a Putative Phosphorylation Site, Is Critical
for Tkv(ca) Ubiquitination and Degradation
(A) Schematic diagram showing the sequence of
the Tkv GS domain, which contains multiple S/T
sites. A series of mutant forms of Tkv(ca)
constructs, in which the S/T sites as indicated
were individually mutated to A, was generated.
(B) The S2 cells were transfected with bamP-luc-
iferase, actinP-Renilla, and actinP-tkv(ca) or
mutant forms of tkv(ca) as indicated.
(C and D) Luciferase reporter analysis and protein
C D E stability assay for Tkv(ca) and Tkv(ca)S238A
proteins revealed that Tkv(ca)S238A has stronger
stability than Tkv(ca).
(E) Ubiquitination analysis for Tkv(ca) and Tkv(ca)
S238A proteins showed that Tkv(ca)S238A protein
is resistant to ubiquitin, compared with Tkv(ca).
(F and G) Ovaries from P{bamP-tkv(ca)} and
P{bamP-tkv(ca)S238A} were stained with anti-
Vasa (green) and anti-Hts (red) antibodies. Scale
bar, 10 mm.
(H) The diagram shows that, in contrast to GSCs
that undergo self-renewal, CBs develop a BMP/
Dpp antagonistic pathway mediated by a Fu/
Smurf complex to degrade Tkv for their differenti-
F G
ation.
(I) Schematic diagram summarizes a conserved
mechanism in the regulation of BMP/TGFb
signaling.
The experiments were carried out by duplicates,
and the standard deviations were calculated by
Excel. See also Figure S5.

H I

understanding the mechanism of how


Tkv is regulated by searching for the
specific S/T site(s) in Tkv(ca). Of interest,
a previous study has implicated that
several S/T sites in the GS domain of
TGFb type I receptor were subjected to
phosphorylation in cell culture assays
(Wrana et al., 1994). We therefore specu-
lated that one of the corresponding sites
in the GS domain of Tkv might be impor-
tant for Tkv ubiquitination and degrada-
ubiquitination of Tkv, whereas those from S2 cells treated with tion. To test this hypothesis, we generated a series of mutant
dsRNA of fu showed significantly reduced activity toward Tkv forms of Tkv(ca) constructs in which the S/T sites, as indicated
ubiquitination (Figures 5H and 5I), suggesting that Smurf ubiqui- in Figure 6A and Figure S5A were individually mutated to A.
tinates Tkv in a Fu-dependent manner. To verify the importance We investigated whether these mutant forms of Tkv(ca) affected
of the coordination between Fu and Smurf in vivo, we performed the response of bamP-luc reporter in S2 cells. As shown Figures
a genetic assay and found that co-overexpression of Smurf and 6B and 6C and Figure S5B, one of the mutant forms of Tkv(ca),
SRC-Fu strongly suppressed Tkv(ca) overexpression as indi- Tkv(ca)S238A, exhibited the strongest transcriptional silencing
cated by the presence of the branched fusomes and expression activity on the bamP-luc reporter. To evaluate whether the
of Bam protein, as well as nearly 50% of ovarioles (n > 100) S238 site is responsible for controlling the ubiquitination and
carrying normal egg chambers (Figures 5C–5E). stability of Tkv(ca), we performed ubiquitination assays on Tkv
(ca) and Tkv(ca)S238A. As shown in Figures 6D and 6E,
The Putative Phosphorylation Site of Tkv, S238, compared to Tkv(ca), Tkv(ca)S238A showed much stronger
Is Responsible for Tkv Ubiquitination and Degradation stability and appeared resistant to ubiquitination. Together with
Given that Fu regulates Tkv ubiquitination and degradation in the data in Figures 3B and 3C and Figure 5G, our findings
a kinase-dependent manner, we then turned our attention to support the notion that S238, a putative phosphorylation site,

Cell 143, 978–990, December 10, 2010 ª2010 Elsevier Inc. 985
is important for Tkv to respond to Fu and critical for Tkv ubiquiti- morphology, and only 24% and 18% of embryos showed D1
nation and degradation. and D2 dorsalization, respectively (Figure 7U). These results indi-
To determine the biological function of the S238 site, we cate that fu overexpression is able to inhibit Nodal-induced dors-
generated a transgene fly P{bamP-tkv(ca)S238A} that expresses alization. In contrast, upregulation of BMP signaling activity by
a mutant form of Tkv(ca) carrying the S238A mutation by the bam injecting bmp2b mRNA led to embryonic ventralization at
promoter. As shown in Figures 6F and 6G, ovaries from P{bamP- 24 hpf, with 28% (n = 141) exhibiting an onion-like shape, the
tkv(ca)} showed normal germline development, whereas in P strongest ventralized phenotype (V1); 27% having an enlarged
{bamP-tkv(ca)S238A} ovaries, expression of a ubiquitin-resistant tail and no head (V2, severely ventralized); and 44% showing
form of Tkv(ca), Tkv(ca)S238A, resulted in a tumorous germarium a smaller head (V3, moderate ventralization) (Figures 7P–7R
phenotype, demonstrating the biological importance of the S238 and 7U). Coinjection of fu and bmp2b mRNAs resulted in 81%
site of Tkv in germ cell differentiation. of embryos (n = 69) developing normally (Figure 7U), indicating
that fu overexpression also antagonizes bmp2b-induced
Fu/STK36 Has a Conserved Role in Regulating the BMP/ ventralization.
TGFb Signaling Pathway in Human Cell Cultures and in To test whether Fu has a role in the degradation of BMP recep-
Zebrafish during Embryonic Development tors in zebrafish, we made a zebrafish alk3a and GFP fusion
Given that FU (also called STK36 in vertebrates) is an evolution- mRNA (zalk3a-GFP). Consistent with the Drosophila data that
arily conserved protein in flies and vertebrates, we explored ectopic expression of Src:Fu downregulated Tkv(ca):GFP in
whether FU has a role in the regulation of BMP signaling in the early embryo (Figures S2E and S2F), as shown in Figures
human cell cultures. As shown in Figures S5C–S5H, in agree- S6G–S6J, coinjection with fu mRNA resulted in much weaker
ment with the data from Drosophila, FU/STK36 physically inter- fluorescence, compared with zalk3a-GFP mRNA injection alone,
acts with both SMURF proteins and ALK3, the type I receptor suggesting that fu might play a conserved role in degrading BMP
of BMP signaling (Figures S5C and S5D). Knockdown of receptors.
FU/STK36 reduced the ubiquitination of ALK3 (Figures S5E To further study the genetic relationship between Fu and BMP
and S5F) and accordingly enhanced the transcriptional response receptors, we used a well-defined dominant-negative form of
of BRE-luciferase (Figures S5G and S5H). These findings sug- BMP type I receptor (tBr). As shown in Figures S6K–S6Y, coin-
gested that FU/STK36 might have a conserved role in SMURF- jection of fu with tBr mRNA partially rescued the tBr-induced
mediated regulation of BMP signaling in mammals. dorsalized phenotype, whereas coinjection of fu-MO and tBr
To further explore the in vivo function of Fu/Stk36 in vertebrates, mRNA had no rescue effect. Considering that Nodal and BMP
we investigated the developmental roles of fu in zebrafish signals have opposite effects in DV patterning (Schier and
embryos. As shown in Figures S6A–S6F, the fu transcripts were Talbot, 2005), these results suggest that Fu antagonizes Nodal
present from the one-cell stage up to 24 hr postfertilization (hpf). signaling when BMP signaling is downregulated.
Knockdown of fu with a morpholino (fu-MO) (Wolff et al., 2003) Taken together, our results support that fu functions as
caused severe neural necrosis and growth retardation at 24 hpf a modulator in zebrafish DV patterning by antagonizing both
(Figure 7B), which was largely due to nonspecific activation of BMP and Nodal signaling.
the p53 signaling pathway (Robu et al., 2007) because
coinjection with p53MO reduced neural necrosis (Figure 7C). DISCUSSION
However, in contrast to the fu-cMO/p53MO coinjected embryos
(Figure 7A), fu-MO/p53MO coinjection resulted in dorsalized Previous studies have demonstrated that BMP/Dpp signals from
phenotypes that manifested as a shortened trunk (Figure 7C). the niche play primary roles in the self-renewal of GSCs by
The expression of gata1 in ventral mesoderm-derived hematopoi- silencing bam transcription (Chen and McKearin, 2003a; Song
etic progenitors was inhibited in the fu morphants (Figures 7F, 7G, et al., 2004). However, the mechanism by which the differenti-
and 7S), whereas the expression of the dorsal organizer marker ating CBs avoid the control of BMP/Dpp and activate bam
gsc in the morphants was expanded variably at the shield stage remains poorly understood. In this study, we have provided
(Figures 7J, 7K, and 7T). On the other hand, embryos injected direct evidence that the differentiating daughter cells of GSCs,
with fu mRNA exhibited a slight expansion of blood island, small known as CBs, become resistant to BMP signaling through
or fused eyes, and an abnormal notochord at 24 hpf (Figure 7D), degradation of Tkv in CBs. We showed that Fu functions as an
indicative of ventralization. In a high proportion of embryos injected antagonistic factor in BMP/Dpp signaling by regulating Tkv
with fu mRNA, gata1 expression was enhanced (Figures 7H and degradation during the differentiation of CBs. Moreover, we
7S) and gsc expression slightly reduced (Figures 7L and 7T). These provided both genetic and biochemical evidence that Fu acts
findings reveal that fu may be involved in the dorsoventral (DV) in concert with Smurf, a HECT domain-containing ubiquitin E3
patterning of zebrafish embryos. ligase, to regulate the ubiquitination of Tkv in the CB, thereby
We then investigated whether fu controls DV patterning by generating a steep gradient of response to BMP signaling
regulating Nodal/BMP signaling. Overexpression of sqt, a between GSCs and CBs for their fate determination (Figure 6H).
zebrafish Nodal ligand, caused variable degrees of dorsalized Finally, we showed a conserved role for fu in antagonizing BMP/
phenotypes at 24 hpf with 73% of embryos showing severe TGFb signals in zebrafish embryonic development as well as in
dorsalization (D1) and 20% showing relatively mild dorsalization human cell cultures. Our findings not only reveal a conserved
(D2) (n = 63; Figures 7N, 7O, and 7U). When fu and sqt mRNAs function of fu in controlling BMP/TGFb signal-mediated develop-
were coinjected, 58% of embryos (n = 62) had almost normal mental processes, but also provide a comprehensive view of

986 Cell 143, 978–990, December 10, 2010 ª2010 Elsevier Inc.
Figure 7. fu Participates in Dorsoventral
Patterning by Regulating both Nodal and
BMP Signaling Pathways in Zebrafish
(A and B) Embryonic morphology at 24 hpf after
downregulating or upregulating Fu activity.
Embryos injected with 5 ng fu-MO exhibited
more severe necrosis (B) than those injected with
5 ng fu-cMO/p53MO (A).
(C) Coinjection of 5 ng p53MO with 5 ng fu-MO
alleviated necrosis as observed in (B) but caused
dorsalized phenotypes.
(D) Overexpression of 300 pg fu mRNA led to ven-
tralized phenotypes.
(E–L) Examination of dorsoventral marker genes
gata1 (24 hpf) and gsc (shield stage). Compared
to control embryos injected with fu-cMO and
p53MO (E and I), 5 ng fu-MO injected alone (F
and J) or coinjected with 5 ng p53MO (G and K)
led to both gata1 inhibition and gsc expansion.
A 300 pg fu mRNA injection (H and L) led to an
expansion of gata1 and a slight reduction of gsc.
Statistical data are shown in (S) and (T). Embryo
orientations: lateral views with head to the left for
gata1; dorsal views with animal pole to the top
for gsc.
(M–R) Compared with the uninjected control (M),
embryos injected with 0.75 pg sqt mRNA were
classified into D1 and D2 groups of dorsalization
(N and O). Embryos injected with 10 pg bmp2b
mRNA were classified into V1–V3 groups of ven-
tralization (P, Q, and R).
(U) Statistical data for rescue experiments in which
300 pg fu mRNA was coinjected with 0.75 pg sqt or
10 pg bmp2b mRNA. Coinjection of fu mRNA
rescues sqt- or bmp2b-induced dorsoventral
patterning defects.
See also Figure S6.

early germ cell proliferation and a


tumorous germarium phenotype (Nar-
bonne-Reveau et al., 2006) and that our
biochemical evidence showed that Fu
forms a complex with Tkv and affects its
stability, we subsequently identified that
Fu as a component negatively regulates
BMP/Dpp signaling by interacting with
the BMP/Dpp type I receptor, Tkv.
BMP/TGFb signals play pivotal roles in
controlling diverse normal developmental
and cellular processes (Wu and Hill,
mechanisms that produce both self-renewal and asymmetry in 2009). In the canonical BMP/TGFb pathway, the receptors and
the division of stem cells. Smad proteins are the essential components for BMP/TGFb
signal transduction. However, this pathway is known to be
A Role for Fu in Smurf-Mediated Ubiquitination modulated by additional factors to reach physiological levels in
of BMP/TGFb Signaling a cellular context-dependent manner (Kitisin et al., 2007). Smurfs
Observations of the existence of a BMP resistance mechanism and HECT domain-containing proteins have been shown to
that controls the proper division of GSCs through the regulation antagonize BMP/TGFb signals through the regulation of the
of Tkv prompted us to explore how Tkv was regulated. Using stability of either receptors or Smads in vertebrates (Ebisawa
immunoprecipitation followed by mass spectrometry analysis, et al., 2001; Murakami et al., 2003). In Drosophila, Smurf has
we identified that Fu associates with the Tkv protein. Given previously been implicated in regulating proteolysis of phosphor-
that previous studies demonstrated that a loss of fu leads to ylated Smad proteins in somatic cells (Liang et al., 2003; Podos

Cell 143, 978–990, December 10, 2010 ª2010 Elsevier Inc. 987
et al., 2001). In the ovary, Smurf was also proposed to downre- determine whether the Fu/Smurf complex also plays roles in
gulate the level of BMP to promote CB differentiation (Casa- other signaling pathways.
nueva and Ferguson, 2004). The mechanism underlying the
action of Smurf in Drosophila early germline cells remains EXPERIMENTAL PROCEDURES
elusive. In this study, we showed that Fu, Smurf, and Tkv could
form a trimeric complex in S2 cells. Importantly, both Fu and Drosophila Strains
Fly stocks used in this study were maintained under standard culture condi-
Smurf are required for ubiquitination of Tkv in S2 cells and for
tions. The w1118 strain was used as the host for all P element-mediated
turnover of Tkv in germ cells. Combined with our genetic transformations. Strains P{bamP-gal4:vp16}, P{uasp-tkv(ca)} P{bamP-gfp},
evidence, we proposed that Fu and Smurf likely function in P{dad-lacZ}, smurf15c, and P{nosP-gal4:vp16} have been described previously
a common biochemical process by controlling Tkv degradation. (Casanueva and Ferguson, 2004; Chen and McKearin, 2003b; Van Doren et al.,
The present study reveals a mechanism by which Fu serves as 1998). Strains P{uasp-SRC-fu}, P{uasp-smurf}, P{bamP-tkv(ca)}, P{bamP-
an essential component in the Smurf-mediated degradation of tkv:gfp}, and P{bamP-tkv(ca):gfp} were made in this study. The fuA mutant
and the rescue transgene for the fu mutant, P{fuP-fu}, were a gift from Dr. Jin
the BMP/TGFb receptor, thereby terminating BMP/TGFb
Jiang. The transgene line, P{fuP-fuKD}, was generated to express the kinase
signaling and negatively regulating the downstream target genes dead form of Fu (FuG13V) in which the conserved glycine (G13) site of Fu was
of BMP/TGFb (Figure 6I). changed into a valine. The fu knockdown transgene line, P{uasp-shmiR-fu},
Because Fu is a putative serine/threonine protein kinase, the was generated according to the method described previously (Haley et al.,
question becomes how Fu acts on Tkv regulation in concert 2008). The detailed information of primers was described in the Extended
with Smurf. Given that knockdown of fu does not significantly Experimental Procedures.
change the pattern of autoubiquitination of Smurf itself (data
Immunohistochemistry for Drosophila Ovary
not shown), it is therefore likely that Tkv is a strong candidate
Ovaries were prepared for immunohistochemistry as described previously
substrate for Fu kinase. Although there is no assay system for (Chen and McKearin, 2005). The following primary antibody dilutions were
analyzing the kinase activity of Fu presently, in this study, we per- used: rabbit anti-GFP (1:5000, Invitrogen); mouse anti-Hts (1:500, DSHB);
formed mutagenesis assays and identified that the S238 in Tkv is rabbit and mouse anti-BamC (1:1000); rabbit anti-Vasa (1:1000, Santa Cruz);
important for Tkv(ca) to respond to Fu and is critical for Tkv(ca) and mouse anti-b Gal (1:1000 Promega). The following secondary antibodies
ubiquitination and degradation. Of note, we found that the ubiq- were used at a 1:200 dilution: goat anti-mouse Alexa568 and goat anti-rabbit
Alexa488 (Molecular Probes).
uitin-resistant form of Tkv(ca) [Tkv(ca)S238A] blocks CB differen-
tiation. A previous study has shown that the S189 site in TGF-b
Phenotypic Analysis
type-I receptor, the corresponding site of S238 in Tkv, was phos- Ovaries isolated from 3-day-old flies were incubated with Hts antibody, and
phorylated in the cell culture system (Wrana, et al., 1994). Our images were collected on a Zeiss LSM 510 Meta confocal microscope to count
results suggest that Fu likely acts on Tkv through targeting and the number of spherical spectrosomes/fusomes and to identify differentiated
phosphorylating the S238 site and subsequently leads to Tkv cysts with branched fusomes. This protocol was described previously (Cox
ubiquitination and degradation by Smurf. Nevertheless, it would et al., 2000).
be advantageous to develop a kinase assay system for Fu to
Anti-Fu and Anti-Smurf Antibodies
determine whether the S238 site in Tkv is an authentic phosphor-
The anti-Fu antibody was generated by immunizing rabbit with the recombi-
ylation site for Fu kinase in the future. nant protein His6-Fu (amino acids 260–431) produced in E. coli, and the
anti-Smurf antibody was generated by immunizing mice with the recombinant
A Conserved Role for Fused in the Regulation of BMP/ protein His6-Smurf protein (amino acids 1–300) produced in E. coli.
TGFb Signals
Previous genetic analyses revealed that Fu plays an evolution- Cell Culture, Immunoprecipitation, and Western Blot Analysis
arily conserved role in the proper activation of the Hh pathway S2 cells were cultured in Schneider’s Drosophila medium (Sigma). Transfec-
tion was performed using the calcium phosphate transfection method. Immu-
and functions downstream of the Hh receptor (Jiang and Hui,
noprecipitation and western blots were performed using protocols previously
2008; Sánchez-Herrero et al., 1996; Ruel et al., 2003; Wilson described (Jiang et al., 2008). The following reagents were used: rabbit and
et al., 2009). Increasing evidence has shown that the kinase Fu mouse anti-Myc and rabbit anti-HA (Santa Cruz); rabbit and mouse anti-Flag
regulates the Hh-signaling complex by targeting Cos2 and anti-Flag M2 affinity gel (Sigma); and rabbit anti-a-tubulin (Abcam).
(Liu et al., 2007; Nybakken et al., 2002; Ruel et al., 2007; Ruel A detailed procedure for the two-step immunoprecipitation assay is given in
et al., 2003). However, the function of Fu as a component in the Extended Experimental Procedures.
the Hh pathway is not consistent with its spatiotemporal expres-
S2 Cell Reporter Gene Assay
sion pattern during development. For example, Hh signaling only
The bam transcription reporter assay in S2 cells was performed by using the
plays a role in zebrafish embryonic development at late stages, bamP-luciferase construct in which the luciferase coding sequence was
but Fu is expressed ubiquitously at both the early and the late placed under the control of the bam promoter. For normalizing the efficiency
stages of zebrafish embryonic development. These findings of the transfection, the actinP-lacZ or actinP-Renilla construct was used.
suggest that Fu may have Hh-independent functions in different The luciferase and b-galactosidase assays were performed as standard
physiological conditions. In this study, by using several different procedures and measured on a luminometer.
systems, including Drosophila germline, zebrafish embryo, and
In Vivo and In Vitro Ubiquitination Assays
human tissue cultures, we demonstrated that Fu is indeed
For the in vivo ubiquitination assay, S2 cells were transfected with DNA
required for balancing proper BMP/TGFb signals in different constructs and also treated with dsRNA according to the protocols described
developmental processes. Given that both Fu and Smurf are previously (Chen et al., 2009). In brief, at 48 hr posttransfection, MG132 (final
evolutionarily conserved proteins, it would be interesting to concentration 50 mM) was added into the media. Cells were harvested 4 hr later

988 Cell 143, 978–990, December 10, 2010 ª2010 Elsevier Inc.
and lysed with a lysis buffer (50 mM Tris [pH 7.5], 120 mM NaCl, and 0.5% REFERENCES
NP40) containing 1% (w/v) sodium dodecyl sulfate (SDS) that was preheated
to 100 C. Before binding with the anti-Myc beads, the concentrations of NaCl Casanueva, M.O., and Ferguson, E.L. (2004). Germline stem cell number in the
and SDS in the binding buffer were adjusted to 500 mM and 0.1%, respec- Drosophila ovary is regulated by redundant mechanisms that control Dpp
tively. After pull-down with anti-Myc beads, the beads were then washed signaling. Development 131, 1881–1890.
with lysis buffer containing 0.1% SDS and were subjected to immunoblot
Chen, D., and McKearin, D. (2003a). Dpp signaling silences bam transcription
analysis.
directly to establish asymmetric divisions of germline stem cells. Curr. Biol. 13,
For the in vitro ubiquitination assay, Myc:TkvC protein was synthesized by
1786–1791.
the in vitro transcription-coupled translation method. To test whether the ubiq-
uitination of Tkv was coordinately supported by Smurf and Fu proteins, E1, E2 Chen, D., and McKearin, D.M. (2003b). A discrete transcriptional silencer in the
(His-UCH5C), E3 (Smurf complexes with Fu or without Fu), substrate bam gene determines asymmetric division of the Drosophila germline stem
(Myc:TkvC), and HA:Ub were then incubated at 30 C for 2 hr in a 40 ml ubiqui- cell. Development 130, 1159–1170.
tination reaction (50 mM Tris-HCl [pH 7.5], 1 mM dithiothreitol, 50 mM NaCl, 5 Chen, D., and McKearin, D. (2005). Gene circuitry controlling a stem cell niche.
mM MgCl2, and 2 mM ATP) with 0.2 mg of E1, 10 mg of ubiquitin (both from Curr. Biol. 15, 179–184.
Upstate). Reactions were terminated with SDS sample buffer and analyzed
Chen, D., Wang, Q., Huang, H., Xia, L., Jiang, X., Kan, L., Sun, Q., and Chen, D.
by western blotting with anti-Myc antibody.
(2009). Effete-mediated degradation of Cyclin A is essential for the mainte-
nance of germline stem cells in Drosophila. Development 136, 4133–4142.

Mammalian Cell Culture, Transient Transfection, and Claret, S., Sanial, M., and Plessis, A. (2007). Evidence for a novel feedback
Immunoprecipitation loop in the Hedgehog pathway involving Smoothened and Fused. Curr. Biol.
Human HEK293T and HepG2 cells were maintained in Dulbecco’s modified 17, 1326–1333.
Eagle’s medium (DMEM) supplemented with 10% fetal bovine serum (FBS) Cox, D.N., Chao, A., and Lin, H. (2000). piwi encodes a nucleoplasmic factor
at 37 C in a humidified incubator containing 5% CO2. Calcium phosphate or whose activity modulates the number and division rate of germline stem cells.
lipofectine was used for plasmid transfection. For the reporter assay, 36 hr Development 127, 503–514.
after transfection, cells were fed with fresh medium containing 0.2% FBS
Ebisawa, T., Fukuchi, M., Murakami, G., Chiba, T., Tanaka, K., Imamura, T.,
and were treated with 10 ng of ligands for another 12 hr. The luciferase and
and Miyazono, K. (2001). Smurf1 interacts with transforming growth factor-
Renilla assays were performed as standard procedures and measured on
beta type I receptor through Smad7 and induces receptor degradation.
a luminometer.
J. Biol. Chem. 276, 12477–12480.
Feng, X.H., and Derynck, R. (2005). Specificity and versatility in tgf-beta
signaling through Smads. Annu. Rev. Cell Dev. Biol. 21, 659–693.
Zebrafish Embryo Assay
All of the zebrafish embryos were derived from the Tübingen strain. Embryos Haley, B., Hendrix, D., Trang, V., and Levine, M. (2008). A simplified miRNA-
were incubated in Holtfreter’s solution at 28.5 C and staged. The mRNAs based gene silencing method for Drosophila melanogaster. Dev. Biol. 321,
were synthesized in vitro with the mMESSAGE mMACHINE Kit (Ambion). An 482–490.
RNeasy Mini Kit (QIAGEN) was used for mRNA purification. The fu-MO and Itoh, S., and ten Dijke, P. (2007). Negative regulation of TGF-beta receptor/
fu-cMO morpholinos have been described previously (Wolff et al., 2003) with Smad signal transduction. Curr. Opin. Cell Biol. 19, 176–184.
sequences of 50 -TGG TAC TGA TCC ATC TCC AGC GAC G-30 (fu-MO) and
50 -TGC TAG TGA TCG ATC TCC ACC GTC G-30 (fu-cMO). The fu-cMO was Jia, J., Tong, C., and Jiang, J. (2003). Smoothened transduces Hedgehog
a mismatch (italicized) control for fu-MO. The p53MO used to suppress signal by physically interacting with Costal2/Fused complex through its
nonspecific activation of morpholino oligonucleotides (Robu et al., 2007) C-terminal tail. Genes Dev. 17, 2709–2720.
was purchased from Gene Tools, LLC. The mRNA and morpholino were in- Jiang, J., and Hui, C.C. (2008). Hedgehog signaling in development and
jected into the yolk of the embryos at the one- or two-cell stage. Digoxige- cancer. Dev. Cell 15, 801–812.
nin-UTP-labeled antisense RNA probes were generated by in vitro transcrip-
Jiang, X., Xia, L., Chen, D., Yang, Y., Huang, H., Yang, L., Zhao, Q., Shen, L.,
tion. Whole-mount in situ hybridization was carried out following standard
Wang, J., and Chen, D. (2008). Otefin, a nuclear membrane protein, deter-
procedures.
mines the fate of germline stem cells in Drosophila via interaction with Smad
complexes. Dev. Cell 14, 494–506.
King, F.J., Szakmary, A., Cox, D.N., and Lin, H. (2001). Yb modulates the divi-
SUPPLEMENTAL INFORMATION
sions of both germline and somatic stem cells through piwi- and hh-mediated
mechanisms in the Drosophila ovary. Mol. Cell 7, 497–508.
Supplemental Information includes Extended Experimental Procedures and
six figures and can be found with this article online at doi:10.1016/j.cell. Kitisin, K., Saha, T., Blake, T., Golestaneh, N., Deng, M., Kim, C., Tang, Y.,
2010.11.022. Shetty, K., Mishra, B., and Mishra, L. (2007). Tgf-Beta signaling in develop-
ment. Sci. STKE 2007, cm1.
Li, L., and Xie, T. (2005). Stem cell niche: structure and function. Annu. Rev.
ACKNOWLEDGMENTS Cell Dev. Biol. 21, 605–631.
Liang, Y.Y., Lin, X., Liang, M., Brunicardi, F.C., ten Dijke, P., Chen, Z., Choi,
We thank Drs. Dennis McKearin, Duojia Pan, Peng Jin, and Zongping Xia for K.W., and Feng, X.H. (2003). dSmurf selectively degrades decapentaplegic-
critical readings of the manuscript. This work was supported by grants from activated MAD, and its overexpression disrupts imaginal disc development.
the National Basic Research Program of China (2007CB947502 and J. Biol. Chem. 278, 26307–26310.
2007CB507400 to D.C.) and from the NSFC (#30630042 and 30825026 to
D.C. and #30830068 to A.M.). Liu, Y., Cao, X., Jiang, J., and Jia, J. (2007). Fused-Costal2 protein complex
regulates Hedgehog-induced Smo phosphorylation and cell-surface accumu-
lation. Genes Dev. 21, 1949–1963.
Received: March 5, 2010
Revised: July 27, 2010 Murakami, G., Watabe, T., Takaoka, K., Miyazono, K., and Imamura, T. (2003).
Accepted: November 9, 2010 Cooperative inhibition of bone morphogenetic protein signaling by Smurf1 and
Published: December 9, 2010 inhibitory Smads. Mol. Biol. Cell 14, 2809–2817.

Cell 143, 978–990, December 10, 2010 ª2010 Elsevier Inc. 989
Narbonne-Reveau, K., Besse, F., Lamour-Isnard, C., Busson, D., and Pret, Song, X., Wong, M.D., Kawase, E., Xi, R., Ding, B.C., McCarthy, J.J., and Xie,
A.M. (2006). fused regulates germline cyst mitosis and differentiation during T. (2004). Bmp signals from niche cells directly repress transcription of a differ-
Drosophila oogenesis. Mech. Dev. 123, 197–209. entiation-promoting gene, bag of marbles, in germline stem cells in the
Nishikawa, S.I., Osawa, M., Yonetani, S., Torikai-Nishikawa, S., and Freter, R. Drosophila ovary. Development 131, 1353–1364.
(2008). Niche required for inducing quiescent stem cells. Cold Spring Harb. Spradling, A., Drummond-Barbosa, D., and Kai, T. (2001). Stem cells find their
Symp. Quant. Biol. 73, 67–71. niche. Nature 414, 98–104.
Nybakken, K.E., Turck, C.W., Robbins, D.J., and Bishop, J.M. (2002). Spradling, A.C., Nystul, T., Lighthouse, D., Morris, L., Fox, D., Cox, R., Tootle,
Hedgehog-stimulated phosphorylation of the kinesin-related protein Costal2 T., Frederick, R., and Skora, A. (2008). Stem cells and their niches: integrated
is mediated by the serine/threonine kinase fused. J. Biol. Chem. 277, units that maintain Drosophila tissues. Cold Spring Harb. Symp. Quant. Biol.
24638–24647. 73, 49–57.
Ohlstein, B., and McKearin, D. (1997). Ectopic expression of the Drosophila Szakmary, A., Cox, D.N., Wang, Z., and Lin, H. (2005). Regulatory relationship
Bam protein eliminates oogenic germline stem cells. Development 124, among piwi, pumilio, and bag-of-marbles in Drosophila germline stem cell self-
3651–3662. renewal and differentiation. Curr. Biol. 15, 171–178.
Ohlstein, B., Kai, T., Decotto, E., and Spradling, A. (2004). The stem cell niche: Van Doren, M., Williamson, A.L., and Lehmann, R. (1998). Regulation of zygotic
theme and variations. Curr. Opin. Cell Biol. 16, 693–699. gene expression in Drosophila primordial germ cells. Curr. Biol. 8, 243–246.
Podos, S.D., Hanson, K.K., Wang, Y.C., and Ferguson, E.L. (2001). The Wieser, R., Wrana, J.L., and Massagué, J. (1995). GS domain mutations that
DSmurf ubiquitin-protein ligase restricts BMP signaling spatially and tempo- constitutively activate T beta R-I, the downstream signaling component in
rally during Drosophila embryogenesis. Dev. Cell 1, 567–578. the TGF-beta receptor complex. EMBO J. 14, 2199–2208.
Robu, M.E., Larson, J.D., Nasevicius, A., Beiraghi, S., Brenner, C., Farber,
Wilson, C.W., Nguyen, C.T., Chen, M.H., Yang, J.H., Gacayan, R., Huang, J.,
S.A., and Ekker, S.C. (2007). p53 activation by knockdown technologies.
Chen, J.N., and Chuang, P.T. (2009). Fused has evolved divergent roles in
PLoS Genet. 25, e78.
vertebrate Hedgehog signalling and motile ciliogenesis. Nature 459, 98–102.
Ruel, L., Rodriguez, R., Gallet, A., Lavenant-Staccini, L., and Thérond, P.P.
Wolff, C., Roy, S., and Ingham, P.W. (2003). Multiple muscle cell identities
(2003). Stability and association of Smoothened, Costal2 and Fused with Cu-
induced by distinct levels and timing of hedgehog activity in the zebrafish
bitus interruptus are regulated by Hedgehog. Nat. Cell Biol. 5, 907–913.
embryo. Curr. Biol. 13, 1169–1181.
Ruel, L., Gallet, A., Raisin, S., Truchi, A., Staccini-Lavenant, L., Cervantes, A.,
Wrana, J.L., Attisano, L., Wieser, R., Ventura, F., and Massagué, J. (1994).
and Thérond, P.P. (2007). Phosphorylation of the atypical kinesin Costal2 by
Mechanism of activation of the TGF-beta receptor. Nature 370, 341–347.
the kinase Fused induces the partial disassembly of the Smoothened-
Fused-Costal2-Cubitus interruptus complex in Hedgehog signalling. Develop- Wu, M.Y., and Hill, C.S. (2009). Tgf-beta superfamily signaling in embryonic
ment 134, 3677–3689. development and homeostasis. Dev. Cell 16, 329–343.
Sánchez-Herrero, E., Couso, J.P., Capdevila, J., and Guerrero, I. (1996). The fu Xie, T., and Spradling, A.C. (1998). decapentaplegic is essential for the main-
gene discriminates between pathways to control dpp expression in Drosophila tenance and division of germline stem cells in the Drosophila ovary. Cell 94,
imaginal discs. Mech. Dev. 55, 159–170. 251–260.
Schier, A.F., and Talbot, W.S. (2005). Molecular genetics of axis formation in Yamashita, Y.M., Fuller, M.T., and Jones, D.L. (2005). Signaling in stem cell
zebrafish. Annu. Rev. Genet. 39, 561–613. niches: lessons from the Drosophila germline. J. Cell Sci. 118, 665–672.

990 Cell 143, 978–990, December 10, 2010 ª2010 Elsevier Inc.
Functional Overlap and Regulatory Links
Shape Genetic Interactions
between Signaling Pathways
Sake van Wageningen,1,5 Patrick Kemmeren,1,5 Philip Lijnzaad,1,4 Thanasis Margaritis,1 Joris J. Benschop,1
Inês J. de Castro,1 Dik van Leenen,1 Marian J.A. Groot Koerkamp,1 Cheuk W. Ko,1 Antony J. Miles,1 Nathalie Brabers,1
Mariel O. Brok,1 Tineke L. Lenstra,1 Dorothea Fiedler,2 Like Fokkens,3 Rodrigo Aldecoa,1 Eva Apweiler,1
Virginia Taliadouros,1 Katrin Sameith,1 Loes A.L. van de Pasch,1 Sander R. van Hooff,1 Linda V. Bakker,1,4
Nevan J. Krogan,2 Berend Snel,3 and Frank C.P. Holstege1,*
1Molecular Cancer Research, University Medical Centre Utrecht, Universiteitsweg 100, Utrecht, The Netherlands
2Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA
3Theoretical Biology and Bioinformatics, Department of Biology, Science Faculty, Utrecht University, Padualaan 8,

3584 CH Utrecht, The Netherlands


4Netherlands Bioinformatics Centre, Geert Grooteplein 28, 6525 GA, Nijmegen, The Netherlands
5These authors contributed equally to this work

*Correspondence: f.c.p.holstege@umcutrecht.nl
DOI 10.1016/j.cell.2010.11.021

SUMMARY in depth knowledge about the ways in which different pathways


work together.
To understand relationships between phosphoryla- Due to the extensive role of signaling, perturbation of different
tion-based signaling pathways, we analyzed 150 pathways leads to diverse phenotypes. Different pathways have
deletion mutants of protein kinases and phospha- therefore often been studied in isolation, frequently using
tases in S. cerevisiae using DNA microarrays. Down- different readouts for different pathways and thereby confound-
stream changes in gene expression were treated as ing systematic comparisons of pathways. This can be overcome
by using a single assay that is detailed enough to reveal differ-
a phenotypic readout. Double mutants with synthetic
ences and at the same time comprehensive enough to reveal
genetic interactions were included to investigate
the workings of many different pathways simultaneously. Pheno-
genetic buffering relationships such as redundancy. types are often accompanied by changes in gene expression
Three types of genetic buffering relationships are and genome-wide mRNA expression profiling can reveal rela-
identified: mixed epistasis, complete redundancy, tionships between pathway components (Capaldi et al., 2008;
and quantitative redundancy. In mixed epistasis, Roberts et al., 2000). Here, we have applied expression profile
the most common buffering relationship, different phenotypes to systematically investigate relationships between
gene sets respond in different epistatic ways. Mixed many different signaling pathways that are simultaneously active
epistasis arises from pairs of regulators that have under a single growth condition in the yeast Saccharomyces
only partial overlap in function and that are coupled cerevisiae.
by additional regulatory links such as repression of Analysis of pathway activity using mutants also requires buff-
ering interactions between genes to be considered. Genetic
one by the other. Such regulatory modules confer
buffering results in masking of the phenotypic consequences
the ability to control different combinations of pro-
of mutations (Hartman et al., 2001). The best appreciated buff-
cesses depending on condition or context. These ering relationship is redundancy, often defined as genes that
properties likely contribute to the evolutionary main- can compensate for each other’s loss by their ability to share
tenance of paralogs and indicate a way in which and takeover the exact same function. Redundancy is frequently
signaling pathways connect for multiprocess control. associated with paralogs that are more likely to share an identical
biochemical function (Prince and Pickett, 2002). Nonhomolo-
INTRODUCTION gous genes are less likely to share function but can still exhibit
genetic buffering in the form of growth-rate compensation. The
Protein kinases and protein phosphatases are key components relative contribution of paralogs versus nonhomologs toward
of regulatory pathways, many of which have been studied in buffering is under debate (Gu et al., 2003; Ihmels et al., 2007;
detail. This has revealed the pleiotropic role of signaling in Papp et al., 2004; Wagner, 2000), but systematic analysis of
cellular regulation, its involvement in disease and how pathway synthetic genetic interactions (SGIs) is revealing extensive
architecture underlies mechanistic aspects such as specificity. buffering between nonhomologs (Costanzo et al., 2010). How
Understanding the complexity of cellular regulation also requires nonhomologous pairs compensate for loss of each other’s

Cell 143, 991–1004, December 10, 2010 ª2010 Elsevier Inc. 991
function is not well understood and the molecular mechanisms Individual mutants vary considerably with regard to the extent
behind such genetic relationships are relatively uncharacterized. of gene expression changes (Figures 1A and 1B). None of the WT
Also enigmatic is the question of why paralogs are stably profiles exhibit more than eight genes changing significantly.
maintained during evolution, often remaining redundant, despite Applying this threshold on the mutants indicates that 71% of
evolutionary pressure against seemingly superfluous copies the kinase deletions behave like WT under this growth condition
(Dean et al., 2008; Vavouri et al., 2008). Resolving these (Figure 1A). For phosphatase deletions this number is even
questions likely requires more detailed characterization of the higher (85%, Figure 1B). Taking into account essential genes,
mechanisms that underlie buffering interactions, including this means that more than 60% of kinase/phosphatase genes
redundancy. can be individually removed under a single growth condition
The yeast Saccharomyces cerevisiae has 141 genes encoding without defects in growth or in gene expression. Analysis of
protein kinases and 38 genes encoding protein phosphatases. mutants with profiles that differ from WT indicates that lack of
Here, kinase and phosphatase function is systematically com- sensitivity is not the cause of apparent inactivity. For example,
pared by generating DNA microarray expression profiles for all mutations in the kinase cascades that control mating and osmo-
150 viable protein kinase and phosphatase knockout strains regulation result in significant changes in mRNA expression,
under a single growth condition. To take buffering interactions related according to the pathways (Figure 1C). This reflects linear
into account, SGI data is exploited by profiling double mutants relationships between components of kinase cascades and indi-
that show greater than expected fitness reduction (Fiedler cates that the approach is sensitive enough to analyze pathways
et al., 2009). This provides a detailed and systematic character- active even at uninduced basal levels (see Figure S1, available
ization of different genetic buffering relationships. The molecular online, for all mutant profiles that differ from WT).
mechanisms of each type are studied in detail, including analysis
of a phosphatase that buffers kinase deletions. An important Profiling Negative Synthetic Genetic Interactions
outcome is identification of a recurrent regulatory module for For many mutants, similarity to WT is likely due to absence
signaling pathways. This module consists of pairs of regulators or inactivity of the protein under a single growth condition. The
that have partial overlap in function and that are also linked by goal of comparing many pathways active under a single
additional regulatory relationships such as repression or inhibi- condition also requires genetic buffering interactions such as
tion of one partner by the other. The module offers insight into redundancy to be considered, since this may mask activity of
how signaling pathways may regulate different combinations of components whereby deletion has no effect. To include redun-
processes in a flexible yet coordinate manner and plausibly dancy relationships that influence fitness, we exploited SGI
explains why apparently redundant components of regulatory data for kinase/phosphatase genes (Fiedler et al., 2009). Selec-
pathways are maintained during evolution. tion was based on a greater than expected growth defect in a
double mutant compared to the singles. An additional criterion
was applied that consisted of one of the single mutants not
RESULTS showing an expression profile different from WT, resulting in
24 pairs. These double mutants were first remade in the genetic
Expression Profiles of Kinase and Phosphatase Gene background used here and the SGIs were retested for the liquid
Deletions culture growth used for expression profiling. Despite differences
To compare signaling pathways, DNA microarray gene expres- with colony growth (Fiedler et al., 2009), correspondence
sion profiles were generated for all 150 viable protein kinase/ between the previous study is strong, with 20 of the 24 pairs
phosphatase deletions in S. cerevisiae under a single growth also showing a greater than expected growth defect in liquid
condition (synthetic complete medium with 2% glucose). Each culture (Table S1). Two previously established redundant pairs
mutant was profiled four times, from two independent cultures (FUS3-KSS1, YPK1-YPK2) were added to the selection, and all
on dual-channel microarrays using a batch of wild-type (WT) viable double mutants were expression profiled.
RNA as common reference. To further control for technical and Genetically buffered gene pairs, such as redundant partners,
biological variation, additional WT cultures were grown along- were expected to show more gene expression changes as
side sets of mutants on each day. These ‘‘same-day’’ WTs a double mutant compared to the two singles combined. Dele-
were processed in parallel to the mutants, all using automated, tion of the kinase ARK1, shows an expression profile similar to
robotic procedures. Comparison of the many WT profiles yields WT (Figure 2A). Similarly, prk1D also has few genes changing
insight into the expression variation of each gene. Statistical significantly (Figure 2B). The ark1D prk1D double mutant has
modeling results in an average profile for each mutant, consist- many genes with expression deviating significantly from WT
ing of p values and changes in mRNA expression for each (Figure 2C) and the profile therefore concurs with the previously
gene, relative to the expression in the 200 WT cultures (Experi- reported redundancy (Cope et al., 1999). Likewise, the profile of
mental Procedures). Throughout the manuscript ‘‘significant’’ the phosphatase double mutant ptp2D ptp3D also agrees with
indicates statistically significant. A p value of 0.05, in combina- redundancy (Figures 2D–2F) (Jacoby et al., 1997; Wurgler-
tion with a fold change (FC) of 1.7, is applied as a threshold for Murphy et al., 1997). Figure S2 depicts all scatter plots indicative
calling a change in mRNA expression significant. Aneuploidy, of a buffering effect. Systematic analysis (Extended Experi-
incorrect deletions, and spurious mutations were identified in mental Procedures) shows that of the pairs successfully
11% of the mutant strains (Experimental Procedures). These analyzed, 21 have expression profiles that support buffering
strains were remade and reprofiled. (Table 1), with more genes changing expression in the double

992 Cell 143, 991–1004, December 10, 2010 ª2010 Elsevier Inc.
A B

4
4

2
2
M (log2(mt/wt))

M (log2(mt/wt))
0

0
−2

−2
−4

−4
ptc1Δ
sit4Δ
yvh1Δ
oca1Δ
siw14Δ
msg5Δ
pph3Δ
mih1Δ
ptc3Δ
ptc4Δ
ptp3Δ
oca2Δ
ppg1Δ
ppt1Δ
ppz1Δ
psr1Δ
ppq1Δ
ptc7Δ
nem1Δ
psr2Δ
ptc2Δ
cna1Δ
ltp1Δ
pph21Δ
pph22Δ
ppz2Δ
ptc5Δ
ych1Δ
cmp2Δ
pps1Δ
ptp1Δ
ptp2Δ
sdp1Δ
ctk1Δ
ssn3Δ
vps15Δ
ypk1Δ
pho85Δ
cka2Δ
fus3Δ
mck1Δ
ste7Δ
ste11Δ
elm1Δ
dun1Δ
kin3Δ
fab1Δ
pbs2Δ
ste20Δ
hog1Δ
ssk2Δ
yck3Δ
sky1Δ
snf1Δ
ire1Δ
ksp1Δ
tpk2Δ
cla4Δ
ptk2Δ
rim15Δ
chk1Δ
cka1Δ
cmk2Δ
bck1Δ
rim11Δ
sat4Δ
ssk22Δ
tpk3Δ
cmk1Δ
lcb5Δ
slt2Δ
tel1Δ
ygk3Δ
kkq8Δ
kss1Δ
npr1Δ
tor1Δ
fpk1Δ
ark1Δ
fmp48Δ
kin82Δ
prr2Δ
ptk1Δ
skm1Δ
ybr028cΔ
ykl161cΔ
ypl141cΔ
ypl150wΔ
isr1Δ
abc1Δ
hal5Δ
hsl1Δ
kin1Δ
kns1Δ
mkk2Δ
prr1Δ
psk1Δ
swe1Δ
tpk1Δ
vhs1Δ
yak1Δ
yck1Δ
ypk2Δ
atg1Δ
cki1Δ
gcn2Δ
hrk1Δ
iks1Δ
mek1Δ
mrk1Δ
pkh1Δ
prk1Δ
scy1Δ
sks1Δ
sps1Δ
twf1Δ
ykl171wΔ
akl1Δ
alk1Δ
alk2Δ
dbf20Δ
eki1Δ
gin4Δ
ime2Δ
kcc4Δ
kin2Δ
kin4Δ
lcb4Δ
mkk1Δ
pkh3Δ
psk2Δ
rck1Δ
rck2Δ
sak1Δ
smk1Δ
tos3Δ
yck2Δ
ydl025cΔ
pkp2Δ
pkp1Δ
ylr253wΔ
tda1Δ
ypl109cΔ
env7Δ
C
MF(ALPHA)1

MF(ALPHA)2

MATALPHA1
YMR173W-A

YGR109W-B
YGR109W-A
YDR261W-B
YER138W-A

YPR158W-A
YMR158C-A

YGR161C-D
YCL021W-A
YDR210C-D

YDR261C-D
YDR381C-A
YDR379C-A

YDR098C-B
YPR158C-D

YBL107W-A
YJL127W-A

YHR214W
YMR046C
YLR400W
YCR102C

YCR013C

YAR009C
YLR460C

YDL228C

YLR042C

YLR040C

YDL158C
YLL053C
YJL107C

YIL080W
YIL169C
PRM10

COS12

PHO12

DDR48
FMP48

SNR10

RNA14

STE20
STE11
STE12
CWP1

HOG1
PGM2
HOR2

PHM6

PRM5

PRM6
GRE2

RHR2

GPH1

MSB2
CHA1
YGP1

NCA3
BDH2

AQY2

GSY2

GPA1

SAG1

SRD1

AGA1

DAD4
PNS1
SED1

PYC1

YHB1

PRY2

CTR3

KAR4

SSK2
PBS2
KSS1
FUS3
HXT8
HXT1

HPF1

VTC1
VTC4

FAR1

TEC1

FUS1

FRE7

STE7
CTT1

ZRT1

ALD4

SST2
STE3

NDJ1
STF2

LSP1

FIG1
SPI1

TIP1
FIT3

1 ste20Δ
2 ste11Δ
3 ste7Δ
4 fus3Δ kss1Δ

5 ssk2Δ
6 pbs2Δ
7 hog1Δ

-2.5 0 2.5

Fold change

Figure 1. Expression Profiles of Kinase/Phosphatase Single Gene Deletions


(A and B) Activity profiles of all deletion strains, ranked as box-whisker plots for kinases (A) and phosphatases (B), showing fold changes (vertical axis), with
significantly changing genes (p < 0.05, FC > 1.7) as red dots and unresponsive genes as black dots. Green triangles indicate the doubling time of each mutant
(-log2 relative to WT). Dashed gray lines indicate 1.7-fold change. The solid gray line is the threshold for distinguishing deletions with significant profiles (R8 genes
changing) versus deletions that behave similarly to WT (<8 genes changing). This threshold is based on the maximum number of changes observed in the 200 WT
profiles, excluding the WT variable genes (Experimental Procedures).
(C) Lanes 1–7 are expression profiles of strains indicated to the right. All genes with significantly changed expression in any single mutant (p < 0.05, FC > 1.7) are
depicted, with gene names on top. STE20, STE11, STE7 and FUS3 are the MAPK components of the mating pheromone response pathway. FUS3 is redundant
with KSS1 and the profile of the double mutant is therefore shown in lane 4. Profiles of the single mutants are depicted in Figure 4C. SSK2, PBS2 and HOG1 are
MAPK components of the HOG pathway. The opposite effects of the HOG pathway on some of the genes affected by the mating pathway agrees with inhibition of
the mating pathway by the HOG pathway (Chen and Thorner, 2007).
See also Figure S1.

mutant versus the two single mutants combined. This includes Buffering between a Kinase and Phosphatase Is due
all the pairs that showed a negative SGI in liquid culture to Phosphatase-Mediated Inhibitory Crosstalk
(Table S1). between Kinase Pathways
Redundancy involves overlap of function and is often associ- Bck1 and Slt2 are mitogen-activated protein kinase (MAPK)
ated with paralogs. Phylogenetic analysis reveals that less than components of the cell-wall integrity (CWI) pathway (Chen and
one third of the buffering relationships observed here are derived Thorner, 2007). Both kinases show buffering with the phospha-
from close paralogs, that is from duplication events that tase PTP3, likely reflecting the fact that both kinases belong to
occurred less than approximately 600 million years ago (Table 1, the same MAPK cascade (Figures 2G–2K). In both kinase-phos-
Figure S3, Extended Experimental Procedures). More than half phatase double mutants the same genes change (Figure 2L).
of the interactions are between pairs that arose from ancient Ptp3 dephosphorylates Hog1, resulting in inactivation of Hog1
duplications (an estimated 2 billion years ago) or between non- (Jacoby et al., 1997). Most of the bck1D ptp3D and slt2D
homologs, in five cases even between kinase-phosphatase ptp3D double deletion profiles consist of upregulated genes
pairs. Buffering between nonhomologs has been noted before (Figures 2J and 2K). This includes established Hog1 downstream
(Gu et al., 2003; Ihmels et al., 2007; Papp et al., 2004; Wagner, target genes (Rodrı́guez-Peña et al., 2005), indicating that buff-
2000), but the underlying mechanisms are often not investigated. ering may be related to defective inhibition of Hog1. To test
Therefore, we selected an example for further analysis, focusing this, the double deletion strains were first assayed for pheno-
on the intriguing buffering between kinases and phosphatases. types associated with increased Hog1 activity such as elevated

Cell 143, 991–1004, December 10, 2010 ª2010 Elsevier Inc. 993
Figure 2. Expression Profiles of Genetically Buffered Pairs
(A–K) Single and double deletion gene expression scatter plots of four genetically buffered pairs. In each scatter plot the normalized, dye-bias corrected and
statistically modeled fluorescent intensity value is plotted for each gene. For each mutant this is the average of four measurements. For WT this is the average
of 200 cultures grown throughout the project. Genes with significant increase or decrease in mRNA expression (p < 0.05, FC > 1.7) are represented by yellow and
blue dots respectively. Gray dots are all other genes.
(L) Scatter plot of all genes that have a significant change in mRNA expression in either bck1D ptp3D (J), slt2D ptp3D (K) or in both double mutants. The log2 FC is
plotted for each of these genes in both double deletions, showing that the same mRNAs are changing in both strains.
See also Figure S2.

994 Cell 143, 991–1004, December 10, 2010 ª2010 Elsevier Inc.
Table 1. Buffering Relationships between Kinases and Phosphatases
Gene 1 Gene 2 Type Duplication Time (Years Ago) Buffering Relationship
HAL5 SAT4 kk old 600 M – 2 G complete redundancy
ARK1 PRK1 kk whole genome 125 M complete redundancy
PTP2 PTP3 pp recent 125 M – 600 M complete redundancy
YCK1 YCK2 kk whole genome 125 M complete redundancya
PTC1 PTC2 pp old 600 M – 2 G quantitative redundancy
PTC1 PPH3 pp not homologous quantitative redundancy
PBS2 PTK2 kk ancient >2G mixed epistasis
CLA4 SLT2 kk ancient >2G mixed epistasis
CLA4 HSL1 kk ancient >2G mixed epistasis
SNF1 RIM11 kk ancient >2G mixed epistasis
BCK1 PTP3 kp not homologous mixed epistasis
SLT2 PTP3 kp not homologous mixed epistasis
FUS3b KSS1 kk recent 125 M – 600 M mixed epistasis
ELM1 MIH1 kp not homologous mixed epistasisc
CLA4 BCK1 kk ancient >2G mixed epistasisc
DUN1 PPH3 kp not homologous mixed epistasisc
CKA2 CKA1 kk recent 125 M – 600 M not classifieda
b
YPK1 YPK2 kk whole genome 125 M not classifieda
PTK1 PTK2 kk whole genome 125 M not classifieda
HSL1 MIH1 kp not homologous not classifieda
SKY1 PTK2 kk ancient >2G not classifieda
a
Double mutant is inviable, confirming a buffering effect.
b
Included based on previously reported redundancy.
c
Double mutant was aneuploid; aneuploid chromosomes were excluded from analysis.
Determination of paralogy relative to important radiations and events was performed by integration of information available in several orthology and
homology databases. The timings in years are estimates derived from literature (Extended Experimental Procedures).
k, kinase; p, phosphatase. See also Table S1 and Figure S3.

temperature (Figure 3A) (Winkler et al., 2002) and sensitivity to (Kelley and Ideker, 2005). In this case the parallel pathways
the cell wall disrupting agent zymolyase (Figure 3B) (Bermejo converge on Hog1 through two redundant phosphatases, one
et al., 2008). That the buffering observed between the BCK1, of which, Ptp2, is likely activated by the CWI pathway.
SLT2 kinases and PTP3 phosphatase indeed involves Hog1 is
confirmed by monitoring Hog1 phosphorylation, which is higher Expression Profiling Reveals Three Different Genetic
in both bck1D ptp3D and slt2D ptp3D double mutants compared Buffering Relationships
to ptp3D or WT (Figure 3C). Division into paralogous and nonhomologous pairs is one type of
Since it is unlikely that the kinases are directly responsible for classification that can be applied to genetic buffering. The data
dephosphorylation of Hog1, a second phosphatase was postu- also prompted a new characterization of genetic buffering rela-
lated to be involved. Candidates included Ptc1, Ptp2, and tionships, based on the single- and double mutant expression
Ptc2, all also capable of dephosphorylating Hog1 (Jacoby profiles. Intriguingly, these can be classified into three types:
et al., 1997; Warmka et al., 2001; Wurgler-Murphy et al., 1997; complete redundancy, quantitative redundancy and mixed epi-
Young et al., 2002). PTP3-phosphatase double mutant expres- stasis (Figure 4, systematic classification is described in detail
sion profiles were analyzed. Only the ptp2D ptp3D double in Extended Experimental Procedures). Complete redundancy
mutant expression profile shows a buffering effect whereby the is exemplified by the ark1D, prk1D scatter plots (Figures 2A–
majority of mRNAs that change in the CWI kinase-phosphatase 2C). There are no changes in single deletions (less than eight
double mutants are also similarly changing in the ptp2D ptp3D genes changing significantly compared to WT), but an effect is
double phosphatase mutant (Figure 3D). In addition, Hog1 phos- observed in the double mutant. Four redundant pairs show
phorylation levels are increased in the ptp3D ptp2D double complete redundancy (Figure 4A). Besides ARK1-PRK1, this
mutant (Figure 3E). Buffering between the CWI pathway kinases includes the kinase pairs HAL5-SAT4, YCK1-YCK2 and the
and the PTP3 phosphatase is therefore likely reflecting redun- phosphatase pair PTP2-PTP3.
dancy between PTP2 and PTP3 (Figure 3F) (Jacoby et al., A second type of redundancy is evident from the quantitative
1997; Wurgler-Murphy et al., 1997). This agrees with the infre- effects observed in the phosphatase pairs PTC2-PTC1 and
quently tested notion that SGIs arise from parallel pathways PPH3-PTC1 (Figure 4B). Here, one single mutant shows no

Cell 143, 991–1004, December 10, 2010 ª2010 Elsevier Inc. 995
A D

pt Δ

pt Δ
Δ

Δ
3

3
t2 ptp
p3

t2 ptp
p3
pt Δ

pt Δ

Δ
Δ

Δ
1 ptp2Δ

Δ
k1

k1

k1

k1
p3

p3
t2

t2
bc

bc

bc

bc
t

t
2 bck1Δ
sl

sl

sl

sl
w

w
3 slt2Δ
4 ptp3Δ
5 bck1Δ ptp3Δ
6 slt2Δ ptp3Δ
7 ptp2Δ ptp3Δ
30 oC 37 oC

B E
12


ptp


10

ptp

ptp

ptp
wt
wt
8 ptp3Δ Hog1- p
slt2Δ
OD600

6 bck1Δ Hog1
ptp3Δ slt2Δ
4 ptp3Δ bck1Δ Tubulin
2
F
0 Bck1
p
0 0.01 0.025 0.1
zymolyase units/ml
Mkk1/2 Mkk1/2
C p
Cl
Na

Slt2 Slt2

p
ptp
.4M

ptp
Δ

Δ
+0


k1

k1
ptp

bc

bc
slt

slt
wt

wt

wt

Hog1- p
Ptp2 Ptp3
Hog1

Tubulin Hog1 Hog1


p

Figure 3. Kinase-Phosphatase Buffering Is Caused by Phosphatase-Mediated Inhibitory Crosstalk between Kinase Pathways
(A) The bck1D ptp3D and slt2D ptp3 kinase-phosphatase double mutants are sensitive to elevated temperature. Ten-fold dilutions of cultures were spotted
onto plate and incubated at 30 C or 37 C.
(B) The bck1D ptp3D and slt2D ptp3 kinase-phosphatase double mutants show more sensitivity to zymolyase. Bars and standard deviations are based on the
average of three.
(C) Active, phosphorylated Hog1 is increased in the bck1D ptp3D and slt2D ptp3 kinase-phosphatase double mutants. Immunoblots for phosphorylated Hog1
(top), all Hog1 (middle) and Tubulin (bottom). Lane 1 is a positive control of WT exposed to 0.4 M NaCl for five minutes prior to harvesting.
(D) All genes with significant changes in bck1D ptp3D or slt2D ptp3D (p < 0.05, FC > 1.7) are depicted. Lane 7 shows the same genes for the ptp2D ptp3D
expression profile.
(E) As in (C).
(F) Model of interactions for the buffering observed between PTP3-SLT2 and PTP3-BCK1. Gray lines indicate buffering. Black line indicates redundancy.
The two arrows between Slt2 and Ptp2 indicate that this activation may be direct or indirect.

effect (less than eight gene changes), but the other single mutant Changing thresholds would result in a different classification
does. The term quantitative is applicable because the effect for some of the pairs. The thresholds were kept identical to those
observed in the single mutant is amplified in the double mutant used for identification of which mutants behave as WT (Figure 1).
(see also Figure 4E) but without involving additional gene sets. In this way sixteen of the twenty-one gene pairs exhibiting
Complete and quantitative redundancy are intuitive in their genetic buffering are classified: four as complete, two as quan-
classification and as is demonstrated below, both can be under- titative and ten as mixed epistatic. In six cases, the double
stood through simple molecular mechanisms. This is not true mutant is inviable (Table 1), hindering classification of CKA1-
because of the third buffering relationship, which we call mixed CKA2, PTK2-PTK1, PTK2-SKY1, HSL1-MIH1, and YPK1-YPK2
epistasis for the different types of epistatic effects observed on (Figure 4D). One case of inviability (YCK1-YCK2) can be unam-
different gene sets (Figure 4C). Whereas some gene sets biguously classified as complete redundancy (Figure 4A).
respond as in complete or quantitative redundancy, other gene The ten pairs showing mixed epistasis are the kinase pairs
sets behave in completely different ways. These typically show KSS1-FUS3, HSL1-CLA4, SNF1-RIM11, BCK1-CLA4, SLT2-
expression changes in single mutants that disappear or even CLA4 and the kinase-phosphatase pairs PBS2-PTK2, ELM1-
show an opposite effect in the double mutant. The classification MIH1, DUN1-PPH3, BCK1-PTP3, SLT2-PTP3. Mixed epistasis
scheme (Extended Experimental Procedures) depends on is therefore exhibited by paralogous as well as nonhomologous
thresholds for identification of differently behaving gene sets. pairs. Besides the mixed epistasis itself, it is striking that this

996 Cell 143, 991–1004, December 10, 2010 ª2010 Elsevier Inc.
buffering interaction is the most common. Redundancy is not the three strains (Figure 4C). To understand mixed epistasis,
necessarily complete. Partial overlap in function is expected to we focused on two such gene sets. The first set behaves as in
result in single mutants exhibiting effects on their own, with these complete redundancy, with downregulation only in the double
same effects reflected in the double mutant, alongside additional mutant (Figure 5A). The second set shows upregulation in
genes changing due to loss of the shared function. It is remark- fus3D only. Together, these two gene sets form a minimal mixed
able that no very clear example of this expected partial redun- epistasis pattern, shared by the majority of pairs classified as
dancy pattern is observed. As is made clear below, this is related such (Figure 4C).
to the finding of mixed epistasis. A model that explains the different epistatic behavior of the
two responder gene sets (Figures 5B and 5C) is based on data
Mechanisms Underlying Complete and Quantitative presented here (Figure 5A) as well as on many previous studies
Redundancy of these pathways (Chen and Thorner, 2007). FUS3 and KSS1
We next considered molecular mechanisms. Complete and are redundant paralogs but the redundancy is only partial (Elion
quantitative redundancy can be explained by similar models et al., 1991). The two pathways work through two downstream
whereby redundant partners function on the same targets (Fig- transcription factors, Ste12 and Tec1 (Chen and Thorner,
ures 4F and 4H). As an example, Ark1 and Prk1 are previously es- 2007; Chou et al., 2006; Madhani and Fink, 1997). The promoters
tablished redundant kinases that regulate endocytosis and the of the two gene sets are differentially enriched for Ste12 and
actin cytoskeleton (Smythe and Ayscough, 2003). ARK1-PRK1 Tec1 binding sites (Figure 5A). The first gene set consists of
demonstrate complete redundancy (Figures 2A–2C). The endo- mating genes, enriched for pheromone response elements
cytic adaptor protein Sla1 is an established direct target of that bind homodimerized Ste12. The second gene set is en-
both kinases (Zeng et al., 2001). The sla1D expression profile riched for the filamentation response element that binds the
reflects this, with the changes in mRNA expression forming Ste12-Tec1 heterodimer. In agreement with previous studies
a perfect subset of the ark1D prk1D expression profile (Fig- (Chen and Thorner, 2007), Kss1 is inactive under noninducing
ure 4G). This illustrates that kinase targets can in some cases conditions and kss1D has virtually no effect (Figure 5A). The
be identified by comparative expression profiling and indicates mating pathway (Fus3) is active at low basal levels under nonin-
here that Ark1 and Prk1 likely have more than one target. ducing conditions. Fus3 is an activating kinase for Ste12 and an
It is similarly intuitive that pairs showing quantitative redun- inactivating kinase for Tec1, whereby Tec1 phosphorylation
dancy have identical targets, since the same genes are affected leads to its degradation (Chen and Thorner, 2007; Chou et al.,
in single and double mutants, but to different degrees (Figures 2004). KSS1 is a target of Tec1 in this model. Upon deletion of
4B and 4E). Quantitative redundancy may reflect a quantitatively FUS3, Tec1 is no longer degraded. KSS1 becomes upregulated
different effect on the target. To test this, we investigated the and because of their redundancy, Kss1 can (partially) take over
phosphatase pair PTC1-PTC2 (Figure 4B). Hog1 is a shared the role of Fus3 (Figure 5C). Kss1 takes over the role of activating
target of Ptc1 and Ptc2 (Young et al., 2002). In agreement with Ste12 (Madhani et al., 1997). No change is therefore observed in
the hypothesis, the degree to which Ptc1 and Ptc2 dephosphor- the mating genes, which remain active at basal levels (Figure 5A).
ylate Hog1 differs (Figure 4I). Levels of phosphorylated Hog1 in Kss1 does not take over the inactivating role of Fus3 toward Tec1
the different mutants match the quantitative effects observed (Chou et al., 2004), leading to activation of the filamentous
in the expression profiles (Figure 4B). This supports the proposal gene cluster in fus3D (Figure 5A). This effect is lost in the double
that quantitative redundancy is caused by identical target mutant and the filamentous gene set reverts back to WT levels
specificity combined with a quantitatively different effect on the (Figure 5A). The mating gene set is down in the double mutant
target. This could be due to differences in enzyme efficiency or (Figure 5A) because neither Kss1 nor Fus3 are present to activate
through differences in expression levels of redundant partners. Ste12.
Due to the selection criteria, the effects observed here always The two pivotal elements that explain the mixed epistatic
involve one single mutant showing an expression profile similar effects are therefore partial redundancy and the negative regula-
to WT. This implies that the enzyme that does show a single- tion of KSS1 by Fus3. A negative effect of Fus3 on KSS1 has
deletion phenotype is overabundantly active under this growth been described for activating conditions (Chou et al., 2006).
condition. The promoter of KSS1 contains binding sites for Tec1 (Figure 5A)
and, as predicted, KSS1 indeed becomes upregulated in fus3D
Mixed Epistasis of FUS3-KSS1 Is a Result of Partial (Figure 5A). The involvement of the two downstream transcrip-
Redundancy Coupled to Unidirectional Repression tion factors (Chen and Thorner, 2007) is supported by the differ-
Mixed epistasis is the most frequently observed buffering inter- ential enrichment of binding sites (Figure 5A) and was tested by
action (Figure 4C, Table 1). To investigate mechanism, we first analyzing tec1D and ste12D (Figure S4).
focused on the FUS3-KSS1 kinase pair (reviewed in Chen and
Thorner, 2007). The Fus3 MAPK is responsible for activation of Boolean Modeling Reveals Two General Properties
mating genes in response to pheromone. Kss1 is the MAPK of of Mixed Epistasis: Partial Overlap in Function
the filamentous growth pathway that activates a nutrient starva- and Regulatory Coupling
tion response whereby yeast cells change polarity and shape, Mixed epistasis similar to FUS3-KSS1 occurs in 10 out of the 16
resulting in filamentous colony outgrowth that enables foraging pairs that can be classified (Figure 4C). To determine whether
for nutrients. The fus3D, kss1D and fus3D kss1D profiles consist similar mechanisms underlie all such cases, we asked which
of several responder gene sets that behave in different ways in regulatory network topologies lead to such phenotypes. By

Cell 143, 991–1004, December 10, 2010 ª2010 Elsevier Inc. 997
A
ark1Δ
prk1Δ
ark1Δ prk1Δ
ptp2Δ hal5Δ yck1Δ
ptp3Δ sat4Δ yck2Δ
ptp2Δ ptp3Δ hal5Δ sat4Δ inviable yck1Δ yck2Δ

B
ptc2Δ
ptc1Δ
ptc1Δ ptc2Δ
pph3Δ
ptc1Δ
ptc1Δ pph3Δ

C
fus3Δ dun1Δ hsl1Δ
kss1Δ pph3Δ cla4Δ
fus3Δ kss1Δ dun1Δ pph3Δ* hsl1Δ cla4Δ
bck1Δ bck1Δ mih1Δ
cla4Δ ptp3Δ elm1Δ
bck1Δ cla4Δ* bck1Δ ptp3Δ *
mih1Δ elm1Δ

slt2Δ ptk2Δ rim11Δ slt2Δ


cla4Δ pbs2Δ snf1Δ ptp3Δ
slt2Δ cla4Δ pbs2Δ ptk2Δ snf1Δ rim11Δ slt2Δ ptp3Δ

D
cka1Δ ptk2Δ ptk2Δ hsl1Δ ypk2Δ
cka2Δ ptk1Δ sky1Δ mih1Δ ypk1Δ
inviable cka1Δ cka2Δ inviable ptk1Δ ptk2Δ inviable sky1Δ ptk2Δ inviable hsl1Δ mih1Δ inviable ypk1Δ ypk2Δ

E
ptc1Δ ptc1Δ ptc2Δ ptc1Δ ptc1Δ pph3Δ
1.0
0.8

0.8
0.6

Density
0.6
Density
0.4

0.4
0.2

0.2
0.0

0.0

-3 -2 -1 0 1 2 3 -3 -2 -1 0 1 2 3

M (log2(mt/wt)) M (log2(mt/wt))
F H
Ark1 Prk1 Ptc1 Ptc2

Sla1 Sla1 Hog1 Hog1


p p
G I
Δ
2
ptc


1
ptc

ptc

ptc
wt

ark1Δ prk1Δ
Hog1- p
sla1Δ

Hog1

Tubulin

Figure 4. Expression Profiling Reveals Three Different Genetic Buffering Interactions


For each set of three profiles all genes with changes in mRNA expression in any single profile are shown (p < 0.05, FC > 1.7).
(A) Complete redundancy whereby the single mutants have less than eight genes changing significantly and the double have more than eight.
(B) Quantitative redundancy, whereby one single mutant shows no significant profile (<8 genes p < 0.05, FC > 1.7), the other single mutant has a significant profile
and in the double the same genes change to a higher degree.
(C) Mixed epistasis. Here at least 8 more genes change significantly in the double versus the two singles, with at least 8 genes behaving in other ways than in
complete or quantitative phenotypes. The two bars below the FUS3-KSS1 profiles indicate the two gene sets selected for modeling (Figure 5).
(D) Unclassified buffering interactions due to inviability of the double mutant (Table 1).

998 Cell 143, 991–1004, December 10, 2010 ª2010 Elsevier Inc.
definition, all the cases of mixed epistasis contain at least two achieving overlap in function explains how functionally distinct
differently responding gene sets. We therefore considered nonhomologous pairs such as kinase-phosphatase pairs, can
models consisting of four nodes: two gene sets and two regula- nevertheless still have buffering effects. That the Boolean solu-
tors. To arrive at all possible solution models rather than a single tions encompass both direct and indirect ways of achieving
optimized solution, modeling was performed with Boolean oper- overlapping function fits well with the observation that mixed
ators (Albert et al., 2008; Ma et al., 2009). Since two nodes can be epistasis is exhibited by paralogous as well as nonhomologous
linked by different combinations of positive and negative regula- pairs (Table 1).
tory edges going in different directions, any two nodes can be Modeling shows that mixed epistasis arises through partial
connected in nine different ways. This leads to 794,176 models overlap in function combined with regulatory links from one
(Experimental Procedures), of which 106 result in the minimal partner to the other. The majority of genetic buffering interactions
mixed epistasis pattern (Figure 5A, Table S5). These steady- are mixed epistatic (Table 1). This indicates that the majority of
state solution models were pruned by removing superfluous genetically buffered kinase/phosphatase pairs have partial
edges (Figure S4C), revealing 28 root models that all exhibit overlap in function and regulatory links. As is explained below,
the experimentally observed mixed epistasis (Table S2). this has implications for understanding multiprocess control
Two important general properties emerge from these models. and for explaining the evolutionary maintenance of redundant
The first is inhibition or repression of one regulator by the other paralogs.
(Table S2 and Table S3). Different ways of achieving these unidi-
rectional negative effects are exemplified by the model solution Regulatorily Linked Pairs with Partial Overlap
that most closely resembles the literature-derived model for in Function Form Modules for Controlling
FUS3-KSS1 (Figures 5D and 5E). Besides encompassing all Different Combinations of Processes
the regulatory edges contained in the experimentally derived A consequence of the network topologies that explain the
scheme, including repression of kinase 2 expression by kinase minimal mixed epistasis pattern is that two distinct responses
1, in this Boolean model, kinase 1 also inhibits kinase 2. Previous can be regulated in either coupled or uncoupled manners.
experiments have suggested the existence of an inhibitory effect Depending on which regulator is active, a single process, or a
of Fus3 toward Kss1, albeit indirectly through Fus3-mediated second process in combination with the first, can be coordi-
activation of a Kss1-inhibitory phosphatase (Chen and Thorner, nately regulated. This feature is illustrated by FUS3-KSS1.
2007). Although this Boolean solution closely resembles the Although the mating pheromone response (Fus3) and the fila-
experimentally derived model (Figure 5C), it should be noted mentous growth starvation response (Kss1) are often treated
that this is not a root model and can be pruned by removal of as distinct, it has been reported that Kss1 is briefly activated
two edges without affecting outcome (Figures 5F and 5G). That during pheromone treatment (Ma et al., 1995). Furthermore,
the experimentally derived model contains seemingly super- under low mating pheromone concentrations, yeast cells display
fluous edges indicates that these features are required for a Kss1-dependent filamentation response that allows outgrowth
aspects of FUS3-KSS1 not modeled here, such as regulatory toward cells of the opposite mating type (Erdman and Snyder,
dynamics and the different behavior of other gene sets 2001). This is similar to Kss1-dependent filamentous growth
(Figure 4C). during nutrient starvation and suggests that under certain
A second general property of all the Boolean solutions is conditions, such as low pheromone concentration, aspects of
partial overlap in function. As with the negative effects, the filamentous growth are indeed regulatorily coupled to the mating
models indicate that partial overlap in function can also be response.
achieved in different ways. The least complex models, the two It is not well understood why redundant pairs such as paralogs
solutions that consist of only four edges, illustrate direct (Fig- are evolutionarily maintained (Vavouri et al., 2008). The ability to
ure 5F) and indirect ways (Figures 5H and 5I) in which partial flexibly couple and uncouple regulation of distinct processes is
overlap in function can be achieved. In the first root model (Fig- intuitively advantageous as a multiprocess control mechanism
ure 5F) both kinases have activating edges toward the first for responding to a large variety of different (combinations of)
responder gene set. This indicates redundancy and fits best conditions. If this ability is a driving force behind the evolutionary
with the expected action of redundant paralogs. The partial maintenance of redundant pairs, then one prediction is that the
nature of the redundancy is represented by different edges to gene sets that behave in different ways in mixed epistatic inter-
the other responder gene set. In the second simple Boolean actions should correspond to distinct processes. This prediction
root model (Figure 5H), partial overlap in function is achieved in is confirmed by Gene Ontology (GO) analysis of the groups of
a different, indirect way, with kinase 2 indirectly acting on one genes contained within the mixed epistasis profiles (Figure 6).
responder gene set through the other. This indirect manner of Presentation of this enrichment analysis as a network also

(E) Quantification of the profiles shown in B, plotted for all genes with significant (p < 0.05, FC > 1.7) changes in mRNA expression in any one single or double
mutant strain. M is the log2 ratio of normalized fluorescent mRNA expression in the mutant divided by WT. Asterisks indicate strains showing aneuploidy in the
double mutant whereby all genes on aneuploid chromosomes were excluded from analyses.
(F) Complete redundancy can result from two proteins able to directly substitute for all of each other’s activity.
(G) Expression profiles of the ark1D prk1D double mutant and the target sla1D. All genes are depicted with significant changes (p < 0.05, FC > 1.7) in mRNA
expression in any profile.
(H) Quantitative redundancy resulting from the ability of two proteins to directly substitute for each others activity qualitatively, but not quantitatively.
(I) Immunoblot as described in Figure 3C.

Cell 143, 991–1004, December 10, 2010 ª2010 Elsevier Inc. 999
A D

MF(ALPHA)1

YHR214W-A
K1 K2

YHR214W
YHR177W
YAR060C
AIM38
PGU1
AGA1

GPA1
SAG1
FAR1

TEC1

KTR2

KSS1
STE3

SST2

SRL1

FYV6
YJU2
OR

1 fus3Δ OR AND
2 kss1Δ
3 fus3Δ kss1Δ R1 R2

promoter (base pairs)


-800 -700 -600 -500 -400 -300 -200 -100
E
R1 (relative) R2 (relative)
k1Δ
k2Δ
k1Δ k2Δ
K1 (absolute) K2 (absolute)
wt
mating filamentous growth k1Δ
k2Δ
Tec1 binding site
k1Δ k2Δ
Ste12 binding site t 1 2 3 4 5 t 1 2 3 4 5
F
K1 K2

B
OR
Ste11
p
R1 R2
Ste7 Ste7 p
G
R1 (relative) R2 (relative)
Fus3 Fus3 p Kss1 k1Δ
k2Δ
k1Δ k2Δ
K1 (absolute) K2 (absolute)
Ste12 Ste12 wt
Tec1 Tec1
Ste12 Ste12 p k1Δ
p
k2Δ
k1Δ k2Δ
t 1 2 3 4 5 t 1 2 3 4 5
degradation
H
mating genes filamentous growth genes K1 K2

C
Ste11p
OR
Ste7 Ste7 p R1 R2

I
Kss1 Kss1
p R1 (relative) R2 (relative)
k1Δ
k2Δ
k1Δ k2Δ
Ste12 Ste12 Tec1 Tec1
Ste12 K1 (absolute) K2 (absolute)
Ste12 Ste12 Ste12 p wt
p
k1Δ
k2Δ
KSS1 k1Δ k2Δ
t 1 2 3 4 5 t 1 2 3 4 5
mating genes filamentous growth genes

Figure 5. Mechanisms of Mixed Epistasis: Partial Overlap in Function Coupled to Unidirectional Repression
(A) A minimal mixed epistasis pattern consisting of two gene sets selected from the FUS3-KSS1 profiles (Figure 4C). The names ‘‘mating’’ and
‘‘filamentous growth’’ are based on the enrichment for Ste12 and Tec1 transcription factor binding sites respectively, upstream of each gene, as indicated in
the vertical bars.
(B) Experimentally-derived/literature-based model for regulation of the mating and filamentous growth gene sets under basal, unactivated conditions in WT cells.
The model omits details such as activation of Ste12 and Tec1 transcription factor complexes through phosphorylation of the Dig1, Dig2 repressors (Chen and
Thorner, 2007). The black line between Kss1 and Fus3 indicates redundancy.
(C) Model for fus3D.
(D, F, and H) Boolean solution models for a minimal mixed epistasis pattern.
(E, G, and I) The accompanying state transitions for one of the eight simulated initial states (Experimental Procedures). R1 and R2 indicates the activities of the two
responder gene sets, depicted for the mutants relative to WT, similarly to the expression profiles, with blue indicating decrease, black no change and yellow

1000 Cell 143, 991–1004, December 10, 2010 ª2010 Elsevier Inc.
vacuolar
protein
septin catabolic
regulation
pheromone-dependent ring STB1 process
response organization of cell
signal
hexose STE12 to stress division
transduction iron
glycerol transport DIG1 transposition, involved response TEC1
RNA-mediated to
regulation SWI6 assimilation
biosynthetic HAP4 in of by
sexual response stimulus cellular
processresponse conjugation regulation ascospore establishment response reduction
to nucleotide reproduction response with of cell to
formation
energy
and
vacuole STE12 pheromone reserve or to DNA MBP1
osmotic metabolic cellular cycle maintenance damage transport
fusion, to DIG1 HAP4 during metabolic
response DNA
non-autophagic
stress process pheromone fusionreproduction regulation conjugation aging process
of cell stimulus
to iron replication
during cell death of mitotic with cellular polarity
microautophagy oxidative pheromone conjugation conjugation
DNA assimilation MBP1
cell cycle cellular nitrogen cellular phospholipid replication
phosphorylation with cell with cell DNA mitotic
polyphosphate nucleotide fusion compound response catabolic DNA
ion cellular adhesion cellular ion division conformation iron ion sister
metabolic conjugation metabolic metabolic to heat process repair
transport fusion fusion transport change transport chromatid
process process process
cohesion
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25

kss1 fus3 fus3 kss1 pbs2 pbs2 ptk2 pph3 d u n 1 dun1 pph3 e l m 1 mih1 elm1 slt2 slt2 ptp3 slt2 cla4 hsl1 cla4 cla4 bck1 cla4 bck1 bck1 ptp3 snf1 snf1 rim11

26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50

DNA beta-glucan pre-replicative regulation nucleotide


ribosomal Propanoate
replication
ribosomal
small biosynthetic complex cellular nucleosome metabolism
of metabolic ion cellular
autophagy
oxidation subunit translation process assembly response organization molecular ribosome process transport response
reduction export subunit function biogenesis
transposition, chromatin vacuolar to water vacuolar
from biogenesis to heat mitochondrial
deoxyribonucleotide RNA-mediated S phase assembly protein cell deprivation protein
biosynthetic nucleus ribosomal of mitotic osmosensory catabolic ncRNA RNA ATP
small or division catabolic
process viral cell cycle signaling disassembly process processing metabolic synthesis
ribosome subunit procapsid DNA process coupled RNA reproduction process
pathway oxidation
biogenesis assembly maturation replication electron processing ribosomal
FHL1 initiation protein-DNA reduction chromatin
fungal-type transport large
complex organization
DNA YOX1 cell wall assembly cellular HAP2 ribosomal subunit
integration MCM1 organization catabolic MIG1 large biogenesis
HAP4
YHP1 response process subunit
to assembly
cellular
temperature
response
stimulus
to heat

Figure 6. Multiprocess Control through Signaling Components with Mixed Epistasis


Yellow circular nodes represent the single and double mutant profiles for the pairs with mixed epistasis (Table 1). Single mutants with no significant changes
are not shown. Square nodes (numbered 1–50) indicate gene sets that show differential expression patterns across this set of mutants, obtained by QT clustering
all genes with a significant change (p < 0.05, FC > 1.7) in any one profile. Yellow edges between mutants and gene sets indicate that a gene set is upregulated in
the mutant, blue indicates downregulation. Diamonds indicate significant (p < 0.05) enrichment of a particular GO category in the gene set. Only the top three
categories are shown. Three-quarters of the gene sets are significantly enriched for at least one GO category. Triangles depict enrichment for transcription factor
binding sites in the gene set, indicating which transcription factor may be mediating the response. See also Figure S5.

illustrates the potential advantage of wiring together several molecular mechanisms underlying such interactions are rela-
such regulatory-coupled redundancy modules for multiprocess tively uncharacterized (Kelley and Ideker, 2005). Expression
control. Many different responses, represented by the square profiling provides detailed insight into the consequences of
nodes of coregulated genes, are influenced by several different mutations. This is exemplified here by the classification of a
regulatorily coupled regulators (Figure 6). In this way a large single type of SGI into three classes. Mixed epistasis is the
number of distinct combinations of processes can be regulated most unanticipated and it is also striking that it is the most
through different activity mixes of a relatively small number of common. The term epistasis is applied here in the broad, Fisher-
pathways. ian definition of any genetic interaction (Roth et al., 2009). To the
best of our knowledge, the simultaneous occurrence of different
DISCUSSION types of epistatic interactions between two genes has not been
generally described before. This is likely because the phenotyp-
Mixed Epistasis and Synthetic Genetic Interactions ical readout used here is more detailed than a fitness defect.
In model organisms, genetic buffering interactions are most
readily uncovered by measuring fitness under a standard growth Paralogous versus Nonhomologous Buffering
condition. Systematic determination of SGIs across all genes Redundancy is often associated with pairs of highly related
has only recently been initiated (Costanzo et al., 2010) and the genes (Prince and Pickett, 2002). One outcome of recently

increase in expression. K1 and K2 indicate the absolute activities of the regulator nodes with red for True and white for False. The numbers at the bottom indicate
the first five time steps of simulation.
See also Figure S4, Table S2, and Table S3.

Cell 143, 991–1004, December 10, 2010 ª2010 Elsevier Inc. 1001
initiated genome-wide mapping of genetic interactions is the Recurrent Modules and Pathway Connectivity
contribution of nonhomologous genes toward buffering (Cos- Recurrent motifs with important properties have previously been
tanzo et al., 2010). The relative contributions of nonhomologs described for transcription regulatory networks (Alon, 2007). The
versus duplicate pairs is under debate (Gu et al., 2003; Papp extent of signaling pathway connectivity has recently been
et al., 2004; Wagner, 2000), with a recent estimate as high as highlighted by systematic analysis of protein interactions (Breitk-
75% for nonhomologs (Ihmels et al., 2007). The gene pairs inves- reutz et al., 2010). Common regulatory motifs within signaling
tigated here were selected from a comprehensive kinase/phos- networks are not well established and little is known in general
phatase genetic interaction study (Fiedler et al., 2009). Half are about multiprocess control. Our analyses indicate that regulato-
either unambiguously nonhomologous or have arisen from rily coupled pairs with partial overlap in function form a common
ancient duplication events (over two billion years ago, Table 1). module for contributing to the control of different combinations
This agrees with a strong contribution of nonhomologous pairs of processes (Figure 6).
toward genetic buffering (Ihmels et al., 2007) and indicates that One of the regulatory links is repression of one regulator by the
redundancy is not merely the transient by-product of gene dupli- other, as exemplified by FUS3-KSS1. The dataset contains other
cations, since overlaps in cellular function have evolved from examples where inactivation of one redundant gene leads to
nonhomologous genes too. increase in expression of its partner (Figure S5). This regulatory
link contributes to differential expression of paralogs (Kafri
The Selective Advantage of Kinase/Phosphatase et al., 2005) and to paralog-responsiveness (DeLuna et al.,
Redundancy Is Superior Regulatory Systems 2010). The minimal mixed epistasis pattern modeled here
Other arguments in favor of an important functional role for consists of only two gene sets (Figure 5). Besides such gene
redundancy include the stable evolutionary maintenance of pa- sets, most mixed epistasis profiles also have additional gene
ralogs and the persistent nature of redundancy (Dean et al., sets behaving in different epistatic ways (Figure 4C). This implies
2008; Vavouri et al., 2008). Different types of selective advan- that wiring of such pairs also occurs in more ways than unidirec-
tages have been proposed for the maintenance of redundant tional repression and likely involves other mechanisms, including
paralogs, including robustness against mutation and robustness differential dose-response effects for other gene sets. The data
against stochastic fluctuations in gene expression (Kafri et al., forms a basis for unraveling such modules further and will be
2006; Nowak et al., 1997; Prince and Pickett, 2002). Backup useful for engineering different types of combinatorial control in
models lack explanation of why only some genes have backups synthetic signaling pathways (Kiel et al., 2010). Although the
and why redundancy is present in diploid organisms too. The number of pairs described here is likely an underestimate, it
partial nature of most redundancy, observed here and elsewhere should be noted that these were selected based on SGIs and
(Ihmels et al., 2007), as well as the condition-dependence of form only a distinct subset of all possible kinase/phosphatase
paralogous redundancy (Musso et al., 2008), also argue against pairs. Connectivity between signaling pathways therefore
backup function. Instead, the results favor superior control occurs in more ways. It can be anticipated that besides regula-
mechanisms as a selective advantage. The lack of phenotypes torily coupled pairs with partial overlapping function, more
expected for simple partial functional redundancy relationships recurrent modules will be uncovered by combinatorial analyses
(Figure 4) is particularly interesting since this indicates that pairs (Kelley and Ideker, 2005), especially of datasets that are starting
with partial overlap in function are always connected through to reveal the full scale of pathway connectivity (Breitkreutz et al.,
additional links. One property of such modules is that dependent 2010; Costanzo et al., 2010).
on which member of a pair is active, distinct processes can be
regulated in coupled or uncoupled manners. EXPERIMENTAL PROCEDURES
The formation of regulatory modules with superior control
potential may also have other implications for understanding All procedures are described in detail in the Extended Experimental
the evolution of gene duplications. Models explaining the main- Procedures.
tenance of paralogs include neo- and subfunctionalisation of
duplicate copies (DeLuna et al., 2008; Innan and Kondrashov, Expression Profiling and Deletion Strains
2010). Recent systematic studies indicate that neofunctionalisa- Each mutant strain, BY4742 (Table S4), was profiled four times from two
tion does not play a large role (Dean et al., 2008). The regulatory independently inoculated cultures. Sets of mutants were grown alongside
modules described here fit best with subfunctionalisation, but WT cultures, all processed in parallel. Dual-channel 70-mer oligonucleotide
arrays were employed with a common reference WT RNA. All steps after
the finding that partially redundant pairs are also coupled by
RNA isolation were automated using robotic liquid handlers. These proce-
regulatory links to each other may require additional subclassifi- dures were first optimized for accuracy (correct fold change) and precision
cation of these models (Innan and Kondrashov, 2010). (reproducible result), using spiked-in RNA calibration standards (van Bakel
Quantitatively redundant pairs may also confer superior regu- and Holstege, 2004). After quality control, normalization and dye-bias correc-
latory properties or may simply indicate requirement for a higher tion (Margaritis et al., 2009), statistical analysis was performed for each mutant
enzymatic capacity than can otherwise be reached with only versus the collection of 200 WT cultures. The reported fold change is the
a single copy. Complete redundancy phenotypes are a minority average of the four replicate mutant profiles versus the average of all WTs.
76 genes showed stochastic changes in WT profiles and were excluded
(Table 1). The selective advantage of such pairs remains enig-
from the analyses. Incorrect strains from the collection as indicated by aneu-
matic. Growth condition dependency of redundancy (Musso ploidy (5%), incorrect deletion (3%) or additional spurious mutation affecting
et al., 2008) suggests that if profiled under other conditions, the profile (3%), were remade and reprofiled (Table S4). None of the WT
such pairs may exhibit one of the other phenotypes. profiles had more than eight genes changing compared to the average WT

1002 Cell 143, 991–1004, December 10, 2010 ª2010 Elsevier Inc.
(p < 0.05, FC > 1.7). A threshold of fewer than eight genes changing was Chen, R.E., and Thorner, J. (2007). Function and regulation in MAPK signaling
therefore applied to determine whether a mutant had a significant profile. pathways: lessons learned from the yeast Saccharomyces cerevisiae. Bio-
chim. Biophys. Acta 1773, 1311–1340.
Double Mutants Chou, S., Huang, L., and Liu, H. (2004). Fus3-regulated Tec1 degradation
SGI data (Fiedler et al., 2009) were converted to Z-scores and double mutants through SCFCdc4 determines MAPK signaling specificity during mating in
were selected based on exhibiting a negative SGI, a Z-score significance of p < yeast. Cell 119, 981–990.
0.05 after multiple testing correction (46 pairs) and one of the mutants having
Chou, S., Lane, S., and Liu, H. (2006). Regulation of mating and filamentation
an expression profile similar to WT (24 pairs). Double mutants were all remade
genes by two distinct Ste12 complexes in Saccharomyces cerevisiae. Mol.
in an identical genetic background as the single mutants. Six were inviable,
Cell. Biol. 26, 4794–4805.
consistent with buffering. One double mutant (dun1D chk1D) had different
degrees of aneuploidy in different isolates and buffering could not be confi- Cope, M.J., Yang, S., Shang, C., and Drubin, D.G. (1999). Novel protein
dently determined from the profile (Table S1). kinases Ark1p and Prk1p associate with and regulate the cortical actin
cytoskeleton in budding yeast. J. Cell Biol. 144, 1203–1218.
Boolean Modeling Costanzo, M., Baryshnikova, A., Bellay, J., Kim, Y., Spear, E.D., Sevier, C.S.,
Given four nodes and no self-edges, topologies were constrained to be Ding, H., Koh, J.L., Toufighi, K., Mostafavi, S., et al. (2010). The genetic
completely connected and have at least two edges from the regulator nodes landscape of a cell. Science 327, 425–431.
(K1, K2) to the responder nodes (R1, R2). The number of incoming edges on Dean, E.J., Davis, J.C., Davis, R.W., and Petrov, D.A. (2008). Pervasive and
any node was limited to two. Influence of two incoming edges could be persistent redundancy among duplicated genes in yeast. PLoS Genet. 4,
Boolean AND or OR. Synchronous Boolean simulations were run for all e1000113.
possible initial states of K2, R1, and R2. The initial state of K1 was True. Solu-
DeLuna, A., Springer, M., Kirschner, M.W., and Kishony, R. (2010). Need-
tion models were those that converged to a steady state under all initial state
based up-regulation of protein levels in response to deletion of their duplicate
settings and had the final states of wild-type: R1 = True, R2 = False; k1D: R1 =
genes. PLoS Biol. 8, e1000347.
True, R2 = True; k2D: R1 = True, R2 = False; k1D k2D: R1 = False, R2 = False.
DeLuna, A., Vetsigian, K., Shoresh, N., Hegreness, M., Colón-González, M.,
Chao, S., and Kishony, R. (2008). Exposing the fitness contribution of
ACCESSION NUMBERS
duplicated genes. Nat. Genet. 40, 676–681.
All microarray gene expression data have been deposited in the public Elion, E.A., Brill, J.A., and Fink, G.R. (1991). FUS3 represses CLN1 and CLN2
data repositories ArrayExpress (accession numbers E-TABM-907 [mutants] and in concert with KSS1 promotes signal transduction. Proc. Natl. Acad. Sci.
and E-TABM-773 [200 WT replicates]) and GEO (GSE25644 [mutants]). The USA 88, 9392–9396.
data are also available as flat-file or in TreeView format from http://www. Erdman, S., and Snyder, M. (2001). A filamentous growth response mediated
holstegelab.nl/publications/sv/signaling_redundancy/. by the yeast mating pathway. Genetics 159, 919–928.
Fiedler, D., Braberg, H., Mehta, M., Chechik, G., Cagney, G., Mukherjee, P.,
SUPPLEMENTAL INFORMATION Silva, A.C., Shales, M., Collins, S.R., van Wageningen, S., et al. (2009).
Functional organization of the S. cerevisiae phosphorylation network. Cell
Supplemental Information includes Extended Experimental Procedures, 136, 952–963.
five figures, and five tables and can be found with this article online at Gu, Z., Steinmetz, L.M., Gu, X., Scharfe, C., Davis, R.W., and Li, W.H. (2003).
doi:10.1016/j.cell.2010.11.021. Role of duplicate genes in genetic robustness against null mutations. Nature
421, 63–66.
ACKNOWLEDGMENTS
Hartman, J.L., 4th, Garvik, B., and Hartwell, L. (2001). Principles for the
buffering of genetic variation. Science 291, 1001–1004.
This work was supported by the Netherlands Bioinformatics Centre (NBIC) and
the Netherlands Organization of Scientific Research (NWO), grants Ihmels, J., Collins, S.R., Schuldiner, M., Krogan, N.J., and Weissman, J.S.
016.108.607, 817.02.015, 050.71.057, 911.06.009, 021.002.035 (T.L.L.), (2007). Backup without redundancy: genetic interactions reveal the cost of
863.07.007 (P.K.), 700.57.407 (J.J.B.). duplicate gene loss. Mol. Syst. Biol. 3, 86.
Innan, H., and Kondrashov, F. (2010). The evolution of gene duplications:
Received: January 29, 2010 classifying and distinguishing between models. Nat. Rev. Genet. 11, 97–108.
Revised: September 20, 2010 Jacoby, T., Flanagan, H., Faykin, A., Seto, A.G., Mattison, C., and Ota, I.
Accepted: November 9, 2010 (1997). Two protein-tyrosine phosphatases inactivate the osmotic stress
Published: December 9, 2010 response pathway in yeast by targeting the mitogen-activated protein kinase,
Hog1. J. Biol. Chem. 272, 17749–17755.
REFERENCES
Kafri, R., Bar-Even, A., and Pilpel, Y. (2005). Transcription control reprogram-
ming in genetic backup circuits. Nat. Genet. 37, 295–299.
Albert, I., Thakar, J., Li, S., Zhang, R., and Albert, R. (2008). Boolean network
simulations for life scientists. Source Code Biol. Med. 3, 16. Kafri, R., Levy, M., and Pilpel, Y. (2006). The regulatory utilization of genetic
Alon, U. (2007). Network motifs: theory and experimental approaches. Nat. redundancy through responsive backup circuits. Proc. Natl. Acad. Sci. USA
Rev. Genet. 8, 450–461. 103, 11653–11658.

Bermejo, C., Rodrı́guez, E., Garcı́a, R., Rodrı́guez-Peña, J.M., Rodrı́guez de la Kelley, R., and Ideker, T. (2005). Systematic interpretation of genetic interac-
Concepción, M.L., Rivas, C., Arias, P., Nombela, C., Posas, F., and Arroyo, J. tions using protein networks. Nat. Biotechnol. 23, 561–566.
(2008). The sequential activation of the yeast HOG and SLT2 pathways is Kiel, C., Yus, E., and Serrano, L. (2010). Engineering signal transduction
required for cell survival to cell wall stress. Mol. Biol. Cell 19, 1113–1124. pathways. Cell 140, 33–47.
Breitkreutz, A., Choi, H., Sharom, J.R., Boucher, L., Neduva, V., Larsen, B., Lin, Ma, D., Cook, J.G., and Thorner, J. (1995). Phosphorylation and localization of
Z.Y., Breitkreutz, B.J., Stark, C., Liu, G., et al. (2010). A global protein kinase Kss1, a MAP kinase of the Saccharomyces cerevisiae pheromone response
and phosphatase interaction network in yeast. Science 328, 1043–1046. pathway. Mol. Biol. Cell 6, 889–909.
Capaldi, A.P., Kaplan, T., Liu, Y., Habib, N., Regev, A., Friedman, N., and Ma, W., Trusina, A., El-Samad, H., Lim, W.A., and Tang, C. (2009). Defining
O’Shea, E.K. (2008). Structure and function of a transcriptional network network topologies that can achieve biochemical adaptation. Cell 138,
activated by the MAPK Hog1. Nat. Genet. 40, 1300–1306. 760–773.

Cell 143, 991–1004, December 10, 2010 ª2010 Elsevier Inc. 1003
Madhani, H.D., and Fink, G.R. (1997). Combinatorial control required for the Smythe, E., and Ayscough, K.R. (2003). The Ark1/Prk1 family of protein
specificity of yeast MAPK signaling. Science 275, 1314–1317. kinases. Regulators of endocytosis and the actin skeleton. EMBO Rep. 4,
Madhani, H.D., Styles, C.A., and Fink, G.R. (1997). MAP kinases with distinct 246–251.
inhibitory functions impart signaling specificity during yeast differentiation. van Bakel, H., and Holstege, F.C. (2004). In control: systematic assessment of
Cell 91, 673–684. microarray performance. EMBO Rep. 5, 964–969.
Margaritis, T., Lijnzaad, P., van Leenen, D., Bouwmeester, D., Kemmeren, P., Vavouri, T., Semple, J.I., and Lehner, B. (2008). Widespread conservation of
van Hooff, S.R., and Holstege, F.C. (2009). Adaptable gene-specific dye bias genetic redundancy during a billion years of eukaryotic evolution. Trends
correction for two-channel DNA microarrays. Mol. Syst. Biol. 5, 266. Genet. 24, 485–488.
Musso, G., Costanzo, M., Huangfu, M., Smith, A.M., Paw, J., San Luis, B.J., Wagner, A. (2000). Robustness against mutations in genetic networks of yeast.
Boone, C., Giaever, G., Nislow, C., Emili, A., and Zhang, Z. (2008). The Nat. Genet. 24, 355–361.
extensive and condition-dependent nature of epistasis among whole-genome Warmka, J., Hanneman, J., Lee, J., Amin, D., and Ota, I. (2001). Ptc1, a type 2C
duplicates in yeast. Genome Res. 18, 1092–1099. Ser/Thr phosphatase, inactivates the HOG pathway by dephosphorylating the
Nowak, M.A., Boerlijst, M.C., Cooke, J., and Smith, J.M. (1997). Evolution of mitogen-activated protein kinase Hog1. Mol. Cell. Biol. 21, 51–60.
genetic redundancy. Nature 388, 167–171. Winkler, A., Arkind, C., Mattison, C.P., Burkholder, A., Knoche, K., and Ota, I.
Papp, B., Pál, C., and Hurst, L.D. (2004). Metabolic network analysis of the (2002). Heat stress activates the yeast high-osmolarity glycerol mitogen-
causes and evolution of enzyme dispensability in yeast. Nature 429, 661–664. activated protein kinase pathway, and protein tyrosine phosphatases are
Prince, V.E., and Pickett, F.B. (2002). Splitting pairs: the diverging fates of essential under heat stress. Eukaryot. Cell 1, 163–173.
duplicated genes. Nat. Rev. Genet. 3, 827–837. Wurgler-Murphy, S.M., Maeda, T., Witten, E.A., and Saito, H. (1997).
Roberts, C.J., Nelson, B., Marton, M.J., Stoughton, R., Meyer, M.R., Bennett, Regulation of the Saccharomyces cerevisiae HOG1 mitogen-activated protein
H.A., He, Y.D., Dai, H., Walker, W.L., Hughes, T.R., et al. (2000). Signaling and kinase by the PTP2 and PTP3 protein tyrosine phosphatases. Mol. Cell. Biol.
circuitry of multiple MAPK pathways revealed by a matrix of global gene 17, 1289–1297.
expression profiles. Science 287, 873–880. Young, C., Mapes, J., Hanneman, J., Al-Zarban, S., and Ota, I. (2002). Role of
Rodrı́guez-Peña, J.M., Pérez-Dı́az, R.M., Alvarez, S., Bermejo, C., Garcı́a, R., Ptc2 type 2C Ser/Thr phosphatase in yeast high-osmolarity glycerol pathway
Santiago, C., Nombela, C., and Arroyo, J. (2005). The ‘yeast cell wall chip’ - inactivation. Eukaryot. Cell 1, 1032–1040.
a tool to analyse the regulation of cell wall biogenesis in Saccharomyces Zeng, G., Yu, X., and Cai, M. (2001). Regulation of yeast actin cytoskeleton-
cerevisiae. Microbiology 151, 2241–2249. regulatory complex Pan1p/Sla1p/End3p by serine/threonine kinase Prk1p.
Roth, F.P., Lipshitz, H.D., and Andrews, B.J. (2009). Q&A: epistasis. J. Biol. 8, 35. Mol. Biol. Cell 12, 3759–3772.

1004 Cell 143, 991–1004, December 10, 2010 ª2010 Elsevier Inc.
Theory

An Integrated Approach
to Uncover Drivers of Cancer
Uri David Akavia,1,2,5 Oren Litvin,1,2,5 Jessica Kim,3,4 Felix Sanchez-Garcia,1 Dylan Kotliar,1 Helen C. Causton,1
Panisa Pochanard,3,4 Eyal Mozes,1 Levi A. Garraway,3,4 and Dana Pe’er1,2,*
1Department of Biological Sciences, Columbia University, 1212 Amsterdam Avenue, New York, NY 10027, USA
2Center for Computational Biology and Bioinformatics, Columbia University, 1130 St. Nicholas Avenue, New York, NY 10032, USA
3Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 44 Binney Street, Boston, MA 02115, USA
4Broad Institute of Harvard and MIT, 7 Cambridge Center, Cambridge, MA 02142, USA
5These authors contributed equally to this work

*Correspondence: dpeer@biology.columbia.edu
DOI 10.1016/j.cell.2010.11.013

SUMMARY of which only a few drive proliferation and metastasis. Thus,


identifying driver mutations (genetic changes that promote
Systematic characterization of cancer genomes has cancer progression) and distinguishing them from passengers
revealed a staggering number of diverse aberrations (those with no selective advantage) has emerged as a major
that differ among individuals, such that the functional challenge in the genomic characterization of cancer.
importance and physiological impact of most tumor The most widely used approaches are based on the frequency
genetic alterations remain poorly defined. We devel- that an aberration occurs: if a mutation provides a fitness advan-
tage in a given tumor type, its persistence will be favored, and it is
oped a computational framework that integrates
likely to be found in multiple tumors. For example, GISTIC iden-
chromosomal copy number and gene expression
tifies regions of the genome that are aberrant more often than
data for detecting aberrations that promote cancer would be expected by chance and has been used to analyze
progression. We demonstrate the utility of this a number of cancers (Beroukhim et al., 2007, 2009; Lin et al.,
framework using a melanoma data set. Our analysis 2008). However, there are limitations to analytical approaches
correctly identified known drivers of melanoma and based on CNA data alone: CNA regions are typically large and
predicted multiple tumor dependencies. Two depen- contain many genes, most of which are passengers that are
dencies, TBC1D16 and RAB27A, confirmed empiri- indistinguishable in copy number from the drivers. CNA data
cally, suggest that abnormal regulation of protein have statistical power to detect only the most frequently recur-
trafficking contributes to proliferation in melanoma. ring drivers above the large number of unrelated chromosomal
Together, these results demonstrate the ability of aberrations that are typical in cancer. Finally, these approaches
rarely elucidate the functional importance or physiological
integrative Bayesian approaches to identify candi-
impact of the genetic alteration on the tumor. These limitations
date drivers with biological, and possibly thera-
highlight the need for new approaches that can integrate addi-
peutic, importance in cancer. tional data to identify drivers of cancer. Gene expression is
readily available for many tumors, but how best to combine it
INTRODUCTION with information on CNA is not obvious.
We postulate that driver mutations coincide with a ‘‘genomic
Large-scale initiatives to map chromosomal aberrations, muta- footprint’’ in the form of a gene expression signature. We devel-
tions, and gene expression have revealed a highly complex oped an algorithm that integrates chromosomal copy number
assortment of genetic and transcriptional changes within indi- and gene expression data to find these signatures and identify
vidual tumors. For example, copy number aberrations (CNAs) likely driver genes located in regions that are amplified or deleted
occur frequently in cancer due to genomic instability. Genomic in tumors. Each potential driver gene is altered in some, but not
data have been collected for thousands of tumors at high reso- all, tumors and, when altered, is considered likely to play
lution using array comparative genomic hybridization (aCGH) a contributing role in tumorigenesis. Unique to our approach,
(Pinkel et al., 1998), high-density single-nucleotide polymor- each driver is associated with a gene module, which is assumed
phism (SNP) microarrays (Beroukhim et al., 2010; Lin et al., to be altered by the driver. We sometimes gain insight into the
2008), and massively parallel sequencing (Pleasance et al., likely role of a candidate driver based on the annotation of the
2010). Although multiple new genes have been implicated in genes in the associated module. We demonstrate the utility of
cancer through sequencing and CNA analysis (Garraway et al., our method using a data set (Lin et al., 2008) that includes paired
2005), these studies have also revealed enormous diversity in measurements of gene expression and copy number from 62
genomic aberrations in tumors among individuals. Each tumor melanoma samples. Our analysis correctly identified known
is unique and typically harbors a large number of genetic lesions, drivers of melanoma and connected them to many of their

Cell 143, 1005–1017, December 10, 2010 ª2010 Elsevier Inc. 1005
A B C Other
Aberrant region Copy Number Driver Copy Number Factors

same chromosome
Normal Amplified

Driver

Genes in an aberrant region Target


Genes
Normal Malignant
Passenger Driver Phenotype Phenotype

D Chr17:68172496-73084144

CNA:
Expression:
Normal
-2 0 2
Deleted Log Change

Figure 1. Modeling Assumptions


For all heat maps, each row represents a gene and each column represents a tumor sample.
(A) The same chromosome in different tumors; orange represents amplified regions. The box shows regions amplified in multiple tumors.
(B) An idealized signature in which the target genes are upregulated (red) when the DNA encoding the driver is amplified (orange).
(C) A driver may be overexpressed due to amplification of the DNA encoding it or due to the action of other factors. The target genes correlate with driver gene
expression (middle row), rather than driver copy number (top row).
(D) Data representing amplified region on chromosome 17. Heat maps of expression for 10 of 24 genes that passed initial expression filtering (Extended Exper-
imental Procedures).
Samples are ordered according to amplification status of the region (orange, amplified; blue, deleted). These genes are identical in their amplification status, and
though gene expression is correlated with amplification status to some degree, the expression of each gene is unique. It is these differences that facilitate the
identification of the driver. See also Extended Experimental Procedures, Figure S1, and Table S1.

targets and biological functions. In addition, it predicted novel mutation might be associated with a characteristic gene expres-
melanoma tumor dependencies, two of which, TBC1D16 and sion signature or other phenotypic output representing a group
RAB27A, were confirmed experimentally. Both of these genes of genes whose expression is modulated by the driver. In addi-
are involved in the regulation of vesicular trafficking, which high- tion, CNAs do not typically alter the coding sequence of the
lights this process as important for proliferation in melanoma. driver and so are expected to influence cellular phenotype via
changes in the driver’s expression. In consequence, changes
RESULTS in expression of the driver are important, so approaches that
measure association between the expression of a candidate
The Genomic Signature of a Driver driver (as opposed to its copy number) and that of the genes in
We define a ‘‘driver mutation’’ to be a genetic alteration that the corresponding module are likely to promote the identification
provides the tumor cell with a growth advantage during carcino- of drivers.
genesis or tumor progression (Stratton et al., 2009). We Gene expression is particularly useful for identifying candidate
reasoned that driver mutations might leave a genomic ‘‘foot- drivers within large amplified or deleted regions of a chromo-
print’’ that can assist in distinguishing between driver and some: whereas genes located in a region of genomic copy
passenger mutations based on the following assumptions: (1) gain/loss are indistinguishable in copy number, expression
a driver mutation should occur in multiple tumors more often permits the ranking of genes based on how well they correspond
than would be expected by chance (Figure 1A); (2) a driver with the phenotype (Figure 1D). CNA data aids in determining the
mutation may be associated (correlated) with the expression of direction of influence, which cannot be derived based on corre-
a group of genes that form a ‘‘module’’ (Figure 1B); (3) copy lation in gene expression alone (Figure 3A). This permits an unbi-
number aberrations often influence the expression of genes in ased approach for identifying candidate drivers from any func-
the module via changes in expression of the driver (Figure 1C). tional family, beyond transcription factors or signaling proteins.
Driver mutations are frequently associated with the abnormal
regulation of processes such as proliferation, differentiation, A Bayesian Network-Based Algorithm
motility, and invasion. Given that many cancer phenotypes are to Identify Driver Genes
reflected in coordinated differences in the expression of multiple We developed a computational algorithm, copy number and
genes (a module) (Golub et al., 1999; Segal et al., 2004), a driver expression in cancer (CONEXIC), that integrates matched copy

1006 Cell 143, 1005–1017, December 10, 2010 ª2010 Elsevier Inc.
Figure 2. The Highest-Scoring Modulators Identi-
fied by CONEXIC
Gene names are color coded based on the role of the gene
in cancer. Ten genes have been previously identified as
oncogenes or tumor suppressors (blue); of these, two in
melanoma (brown). Column 3 represents chromosomal
location, orange represents amplification, and blue repre-
sents deletion. These genes were identified within regions
containing multiple genes, and the number of genes in
each aberrant region is listed in column 4. Column 5 lists
the p value for modulator validation in independent data
(for a full list, see Table S2 and Figure S3C). p values are
shown for the Johansson data set unless the modulator
was missing from this data set, and then p value from
the Hoek data set is shown. See also Extended Experi-
mental Procedures, Table S2, and Figure S3.

nomas (Lin et al., 2008). A list of candidate


drivers was generated using copy number data
available for 101 melanoma samples by
applying a modified version (Sanchez-Garcia
et al., 2010) of GISTIC (Beroukhim et al., 2007)
(see Table S1). Next, we integrated copy
number and gene expression data (available
for 62 tumors) to identify the most likely drivers
(Extended Experimental Procedures). Statistical
power is gained by integrating all data and by
combining statistical tests on thousands of
genes to support the selected modulators.
This resulted in the identification of 64 modula-
number (amplifications and deletions) and gene expression tors that explain the behavior of 7869 genes. We consider the
data from tumor samples to identify driver mutations and top 30 scoring modulators, presented in Figure 2, as likely drivers
the processes that they influence. CONEXIC is inspired by (see Table S2 for the complete list).
Module Networks (Segal et al., 2003) but has been augmented
by a number of critical modifications that make it suitable for Many Modulators Are Involved in Pathways Related
identifying drivers (see Extended Experimental Procedures avail- to Melanoma
able online). CONEXIC uses a score-guided search to identify The top 30 modulators (likely drivers) include 10 known
the combination of modulators that best explains the behavior oncogenes and tumor suppressors (Figure 2). In many cases,
of a gene expression module across tumor samples and CONEXIC chose the cancer-related gene out of a large aberrant
searches for those with the highest score within the amplified region containing many genes. For example, DIXDC1, a gene
or deleted regions (Extended Experimental Procedures and known to be involved in the induction of colon cancer (Wang
Figure S1). et al., 2009b), was selected among 17 genes in an aberrant
The resulting output is a ranked list of high-scoring modulators region (Figure S2). CCNB2, a cell-cycle regulator, was selected
that both correlate with differences in gene expression modules from a large amplified region containing 33 genes. The modula-
across samples and are located in amplified or deleted regions in tors span diverse functional classes, including signal trans-
a significant number of these samples. The fact that the modula- ducers (TRAF3), transcription factors (KLF6), translation factors
tors are amplified or deleted indicates that they are likely to (EIF5), and genes involved in vesicular trafficking (RAB27A).
control the expression of the genes in the corresponding Performing a comprehensive literature search for all genes
modules (see Figure 3). Because the modulators are amplified is tedious and time consuming, so we developed an automated
or deleted in a significant number of tumors, it is reasonable to procedure, literature vector analysis (LitVAn), which searches
assume that expression of the modulator (altered by copy for overrepresented terms in papers associated with genes
number) contributes a fitness advantage to the tumor. Therefore, in a gene set. LitVAn uses a manually curated database (NCBI
the modulators likely include genes whose alteration provides Gene) to connect genes with terms from the complete text of
a fitness advantage to the tumor. more than 70,000 published scientific articles (Extended Exper-
imental Procedures). LitVAN found a number of overrepresented
Identifying Candidate Driver Genes in Melanoma terms (Figure S3E) among the top 30 modulators, including
We applied the CONEXIC algorithm to paired gene expression ‘‘PI3K’’ and ‘‘MAPK,’’ which are known to be activated in mela-
and CNA data from 62 cultured (long- and short-term) mela- noma; ‘‘cyclin,’’ representing proliferation, which is common in

Cell 143, 1005–1017, December 10, 2010 ª2010 Elsevier Inc. 1007
all cancers; and ‘‘RAB.’’ Rabs regulate vesicular trafficking, a the 80 targets identified by Hoek et al. (p value < 1.5 3 10 78).
process not previously implicated in melanoma. Similar target sets are not available for any other modulator,
precluding a more rigorous evaluation of our other predictions.
The Association between a Modulator and the Genes
in a Module MITF Expression Correlates with Targets Better
Beyond generating a list of likely drivers (modulators), the Than Copy Number
CONEXIC output includes groups of genes that are associated Expression of MITF correlates with the expression of its targets
with each modulator (modules). We tested how reproducible better than MITF copy number, though both correlations are
the modulators and their associated modules are using gene statistically significant (p value of 0.0001 versus 0.04; Figures 4A
expression data from two other melanoma cohorts with 45 and 4B). This relationship is unidirectional: MITF is significantly
(Hoek et al., 2006) and 63 (Johansson et al., 2007) samples overexpressed when its DNA is amplified (p value 0.0004), but
(see Extended Experimental Procedures and Figure S3). We overexpressed MITF does not always correspond with MITF
found that 51 of 64 (80%) of the selected modulators are amplification. We find that MITF is less correlated with its copy
conserved across data sets in a statistically significant manner. number (rank 294th) than most other genes in aberrant regions
Modules (statistically associated genes) are likely enriched with (see Table S1C), and more than half of the tumors that overex-
genes whose expression is biologically affected by the modu- press MITF do not have a CNA that spans the MITF gene. Compar-
lator (Figure 3). In consequence, the processes and pathways ison of MITF target expression between samples with and without
represented by genes in a module can help us to gain insight MITF amplification did not show an effect of DNA amplification on
into how an aberration in the modulator might alter the cellular expression of the targets (Extended Experimental Procedures).
physiology and contribute to the malignant phenotype.
Annotation of data-derived sets of genes is typically carried out MITF Correctly Annotated with Its Known Role
based on gene set enrichment using Gene Ontology (GO) annota- in Melanoma
tion. Although this approach is useful, there are modules for which We used GO gene set enrichment to identify the biological
GO annotation does not capture the known biology. For example, processes and pathways represented in each module associ-
the ‘‘TNF module’’ is enriched with the GO terms ‘‘developmental ated with MITF. The module containing the genes most signifi-
process’’ and ‘‘cell differentiation’’ (q value = 0.0014 and 0.004, cantly upregulated by MITF (Figure 4B and Figure S4A) is signif-
respectively). We used LitVAn to carry out a systematic literature icantly enriched for the terms ‘‘melanosome’’ and ‘‘pigment
search and found 11 of 20 genes in the module related to the granule’’ (q value = 4.86e 6 for each). It includes targets involved
TNF pathway, inflammation, or both (Figure 3C and Table S3), in proliferation such as CDK2, consistent with the observation
although only two of these genes were annotated for these that MITF can promote proliferation via lineage-specific regula-
processes in GO. TRAF3, the modulator chosen by CONEXIC, is tion of CDK2 (Du et al., 2004). The module containing genes
known to regulate the NF-kB pathway (Vallabhapurapu et al., most strongly inhibited by MITF (Figure 4B and Figure S4B)
2008), a major downstream target of TNF. Although TRAF3 has has a metastatic signature strongly associated with invasion,
not been previously implicated in melanoma, the importance of angiogenesis, the extracellular matrix, and NF-kB signaling.
the NF-kB pathway in melanoma is well supported (Chin et al., These modules and their annotation suggest that MITF serves
2006). as a developmental switch between two types of melanoma, in
which high MITF expression promotes proliferation and low
A Known Driver, MITF, Is Correctly Associated MITF expression promotes invasion. Thus, our automated,
with Target Genes computationally derived findings dissect a complex response
CONEXIC identified microphthalmia-associated transcription and accurately recapitulate the known literature, including the
factor (MITF) as the highest-scoring modulator. MITF is a master experimental characterization of MITF (Hoek et al., 2008a).
regulator of melanocyte development, function, and survival LitVAN annotated additional modulators with their known role
(Levy et al., 2006; Steingrı́msson et al., 2004), and the overex- (e.g., CCNB2 with cell cycle and mitosis; data not shown). The
pression of MITF is known to have an adverse effect on patient detailed match between the CONEXIC output and empirically
survival (Garraway et al., 2005). derived knowledge of the role of known modulators in melanoma
To test the association between modulator and module, we ob- provides confidence in CONEXIC’s predictions for modulators
tained an experimentally derived list of MITF targets (Hoek et al., that are not well characterized.
2008b) and asked whether the modules identified by CONEXIC
associate MITF with its known targets. The MITF-associated Identification of TBC1D16 as a Tumor Dependency
modules contained 45 of 80 previously identified targets in Melanoma
(p value < 1.5 3 10 45) supporting a match between the transcrip- The second highest-scoring modulator identified by CONEXIC
tion factor (TF) and its known targets. However, a few targets is TBC1D16, a Rab GTPase-activating protein of unknown
(TBC1D16, ZFP106, and RAB27A) are both associated with biological function. Rabs are small monomeric GTPases
MITF and are themselves modulators of additional modules. involved in membrane transport and trafficking. TBC1D16 is
CONEXIC limits each gene to a single module, so association well conserved, and although its targets are not known, a close
with an MITF target would preclude association with MITF. If we paralog, TBC1D15, regulates RAB7A (also selected as a modu-
permit indirect association to MITF through the modules of lator; Figure 2) (Itoh et al., 2006). We used a module associated
these additional modulators, CONEXIC correctly identifies 76 of with TBC1D16 to infer its potential role in melanoma (Figure 5A)

1008 Cell 143, 1005–1017, December 10, 2010 ª2010 Elsevier Inc.
A
Copy Number of gene A
A B Other
Factors
B A
C A

A B B
B Modulator X
Cell
Modulator Y
Down Up
Modulator X Processes Modulator X
(Metabolism,
Growth, etc.)

Factors

Underlying
Biology
-2 0
Log Change
2
(OR)
Modulation detected Indirect Modulation Joint Modulator
by Conexic

C TRAF3
Exp
CNA

MITF
Exp
CNA

TNF - By GO
TNF

Figure 3. Associating Modulators to Genes


(A) Three scenarios could explain a correlation between a candidate driver (gene A) and its target (gene B): A could influence B, B could influence A, or both could
be regulated by a common third mechanism (Pearl, 2000). The availability of both gene expression and chromosomal copy number data allows us to establish the
likely direction of influence. If the expression of gene A is correlated with its DNA copy number and the copy number is altered in a large number of tumors, it is
likely that the copy number alteration results in a change in expression of A in these tumors. So the model in which A influences the expression of B and other
correlated genes is the most likely. In this way, examination of both copy number and gene expression in a single integrated computational framework facilitates
identification of candidate drivers.
(B) Modulator influence on a module can go beyond direct transcriptional cascades involving transcription factors or signaling proteins and their targets. Genetic
alteration of any gene (e.g., a metabolic enzyme) can alter cell physiology, which is sensed by the cell and subsequently leads to a transcriptional response
through a cascade of indirect influences and mechanisms. Whereas modules are typically enriched for genes influenced by the modulator, they also contain
genes that are coexpressed with the modulator (‘‘joint modulator’’). Both types are helpful for annotating the module and determining the functional role of
the modulator.
(C) The TNF module. The modulators include TRAF3 and MITF, wherein high TRAF3 and low MITF are required for upregulation of the genes in the module. The
annotation for each gene is represented in a color-coded matrix. Blue and orange squares represent literature-based annotation (see Table S3); green and brown
are from GO. LitVAN associated the genes in this module with TNF and the inflammatory response.
See also Figure S2 and Table S3.

Cell 143, 1005–1017, December 10, 2010 ª2010 Elsevier Inc. 1009
B MITF-Expression Figure 4. MITF Expression Correlates with Expres-
A MITF-CNA
sion of the Genes in the Associated Module
(A) Each row represents the gene expression of 1 of 78
MITF targets identified by Hoek (Hoek et al., 2008b); the
tumor samples are split into two groups based on the
copy number of MITF (Welch t test p value = 0.04).
(B) The rows represent the same genes, in the same order
as in (A), but here, the tumor samples are split into a group
Hoek MITF Targets

of samples that express MITF at high (n = 46) or low levels


(n = 16) (Welch t test p value = 0.0001).
(C) Two modules associated with MITF, showing a
selected subset of genes. LitVAN annotation for the genes
in each module is shown below the heat map. The com-
plete modules with all genes are available in Figure S4.

CNA:
Expression:
We carried out western blotting and RT-PCR on
Normal -2 some of the short-term cultures (STCs) used to
0 2
Deleted Log Change generate the Lin data set and asked whether
the TBC1D16 transcript correlates with protein
C levels. The results confirmed that the expression
MITF
Exp of TBC1D16 corresponds well with the amount
CNA
Low Expression High Expression of the 45 kD isoform of TBC1D16 (data not
STX7, MYO5A, shown). These results suggest that knockdown
76 Genes

RAB27A, RAB7A, of TBC1D16 expression in tumors that have


RAB38, SORT1, high levels of TBC1D16 will lead to a reduction
CDK2, MLANA, in proliferation.
DCT...
Genes overexpressed Vesicular Trafficking
when MITF is high Melanogenesis TBC1D16 Is Required for Proliferation
are involved in: Lysosome/Endosome To test whether TBC1D16 is required for prolif-
Known MITF Targets eration of melanoma cultures, we carried out
SMAD3, CTGF, a knockdown experiment. We selected two
84 Genes

SMURF2, CCL2, STCs with high levels of TBC1D16, WM1960


NFKBIA, ITGA3, (16-fold higher expression than WM1346, DNA
CXCL1, ITGA5...
not amplified) and WM1976 (34-fold higher
Genes overexpressed NFkB/TNF
expression, amplified DNA) and control STCs,
when MITF is low Invasion/Migration WM262 and WM1346 that express TBC1D16
are involved in: Angiogenesis at a lower level. We used two shRNAs to knock
down TBC1D16 expression in each of the four
STCs and measured growth over 8 days
(Extended Experimental Procedures). RT-PCR
and discovered that diverse biological processes are repre- was used to confirm that the reduction in the amount of the
sented by genes in the module and that more than half are anno- TBC1D16 transcript was similar for all of the STCs (Figure S5).
tated for processes such as melanogenesis, vesicular trafficking, Knockdown of TBC1D16 expression reduced cell growth in
and survival/proliferation (Table S4A). This suggests that WM1960 and WM1976 to 16% and 40%, respectively, relative
TBC1D16 plays a role in cell survival and proliferation. to controls infected with GFP shRNA in the same STCs (Figures
TBC1D16 is an uncharacterized gene located in an amplified 5B–5D). This result is specific for cultures with high levels of
region that contains 23 other genes, including CBX4, which is TBC1D16, as the controls, WM262 and WM1346, grow at similar
known to play a role in cancer (Satijn et al., 1997). Expression rates to cultures infected with shGFP (75%–90%). As predicted,
of TBC1D16 is not highly correlated with TBC1D16 copy growth inhibition at day 8 is proportional to the amount of the
number compared to other genes in the region (ranked 7th out TBC1D16 transcript and is independent of TBC1D16 copy
of 24) or to all candidate drivers (252th out of 428). Nevertheless, number (Figures 5C and 5D). Taken together, these results
TBC1D16 is the top-scoring gene in the region and the second support CONEXIC’s prediction that TBC1D16 is required for
highest-scoring modulator, so it was selected for experimental proliferation in melanomas that overexpress the gene.
verification.
The module exhibits a dose-response relationship between RAB27A Identified and Experimentally Confirmed
TBC1D16 expression and the expression of genes in the module as a Tumor Dependency
such that higher expression of TBC1D16 is correlated with higher The TBC1D16 module contains a second modulator, RAB27A,
expression of genes in the module (correlation coefficient 0.76). also known to be involved in vesicular trafficking (Figure 5A).

1010 Cell 143, 1005–1017, December 10, 2010 ª2010 Elsevier Inc.
A TBC1D16 B
Exp WM262 WM1346
CNA 1400 1600
RAB27A DNA: Normal DNA: Normal
Exp Expression: Low Expression: Low

# Cells (in 1000)


CNA 1200

CONTROL
1000

TBC1D16 - sh302
TBC1D16 - sh1490 800
600 Control - shGFP

400
200

WM1960 WM1976
1200 250
DNA: Normal DNA:
Expression: High Expression: High

# Cells (in 1000)


200

800
150

TEST
100
400

50

0 2 4 6 8 0 2 4 6 8

C sh302 D

CNA:

Normal
Deleted
Expression:

-2 0 2
Log Change Time (days)
TBC1D16 transcript

Figure 5. TBC1D16 Is Necessary for Melanoma Growth


(A) A module associated with TBC1D16 and RAB27A. The genes in the module are involved in melanogenesis, survival/proliferation, lysosome, and protein traf-
ficking (see Table S4A for details).
(B) Representative growth curves for each of the four STCs infected with TBC1D16 shRNA. Each curve represents three technical replicates. RT-PCR was used to
confirm that the reduction in the amount of the TBC1D16 transcript was similar for all of the STCs (Figure S5).
(C) Change in growth over time, relative to the number of cells plated, averaged over all replicates (Extended Experimental Procedures). Mean over three bio-
logical replicates 3 three technical replicates for each STC. See Figure S5 and Table S4B for additional replicates and hairpins.
(D) Growth inhibition at 8 days is directly proportional to the amount of the TBC1D16 transcript and is independent of the TBC1D16 copy number.

RAB27A functions with RAB7A to control melanosome transport amplified) and WM1960 (38-fold higher expression, DNA not
and secretion. RAB7A localizes to early melanosomes, whereas amplified) and two controls that express RAB27A at a lower level
RAB27A is found in mature melanosomes (Jordens et al., 2006). (A375 and WM1930). Western blots show that expression of
CONEXIC selected both RAB27A and RAB7A as modulators. RAB27A correlates with expression of the cognate gene in these
RAB27A is in an amplified region that did not pass the standard cultures (data not shown).
GISTIC q value threshold for significance, and expression of Knockdown of RAB27A expression using shRNA was similar
the gene is not highly correlated with RAB27A copy number for all cultures (Figure S6) but only reduced cell growth signifi-
compared to other candidate drivers (323th out of 428). Never- cantly in the STCs that overexpress RAB27A (18% or 35% in
theless, CONEXIC identified it as the top-scoring modulator out WM1385 or WM1960 relative to the same cultures infected
of the 33 genes in this region and ranked it 8th out of 64 modu- with GFP shRNA). RAB27A shRNA had less impact (growth rates
lators, and it was therefore selected for empirical assessment. of 65%–80%) in the control STCs that have low RAB27A (Figures
To test the prediction that RAB27A is important for prolifera- 6A and 6B). Growth inhibition at 6 days is correlated with the
tion in tumors with high levels of RAB27A, we tested the effect amount of the RAB27A transcript and is independent of
of shRNA knockdown of the RAB27A transcript on proliferation. RAB27A copy number (Figures 6B and 6C). Taken together,
We chose two STCs in which the gene is highly expressed these results support CONEXIC’s prediction that RAB27A is
WM1385 (28-fold higher expression compared with A375, DNA a tumor dependency in melanomas that overexpress RAB27A.

Cell 143, 1005–1017, December 10, 2010 ª2010 Elsevier Inc. 1011
A A375 WM1930 Figure 6. RAB27A Is Necessary for Melanoma
1400 1000 Growth
DNA: Normal DNA: Normal (A) Representative growth curves for each of the four STCs
Expression: Low 800 Expression: Low infected with RAB27A shRNA. Each curve represents
# Cells (in 1000)
CONTROL

1000 three technical replicates. RT-PCR was used to confirm


600 that the reduction in the amount of the RAB27A transcript
RAB27A - sh865 was similar in all of the STCs (Figure S6).
600 RAB27A - sh477 (B) Change in growth over time, relative to the number of
400
Control - shGFP
cells plated, averaged over all replicates. Knockdown of
200 RAB27A expression in cells that express this gene at
200
high levels reduces proliferation. Data averaged over three
biological replicates 3 three technical replicates for each
STC. See Figure S6 and Table S5 for all data.
WM1960 WM1385
300 350 (C) Growth inhibition at 6 days is dependent on the amount
DNA: Normal DNA: High of the RAB27A transcript and is independent of RAB27A
250 Expression: High Expression: High copy number.
# Cells (in 1000)

250
200
TEST

150
150
100

50
We used LitVAN to identify the biological
50
processes and pathways represented among
the DEGs. Cell cycle-related terms are signifi-
0 2 4 6 0 2 4 6 cant among the downregulated genes, which
might be expected given the reduced growth
B 1.2 C after RAB27A knockdown. In addition, we found
sh865 sh865 that genes annotated for the ERK pathway
1
are upregulated (including MYC, FOSL1, and
0.8 DUSP6). We used GSEA to measure enrichment
0.6 of an experimentally derived set of genes that
respond to MEK inhibition in melanoma (Pratilas
0.4 A375
WM1930 et al., 2009). The resulting p value < 4.7 3 10 5
0.2 WM1385 suggests that ERK signaling is altered after
WM1960
RAB27A knockdown in these STCs.
0
0 2 4 6
TBC1D16 Influences the Expression
of Genes in Associated Modules
We carried out microarray profiling after knock-
down of TBC1D16 to evaluate whether expres-
RAB27A Affects the Expression of Genes sion of TBC1D16 affects the expression of genes in the four
in Associated Modules modules associated with it. We used two shRNAs to knock
To test whether RAB27A affects the expression of genes in asso- down TBC1D16 in the test STCs (WM1960 and WM1976) and
ciated modules, as predicted by CONEXIC, we carried out compared the gene expression to controls infected with GFP
microarray profiling after knockdown of RAB27A in the test shRNA (in the same STCs). GSEA analysis established that all
STCs (WM1385 and WM1960). We compared the expression four modules are significantly enriched for genes affected by
profile after RAB27A knockdown to a control profile generated differences in TBC1D16 expression (p values < 10 5, 0.0002,
by infecting the same STC with GFP shRNA. We used gene set 0.008, and 0.009, respectively; see Figure 7). Two modules
enrichment analysis (GSEA) (Subramanian et al., 2005) to test responded to TBC1D16 knockdown in the direction predicted
whether each of the three modules associated with RAB27A by CONEXIC. In addition, GSEA analysis ranked genes in
are enriched with genes that are differentially expressed (DEG) the TBC1D16 module (Module 25) highest out of 177 (based
after knockdown (see Extended Experimental Procedures). We on the GSEA p value), demonstrating that the genes in this
found that all three RAB27A-associated modules are signifi- module are the most highly differentially expressed genes in
cantly enriched for genes affected by RAB27A (p values < 10 5 the data set.
for all three modules; see Figure 7C) and that these modules The function of TBC1D16 is unknown, but it is predicted to be
responded in the direction predicted by CONEXIC. involved in vesicular trafficking. In our knockdown analysis,
These results support our computational prediction that the LitVAN annotated the upregulated genes with terms related to
expression of RAB27A affects the expression of the genes in vesicular trafficking. These include RAB3C, RAB7A, CHMP1B,
the associated modules. We note that RAB27A functions as a RAB18, SNX16, COPB1, and CAV1 (see Table S6A). However,
vesicular trafficking protein, suggesting that it influences gene it is not clear how TBC1D16 affects gene expression or how
expression through an unknown and likely indirect mechanism. changes in expression affect vesicular trafficking.

1012 Cell 143, 1005–1017, December 10, 2010 ª2010 Elsevier Inc.
A Figure 7. Results of Knockdown Microarrays for
RAB27A Low High
RAB27A and TBC1D16
(A) To the left is one of the modules associated with
RAB27A, and to the right are data generated following
knockdown (KD) of RAB27A for the same genes in the
STCs indicated (pink and blue). The expression of genes
in the module goes down relative to shGFP, as predicted.
KD expression heat map shows Z scores (see Extended
Experimental Procedures) showing that these are some
of the most differentially expressed genes (DEGs) in the

GSEA p-value <10


genome.
(B) To the left is one of the modules associated with
TBC1D16, and to the right are data generated following
KD of TBC1D16 in the STCs indicated. The expression
of genes in the module goes up relative to shGFP, as
2 2
predicted. The test STCs (blue) and control STCs (pink)
Log Change

Z-Score
respond differently, demonstrating the importance of
-2 -2 context (TBC1D16 overexpression status) in determining
Conexic Results - Module 3 RAB27A KD the response.
(C) GSEA p value and ranking (relative to 177 CONEXIC
modules) for RAB27A- and TBC1D16-associated modules
B TBC1D16 Low High (see Figure S7 for data). GSEA was calculated using the
median of four profiles (two cell lines 3 two hairpins) on
the test STCs. Significant p values indicate that knock-
down of RAB27A and TBC1D16 each affects the subset
of genes predicted by CONEXIC (note that 10 5 is the
smallest p value possible given that 100,000 permutations
are used). The color of the module name represents the
predicted direction of response to knockdown (red and
green represent up- and downregulated, respectively).
The arrow represents the observed response to knock-
down. The direction of response was correctly predicted
for two of four TBC1D16 modules and for all RAB27A
GSEA p-value 0.009

modules.
See also Figure S7 and Table S6.

and to identify those that are likely to be drivers.


2 2 The combination of data types allows us to iden-
Log Change

Z-Score

tify regions that would be overlooked using


-2 -2
methods based on DNA copy number alone.
Conexic Results - Module 127 TBC1D16 KD

C Expression of a Driver, Not Its Copy


Number, Drives Phenotype
RAB27A Moduled Modules TBC1D16 Moduled Modules
The novelty of our method and the key to its
Module GSEA p-value Rank Module GSEA p-value Rank success is our modeling paradigm: the expres-
Module 3 <10 3 Module 25 <10 1 sion of a driver should correspond with the
Module 75 0.008 21 expression of genes in an associated module.
Module 31 <10 2
Module 147 2x10 5 Examination of MITF and its targets supports
Module 75 <10 7 our assumptions. Expression of MITF best
Module 127 0.009 22
correlates with the expression of its targets,
but MITF overexpression does not always
correspond with MITF amplification. A change
DISCUSSION in DNA copy number is only one of many ways that gene expres-
sion can be altered. For example, MITF expression can be upre-
We have demonstrated that combining tumor gene expression gulated via signaling from the Ras/Raf (oncogenic BRAF occurs
and copy number data into a single framework increases our frequently in melanoma) (Wellbrock et al., 2008) and Frizzled/Wnt
ability to identify likely drivers in cancer and the processes pathways (Chin et al., 2006).
affected by them. Gene expression allows us to distinguish Most methods for identifying drivers within aberrant regions
between multiple genes in an amplified or deleted region focus on genes whose expression is well correlated with the
(many of which are indistinguishable based on copy number) copy number of the cognate DNA (Lin et al., 2008; Turner

Cell 143, 1005–1017, December 10, 2010 ª2010 Elsevier Inc. 1013
et al., 2010). The expression of many of the predicted drivers genetic context has a fundamental impact on the effect of
that we identify is poorly correlated with their copy number, rela- a perturbation.
tive to other genes in the region and to all other candidate
drivers MITF (294th), TBC1D16 (252th) and RAB27A (323th) Genes Involved in Trafficking Are Important
(see Table S1C). We believe that the discrepancies between in Melanoma
CNA and expression arise because there are multiple ways to Of the top 30 drivers selected by CONEXIC, three genes
up- or downregulate a gene. For example, TBC1D16 and (TBC1D16, RAB27A, and RAB7A) are known to be involved in
RAB27A were both identified as transcriptional targets of MITF vesicular trafficking (Itoh et al., 2006; Jordens et al., 2006). All of
(Chiaverini et al., 2008; Hoek et al., 2008b) and are therefore these genes are amplified (DNA) and highly expressed (RNA) in
upregulated when MITF is overexpressed. Moreover, we postu- multiple melanomas. There is increasing evidence that genes
late that many drivers are less correlated with their copy number controlling trafficking play a role in melanoma. Germline variation
than passengers due to selective pressure; if there is a fitness in Golgi phosphoprotein 3 (GOLPH3), a gene involved in vesicular
advantage to up- or downregulate expression, the tumor will trafficking, is associated with multiple cancers (Scott et al., 2009).
find a mechanism to do so. Our data identify two novel dependencies that are encoded
in somatic CNAs, demonstrate the dependency of melanoma
TBC1D16 and RAB27A Are Required for Proliferation on TBC1D16 and RAB27A expression for proliferation, and high-
We tested two drivers predicted by CONEXIC with knockdown light the potential role of vesicular trafficking in this malignancy.
experiments and showed that tumors that express either The role of vesicular trafficking in melanoma has yet to be
TBC1D16 or RAB27A at high levels are dependent on the corre- characterized. Vesicular trafficking regulates many receptor
sponding gene for growth. Our results demonstrate that these tyrosine kinases (RTKs) both spatially and temporally and thus
dependencies are determined by expression of the gene (in determines both the duration and intensity of signaling (Ying
both cases), rather than DNA amplification status, further sup- et al., 2010). For example, RAB7A is involved in the regulation
porting the assumptions underlying our approach. Thus, we of ERK signaling (Taub et al., 2007), and ERK is known to play
not only identify tumor dependencies, but also the tumors in an important role in melanoma (Chin et al., 2006). Tight control
which these genes are crucial for proliferation. Identifying depen- of ERK expression could potentially be important in melanocytes
dencies that are critical for tumor survival is needed for drug- because of its influence on MITF: ERK is required for the activa-
targeted therapies; for example, FLT3 inhibitors in AML, which tion of MITF, but high levels of ERK lead to MITF degradation
have had successful phase II trials (Fischer et al., 2010). Our (Wellbrock et al., 2008). It is possible that recurrent aberrations
approach is unbiased with respect to protein function and in vesicular trafficking genes might involve control of ERK
does not incorporate prior knowledge, thus enabling the identifi- signaling intensity. This is further supported by the upregulation
cation of dependencies in genes involved with vesicular traf- of an ERK signature (Pratilas et al., 2009) following RAB27A
ficking. TBC1D16 and RAB27A validate the ability of our knockdown in our data (p value < 4.7 3 10 5).
approach to correctly identify tumor dependencies and the
genes that they affect. CONEXIC and Other Approaches
CONEXIC differs from other methods in a number of ways. First,
Association between Modulator and Module it uses the gene expression of a candidate driver, rather than its
A key feature of our approach is that CONEXIC goes beyond copy number, as a proxy to report on the status of the gene, e.g.,
identifying drivers. By associating candidate drivers with gene two tumors that overexpress a driver are treated equivalently
modules and annotating them using information from the litera- even if there is amplification in the DNA of only one of them.
ture, CONEXIC provides insight into the physiological roles of Second, it associates a candidate driver with a module of genes
drivers and associated genes. We used LitVAn to find biological whose expression corresponds with that of the predicted driver,
processes and pathways overrepresented in each module and which was critical for identification of TBC1D16 as a modulator.
to associate drivers with functions, accurately identifying targets Third, combining copy number and gene expression provides
of MITF and annotating the functions of known drivers (MITF, greater sensitivity for identifying significantly aberrant regions
CCBN2, and TRAF3). that would not be selected based on DNA alone; this was critical
The results of microarray profiling following knockdown for the identification of RAB27A.
further support the association between modulator and module Methods based on copy number data are limited to detecting
and confirm our ability to identify genes affected by TBC1D16 large regions containing multiple genes, such that the driver
and RAB27A. We successfully connected genes involved in cannot be readily identified among them. Recent efforts have
vesicular trafficking to their effects on gene expression, likely focused on integrating additional sources of information into
through a cascade of indirect influences. In addition to profiling the analysis. Some methods use prior information, such as the
the STCs that highly express each of these genes (test STCs), role of a gene in other cancers (Beroukhim et al., 2010). Others,
we also profiled two lower-expressing STCs (control STCs), like CONEXIC, integrate gene expression data (Adler et al.,
in which the effect of knockdown is less detrimental to 2006), but the results of these methods fall short of CONEXIC’s.
growth. For TBC1D16, there is substantial overlap in the DEGs We systematically compared CONEXIC to other methods using
in the test STCs (p value < 6.6 3 10 22), but not in the DEGs the same data and found that they did not identify MITF or any
between control and test STCs (p value > 0.76). This reflects other known driver in melanoma (see Extended Experimental
the complexity of the transformed state and demonstrates that Procedures).

1014 Cell 143, 1005–1017, December 10, 2010 ª2010 Elsevier Inc.
Statistical dependencies in gene expression have been used sible for tumorigenesis and to find those that relate to any other
to connect a regulator to its target (Friedman et al., 2000; Lee measurable phenotype, such as the resistance of tumors to
et al., 2006; Segal et al., 2003) and for uncovering important drugs. We anticipate that our approach will make an important
regulators in cancer (Adler et al., 2006; Carro et al., 2010; contribution toward a basic mechanistic understanding of
Wang et al., 2009a). These approaches typically only detect tran- cancer and in revealing associations of clinical significance.
scription factors and signaling molecules and do not connect the Cancer is a heterogeneous disease in which we are only just
altered regulatory networks to upstream genetic aberrations. beginning to appreciate the importance of genetic background
Incorporating information on amplification or deletion status and the myriad ways in which the cellular machinery can be re-
allows us to consider any functional class of genes and thus directed toward the transformed state. Methods that begin to
permits detection of vesicular trafficking genes that would not dissect this complexity move us another step closer to a world
be identified using other methods. It also allows us to relate where personalized therapies are routine.
the malignant phenotype to genetic aberrations from which it is
likely to have originated. EXPERIMENTAL PROCEDURES
We tuned our method toward reducing the selection of modu-
lators that are not drivers. To gain this specificity, we do not Statistical Methods
A detailed description of the statistical methods and computational algorithms
detect all genes and pathways that drive tumors. First, some
used can be found in the Extended Experimental Procedures. The CONEXIC
drivers in amplified and deleted regions do not pass the stringent and LitVAN algorithms were developed for this research, and the software is
statistical tests employed in our method. Second, CONEXIC only available at http://www.c2b2.columbia.edu/danapeerlab/html/software.html.
identifies candidate drivers that are encoded in amplified or
deleted regions. In consequence, it would not detect drivers of Experimental Methods
melanoma such as BRAF and NRAS that are typically associated Cells were grown using standard culture conditions, and knockdown was
with point mutations. Third, CONEXIC detects drivers based on carried out by infection with lentivirus using RNAi sequences designed by
the RNAi Consortium. shRNA lentivirus were prepared according to TRC
the assumptions delineated above; though these hold for many
protocols (http://www.broadinstitute.org/rnai/trc), with minor modifications.
drivers, it is likely that they are not appropriate for all drivers. Cell proliferation assays, RT-PCR, microarrays, and immunoblotting were
To meet the challenge of finding all driving alterations in carried out using standard techniques. Primer sequences and detailed
cancer, a number of complementary approaches are needed. methods can be found in the Extended Experimental Procedures.
Experimental approaches such as screening using pooled short
hairpin RNAs (shRNAs) (Bric et al., 2009; Zender et al., 2008) are ACCESSION NUMBERS
likely to detect a set of drivers different from those detected by
CONEXIC. These screens are dependent on the genetic back- All primary data are available at the Gene Expression Omnibus (GSE23884).
ground and are limited to drivers that influence processes that
can be readily measured, such as proliferation, whereas SUPPLEMENTAL INFORMATION

CONEXIC scans all of the genetic data together and can poten-
Supplemental Information includes Extended Experimental Procedures, eight
tially identify drivers of any function across different genetic figures, and six tables and can be found with this article online at doi:10.1016/
backgrounds. In the future, we envision that CONEXIC will be j.cell.2010.11.013.
used to guide in vivo screening initiatives and to assist in the
choice of regions, functional assays, and genetic backgrounds ACKNOWLEDGMENTS
probed.
The authors will like to thank Nir Hacohen, Antonio Iavarone, Daphne Koller, Liz
Miller, Itsik Pe’er, Suzanne Pfeffer, Neal Rosen, and Olga Troyanskaya for
Beyond Melanoma
valuable comments. This research was supported by the National Institutes
The challenge of finding candidate drivers is considerable: of Health Roadmap Initiative, NIH Director’s New Innovator Award Program
tumors are heterogeneous, the data are noisy and highly corre- through grant number 1-DP2-OD002414-01, and National Centers for
lated, and there are a large number of possible combinations Biomedical Computing Grant 1U54CA121852-01A1. D.P. holds a Career
of drivers and genes in modules. Our approach is successful Award at the Scientific Interface from the Burroughs Wellcome Fund and
because it couples simple modeling assumptions with powerful Packard Fellowship for Science and Engineering.
computational search techniques and rigorous statistical evalu-
Received: May 13, 2010
ation of the results at each step.
Revised: August 31, 2010
Both the principles underlying CONEXIC and the software can Accepted: October 22, 2010
be applied to any tumor cohort containing matched data for Published online: December 2, 2010
copy number aberrations and gene expression. The principle
of associating any type of mutation (e.g., epigenetic alterations REFERENCES
and coding sequence) with gene expression signatures or
Adler, A.S., Lin, M., Horlings, H., Nuyten, D.S., van de Vijver, M.J., and Chang,
other phenotypic outputs that differ among samples will be of
H.Y. (2006). Genetic regulators of large-scale transcriptional signatures in
increasing importance as sequence and epigenetic data accu- cancer. Nat. Genet. 38, 421–430.
mulate. Not only does this help to distinguish between driver
Beroukhim, R., Getz, G., Nghiemphu, L., Barretina, J., Hsueh, T., Linhart, D.,
and passenger mutations, but the genes in the associated Vivanco, I., Lee, J.C., Huang, J.H., Alexander, S., et al. (2007). Assessing the
module can also provide insight into the role of the driver. This significance of chromosomal aberrations in cancer: methodology and applica-
approach can be used to identify the genetic aberrations respon- tion to glioma. Proc. Natl. Acad. Sci. USA 104, 20007–20012.

Cell 143, 1005–1017, December 10, 2010 ª2010 Elsevier Inc. 1015
Beroukhim, R., Brunet, J.-P., Di Napoli, A., Mertz, K.D., Seeley, A., Pires, M.M., Jordens, I., Westbroek, W., Marsman, M., Rocha, N., Mommaas, M., Huizing,
Linhart, D., Worrell, R.A., Moch, H., Rubin, M.A., et al. (2009). Patterns of gene M., Lambert, J., Naeyaert, J.M., and Neefjes, J. (2006). Rab7 and Rab27a
expression and copy-number alterations in von-hippel lindau disease- control two motor protein activities involved in melanosomal transport.
associated and sporadic clear cell carcinoma of the kidney. Cancer Res. 69, Pigment Cell Res. 19, 412–423.
4674–4681. Lee, S.-I., Pe’er, D., Dudley, A.M., Church, G.M., and Koller, D. (2006). Identi-
Beroukhim, R., Mermel, C.H., Porter, D., Wei, G., Raychaudhuri, S., Donovan, fying regulatory mechanisms using individual variation reveals key role for
J., Barretina, J., Boehm, J.S., Dobson, J., Urashima, M., et al. (2010). The land- chromatin modification. Proc. Natl. Acad. Sci. USA 103, 14062–14067.
scape of somatic copy-number alteration across human cancers. Nature 463, Levy, C., Khaled, M., and Fisher, D.E. (2006). MITF: master regulator of
899–905. melanocyte development and melanoma oncogene. Trends Mol. Med. 12,
Bric, A., Miething, C., Bialucha, C.U., Scuoppo, C., Zender, L., Krasnitz, A., 406–414.
Xuan, Z., Zuber, J., Wigler, M., Hicks, J., et al. (2009). Functional identification Lin, W.M., Baker, A.C., Beroukhim, R., Winckler, W., Feng, W., Marmion, J.M.,
of tumor-suppressor genes through an in vivo RNA interference screen in Laine, E., Greulich, H., Tseng, H., Gates, C., et al. (2008). Modeling genomic
a mouse lymphoma model. Cancer Cell 16, 324–335. diversity and tumor dependency in malignant melanoma. Cancer Res. 68,
Carro, M.S., Lim, W.K., Alvarez, M.J., Bollo, R.J., Zhao, X., Snyder, E.Y., 664–673.
Sulman, E.P., Anne, S.L., Doetsch, F., Colman, H., et al. (2010). The transcrip- Pearl, J. (2000). Causality: models, reasoning, and inference (Cambridge, U.K.;
tional network for mesenchymal transformation of brain tumours. Nature 463, New York: Cambridge University Press).
318–325. Pinkel, D., Segraves, R., Sudar, D., Clark, S., Poole, I., Kowbel, D., Collins, C.,
Chiaverini, C., Beuret, L., Flori, E., Busca, R., Abbe, P., Bille, K., Bahadoran, P., Kuo, W.-L., Chen, C., Zhai, Y., et al. (1998). High resolution analysis of DNA
Ortonne, J.-P., Bertolotto, C., and Ballotti, R. (2008). Microphthalmia-associ- copy number variation using comparative genomic hybridization to microar-
ated transcription factor regulates RAB27A gene expression and controls rays. Nat. Genet. 20, 207–211.
melanosome transport. J. Biol. Chem. 283, 12635–12642. Pleasance, E.D., Cheetham, R.K., Stephens, P.J., McBride, D.J., Humphray,
Chin, L., Garraway, L.A., and Fisher, D.E. (2006). Malignant melanoma: S.J., Greenman, C.D., Varela, I., Lin, M.-L., Ordóñez, G.R., Bignell, G.R.,
genetics and therapeutics in the genomic era. Genes Dev. 20, 2149–2182. et al. (2010). A comprehensive catalogue of somatic mutations from a human
cancer genome. Nature 463, 191–196.
Du, J., Widlund, H.R., Horstmann, M.A., Ramaswamy, S., Ross, K., Huber,
W.E., Nishimura, E.K., Golub, T.R., and Fisher, D.E. (2004). Critical role of Pratilas, C.A., Taylor, B.S., Ye, Q., Viale, A., Sander, C., Solit, D.B., and Rosen,
CDK2 for melanoma growth linked to its melanocyte-specific transcriptional N. (2009). (V600E)BRAF is associated with disabled feedback inhibition of
regulation by MITF. Cancer Cell 6, 565–576. RAF-MEK signaling and elevated transcriptional output of the pathway.
Proc. Natl. Acad. Sci. USA 106, 4519–4524.
Fischer, T., Stone, R.M., Deangelo, D.J., Galinsky, I., Estey, E., Lanza, C., Fox,
Sanchez-Garcia, F., Akavia, U.D., Mozes, E., and Pe’er, D. (2010). JISTIC:
E., Ehninger, G., Feldman, E.J., Schiller, G.J., et al. (2010). Phase IIB trial of oral
identification of significant targets in cancer. BMC Bioinformatics 11, 189.
Midostaurin (PKC412), the FMS-like tyrosine kinase 3 receptor (FLT3) and
multi-targeted kinase inhibitor, in patients with acute myeloid leukemia and Satijn, D.P., Olson, D.J., van der Vlag, J., Hamer, K.M., Lambrechts, C.,
high-risk myelodysplastic syndrome with either wild-type or mutated FLT3. Masselink, H., Gunster, M.J., Sewalt, R.G., van Driel, R., and Otte, A.P.
J. Clin. Oncol. 28, 4339–4345. (1997). Interference with the expression of a novel human polycomb protein,
hPc2, results in cellular transformation and apoptosis. Mol. Cell. Biol. 17,
Friedman, N., Linial, M., Nachman, I., and Pe’er, D. (2000). Using Bayesian
6076–6086.
networks to analyze expression data. J. Comput. Biol. 7, 601–620.
Scott, K.L., Kabbarah, O., Liang, M.-C., Ivanova, E., Anagnostou, V., Wu, J.,
Garraway, L.A., Widlund, H.R., Rubin, M.A., Getz, G., Berger, A.J., Ramasw- Dhakal, S., Wu, M., Chen, S., Feinberg, T., et al. (2009). GOLPH3 modulates
amy, S., Beroukhim, R., Milner, D.A., Granter, S.R., Du, J., et al. (2005). Integra- mTOR signalling and rapamycin sensitivity in cancer. Nature 459, 1085–1090.
tive genomic analyses identify MITF as a lineage survival oncogene amplified
Segal, E., Shapira, M., Regev, A., Pe’er, D., Botstein, D., Koller, D., and
in malignant melanoma. Nature 436, 117–122.
Friedman, N. (2003). Module networks: identifying regulatory modules and
Golub, T.R., Slonim, D.K., Tamayo, P., Huard, C., Gaasenbeek, M., Mesirov, their condition-specific regulators from gene expression data. Nat. Genet.
J.P., Coller, H., Loh, M.L., Downing, J.R., Caligiuri, M.A., et al. (1999). Molec- 34, 166–176.
ular classification of cancer: class discovery and class prediction by gene
Segal, E., Friedman, N., Koller, D., and Regev, A. (2004). A module map
expression monitoring. Science 286, 531–537.
showing conditional activity of expression modules in cancer. Nat. Genet.
Hoek, K.S., Eichhoff, O.M., Schlegel, N.C., Döbbeling, U., Kobert, N., 36, 1090–1098.
Schaerer, L., Hemmi, S., and Dummer, R. (2008a). In vivo switching of human
Steingrı́msson, E., Copeland, N.G., and Jenkins, N.A. (2004). Melanocytes
melanoma cells between proliferative and invasive states. Cancer Res. 68,
and the microphthalmia transcription factor network. Annu. Rev. Genet. 38,
650–656.
365–411.
Hoek, K.S., Schlegel, N.C., Eichhoff, O.M., Widmer, D.S., Praetorius, C., Stratton, M.R., Campbell, P.J., and Futreal, P.A. (2009). The cancer genome.
Einarsson, S.O., Valgeirsdottir, S., Bergsteinsdottir, K., Schepsky, A., Nature 458, 719–724.
Dummer, R., and Steingrimsson, E. (2008b). Novel MITF targets identified
Subramanian, A., Tamayo, P., Mootha, V.K., Mukherjee, S., Ebert, B.L.,
using a two-step DNA microarray strategy. Pigment Cell Melanoma Res 21,
Gillette, M.A., Paulovich, A., Pomeroy, S.L., Golub, T.R., Lander, E.S., and
665–676.
Mesirov, J.P. (2005). Gene set enrichment analysis: a knowledge-based
Hoek, K.S., Schlegel, N.C., Brafford, P., Sucker, A., Ugurel, S., Kumar, R., approach for interpreting genome-wide expression profiles. Proc. Natl.
Weber, B.L., Nathanson, K.L., Phillips, D.J., Herlyn, M., et al. (2006). Metastatic Acad. Sci. USA 102, 15545–15550.
potential of melanomas defined by specific gene expression profiles with no
Taub, N., Teis, D., Ebner, H.L., Hess, M.W., and Huber, L.A. (2007). Late endo-
BRAF signature. Pigment Cell Res. 19, 290–302.
somal traffic of the epidermal growth factor receptor ensures spatial and
Itoh, T., Satoh, M., Kanno, E., and Fukuda, M. (2006). Screening for target temporal fidelity of mitogen-activated protein kinase signaling. Mol. Biol. Cell
Rabs of TBC (Tre-2/Bub2/Cdc16) domain-containing proteins based on their 18, 4698–4710.
Rab-binding activity. Genes Cells 11, 1023–1037. Turner, N., Lambros, M.B., Horlings, H.M., Pearson, A., Sharpe, R., Natrajan,
Johansson, P., Pavey, S., and Hayward, N. (2007). Confirmation of a BRAF R., Geyer, F.C., van Kouwenhove, M., Kreike, B., Mackay, A., et al. (2010). Inte-
mutation-associated gene expression signature in melanoma. Pigment Cell grative molecular profiling of triple negative breast cancers identifies amplicon
Res. 20, 216–221. drivers and potential therapeutic targets. Oncogene 29, 2013–2023.

1016 Cell 143, 1005–1017, December 10, 2010 ª2010 Elsevier Inc.
Vallabhapurapu, S., Matsuzawa, A., Zhang, W., Tseng, P.-H., Keats, J.J., Wellbrock, C., Rana, S., Paterson, H., Pickersgill, H., Brummelkamp, T., and
Wang, H., Vignali, D.A.A., Bergsagel, P.L., and Karin, M. (2008). Nonredundant Marais, R. (2008). Oncogenic BRAF regulates melanoma proliferation through
and complementary functions of TRAF2 and TRAF3 in a ubiquitination the lineage specific factor MITF. PLoS ONE 3, e2734.
cascade that activates NIK-dependent alternative NF-kappaB signaling. Nat.
Immunol. 9, 1364–1370. Ying, H., Zheng, H., Scott, K., Wiedemeyer, R., Yan, H., Lim, C., Huang, J.,
Dhakal, S., Ivanova, E., Xiao, Y., et al. (2010). Mig-6 controls EGFR
Wang, K., Saito, M., Bisikirska, B.C., Alvarez, M.J., Lim, W.K., Rajbhandari, P.,
trafficking and suppresses gliomagenesis. Proc. Natl. Acad. Sci. USA 107,
Shen, Q., Nemenman, I., Basso, K., Margolin, A.A., et al. (2009a). Genome-
6912–6917.
wide identification of post-translational modulators of transcription factor
activity in human B cells. Nat. Biotechnol. 27, 829–839. Zender, L., Xue, W., Zuber, J., Semighini, C.P., Krasnitz, A., Ma, B., Zender, P.,
Wang, L., Cao, X.X., Chen, Q., Zhu, T.F., Zhu, H.G., and Zheng, L. (2009b). Kubicka, S., Luk, J.M., Schirmacher, P., et al. (2008). An oncogenomics-based
DIXDC1 targets p21 and cyclin D1 via PI3K pathway activation to promote in vivo RNAi screen identifies tumor suppressors in liver cancer. Cell 135,
colon cancer cell proliferation. Cancer Sci. 100, 1801–1808. 852–864.

Cell 143, 1005–1017, December 10, 2010 ª2010 Elsevier Inc. 1017
Resource

Comprehensive Polyadenylation Site


Maps in Yeast and Human Reveal
Pervasive Alternative Polyadenylation
Fatih Ozsolak,1,* Philipp Kapranov,1 Sylvain Foissac,2 Sang Woo Kim,3 Elane Fishilevich,3 A. Paula Monaghan,4
Bino John,3 and Patrice M. Milos1,*
1HelicosBioSciences Corporation, Cambridge, MA 02139, USA
2Integromics,Madrid 28760, Spain
3Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA
4Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15260, USA

*Correspondence: fatihozsolak@gmail.com (F.O.), pmilos@helicosbio.com (P.M.M.)


DOI 10.1016/j.cell.2010.11.020

SUMMARY predictions relying on polyadenylation-associated motif


elements (Graber et al., 2002; Lutz, 2008; Tian et al., 2005). EST
The emerging discoveries on the link between polya- databases are valuable but insufficient for in-depth mapping of
denylation and disease states underline the need to polyadenylation sites due to data quality problems, such as low
fully characterize genome-wide polyadenylation numbers of full-length ESTs, chimeric sequences (due to cDNA
states. Here, we report comprehensive maps of global template switching; Cocquet et al., 2006), internal cDNA priming
polyadenylation events in human and yeast generated events leading to cloning of incomplete transcripts, and low-
quality sequences at the ends of ESTs (Zhang et al., 2005a,
using refinements to the Direct RNA Sequencing tech-
2005b). For applications requiring identification of polyadenyla-
nology. This direct approach provides a quantitative
tion site usage frequency changes across biological conditions,
view of genome-wide polyadenylation states in EST databases, motif searches, and classic polyadenylation
a strand-specific manner and requires only attomole site mapping approaches (Slomovic et al., 2008), such as
RNA quantities. The polyadenylation profiles revealed RACE, RT-PCR, and nuclease sensitivity assays, do not provide
an abundance of unannotated polyadenylation sites, the required simplicity, sensitivity, depth, and quantitative
alternative polyadenylation patterns, and regulatory genome-wide view. Annotation of the 30 ends of yeast genes
element-associated poly(A)+ RNAs. We observed were attempted previously with RNA-seq (Nagalakshmi et al.,
differences in sequence composition surrounding 2008) and microarray-based (David et al., 2006) approaches,
canonical and noncanonical human polyadenylation but these studies did not have sufficient resolution to map indi-
sites, suggesting novel noncoding RNA-specific poly- vidual cleavage sites for polyadenylation. Furthermore, despite
much interest devoted to overlapping transcription, we still do
adenylation mechanisms in humans. Furthermore, we
not have a complete understanding of sense/antisense transcrip-
observed the correlation level between sense and
tion (reviewed by Faghihi and Wahlestedt, 2009). To date, our
antisense transcripts to depend on gene expression knowledge in this area comes from methods relying on reverse
levels, supporting the view that overlapping transcrip- transcription that suffers from spurious second-strand cDNA
tion from opposite strands may play a regulatory role. products (Gubler, 1987; Spiegelman et al., 1970), complicating
Our data provide a comprehensive view of the polya- analyses requiring unambiguous determination of RNA strand.
denylation state and overlapping transcription. Although methods have recently been developed that preserve
the RNA strand information through RNA-level modifications,
such as bisulfite treatment or RNA-level adaptor ligation (He
INTRODUCTION et al., 2008; Mamanova et al., 2010), these still rely on cDNA
synthesis, ligation, and amplification steps that may introduce
The known regulatory role of 30 untranslated regions (30 UTRs) and artifacts and complicate the quantitation of various RNA species.
poly(A) tails in mRNA localization, stability, and translation (re- To avoid the known biases and artifacts introduced to RNA
viewed by Andreassi and Riccio, 2009), and polyadenylation measurements during reverse transcription (Cocquet et al.,
regulation defects leading to human diseases such as oculophar- 2006; Liu and Graber, 2006; Mamanova et al., 2010; Wu et al.,
yngeal muscular dystrophy, thalassemias, thrombophilia, and 2008) or other sample manipulation steps, we recently devel-
IPEX syndrome (Bennett et al., 2001; Brais et al., 1998; Gehring oped the direct RNA sequencing (DRS) technology (Ozsolak
et al., 2001; Higgs et al., 1983; Lin et al., 1998; Orkin et al., et al., 2009). DRS sequences RNA molecules in a massively
1985) underscores the need to fully characterize polyadenylation parallel manner without its prior conversion to cDNA or the
sites and mechanisms. Our knowledge in this area primarily orig- need for biasing ligation or amplification steps. Since this
inates from expressed sequence tag (EST) databases and proof-of-concept study, we have improved and adapted DRS

1018 Cell 143, 1018–1029, December 10, 2010 ª2010 Elsevier Inc.
for use with the Helicos Genetic Analysis System. DRS produces Genome-wide 30 Polyadenylation State in Yeast
alignable reads up to 55 nt (mean read length, 33–34 nt). Unlike We obtained 7,036,730 DRS reads uniquely aligned to the yeast
other RNA analysis approaches, which require multiple nucleic genome, each read representing a polyadenylation site of an
acid manipulation steps, DRS only requires polyadenylated independent transcript, to deduce the yeast polyadenylation
and 30 blocked RNA templates for sequencing. landscape (Table S2). To verify our findings, we compared the
We here applied DRS to generate a comprehensive and high- polyadenylation sites identified here to the sites identified previ-
resolution map of polyadenylation sites of human and yeast ously for 11 yeast genes using classic approaches, observing
transcripts. Using multiple independent approaches, we vali- high overlap (Figure 1B). Because of its higher resolution, DRS
dated our findings and demonstrated the usefulness of the found the frequently used cleavage locations reported previ-
approach to identify alternative polyadenylation events. We ously and other generally lower-frequency cleavage positions
observed many unannotated polyadenylation sites and novel (Figure 1A and Figure S1B). In addition, DRS data agreed well
RNA species associated with open chromatin sites that may with the polyadenylation sites mapped previously for ten genes
function to regulate gene expression. We also observed that and seven snoRNAs using PCR amplification of 30 transcript
sequence and motif contexts surrounding novel intergenic and ends in a manner that preserves the variability in the 30 ends,
genic sense/antisense polyadenylation sites away from 30 ends followed by high-throughput DNA sequencing of the RT-PCR
of known genes exhibit significant differences than sequence products (Ozsolak et al., 2009). Furthermore, we validated four
and motif contexts surrounding polyadenylation sites near previously unannotated intergenic and genic polyadenylation
known gene 30 ends. This observation suggests alternative locations using cloning and RACE approaches (Figure S1C and
mechanisms and/or purposes of RNA polyadenylation. In addi- Table S3). We also compared DRS reads to the 60,218 30 end
tion, we have examined overlapping transcription patterns of tags, which constitute 0.2% of RNA-seq reads, are analogous
poly(A)+ transcripts. Between the steady-state quantities of to DRS reads and mark yeast polyadenylation sites (Naga-
sense and antisense transcripts, we observed a complex corre- lakshmi et al., 2008), observing 53,849 (89.4%) of end tags to
lation pattern that depends on gene expression levels. be within 5 nt of DRS read start locations. The difference
observed in the remaining 10% may be due to differences in
RESULTS the resolution of both methods, different yeast strains and RNA
preparation approaches used in both studies.
Mapping Global 30 Polyadenylation Sites with DRS The median length of the 30 UTRs of 5759 yeast open reading
To determine polyadenylation locations, 200–300 picograms of frames (ORFs) was 166 nt (Figure 2A and Table 1). With the
human liver and yeast poly(A)+ RNAs, and 3 ng of human brain number of reads and depth we generated for this study, we
total RNA blocked at their 30 ends were used per sequencing observed that 72.1% of the yeast genes exhibited polyadenyla-
channel. Given that poly(A)+ RNA species already contain tion locations separated by at least 50 nt, and frequently more,
a natural poly(A) tail, additional polyadenylation was not needed. and thus have multiple polyadenylation sites. The higher levels
After the capture of poly(A)+ RNA species on poly(dT)-coated flow observed here relative to the 10%–15% level reported previously
cell surfaces by hybridization, a ‘‘fill’’ step with natural dTTP and (Nagalakshmi et al., 2008) may be due to the higher resolution of
a ‘‘lock’’ step with fluorescently labeled proprietary Virtual Termi- the approach presented here and the higher number of tran-
nator (VT)-A, -C, and -G nucleotides were performed. These scripts analyzed. Similar to previous reports (Nagalakshmi
steps correct for any misalignments that may be present in poly et al., 2008), we observed 14% of genes to be orientated in
(A/T) duplexes and ensure that the sequencing starts in the tail-to-tail orientation and have overlapping 30 ends (see below).
template rather than the poly(A) tail. After the completion of fill Fourteen percent of yeast DRS reads mapped to regions within
and lock steps, DRS was initiated. The 50 ends of DRS reads the yeast ORFs either in exons or introns (Table 1). Intronic poly-
signify cleavage locations. The resolution for identification of adenylation sites are possibly due to a dynamic interplay
the polyadenylation cleavage nucleotide is dependent on fill between splicing and polyadenylation (Tian et al., 2007) and
and lock efficiency and the ability of the sequencing reaction to may represent transcripts encoding shorter proteins.
start immediately upstream of the poly(A) tails. We measured 10.6% of yeast DRS reads did not map downstream of anno-
this efficiency using polyadenylated oligoribonucleotides and tated yeast 30 ends or within the ORFs. To examine the degree of
determined the resolution to be ±2 nt (see Figure S1A available association of yeast poly(A)+ transcripts with regulatory regions,
online). To determine whether our results might have been nega- we took advantage of the regulatory protein binding sites defined
tively affected by potential internal priming events, we performed recently by DNase I hypersensitive site (DHS) mapping (Hessel-
experiments to observe the sequencing behavior of templates berth et al., 2009). We observed a significant enrichment of diver-
containing internal poly(A) stretches with 30 noncomplementary gent transcripts (e.g., transcribed away from DHSs) in regions
overhangs and examined the fraction of polyadenylation regions that are in proximity to intergenic DHSs (p = 8.041e-07, nonpara-
containing downstream poly(A)-rich regions. We observed rare or metrical two-sample Kolmogorov-Smirnov test) (Figure S2).
no occurrence of internal priming events (Table S1 and Extended
Experimental Procedures). Thus, the technology is capable of Genome-wide 30 Polyadenylation State in Humans
mapping the extensive 30 end heterogeneity we and others (Iseli A total of 11,882,580 uniquely mapping reads were obtained
et al., 2002; Lopez et al., 2006; Muro et al., 2008) observed in from human liver poly(A)+ RNA, of which 1,322,970 were derived
the majority of yeast and human genes in a genome-wide manner from mitochondria and 2,570 reads from rRNA. This is consistent
and at nucleotide resolution (Figures 1A–1D). with the observations that human mitochondrial transcripts and

Cell 143, 1018–1029, December 10, 2010 ª2010 Elsevier Inc. 1019
A B
300 5’ ends of DRS reads corresponding DRS, same direction as HIS3
250 to (+) strand transcripts 50

40
200 30

20
150
10
100
0

50
5

4
Nagalakshmi et al, same direction as HIS3
3
HIS3 (+) 2

722,500 722,550 722,600 722,650 722,700 722,750 722,800 722,850 722,900 0

722,660 722,680 722,700 722,720 722,740


300
5’ ends of DRS reads corresponding
250
to (-) strand transcripts 50
Mahadevan et al. sites
200 DRS, opposite direction of HIS3
40

150 30

20
100
10
50 0

Nagalakshmi et al, opposite direction of HIS3


5

3
2

C D
100 5’ ends of DRS reads corresponding
4000 5’ ends of DRS reads corresponding 90
to (+) strand transcripts
to (+) strand transcripts 80
3000 70
60
50
2000 40
30
1000 20
10

70,380,000 70,380,200 70,380,400 70,380,600 70,380,800 70,381,000


70,380,000 70,380,200 70,380,400 70,380,600 70,380,800 70,381,000 100
5’ ends of DRS reads corresponding
90
4000 5’ ends of DRS reads corresponding 80 to (-) strand transcripts
70
to (-) strand transcripts 60
3000 50
40
2000 30
20
10
1000

UGT2B4(-)
UGT2B4(-)

Figure 1. Polyadenylation Site Detection in Yeast and Human


(A) The blue and black panels show the DRS reads emanating from transcripts in the + and – direction, respectively. The major peaks in the blue panel correspond
to the 13 polyadenylation sites at locations 722690, 722692, 722695, 722710, 722716, 722718, 722723, 722726, 722746, 722750, 722752, 722775, and 722777
previously identified for HIS3 (Mahadevan et al., 1997) using 30 RACE-PCR.
(B) Zoomed-in view of (A). y axis was reduced from 0–300 scale to 0–50. x axis was reduced from 722,500–722,900 scale to 722,660–722,740. All ‘‘end tags’’
identified by Nagalakshmi et al. (2008) in this region are also shown (y axis for these tags is on the scale of 0–5). Arrows mark the sites identified by Mahadevan
et al. (1997) in the region shown.
(C and D) Overview (B) and a zoomed-in view (C) of reads mapping to UGT2B4 30 annotated ends. Multiple potential polyadenylation sites are evident in panel C
(see also Figure S2 and Table S1).

a fraction of rRNAs are polyadenylated, perhaps for the regions that are at least 5 kb away from known genes and thus
purposes of degradation (Nagaike et al., 2005; Slomovic et al., likely to represent novel RNAs; 37% of intergenic reads in hu-
2010); 56.1% of DRS reads overlapped with 19,871 of 28,858 mans are within 5 kb of known transcripts, and 42% are within
polyadenylation sites previously annotated using EST databases 10 kb. Thus, a considerable fraction of intergenic reads are in
and motif searches (Zhang et al., 2005a). The differences proximity to known genes (van Bakel et al., 2010). An additional
observed may be due to the single tissue examined here, 14.7% of reads fall within introns on either strand. Polyadenyla-
whereas EST database searches include data from multiple tion events near the 30 ends of known genes tend to happen more
tissue types. More than half (55.7%) of liver DRS reads emerged frequently in 30 UTR regions rather than the region immediately
from within 10 nt of annotated 30 ends of UCSC Genes (Figure 2B, downstream of the 30 ends of genes (Table 1 and Figure S3A).
Figure S3A, and Table 1). The remaining 44.3% of the reads This may be caused by degradation intermediates of prema-
represent either novel RNAs or alternative polyadenylation sites turely terminated transcripts, or the 30 end annotations gener-
of known mRNAs. Although estimation of the extent of noncod- ated from EST databases favoring more downstream polyade-
ing transcription based on this data is difficult because the full nylation locations over upstream ones due to concerns such
structures of transcripts represented by the DRS reads are not as incomplete cDNA clones and sequences, and thus, underre-
known, at the very least, 9% of reads are located in intergenic presenting the diversity of polyadenylation sites.

1020 Cell 143, 1018–1029, December 10, 2010 ª2010 Elsevier Inc.
70,736,000 70,738,000 70,740,000 70,742,000 70,744,000 70,746,000
A 4 C
5’ ends of DRS reads from brain corresponding
Fraction of DRS reads (%)

400 to (-) strand transcripts


300
3
200 A3 A2 A1
100
2 0

400
5’ ends of DRS reads from liver corresponding
1 300 to (-) strand transcripts
200
100
0
0 200 400 600 800 0 3’ ends of ADD2 (-) from UCSC Genes
Distance to annotated 3' ends of
S. cerevisiae ORFs (bps)
B D
5’ ends of DRS reads from brain corresponding
100
to (+) strand transcripts
25
Fraction of DRS reads (%)

80
60 A1 A2
40
20
20
0
15
5’ ends of DRS reads from liver corresponding
to (+) strand transcripts
10 100
80
5 60
40
20
0 0
-1000 -500 0 500 1000
Distance to annotated 3' ends of 3’ end of BBOX1 (+) from UCSC Genes
human known genes (bps)
27,105,650 27,105,700 27,105,750 27,105,800 27,105,850 27,105,900

Figure 2. Characteristics of Polyadenylation Sites in Yeast and Human


(A and B) Y-axes indicate the fraction of DRS reads aligning at x-distances (in 10 bp bins) relative to the annotated 30 ends of yeast ORFs (A) and the annotated 30
ends of human UCSC genes (B).
(C and D) ADD2 (C) and BBOX1 (D) polyadenylation sites in human liver and brain. The polyadenylation sites identified (indicated as A1, A2 ,and A3) for both genes
agree well with previous findings (Costessi et al., 2006; Rigault et al., 2006) (see also Figure S3 and Table S3).

To exemplify the ability of DRS to identify alternative polyade- 41.2% and the ORFs with antisense transcripts increased to
nylation events, we profiled human brain total RNA. ADD2 4641 (80.4%), in part due to the genes with overlapping 30 ends.
mRNAs were found to have one major and additional minor poly- In the human liver RNA, at least 19,680/65,260 (30.2%) of all
adenylation sites in brain but none in liver (Figure 2C), as reported annotated transcripts were found to have antisense transcription
previously (Costessi et al., 2006). In addition, in concordance as defined by at least 10 antisense reads either in exons or
with previous results (Rigault et al., 2006), we observed two poly- introns (Figures S3D and S3E). Although prevalent, the antisense
adenylation sites for BBOX1 and a higher quantity of the ‘‘short’’ transcription is still a minority in terms of transcript abundance:
versus the ‘‘long’’ transcript in both tissues (Figure 2D). 8% of all reads that overlap an annotated transcript are anti-
sense to it. This number is similar to the 11% reported previously
(He et al., 2008). Importantly, these numbers were obtained from
Sense/Antisense Poly(A)+ Transcripts in Yeast and
poly(A)+ RNA and do not represent the extent of poly(A) anti-
Human
sense transcription (Dutrow et al., 2008; Kiyosawa et al., 2005).
DRS can not only pinpoint the sites of sense and antisense tran-
scription, but also provide quantification of such transcripts
without biases introduced by steps such as ligation, amplifica- Quantification of Sense/Antisense Poly(A)+
tion, and other manipulations. Of the 5769 annotated yeast Transcriptome
ORFs, at least 3492 (60.5%) had an antisense transcript, as evi- We then explored the correlation between the quantities of sense
denced by at least 10 antisense reads within the annotated ORFs and antisense transcripts. This analysis was attempted to
(Figures S3B and S3C). These antisense reads compose 9.2% of observe the relationship between sense and antisense tran-
the total DRS reads. When we considered the ambiguity in yeast scripts encoded by the same genomic region, given the pres-
30 end annotations and included regions 200 nt downstream of ence of certain biological constraints such as transcription
the 30 annotation, the fraction of antisense reads increased to in both directions in a locus and pathways degrading

Cell 143, 1018–1029, December 10, 2010 ª2010 Elsevier Inc. 1021
Table 1. Distribution of Yeast and Human Liver Reads across Genomic Regions
Human 50 UTR 30 UTR CDS Introns Transcripts ±200 nt of 50 Ends ±200 nt of 30 Ends ±10 nt of 30 Ends
Sense 6.46 79.38 1.02 8.8 83.94 0.59 71.32 55.7
Antisense 0.18 2.1 0.23 5.86 7.98 0.1 2.96 1.12
Yeast CDS Introns Transcripts ±1000 nt of 30 Ends of ORFs
Sense 4.68 0.19 4.86 91.36
Antisense 9.16 0.04 9.19 53.04
The numbers indicate percentages of uniquely aligned yeast and human DRS reads (Table S2) as provided by the SeqSolve software (Integromics). The
categories shown are not exclusive, and each proportion was computed independently. Hence, proportions are not expected to add up to 100%. The
relatively high percentage of reads in the category of antisense yeast reads within 1000 nt of 30 ORF ends is due to 2000 yeast ORFs whose 30 ends are
close to each other. CDS: coding sequence, UTR: untranslated region, ORF: open reading frame, Transcripts: within annotated gene boundaries (see
also Figure S1 and Table S2).

complementary RNA species, such as microRNA or similar path- counts are indeed highly significant (p < 0.001). Similar trends
ways in human. The distribution of the sense and antisense were observed when converging genes in yeast were omitted
counts for yeast and human did not represent the normal distri- from the analyses (Table S4).
bution (Shapiro-Wilk test, p < 0.0001), even after converting the
values into log space. Thus, we used the nonparametric Sequence Structure Surrounding Polyadenylation Sites
Spearman correlation for this analysis based on the raw (non- Having generated an in-depth view of polyadenylation cleavage
log converted) values of sense and antisense expression levels locations, we examined the sequence patterns potentially gov-
of annotated genes. We separated the annotated genes into erning transcription termination and polyadenylation. We first
four quartiles according to their sense expression levels (Table 2). performed a de novo search for motifs near human polyadenyla-
We did find a weak, but significant (see below), negative correla- tion locations and detected three novel motifs and the canonical
tion between the levels of sense and antisense polyadenylated signal (Figure 3). For this analysis, we used confident polyadeny-
RNAs in the top quartile (Q1). The correlation became progres- lation sites we defined using a clustering approach and
sively more positive as the levels of the sense transcripts supported by multiple reads (Figure S4, Table S5, and Extended
decreased, as exemplified by the positive correlation for the Experimental Procedures). We identified a novel TTTTTTTTT
bottom fourth and third quartile of expression for the yeast and motif (e = 10158) (Figure 3A) and an AAWAAA motif closely
human samples, respectively. Because the expression levels resembling the canonical AWTAAA signal (e = 10112) (Figure 3C)
of transcripts that do not overlap in the genome could also corre- upstream of the polyadenylation sites (Zhao et al., 1999). We
late and the negative correlations obtained for the high expres- examined the distribution of these motifs across five polyadeny-
sors could be influenced by the extreme values, we introduced lation site categories (C1-5) generated depending on site
a permutation test whereby pairing of sense-antisense values orientation (e.g., sense or antisense) and proximity relative to
for each gene was reassigned: for each annotated gene, the known 30 ends of genes (Figure 3 and Experimental Procedures).
sense value was kept the same, the antisense value was Just like the canonical AWTAAA signal (Figure 3D and
randomly chosen from another gene, and the Spearman correla- Table S6), TTTTTTTTT occurs in a highly position-specific
tion was calculated. This test shows that all (even the lowest) manner 21 nt upstream of the polyadenylation site (Figure 3B),
correlations found between the real sense and antisense reads suggesting that these motifs are mechanistically important for

Table 2. Spearman Correlation Coefficients between Sense and Antisense Transcript Levels
Yeast
Q1 Q2 Q3 Q4
Actual correlation 0.11 0 0.01 0.36
1000 permutations, minimum 2.39E-05 5.55E-05 3.79E-05 6.78E-05
1000 permutations, maximum 9.05E-05 7.01E-05 8.47E-05 7.84E-05
Human Liver
Q1 Q2 Q3
Actual Correlation 0.11 0.02 0.12
1000 permutations, minimum 9.59E-05 3.40E-05 9.00E-05
1000 permutations, maximum 9.25E-05 9.80E-06 5.69E-07
Q1–4 indicates quartiles, with Q1 indicating the genes with highest sense expression values. For the human liver sample, we performed the analysis
only for the top three quartiles since genes with zero expression level dominated the fourth quartile. The minimum and maximum correlation coeffi-
cients obtained after 1000 permutations were reported (thus p < 0.001). Similar trends were observed for yeast after the removal of potentially over-
lapping transcripts (see also Table S4).

1022 Cell 143, 1018–1029, December 10, 2010 ª2010 Elsevier Inc.
A B

T T TT T T
2
8

Fraction of motifs (%)


Within genes, sense (C3)
Within genes, antisense (C4)
1 4 Intergenic (C5)

A
C 0
0 CG A C
0 50 100 150 200

10
1
2
3
4
5
6
7
8
9
Base location

C D

A AA
2 12
Within 5 nts of annoated 3’ ends, sense (C1)

Fraction of motifs (%)


In the last exon and 1000 nts downstream
8 of annotated 3’ ends, sense (C2)

1 Within genes, sense (C3)


Within genes, antisense (C4)

T
4
Intergenic (C5)

A A A
0
T
G
C
G
C
T
G
C
0
1
2
3
4
5
6
7
8

0 50 100 150 200


Base location

E F
6 Within genes, sense (C3)
2
Fraction of motifs (%)

CCAG CTGG
Within genes, antisense (C4)
Intergenic (C5)

3
1

0
C
T
G
A
AA G TA
G
C
A
T
AAA
G G
C
A
0
0 50 100 150 200
10
1

Base location

G H

G GTG
2
6 Within genes, sense (C3)
Fraction of motifs (%)

Within genes, antisense (C4)

Intergenic (C5)

A TGCA
1
3

0 G CATG
AG G A A
10
1
2
3
4
5
6
7
8
9

0
0 50 100 150 200
Base location

Figure 3. Polyadenylation Motif Analyses


Panels (A), (C), (E), and (G) indicate human motif elements identified. TTTTTTTTT (B), AWTAAA (D), CCAGSCTGG (F), and RGYRYRGTGG (H) distance distribution
are shown in respective panels. Human categories were defined as sites that are within 5 nucleotides of annotated 30 ends of known human genes in sense orien-
tation (category 1), in the last exon and 1 kb downstream of annotated 30 ends of human known genes in sense orientation (category 2), located anywhere within
the transcripts in sense orientation except in categories 1 and 2 (category 3), antisense to genes (category 4) and in intergenic regions (category 5). In distance
plots, y axis indicates the fraction of motifs (in percentages) at x-distances relative to the polyadenylation location (at base location 101) in each category.
X-distances were calculated between the polyadenylation location identified with DRS and the first base immediately before the motif element. In panels B,
F, and H, only the categories 3, 4, and 5 representing genic and intergenic sites were shown, because less than 10% (250–350) of these motifs were in categories
1 and 2, and not in sufficient numbers to be plotted in the graphs. Absolute numbers of motif counts for these latter three panels across all five human categories
were provided in Figures S6A–S6C (see also Figure S4 and Table S5).

polyadenylation. However, the TTTTTTTTT motif is largely We also detected a novel palindromic sequence,
present in the genic and intergenic regions (C3-5 in Figure 3), CCAGSCTGG (e = 1033) (Figure 3E), downstream of the polya-
unlike the canonical motif which is largely present near the anno- denylation sites that manifests a strong position-specific pattern
tated 30 ends of genes (C1-2). (Figure 3F). Further analysis using less stringent motif scans led

Cell 143, 1018–1029, December 10, 2010 ª2010 Elsevier Inc. 1023
to the identification of RGYRYRGTGG (Figure 3G) that co-occur Brem, 2010). Compared to these approaches, the DRS-based
(p = 0) with the CCAGSCTGG motif at a frequency of 45%, approach is in quantitative nature, free of reverse transcription
and localizes 31 nt downstream from the polyadenylation loca- and ligation artifacts, and requires only minute RNA quantities.
tion (Figure 3H). Notably, we found that CCAGSCTGG and The nucleotide resolution of the approach is similar to other
RGYRYRGTGG also strongly co-occur with the TTTTTTTTT classic methods of polyadenylation site mapping. However,
motif (p = 0) in the intergenic and genic regions (C3-5), whereas just like these other approaches, the DRS-based approach
these motifs does not co-occur and anticorrelate with the canon- cannot truly differentiate cases where the template cleavage
ical AWTAAA localization (p = 0). The pervasive presence of the may occur right after an A-residue. Such sites may cause the
TTTTTTTTT motif in the novel genic and intergenic polyadenyla- resolution of the approach to elevate from its current level
tion sites, its similarity to the AWTAA signal with respect to of ±2 nt. Because sequencing technologies available or in devel-
its positional preference, its anticorrelation to AWTAAA localiza- opment today, including DRS, do not provide the full transcript
tion, and its co-occurrence with the CCAGSCTGG and sequence, it is not possible to know the sequence of the entire
RGYRYRGTGG motifs are intriguing and may point to uncharac- RNA molecule represented by each read by any sequencing
terized polyadenylation mechanism(s) in humans. We applied technology. It is therefore possible that the reads found around
similar approaches to yeast, detecting no additional motifs the annotated transcriptional start and polyadenylation sites
beyond the previously characterized positioning (PE, AAWAAA) may partly represent short poly(A)+ RNAs previously found to
and upstream efficiency (EE, TAYRTA) elements (Zhao et al., be associated with the gene termini (Kapranov et al., 2007a,
1999). The general positioning of the upstream PE motif (Fig- 2010; Affymetrix ENCODE Transcriptome Project, 2009). A frac-
ure S5A) were closer to the cleavage site than the localization tion of reads found around the annotated polyadenylation site of
of the EE motif (Figure S5B), as expected (Zhao et al., 1999). known messages may not represent the annotated form, but
We then examined the nucleotide composition around the pol- other isoforms or correspond to other overlapping transcripts
yadenylation cleavage locations in each group. We observed that share the same polyadenylation region. Furthermore, polya-
a difference in the profiles of nucleotide frequency distributions denylation sites observed downstream of annotated 30 ends may
surrounding human cleavage sites in regions near 30 known represent alternative polyadenylation events or transcription
gene ends (C1-2) and in genic and intergenic regions (C3-5, Fig- termination products (Kim et al., 2004; Teixeira et al., 2004;
ure 4). As expected, the categories 1 and 2 had the T-rich down- West et al., 2004).
stream sequence element (DSE) 20-30 bases downstream of the Our results show that most yeast and human transcripts have
polyadenylation sites and A-rich sequences upstream (Zhao yet uncharacterized polyadenylation sites. This dataset, along
et al., 1999). On the other hand, the nucleotide profiles around with additional biological replicates and data from different cell
the sites in the categories 3–5 were different and similar to the types and states, will allow empirical annotation of such sites
yeast sites (Figures S5C–S5F) with the pronounced upstream and provide the substrate for biological experimentation exam-
T-rich sequences, in line with the TTTTTTTTT motif identified in ining changes in these sites. The enrichment of reads in yeast in-
the upstream regions above (Figure 4). The presence of a T-rich tergenic functional transcription factor-binding sites and DHSs
polyadenylation enhancer sequence element upstream of the suggests that these potential regulatory regions may indeed
AATAAA motif is common among viruses and has been previ- encode for RNAs. The presence of RNAs from a subset of poten-
ously found in a few human genes (Bhat and Wold, 1987; Moreira tial mammalian enhancers (eRNAs) and open chromatin regions
et al., 1995). However, the T-rich pattern observed here is imme- has recently been described (De Santa et al., 2010; Kim et al.,
diately upstream of the sense/antisense genic and intergenic 2010; van Bakel et al., 2010). Unlike the report by Kim et al.,
cleavage sites, and therefore represents a different and novel (2010), which found eRNAs to lack poly(A) tails, our results
observation. This latter similarity at the yeast and human nucle- indicate the potential existence of poly(A)-tail containing RNAs
otide profiles prompted us to examine yeast motif presence in associated with regulatory elements in yeast. We speculate
humans. Interestingly, we observed an enrichment of the yeast that these regulatory region-associated reads may represent
EE motif immediately upstream of the human cleavage sites in a recently described class of polyadenylated noncoding RNAs
categories 3–5, but not in categories 1 and 2 (Figure 5). The yeast that regulate gene expression (Bumgarner et al., 2009; Orom
EE motif however does not co-occur with the novel et al., 2010). They may also represent divergent transcription
CCAGSCTGG, RGYRYRGTGG, and TTTTTTTTT motifs identi- events from unannotated promoters (Neil et al., 2009; Seila
fied above, and thus may be present in an independent subset et al., 2008; Xu et al., 2009). Alternatively, given the likely associ-
of genic and intergenic sites. This latter finding may point to ation of RNA polymerase II with the transcriptional factors
the existence of another, perhaps yeast-like, polyadenylation binding to these regions, these RNAs may emerge from tran-
sequence structure in a subset of human polyadenylation sites. scriptional noise events postulated to occur (Struhl, 2007). The
lack of comprehensive transcription factor-binding site and
DISCUSSION enhancer maps in humans prevented us from examining such
RNAs in our human studies. However, the relatively high fraction
This study presents genome-wide polyadenylation maps that of intergenic DRS reads obtained in the human samples suggest
incorporate the accuracy of a high-throughput sequencing- that at least a fraction of these reads may emerge from
based methodology and true strand-specificity. Other enhancers. Further studies are needed to delineate the func-
sequencing-based polyadenylation mapping approaches have tions, if any, of these RNAs and how they may be contributing
recently become available (Mangone et al., 2010; Yoon and to regulatory function.

1024 Cell 143, 1018–1029, December 10, 2010 ª2010 Elsevier Inc.
A C
Within 5 nts of annotated 3' ends, sense (C1) Within genes, sense (C3)
60 60
Fraction of Each Base (%)

Fraction of Each Base (%)


30 30

0 0
0 50 100 150 200 0 50 100 150 200

Base location Base location

B D
In the last exon and 1000 nts downstream of Within genes, antisense (C4)
annotated 3' ends, sense (C2) 60
Fraction of Each Base (%)

60
Fraction of Each Base (%)

30
30

0 0
0 50 100 150 200 0 50 100 150 200

Base location Base location

T E Intergenic (C5)
60
G
Fraction of Each Base (%)

C
A
30

0
0 50 100 150 200

Base location

Figure 4. The Nucleotide Composition Surrounding Polyadenylation Cleavage Locations in Humans


(A–E) Category descriptions were provided in Figure 3. y axis indicates the nucleotide composition (in percentages) at x-locations relative to the cleavage posi-
tions (at base location 101). Dark blue (diamond), blue (rectangle), green (triangle), and red (cross) lines indicate T, G, C, and A nucleotides, respectively. Poly-
adenylation locations in C3-5 differ from those in C1-2, and exhibit elevated T and A content in 40–50 nt upstream of polyadenylation cleavage positions (see also
Figure S5 and Table S6).

Our observation of novel polyadenylation patterns including to sites in proximity to the 30 ends of known genes suggest inter-
novel co-occurring motifs (CCAGSCTGG, RGYRYRGTGG, and esting possibilities for human polyadenylation. Particularly, the
TTTTTTTTT) and enrichment of T-rich and yeast EE motif anticorrelation we observed between the localizations of the
sequences near sites corresponding to noncoding transcript three novel motifs above and the canonical AWTAAA suggests
categories (antisense, sense genic, and intergenic) compared alternative and yet to be characterized mechanisms of

Cell 143, 1018–1029, December 10, 2010 ª2010 Elsevier Inc. 1025
0.50 Within 5 nts of annotated 3’ ends, sense (C1) Figure 5. Distance Distribution of Yeast EE
(TAYRTA) Motif across Human Categories
In the last exon and 1000 nts downstream y axis indicates the fraction of motifs (in percent-
Fraction of motifs (%)

of annotated 3’ ends, sense (C2) ages) at x-distances relative to the cleavage posi-
Within genes, sense (C3) tions (at base location 101) in each category.
X-distances were calculated between the
Within genes, antisense (C4) cleavage location identified with DRS and the first
0.25
base immediately before the motif element.
Intergenic (C5)
Human category descriptions were provided in
Figure 3 legend. The enrichment of the EE motif
immediately upstream of the cleavage sites in
human categories 3, 4, and 5, but not in categories
1 and 2, is in parallel to the upstream human
0.00 T-enrichment pattern shown in Figure 4 (see also
0 50 100 150 200 Figure S6).
Base location

transcription termination, cleavage, and polyadenylation. Given hand, the genes in the bottom quartile show a positive correla-
that RNAs in these regions are likely to be noncoding, perhaps tion between the sense and antisense transcription. Although
alternative modes of polyadenylation exist for noncoding both results are significant, the former effect is relatively small
RNAs. These three novel motifs are present in a relatively small and similar to what has been detected previously (Chen et al.,
fraction of polyadenylation sites and cleavage events 2005), whereas the latter effect is the strongest (at least in yeast)
(Table S6C). This may partly be explained by the relatively low and is similar to the results obtained in Schizosaccharomyces
fraction of polyadenylated noncoding RNAs relative to mRNAs pombe (Dutrow et al., 2008) and mouse (Katayama et al.,
of protein-coding genes in terms of mass. Combined with the 2005), where positive correlation was found. In view of these
recent observation that even very low abundance noncoding results, it is perhaps not surprising that the correlation of sense
RNAs, as low as four copies per cell, can regulate target genes and antisense transcripts has remained a controversial issue
(Wang et al., 2008), these new motifs may be specific to such as often both were found to be positively correlated (Kapranov
a subset of noncoding RNAs. Further in-depth de novo motif et al., 2007b). The relatively low negative correlation values
analyses in these novel regions and the identification of the most likely reflect the fact the overlapping positioning in the
components of this potential alternative polyadenylation genome is only one of many ways to regulate stable levels of pol-
machinery would open a number of conceptual and experi- ydenylated RNAs species. It is however tempting to speculate
mental possibilities. First, we may learn more about the RNAs that in highly expressed genes, the physical interference of
they process, which may include various species (Buratowski, converging RNA polymerase complexes could exert a dominant
2008) such as promoter-associated RNAs (Core et al., 2008; effect, whereas this possibility may be less of a factor in the
Neil et al., 2009; Seila et al., 2008), cryptic unstable RNAs (Preker genes with lower transcriptional activity. In the latter cases, other
et al., 2008; Wyers et al., 2005), long intergenic noncoding RNAs factors, such as chromatin accessibility that could permit tran-
(Guttman et al., 2009), and polyadenylated RNAs resulting from scription from both strands could be a larger determining factor.
degradation events (Slomovic et al., 2010). Second, we may To what extent the observed negative correlation is due to
get a more mechanistic understanding of polyadenylation and sense/antisense transcripts occupying the same genomic space
its connection with other cellular processes. For instance, the and/or other transcriptional control mechanisms needs further
CCAGSCTGG palindromic motif identified here is a candidate exploration.
binding site for human topoisomerase II (topo-II) (Spitzner and This study represents the first step for the adaptation of the
Muller, 1988). Topo-II is part of the RNA polymerase II holoen- direct RNA sequencing technology to decipher the genome
zyme and relaxes the superhelical tension that accumulates and its functions. Future studies will focus on the functional char-
during transcription elongation (Mondal and Parvin, 2001). acterization of novel poly(A)+ regulatory region-associated
Perhaps the presence of such a motif downstream of polyadeny- RNAs, antisense transcripts, and polyadenylation sites identified
lation sites is to ensure that transcriptional superhelical tension in this study, and the adaptation of DRS for other existing and
does not extend beyond the boundaries of the transcripts and novel RNA applications.
thus do not disturb downstream regions.
In line with previous studies (He et al., 2008; Kapranov et al.,
2007b), we observed that antisense transcription is prevalent EXPERIMENTAL PROCEDURES
in the yeast and human genomes and that the quantities of
steady-state levels of sense and antisense transcripts occupying Sample Preparation for DRS
Yeast (Saccharomyces cerevisiae) and human liver poly(A)+ RNAs were
the same genomic space can negatively correlate with each
obtained from Clontech, CA (USA). Human brain total RNA was from Ambion.
other. Our results indicate a complex picture where the highly The 30 blocking reaction was performed with poly(A) tailing kit (Ambion, TX,
expressed genes in the top quartile tend to negatively correlate USA) and 30 deoxyATP (Jena Biosciences, Germany), incubating the reaction
with the expression of antisense transcripts. On the other mixture at 37 C for 30 min. The blocked RNA was hybridized to flow cell

1026 Cell 143, 1018–1029, December 10, 2010 ª2010 Elsevier Inc.
surfaces for sequencing with DRS without additional cleaning steps (Ozsolak ACCESSION NUMBERS
et al., 2009).
Sequencing datasets described in this study have been deposited at the
Data Analysis National Center for Biotechnology Information (NCBI) Sequence Read Archive
Raw DRS reads were filtered using a suite of Helicos tools available at http:// (SRA), accession no SRA012232. The datasets are also available as wiggle
open.helicosbio.com/mwiki/index.php/Releases and described at http:// files at the Helicos open access website (HeliSphere, http://open.helicosbio.
open.helicosbio.com/helisphere_user_guide/. Alignments were conducted com/) along with yeast and human polyadenylation sites defined in this study.
with indexDPgenomic available on the Helicos website (http://open.
helicosbio.com/mwiki/index.php/Releases). For the genomic alignments, reads
SUPPLEMENTAL INFORMATION
were aligned to the yeast SGD/sacCer2 and human NCBIv36 version of the
genome supplemented with the complete ribosomal repeat unit (GenBank
Supplemental Information includes Extended Experimental Procedures, six
accession number U13369.1). Reads with a minimal length of 25 nt and align-
figures, and six tables and can be found with this article online at doi:10.
ment score of 4.3 and above were allowed. Aligned reads were further filtered
1016/j.cell.2010.11.020.
for reads having a unique best alignment score. Total raw per base error rate
was 4%–5%, dominated by missing base errors (2%–3%).
Downstream analysis was performed with the SeqSolve NGS software (In- ACKNOWLEDGMENTS
tegromics, Spain). Annotated yeast or human transcriptome was defined as
either the SGD Genes from Saccharomyces Genome Database track or We thank our Helicos and Integromics colleagues for technical assistance and
UCSC Genes track on the UCSC Genome Browser. Counts within each anno- discussions. This work was supported by the National Human Genome
tation were derived from either the sense or antisense strand using the posi- Research Institute (grant R01-HG005230 to F.O. and P.M.M.). B.J. is sup-
tions of the 50 ends of reads aligned to the appropriate strand. Yeast median ported by the National Institutes of Health (grant GM079756) and the American
UTR length was calculated by taking the median of the distances between Cancer Society (grant RSG0905401), A.P.M. is supported by the National Insti-
the annotated 30 end locations of yeast ORFs and the reads that map in the tutes of Health (grant MH60774), and S.F. is supported by the Spanish Ministry
sense orientation and within 1000 bp downstream of ORF 30 ends. of Science and Innovation—FEDER (CDTI loan IDI-20091293). F.O., P.K., and
For the sequence composition surrounding polyadenylation cleavage site P.M.M. are employees of Helicos BioSciences Corporation. S.F. is an
analysis, the 50 ends of reads representing the 30 cleavage sites were grouped employee of Integromics.
based on overlap with the genomic annotation, as described in Figure 3 and
Figure S5. Mitochondrial reads were not used for the sequence analysis. These Received: May 26, 2010
categories for human were (1) sense cleavage locations that are within 5 bases Revised: September 28, 2010
of annotated 30 ends, (2) sense cleavage locations that are not in category #1 Accepted: November 9, 2010
and are in the last exons or 1 kb downstream of the annotated 30 ends, (3) Published: December 9, 2010
sense cleavage locations that are not in categories 1 and 2 and are within
annotated genes, (4) antisense cleavage locations that are within annotated
REFERENCES
genes, and (5) intergenic cleavage locations that are not in categories 1–4.
The categories for yeast were (1) sense cleavage locations that are located
Andreassi, C., and Riccio, A. (2009). To localize or not to localize: mRNA fate is
within 200 bp downstream of the annotated 30 ends of yeast ORFs, (2) sense
in 30 UTR ends. Trends Cell Biol. 19, 465–474.
cleavage locations that are not within category 1 and are within bodies of
ORFs, (3) antisense cleavage locations that are not within category 1 and Bailey, T.L., Williams, N., Misleh, C., and Li, W.W. (2006). MEME: discovering
are within bodies of ORFs, and (4) intergenic cleavage locations that are not and analyzing DNA and protein sequence motifs. Nucleic Acids Res. 34,
in categories 1, 2, and 3, and are at least 1 kb away from the 30 ends of yeast W369–W373.
ORFs. Reads in each category were then collapsed according to their unique Bennett, C.L., Brunkow, M.E., Ramsdell, F., O’Briant, K.C., Zhu, Q., Fuleihan,
50 ends representing unique polyadenylation cleavage locations. Sequences R.L., Shigeoka, A.O., Ochs, H.D., and Chance, P.F. (2001). A rare polyadeny-
100 bases on each side of each collapsed locations were analyzed as lation signal mutation of the FOXP3 gene (AAUAAA/AAUGAA) leads to the
described in the text. IPEX syndrome. Immunogenetics 53, 435–439.
Bhat, B.M., and Wold, W.S. (1987). A small deletion distant from a splice or pol-
Detection of Novel Motifs yadenylation site dramatically alters pre-mRNA processing in region E3 of
To investigate the presence of new sequence motifs, upstream and down- adenovirus. J. Virol. 61, 3938–3945.
stream genomic sequences (50 bases) of novel polyadenylation sites (Fig-
Brais, B., Bouchard, J.P., Xie, Y.G., Rochefort, D.L., Chretien, N., Tome, F.M.,
ure S4) were scanned independently using MEME (Bailey et al., 2006). To
Lafreniere, R.G., Rommens, J.M., Uyama, E., Nohira, O., et al. (1998). Short
reduce the occurrence of spurious motifs, motif searches were performed
GCG expansions in the PABP2 gene cause oculopharyngeal muscular
using a highly stringent E-value (1025) threshold, based on a nonredundant
dystrophy. Nat. Genet. 18, 164–167.
set of 1000 sequences that were sampled uniformly from the complete set
of upstream/downstream sequences. The threshold (1025) was used Bumgarner, S.L., Dowell, R.D., Grisafi, P., Gifford, D.K., and Fink, G.R. (2009).
because even when sites across each chromosome was separately analyzed Toggle involving cis-interfering noncoding RNAs controls variegated gene
(24 control experiments) to rule out dataset artifacts, the three human motifs expression in yeast. Proc. Natl. Acad. Sci. USA 106, 18321–18326.
were consistently detected. The various motif variants were manually in- Buratowski, S. (2008). Transcription: gene expression–where to start? Science
spected to select a single motif for display representation. For additional vali- 322, 1804–1805.
dations of the motifs, the up/downstream occurrences and co-occurrences
Chen, J., Sun, M., Hurst, L.D., Carmichael, G.G., and Rowley, J.D. (2005).
were analyzed. Total occurrences of motifs in up/downstream sequences
Genome-wide analysis of coordinate expression and evolution of human cis-
were determined by searching for all short strings that matched (>90%) the
encoded sense-antisense transcripts. Trends Genet. 21, 326–329.
position-specific scoring (log-odds) matrix profile of the motifs detected by
MEME. To test the statistical significance of co-occurrence between two Cocquet, J., Chong, A., Zhang, G., and Veitia, R.A. (2006). Reverse transcrip-
motifs, hypergeometric tests (Lee et al., 2007) were performed based on the tase template switching and false alternative transcripts. Genomics 88,
total number of occurrences of the two motifs in the complete set of nonredun- 127–131.
dant sequences. Because only four motifs were compared (six comparisons) Core, L.J., Waterfall, J.J., and Lis, J.T. (2008). Nascent RNA sequencing
to each other for co-occurrence analysis, and because the reported p values reveals widespread pausing and divergent initiation at human promoters.
are close to zero, the Bonferroni correction factor of 6 was not used. Science 322, 1845–1848.

Cell 143, 1018–1029, December 10, 2010 ª2010 Elsevier Inc. 1027
Costessi, L., Devescovi, G., Baralle, F.E., and Muro, A.F. (2006). Brain-specific Lee, J., Li, Z., Brower-Sinning, R., and John, B. (2007). Regulatory circuit of
promoter and polyadenylation sites of the beta-adducin pre-mRNA generate human microRNA biogenesis. PLoS Comput. Biol. 3, e67.
an unusually long 30 -UTR. Nucleic Acids Res. 34, 243–253. Lin, C.L., Bristol, L.A., Jin, L., Dykes-Hoberg, M., Crawford, T., Clawson, L.,
David, L., Huber, W., Granovskaia, M., Toedling, J., Palm, C.J., Bofkin, L., and Rothstein, J.D. (1998). Aberrant RNA processing in a neurodegenerative
Jones, T., Davis, R.W., and Steinmetz, L.M. (2006). A high-resolution map of disease: the cause for absent EAAT2, a glutamate transporter, in amyotrophic
transcription in the yeast genome. Proc. Natl. Acad. Sci. USA 103, 5320–5325. lateral sclerosis. Neuron 20, 589–602.
De Santa, F., Barozzi, I., Mietton, F., Ghisletti, S., Polletti, S., Tusi, B.K., Muller, Liu, D., and Graber, J.H. (2006). Quantitative comparison of EST libraries
H., Ragoussis, J., Wei, C.L., and Natoli, G. (2010). A large fraction of extragenic requires compensation for systematic biases in cDNA generation. BMC Bioin-
RNA pol II transcription sites overlap enhancers. PLoS Biol. 8, e1000384. formatics 7, 77.
Dutrow, N., Nix, D.A., Holt, D., Milash, B., Dalley, B., Westbroek, E., Parnell, Lopez, F., Granjeaud, S., Ara, T., Ghattas, B., and Gautheret, D. (2006). The
T.J., and Cairns, B.R. (2008). Dynamic transcriptome of Schizosaccharomyces disparate nature of ‘‘intergenic’’ polyadenylation sites. RNA 12, 1794–1801.
pombe shown by RNA-DNA hybrid mapping. Nat. Genet. 40, 977–986.
Lutz, C.S. (2008). Alternative polyadenylation: a twist on mRNA 30 end forma-
Faghihi, M.A., and Wahlestedt, C. (2009). Regulatory roles of natural antisense
tion. ACS Chem. Biol. 3, 609–617.
transcripts. Nat. Rev. Mol. Cell Biol. 10, 637–643.
Mahadevan, S., Raghunand, T.R., Panicker, S., and Struhl, K. (1997). Charac-
Gehring, N.H., Frede, U., Neu-Yilik, G., Hundsdoerfer, P., Vetter, B., Hentze,
terisation of 30 end formation of the yeast HIS3 mRNA. Gene 190, 69–76.
M.W., and Kulozik, A.E. (2001). Increased efficiency of mRNA 30 end formation:
a new genetic mechanism contributing to hereditary thrombophilia. Nat. Mamanova, L., Andrews, R.M., James, K.D., Sheridan, E.M., Ellis, P.D., Lang-
Genet. 28, 389–392. ford, C.F., Ost, T.W., Collins, J.E., and Turner, D.J. (2010). FRT-seq: amplifica-
tion-free, strand-specific transcriptome sequencing. Nat. Methods 7,
Graber, J.H., McAllister, G.D., and Smith, T.F. (2002). Probabilistic prediction
130–132.
of Saccharomyces cerevisiae mRNA 30 -processing sites. Nucleic Acids Res.
30, 1851–1858. Mangone, M., Manoharan, A.P., Thierry-Mieg, D., Thierry-Mieg, J., Han, T.,
Gubler, U. (1987). Second-strand cDNA synthesis: mRNA fragments as Mackowiak, S.D., Mis, E., Zegar, C., Gutwein, M.R., Khivansara, V., et al.
primers. Methods Enzymol. 152, 330–335. (2010). The landscape of C. elegans 30 UTRs. Science 329, 432–435.

Guttman, M., Amit, I., Garber, M., French, C., Lin, M.F., Feldser, D., Huarte, M., Mondal, N., and Parvin, J.D. (2001). DNA topoisomerase IIalpha is required for
Zuk, O., Carey, B.W., Cassady, J.P., et al. (2009). Chromatin signature reveals RNA polymerase II transcription on chromatin templates. Nature 413,
over a thousand highly conserved large non-coding RNAs in mammals. Nature 435–438.
458, 223–227. Moreira, A., Wollerton, M., Monks, J., and Proudfoot, N.J. (1995). Upstream
He, Y., Vogelstein, B., Velculescu, V.E., Papadopoulos, N., and Kinzler, K.W. sequence elements enhance poly(A) site efficiency of the C2 complement
(2008). The antisense transcriptomes of human cells. Science 322, 1855–1857. gene and are phylogenetically conserved. EMBO J. 14, 3809–3819.
Hesselberth, J.R., Chen, X., Zhang, Z., Sabo, P.J., Sandstrom, R., Reynolds, Muro, E.M., Herrington, R., Janmohamed, S., Frelin, C., Andrade-Navarro,
A.P., Thurman, R.E., Neph, S., Kuehn, M.S., Noble, W.S., et al. (2009). Global M.A., and Iscove, N.N. (2008). Identification of gene 30 ends by automated
mapping of protein-DNA interactions in vivo by digital genomic footprinting. EST cluster analysis. Proc. Natl. Acad. Sci. USA 105, 20286–20290.
Nat. Methods 6, 283–289.
Nagaike, T., Suzuki, T., Katoh, T., and Ueda, T. (2005). Human mitochondrial
Higgs, D.R., Goodbourn, S.E., Lamb, J., Clegg, J.B., Weatherall, D.J., and mRNAs are stabilized with polyadenylation regulated by mitochondria-specific
Proudfoot, N.J. (1983). Alpha-thalassaemia caused by a polyadenylation poly(A) polymerase and polynucleotide phosphorylase. J. Biol. Chem. 280,
signal mutation. Nature 306, 398–400. 19721–19727.
Iseli, C., Stevenson, B.J., de Souza, S.J., Samaia, H.B., Camargo, A.A., Nagalakshmi, U., Wang, Z., Waern, K., Shou, C., Raha, D., Gerstein, M., and
Buetow, K.H., Strausberg, R.L., Simpson, A.J., Bucher, P., and Jongeneel, Snyder, M. (2008). The transcriptional landscape of the yeast genome defined
C.V. (2002). Long-range heterogeneity at the 30 ends of human mRNAs. by RNA sequencing. Science 320, 1344–1349.
Genome Res. 12, 1068–1074.
Neil, H., Malabat, C., d’Aubenton-Carafa, Y., Xu, Z., Steinmetz, L.M., and Jac-
Kapranov, P., Cheng, J., Dike, S., Nix, D.A., Duttagupta, R., Willingham, A.T., quier, A. (2009). Widespread bidirectional promoters are the major source of
Stadler, P.F., Hertel, J., Hackermuller, J., Hofacker, I.L., et al. (2007a). RNA cryptic transcripts in yeast. Nature 457, 1038–1042.
maps reveal new RNA classes and a possible function for pervasive transcrip-
tion. Science 316, 1484–1488. Orkin, S.H., Cheng, T.C., Antonarakis, S.E., and Kazazian, H.H., Jr. (1985).
Thalassemia due to a mutation in the cleavage-polyadenylation signal of the
Kapranov, P., Willingham, A.T., and Gingeras, T.R. (2007b). Genome-wide
human beta-globin gene. EMBO J. 4, 453–456.
transcription and the implications for genomic organization. Nat. Rev. Genet.
8, 413–423. Orom, U.A., Derrien, T., Beringer, M., Gumireddy, K., Gardini, A., Bussotti, G.,
Lai, F., Zytnicki, M., Notredame, C., Huang, Q., et al. (2010). Long noncoding
Kapranov, P., Ozsolak, F., Kim, S.W., Foissac, S., Lipson, D., Hart, C., Roels,
RNAs with enhancer-like function in human cells. Cell 143, 46–58.
S., Borel, C., Antonarakis, S.E., Monaghan, P., et al. (2010). New class of gene-
termini-associated human RNAs suggests a novel RNA copying mechanism. Ozsolak, F., Platt, A.R., Jones, D.R., Reifenberger, J.G., Sass, L.E., McInerney,
Nature 466, 642–646. P., Thompson, J.F., Bowers, J., Jarosz, M., and Milos, P.M. (2009). Direct RNA
Katayama, S., Tomaru, Y., Kasukawa, T., Waki, K., Nakanishi, M., Nakamura, sequencing. Nature 461, 814–818.
M., Nishida, H., Yap, C.C., Suzuki, M., Kawai, J., et al. (2005). Antisense tran- Preker, P., Nielsen, J., Kammler, S., Lykke-Andersen, S., Christensen, M.S.,
scription in the mammalian transcriptome. Science 309, 1564–1566. Mapendano, C.K., Schierup, M.H., and Jensen, T.H. (2008). RNA exosome
Kim, M., Krogan, N.J., Vasiljeva, L., Rando, O.J., Nedea, E., Greenblatt, J.F., depletion reveals transcription upstream of active human promoters. Science
and Buratowski, S. (2004). The yeast Rat1 exonuclease promotes transcription 322, 1851–1854.
termination by RNA polymerase II. Nature 432, 517–522. Affymetrix ENCODE Transcriptome Project; Cold Spring Harbor Laboratory
Kim, T.K., Hemberg, M., Gray, J.M., Costa, A.M., Bear, D.M., Wu, J., Harmin, ENCODE Transcriptome Project. (2009). Post-transcriptional processing
D.A., Laptewicz, M., Barbara-Haley, K., Kuersten, S., et al. (2010). Widespread generates a diversity of 50 -modified long and short RNAs. Nature 457, 1028–
transcription at neuronal activity-regulated enhancers. Nature 465, 182–187. 1032.
Kiyosawa, H., Mise, N., Iwase, S., Hayashizaki, Y., and Abe, K. (2005). Rigault, C., Le Borgne, F., and Demarquoy, J. (2006). Genomic structure, alter-
Disclosing hidden transcripts: mouse natural sense-antisense transcripts native maturation and tissue expression of the human BBOX1 gene. Biochim.
tend to be poly(A) negative and nuclear localized. Genome Res. 15, 463–474. Biophys. Acta 1761, 1469–1481.

1028 Cell 143, 1018–1029, December 10, 2010 ª2010 Elsevier Inc.
Seila, A.C., Calabrese, J.M., Levine, S.S., Yeo, G.W., Rahl, P.B., Flynn, R.A., Wang, X., Arai, S., Song, X., Reichart, D., Du, K., Pascual, G., Tempst, P.,
Young, R.A., and Sharp, P.A. (2008). Divergent transcription from active Rosenfeld, M.G., Glass, C.K., and Kurokawa, R. (2008). Induced ncRNAs allo-
promoters. Science 322, 1849–1851. sterically modify RNA-binding proteins in cis to inhibit transcription. Nature
Slomovic, S., Portnoy, V., and Schuster, G. (2008). Detection and characteriza- 454, 126–130.
tion of polyadenylated RNA in Eukarya, Bacteria, Archaea, and organelles. West, S., Gromak, N., and Proudfoot, N.J. (2004). Human 50 –> 30 exonuclease
Methods Enzymol. 447, 501–520. Xrn2 promotes transcription termination at co-transcriptional cleavage sites.
Slomovic, S., Fremder, E., Staals, R.H., Pruijn, G.J., and Schuster, G. (2010). Nature 432, 522–525.
Addition of poly(A) and poly(A)-rich tails during RNA degradation in the cyto- Wu, J.Q., Du, J., Rozowsky, J., Zhang, Z., Urban, A.E., Euskirchen, G., Weiss-
plasm of human cells. Proc. Natl. Acad. Sci. USA 107, 7407–7412. man, S., Gerstein, M., and Snyder, M. (2008). Systematic analysis of tran-
Spiegelman, S., Burny, A., Das, M.R., Keydar, J., Schlom, J., Travnicek, M., scribed loci in ENCODE regions using RACE sequencing reveals extensive
and Watson, K. (1970). DNA-directed DNA polymerase activity in oncogenic transcription in the human genome. Genome Biol. 9, R3.
RNA viruses. Nature 227, 1029–1031.
Wyers, F., Rougemaille, M., Badis, G., Rousselle, J.C., Dufour, M.E., Boulay,
Spitzner, J.R., and Muller, M.T. (1988). A consensus sequence for cleavage by J., Regnault, B., Devaux, F., Namane, A., Seraphin, B., et al. (2005). Cryptic
vertebrate DNA topoisomerase II. Nucleic Acids Res. 16, 5533–5556. pol II transcripts are degraded by a nuclear quality control pathway involving
Struhl, K. (2007). Transcriptional noise and the fidelity of initiation by RNA poly- a new poly(A) polymerase. Cell 121, 725–737.
merase II. Nat. Struct. Mol. Biol. 14, 103–105.
Xu, Z., Wei, W., Gagneur, J., Perocchi, F., Clauder-Munster, S., Camblong, J.,
Teixeira, A., Tahiri-Alaoui, A., West, S., Thomas, B., Ramadass, A., Martianov, Guffanti, E., Stutz, F., Huber, W., and Steinmetz, L.M. (2009). Bidirectional
I., Dye, M., James, W., Proudfoot, N.J., and Akoulitchev, A. (2004). Autocata- promoters generate pervasive transcription in yeast. Nature 457, 1033–1037.
lytic RNA cleavage in the human beta-globin pre-mRNA promotes transcrip-
Yoon, O.K., and Brem, R.B. (2010). Noncanonical transcript forms in yeast and
tion termination. Nature 432, 526–530.
their regulation during environmental stress. RNA 16, 1256–1267.
Tian, B., Hu, J., Zhang, H., and Lutz, C.S. (2005). A large-scale analysis of
mRNA polyadenylation of human and mouse genes. Nucleic Acids Res. 33, Zhang, H., Hu, J., Recce, M., and Tian, B. (2005a). PolyA_DB: a database for
201–212. mammalian mRNA polyadenylation. Nucleic Acids Res. 33, D116–D120.

Tian, B., Pan, Z., and Lee, J.Y. (2007). Widespread mRNA polyadenylation Zhang, H., Lee, J.Y., and Tian, B. (2005b). Biased alternative polyadenylation in
events in introns indicate dynamic interplay between polyadenylation and human tissues. Genome Biol. 6, R100.
splicing. Genome Res. 17, 156–165. Zhao, J., Hyman, L., and Moore, C. (1999). Formation of mRNA 30 ends in
van Bakel, H., Nislow, C., Blencowe, B.J., and Hughes, T.R. (2010). Most ‘‘dark eukaryotes: mechanism, regulation, and interrelationships with other steps
matter’’ transcripts are associated with known genes. PLoS Biol. 8, e1000371. in mRNA synthesis. Microbiol. Mol. Biol. Rev. 63, 405–445.

Cell 143, 1018–1029, December 10, 2010 ª2010 Elsevier Inc. 1029
Scientific Editor, Cell
Cell is seeking a scientist to join its editorial team. The minimum qualification for this
position is a PhD in a relevant area of biomedical research, although additional post-
doctoral or editorial experience is preferred. This is a superb opportunity for a talented
individual to play a critical role in promoting science by helping researchers disseminate
their findings to the wider community.

As an editor, you would be responsible for assessing submitted research papers and
overseeing the refereeing process, and you would commission and edit material for
Cell's Leading Edge. You would also travel frequently to scientific conferences to follow
developments in research and to establish and maintain close ties with the scientific
community. You would have opportunities to pioneer and contribute to new trends in
scientific publishing. The key qualities we look for are breadth of scientific interest and
the ability to think critically about a wide range of scientific issues. The successful
candidate will be highly motivated and creative and able to work independently as well
as in a team.

This is a full-time, in-house editorial position, based at the Cell Press office in Kendall
Square near MIT in Cambridge, Massachusetts. Cell Press offers an attractive salary
and benefits package and a stimulating working environment that encourages innova-
tion.

To apply, please visit


http://reedelsevier.taleo.net/careersection/51/jobdetail.ftl?lang=en&job=SCI0005W.
Scientific Editor, Cell Press
Cell Press seeks to appoint three Scientific Editors with dual roles covering scientific
editing and the review material. These positions will be associated with the Cell Press
titles Cancer Cell, Current Biology, Developmental Cell, and Neuron, and expertise in
any of the relevant areas covered by these journals will be considered. Working closely
with the research community, you will be acquiring, managing, and developing new
editorial content for the Cell Press research titles. These positions will also work closely
with other aspects of the business, including production, business development,
marketing, and commercial sales, and, therefore, provide an excellent entry opportunity
to science publishing. You will work as part of a highly dynamic and collaborative
editorial group in the Cambridge, MA office. These positions are an exciting opportunity
to stay at the forefront of the latest scientific advances while developing a new career in
an exciting publishing environment.

Minimum qualifications are a PhD in a relevant life science discipline, and additional
postdoctoral or other experience is a plus. Ideal candidates would have a strong
scientific background and broad research interests, excellent writing and communica-
tion skills, strong organizational and interpersonal skills, as well as creative energy and
enthusiasm for science and science communication. Prior publishing or editorial
experience is an advantage but is not a requirement.

To apply
Please submit to the url below a CV and cover letter explaining your interest in an
editorial position and describing your qualifications, research interests, and reasons for
pursuing a career in scientific publishing. Applications will be accepted on an ongoing
basis through January 10, 2011.

http://reedelsevier.taleo.net/careersection/51/jobdetail.ftl?lang=en&job=SCI00063.

No phone inquiries. Elsevier-Cell Press is an Equal Opportunity Employer.


Scientific Editor, Molecular Cell
Molecular Cell is seeking a full-time scientific editor to join its editorial team. We will
consider qualified candidates with scientific expertise in any area that the journal
covers. The minimum qualification for this position is a PhD in a relevant area of
biomedical research, although additional experience is preferred. This is a superb
opportunity for a talented individual to play a critical role in the research community
away from the bench.

As a scientific editor, you would be responsible for assessing submitted research


papers, overseeing the refereeing process, and choosing and commissioning review
material. You would also travel frequently to scientific conferences to follow develop-
ments in research and establish and maintain close ties with the scientific community.
The key qualities we look for are breadth of scientific interest and the ability to think
critically about a wide range of scientific issues. The successful candidate will also be
highly motivated and creative and able to work independently as well as in a team.

This is a full-time in-house editorial position, based at the Cell Press office in Cambridge,
Massachusetts. Cell Press offers an attractive salary and benefits package and a
stimulating working environment. Applications will be held in the strictest of confidence
and will be considered on an ongoing basis until the position is filled.

To apply
Please submit a CV and cover letter describing your qualifications, research interests,
and reasons for pursuing a career in scientific publishing, as soon as possible, to our
online jobs site:
http://www.elsevier.com/wps/find/job_search.careers. Click on “search for US jobs”
and select “Massachusetts.”
Or:
http://reedelsevier.taleo.net/careersection/51/jobdetail.ftl?lang=en&job=SCI0005X.

No phone inquiries, please. Cell Press is an equal opportunity/affirmative action


employer, M/F/D/V.
Scientific Editor, Cell Metabolism
Cell Metabolism is seeking a full-time scientific editor to join its editorial team. Cell
Metabolism publishes metabolic research with an emphasis on molecular mechanisms
and translational medicine. The minimum qualification for this position is a PhD in a
relevant area of biomedical research, although additional postdoctoral and/or editorial
experience is preferred. This is a superb opportunity for a talented individual to play a
critical role in promoting science by helping researchers shape and disseminate their
findings to the wider community.

The scientific editor is responsible for assessing submitted research papers, overseeing
the refereeing process, and choosing, commissioning, and editing review material. The
scientific editor frequently travels to scientific conferences to follow developments in
research and establish and maintain close ties with the scientific community. The key
qualities we look for are breadth of scientific interest, the ability to think critically about
a wide range of scientific issues, and strong communication skills. The successful
candidate will also be highly motivated and creative and able to work independently
as well as in a team and should have opportunities to pioneer and contribute to new
trends in scientific publishing.

This is a full-time in-house editorial position, based at the Cell Press office in Cambridge,
Massachusetts. Cell Press offers an attractive salary and benefits package and a
stimulating working environment that encourages innovation.

Please submit a CV and cover letter describing your qualifications, general research
interests, and motivation for pursuing a career in scientific publishing. Applications will
be considered on an ongoing basis until the closing date of January 20, 2011.

To apply, visit
http://reedelsevier.taleo.net/careersection/51/jobdetail.ftl?lang=en&job=SCI0005Y.

No phone inquiries. Elsevier-Cell Press is an Equal Opportunity Employer.


Scientific Editor, Neuron
Neuron is seeking an additional full-time scientific editor to join its editorial team based
in Cambridge, Massachusetts. Neuron publishes across a range of disciplines includ-
ing developmental, molecular, cellular, systems, and cognitive neuroscience.

As a scientific editor, you would be responsible for assessing submitted research


manuscripts, overseeing the review process, and commissioning and editing review
material for the journal. You would also travel frequently to scientific conferences to
follow developments in research and to establish and maintain close ties with the
scientific community.

The minimum qualification for this position is a PhD in a relevant area of biomedical
research, although previous editorial experience is beneficial. This is a superb opportu-
nity for a talented individual to play a critical role in the research community away from
the bench. The key qualities we are looking for are breadth of scientific interest and the
ability to think critically about a wide range of scientific issues. The successful candi-
date will also be highly motivated and creative, possess strong communication skills,
and be able to both work independently and as part of a team.

This is a full-time, in-house editorial position, based at Cell Press headquarters in


Cambridge, Massachusetts. Cell Press offers an attractive salary and benefits package
and a stimulating work environment. Applications will be held in the strictest of confi-
dence and will be considered on an ongoing basis.

To apply
Please submit a cover letter describing your background, interests, and a candid
appraisal of the strengths and weaknesses of Neuron, along with your CV, to
http://reedelsevier.taleo.net/careersection/51/jobdetail.ftl?lang=en&job=SCI0006F.
Applications will be accepted through January 10, 2011.
The American Journal of Human Genetics
Editor Position Available

The American Society of Human Genetics is seeking an Editor for The American Journal of Human
Genetics. The Editor leads one of the world’s oldest and most prestigious journals publishing pri-
mary human genetics research.

Among the Editor’s responsibilities are determining the scope and direction of the scientific con-
tent of The Journal, overseeing manuscripts submitted for review and their publication, selecting
and supervising a staff consisting of an Editorial Assistant and doctoral-level Deputy Editor, direct-
ing interactions with the publisher (currently Cell Press), reviewing quarterly reports provided by the
publisher, evaluating the performance of the publisher, and if required, supervising the process of
the selection a new publisher. The Editor serves as a member of the Board of Directors of the Ameri-
can Society of Human Genetics (ASHG), as well as the ASHG Finance Committee, and presents
semiannual reports to the Board. All Associate Editors of The Journal are appointed by the Editor,
who also determines their duties. At the ASHG annual meeting, the Editor presides over a meeting
of the Associate Editors and presents an annual report to the ASHG membership.

The term of the appointment is five years and includes a yearly stipend. The new Editor will be
selected by the end of 2010 and will begin receiving manuscripts approximately in September 2011;
there will be partial overlap with the Boston office. Applicants should be accomplished scientists
in the field of human genetics and should have a broad knowledge and appreciation of the field.
Nominations, as well as applications consisting of a letter of interest and curriculum vitae, should be
sent to:

AJHG Editorial Search Committee


American Society of Human Genetics
9650 Rockville Pike
Bethesda, MD 20814
Announcements
Positions Available

UAB Stem Cell Institute


Department of Biochemistry and Molecular Genetics
Senior Faculty Position in Colorectal Cancer Biology University of Alabama at Birmingham
Department of Cancer Biology Schools of Medicine and Dentistry
Lerner Research Institute Tenure track junior faculty positions and tenured senior faculty
The Department of Cancer Biology is seeking a senior level positions are available for investigators focused on stem cell
biology, biochemistry, epigenetics and transplantation biology.
cancer biologist (Associate/Full Professor) with major basic
Areas of special emphasis include, but are not limited to,
research focus in colorectal cancer, animal models, and mechanistic studies of stem cell self-renewal and lineage
translational research. Research in colorectal cancer also specification and mechanisms of somatic cell reprogramming to
spans the Cleveland Clinic’s Department of Colorectal pluripotency. Structural biology of stem cell proteins by X-ray
Surgery (Digestive Diseases Institute), Department of crystallography and high-field NMR is an additional area of
Pathology, the Taussig Cancer Institute, and the Case interest. State of the art X-ray crystallography instrumentation and
Comprehensive Cancer Center. The position provides an a new 800MHz NMR system with a cryoprobe are available for
exceptional opportunity to integrate and translate discoveries Departmental faculty and Institute members. Nationally
competitive salaries, start-up packages and space allocations will
through collaborative interactions with outstanding basic and be offered to successful candidates. UAB is a highly interactive
clinical programs supported by a substantial network of Core environment with strong basic and clinical sciences. Birmingham
facilities, and comes with generous startup funds. is a beautiful and affordable city with many cultural attractions.
To be considered, applicants should have M.D., M.D./Ph.D., Applicants should send a C.V., a summary of research interests
or Ph.D. degrees and must have ongoing grant support and and the names of three references before January 31, 2011 to:
an accomplished research program in colorectal cancer. Dr. Tim Townes
Director, UAB Stem Cell Institute
Candidates should submit a curriculum vitae, summary of Chairman, Department of Biochemistry and
research plans, and three references, via e-mail to: Loreen Molecular Genetics, University of Alabama at Birmingham
Burke at burkel2@ccf.org. Kaul Genetics Building, Room 502
For further information see: 720 20th Street South
Birmingham, AL 35294
http://www.lerner.ccf.org/cancerbio/
Email: ttownes@uab.edu
Cleveland Clinic is an Equal Opportunity/ UAB is an Equal Opportunity Employer committed to
Affirmative Action Employer building a culturally diverse environment.

Look Again. Discover More.


• Access to the 14 Cell Press
primary research journals and
14 Trends reviews title, all on
the same platform
• Improved, more robust article and
author search

• Easy to navigate home page,


articles pages and archive

www.cell.com
Cell Press Business Project Editor
Position Available

Cell Press is seeking a Business Project Editor to plan, develop, and implement projects that have
commercial or sponsorship potential. By drawing on existing content or developing new material, the
Editor will work with Cell Press’s commercial sales group to create collections of content in print or
online that will be attractive to readers and sponsors. The Editor will also be responsible for leverag-
ing new online opportunities for engaging the readers of Cell Press journals.

The successful candidate will have a PhD in the biological sciences, broad scientific interests, a
fascination with technology, good commercial instincts, and a true passion for both science and
science communication. They should be highly organized and dedicated, with excellent written and
oral communication skills, and should be willing to work to tight deadlines.

The position is full time and based in Cambridge, MA. Cell Press offers an attractive salary and
benefits package and a stimulating work environment. Applications will be considered on a rolling
basis. For consideration, please apply online and include a cover letter and resume. To apply, visit
the career page at http://www.elsevier.com and search on keywords “Business Project Editor.”
EDITOR-IN-CHIEF SENIOR EDITORS ASSOCIATE EDITORS
F.E. Bloom J.F. Baker G. Aston-Jones T.A. Milner
La Jolla, CA, USA Chicago, IL, USA Charleston, SC, USA New York, NY, USA
P.R. Hof J.S. Baizer S.D. Moore
New York, NY, USA Buffalo, NY, USA Durham, NC, USA
G.R. Mangun J.D. Cohen T.H. Moran
Davis, CA, USA Princeton, NJ, USA Baltimore, MD, USA
J.I. Morgan B.M. Davis T.F. Münte
Memphis, TN, USA Pittsburgh, PA, USA Magdeburg, Germany
F.R. Sharp J. De Felipe K-C. Sonntag
Sacramento, CA, USA Madrid, Spain Belmont, MA, USA
R.J.Smeyne M.A. Dyer R.J. Valentino
Memphis, TN, USA Memphis, TN, USA Philadelphia, PA, USA
A.F. Sved M.S. Gold C.L. Williams
Pittsburgh, PA, USA Pittsburgh, PA, USA Durham,NC, USA
G.F. Koob
La Jolla, CA, USA

1
23 Twenty-three to
the Power of One.
One re-unified journal, nine specialist sections, 23 receiving Editors ←
Authors receive first editorial decision within 30 days of submission ←
“Young Investigator Awards” for innovative work by a new generation of researchers ←

Brain Research take another look


www.elsevier.com/locate/brainres
SnapShot: Inositol Phosphates
Ace J. Hatch and John D. York
HHMI, Pharmacology and Cancer Biology, Biochemistry, Duke University, Durham, NC 27710, USA

PLC-dependent IP code
GPCR RTK O

O O O O O
5-PP-IP 4 IP 4 5-IP 7
O O O O O O
PIP2 O IP6K O IP6K O VIP1
2 O O O
3 1
O ITPK1 O
O
PLC 2 O O O O O O O O
4 6 3 1
O 5 IP 3
6
IPMK IP 4 IPMK IP 5 IPK1 IP 6 1,5-IP8
4
O O 5 O O O O O O O O
ENZYMES O O O O O
VIP1 IP6K O
YEAST MAMMALIAN IP3K
IPMK O
PLC1 PLCβ, γ, δ, ε, ζ, η
- IP3KA, B, C
O O O O O O O O
- ITPK1 (IP56K)
IPK2(ARG82) IPMK (IPK2) IP 4 IP 3 IP 4 1-IP 7
IPK1 IPK1 (IP5K) INPP5 ITPK1
KCS1 IP6K1, 2, 3 O O O O O O
VIP1 VIP1, 2 (PPIP5K1, 2) O O

Ion channels Phosphate sensing Transcription


Cl- Abundant phosphate
MCM1 ARG80
CIC3 Pho80
P
PLASMA MEMBRANE
Cl- channel Pho4 Kinase
Kinase Assembly independent
Pho85 activity
CYTOPLASM
2 O PIP2 Pho81
3 1 CYTOPLASM NUCLEUS IPK2 ARG81
MCM1-ArgR
O
4
5
6
O Phosphate starvation complex
IP 4 O
O
O
O O O O
O O 1-IP 7 Kinase
Activation dependent
IP 3 O O Transcription O
O O activated
Pho80 IP 4
O
Pho85 Kinase activity
X Pho4 O O

blocked O
IP3 receptor ENDOPLASMIC Pho81
RETICULUM
Ca2+ CYTOPLASM NUCLEUS NUCLEUS

mRNA export and translation Insulin secretion and AKT Embryonic development


Effects of IP kinase deficiency
Translation termination
O IPMK (IPK2): Multiple defects, death by
embryonic day 10 (mice)
O O
Insulin IPK1: Cillia are shortened and immotile
IP 6 AKT resistance causing patterning defects (zebrafish)
O O
Multiple defects, death by
Ribosome O
embryonic day 8.5 (mice)
GleI eRF1 Insulin GSK3β
Dbp5 ITPK1 (IP56K): Neural tube
STOP defects (mice)
AAAAA O
IP3K: Sterility (nematodes)
mRNA O O
eRF3
5-IP 7 Multiple defects in immune
CYTOPLASM and neural cell development (mammals)
O O Adipogenesis
Nuclear mRNA export O IP6K2: Misregulated hedgehog signaling
O
O O
results in patterning defects (zebrafish)
O
IP 6 CYTOPLASM
O O
Other roles
GleI O
CYTOPLASM Dbp5 O
O O Inositol diphosphates can transfer phosphate
nonenzymatically to phosphoserine to
IP 6 Ca 2+, final release generate diphosphate modified proteins
NUCLEUS Nuclear pore O O
complex IP6K1 (KCS1) generated inositol diphosphates
mRNA O are required for proper regulation of telomere
length
RRP vesicles
IP structural cofactors Ipk2 regulates activity of Swi/Snf and Ino80
O
Insulin chromatin-remodeling complexes in yeast
O O
IP6K1 (KCS1) is required for proper vacuole
5-IP 7 morphology and responses to osmotic stress
O O Secretory
vesicles IP6 stimulates nonhomologous end joining
O
through interactions with Ku
O
IP6 (phytate) is important in plant biology
and agriculture as a major phosphate store
ADAR2 TIR1 β CELL

1030 Cell 143, December 10, 2010 ©2010 Elsevier Inc. DOI 10.1016/j.cell.2010.11.045 See online version for legend and references.
© 2010 Thermo Fisher Scientific Inc. All rights reserved.

Simplify Over-expression with Fully Sequenced cDNAs and ORFs.

Thermo Scientific Open Biosystems products include the world’s


largest and most complete collections of pre-made, full-length,
sequence-verified cDNA and open reading frame (ORF) clones for
reliable gene over-expression. Express your genes in less time, at a
better value with higher quality cDNA and ORF clones.

• Convenient – Pre-made clones, competitively priced,


ready to ship in 48 hours
• Innovative – Powerful lentiviral delivery with visual tracking
extends gene analysis to primary and non-dividing cells
• Comprehensive – The world’s largest collections of full-
length cDNA and ORF clones New! Thermo Scientific Open
Biosystems Precision LentiORF
• Guaranteed* sequence
Collection.
Expand your delivery options with genome-
Use our gene search and BLAST tool to find your gene of interest: wide, expression-ready lentiviral ORFs.
www.thermoscientific.com/GeneExpression
*The sequence of a cDNA clone is guaranteed to contain the BC accession sequence
associated with the clone using the gene search tool.

Moving science forward


better count fast
easy and reliable cell counting

Countess® Automated Cell Counter


The Countess® Automated Cell Counter takes subjectivity and tedium out of a fundamental cell
culture step—counting live and dead cells. With a proven track record and superior user interface,
now it's easy to visualize counted cells and verify performance, even with mixed-size cell populations.

go to www.invitrogen.com/gocountess
download the free mobile app at http://gettag.mobi
scan the barcode to instantly access more information about countess® cell counter

Life Technologies offers a breadth of products DNA | RNA | protein | cell culture | instruments
FOR RESEARCH USE ONLY. NOT INTENDED FOR ANY ANIMAL OR HUMAN THERAPEUTIC OR DIAGNOSTIC USE, UNLESS OTHERWISE STATED.
© 2010 Life Technologies Corporation. All rights reserved. The trademarks mentioned herein are the property of Life Technologies Corporation or their respective owners, unless otherwise noted.

You might also like