Download as pdf or txt
Download as pdf or txt
You are on page 1of 55

G protein-coupled receptors : signaling,

trafficking and regulation First Edition


Shukla
Visit to download the full and correct content document:
https://ebookmass.com/product/g-protein-coupled-receptors-signaling-trafficking-and-
regulation-first-edition-shukla/
Methods in Cell
Biology
G Protein-Coupled Receptors:
Signaling, Trafficking and
Regulation

Volume 132
Series Editors

Leslie Wilson
Department of Molecular, Cellular and Developmental Biology
University of California
Santa Barbara, California

Phong Tran
University of Pennsylvania
Philadelphia, USA &
Institut Curie, Paris, France
Methods in Cell
Biology
G Protein-Coupled Receptors:
Signaling, Trafficking and
Regulation

Volume 132

Edited by

Arun K. Shukla
Department of Biological Sciences and Bioengineering,
Indian Institute of Technology, Kanpur, India

AMSTERDAM • BOSTON • HEIDELBERG • LONDON


NEW YORK • OXFORD • PARIS • SAN DIEGO
SAN FRANCISCO • SINGAPORE • SYDNEY • TOKYO

Academic Press is an imprint of Elsevier


Academic Press is an imprint of Elsevier
50 Hampshire Street, 5th Floor, Cambridge, MA 02139, USA
525 B Street, Suite 1800, San Diego, CA 92101-4495, USA
125 London Wall, London EC2Y 5AS, UK
The Boulevard, Langford Lane, Kidlington, Oxford OX5 1GB, UK
First edition 2016
Copyright © 2016 Elsevier Inc. All Rights Reserved.

No part of this publication may be reproduced or transmitted in any form or by any means,
electronic or mechanical, including photocopying, recording, or any information storage
and retrieval system, without permission in writing from the publisher. Details on how to
seek permission, further information about the Publisher’s permissions policies and our
arrangements with organizations such as the Copyright Clearance Center and the
Copyright Licensing Agency, can be found at our website: www.elsevier.com/permissions.
This book and the individual contributions contained in it are protected under copyright by
the Publisher (other than as may be noted herein).

Notices
Knowledge and best practice in this field are constantly changing. As new research and
experience broaden our understanding, changes in research methods, professional
practices, or medical treatment may become necessary.
Practitioners and researchers must always rely on their own experience and knowledge in
evaluating and using any information, methods, compounds, or experiments described
herein. In using such information or methods they should be mindful of their own safety
and the safety of others, including parties for whom they have a professional responsibility.

To the fullest extent of the law, neither the Publisher nor the authors, contributors, or
editors, assume any liability for any injury and/or damage to persons or property as a
matter of products liability, negligence or otherwise, or from any use or operation of any
methods, products, instructions, or ideas contained in the material herein.

ISBN: 978-0-12-803595-5
ISSN: 0091-679X

For information on all Academic Press publications


visit our website at http://store.elsevier.com
Contributors

Agnes M. Acevedo Canabal


Institute of Neurobiology, University of Puerto Rico Medical Sciences Campus,
San Juan, PR, USA; Department of Anatomy and Neurobiology, School of
Medicine, University of Puerto Rico, San Juan, PR, USA
D. Agranovich
Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center,
Jerusalem, Israel
Stefan Amisten
Diabetes Research Group, King’s College London, London, UK
Gabriela Antunes
Laboratory of Neural Systems (SisNE), Department of Physics, Faculdade de
Filosofia Ciências e Letras de Ribeirão Preto, Universidade de São Paulo,
Ribeirão Preto, Brazil
Chaitanya A. Athale
Division of Biology, IISER Pune, Pune, India
Nicolas Audet
Department of Pharmacology and Therapeutics, McGill University, Montreal, QC,
Canada
Mohammed Akli Ayoub
Biologie et Bioinformatique des Systèmes de Signalisation, Institut National de la
Recherche Agronomique, UMR85, Unité Physiologie de la Reproduction et des
Comportements; CNRS, UMR7247, Nouzilly, France; LE STUDIUMÒ Loire Valley
Institute for Advanced Studies, Orléans, France
R. Bar-Shavit
Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center,
Jerusalem, Israel
Damian Bartuzi
Department of Synthesis and Chemical Technology of Pharmaceutical
Substances with Computer Modelling Lab, Faculty of Pharmacy with Division of
Medical Analytics, Medical University of Lublin, Lublin, Poland
Maik Behrens
Department of Molecular Genetics, German Institute of Human Nutrition
Potsdam-Rehbruecke, Nuthetal, Germany

xiii
xiv Contributors

Nicolas F. Berbari
Department of Biology, Indiana University-Purdue University Indianapolis,
Indianapolis, IN, USA
Hélène Bonin
Department of Biochemistry and Molecular Medicine, Institute for Research in
Immunology and Cancer, Université de Montréal, Montreal, QC, Canada
Michel Bouvier
Department of Biochemistry and Molecular Medicine, Institute for Research in
Immunology and Cancer, Université de Montréal, Montreal, QC, Canada
Amitabha Chattopadhyay
CSIR-Center of Cellular and Molecular Biology, Hyderabad, India
Linjie Chen
Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang
University, Hangzhou, Zhejiang, China
Santiago Cuevas
Division of Renal Diseases & Hypertension, Department of Medicine, The George
Washington University School of Medicine and Health Sciences, WA, USA
Francheska Delgado-Peraza
Institute of Neurobiology, University of Puerto Rico Medical Sciences Campus,
San Juan, PR, USA; Department of Anatomy and Neurobiology, School of
Medicine, University of Puerto Rico, San Juan, PR, USA
Dominic Devost
Department of Pharmacology and Therapeutics, McGill University, Montréal, QC,
Canada
Antonella Di Pizio
Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith
Faculty of Agriculture, Food and Environment, The Hebrew University, Rehovot,
Israel
Shalini Dogra
Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, Uttar
Pradesh, India
Zyanya P. Espinosa-Riquer
Departamento de Farmacobiologı́a, Centro de Investigación y de Estudios
Avanzados del IPN, México D.F., Mexico
Timothy N. Feinstein
Department of Developmental Biology, University of Pittsburgh School of
Medicine, Pittsburgh, PA, USA
Contributors xv

Colleen A. Flanagan
School of Physiology and Medical Research Council Receptor Biology Research
Unit, Faculty of Health Sciences, University of the Witwatersrand, Wits Parktown,
Johannesburg, South Africa
Alexandre Gidon
Molecular Mechanisms of Mycobacterial Infection, Center for Molecular
Inflammation Research, Norwegian University of Science and Technology,
Trondheim, Norway
Claudia González-Espinosa
Departamento de Farmacobiologı́a, Centro de Investigación y de Estudios
Avanzados del IPN, México D.F., Mexico
S. Grisaru-Granovsky
Department of Obstetrics and Gynecology, Shaare Zedek, Jerusalem, Israel
Aylin C. Hanyaloglu
Institute of Reproductive and Developmental Biology, Imperial College London,
London, UK
Terence E. Hébert
Department of Pharmacology and Therapeutics, McGill University, Montréal, QC,
Canada
Mellisa M. Hege
Department of Biology, Indiana University-Purdue University Indianapolis,
Indianapolis, IN, USA
Ilpo Huhtaniemi
Institute of Reproductive and Developmental Biology, Imperial College London,
London, UK
M. Jaber
Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center,
Jerusalem, Israel
Kim C. Jonas
Institute of Reproductive and Developmental Biology, Imperial College London,
London, UK; Institute of Medical and Biomedical Education, St George’s
University of London, London, UK
Pedro A. Jose
Division of Renal Diseases & Hypertension, Department of Medicine, The George
Washington University School of Medicine and Health Sciences, WA, USA
Manali Joshi
Savitribai Phule Pune University, Pune, India
xvi Contributors

Agnieszka A. Kaczor
Department of Synthesis and Chemical Technology of Pharmaceutical
Substances with Computer Modelling Lab, Faculty of Pharmacy with Division of
Medical Analytics, Medical University of Lublin, Lublin, Poland; School of
Pharmacy, University of Eastern Finland, Kuopio, Finland
A. Kancharla
Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center,
Jerusalem, Israel
Rafik Karaman
Bioorganic Chemistry Department, Faculty of Pharmacy, Al-Quds University,
Jerusalem, Israel
Hiroyuki Kobayashi
Department of Biochemistry and Molecular Medicine, Institute for Research in
Immunology and Cancer, Université de Montréal, Montreal, QC, Canada
Ajeet Kumar
Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, Uttar
Pradesh, India
Christian Le Gouill
Department of Biochemistry and Molecular Medicine, Institute for Research in
Immunology and Cancer, Université de Montréal, Montreal, QC, Canada
Anat Levit
Department of Pharmaceutical Chemistry, University of California e San
Francisco, San Francisco, CA, USA
Bin Lu
Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang
University, Hangzhou, Zhejiang, China
Viktorya Lukashova
Department of Biochemistry and Molecular Medicine, Institute for Research in
Immunology and Cancer, Université de Montréal, Montreal, QC, Canada
Marina Macı́as-Silva
Departamento de Biologı́a Celular y Desarrollo, Instituto de Fisiologı́a Celular,
Universidad Nacional Autónoma de México, México D.F., Mexico
M. Maoz
Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center,
Jerusalem, Israel
Contributors xvii

Dariusz Matosiuk
Department of Synthesis and Chemical Technology of Pharmaceutical
Substances with Computer Modelling Lab, Faculty of Pharmacy with Division of
Medical Analytics, Medical University of Lublin, Lublin, Poland
Jeremy C. McIntyre
Department of Neuroscience, University of Florida, Gainesville, FL, USA; Center
for Smell and Taste, University of Florida, Gainesville, FL, USA
Masha Y. Niv
Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith
Faculty of Agriculture, Food and Environment, The Hebrew University, Rehovot,
Israel; Fritz Haber Center for Molecular Dynamics, The Hebrew University,
Jerusalem, Israel
Carlos Nogueras-Ortiz
Institute of Neurobiology, University of Puerto Rico Medical Sciences Campus,
San Juan, PR, USA
Melanie Philipp
Institute for Biochemistry and Molecular Biology, Ulm University, Ulm, Germany
Cristina Roman-Vendrell
Institute of Neurobiology, University of Puerto Rico Medical Sciences Campus,
San Juan, PR, USA; Department of Physiology, School of Medicine, University of
Puerto Rico, San Juan, PR, USA
Ewelina Rutkowska
Department of Biopharmacy, Faculty of Pharmacy with Division of Medical
Analytics, Medical University of Lublin, Lublin, Poland
Jana Selent
Research Programme on Biomedical Informatics (GRIB), Universitat Pompeu
Fabra, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
Durba Sengupta
CSIR-National Chemical Laboratory, Pune, India
Ying Shi
Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang
University, Hangzhou, Zhejiang, China
Fabio Marques Simoes de Souza
Center for Mathematics, Computation and Cognition, Federal University of ABC,
São Bernardo do Campo, Brazil
xviii Contributors

Michal Slutzki
Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith
Faculty of Agriculture, Food and Environment, The Hebrew University, Rehovot,
Israel
Chandan Sona
Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, Uttar
Pradesh, India
Katarzyna M. Targowska-Duda
Department of Biopharmacy, Faculty of Pharmacy with Division of Medical
Analytics, Medical University of Lublin, Lublin, Poland
Teresa Casar Tena
Institute for Biochemistry and Molecular Biology, Ulm University, Ulm, Germany
B. Uziely
Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center,
Jerusalem, Israel
Genaro Vázquez-Victorio
Departamento de Biologı́a Celular y Desarrollo, Instituto de Fisiologı́a Celular,
Universidad Nacional Autónoma de México, México D.F., Mexico
Jean-Pierre Vilardaga
Laboratory for GPCR Biology, Department of Pharmacology & Chemical Biology,
University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
Van Anthony M. Villar
Division of Renal Diseases & Hypertension, Department of Medicine, The George
Washington University School of Medicine and Health Sciences, WA, USA
Richard Wargachuk
Department of Pharmacology and Therapeutics, McGill University, Montréal, QC,
Canada
Kunhong Xiao
Laboratory for GPCR Biology, Department of Pharmacology & Chemical Biology,
University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
Prem N. Yadav
Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, Uttar
Pradesh, India
Guillermo A. Yudowski
Institute of Neurobiology, University of Puerto Rico Medical Sciences Campus,
San Juan, PR, USA; Department of Anatomy and Neurobiology, School of
Medicine, University of Puerto Rico, San Juan, PR, USA
Contributors xix

Yaping Zhang
Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang
University, Hangzhou, Zhejiang, China
Xiaoxu Zheng
Division of Renal Diseases & Hypertension, Department of Medicine, The George
Washington University School of Medicine and Health Sciences, WA, USA
Cynthia Zhou
Department of Pharmacology and Therapeutics, McGill University, Montréal, QC,
Canada
Naiming Zhou
Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang
University, Hangzhou, Zhejiang, China
Preface

G proteinecoupled receptors (GPCRs) also referred as seven transmembrane


receptors (7TMRs) lie at the heart of almost every physiological and pathophysio-
logical process in our body. These receptors bind to and get activated by a wide
range of ligands ranging from small molecules, hormones, peptides, proteins to
lipids. The overall activation and signal transduction mechanisms of GPCRs are
highly conserved where binding of an agonist results in a conformational change
in the receptor followed by activation of heterotrimeric G proteins and subsequent
generation of second messengers and downstream signaling. Downregulation of
GPCRs is also primarily a conserved process where activated receptors are
phosphorylated by GRKs (GPCR kinases) followed by binding of beta arrestins
which leads to receptor desensitization and internalization. GPCRs are targeted by
about one-third of the currently prescribed drugs which include angiotensin blockers
for hypertension, beta-blockers for heart failure, antihistamines for allergy manage-
ment, and opioid agonists as analgesic medication.
In this volume of Methods in Cell Biology, we cover multiple aspects of GPCR
signaling, trafficking, regulation, and cellular assays in a form of either an over-
view or as step-by-step protocol. This is an effort to bring together different
domains of GPCR pharmacology and signaling on to a common platform and high-
light the incredibly versatile nature and diverse functional manifestation of
GPCRs. Section I includes chapters on GPCR trafficking in lipid rafts and cilia,
imaging endogenous receptor in neurons, single molecule imaging of GPCRs,
and a comprehensive analysis of GPCRs in adipose tissue. In Section II, we cover
topics ranging from GPCR signaling from endosomes, olfactory receptor signal
transduction, studies of a specialized GPCR smoothened in zebra fish model,
and the outcome of GPCR signaling in cytoskeletal dynamics. In recent years, a
key focus area in GPCR biology has been the development of novel and more sen-
sitive cellular assays to investigate GPCR expression, signaling, and downregula-
tion. Section III of this volume is focused on GPCR assays which include classical
radioligand binding, label-free, biosensor and fluorescenceebased approaches to
study GPCR trafficking and signaling, and TANGO assay for measuring GPCR-
beta-arrestin interaction. Finally, Section IV consists of chapters on structural
and computational aspects of protease-activated receptors, bitter taste receptors,
and GPCR dimerization.
I would like to thank all the authors who have contributed to this focused volume
despite their busy schedule. I also express my sincere gratitude to the journal edito-
rial staff and production team for a wonderful job in putting this volume together in a
timely fashion. With this brief background, on behalf of the entire Methods in Cell

xxi
xxii Preface

Biology Team, I present to you this volume entitled “G ProteineCoupled Receptors:


Signaling, Trafficking, and Regulation.” I sincerely hope that you enjoy the topics
covered in this issue and please feel free to share your feedback with us.

Arun K. Shukla
Indian Institute of Technology, Kanpur, India
CHAPTER

Localization and signaling


of GPCRs in lipid rafts

Van Anthony M. Villar1, Santiago Cuevas, Xiaoxu Zheng, Pedro A. Jose1


1
Division of Renal Diseases & Hypertension, Department of Medicine, The George Washington
University School of Medicine and Health Sciences, WA, USA
1
Corresponding authors: E-mail: vvillar@gwu.edu; pjose@mfa.gwu.edu

CHAPTER OUTLINE
Introduction ................................................................................................................ 4
1. Localization of GPCRs in Lipid Rafts ........................................................................ 6
1.1 Isolation of Lipid Rafts ............................................................................ 7
1.1.1 Detergent-free method.......................................................................... 7
1.1.2 Detergent-based method ...................................................................... 9
1.1.3 Immunoblotting and data interpretation............................................... 10
1.2 Localization of GPCRs in Lipid Rafts....................................................... 11
1.2.1 Cells in suspension............................................................................. 13
1.2.2 Adherent cells .................................................................................... 14
2. GPCR Signaling in Lipid Rafts ............................................................................... 15
2.1 Perturbation of Raft Stability.................................................................. 15
2.2 Changing the Cholesterol Content ........................................................... 16
2.3 Fluorescence Imaging............................................................................ 16
References ............................................................................................................... 18

Abstract
The understanding of how biological membranes are organized and how they function
has evolved. Instead of just serving as a medium in which certain proteins are found,
portions of the lipid bilayer have been demonstrated to form specialized platforms that
foster the assembly of signaling complexes by providing a microenvironment that is
conducive for effective proteineprotein interactions. G protein-coupled receptors
(GPCRs) and relevant signaling molecules, including the heterotrimeric G proteins, key
enzymes such as kinases and phosphatases, trafficking proteins, and secondary messen-
gers, preferentially partition to these highly organized cell membrane microdomains,
called lipid rafts. As such, lipid rafts are crucial for the trafficking and signaling of
GPCRs. The study of GPCR biology in the context of lipid rafts involves the localization
of the GPCR of interest in lipid rafts, at the basal state and upon receptor agonism, and

Methods in Cell Biology, Volume 132, ISSN 0091-679X, http://dx.doi.org/10.1016/bs.mcb.2015.11.008 3


© 2016 Elsevier Inc. All rights reserved.
4 CHAPTER 1 GPCRs in lipid rafts

the evaluation of the biological functions of the GPCR in appropriate cell lines. The lack
of standardized methodology to study lipid rafts, in general, and of the workings of
GPCRs in lipid rafts, in particular, and the inherent drawbacks of current methods have
hampered the complete understanding of the underlying molecular mechanisms. Newer
methodologies that allow the study of GPCRs in their native form are needed. The use of
complementary approaches that produce mutually supportive results appear to be the best
way for drawing conclusions with regards to the distribution and activity of GPCRs in
lipid rafts.

INTRODUCTION
Lipid Raft Microdomains. The plasma membrane is a semipermeable, biological
membrane that demarcates the intracellular milieu from the extracellular environ-
ment. Amphipathic lipids, such as phospholipids and sphingolipids, are the building
blocks of these bilipid membranes because of their aggregative properties, i.e., their
hydrophobic tails associate together, while their hydrophilic heads interact with both
extra- and intracellular aqueous environments (Sonnino & Prinetti, 2013). The
fluidity of the fatty acyl groups of phospholipids at 37  C enables the membranes
to act as a medium in which dissolved membrane proteins are afforded ample lateral
mobility, especially in response to environmental cues. Since the first description of
an “organization of the lipid components of membranes into domains” (Karnovsky
et al., 1982) and the elaboration of the “lipid raft hypothesis” by Simons and van
Meer (van Meer & Simons, 1988; Simons & Ikonen, 1997; Simons & van Meer
1998), the existence of lipid rafts is now established.
Lipid rafts are tightly packed, highly organized plasma membrane microdomains
that are enriched in phospholipids, glycosphingolipids, and cholesterol and serve as
a platform for the organization and dynamic interaction of biomolecules involved in
various biological processes (Figure 1). The cholesterol bestows a semblance of
rigidity and order by intertwining into the hydrophobic gaps between the phospho-
lipid acyl chains. Certain structural proteins abound in lipid rafts to serve as scaffold
or anchor for other proteins, including caveolins (Head, Patel, & Insel, 2014; Quest,
Leyton, & Párraga, 2004; Yu, Villar, & Jose, 2013; Yu et al., 2004), flotillins
(Rajendran, Le Lay, & Illges, 2007; Yu et al., 2004) and tetraspanins (Hemler,
2005), and glycosylphosphatidylinositol-linked (GPI-linked) proteins. The spatial
concentration and organization of specific sets of membrane proteins allow greater
efficiency and specificity of signal transduction by facilitating proteineprotein
interactions and by preventing crosstalk between competing pathways. The
nonhomogeneous lateral distribution of membrane components helps explain the
differences in composition between apical and basolateral membrane domains of
polarized epithelial cells (Sonnino & Prinetti, 2013).
The best characterized lipid raft microdomains are the caveolae, which were first
described by Palade and Yamada in the 1950s (Palade, 1953; Yamada, 1955). These
are small (60e80 nm) invaginations of the plasma membrane formed by the
polymerization of caveolins with cholesterol (Parton & del Pozo, 2013). Caveolae
Introduction 5

FIGURE 1 A Lipid Raft Membrane Microdomain.


Lipid rafts are highly organized plasma membrane microdomains enriched in phospholipids,
glycosphingolipids, and cholesterol, and serve as matrix for receptors, such as G protein-
coupled receptors (GPCRs), and other signaling molecules. (See color plate)
Van Anthony M. Villar, MD, PhD.

have been implicated in a variety of cellular processes, including signal transduc-


tion, endocytosis, transcytosis, and cholesterol trafficking (Barnett-Norris, Lynch, &
Reggio, 2005). Lipid rafts accumulate in the apical plasma membrane in polarized
epithelial cells and in axonal membranes in neurons. Basolateral and dendritic
membranes contain lipid rafts but in more limited quantities (Simons & Ikonen,
1997). Interestingly, caveolae are found mostly at the basolateral membrane that
faces the blood supply and is more active during signal transduction (Simons &
Toomre, 2000). Lipid rafts are mostly found at the plasma membrane; however,
they may also be found in intracellular membranes involved in the biosynthetic
and endocytic pathways. Lipid raft microdomains play a crucial role in cellular pro-
cesses such as membrane sorting, receptor trafficking, signal transduction, and cell
adhesion.
GPCR Signaling and Trafficking. G protein-coupled receptors (GPCRs)
constitute the largest superfamily of seven transmembrane proteins that respond
to a myriad of environmental stimuli that are transduced intracellularly as meaning-
ful signals through secondary messengers. Agonist stimulation of a GPCR leads to a
conformational change that promotes the exchange of GDP for GTP on the Ga sub-
unit of the G protein, resulting in the uncoupling of the G protein from the GPCR and
the dissociation of Ga and Gbg subunits. The Ga subunit either activates or inhibits
intracellular signaling pathways depending on the receptor subtype, while the Gbg
subunit recruits G protein-coupled receptor kinases which selectively phosphorylate
serine and threonine residues localized within the third intracellular loop and
carboxyl-terminal tail domains of the receptor to promote the binding of cytosolic
cofactor proteins called arrestins (Lefkowitz, 1998). The b-arrestins play a pivotal
role in the uncoupling process and in the sequestration and internalization of GPCRs
6 CHAPTER 1 GPCRs in lipid rafts

through a dynamin-dependent, clathrin-mediated endocytosis. Once internalized,


the GPCRs, in vesicles termed as early endosomes, are sorted by sorting nexins
and follow divergent pathways (Worby & Dixon, 2002). The receptors are sorted
into recycling endosomes for their return to the cell membrane (recycling and
resensitization), accumulate in late endosomes which target the lysosomes for their
subsequent degradation, or transported initially to the perinuclear endosomes (trans-
Golgi network) and then to the late endosomes for eventual lysosomal degradation.
Additional proteolytic mechanisms, such as proteasomes or cell-associated endo-
peptidases, are also implicated in mediating the downregulation of certain GPCRs
(von Zastrow, 2003).
The signal transduction that follows ligand occupation of the GPCR is highly
regulated to ensure the specificity of the cellular response, both temporally and
spatially. The signal transduction can be attenuated with relatively fast kinetics
through a process called desensitization or through a much slower process of down-
regulation following prolonged or repeated exposure to an agonist. Desensitization,
or the waning of a receptor’s responsiveness to agonist with time, is an inherent
molecular “feedback” mechanism that prevents receptor overstimulation and helps
in creating an integrated and meaningful signal by filtering out information from
weaker GPCR-mediated signals (Ferguson, 2001).
It is accomplished through two complementary mechanisms, i.e., the functional
uncoupling of GPCRs from their cognate G proteins, which occurs without any
detectable change in the number of cell surface receptors, and GPCR phosphoryla-
tion, sequestration, and internalization/endocytosis. GPCR resensitization protects
the cells from prolonged desensitization and is carried out via dephosphorylation
by phosphatases as the GPCR traffics through the endosomal pathway. GPCR activ-
ity is the net result of a coordinated balance between receptor desensitization and
resensitization.
It is now established that lipid rafts serve as dynamic platforms for GPCRs and
pertinent signaling molecules such as G proteins, enzymes, and adaptors (Barnett-
Norris et al., 2005; Lingwood & Simons, 2010). However, understanding the
molecular mechanisms involved has been hampered by the lack of standardized
methodology to study lipid rafts, in general, and of the workings of GPCRs in lipid
rafts, in particular. Moreover, the minute size of lipid rafts has made lipid rafts
difficult to resolve by standard light microscopy, unless the lipid raft components
are cross-linked with antibodies or lectins (Simons & Toomre, 2000). Studying
how GPCR works in lipid rafts may be accomplished by determining if the
GPCR of interest localizes to the lipid rafts and by evaluating if GPCR signaling
and activity are lost when lipid rafts are disrupted.

1. LOCALIZATION OF GPCRs IN LIPID RAFTS


Several techniques are available for the detection and localization of GPCRs in lipid
raft microdomains in cells. The most commonly employed approach utilizes cell
1. Localization of GPCRs in lipid rafts 7

fractionation procedures that break the cells apart and destroy cell morphology
before GPCR analysis using biochemical or immunological assays. A complemen-
tary biophysical approach involves the visualization of GPCRs in intact cell
membranes.

1.1 ISOLATION OF LIPID RAFTS


Lipid rafts are characterized by their relative insolubility in nonionic detergents at
4  C and light buoyant density on sucrose gradient (Schnitzer, McIntosh, Dvorak,
Liu, & Oh, 1995). The isolation of lipid rafts can be performed using either
detergent-based or detergent-free methods (Yu et al., 2013), with the latter generating
a greater fraction of inner leaflet membrane rafts and producing more replicable
results (Pike, 2004). Schnitzer et al. (1995) employed a detergent-free method to
isolate lipid rafts using cationic colloidal silica particles, which is appropriate for
non-cell culture studies. Lipid rafts may be extracted from total cell membranes
(Song et al., 1996) or just from surface plasma membranes (Smart, Ying, Mineo, &
Anderson, 1995). Detergent insolubility results from the segregation of membrane-
associated proteins into the lipid rafts, which are abundant in cholesterol and
glycosphingolipids. Nonionic detergents, such as Triton X-100, b-octyl glucoside,
CHAPS, deoxycholate, Lubrol WX, Lubrol PX, Brij 58, Brij 96, and Brij 98, have
been used to prepare lipid raft fractions (Macdonald & Pike, 2005), resulting in
varying yields of proteins. Samples obtained by detergent-based methods are termed
detergent-resistant membranes or detergent-insoluble fractions. Different detergents
may yield different lipid raft components because of the varying degrees of resis-
tance by the proteins to extraction using different reagents. The methods detailed
below are based on Yu et al. (2013).

1.1.1 Detergent-free method


Materials
2-N-morpholino ethanesulfonic acid (Mes), 250 mM, pH ¼ 6.8
Mes-buffered solution (MBS), 25 mM Mes þ 150 mM NaCl
Sodium citrate, 500 mM, pH w 11 (add protease inhibitors)
Sucrose, 5%, 35%, and 80% in MBS solution (add protease inhibitors)
Methyl-b-cyclodextrin (b-MCD), 2% dissolved in cell culture media
Cholesterol þ b-MCD (Sigma catalog #C4951), dissolved in cell culture
media
1X PBS, for washing
1. Cell culture and cell pellet collection. To obtain sufficient amounts of lipid raft
fraction, cells should be grown in 150-mm dishes until almost confluent using
the appropriate media at 37  C with 95% air and 5% CO2. Separate dishes of
cells should also be treated for cholesterol depletion and repletion as experi-
mental controls (Figure 2). Cholesterol depletion to disrupt the lipid rafts is
commonly performed by pretreatment with b-MCD for 1 h at 37  C.
8 CHAPTER 1 GPCRs in lipid rafts

FIGURE 2 Comparison groups for GPCR localization in lipid rafts.

Methyl-a-cyclodextrin (a-MCD) may be used as control for b-MCD (Vial &


Evans, 2005). Cholesterol repletion is performed by pretreating with choles-
terol/b-MCD solution for 1 h at 37  C. Cholestane-3,5,6-triol, an inactive
analog of cholesterol, may be used as control for the use of exogenous
cholesterol (Murtazina, Kovbasnjuk, Donowitz, & Li, 2006). To determine the
effect of agonist or antagonist treatment, cells should be serum-starved for at
least 1 h prior to treatment to achieve “basal” conditions prior to treatment.
Additional controls, such as the use of the drug vehicle, should be concomi-
tantly performed.
1.1 Wash cells with cold PBS and scrape the cells using a rubber-tipped cell
scraper.
1.2 Transfer cell suspension into 15-mL tube and spin at 2000 g for 5 min.
1.3 Decant the supernatant to obtain the cell pellet.
2. Cell homogenate preparation. All steps are carried out at 4  C.
2.1 To the cell pellet, add 1.5 mL 500 mM sodium carbonate and vortex.
2.2 Homogenize the cell suspension by sonication using five 20-s bursts on ice.
2.3 Add 1.5 mL of 80% sucrose and mix by vortex and sonication (three 20-s
bursts) on ice. Protein concentration may be determined at this time using a
BCA kit.
3. Sucrose gradient ultracentrifugation. Prepare 5%, 35%, and 80% sucrose
solutions in MBS solution. The use of MBS solution with pH close to 7.0 may
be advantageous for most proteins.
3.1 Place 3 mL of cell homogenates into the bottom of precooled 12-mL
ultracentrifuge tubes.
3.2 Overlay sequentially 4.5 mL of 35% sucrose and 4.5 mL of 5% sucrose to
each tube.
3.3 With the tubes securely balanced in an SW-41 bucket, spin at 180,000 g
(38,000 rpm) for 16 h at 4  C in a Beckman SW-41 centrifuge.
4. Lipid raft fraction preparation. A light-scattering band that is enriched with
caveolae/lipid rafts can be observed between the 5% and 35% sucrose gradients
and corresponds to the fourth fraction.
4.1 Carefully aspirate 12 1-mL fractions from the top of the tube and transfer
into prelabeled 1.5 microcentrifuge tubes.
1. Localization of GPCRs in lipid rafts 9

4.2 Prepare 0.5 mL of each fraction by adding 0.1 mL 6X sample buffer, vortex,
and boil for 5 min before use for immunoblotting. These samples can be
stored at 20  C, while the rest of the fractions without the 6X sample
buffer can be stored at 80  C.

1.1.2 Detergent-based method


Materials
50% Optiprep Stock solution (45 mL of 60% Optiprep þ 9 mL of Optiprep
diluent)
MBSTS buffer (MBS þ 0.5% Triton X-100 þ protease inhibitors in 10% sucrose)
Sucrose solutions (Table 1):
Table 1 Preparation of Optiprep Gradient Solutions
Solution
(5 mL total
volume) 30% Sucrose 20% Sucrose 10% Sucrose 5% Sucrose
50% Optiprep 3.0 2.0 1.0 0.5
(mL)
MBSTS (mL) 2.0 3.0 4.0 4.5

1. Cell culture and cell pellet preparation. The same as with the detergent-free
method.
2. Cell extract preparation.
2.1 Add 0.3 mL ice-cold MBSTS to cell pellet and push through a 25G
needle 10.
2.2 Adjust cell extract (w0.4 mL; cell pellet volume is w0.1 mL) to 40%
Optiprep by adding 0.8 mL of cold 60% Optiprep and vortex. Determine
protein concentration using a BCA kit.
3. Optiprep gradient ultracentrifugation.
3.1 Place 1 mL of the cell extract into the bottom of precooled 5-mL ultra-
centrifuge tubes.
3.2 Overlay with 1 mL each of 30%, 25%, 20%, and 0% Optiprep solutions in
MBSTS buffer.
3.3 Secure each tube in a Beckman SW 50.1 bucket and spin at 175,000 g
(42,000 rpm) at 4  C for 4 h. Other rotors may be used, such as the SW 55
(170,000 g for 4 h) or TLS55 (250,000 g for 2.5 h).
4. Lipid raft fraction preparation.
4.1 Carefully aspirate ten 0.5-mL fractions from the top of the tube and transfer
into prelabeled 1.5 microcentrifuge tubes.
4.2 Prepare 0.25 mL of each fraction by adding 0.5 mL 6X sample buffer,
vortex, and boil for 5 min before use for immunoblotting. These samples
can be stored at 20  C, while the rest of the fractions without the 6X
sample buffer can be stored at 80  C.
10 CHAPTER 1 GPCRs in lipid rafts

1.1.3 Immunoblotting and data interpretation


Western blot is the most commonly used method to determine the lipid raft distribu-
tion of proteins, such as GPCRs. Antibody specificity is crucial for the identification
of the GPCR of interest. The lipid raft proteins are found in the more buoyant
fractions (top 5e6 fractions); however, their distribution among these fractions is
not uniform. Immunoblotting for lipid raft markers may help in determining the
fractions where the lipid rafts are most abundant. Caveolin-1 is the most commonly
used protein marker for lipid rafts, specifically for caveolae (Insel et al., 2005;
Lingwood & Simons, 2010). There are several other markers for lipid rafts, such
as flotillin-1, CD55, alkaline phosphatase, and pore-forming toxins, such as cholera
toxin subunit B (CTxB), equinatoxin II, perfringolysin (Foster, De Hoog, & Mann,
2003; Salzer & Prohaska, 2001; Skocaj et al., 2013). Flotillin-1 has been used as a
lipid raft marker protein in cells that do not contain caveolae, i.e., blood cells
(Salzer & Prohaska, 2001), neural cells (Huang et al., 2007), and rat renal proximal
tubule cells (Breton, Lisanti, Tyszkowski, McLaughlin, & Brown, 1998; Riquier, Lee,
& McDonough, 2009) and human embryonic kidney (HEK)-293 cells (Yu et al.,
2004). There is species specificity because human renal proximal tubule cells
express caveolin-1 (Gildea et al., 2009), while HEK-293 cells express caveolin-2.
These markers may also be used to indicate the integrity of lipid rafts in cholesterol
depletion or repletion experiments. In general, these markers should be distributed in
the more buoyant fractions and should redistribute into the less buoyant fractions
(fractions 7e12) after cholesterol depletion with b-MCD (Figure 3). Cholesterol
repletion reconstitutes the lipid rafts and thus, these markers should be observed in
the more buoyant fractions.

FIGURE 3 Lipid Raft Distribution of Caveolin-1 and D1R.


Lipid raft and non-lipid raft fractions from human renal proximal tubule cells treated with
b-MCD, a cholesterol-depleting and lipid raft-disrupting agent, were prepared by detergent-
free method and sucrose gradient ultracentrifugation. The distribution of caveolin-1, a lipid
raft marker, and the dopamine D1 receptor (D1R), a GPCR, is shown in the immunoblots.
Images are courtesy of Peiying Yu, MD.
1. Localization of GPCRs in lipid rafts 11

1.2 LOCALIZATION OF GPCRs IN LIPID RAFTS


Another way to demonstrate the distribution of GPCRs in lipid rafts is by visualizing
them in intact cells, living or fixed, and tissues. There are now commercially avail-
able kits that have been developed for labeling the lipid rafts using the CTxB that is
tagged with fluorophores (Figure 4). CTxB binds to the pentasaccharide chain of
ganglioside GM1, which selectively partitions into lipid rafts. For visualizing lipid
rafts, cells are labeled with CTxB tagged with Alexa FluorÒ 488, Alexa FluorÒ

FIGURE 4 Colocalization of the D1 dopamine receptor (D1R) in Lipid Rafts of Human Renal
Proximal Tubule Cells.
Human renal proximal tubule cells were grown on a poly-L-Lysine-coated cover slip to 50%
confluence and serum-starved for 1 h to determine the basal distribution of D1R prior to
fixation with 4% paraformaldehyde and permeabilization with 0.5% Triton X-100. The lipid
rafts were labeled using cholera toxin subunit B (CTxB) tagged with Alexa FluorÒ 555
(Molecular Probes), while the endogenous D1R was immunostained using a proprietary
rabbit-anti-D1R antibody and a donkey anti-rabbit secondary antibody tagged with Alexa
FluorÒ 488 (Molecular Probes). DAPI was used to visualize the nucleus. At the basal state,
most of the D1R were found intracellularly, just below the inner leaflet of the plasma
membrane, although some colocalized with the lipid rafts (yellow areas pointed at by arrows).
The raw images were captured via laser scanning confocal microscope using separate
channels and the composite image was obtained using Zen 2011 software. 630X
magnification, scale bar ¼ 10 mm. (See color plate)
Van Anthony M. Villar, MD, PhD.
12 CHAPTER 1 GPCRs in lipid rafts

555, or Alexa FluorÒ 647 before cross-linking with an anti-CTxB to maintain the
in situ protein distribution. To demonstrate the lipid raft distribution of GPCRs, coloc-
alization experiments may be performed via laser scanning confocal microscopy by
labeling the lipid rafts using CTxB and immunostaining the GPCR of interest using
specific antibodies on the same cell. CTxB labeling may also be used to demonstrate
lipid raft endocytosis upon agonist stimulation in live cells (Qi, Mullen, Baker, &
Holl, 2010) and cultured explants (Hansen et al., 2005). The c-subunit of cytolethal
distending toxin (cdt) may also be utilized for lipid raft colocalization experiments
(the protocol is detailed in Boesze-Battaglia, 2006). Other pore-forming toxins, be-
sides CTxB, used to visualize lipid rafts include equinatoxin II which binds dispersed
sphingomyelin, lysenin which binds clustered sphingomyelin, perfringolysin O which
binds to cholesterol, and ostreolysin which binds to the combination of sphingomyelin
and cholesterol (Makino et al., 2015; Skocaj et al., 2013).
An alternative to using CTxB, cdt, and other pore-forming toxins is to use
antibodies that specifically target the lipid raft protein markers, such as
caveolin-1, caveolin-3, and flotillin-1. Conversely, transferrin receptors, CD71,
and geranylated proteins are non-lipid raft markers (Boesze-Battaglia, 2006;
Magee, Adler, & Parmryd, 2005). The ganglioside GM1 may be labeled with single
quantum dots to measure the lateral mobility and extent of movement of the lipid
rafts (Chang & Rosenthal, 2012). Recently, GPI-anchored proteins that segregate
into lipid rafts have been visualized using a novel method called enzyme-mediated
activation of radical sources (Miyagawa-Yamaguchi, Kotani, & Honke, 2015).
Probes that target the lipid content of lipid rafts have also been used to visualize
these membrane microdomains. Laurdan (6-dodecanoyl-2-(dimethylamino)-
naphthalene) and C-laurdan (6-dodecanoyl-2-[N-methyl-N-(carboxymethyl)
amino]-naphthalene), which are membrane probes that are sensitive to membrane
polarity, allow the observation of lipid rafts via two-photon microscopy (Gaus,
Zech, & Harder, 2006; Kim et al., 2007, 2008). A fluorophore-tagged domain
D4 of perfringolysin O, a cholesterol-binding cytolysin produced by Clostridium
perfringens, has been used as probe to study membrane cholesterol (Ohno-
Iwashita et al., 2004).
Aside from confocal microscopy, other biophysical approaches may also be
employed to study labeled GPCRs and/or lipid rafts. Single fluorophore tracking
microscopy (Schütz, Kada, Pastushenko, & Schindler, 2000) and fluorescence
recovery after photobleaching (Kenworthy, 2007) may be used to monitor lateral
diffusion of lipid raft-anchored GPCRs, while fluorescence lifetime imaging
microscopyefluorescence resonance energy transfer (FLIM-FRET) (Kenworthy,
Petranova & Edidin, 2000; Thaa, Herrmann, & Veit, 2010) may be used to deter-
mine the proximity of GPCRs with other proteins of interest, or of lipid raft sizes
depending on membrane composition (de Almeida, Loura, Fedorov, & Prieto,
2005). Atomic force microscopy may be used to visualize the effects of detergent
solubilization of membranes during lipid raft studies (Garner, Smith, & Hooper,
2008). Lipid rafts can now be visualized using superresolution imaging below
the 200 nm limit of conventional microscopes, e.g., including structured
1. Localization of GPCRs in lipid rafts 13

illumination microscopy, stimulated emission depletion (STED) microscopy, near-


field scanning optical microscopy, photoactivated localization microscopy
(PALM), and stochastic optical reconstruction microscopy (dSTORM) (Owen &
Gaus, 2013; Tobin et al., 2014; Wu et al., 2013).
Materials
VybrantÒ Lipid Raft Labeling Kits (Catalog #V-34403, V-34404, or V-34405)
prepare fresh working solutions according to manufacturer’s instructions
Primary antibody against the GPCR of interest
Secondary antibody against the host of the primary antibody
10% bovine serum albumin (BSA) solution
4% Paraformaldehyde in PBS
Mounting medium (EMS catalog #17985) without 40 ,6-diamidino-2-
phenylindole (DAPI)
DAPI, a nuclear stain, 10 mM stock solution
Triton X-100, 20% stock solution in deionized water
1X PBS for washing

1.2.1 Cells in suspension


Colocalization of GPCRs with lipid rafts can now be accomplished with the concom-
itant use of CTxB and an antibody against the GPCR of interest on cells. The cells can
be labeled in suspension and then mounted on glass slides for imaging, or the cells can
be grown and labeled on cover slips or in TranswellsÒ cell culture inserts when cell
polarity is important to distinguish between apical versus basolateral membranes.
1. Fluorescent labeling of cells.
1.1 Spin cells at 2000 g for 5 min and decant the medium.
1.2 Resuspend the cells in cold medium, spin, and decant the medium.
1.3 Resuspend the cells in 2 mL of CTxBeAlexa FluorÒ working solution at
4  C for 10 min. The primary antibody against the GPCR of interest may
be added to this working solution at 1:100 dilution. The primary antibody
against the GPCR should be raised in mouse, goat, rat, or chicken but not in
rabbit when using the VybrantÒ Lipid Raft Labeling Kits. Alternatively,
the primary antibody against the GPCR (especially if only a rabbit anti-
body is available) may be prelabeled with a Fluor other than the one used
for CTxB. Directly labeling the primary antibody precludes the use of a
secondary antibody (in step 1.5).
1.4 Gently wash cells 3 with cold PBS. Spin cells and decant wash buffer.
1.5 Resuspend in 2 mL of the rabbit CTxB antibody working solution at 4  C
for 30 min. The rabbit CTxB antibody cross-links it to the lipid raft do-
mains. The secondary antibody against the primary antibody may be added
to this working solution at 1:100 dilution. The secondary antibody should
be tagged with a Fluor other than the one used to label the CTxB.
As counterstain, 300 nM DAPI may also be added to this working solution.
1.6 Gently wash cells 3 with cold PBS. Spin cells and decant wash buffer.
14 CHAPTER 1 GPCRs in lipid rafts

2. Mounting and imaging.


2.1 (Optional) Fix cells with 4% paraformaldehyde at room temperature for
15 min. Paraformaldehyde is a cross-linker fixative that preserves the
architecture of the cell but may reduce the antigenicity of some cell
components and thus, requires an additional permeabilization step if
additional intracellular proteins are needed to be visualized. Fixation may
also be achieved using organic solvents, such as alcohols and acetone, but
these remove lipids and precipitate the proteins and often disrupt the cell
structure.
2.2 Mount live cells in cold PBS or fixed cells in mounting medium on glass
slide and cover with cover slip.
2.3 Image the cells using a laser scanning confocal microscope. The appropriate
filters should be used depending on the Alexa FluorÒ dye that was used and
whether DAPI was used as a nuclear stain or not (Table 2).

1.2.2 Adherent cells


1. Cell culture on cover slips.
1.1 Grow cells on 12-mm cover slips placed in a 24-well tissue culture plate to
w50% confluence using complete cell culture medium at 37  C in 95% air
and 5% CO2. Cover slips coated with lysine, laminin, or collagen may
improve cell attachment for cells that easily detach, such as HEK-293 cells.
To determine the effect of agonist/antagonist treatment on GPCR
trafficking, cells should be serum-starved for at least 1 h prior to treatment
to achieve “basal” conditions prior to treatment. Additional controls, such
as vehicle treatment, should be performed.
1.2 Draw off the medium and wash cells with cold PBS. Place the cell culture
plate on ice to stop further receptor endocytosis and endosomal trafficking.
2. Fluorescent labeling, fixation, and permeabilization.
2.1 Add 0.3 mL of CTxBeAlexa FluorÒ working solution at 4  C for 10 min.
2.2 Draw off the solution and wash cells with cold PBS.
2.3 Fix cells with 0.3 mL of 4% paraformaldehyde at room temperature for
15 min.
2.4 Wash cells with PBS. Subsequent steps can be performed at room
temperature.

Table 2 Fluorescence Spectra of CTxB Conjugates


Maximum Absorption and Emission
CTxB Fluor Conjugate (Catalog #) (nm)
Alexa FluorÒ 488 (V-34403) 495/519
Alexa FluorÒ 555 (V-34404) 555/565
Alexa FluorÒ 594 (V-34405) 590/617
The maximum absorption and emission for DAPI are 358/461 nm.
2. GPCR signaling in lipid rafts 15

2.5 Permeabilize the cells with 0.3 mL of 0.5% Triton X-100 in deionized water
for 10 min. Permeabilization provides access to intracellular antigens.
Triton X-100 can effectively solvate cellular membranes without disturb-
ing proteineprotein interactions. Other detergents such as saponin,
Tween-20, or sodium dodecyl sulfate may also be used.
2.6 Wash cells with PBS.
3. Immunostaining.
3.1 Add 0.3 mL of the primary antibody against the GPCR of interest dissolved
in 10% BSA (1:100e200 dilution) for 30e60 min.
3.2 Wash cells 3X with PBS.
3.3 Add 0.3 mL of the secondary antibody (against the host of the primary
antibody used in step 3.1) in 10% BSA. The secondary antibody should be
tagged with a Fluor other than the one used to label the CTxB. As coun-
terstain, 300 nM DAPI may also be added to this working solution.
3.4 Wash 2X with PBS and once with deionized water. The use of deionized
water washes away the residual NaCl crystals from PBS.
3.5 Mount cover slips using a mounting medium on glass slide. Gently remove
excess mounting medium by aspiration. Allow the mounting medium to
harden completely.
3.6 Image the cells using a laser scanning confocal microscope. The appropriate
filters should be used depending on the Alexa FluorÒ dye that was used and
whether DAPI was used as a nuclear stain.

2. GPCR SIGNALING IN LIPID RAFTS


There are many established protocols available that allow the study of GPCR activity
per se using commercially available kits or, less commonly, proprietary materials.
Studying the activity of GPCRs in the context of their residency in lipid rafts often
requires additional steps that would disrupt the integrity of the lipid raft microdo-
main or dissociate the protein of interest from the rafts. Most of the current strategies
to disrupt lipid raft involves either perturbation of the raft stability or modifying the
cholesterol content of the lipid rafts. Most of these treatments are performed on cells
prior to agonist/antagonist treatment and functional assays, such as cAMP produc-
tion, sodium transport, and NADPH oxidase activity (Gildea et al., 2009; Han et al.,
2008; Yu et al., 2004, 2014).

2.1 PERTURBATION OF RAFT STABILITY


Lipid rafts are dynamic assemblies of phospholipids and glycosphingolipids that
contain mostly saturated hydrocarbon chains which allow cholesterol to intercalate
between the fatty acyl chains. The surrounding membrane has greater fluidity
because of the preponderance of phospholipids with unsaturated acyl groups. The
addition of exogenous gangliosides (Webb, Hermida-Matsumoto, & Resh, 2000)
16 CHAPTER 1 GPCRs in lipid rafts

and polyunsaturated fatty acids (Simons et al., 1999), such as docosahexaenoic acid
(Ravicci et al., 2013), in the growth medium results in a change in the lipid raft
composition and the dissociation of proteins from the lipid raft. Inhibition of the
biosynthesis of glycosphingolipids and sphingomyelins using the fungal metabolite
fumonisin B1 (Lipardi, Nitsch, & Zurzolo, 2000; Nakai & Kamiguchi, 2002) may
also perturb the integrity of lipid rafts. Supplementation with 7-ketocholesterol,
which differs from cholesterol by the additional ketone group that protrudes perpen-
dicularly to the cyclopentano-perhydro-phenanthrene ring, decreases lipid raft order,
and increases membrane polarity (Rentero et al., 2008; Schieffer, Naware, Bakun, &
Bamezai, 2014). Interestingly, the nonsteroidal, anti-inflammatory drug aspirin has a
high affinity for phospholipid membranes and partitions into the lipid head groups.
This interaction impairs the molecular organization brought about by cholesterol
and thus, leads to increased mobility in a lipid raft model (Alsop et al., 2015; Kyrikou,
Hadjikakou, Kovala-Demertzi, Viras, & Mavromoustakos, 2004). The use of short-
chain ceramides, i.e., C2-ceramide and C6-ceramide, decreases the plasma mem-
brane lipid order and disrupts the lipid rafts as indicated by a reduction in the extent
of FRET between lipid raft markers (Gidwani, Brown, Holowka, & Baird, 2003).

2.2 CHANGING THE CHOLESTEROL CONTENT


Cholesterol is an integral component of lipid rafts in mammalian cell membranes,
and membrane cholesterol levels are crucial in determining the stability and organi-
zation of lipid rafts (Silvius, 2003). Thus, modifying the content of cholesterol in the
plasma membrane is another option to disrupt the lipid raft and evaluate the function
of GPCRs. The antifungal polyene antibiotics filipin (Brown & London, 2000;
Drake & Braciale, 2001), nystatin (Oakley, Smith, & Engelhardt, 2009), and ampho-
tericin (Wysoczynski et al., 2005) disrupt lipid rafts by binding and sequestering
cholesterol within the plasma membrane. Pore-forming agents such as saponin
(Hering, Lin, & Sheng, 2003; Schroeder, Ahmed, Zhu, London, & Brown, 1998),
digitonin (Oliferenko et al., 1999), and streptolysin O (Fernandez-Lizarbe, Pascual,
Gascon, Blanco, & Guerri, 2008) may also be used. b-MCD is one of the most
frequently used agents to deplete the endogenous cholesterol content of lipid rafts
(Han et al., 2008; Yu et al., 2014, 2013). One advantage in using b-MCD is the avail-
ability of a control for its use, i.e., a-MCD (Vial & Evans, 2005). Inhibitors of the
rate-limiting enzyme for cholesterol synthesis, the HMG-CoA reductase, may be
used also to inhibit endogenous cholesterol biosynthesis. These include drugs
such as simvastatin (Drake & Braciale, 2001) and lovastatin (Meszaros, Klappe,
Hummel, Hoekstra, & Kok, 2011). A summary of protocols using these approaches
is found in Table 3.

2.3 FLUORESCENCE IMAGING


The advent of FRET and BRET (bioluminescence resonance energy transfer)
biosensors has allowed the study of GPCR activity in lipid rafts in living cells.
2. GPCR signaling in lipid rafts 17

Table 3 Common Strategies to Disrupt the Lipid Raft


Strategies Protocol
A. Disruption of raft stability
Addition of gangliosides 10e100 mM for 1 h (Simons et al., 1999)
Addition of PUFAs 50 mM, overnight (Webb et al., 2000)
Fumonisin B1 25 ug/mL, 48e72 h (Lipardi et al., 2000)
7-cholesterol 35e70 mM for 5 min to 2 h (Schieffer et al.,
2014)
Aspirin 10% For 30e60 min in noncellular
experiments (Alsop et al., 2015)
C2- and C6-ceramide 32 and 8 mM, respectively (Gidwani et al.,
2003)
B. Changing the cholesterol content
Filipin 2.5e5 mg/mL for 15 min (stock solution:
5 mg/mL in ethanol) (Drake & Braciale,
2001)
Nystatin 20e50 mg/mL for 1 h (Oakley et al., 2009)
Amphotericin 10 mg/mL for 1 h (Wysoczynski et al., 2005)
Saponin 5% in 20 mM phosphate buffer, pH 7.4, at
4  C for 10 min followed by extraction in
0.5% Triton X-100 at 4  C (Hering et al.,
2003)
Digitonin 0.003% for 30 min on ice (Oliferenko et al.,
1999)
Streptolysin O 500 ng/mL for 2 h (Fernandez-Lizarbe
et al., 2008)
b-MCD 2% for 1 h (Han et al., 2008; Yu et al., 2014,
2013)
10 mM for 1 h (Simons, 1999)
Lovastatin 1 mg/mL for 20 h (Meszaros, 2011)
Simvastatin Use 5 mg/mL for 12 h (Drake & Braciale,
2001)
PUFAs, Polyunsaturated Fatty Acids.

N-Way FRET microscopy can quantify interacting and noninteracting FRET pairs in
live cells (Hoppe, Scott, Welliver, Straight, & Swanson, 2013). The freely diffusible
FRET sensor Epac2-camps has been used to measure global cAMP responses of
lipid raft-associated receptors since it responds to changes in cAMP occurring
throughout the cytosolic compartment of cells (Agarwal et al., 2014). Moreover, ver-
sions of the Epac2-camps probe allow the selective targeting to lipid raft (Epac2-
MyrPalm) and nonraft (Epac2-CAAX) domains, which are useful in monitoring
local cAMP production near the plasma membrane (Agarwal et al., 2014). PALM,
as indicated above, and dSTORM have also been used to track the reorganization
18 CHAPTER 1 GPCRs in lipid rafts

of lipid rafts (Tobin et al., 2014; Wu et al., 2013). Movement of single molecules in
living cells could also be tracked (single molecule tracking) (Scarselli et al., 2015).
In addition, fluorescent nanosensors that measure sodium in real time are reversible
and completely selective over other cations (Dubach, Das, Rosenzweig, & Clark,
2009). Real-time monitoring of sodium transport in response to stimulation or inhi-
bition of GPCRs in intact or disrupted lipid rafts has become feasible.
Current biochemical and biophysical techniques for studying GPCRs in lipid
rafts, while helpful in many instances, are still rife with methodological drawbacks
and limitations. These include the requirement for cell membrane disruption, the
reliance on antibodies that are specific for the GPCR of interest, the inability to study
native proteins, and the use of exogenous, often tagged, proteins. Newer methodol-
ogies that allow the study of GPCRs in their native form in intact cells are needed,
such as the FRET biosensors for cAMP monitoring. Meanwhile, the use of comple-
mentary approaches that yield mutually supportive results may be the most judicious
way for drawing conclusions regarding the distribution and activity of GPCRs in
lipid rafts.

REFERENCES
Agarwal, S. R., Yang, P. C., Rice, M., Singer, C. A., Nikolaev, V. O., Lohse, M. J., et al.
(2014). Role of membrane microdomains in compartmentation of cAMP signaling.
PLoS One, 9(4), e95835.
de Almeida, R. F., Loura, L. M., Fedorov, A., & Prieto, M. (2005). Lipid rafts have different
sizes depending on membrane composition: a time-resolved fluorescence resonance
energy transfer study. Journal of Molecular Biology, 346(4), 1109e1120.
Alsop, R. J., Toppozini, L., Marquardt, D., Kucerka, N., Harroun, T. A., & Rheinstädter, M. C.
(2015). Aspirin inhibits formation of cholesterol rafts in fluid lipid membranes.
Biochimica et Biophysica Acta, 1848(3), 805e812.
Barnett-Norris, J., Lynch, D., & Reggio, P. H. (2005). Lipids, lipid rafts and caveolae: their
importance for GPCR signaling and their centrality to the endocannabinoid system.
Life Sciences, 77(14), 625e639.
Boesze-Battaglia, K. (2006). Isolation of membrane rafts and signaling complexes. Methods
in Molecular Biology, 332, 169e179.
Breton, S., Lisanti, M. P., Tyszkowski, R., McLaughlin, M., & Brown, D. (1998). Basolat-
eral distribution of caveolin-1 in the kidney. Absence from Hþ-ATPase-coated
endocytic vesicles in intercalated cells. Journal of Histochemistry and Cytochemistry,
46(2), 205e214.
Brown, D. A., & London, E. (2000). Structure and function of sphingolipid- and cholesterol-
rich membrane rafts. Journal of Biological Chemistry, 275(23), 17221e17224.
Chang, J. C., & Rosenthal, S. J. (2012). Visualization of lipid raft membrane compartmental-
ization in living RN46A neuronal cells using single quantum dot tracking. ACS Chemical
Neuroscience, 3(10), 737e743.
Drake, D. R., 3rd, & Braciale, T. J. (2001). Cutting edge: lipid raft integrity affects the
efficiency of MHC class I tetramer binding and cell surface TCR arrangement on
CD8þ T cells. Journal of Immunology, 166(12), 7009e7013.
References 19

Dubach, J. M., Das, S., Rosenzweig, A., & Clark, H. A. (2009). Visualizing sodium dynamics
in isolated cardiomyocytes using fluorescent nanosensors. Proceedings of the National
Academy of Sciences of the United States of America, 106(38), 16145e16150.
Ferguson, S. G. (2001). Evolving concepts in G protein-coupled receptor endocytosis: the role
in receptor desensitization and signaling. Pharmacological Reviews, 53, 1e24.
Fernandez-Lizarbe, S., Pascual, M., Gascon, M. S., Blanco, A., & Guerri, C. (2008). Lipid
rafts regulate ethanol-induced activation of TLR4 signaling in murine macrophages.
Molecular Immunology, 45(7), 2007e2016.
Foster, L. J., De Hoog, C. L., & Mann, M. (2003). Unbiased quantitative proteomics of lipid
rafts reveals high specificity for signaling factors. Proceedings of the National Academy of
Sciences of the United States of America, 100(10), 5813e5818.
Garner, A. E., Smith, D. A., & Hooper, N. M. (2008). Visualization of detergent solubilization of
membranes: implications for the isolation of rafts. Biophysical Journal, 94(4), 1326e1340.
Gaus, K., Zech, T., & Harder, T. (2006). Visualizing membrane microdomains by Laurdan
2-photon microscopy. Molecular Membrane Biology, 23(1), 41e48.
Gidwani, A., Brown, H. A., Holowka, D., & Baird, B. (2003). Disruption of lipid order by
short-chain ceramides correlates with inhibition of phospholipase D and downstream
signaling by FcepsilonRI. Journal of Cell Science, 116(Pt 15), 3177e3187.
Gildea, J. J., Israel, J. A., Johnson, A. K., Zhang, J., Jose, P. A., & Felder, R. A. (2009).
Caveolin-1 and dopamine-mediated internalization of NaKATPase in human renal
proximal tubule cells. Hypertension, 54(5), 1070e1076.
Han, W., Li, H., Villar, V. A., Pascua, A. M., Dajani, M. I., Wang, X., et al. (2008). Lipid rafts
keep NADPH oxidase in the inactive state in human renal proximal tubule cells.
Hypertension, 51(2), 481e487.
Hansen, G. H., Dalskov, S. M., Rasmussen, C. R., Immerdal, L., Niels-Christiansen, L. L., &
Danielsen, E. M. (2005). Cholera toxin entry into pig enterocytes occurs via a lipid raft-
and clathrin-dependent mechanism. Biochemistry, 44(3), 873e882.
Head, B. P., Patel, H. H., & Insel, P. A. (2014). Interaction of membrane/lipid rafts with the cyto-
skeleton: impact on signaling and function: membrane/lipid rafts, mediators of cytoskeletal
arrangement and cell signaling. Biochimica et Biophysica Acta, 1838(2), 532e545.
Hemler, M. E. (2005). Tetraspanin functions and associated microdomains. Nature Reviews
Molecular Cell Biology, 6(10), 801e811.
Hering, H., Lin, C. C., & Sheng, M. (2003). Lipid rafts in the maintenance of synapses,
dendritic spines, and surface AMPA receptor stability. Journal of Neuroscience,
23(8), 3262e3271.
Hoppe, A. D., Scott, B. L., Welliver, T. P., Straight, S. W., & Swanson, J. A. (2013). N-way
FRET microscopy of multiple protein-protein interactions in live cells. PLoS One, 8(6),
e64760.
Huang, P., Xu, W., Yoon, S. I., Chen, C., Chong, P. L., & Liu-Chen, L. Y. (2007). Cholesterol
reduction by methyl-beta-cyclodextrin attenuates the delta opioid receptor-mediated
signaling in neuronal cells but enhances it in non-neuronal cells. Biochemical Pharma-
cology, 73(4), 534e549.
Insel, P. A., Head, B. P., Ostrom, R. S., Patel, H. H., Swaney, J. S., Tang, C. M., et al. (2005).
Caveolae and lipid rafts: G protein-coupled receptor signaling microdomains in cardiac
myocytes. Annals of the New York Academy of Sciences, 1047, 166e172.
Karnovsky, M. J., Kleinfeld, A. M., Hoover, R. L., Dawidowicz, E. A., McIntyre, D. E.,
Salzman, E. A., et al. (1982). Lipid domains in membranes. Annals of the New York Acad-
emy of Sciences, 401, 61e75.
20 CHAPTER 1 GPCRs in lipid rafts

Kenworthy, A. K. (2007). Fluorescence recovery after photobleaching studies of lipid rafts.


Methods in Molecular Biology, 398, 179e192.
Kenworthy, A. K., Petranova, N., & Edidin, M. (2000). High-resolution FRET microscopy of
cholera toxin B-subunit and GPI-anchored proteins in cell plasma membranes. Molecular
Biology of the Cell, 11(5), 1645e1655.
Kim, H. M., Choo, H. J., Jung, S. Y., Ko, Y. G., Park, W. H., Jeon, S. J., et al. (2007).
A two-photon fluorescent probe for lipid raft imaging: C-laurdan. Chembiochem, 8(5),
553e559.
Kim, H. M., Jeong, B. H., Hyon, J. Y., An, M. J., Seo, M. S., Hong, J. H., et al. (2008). Two-
photon fluorescent turn-on probe for lipid rafts in live cell and tissue. Journal of the
American Chemical Society, 130(13), 4246e4247.
Kyrikou, I., Hadjikakou, S. K., Kovala-Demertzi, D., Viras, K., & Mavromoustakos, T.
(2004). Effects of non-steroid anti-inflammatory drugs in membrane bilayers. Chemistry
and Physics of Lipids, 132(2), 157e169.
Lefkowitz, R. J. (1998). G protein-coupled receptors. III. New roles for receptor kinases and
beta-arrestins in receptor signaling and desensitization. Journal of Biological Chemistry,
273(30), 18677e18680.
Lingwood, D., & Simons, K. (2010). Lipid rafts as a membrane-organizing principle. Science,
327(5961), 46e50.
Lipardi, C., Nitsch, L., & Zurzolo, C. (2000). Detergent-insoluble GPI-anchored proteins are
apically sorted in fischer rat thyroid cells, but interference with cholesterol or sphingoli-
pids differentially affects detergent insolubility and apical sorting. Molecular Biology of
the Cell, 11(2), 531e542.
Macdonald, J. L., & Pike, L. J. (2005). A simplified method for the preparation of detergent-
free lipid rafts. Journal of Lipid Research, 46(5), 1061e1067.
Magee, A. I., Adler, J., & Parmryd, I. (2005). Cold-induced coalescence of T-cell plasma mem-
brane microdomains activates signalling pathways. Journal of Cell Science, 118(Pt 14),
3141e3151.
Makino, A., Abe, M., Murate, M., Inaba, T., Yilmaz, N., Hullin-Matsuda, F., et al. (2015).
Visualization of the heterogeneous membrane distribution of sphingomyelin associated
with cytokinesis, cell polarity, and sphingolipidosis. FASEB Journal, 29(2), 477e493.
van Meer, G., & Simons, K. (1988). Lipid polarity and sorting in epithelial cells. Journal of
Cellular Biochemistry, 36(1), 51e58.
Meszaros, P., Klappe, K., Hummel, I., Hoekstra, D., & Kok, J. W. (2011). Function of MRP1/
ABCC1 is not dependent on cholesterol or cholesterol-stabilized lipid rafts. Biochemical
Journal, 437(3), 483e491.
Miyagawa-Yamaguchi, A., Kotani, N., & Honke, K. (2015). Each GPI-anchored protein
species forms a specific lipid raft depending on its GPI attachment signal. Glycoconjugate
Journal, 32(7), 531e540.
Murtazina, R., Kovbasnjuk, O., Donowitz, M., & Li, X. (2006). Naþ/Hþ exchanger NHE3
activity and trafficking are lipid Raft-dependent. Journal of Biological Chemistry,
281(26), 17845e17855.
Nakai, Y., & Kamiguchi, H. (2002). Migration of nerve growth cones requires detergent-
resistant membranes in a spatially defined and substrate-dependent manner. Journal of
Cell Biology, 159(6), 1097e1108.
Oakley, F. D., Smith, R. L., & Engelhardt, J. F. (2009). Lipid rafts and caveolin-1 coordinate
interleukin-1beta (IL-1beta)-dependent activation of NFkappaB by controlling endocy-
tosis of Nox2 and IL-1beta receptor 1 from the plasma membrane. Journal of Biological
Chemistry, 284(48), 33255e33264.
References 21

Ohno-Iwashita, Y., Shimada, Y., Waheed, A. A., Hayashi, M., Inomata, M., Nakamur, M.,
et al. (2004). Perfringolysin O, a cholesterol-binding cytolysin, as a probe for lipid
rafts. Anaerobe, 10(2), 125e134.
Oliferenko, S., Paiha, K., Harder, T., Gerke, V., Schwärzler, C., Schwarz, H., et al. (1999).
Analysis of CD44-containing lipid rafts: recruitment of annexin II and stabilization by
the actin cytoskeleton. Journal of Cell Biology, 146(4), 843e854.
Owen, D. M., & Gaus, K. (2013). Imaging lipid domains in cell membranes: the advent of
super-resolution fluorescence microscopy. Frontiers in Plant Science, 4, 503.
Palade, G. E. (1953). The fine structure of blood capillaries. Journal of Applied Physics,
24, 1424.
Parton, R. G., & del Pozo, M. A. (2013). Caveolae as plasma membrane sensors, protectors
and organizers. Nature Reviews Molecular Cell Biology, 14(2), 98e112.
Pike, L. J. (2004). Lipid rafts: heterogeneity on the high seas. Biochemical Journal, 378(Pt 2),
281e292.
Qi, R., Mullen, D. G., Baker, J. R., & Holl, M. M. (2010). The mechanism of polyplex inter-
nalization into cells: testing the GM1/caveolin-1 lipid raft mediated endocytosis pathway.
Molecular Pharmaceutics, 7(1), 267e279.
Quest, A. F., Leyton, L., & Párraga, M. (2004). Caveolins, caveolae, and lipid rafts in cellular
transport, signaling, and disease. Biochemistry and Cell Biology, 82(1), 129e144.
Rajendran, L., Le Lay, S., & Illges, H. (2007). Raft association and lipid droplet targeting of
flotillins are independent of caveolin. Biological Chemistry, 388(3), 307e314.
Ravacci, G. R., Brentani, M. M., Tortelli, T., Jr., Torrinhas, R. S., Saldanha, T., Torres, E. A.,
et al. (2013). Lipid raft disruption by docosahexaenoic acid induces apoptosis in
transformed human mammary luminal epithelial cells harboring HER-2 overexpression.
Journal of Nutritional Biochemistry, 24(3), 505e515.
Rentero, C., Zech, T., Quinn, C. M., Engelhardt, K., Williamson, D., Grewal, T., et al. (2008).
Functional implications of plasma membrane condensation for T cell activation. PLoS
One, 3(5), e2262.
Riquier, A. D., Lee, D. H., & McDonough, A. A. (2009). Renal NHE3 and NaPi2 partition into
distinct membrane domains. American Journal of Physiology Cell Physiology, 296(4),
C900eC910.
Salzer, U., & Prohaska, R. (2001). Stomatin, flotillin-1, and flotillin-2 are major integral
proteins of erythrocyte lipid rafts. Blood, 97(4), 1141e1143.
Scarselli, M., Annibale, P., McCormick, P. J., Kolachalam, S., Aringhieri, S., Radenovic, A.,
et al. (2015). Revealing GPCR oligomerization at the single-molecule level through a
nanoscopic lens: methods, dynamics and biological function. FEBS Journal. http://
dx.doi.org/10.1111/febs.13577 (Epub ahead of print) Review.
Schieffer, D., Naware, S., Bakun, W., & Bamezai, A. K. (2014). Lipid raft-based membrane
order is important for antigen-specific clonal expansion of CD4(þ) T lymphocytes. BMC
Immunology, 15, 58.
Schnitzer, J. E., McIntosh, D. P., Dvorak, A. M., Liu, J., & Oh, P. (1995). Separation of caveolae
from associated microdomains of GPI-anchored proteins. Science, 269(5229), 1435e1439.
Schroeder, R. J., Ahmed, S. N., Zhu, Y., London, E., & Brown, D. A. (1998). Cholesterol
and sphingolipid enhance the Triton X-100 insolubility of glycosylphosphatidylinositol-
anchored proteins by promoting the formation of detergent-insoluble ordered membrane
domains. Journal of Biological Chemistry, 273(2), 1150e1157.
Schütz, G. J., Kada, G., Pastushenko, V. P., & Schindler, H. (2000). Properties of lipid micro-
domains in a muscle cell membrane visualized by single molecule microscopy. EMBO
Journal, 19(5), 892e901.
22 CHAPTER 1 GPCRs in lipid rafts

Silvius, J. R. (2003). Role of cholesterol in lipid raft formation: lessons from lipid model
systems. Biochimica et Biophysica Acta, 1610(2), 174e183.
Simons, M., Friedrichson, T., Schulz, J. B., Pitto, M., Masserini, M., & Kurzchalia, T. V. (1999).
Exogenous administration of gangliosides displaces GPI-anchored proteins from lipid
microdomains in living cells. Molecular Biology of the Cell, 10(10), 3187e3196.
Simons, K., & Ikonen, E. (1997). Functional rafts in cell membranes. Nature, 387(6633),
569e572.
Simons, K., & Toomre, D. (2000). Lipid rafts and signal transduction. Nature Reviews
Molecular Cell Biology, 1(1), 31e39.
Simons, K., & van Meer, G. (1998). Lipid sorting in epithelial cells. Biochemistry, 27(17),
6197e6202.
Skocaj, M., Bakrac, B., Krizaj, I., Macek, P., Anderluh, G., & Sepcic, K. (2013). The sensing
of membrane microdomains based on pore-forming toxins. Current Medicinal Chemistry,
20(4), 491e501.
Smart, E. J., Ying, Y. S., Mineo, C., & Anderson, R. G. (1995). A detergent-free method for
purifying caveolae membrane from tissue culture cells. Proceedings of the National Acad-
emy of Sciences of the United States of America, 92(22), 10104e10108.
Song, K. S., Li, S., Okamoto, T., Quilliam, L. A., Sargiacomo, M., & Lisanti, M. P. (1996). Co-
purification and direct interaction of Ras with caveolin, an integral membrane protein of
caveolae microdomains. Detergent-free purification of caveolae microdomains. Journal of
Biological Chemistry, 271(16), 9690e9697.
Sonnino, S., & Prinetti, A. (2013). Membrane domains and the “lipid raft” concept. Current
Medicinal Chemistry, 20(1), 4e21.
Thaa, B., Herrmann, A., & Veit, M. (2010). Intrinsic cytoskeleton-dependent clustering of
influenza virus M2 protein with hemagglutinin assessed by FLIM-FRET. Journal of
Virology, 84(23), 12445e12449.
Tobin, S. J., Cacao, E. E., Hong, D. W., Terenius, L., Vukojevic, V., & Jovanovic-Talisman, T.
(2014). Nanoscale effects of ethanol and naltrexone on protein organization in the plasma
membrane studied by photoactivated localization microscopy (PALM). PLoS One, 9,
e87225.
Vial, C., & Evans, R. J. (2005). Disruption of lipid rafts inhibits P2X1 receptor-mediated
currents and arterial vasoconstriction. Journal of Biological Chemistry, 280(35),
30705e30711.
Webb, Y., Hermida-Matsumoto, L., & Resh, M. D. (2000). Inhibition of protein palmitoyla-
tion, raft localization, and T cell signaling by 2-bromopalmitate and polyunsaturated fatty
acids. Journal of Biological Chemistry, 275(1), 261e270.
Worby, C. A., & Dixon, J. E. (2002). Sorting out the cellular functions of sorting nexins.
Nature Reviews Molecular Cell Biology, 3(12), 919e931.
Wu, J., Gao, J., Qi, M., Wang, J., Cai, M., Liu, S., et al. (2013). High-efficiency localization of
Na(þ)-K(þ) ATPases on the cytoplasmic side by direct stochastic optical reconstruction
microscopy. Nanoscale, 5(23), 11582e11586.
Wysoczynski, M., Reca, R., Ratajczak, J., Kucia, M., Shirvaikar, N., Honczarenko, M., et al.
(2005). Incorporation of CXCR4 into membrane lipid rafts primes homing-related
responses of hematopoietic stem/progenitor cells to an SDF-1 gradient. Blood, 105(1),
40e48.
Yamada, E. (1955). The fine structure of the gall bladder epithelium of the mouse. Journal of
Biophysical and Biochemical Cytology, 1(5), 445e458.
References 23

Yu, P., Sun, M., Villar, V. A., Zhang, Y., Weinman, E. J., Felder, R. A., et al. (2014).
Differential dopamine receptor subtype regulation of adenylyl cyclases in lipid rafts in hu-
man embryonic kidney and renal proximal tubule cells. Cellular Signalling, 26(11),
2521e2529.
Yu, P., Villar, V. A., & Jose, P. A. (2013). Methods for the study of dopamine receptors within
lipid rafts of kidney cells. Methods in Molecular Biology, 964, 15e24.
Yu, P., Yang, Z., Jones, J. E., Wang, Z., Owens, S. A., Mueller, S. C., et al. (2004). D1 dopa-
mine receptor signaling involves caveolin-2 in HEK-293 cells. Kidney International,
66(6), 2167e2180.
von Zastrow, M. (2003). Mechanisms regulating trafficking of G protein-coupled receptors in
the endocytic pathway. Life Sciences, 74, 217e224.
CHAPTER

Imaging GPCRs
trafficking and signaling
with total internal
reflection fluorescence
2
microscopy in cultured
neurons
Francheska Delgado-Peraza*, x, Carlos Nogueras-Ortiz*,
Agnes M. Acevedo Canabal*,x,
Cristina Roman-Vendrell*, {, Guillermo A. Yudowski*, x, 1
*Institute of Neurobiology, University of Puerto Rico Medical Sciences Campus,
San Juan, PR, USA
x
Department of Anatomy and Neurobiology, School of Medicine, University of Puerto Rico,
San Juan, PR, USA
{
Department of Physiology, School of Medicine, University of Puerto Rico, San Juan, PR, USA
1
Corresponding author: E-mail: Guillermo.yudowski@upr.edu

CHAPTER OUTLINE
1. Image Acquisition ................................................................................................ 27
1.1 Important Considerations before Imaging ................................................ 27
1.2 Materials .............................................................................................. 28
1.2.1 Cell culture......................................................................................... 28
1.2.2 TIRF microscopy equipment and settings............................................ 28
1.3 Imaging................................................................................................ 28
1.3.1 Cell culture......................................................................................... 28
1.3.2 Live cell imaging................................................................................. 29
1.4 Notes ................................................................................................... 30
2. Image Analysis .................................................................................................... 30
3. Final Comments.................................................................................................... 31
Acknowledgments ..................................................................................................... 31
References ............................................................................................................... 31

Methods in Cell Biology, Volume 132, ISSN 0091-679X, http://dx.doi.org/10.1016/bs.mcb.2015.10.002 25


© 2016 Elsevier Inc. All rights reserved.
26 CHAPTER 2 Imaging GPCRs trafficking and signaling

Abstract
Total internal reflection fluorescence (TIRF) microscopy allows probing the cellular
events occurring close and at the plasma membrane. Over the last decade, we have seen a
significant increase in the number of publications applying TIRF microscopy to unravel
some of the fundamental biological questions regarding G protein-coupled receptors
(GPCRs) function such as the mechanisms controlling receptor trafficking, quaternary
structure, and signaling among others. Most of the published work has been performed in
heterologous systems such as HEK293 and CHO cells, where the imaging surface
available is higher and smoother when compared with the narrow processes or the smaller
cell bodies of neurons. However, some publications have expanded our understanding of
these events to primary cell cultures, mostly rat hippocampal and striatal neuronal cul-
tures. Results from these cells provide a bona fide model of the complex events con-
trolling GPCR function in living cells. We believe more work needs to be performed in
primary cultures and eventually in intact tissue to complement the knowledge obtained
from heterologous cell models. Here, we described a step-by-step protocol to investigate
the surface trafficking and signaling from GPCRs in rat hippocampal and striatal primary
cultures.

The unique ability of total internal reflection fluorescence (TIRF) microscopy to


generate an evanescent field and excite fluorophores within a narrow optical section
(100 nm) provides an ideal tool to investigate the multiple and dynamic events
occurring close to and at the plasma membrane of living cells with reduced
phototoxicity and bleaching (Axelrod, 1981, 2003, 2008; Simon, 2009). This
characteristic resulted in the application of TIRF microscopy to multiple biological
areas ranging from single molecule analysis to cell migration. TIRF microscopy has
been particularly used to investigate protein translocation and trafficking to the
membrane, vesicular events, and single molecule analysis among others
(Mattheyses, Simon, & Rappoport, 2010; Reck-Peterson, Derr, & Stuurman,
2010; Roman-Vendrell & Yudowski, 2015; Steyer & Almers, 2001). Since astro-
cytes display a significant surface area, these cells have been used to visualize
and analyze various events such as the molecular mechanisms controlling vesicular
release (Li, Agulhon, Schmidt, Oheim, & Ropert, 2013). However, the application of
TIRF microscopy to investigate G protein-coupled receptors (GPCRs) in neurons is
still very limited. Our laboratory was among the first to use TIRF microscopy to
investigate GPCRs in neurons (Yudowski, Puthenveedu, & von Zastrow, 2006).
Our data showed that receptor recycling to the cell surface can be mediated by
two different vesicular events, a rapid event without diffusion barriers and a slow
event with a kinetic similar to kiss and run exocytosis (Roman-Vendrell et al.,
2014; Yudowski et al., 2006). These kiss and run events were not observed with
the Mu opioid receptor and were dependent on the carboxy terminal sequence of
the B2 adrenergic receptor (Roman-Vendrell & Yudowski, 2015; Yu, Dhavan,
Chevalier, Yudowski, & Zastrow, 2010). TIRF microscopy was also utilized to
demonstrate that GPCR recycling after ligand-induced internalization is actively
controlled by the activation of receptors still at the cell surface. Interestingly,
1. Image acquisition 27

identical results were observed in primary cell cultures and in heterologous systems
supporting the idea that heterologous systems can provide valuable information
(Bowman & Puthenveedu, 2015; Bowman et al., 2015; Roman-Vendrell, Yu, &
Yudowski, 2012; Yudowski, Puthenveedu, Henry, & Von Zastrow, 2009). Others
have utilized TIRF in neuronal cultures to investigate the pathways by which recep-
tors are recycled to the cell surface (Li et al., 2012). Our laboratory also utilized
TIRF microscopy to investigate endogenous GPCR signaling in hippocampal
neurons by loading neurons with calcium sensitive dyes and pharmacologically acti-
vating specific adrenergic receptors (Tzingounis, von Zastrow, & Yudowski, 2010).
More recently, we have investigated how cannabinoid receptor 1 interacts with beta-
arrestins at the cell surface of hippocampal neurons to regulate beta-arrestin
signaling (Flores-Otero et al., 2014). These are some of the applications of TIRF mi-
croscopy to investigate GPCRs. The application of TIRF and superresolution micro-
scopy in combination with novel fluorescent tags and nanobodies should only
expand the toolbox available to further probe the biology of these highly relevant
receptors.

1. IMAGE ACQUISITION
1.1 IMPORTANT CONSIDERATIONS BEFORE IMAGING
To visualize GPCRs at the cell surface, receptors must be tagged with fluorophores
that are ideally resistant to quenching/photobleaching and with a significant quantum
yield (Shaner, Steinbach, & Tsien, 2005). One of the fluorescent tags widely utilized
in TIRF microscopy to investigate GPCRs is the pH-sensitive eGFP-variant super
ecliptic pHluorin (SEP) (Miesenbock, De Angelis, & Rothman, 1998). By attaching
the SEP molecule to the extracellular domain of GPCRs, receptors are highly visible
when they are located at the cell surface (neutral pH) and their fluorescence is
rapidly quenched in intracellular compartments such as endosomes (pH acidic).
The use of GPCRs tagged with SEP at the extracellular domain in TIRF microscopy
is an ideal approach to investigate events at the cell surface minimizing fluorescence
from receptors in intracellular compartments while reducing phototoxicity. Other
available probes such as antibodies conjugated with quantum dots or SNAP tag
fusion proteins have been also utilized to investigate GPCRs surface trafficking
(Calebiro et al., 2013; Maurel et al., 2008; Mikasova, Groc, Choquet, & Manzoni,
2008; Reck-Peterson et al., 2010). However, their application to TIRF microscopy
is not as frequent as conventional genetically coded fluorophores.
It is important to note that regardless of the florescent tag utilized, we strongly
recommend tagging receptors at their extracellular domain and compare their
function with wild-type receptors before any further analysis. Only tagged receptors
with identical pharmacological and functional properties to wild-type receptors must
be used. In our experience, tags in intracellular domains of GPCRs generally disrupt
their signaling and or trafficking, rendering nonphysiological behavior. Finally, TIRF
28 CHAPTER 2 Imaging GPCRs trafficking and signaling

microscopy is not exempt from the general rules and pitfalls from live cell imaging,
image acquisition, and analysis (Frigault, Lacoste, Swift, & Brown, 2009; Jaiswal &
Simon, 2004; North, 2006; Schnell, Dijk, Sjollema, & Giepmans, 2012).

1.2 MATERIALS
1.2.1 Cell culture
1. Striatal primary cultures obtained from embryonic day 17e18 Sprague-Dawley
rat embryos. Alternatively, brain tissue can be purchased from BrainBits LLC
(Springfield, IL).
2. Neuron culture media: Neurobasal medium supplemented with B27 (according
to manufacturer protocol) and 0.5 mM glutaMAXÔ (Life Technologies).
3. Imaging media such as Neurobasal nimus phenol red without serum and
supplemented with 20 mM HEPES (Life Technologies) (see Notes).
4. 30 mm coverslips #1.5 thickness. Coverslips must be acid washed and coated
with fresh poly-D-Lysine (Sigma). Glass bottom dishes (MatTek) can be also
utilized. They must be coated with PDL and the glass thickness must be 1.5.
5. Transfection reagents, Lipofectamine 3000 (Life Technologies).
6. Hippocampal cultures were >90% pure as calculated by MAP2 and GFAP
staining as previously described (Yudowski et al., 2006, Yudowski, Olsen,
Adesnik, Marek, & Bredt, 2013).

1.2.2 TIRF microscopy equipment and settings


1. Motorized Nikon (Melville, NY) Ti-E inverted microscope with a 100 apo-
chromat oil immersion TIRF objective lens (CFI Apo TIRF 100; Nikon), color
correction and a motorized stage with perfect focus (see Notes).
2. Light source: 488 and 561 nm Coherent sapphire lasers (Coherent Inc. Santa
Clara, CA) 50 and 100 mW lasers, respectively.
3. Temperature control is utilized to keep cells at 37  C with a stable Z stage and
objective warmer (Bioptechs, Butler, PA)
4. Interchangeable Coverslip Dish (Bioptechs) (http://www.bioptechs.com/
Products/ICD/coverslipdish.html)
5. Camera: iXonEM þ DU897 back-illuminated electron multiplying charged
coupled device sensor camera (Andor, Belfast, UK).
6. Readout speed: 10 Hz, exposure time: continuous 50e100 ms exposure for
receptor recycling, electron multiplying gain of 300, no binning, bit depth ¼ 14
bits, camera temperature set to minimum and laser power to 10% for 488 nm at
50 mW.

1.3 IMAGING
1.3.1 Cell culture
1. Acid clean coverslips or glass bottom dishes by incubation in 1 M HCl shaking
overnight. Rinse two times with abundant ddH2O and once with 70% ethanol.
1. Image acquisition 29

Leave in 95% ethanol until ready to use. Before coating, UV and dry in the
culture hood no less than 1 h.
2. Coat acid clean coverslips or glass bottom dishes with freshly prepared
100 mg/mL poly-D-lysine (PDL) for 2e4 h at 37  C. Wash 3 times with sterile
water and air dry in sterile environment. It is important to prepare PDL fresh for
every use. Old PDL solutions result in less than ideal cell attachment.
3. Plate 250,000e300,000 neurons per 35 mm dish. Neurons are transfected at 4e5
days in vitro (DIV) and imaged at 15 DIVor later. Expression levels for receptor
trafficking is ideal between 14 and 25 DIV.
4. Transfect cells with DNA constructs using Lipofectamine 3000 or Effectene
according to the manufacturer’s instructions. We perform imaging after DIV 15.
(High expression levels will impair observation of individual events. They can
also result in trafficking artifacts such as reduced internalization). Lentivirus
can be also used to infect targeted cells. In our experience, infected cells do not
look as healthy as transfected cells.
5. Replace conditioned media with 2 mL of freshly prepared Opti-MEM with
HEPES 15e30 min before imaging sessions. Important: Do not to allow
neurons to dry during this process. A small amount of media must be left at the
dish covering all cells during the process.
6. Incubate cells at 37  C >10 min to allow acclimatization to the new media.
7. Transfer neurons to the microscope.

1.3.2 Live cell imaging


1. At least 30 min before any acquisition, turn on the microscope and the tem-
perature controllers. Turn on the laser key and let the laser warm up. Important:
Objective and imaging chamber temperature must be controlled before and
during experiments. Temperature must be 37  C at the glass.
2. Select TIRF objective, add a drop of immersion oil (type LDF, RI: w1.515) and
carefully place cells on the stage (see Notes).
3. To reduce the effects of photobleaching, it is important to find and focus the cells
using transmission light first. Then, find cells expressing tagged receptors using
epifluorescence and then switch to TIRF illumination (see Notes).
4. Add agonist diluted in warm imaging media by automated perfusion system or
manually outside the imaging area to minimize artifacts (see Notes).
5. Acquisition settings for endocytosis: 100e300 msec exposures every 2e3 s.
Total time: 10e30 min.
6. Acquisition settings for recycling: Continuous illumination and acquisition at
50e100 ms exposures for 1e2 min (see Notes).
7. Depolarization with 25 mM KCl is performed to test viability at the end of ex-
periments with Fluo-4.
8. Imaging sessions will generate large amounts of data. Careful data management
must be implemented in advance. Standardized electronic notebooks or
spreadsheets are recommended.
30 CHAPTER 2 Imaging GPCRs trafficking and signaling

1.4 NOTES
1. Healthy primary cultures are essential to obtain reliable and valid results.
Neurons must conserve their integrity, without blebbing or detachments.
2. HEPES is used to maintain the pH constant for up to 45e60 min outside a CO2
incubator.
3. High quality cDNA is highly desired for neuronal transfection. Multiple
transfection agents are commercially available. We utilize lipofectamine 2000
on DIV 4e5 and perform imaging on DIV > 15. This delay results in optimal
expression levels for TIRF imaging.
4. Focal plane must be kept constant during imaging sessions.
5. It is very important that the cells grow in monolayer and are not more than
80e90% confluent on the day of imaging.
6. It is very important that the bottom of the imaging dish is completely dry and
clean. Any liquid or dirt will interfere during TIRF imaging.
7. The most critical step is to find the exact angle for TIRF. To align the laser
properly, focus on the plasma membrane. You can find the cell sharp edges and
use them as reference.
8. If ligands are added manually, extreme care is needed to prevent disturbing the
cells within the imaging area. Controls should be performed to test the effects of
dimethyl sulfoxide and other solvents on surface fluorescence and basal cell
activity.
9. Endocytosis should be visible within 1 or 2 min of agonist addition. Agonist-
induced recycling can be observed 2e3 min after initial exposure. A constant
rate of vesicular fusion is generally observed at w10 min.

2. IMAGE ANALYSIS
Exocytotic events are easily identified by direct visualization of the abrupt increase
(w1 s) in intensity at discrete points in the cell surface. Image sequences can be
analyzed using the acquisition software available from the microscope or by using
the public domain NIH Image program ImageJ/FIJI software, which is freely avail-
able at http://fiji.sc/Fiji. Orthogonal views (kymographs) can be used to distinguish
these events from other vesicular event due to their characteristic kinetics and to
quantify their frequency, decay kinetics, and location. Maximum intensity fluores-
cence can be extracted and plotted to demonstrate lateral diffusion and number of
exocytic molecules among others. More recently, software specifically designed
to identify exocytic events has been developed such as the exocytosis detection
recipe from SVCell (https://www.svcell.com/recipes/exocytosis-detection). Further
development of this and other open source codes should provide better tools to
extract and analyze exocytic events. We recommend performing all the analysis
blindly to reduce bias in the quantification.
Endocytic events are intrinsically more challenging to analyze due to their lower
signal to noise ration. Manual detection was initially utilized following specific
References 31

predetermined clathrin-coated pits behaviors including: (1) events must appear and
disappear within the time series (<250 s); (2) events must display limited movement
(no more than 4 by 4 pixels through their lifetime); and (3) events must not fuse or
collide with each other (Saffarian, Cocucci, & Kirchhausen, 2009). More recently,
particle-tracking algorithms have been developed to follow these events such as
the imageJ 2D-spot tracker plug-in (Sage, Hediger, Gasser, & Unser, 2005) or the
Matlab code developed by the groups of Danuser and Schmid (Loerke et al.,
2009, 2011). These codes were developed to analyze movies with ideal signal to
noise ratio such as those obtained by imaging fluorescently tagged clathrin and
dynamin. Analysis of GPCR endocytosis utilizing these tools has been more chal-
lenging with limited success. Nevertheless, these are fundamental steps toward a
more rigorous and unbiased tool to investigate endocytosis. Finally, to achieve sta-
tistical data, we define individual experiments by the number of cells analyzed from
the same transfection (n ¼ 1 experiment, x number of cells). To achieve statistical
significance, we analyze no less than 10 different cells from more than four different
experiments (different transfection). A minimum of 20 endocytic events are ex-
pected from every cell to secure results from healthy cells.

3. FINAL COMMENTS
The availability of turnkey TIRF microscopes and the development of multiple flu-
orophores have allowed the use of TIRF microscopy to peer into the complex lives of
GPCRs at the cell surface. We have learned that their surface trafficking, signaling,
and general function are more complex and exquisite than previously thought. Novel
layers of regulatory mechanisms and trafficking pathways have been discovered
along with complex associations between receptors and other proteins. This is
only the beginning of a new era of GPCR research in which novel imaging tools
such as TIRF and superresolution microscopy are key to unravel some of the funda-
mental questions in GPCRs biology.

ACKNOWLEDGMENTS
This work was supported by research grants from NIH DA023444 and DA037924 to GY, FDP,
and CNO. MBRS program RISE R25 GM061838 to CRV and AAC. We thank Dr Nevin
Lambert (Medical College of Georgia, Georgia Health Sciences University, Georgia Regents
University) for his feedback.

REFERENCES
Axelrod, D. (1981). Cell-substrate contacts illuminated by total internal reflection
fluorescence. Journal of Cell Biology, 89(1), 141e145. PMID: 7014571.
Axelrod, D. (January 2003). Total internal reflection fluorescence microscopy in cell biology.
Methods in Enzymology, 361(2), 1e33. PMID: 12624904.
32 CHAPTER 2 Imaging GPCRs trafficking and signaling

Axelrod, D. (2008). Total internal reflection fluorescence microscopy. Methods in Cell


Biology, 30, 245e270.
Bowman, S. L., & Puthenveedu, M. A. (January 2015). Postendocytic sorting of adrenergic
and opioid receptors: new mechanisms and functions. Progress in Molecular Biology
and Translational Science, 132, 189e206. PMID: 26055059.
Bowman, S. L., Soohoo, A. L., Shiwarski, D. J., Schulz, S., Pradhan, A. A., &
Puthenveedu, M. A. (March 24, 2015). Cell-autonomous regulation of Mu-opioid receptor
recycling by substance P. Cell Reports, 10(11), 1925e1936. PMID: 25801029.
Calebiro, D., Rieken, F., Wagner, J., Sungkaworn, T., Zabel, U., Borzi, A., et al. (2013).
Single-molecule analysis of fluorescently labeled G-protein-coupled receptors reveals
complexes with distinct dynamics and organization. Proceedings of the National
Academic Sciences of United States of America, 110(2), 743e748. PMID: 23267088.
Flores-Otero, J., Ahn, K. H., Delgado-Peraza, F., Mackie, K., Kendall, D. A., &
Yudowski, G. A. (August 1, 2014). Ligand-specific endocytic dwell times control
functional selectivity of the cannabinoid receptor 1. Nature Communications: Nature
Publishing Group, 5. PMID: 25081814.
Frigault, M. M., Lacoste, J., Swift, J. L., & Brown, C. M. (March 15, 2009). Live-cell
microscopy - tips and tools. Journal of Cell Science, 122(Pt 6), 753e767. PMID:
19261845.
Jaiswal, J. K., & Simon, S. M. (October 2004). Potentials and pitfalls of fluorescent quantum
dots for biological imaging. Trends in Cell Biology, 14(9), 497e504. PMID: 15350978.
Li, D., Agulhon, C., Schmidt, E., Oheim, M., & Ropert, N. (January 2013). New tools for
investigating astrocyte-to-neuron communication. Frontiers in Cellular Neuroscience,
7, 193. PMID: 24194698.
Li, Y., Roy, B. D., Wang, W., Zhang, L., Sampson, S. B., Yang, Y., et al. (May 23, 2012).
Identification of two functionally distinct endosomal recycling pathways for dopamine
D2 receptor. Journal of Neuroscience, 32(21), 7178e7190.
Loerke, D., Mettlen, M., Schmid, S., & Danuser, G. (2011). Measuring the hierarchy of
molecular events during clathrin-mediated endocytosis. Traffic, 12(7), 815e825.
Loerke, D., Mettlen, M., Yarar, D., Jaqaman, K., Jaqaman, H., Danuser, G., et al. (March 17,
2009). Cargo and dynamin regulate clathrin-coated pit maturation. PLoS Biology, 7(3),
e57. PMID: 19296720.
Mattheyses, A. L., Simon, S. M., & Rappoport, J. Z. (2010). Imaging with total internal
reflection fluorescence microscopy for the cell biologist. Journal of Cell Science,
123(Pt 21), 3621e3628. PMID: 20971701.
Maurel, D., Comps-Agrar, L., Brock, C., Rives, M.-L., Bourrier, E., Ayoub, M. A., et al. (June
2008). Cell-surface protein-protein interaction analysis with time-resolved FRET and
snap-tag technologies: application to GPCR oligomerization. Nature Methods, 5(6),
561e567. PMID: 18488035.
Miesenbock, G., De Angelis, D. A., & Rothman, J. E. (1998). Visualizing secretion and
synaptic transmission with pH-sensitive green fluorescent proteins. Nature, 192e195.
Mikasova, L., Groc, L., Choquet, D., & Manzoni, O. J. (November 2008). Altered surface
trafficking of presynaptic cannabinoid type 1 receptor in and out synaptic terminals
parallels receptor desensitization. Proceedings of the National Academy of Sciences of
the United States of America, 105(47), 18596e18601. PMID: 19015531.
North, A. J. (January 2, 2006). Seeing is believing? A beginners’ guide to practical pitfalls in
image acquisition. Journal of Cell Biology, 172(1), 9e18. PMID: 16390995.
References 33

Reck-Peterson, S. L., Derr, N. D., & Stuurman, N. (March 2010). Imaging single molecules
using total internal reflection fluorescence microscopy (TIRFM). Cold Spring Harbor
Protocols, 2010(3). pdb.top73. PMID: 20194477.
Roman-Vendrell, C., Chevalier, M., Acevedo-Canabal, A. M., Delgado-Peraza, F., Flores-
Otero, J., & Yudowski, G. A. (November 3, 2014). Imaging of kiss-and-run exocytosis
of surface receptors in neuronal cultures. Frontiers in Cellular Neuroscience,
8(November), 1e10. PMID: 25404895.
Roman-Vendrell, C., & Yudowski, G. A. (January 2015). Real-time imaging of mu opioid
receptors by total internal reflection fluorescence microscopy. Methods in Molecular
Biology, 1230, 79e86. PMID: 25293317.
Roman-Vendrell, C., Yu, Y. J., & Yudowski, G. A. (February 29, 2012). Fast modulation of
m-Opioid receptor (MOR) recycling is mediated by receptor agonists. Journal of Biolog-
ical Chemistry, 287(18), 14782e14791. PMID: 22378794.
Saffarian, S., Cocucci, E., & Kirchhausen, T. (September 2009). Distinct dynamics of endocytic
clathrin-coated pits and coated plaques. PLoS Biol, 7(9), e1000191. PMID: 19809571.
Sage, D., Hediger, F., Gasser, S. M., & Unser, M. (2005). Automatic tracking of individual
fluorescence particles: application to the study of chromosome dynamics. IEEE Transac-
tions on Image Processing, 14(9), 582e586. PMID: 16190472.
Schnell, U., Dijk, F., Sjollema, K. A., & Giepmans, B. N. G. (February 2012). Immunolabel-
ing artifacts and the need for live-cell imaging. Nature Methods, 9(2), 152e158. PMID:
22290187.
Shaner, N. C., Steinbach, P. A., & Tsien, R. Y. (December 2005). A guide to choosing
fluorescent proteins. Nature Methods, 2(12), 905e909. PMID: 16299475.
Simon, S. (2009). Partial internal reflections on total internal reflection fluorescent
microscopy. Trends in Cell Biology, 19(11), 661e668.
Steyer, J. A., & Almers, W. (April 2001). A real-time view of life within 100 nm of the plasma
membrane. Nature Reviews Molecular Cell Biology, 2(4), 268e275. PMID: 11283724.
Tzingounis, A. V., von Zastrow, M., & Yudowski, G. A. (November 2010). Blocker drugs
mediate calcium signaling in native central nervous system neurons by arrestin-biased
agonism. Proceedings of the National Academy of Sciences, 107(49), 21028e21033.
PMID: 21078978.
Yu, Y. J., Dhavan, R., Chevalier, M. W., Yudowski, G. A., & Zastrow, M. (September 2010).
Rapid delivery of internalized signaling receptors to the somatodendritic surface by
sequence-specific local insertion. Journal of Neuroscience, 30(35), 11703e11714.
PMID: 20810891.
Yudowski, G. A., Olsen, O., Adesnik, H., Marek, K. W., & Bredt, D. S. (2013). Acute inac-
tivation of PSD-95 destabilizes AMPA receptors at hippocampal synapses. Methods, 8(1),
1e9. PMID: 23342049.
Yudowski, G. A., Puthenveedu, M. A., Henry, A. G., & Von Zastrow, M. (2009). Cargo-
mediated regulation of a rapid Rab4-dependent recycling pathway. Molecular Biology
of the Cell, 20, 2774e2784. PMID: 19369423.
Yudowski, G. A., Puthenveedu, M. A., & von Zastrow, M. (May 2006). Distinct modes of
regulated receptor insertion to the somatodendritic plasma membrane. Nature Neurosci-
ence, 9(5), 622e627. PMID: 16604070.
CHAPTER

Trafficking of ciliary
G protein-coupled
receptors 3
Jeremy C. McIntyre*, x, 1, Mellisa M. Hege{, Nicolas F. Berbari{, 1
*Department of Neuroscience, University of Florida, Gainesville, FL, USA
x
Center for Smell and Taste, University of Florida, Gainesville, FL, USA
{
Department of Biology, Indiana University-Purdue University Indianapolis,
Indianapolis, IN, USA
1
Corresponding authors: E-mail: jmcin@ufl.edu; nberbari@iupui.edu

CHAPTER OUTLINE
Introduction .............................................................................................................. 36
1. Rhodopsin as a Ciliary GPCR................................................................................. 38
2. Ciliary GPCRs in Other Neurons ............................................................................. 39
3. Olfactory Sensory Neuron Ciliary GPCRs ................................................................ 41
4. Dynamic Trafficking of Ciliary GPCRs..................................................................... 45
Conclusions and Future Directions ............................................................................. 47
References ............................................................................................................... 48

Abstract
In the last decade highly conserved cellular appendages called cilia have enjoyed a
renewed interest from basic, biomedical scientists, and clinicians alike. This interest has
grown upon the elucidation that cilia throughout the body serve as important sensory and
signaling centers in both development and adult homeostasis. Furthermore, the identifi-
cation of several rare genetic disorders associated with cilia dysfunction has broadened
the field. However, even though their potential role in human health and disease is now
recognized many basic questions about their functions remain. This chapter seeks to
explore the trafficking of cilia-specific G protein-coupled receptors (GPCRs) and
discusses several model systems in which this has been explored. We open the chapter by
briefly discussing cilia and GPCRs then begin discussing some aspects of rhodopsin
trafficking, arguably the most well studied of cilia GPCRs. We continue with sections on
neuronal cilia and olfactory cilia receptor trafficking. Finally, we conclude with the
emerging area of dynamic ciliary GPCR trafficking and speculate about future directions
and some of the questions that remain for ciliary GPCRs.

Methods in Cell Biology, Volume 132, ISSN 0091-679X, http://dx.doi.org/10.1016/bs.mcb.2015.11.009 35


© 2016 Elsevier Inc. All rights reserved.
36 CHAPTER 3 Trafficking of ciliary G protein-coupled receptors

INTRODUCTION
The cilium is a highly conserved, near ubiquitous, small microtubule-based cell
appendage. Historically classified as either motile or immotile (primary), it is clear
that all cilia have the capacity to coordinate specific signaling pathways. Most
mammalian cells possess a singular immotile cilium or “primary” cilium which
is thought to play a crucial role as a complex sensory and signaling center (for a
review on cilia signaling associated pathways, see Berbari, O’Connor, Haycraft,
& Yoder, 2009) (Figure 1). An emerging class of relatively rare genetic diseases,
termed ciliopathies, are now associated with cilia dysfunction. Interestingly, the
clinical features of ciliopathies are broad and diverse. Phenotypes range from

FIGURE 1
Primary cilium schematic. Several of the structural elements are labeled and indicated with
arrows including the basal body, axoneme. The green and red arrows along the axoneme
indicate anterograde and retrograde intraflagellar transport, respectively. Other proteins and
complexes involved in general ciliary trafficking are indicated as well as generic molecules
found in the ciliary membrane such at ciliary-specific GPCRs. (See color plate)
Another random document with
no related content on Scribd:
Gutenberg” is associated) is accessed, displayed, performed,
viewed, copied or distributed:

This eBook is for the use of anyone anywhere in the United


States and most other parts of the world at no cost and with
almost no restrictions whatsoever. You may copy it, give it
away or re-use it under the terms of the Project Gutenberg
License included with this eBook or online at
www.gutenberg.org. If you are not located in the United
States, you will have to check the laws of the country where
you are located before using this eBook.

1.E.2. If an individual Project Gutenberg™ electronic work is


derived from texts not protected by U.S. copyright law (does not
contain a notice indicating that it is posted with permission of the
copyright holder), the work can be copied and distributed to
anyone in the United States without paying any fees or charges.
If you are redistributing or providing access to a work with the
phrase “Project Gutenberg” associated with or appearing on the
work, you must comply either with the requirements of
paragraphs 1.E.1 through 1.E.7 or obtain permission for the use
of the work and the Project Gutenberg™ trademark as set forth
in paragraphs 1.E.8 or 1.E.9.

1.E.3. If an individual Project Gutenberg™ electronic work is


posted with the permission of the copyright holder, your use and
distribution must comply with both paragraphs 1.E.1 through
1.E.7 and any additional terms imposed by the copyright holder.
Additional terms will be linked to the Project Gutenberg™
License for all works posted with the permission of the copyright
holder found at the beginning of this work.

1.E.4. Do not unlink or detach or remove the full Project


Gutenberg™ License terms from this work, or any files
containing a part of this work or any other work associated with
Project Gutenberg™.
1.E.5. Do not copy, display, perform, distribute or redistribute
this electronic work, or any part of this electronic work, without
prominently displaying the sentence set forth in paragraph 1.E.1
with active links or immediate access to the full terms of the
Project Gutenberg™ License.

1.E.6. You may convert to and distribute this work in any binary,
compressed, marked up, nonproprietary or proprietary form,
including any word processing or hypertext form. However, if
you provide access to or distribute copies of a Project
Gutenberg™ work in a format other than “Plain Vanilla ASCII” or
other format used in the official version posted on the official
Project Gutenberg™ website (www.gutenberg.org), you must, at
no additional cost, fee or expense to the user, provide a copy, a
means of exporting a copy, or a means of obtaining a copy upon
request, of the work in its original “Plain Vanilla ASCII” or other
form. Any alternate format must include the full Project
Gutenberg™ License as specified in paragraph 1.E.1.

1.E.7. Do not charge a fee for access to, viewing, displaying,


performing, copying or distributing any Project Gutenberg™
works unless you comply with paragraph 1.E.8 or 1.E.9.

1.E.8. You may charge a reasonable fee for copies of or


providing access to or distributing Project Gutenberg™
electronic works provided that:

• You pay a royalty fee of 20% of the gross profits you derive from
the use of Project Gutenberg™ works calculated using the
method you already use to calculate your applicable taxes. The
fee is owed to the owner of the Project Gutenberg™ trademark,
but he has agreed to donate royalties under this paragraph to
the Project Gutenberg Literary Archive Foundation. Royalty
payments must be paid within 60 days following each date on
which you prepare (or are legally required to prepare) your
periodic tax returns. Royalty payments should be clearly marked
as such and sent to the Project Gutenberg Literary Archive
Foundation at the address specified in Section 4, “Information
about donations to the Project Gutenberg Literary Archive
Foundation.”

• You provide a full refund of any money paid by a user who


notifies you in writing (or by e-mail) within 30 days of receipt that
s/he does not agree to the terms of the full Project Gutenberg™
License. You must require such a user to return or destroy all
copies of the works possessed in a physical medium and
discontinue all use of and all access to other copies of Project
Gutenberg™ works.

• You provide, in accordance with paragraph 1.F.3, a full refund of


any money paid for a work or a replacement copy, if a defect in
the electronic work is discovered and reported to you within 90
days of receipt of the work.

• You comply with all other terms of this agreement for free
distribution of Project Gutenberg™ works.

1.E.9. If you wish to charge a fee or distribute a Project


Gutenberg™ electronic work or group of works on different
terms than are set forth in this agreement, you must obtain
permission in writing from the Project Gutenberg Literary
Archive Foundation, the manager of the Project Gutenberg™
trademark. Contact the Foundation as set forth in Section 3
below.

1.F.

1.F.1. Project Gutenberg volunteers and employees expend


considerable effort to identify, do copyright research on,
transcribe and proofread works not protected by U.S. copyright
law in creating the Project Gutenberg™ collection. Despite
these efforts, Project Gutenberg™ electronic works, and the
medium on which they may be stored, may contain “Defects,”
such as, but not limited to, incomplete, inaccurate or corrupt
data, transcription errors, a copyright or other intellectual
property infringement, a defective or damaged disk or other
medium, a computer virus, or computer codes that damage or
cannot be read by your equipment.

1.F.2. LIMITED WARRANTY, DISCLAIMER OF DAMAGES -


Except for the “Right of Replacement or Refund” described in
paragraph 1.F.3, the Project Gutenberg Literary Archive
Foundation, the owner of the Project Gutenberg™ trademark,
and any other party distributing a Project Gutenberg™ electronic
work under this agreement, disclaim all liability to you for
damages, costs and expenses, including legal fees. YOU
AGREE THAT YOU HAVE NO REMEDIES FOR NEGLIGENCE,
STRICT LIABILITY, BREACH OF WARRANTY OR BREACH
OF CONTRACT EXCEPT THOSE PROVIDED IN PARAGRAPH
1.F.3. YOU AGREE THAT THE FOUNDATION, THE
TRADEMARK OWNER, AND ANY DISTRIBUTOR UNDER
THIS AGREEMENT WILL NOT BE LIABLE TO YOU FOR
ACTUAL, DIRECT, INDIRECT, CONSEQUENTIAL, PUNITIVE
OR INCIDENTAL DAMAGES EVEN IF YOU GIVE NOTICE OF
THE POSSIBILITY OF SUCH DAMAGE.

1.F.3. LIMITED RIGHT OF REPLACEMENT OR REFUND - If


you discover a defect in this electronic work within 90 days of
receiving it, you can receive a refund of the money (if any) you
paid for it by sending a written explanation to the person you
received the work from. If you received the work on a physical
medium, you must return the medium with your written
explanation. The person or entity that provided you with the
defective work may elect to provide a replacement copy in lieu
of a refund. If you received the work electronically, the person or
entity providing it to you may choose to give you a second
opportunity to receive the work electronically in lieu of a refund.
If the second copy is also defective, you may demand a refund
in writing without further opportunities to fix the problem.

1.F.4. Except for the limited right of replacement or refund set


forth in paragraph 1.F.3, this work is provided to you ‘AS-IS’,
WITH NO OTHER WARRANTIES OF ANY KIND, EXPRESS
OR IMPLIED, INCLUDING BUT NOT LIMITED TO
WARRANTIES OF MERCHANTABILITY OR FITNESS FOR
ANY PURPOSE.

1.F.5. Some states do not allow disclaimers of certain implied


warranties or the exclusion or limitation of certain types of
damages. If any disclaimer or limitation set forth in this
agreement violates the law of the state applicable to this
agreement, the agreement shall be interpreted to make the
maximum disclaimer or limitation permitted by the applicable
state law. The invalidity or unenforceability of any provision of
this agreement shall not void the remaining provisions.

1.F.6. INDEMNITY - You agree to indemnify and hold the


Foundation, the trademark owner, any agent or employee of the
Foundation, anyone providing copies of Project Gutenberg™
electronic works in accordance with this agreement, and any
volunteers associated with the production, promotion and
distribution of Project Gutenberg™ electronic works, harmless
from all liability, costs and expenses, including legal fees, that
arise directly or indirectly from any of the following which you do
or cause to occur: (a) distribution of this or any Project
Gutenberg™ work, (b) alteration, modification, or additions or
deletions to any Project Gutenberg™ work, and (c) any Defect
you cause.

Section 2. Information about the Mission of


Project Gutenberg™
Project Gutenberg™ is synonymous with the free distribution of
electronic works in formats readable by the widest variety of
computers including obsolete, old, middle-aged and new
computers. It exists because of the efforts of hundreds of
volunteers and donations from people in all walks of life.

Volunteers and financial support to provide volunteers with the


assistance they need are critical to reaching Project
Gutenberg™’s goals and ensuring that the Project Gutenberg™
collection will remain freely available for generations to come. In
2001, the Project Gutenberg Literary Archive Foundation was
created to provide a secure and permanent future for Project
Gutenberg™ and future generations. To learn more about the
Project Gutenberg Literary Archive Foundation and how your
efforts and donations can help, see Sections 3 and 4 and the
Foundation information page at www.gutenberg.org.

Section 3. Information about the Project


Gutenberg Literary Archive Foundation
The Project Gutenberg Literary Archive Foundation is a non-
profit 501(c)(3) educational corporation organized under the
laws of the state of Mississippi and granted tax exempt status by
the Internal Revenue Service. The Foundation’s EIN or federal
tax identification number is 64-6221541. Contributions to the
Project Gutenberg Literary Archive Foundation are tax
deductible to the full extent permitted by U.S. federal laws and
your state’s laws.

The Foundation’s business office is located at 809 North 1500


West, Salt Lake City, UT 84116, (801) 596-1887. Email contact
links and up to date contact information can be found at the
Foundation’s website and official page at
www.gutenberg.org/contact

Section 4. Information about Donations to


the Project Gutenberg Literary Archive
Foundation
Project Gutenberg™ depends upon and cannot survive without
widespread public support and donations to carry out its mission
of increasing the number of public domain and licensed works
that can be freely distributed in machine-readable form
accessible by the widest array of equipment including outdated
equipment. Many small donations ($1 to $5,000) are particularly
important to maintaining tax exempt status with the IRS.

The Foundation is committed to complying with the laws


regulating charities and charitable donations in all 50 states of
the United States. Compliance requirements are not uniform
and it takes a considerable effort, much paperwork and many
fees to meet and keep up with these requirements. We do not
solicit donations in locations where we have not received written
confirmation of compliance. To SEND DONATIONS or
determine the status of compliance for any particular state visit
www.gutenberg.org/donate.

While we cannot and do not solicit contributions from states


where we have not met the solicitation requirements, we know
of no prohibition against accepting unsolicited donations from
donors in such states who approach us with offers to donate.

International donations are gratefully accepted, but we cannot


make any statements concerning tax treatment of donations
received from outside the United States. U.S. laws alone swamp
our small staff.

Please check the Project Gutenberg web pages for current


donation methods and addresses. Donations are accepted in a
number of other ways including checks, online payments and
credit card donations. To donate, please visit:
www.gutenberg.org/donate.

Section 5. General Information About Project


Gutenberg™ electronic works
Professor Michael S. Hart was the originator of the Project
Gutenberg™ concept of a library of electronic works that could
be freely shared with anyone. For forty years, he produced and
distributed Project Gutenberg™ eBooks with only a loose
network of volunteer support.

Project Gutenberg™ eBooks are often created from several


printed editions, all of which are confirmed as not protected by
copyright in the U.S. unless a copyright notice is included. Thus,
we do not necessarily keep eBooks in compliance with any
particular paper edition.

Most people start at our website which has the main PG search
facility: www.gutenberg.org.

This website includes information about Project Gutenberg™,


including how to make donations to the Project Gutenberg
Literary Archive Foundation, how to help produce our new
eBooks, and how to subscribe to our email newsletter to hear
about new eBooks.

You might also like