Download as pdf or txt
Download as pdf or txt
You are on page 1of 92

THE

JOURNAL OF
EXPERIMENTAL
MEDICINE
SELECTED ARTICLES APRIL 2015 www.jem.org

INNATE IMMUNITY
The Pinnacle of Discovery
Recombinant Proteins for Bioassay

BioLegend has a large variety of recombinant cytokines, chemokines, growth factors, and proteinases (e.g., MMPs, Granzymes A
and B). The high quality of our recombinant proteins anchor your research, and the exceptional pricing allows you to focus on
reaching those lofty research goals.

BioLegend Recombinant Proteins are:


Diverse, with over 400 Exceptionally priced, to Functionally tested and Available in carrier-free
human, mouse, and rat meet even the tightest validated for bioassay. and animal-free formats
proteins. budget. to suit your assay needs.

Recombinant Mouse FLT3L Recombinant Human MMP-9


Mouse FLT3L induces The activity of
BioLegend IL-6 production in monomeric hMMP-9
Competitor murine myeloid was measured with
OD (450 - 570 nm)

leukemia cell line M1. 10 M of fluorogenic


MMP substrate, Mca-
RFU/sec

PLGL-Dpa-AR-NH2 , in
the presence of 2.5,
5.0, 10, 20, 40, and
80 ng of activated
hMMP-9. The activity
of activated hMMP-9 is
>800 pmole/min/g.
ng/mL hMMP-9 (ng)

To learn more, visit: biolegend.com/recombinant_proteins


BioLegend is ISO 9001:2008 and ISO 13485:2003 Certified
Toll-Free Tel: (US & Canada): 1.877.BIOLEGEND (246.5343)
Tel: 858.768.5800
biolegend.com
08-0050-01

World-Class Quality | Superior Customer Support | Outstanding Value


Welcome
I n n a t e I m mu n i t y
The Journal of Experimental Medicine now prints topic-specific mini collections to showcase a handful
of our recent publications. In this installment, we highlight papers focusing on two innate cell subsets,
dendritic cells and macrophages in health and disease.
Immunotherapy has proven successful in many types of cancer treatment, but has also been
associated with dangerous inflammatory responses in some patients. Our collection begins with
an insight by Merghoub and Wolchok discussing the findings of Mirsoian et al. who show that
lethal inflammation in response to anti-CD40 and IL-2 immunotherapy is triggered by increased
inflammatory macrophages in the accumulated visceral fat of aged mice. Similarly, young obese
mice succumbed to treatment while older mice on diets were protected.The study highlights factors
to be considered with use of immunotherapeutics.
In the gut, the immune system must balance recognition of infectious pathogens with minimal responses to commensal
bacteria. An Insight by Giorgio Trinchieri describes these immune challenges and the findings from Longman et al. showing that
in both mice following Citrobacter rodentium infection and in patients with colitis, CX3CR1+ mononuclear phagocytes (MNPs)
are potent producers of IL-23 and IL-1b. The production of these cytokines by MNPs efficiently induces IL-22 production by
group 3 innate lymphoid cells to promote barrier integrity and mucosal protection.
Lambrecht and Guilliams examine genetic fate mapping of myeloid cells that distinguishes macrophages derived from
embryonic precursors vs. circulating monocytes. They highlight the contribution to macrophage biology from Molowi et al.
reporting that cardiac macrophages are initially of embryonic origin, but are progressively replaced by monocyte-derived cells as
mice age. They speculate about how manipulation of different macrophage populations could be used in therapeutics.
Myelin destruction in multiple sclerosis (MS) is mediated by inflammatory macrophages, but the origin of these cells has been
unclear. An Insight from Michael Heneka discusses findings from Yamasaki et al., who use a mouse model of MS to distinguish
tissue-resident microglial cells from infiltrating monocytes. Using double chemokine reporter mice, the authors find that
monocyte-derived macrophages initiate myelin destruction, mainly in the nodes of Ranvier, while microglia-derived macrophages are
involved with clearing debris.
Two papers from the Liu and Nussenzweig groups track the differentiation of human progenitor cells into dendritic cells
(DCs). They show that a granulocyte/monocyte/DC progenitor gives rise to a monocyte-DC progenitor that in turn generates
both monocytes and a common DC progenitor. The common DC progenitor produces the three major subsets of human DCs,
such as CD1c+ cDCs, CD141+ cDCs and pDCs. They go on to identify the immediate precursor of CD1c+ and CD141+ DCs
in the circulation of healthy donors. These precursor cells (hpre-cDC) were detectable in cord blood, bone marrow, blood, and
peripheral lymphoid organs. An Insight by Frederic Geissmann describes the novel in vitro culture system used in the studies to
parse out the development of human DCs as well as a discussion of the developmental similarities between mouse and human DCs.
Together these studies characterize the many roles and functions of innate immune cells. We hope you enjoy this
complimentary copy of our Innate Immunity collection. We invite you to explore additional collections at www.jem.org
and to follow JEM on Facebook, Google+, and Twitter.

Selected Articles April 2015

Immunotherapy and the belly of the beast


Taha Merghoub and Jedd D. Wolchok

Adiposity induces lethal cytokine storm after systemic administration of stimulatory immunotherapy
regimens in aged mice
Annie Mirsoian, Myriam N. Bouchlaka, Gail D. Sckisel, Mingyi Chen, Chien-Chun Steven Pai, Emanuel Maverakis,
Richard G. Spencer, Kenneth W. Fishbein, Sana Siddiqui, Arta M. Monjazeb, Bronwen Martin, Stuart Maudsley,
Charles Hesdorffer, Luigi Ferrucci, Dan L. Longo, Bruce R. Blazar, Robert H. Wiltrout, Dennis D. Taub,
and William J. Murphy
Critical role for CX3CR1+ mononuclear phagocytes in intestinal homeostasis
Giorgio Trinchieri

CX3CR1+ mononuclear phagocytes support colitis-associated innate lymphoid cell production of IL-22
Randy S. Longman, Gretchen E. Diehl,1 Daniel A. Victorio, Jun R. Huh,1 Carolina Galan, Emily R. Miraldi,
Arun Swaminath, Richard Bonneau, Ellen J. Scherl, and Dan R. Littman

Monocytes find a new place to dwell in the niche of heartbreak hotel


Bart Lambrecht and Martin Guilliams

Progressive replacement of embryo-derived cardiac macrophages with age


Kaaweh Molawi, Yochai Wolf, Prashanth K. Kandalla, Jeremy Favret, Nora Hagemeyer, Kathrin Frenzel,
Alexander R. Pinto, Kay Klapproth, Sandrine Henri, Bernard Malissen, Hans-Reimer Rodewald, Nadia A. Rosenthal,
Marc Bajenoff, Marco Prinz, Steffen Jung, and Michael H. Sieweke

Macrophages derived from infiltrating monocytes mediate autoimmune myelin destruction


Michael T. Heneka

Differential roles of microglia and monocytes in the inflamed central nervous system
Ryo Yamasaki, Haiyan Lu, Oleg Butovsky, Nobuhiko Ohno, Anna M. Rietsch, Ron Cialic, Pauline M. Wu,
Camille E. Doykan, Jessica Lin, Anne C. Cotleur, Grahame Kidd, Musab M. Zorlu, Nathan Sun, Weiwei Hu,
LiPing Liu, Jar-Chi Lee, Sarah E. Taylor, Lindsey Uehlein, Debra Dixon, Jinyu Gu, Crina M. Floruta, Min Zhu,
Israel F. Charo, Howard L. Weiner, and Richard M. Ransohoff

The real thing: How to make human DC subsets


Frederic Geissmann

Restricted dendritic cell and monocyte progenitors in human cord blood and bone marrow
Jaeyop Lee, Galle Breton, Thiago Yukio Kikuchi Oliveira, Yu Jerry Zhou, Arafat Aljoufi, Sarah Puhr, Mark J. Cameron,
Rafick-Pierre Skaly, Michel C. Nussenzweig, and Kang Liu

Circulating precursors of human CD1c+ and CD141+ dendritic cells


Galle Breton, Jaeyop Lee, Yu Jerry Zhou, Joseph J. Schreiber, Tibor Keler, Sarah Puhr, Niroshana Anandasabapathy,
Sarah Schlesinger, Marina Caskey, Kang Liu, and Michel C. Nussenzweig
THE
JOURNAL OF
EXPERIMENTAL
MEDICINE
Executive Editor Advisory Editors Tomohiro Kurosaki Ethan M. Shevach
Marlowe S.Tessmer Shizuo Akira Bart N. Lambrecht Roy L. Silverstein
phone (212) 327-8575 Kari Alitalo Klaus F. Ley Jonathan Sprent
email: jem@rockefeller.edu Frederick W. Alt Yong-Jun Liu Janet Stavnezer
Senior Editors K. Frank Austen Clare Lloyd Andreas Strasser
Teodoro Pulvirenti Albert Bendelac Tak Mak Stuart Tangye
Heather L. Van Epps Michael J. Bevan Bernard Malissen Steven L.Teitelbaum
Christine A. Biron James S. Malter Thomas J.Templeton
Scientific Editor
Christian Bogdan Philippa Marrack Kevin J.Tracey
Catarina Sacristn
Hal E. Broxmeyer Diane Mathis Giorgio Trinchieri
Editors Meinrad Busslinger Ira Mellman Shannon Turley
Jean-Laurent Casanova Arturo Casadevall Matthias Merkenschlager Marcel R.M. van den Brink
Douglas T. Fearon Ajay Chawla Sean J. Morrison Ulrich von Andrian
David Holtzman Yongwon Choi Muriel Moser Harald von Boehmer
Lewis L. Lanier Robert L. Coffman Christian Mnz Christopher M.Walker
William A. Muller Daniel J. Cua Cornelis Murre Raymond M.Welsh
Carl Nathan Myron I. Cybulsky Benjamin G. Neel E. John Wherry
Michel Nussenzweig Riccardo Dalla Favera Michael Neuberger Linda S.Wicker
Anne OGarra Glenn Dranoff Victor Nussenzweig Ian Wilson
Alexander Rudensky Michael Dustin John J. OShea Thomas Wynn
Alan Sher Vincent A. Fischetti Paul H. Patterson
Sasha Tarakhovsky Richard A. Flavell Fiona Powrie
Andreas Trumpp Adolfo Garcia-Sastre Lluis Quintana-Murci Monitoring Editors
David Tuveson Patricia Gearhart Klaus Rajewsky Marco Colonna
Editor Emeritus Ronald N. Germain Gwendalyn J. Randolph Jason Cyster
Alan N. Houghton Christopher Goodnow Jeffrey Ravetch Stephen Hedrick
Siamon Gordon Sergio Romagnani Kristin A. Hogquist
Manuscript Coordinator Or Gozani Nikolaus Romani Andrew McMichael
Sylvia F. Cuadrado Sergio Grinstein David L. Sacks Luigi Notarangelo
phone (212) 327-8575
email: jem@rockefeller.edu Philippe Gros Shimon Sakaguchi Anjana Rao
Kristian Helin Matthew D. Scharff Federica Sallusto
Preflight Editor Chyi Hsieh Olaf Schneewind Louis M. Staudt
Rochelle Ritacco Christopher A. Hunter Stephen P. Schoenberger Toshio Suda
Assistant Production Editor Kayo Inaba Hans Schreiber
Shauna OGarro Gerard Karsenty Gerold Schuler
Jay Kolls Robert A. Seder Consulting
Production Editor
Paul Kubes Rafick-P. Skaly Biostatistics Editors
Brianna Caszatt
Vijay K. Kuchroo Charles N. Serhan Glenn Heller
Production Manager Ralf Kuppers Nilabh Shastri Madhu Mazumdar
Camille Clowery
Production Designer
Erinn A. Grady

Copyright to articles published in this journal is held by the authors. Articles are published by
The Rockefeller University Press under license from the authors. Conditions for reuse of the
articles by third parties are listed at http://www.rupress.org/terms

Print ISSN 0022-1007 Online ISSN 1540-9538


INSIGHTS

Immunotherapy and the belly of the beast


Immunotherapy has emerged as an effective means to restore immune recognition of cancer.
Numerous forms of immunotherapy are currently being explored clinically. The most common are
vaccines (dendritic cell, viral, and whole tumor cell based), adoptive T cell therapy and immune
checkpoint blockade. The recent successes in melanoma, renal cancer, and lung cancer have gener-
ated renewed optimism for treatment of multiple cancer types that were believed not amenable to
immune-based therapies. However, immune-related events such as colitis, dermatitis, and, less
frequently, endocrinopathy and pneumonitis have been reported and can be a challenge in the
clinical use of these approaches. Cytokine release syndrome has been reported in the case of CAR Insight from Taha Merghoub (left)
(chimeric antigen receptor) T cell therapy and treatment with agonist antibodies. In this issue, and Jedd Wolchok (right)
Mirsoian et al. provide evidence that adiposity in aged mice induces a lethal cytokine storm follow-
ing systemic administration of stimulatory immunotherapy consisting of anti-CD40 agonist antibody with IL-2.
This is an elegant follow up to a previous study by the same authors in which they showed that systemic immunotherapy adminis-
tration in aged mice resulted in the induction of a rapid and lethal cytokine storm. In the current paper, they find that it is the fat that
accumulates during aging, and mainly the visceral fat, that is associated with toxicity. This results in increased levels of proinflammatory
M1 macrophages within the peritoneal cavity and visceral adi-
pose tissues, leading to heightened production of TNF. In or-
der to confirm that it is indeed the fat that is associated with
lethal effects, the authors repeated their studies in young mice
with genetic (ob/ob) or diet-induced obesity (DIO). While
young obese mice also displayed severe toxicity to the therapy,
it was much more severe in older mice suggesting that age is
also a significant factor. Reciprocally, calorie-restricted aged
mice had lower visceral body fat content and reduced cytokine
levels, with increased survival following immunotherapy.
Obese mice were also protected by macrophage depletion or
TNF blockade. These data demonstrate the need to consider
age and body fat content as variables in preclinical assessment
of therapeutics and when modeling diseases such as cancer.
Since many cancer patients fall into the elderly category,
this study brings up a critical point that aged mice respond dif-
ferently to immunotherapy than younger mice. This also
highlights an important issue when considering potential tox-
icities associated with any therapy, as most animal studies are
performed in young, healthy mice. Another interesting point
is the potential impact of body mass index and adipose accu-
mulation in predicting side effects when investigating and uti-
lizing immunotherapies. This is a very important and impactful
The Murphy group previously showed that immunotherapy with anti-CD40 finding for the field, and further investigations in tumor bear-
and IL-2 leads to a productive immune response in young mice (A) but ing mice with commonly used therapies are warranted. If the
lethal cytokine storm in older mice (B). They now find that the visceral fat findings are confirmed in other therapeutic settings, the inclu-
that accumulates in aged mice (or young obese mice) is the primary trigger
sion of supportive measures, such as TNF blockade, should
of inflammation after immunotherapy, resulting in increased inflammatory
M1 macrophages and toxic cytokine storm (B). Immunotherapy-induced
continue to be considered for early management of immune-
lethality was prevented by blocking TNF or depleting macrophages related toxicities to improve clinical outcomes.
(B). Whether the success of immunotherapy in the treatment of tumor- Mirsoian, A., et al. 2014. J. Exp. Med. http://dx.doi.org/10.1084/
bearing mice will also be impaired by obesity remains to be determined. jem.20140116.

Taha Merghoub and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center: merghout@mskcc.org and wolchokj@mskcc.org

2 INSIGHTS | The Journal of Experimental Medicine


Article

Adiposity induces lethal cytokine storm


after systemic administration of stimulatory
immunotherapy regimens in aged mice
Annie Mirsoian,1* Myriam N. Bouchlaka,1* Gail D. Sckisel,1 Mingyi Chen,2
Chien-Chun Steven Pai,1 Emanuel Maverakis,1 Richard G. Spencer,5
Kenneth W. Fishbein,5 Sana Siddiqui,5 Arta M. Monjazeb,3
Bronwen Martin,5 Stuart Maudsley,5 Charles Hesdorffer,5 Luigi Ferrucci,5
Dan L. Longo,5 Bruce R. Blazar,6 Robert H. Wiltrout,7 Dennis D. Taub,4,8**
and William J. Murphy4**
1Department of Dermatology, 2Department of Pathology and Laboratory Medicine, 3Department of Radiation Oncology,
and 4Department of Dermatology and Internal Medicine, University of California, Davis, Sacramento, CA 95817
5National Institute on Aging-Intramural Research Program, National Institutes of Health, Biomedical Research Center,

Baltimore, MD 21224
6Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455
7National Cancer Institute, Frederick, MD 21702
8Hematology and Immunology Translational Research Center, VA Medical Center, Washington, DC 20422

Aging is a contributing factor in cancer occurrence. We recently demonstrated that sys-


temic immunotherapy (IT) administration in aged, but not young, mice resulted in induction
of rapid and lethal cytokine storm. We found that aging was accompanied by increases in
visceral fat similar to that seen in young obese (ob/ob or diet-induced obese [DIO]) mice.
Yet, the effects of aging and obesity on inflammatory responses to immunotherapeutics are
not well defined. We determine the effects of adiposity on systemic IT tolerance in aged
compared with young obese mice. Both young ob/ob- and DIO-generated proinflammatory
cytokine levels and organ pathologies are comparable to those in aged ad libitum mice
after IT, culminating in lethality. Young obese mice exhibited greater ratios of M1/M2
macrophages within the peritoneal and visceral adipose tissues and higher percentages of
TNF+ macrophages in response to CD40/IL-2 as compared with young lean mice. Macro-
phage depletion or TNF blockade in conjunction with CD40/IL-2 prevented cytokine
storms in young obese mice and protected from lethality. Calorie-restricted aged mice
contain less visceral fat and displayed reduced cytokine levels, protection from organ
pathology, and protection from lethality upon CD40/IL-2 administration. Our data dem-
onstrate that adiposity is a critical factor in the age-associated pathological responses to
systemic anti-cancer IT.

CORRESPONDENCE The use of immunotherapy (IT) in cancer has the discrepancies in observations between clin
William J. Murphy: recently resulted in impressive responses.Yet, the ical and preclinical toxicity merit further study.
wmjmurphy@ucdavis.edu
usages of IT regimens, especially those involving Cancer has been considered a disease of aging,
Abbreviations used: AL, ad cytokine therapies, have also resulted in the in with persons over 65 accounting for over 60%
libitum; ALT, alanine amino duction of severe systemic toxicities often not pre of newly diagnosed malignancies (Balducci and
transferase; CR, calorie restricted; viously characterized in their preclinical animal Ershler, 2005). Consequences of aging include
DIO, diet-induced obese; HD,
high dose; IT, immunotherapy; studies.Because such toxicities often require inten gradual decreases in immunological function,
LD, low dose; MRI, magnetic sive care management of patients, the causes for thymic involution leading to decreased naive
resonance imaging. T cell output, restriction within T cell repertoire
2014 Mirsoian et al. This article is distributed under the terms of an Attribution
*A. Mirsoian and M.N. Bouchlaka contributed equally to NoncommercialShare AlikeNo Mirror Sites license for the first six months after
this paper. the publication date (see http://www.rupress.org/terms). After six months it is
available under a Creative Commons License (AttributionNoncommercialShare
**D.D. Taub and W.J. Murphy contributed equally to Alike 3.0 Unported license, as described at http://creativecommons.org/licenses/
this paper. by-nc-sa/3.0/).

The Rockefeller University Press $30.00


J. Exp. Med. 2014 Vol. 211 No. 12 23732383 2373
www.jem.org/cgi/doi/10.1084/jem.20140116
diversity, increases in genomic instability, and increases in cel study seeks to establish the consequences of increased adipose
lular senescence (Campisi, 2013). However, despite the ma tissue accumulation throughout aging upon IT-induced tox
jority of patients being of this demographic, the overwhelming icities. Here, we find that adiposity results in a skewing toward
majority of preclinical studies are performed in young inbred increased levels of proinflammatory M1 macrophages within
mice.We recently demonstrated that treatment of young mice the peritoneal cavity and visceral adipose tissues that ultimately
with a combination IT regimen consisting of agonistic mono result in heightened production of proinflammatory cytokines,
clonal antibody to CD40 (CD40) given in conjunction with namely TNF, during strong immune stimulation with IT.
IL-2 resulted in synergistic anti-tumor effects and was well Importantly, aged ad libitum (AL)fed mice and young obese
tolerated (Murphy et al., 2003).Yet, administration of the same mice succumb to TNF-mediated pathological responses culmi
regimen in an aged cohort resulted in 100% lethality within nating in rapid lethality, which are prevented through caloric
two days of treatment (Bouchlaka et al., 2013). Importantly, the restriction in the aged or through either macrophage depletion
mortality in the aged was perpetuated by an aberrant cytokine or TNF blockade.
storm mirroring a systemic inflammatory response syndrome
(SIRS), which resulted in multi-organ damage (Bouchlaka et al., RESULTS
2013). These systemic toxicities were closely representative of IT administration in the aged results in rapid lethal cytokine
those previously noted clinically with FDA-approved immu storm and is associated with increased accumulation
nostimulatory IT therapeutics such as high-dose IL-2 and of visceral adipose tissue
IFN- (Lee and Margolin, 2011). The combination IT regimen consisting of CD40 and IL-2
Aging is associated with a gradual decline in immunolog (CD40/IL-2) has previously shown synergistic anti-tumor
ical function but is also accompanied by the coincidence of effects resulting in the regression of metastatic tumors in
a systemic, chronic, and low-grade proinflammatory response young (<6 mo) mouse models (Murphy et al., 2003). Re
termed inflammaging that leads to increased systemic cyto cently, we demonstrated that administration of the therapeu
kine levels, namely IL-1, IL-6, and TNF (Franceschi, 2007). tic dose (high dose [HD]) of CD40/IL-2 into aged mice
Emerging data suggests that inflammaging may be influenced (15 mo) resulted in a heightened cytokine storm that prompted
by age-associated changes in body mass composition such as multi-organ damage and resulted in 100% lethality by day 2
decreased lean muscle mass, increased adiposity, and increases of treatment (Bouchlaka et al., 2013). To address whether the
in overall body mass index (BMI; Franceschi et al., 2000). increased lethality was specific to this therapeutic dosage in
Whether adiposity plays a role in the generation of systemic the aged, we administered CD40/IL-2 at a lower dose (LD),
toxicities or whether it is primarily an age-associated occur less than half, into aged AL-fed mice. Consistent with our
rence remains unknown. previous findings, treatment of aged mice with LD IT re
Like aging, hallmark to the obese microenvironment is sulted in 100% mortality by day 2 of treatment (Fig. 1 A) and
the development of a low-grade chronic meta-inflammatory demonstrated increased serum levels of proinflammatory cy
state. Excess adiposity is associated with increased infiltration tokines TNF, IL-6, and IFN-, indicative of a cytokine storm
of macrophages and proinflammatory mediators such as den (Fig. 1 B). In addition, to ensure that the lethality observed in
dritic cells, NK cells, and T cells into fat depots that ultimately the aged mice was not IT regimenspecific, we administered
results in environmental remodeling. Consequences include an IT combination of CpG and IL-2 (CpG/IL-2) into young
increased circulating levels of C-reactive protein (CRP), IL-6, WT and aged AL mice. Confirming our previous findings,
leptin, and TNF (Ronti et al., 2006;Vzquez-Vela et al., 2008). aged mice treated with CpG/IL-2 therapy incurred a similar
Importantly, the immunomodulatory effects of increased adi cytokine storm that culminated in rapid lethality by day 3
pose tissue extends through the ability for both adipocytes and (Fig. 1, CE), in contrast to young mice which exhibited lower
the stromal vascular fraction to express a variety of immunologi levels of cytokines and demonstrated complete treatment tol
cally important surface receptors, such as the leptin receptor, erance (Fig. 1, CE).
IL-6 receptor, and both p55 and p75 TNF receptors (Ailhaud, Normal aging is inherently associated with changes in body
2000). Systemic consequences of increased adiposity are also mass compositionspecifically with decreases in muscle mass
seen at distance sites through premature induction of aging- and increases in intra-abdominal visceral adiposity (Harris,
associated changes such as thymic involution, which leads to 2002). Equally, within our animal studies an observable differ
the restriction of naive T cell output and ultimately restriction ence between aged and young mice is noted in overall size, in
of the T cell repertoire (Yang et al., 2009). particular increased waist circumference (Fig. 1 F). On average,
Collectively, the field of IT has made great progress in the aged mice weigh twice as much as young (Fig. 1 G).To deter
last decade toward the development of therapeutic candidates mine whether the difference in size is due to increases in fat
against cancer.Yet, these candidates have been met in the clinic deposits, we sought to quantify differences in body mass and
with the induction of severe, often limiting, systemic toxici the presence of visceral adiposity. Young (<6 mo) and aged
ties that hinder their usage. Additionally, given the rise of (15 mo) C57BL/6 mice were analyzed through magnetic
obesity within society, as well as the estimation that cancer is resonance imaging (MRI) to quantify the presence of visceral
a disease primarily of the aged, using lean and young models adiposity. Aged mice exhibited markedly increased body mass
may not accurately reflect patient outcomes. Therefore, this indices and total visceral fat volumes in comparison with young

2374 Obesity induces immunostimulatory therapy toxicity | Mirsoian et al.


Ar ticle

Figure 1. Aged AL mice show increased


proinflammatory cytokines, rapid lethality
to LD IT, and display increased fat volume
ratios in comparison to aged CR mice.
(A) Ad libitumfed C57BL/6 mice were treated
with LD CD40/IL-2 or rIgG/PBS and survival
was monitored, n = 58 mice/group. (B) Serum
cytokines TNF, IL-6, and IFN- were measured
day 2 by CBA after the start of CD40/IL-2 or
rIgG/PBS therapy in aged AL mice, n = 35
mice/group. (C) Young AL (3 mo) and aged AL
(18 mo) mice were treated with CpG/IL-2 IT
and survival was monitored, n = 5 mice/group.
(D and E) Serum cytokine levels of TNF (D) and
IL-6 (E) were measured by CBA on day 2 after
the start of therapy, n = 5 mice/group.
(F) Representative image of aged and young
C57BL/6 mice showing size and waist circum-
ference. (G) Total body weight (left graph) and
visceral fat weight (right graph), was mea-
sured for young (2 mo) and aged AL mice
(20 mo), n = 5 mice/group. (H) Total body
weight (left graph) or visceral fat (right graph)
was measured in naive 15-mo-old C57BL/6
mice on an AL or CR diet, n = 34 mice/group.
(I) Visceral fat in young (4 mo), middle-aged
(9 mo), and aged (15 mo) mice on either an AL
(left) or CR (right) diet was evaluated by MRI.
WS is indicative of water suppression, where
fat is bright. FS is indicative of fat suppression,
fat is dark/black. Data in all panels are repre-
sentative of one of at least five experiments
with similar results. MRI scans are representa-
tive of one of at least three mice imaged per
group with similar results. Survival analysis
was plotted according to the Kaplan-Meier
method, and statistical differences were deter-
mined with the log-rank test. Bar graph (mean
value SEM) statistics were performed using
either one-way or two-way ANOVA with
Bonferroni post-hoc tests. ***, P < 0.001; **,
P < 0.01; *, P < 0.05.

mice (Fig. 1, G and I).These results indicate that aged mice are were similar to young mice (Fig. 1, GI). MRI scans of young
not only heavier and larger in size but that these size differences (4 mo), middle-aged (9 mo), and aged (15 mo) mice that were
are attributed to increases in adipose tissue accumulation. either AL-fed or CR throughout life showed significant dif
Adipose tissue is widely published to be a highly active ferences in adipose accumulation over time (Fig. 1 I). We found
metabolic organ with immune modulatory capabilities. To that with increasing age, AL-fed mice have a proportional in
address whether in our aged AL-fed mice, who were allowed crease in body fat content, whereas mice placed on a CR diet
to freely access food, the increased adiposity noted was con lack increased fat accumulation regardless of age. Aged CR
tributory toward the development of IT-induced toxicities in mice show no significant difference in fat volume ratios to
the aged, we aimed to determine the effects of IT administra their middle-age and young counterparts, and remain similar
tion into an aged calorie-restricted (CR) cohort. CR mice are to young AL mice (Fig. 1 I).
placed on a food restriction diet beginning at 14 wk of age
and maintained throughout life. To ensure that age-matched Caloric restriction in aged mice results in decreased
CR aged mice contained a decreased accumulation of body accumulation of adiposity and protection
fat in comparison with aged AL mice, we quantified total body from IT-induced toxicity
weight and visceral fat through MRI (Fig. 1, H and I). CR To examine the effect of adiposity and aging on the develop
mice weighed significantly less than AL aged mice and exhib ment of systemic toxicities after IT, aged AL, aged CR, and
ited a decreased presence of visceral adiposity to levels that young AL mice were administered IT or a control therapy

JEM Vol. 211, No. 12 2375


Figure 2. Calorie restriction in the aged confers protection by decreasing cytokine levels and IT-associated organ pathology, thereby
allowing increased survival during IT. (AC) Aged AL and age-matched aged CR mice (18 mo) were treated with HD CD40/IL-2 or rIgG/PBS; at day 2
of IT, mice were sacrificed for histological analysis of livers (A) and intestines (C). Serum was collected on day 2 and assessed for ALT (B) levels, n = 3 mice/
group. Representative H&E images of liver and intestines (gut) for aged AL and aged CR groups. Bars, 200 m. Asterisks represent steatosis and arrows
denote areas of severe immune infiltration and/or necrosis. (DF) Serum was analyzed day 2 after treatment for TNF (D), IL-6 (E), and IFN- (F). (G) Aged
(18 mo) C57BL/6 mice on AL or CR diet were treated with either LD or HD of CD40/IL-2 and survival was monitored. Control groups received rIgG/PBS.
All data panels are representative of one of four independent experiments with similar results for all panels. Survival analysis was plotted according to
the Kaplan-Meier method and statistical differences were determined by using the log-rank test. Bar graph (mean value SEM) statistics were performed
using one-way ANOVA with Bonferroni post-hoc test. ****, P < 0.0001; ***, P < 0.001; **, P < 0.01.

(rIgG/PBS). IT resulted in increased lymphocytic infiltrates succumb to mortality on day 2 of treatment (Fig. 2 G). How
to the liver of aged CR and aged AL mice, in contrast to age- ever, 100% of aged CR mice placed on the HD amount of
matched control-treated mice (Fig. 2, AC). To assess whether IT demonstrated complete protection from CD40/IL-2
IT resulted in organ damage, livers and intestines were col mediated lethality (Fig. 2 G). Collectively, these data suggest
lected on day 2 of treatment, the day of mortality in aged AL that increased body fat plays a critical role in the induction of
mice, and assessed for pathological changes. Importantly, livers systemic toxicities by IT. Increased adiposity played a critical
and intestines from aged CR mice demonstrated significantly role in the age-associated cytokine storm and subsequent organ
less inflammation and less necrosis in comparison with aged pathologies after IT and calorie restriction diminished the de
AL mice (Fig. 2, A and C). Serum analysis of alanine amino velopment of such toxicities conferring a protective effect that
transferase (ALT) enzyme levels, a correlate of liver damage, allowed for increased survival in the aged.
showed a significant reduction in ALT levels in aged CR mice
in comparison with aged AL mice (Fig. 2 B).Additionally, serum Increased adipose tissue accumulation through obesity, in the
proinflammatory cytokine analysis of TNF (Fig. 2 D), IL-6 absence of age, results in IT-induced lethal cytokine storm
(Fig. 2 E), and IFN- (Fig. 2 F) resulted in all cytokines being To further dissect the role of adipose tissue in the IT-associated
significantly decreased in aged CR mice in comparison with inflammatory responses seen in aged mice, we next sought to
the aged AL mice. These decreases in cytokine values were to determine the impact of adiposity, in the absence of age, as a
levels that were either lower-than or as-low-as those in young possible cofactor.Young ob/ob mice (B6.Cg-Lepob/ob) are leptin-
AL mice that received IT (Fig. 2, DF). To determine if these deficient and therefore start exhibiting obesity by 3 wk of age.
lower cytokine values during IT therapy would confer protec In comparison to age-matched young WT (2 mo) control mice
tion against IT-induced mortality, aged AL mice and aged CR placed on an AL diet (young AL), ob/ob mice have significantly
mice were administered either LD IT or HD IT and monitored higher body masses and weigh, on average, 2.5 greater
for mortality. Independent of treatment dose, aged AL mice all (Fig. 3 A). MRI analysis of body fat content demonstrated

2376 Obesity induces immunostimulatory therapy toxicity | Mirsoian et al.


Ar ticle

Figure 3. Increased adiposity indepen-


dently of age results in systemic release of
proinflammatory cytokines and organ pa-
thology after IT in young ob/ob (obese)
and aged AL mice. (A) Body weights of naive
young (2 mo) WT and young ob/ob C57BL/6
mice, n = 6 mice/group. Representative pic-
tures of young-AL WT mice and obese ob/ob
naive mice are shown. (B) Total fat content in
naive young WT (2 mo), young ob/ob (2 mo),
and aged (18 mo) C57BL/6 mice was visualized
by MRI. Fat is bright. (C and D) Young AL (2 mo),
young ob/ob (2 mo), and aged AL (18 mo) mice
were treated with HD CD40/IL-2 and liver
(C) and gut (D) were stained with H&E and
scored for histopathology on day 2. Bars,
200 m. Arrows indicate areas of steatosis
and/or immune cell infiltration and asterisks
indicate patchy necrosis and/or fibrosis.
(EG) Young AL (2 mo), young ob/ob (2 mo),
aged AL (18 mo), and aged CR (18 mo) were
treated with LD CD40/IL-2 and serum was
collected day 2 of treatment and analyzed by
CBA for levels of: TNF (E), IL-6 (F), and IFN-
(G), n = 3 mice/group. (H and I) Young AL (2 mo),
young ob/ob (2 mo), aged AL (18 mo), and
aged CR (18 mo) were treated with LD CD40/
IL-2 (H) or CpG/IL-2 (I) or rIgG/PBS control and
survival was monitored. (J) Serum levels of TNF
were measured on day 2 of CpG/IL-2 therapy
by CBA. Data in all panels are representative of
one of at least three experiments with similar
results. MRI scans are representative of at
least three mice imaged per group with similar
results. Survival data were plotted using the
Kaplan-Meier method and statistical differ-
ences were determined with the log-rank test.
Bar graph (mean value SEM) statistics per-
formed using Students t test or one-way
ANOVA with Bonferroni post-hoc tests. ***,
P < 0.001; **, P < 0.01; *, P < 0.05.

that young ob/ob mice have higher total body fat content that was comparable to the aged AL mice and significantly greater
is similar in accumulation to the aged mice, being deposited than the young AL (Fig. 3, C and D).
largely in the intra-abdominal cavity, in comparison with young Because HD IT resulted in significant changes within the
AL mice (Fig. 3 B). obese ob/ob mice, we next administered the lower dose (LD)
To determine if adiposity could solely induce toxic con of IT to young ob/ob, young age-matched AL mice, aged AL,
sequences in aged AL, young ob/ob and age-matched young and aged CR mice. Administration of LD IT still resulted in
AL mice were treated with HD IT and at day 2 of treatment significant increases in TNF and IL-6 proinflammatory cyto
analyzed for the presence of immune-mediated organ patholog kine levels within the young ob/ob that were not significantly
ical changes within the liver and intestines (Fig. 3, C and D). different to the aged AL mice (Fig. 3, E and F). Both ob/ob and
IT resulted in mild inflammatory infiltrates in young AL mice aged AL were significantly greater than both the young AL
but resulted in moderate to severe inflammation in the ob/ob and aged CR mice, where CR in the aged led to significant
mice, which was comparable to the aged within the intestines down-regulation in both TNF and IL-6 cytokine production
(Fig. 3 D).Yet, analysis of IT-induced liver pathology was hin (Fig. 3, E and F). Although the young ob/ob and the aged AL
dered by the presence of extensive steatosis in ob/ob mice, re mice displayed increased levels of IFN-, these levels were not
sulting in a paradoxical interference in determining the intensity significant among the groups (Fig. 3 G). Consistent with the
of immune-mediated inflammatory liver damage (Fig. 3 C). increase in proinflammatory cytokines at day 2 of IT treatment,
IT resulted in both the intestines and liver displaying the most 100% of young ob/ob mice succumbed to IT-induced lethality
immune-mediated inflammatory damage within the ob/ob that within 4 days of treatment in contrast to 100% survival in the

JEM Vol. 211, No. 12 2377


Figure 4. IT toxicity is TNF-dependent through M1 macrophage accumulation. (AD) Young (2 mo) WT and young (2 mo) ob/ob mice were
treated with either control rIgG/PBS or LD CD40/IL-2. Visceral adipose tissue and peritoneal lavages were collected on day 2 and assessed for the pres-
ence of macrophages identified as CD45+CD19F4/80+CD11b+. Ratio of M1/M2 macrophages (A), and total numbers (B) and percentages (C) of TNF-
producing macrophages. Macrophages identified as M1 macrophages were characterized as CD206; M2 macrophages were gated as CD206+. (EH)
Young AL (2 mo) and young ob/ob (2 mo) were treated with either control or macrophage depleting clodronate liposomes before and during LD CD40/IL-2
and survival was monitored; n = 5 mice/group. (F and G) On day 2, visceral adipose tissues and peritoneal lavages were collected and assessed for the
percentages TNF+ macrophages (F and G) as characterized in experiments in AD and serum TNF levels (H), n = 4 mice/group. (IL) Young AL (2 mo) and
young ob/ob (2 mo) mice were treated with either control hIgG/PBS or subcutaneous injection of 1.5 mg/0.1 ml etanercept before and during LD CD40/
IL-2 and survival was monitored (I), n = 5 mice/group. (J and K) On day 2, visceral adipose tissue and peritoneal lavages were collected and assessed by
flow cytometry for the percentages of TNF+ macrophages (J and K) and serum TNF levels (L); n = 4 mice/group. Data in all panels are representative of one
of at least three experiments with similar results. Survival data were plotted using the Kaplan-Meier method and statistical differences were determined
with the log-rank test. Bar graph (mean value SEM) statistics performed using one-way ANOVA with Bonferroni post-hoc tests. **, P < 0.01; *, P < 0.05.

young WT cohort (Fig. 3 H). 100% of aged AL mice yielded This data further supports that increased adiposity plays a
to lethality from the IT by day 2 of treatment, whereas 100% crucial role in the induction of systemic toxicities after IT.
of aged CR mice survived the entire regimen (Fig. 3 H). These data are suggestive that not only does an aging environ
The pathological and lethal response observed within the ment result in lethal toxicities but toxicity may also be depen
young ob/ob mice was not CD40/IL-2 IT administration dent on the increase in fat accumulation as the young ob/ob
specific. Young AL and age-matched young ob/ob mice were mice succumb to cytokine storm induction and mortality after
administered CpG in combination with HD IL-2 and, like IT, similar to that observed in the aged mice.
wise, resulted in lethality of 75% of young ob/ob mice by day 5
of treatment but resulted in complete tolerance by young IT toxicity is TNF- and M1 macrophagedependent within
AL mice (Fig. 3 I). CpG/IL-2 therapy led to significantly in the peritoneum and visceral adipose tissues
creased levels of serum proinflammatory cytokine TNF in an A hallmark of obesity is the increasing infiltration of macro
analogous trend as CD40/IL-2 administration on day 2 of phages into adipose tissues (ATMs) through the secretion of
therapy (Fig. 3 J). MCP-1 by adipocytes. Phenotypically, these macrophages are

2378 Obesity induces immunostimulatory therapy toxicity | Mirsoian et al.


Ar ticle

Figure 5. DIO succumb to cytokine storm,


organ pathology, and lethality similar to aged
AL mice after IT. Young C57BL/6 mice were
placed on a 60% fat diet starting at 3 wk of age
to induce obesity (young DIO). Lean age-matched
controls (young lean) were fed a 10% fat diet
beginning at 3 wk of age. Young mice were ana-
lyzed at 5 mo of age, and aged AL mice were
analyzed at 18 mo of age, n = 6 mice/group. Av-
erage body weight (A), representative pictures (B),
and total fat content as measured by MRI (C) are
shown for each group. (DF) Young lean (5 mo),
young DIO (5 mo), and aged AL (18 mo) mice
were treated with HD CD40/IL-2 or rIgG/PBS.
Serum was collected day 2 after the start of
treatment and analyzed by CBA for levels of TNF
(C), IL-6 (D), and IFN- (E) and survival was ana-
lyzed (F). Data in AC are representative of one of
at least three experiments with similar results and
data in DG are representative of one of at least
four experiments with similar results. Survival
data were plotted using the Kaplan-Meier method
and statistical differences were determined with
the log-rank test. Bar graph (mean value SEM)
statistics performed using one-way ANOVA with
Bonferroni post-hoc tests. ****, P < 0.0001;
***, P < 0.001; **, P < 0.01; *, P < 0.05.

predominately classically activated M1 cells that also express (Fig. 4, IL). Both macrophage depletion and co-treatment
proinflammatory mediators IL-1, TNF, and IL-6 (Lumeng with etanercept during LD CD40/IL-2 resulted in complete
et al., 2007; Fujisaka et al., 2009). As such, we next sought to protection of young ob/ob mice from lethality (Fig. 4, E and I).
determine if increases in M1 macrophages within the perito As expected, treatment with liposomal clodronate led to signif
neal cavity and visceral adipose tissues could account for the icantly decreased levels of TNF-producing macrophages within
pathological increases in systemic TNF during IT administra both the visceral fat and peritoneal lavages (Fig. 4, F and G),
tion.Young AL and age-matched young ob/ob mice were ad which significantly reduced systemic circulating serum levels
ministered LD CD40/IL-2. At day 2 of treatment, visceral of proinflammatory cytokine TNF (Fig. 4 H). Additionally,
adipose tissues and peritoneal lavages were phenotypically as co-administration of LD CD40/IL-2 with etanercept did
sessed by flow cytometry for the presence of M1 (CD206) not reduce the percentages of TNF-expressing macrophages
macrophages, M2 (CD206+) macrophages, and percentages within the visceral fat or peritoneal cavity (Fig. 4, J and K) but
of TNF-expressing macrophages. Consistently, young ob/ob mediated complete protection through an overall reduction
mice demonstrated significant increases in M1 macrophage in systemic TNF levels (Fig. 4 L).
proportions during IT administration, whereas young AL mice Together, these data indicate that proinflammatory M1
were able to maintain consistent M1-to-M2 ratios (Fig. 4 A). macrophages are the cellular mediator in the induction of IT
Additionally, both the visceral adipose tissues and peritoneal toxicity, particularly through increased presence within the peri
lavages of ob/ob mice experienced significant increases in per toneal and visceral adipose tissues. Importantly, these effects are
centages of TNF-expressing macrophages as opposed to young TNF-dependent, as blocking TNF levels either through etan
AL mice (Fig. 4, BD). Significant changes were not observed ercept administration or macrophage depletion resulted in com
in the spleen of either young ob/ob or AL mice (unpublished plete protection of young ob/ob mice from lethality.
data).This data, consistent with obesity literature, suggests that
increases in adipose tissue accumulation within the visceral Diet-induced obese (DIO) mice succumb to cytokine
cavity may impact regulation of M1 and M2 macrophage ra storm, organ pathology, and lethality similar to ob/ob
tios that ultimately may affect TNF responses to immune stim and aged AL mice after IT
ulation with immunotherapeutic regimens. ob/ob mice are congenic for the spontaneous mutation in the
Given the increases in M1 macrophages within the obese OB gene, resulting in leptin hormone deficiency. To extend
mice and dysregulation observed with IT, young ob/ob and our findings, we generated young DIO mice. Mice were placed
young AL mice were treated with LD CD40/IL-2 in conjunc on a purified 60% fat diet to induce obesity beginning at 3 wk
tion with either liposomal clodronate for macrophage deple of age and were determined to be obese when weighing >40 g
tion (Fig. 4, EH) or etanercept (Enbrel) for TNF blockade (DIO). Age-matched counterparts were placed on a matching

JEM Vol. 211, No. 12 2379


purified diet that was 10% fat and served as lean controls. In were observed between young obese and aged AL mice, where
creases in body mass were validated in DIO mice before IT aged mice die by day 2 but young obese mice die at day 4 of
administration (Fig. 5, AC). DIO mice displayed increases in treatment. Therefore, although our data strongly demonstrates
overall body weight and size (Fig. 5, A and B). MRI analysis that adiposity plays a central role in the induction of toxicities,
of body fat content showed increased total body fat accumu aging is also a likely key factor. Therefore, our findings high
lation, largely as increased visceral fat, in young DIO mice that light the importance in considering both the status of aging
was similar to the aged AL mice (Fig. 5 C). and obesity of patients receiving systemic administration of stim
Confirming our previous findings, DIO mice treated with ulatory IT regimens. It will be important to conduct studies
HD CD40/IL-2 displayed increased serum TNF, IL-6, and dissociating the consequences of aging upon increased cytokine
IFN- levels compared with age-matched young mice on a responsiveness, possibly through either investigating epigene
10% fat diet after IT administration (Fig. 5, DF).The increases tic modifications induced by the meta-inflammatory and/or
in proinflammatory cytokines in DIO mice were lower, yet not inflammaging states associated with increased adipose accumu
significantly so, than aged mice after IT (Fig. 4, CE). Young lation or whether aging is accompanied by changes in expres
DIO mice started succumbing to IT lethality at day 3 of treat sion levels of various receptors that allow for a quicker response.
ment (>60%), and <40% were still alive by day 4 (Fig. 5 G). The development of IT regimens continues on a positive
Together, these results suggest that adipose tissue is a criti trajectory with increasing successes within a variety of can
cal component to the development of systemic cytokine storm cers. Active IT has taken a greater comprehensive form focus
and lethality after IT.Yet, due to the observation that treatment ing on activating humoral and cell-mediated immunity, and
of DIO mice did not result in 100% of mice succumbing to these strategies have included antigen-specific vaccines (i.e.,
lethality, aging itself may also likely contribute to the overall MART-1 vaccines), DC-based vaccines (i.e., Sipuleucel-T),
heightened inflammatory responses incurred by IT. inhibitors of checkpoint blockade (i.e., CTLA-4 and PD-1),
and systemic stimulatory regimens through cytokine based
DISCUSSION therapies and application of agonistic antibodies (i.e., CD40,
After initiation of strong immune stimulation with IT, we IFN-, and IL-2). Our data presented herein demonstrates
show that both young obese and aged AL mice demonstrate that both young obese and aged AL mice succumb to rapid
increased levels of adiposity, which led to aberrant increases in lethality during systemic administration of stimulatory thera
both TNF and IL-6 levels that ultimately resulted in multi- pies. Our data strongly suggests that excess adiposity may be a
organ damage. Similar to aging, obesity has been shown to be prognostic factor in determining patient outcome and treat
immunomodulatory, where the cross talk between adipocytes ment tolerance. However, additional studies need to be con
and macrophages is thought to lead toward the development ducted examining the effects of adiposity on the development
and perpetuation of a meta-inflammatory state through con of adverse reactions within other classes of IT regimens, such
tinual NF-B activation, which is currently hypothesized to as checkpoint inhibitors.
be responsible for the induction of metabolic diseases. A study Clinically, we hypothesize that taking into account patient
conducted by Spaulding et al. (1997) examined the effects of BMI, adipose accumulation, and age may better modulate treat
age and calorie restriction on the production of TNF and IL-6 ment type choices as well as the inclusion of supportive thera
in resting mice and demonstrated that serum levels of both peutics such as TNF blockade in conjunction with anti-cancer
cytokines were significantly higher in aged mice in compari therapies that, overall, may increase patient survival and lead
son to young, and yet aged mice subjected to long-term calo to greater anti-cancer responses. Follow-up investigations in
rie restriction resulted in TNF and IL-6 serum levels that tumor-bearing models merit further study.
were comparable to the young mice.These findings lend sup
MATERIALS AND METHODS
port to the protective effects seen within our studies in the
Animals and diets. Female young (25 mo old), middle-aged (912 mo old),
aged CR cohort administered IT. and aged (1522 mo old) C57BL/6 mice were purchased from Charles River
Importantly, the data presented here demonstrate the need or from the animal production area at the National Cancer Institute. Young
for using age and body fat content as variables in preclinical female B6.Cg-Lepob/ob and young age-matched female WT control C57BL/6
assessment of therapeutics and modeling diseases, such as can mice were purchased from The Jackson Laboratory at 812 wk of age.
cer. However, the vast majority of inbred mouse studies are DIO obese and control lean mice were generated from WT C57BL/6
mice purchased at 3 wk of age from Charles River or from the animal produc
housed in specific pathogen-free colonies where normal aging
tion area at the National Cancer Institute. Mice were placed on an open-source
may result in higher body fat content and yet less immune purified diet consisting of either 60% high-fat (D12492) or 10% low-fat
challenge from pathogens making them more susceptible to (D12450J) constitution (Research Diets, Inc.). Age-matched mice were placed
proinflammatory responses and toxicities after immune thera on either a high-fat or low-fat diet beginning at 3 wk of age for 1620 wk
pies. It will therefore be important that future studies ascer and weighed weekly. Mice were deemed obese upon reaching a minimum of
tain responses in mice that have been exposed to pathogens 40 g of weight.
For studies comparing aged AL and CR dietary effects, both female aged
and immune challenges as they age.
CR and age-matched female AL-littermates (1522 mo old) were purchased
Furthermore, our data demonstrate that an obese phenotype from the CR rodent colony at the National Institutes on Aging, National In
resulted in lethal consequences that were ameliorated through stitutes of Health (NIH). In brief, mice were either raised through free access,
calorie restriction in the aged. Also, differences in lethality AL, feeding on the NIH31 regular chow, or placed on a limited daily feeding

2380 Obesity induces immunostimulatory therapy toxicity | Mirsoian et al.


Ar ticle

Table 1. Grading of acute liver damage


Global assessment Criteria
0: None No portal inflammation
I: Minimal/indeterminate Portal inflammatory infiltrate is in a minority of the portal triads, which is minimal
II: Mild Inflammatory infiltrate in 50% of the triads, which is generally mild, and confined within the portal spaces
III: Moderate Inflammatory infiltrate, expanding most or all of the triads, and associated with mild interface and lobular activity
IV: Severe As above for moderate, with spillover into periportal areas and moderate to severe perivenular inflammation that
extends into the hepatic parenchyma and is associated with perivenular hepatocyte necrosis

schedule with the NIH31-fortified chow. CR is initiated at 14 wk of age at Murine macrophage studies. On day 2 of treatment, murine macrophages
10% restriction, increased to 25% restriction at 15 wk, and to 40% restriction were isolated from either the peritoneal cavity or visceral adipose tissue. For
at 16 wk of age where it is maintained throughout the life of the animal. In peritoneal lavages, 4 ml of cold PBS was injected into the peritoneal cavity
formation on survival curves, food intake curves, and body weights of the of each mouse and then aspirated. Lavages were spun down at 1,200 rpm for
NIA strains has been previously published (Turturro et al., 1999). 5 min and the resulting pellet was resuspended in RF10c media and plated
All experimental mice were housed in the Animal Facilities at the Uni for flow cytometry staining.
versity of California, Davis under specific pathogen-free (SPF) conditions. The stromal vascular fraction of visceral adipose tissues was collected day
For the comparison in fat volume ratios, young, middle-aged, and aged AL 2 of treatment. Adipose tissues from the visceral cavity (gonadal) were col
and CR mice were ordered and sent to the National Institute on Aging for lected and then incubated for 60 min in a digestion solution consisting of
MRI analysis and also housed under SPF conditions. All of the animal proto 2 mg/ml type IV collagenase (Sigma-Aldrich) dissolved fresh in a 2% BSA in
cols were approved by the Institutional Animal Care and Use Committees of PBS solution. After incubation samples were centrifuged at 1,200 rpm for
both institutions. Body weights were also measured for several mice at differ 5 min for layer separation of the stromal vascular fraction from adipocytes.
ent ages and on dietary regimens. Mice were euthanized by CO2 asphyxia The resulting stromal vascular fraction was then removed, resuspended into
tion and the visceral fat pads (mesenteric, gonadal, and renal) were collected a single cell suspension, counted, and plated for flow cytometry staining.
and weighed together for each mouse to determine the visceral fat weight.
Flow cytometry analysis of macrophages. Single cell suspensions from
Reagents. The agonistic antimouse CD40 antibody (clone FGK115B3) the peritoneal lavages and visceral adipose tissues were plated in RF10c media
was generated via ascites production in our laboratory as previously described with the addition of GolgiPlug (containing Brefeldin A) and GolgiStop (con
(Murphy et al., 2003; Bouchlaka et al., 2013). The endotoxin level of the taining Monensin) Protein Transport Inhibitors (BD) according to manufac
CD40 was <1 endotoxin unit/mg of antibody as determined by a quantita turers protocols. Plates were incubated for 9 h at 37C and 5% CO2.
tive chromogenic limulus amebocyte lysate kit (QCL-1000; Bio Whittaker). After incubation, cells were washed with PBS and stained for flow analy
Recombinant human IL-2 (rhIL-2; TECIN Teceleukin) was provided by the sis. Single cell suspensions were labeled with Fc block (purified antimouse
National Cancer Institute (NCI). Murine CpG ODN 1826 was purchased CD16/32; BD) for 10 min, followed by labeling with antibodies for 20 min
from InvivoGen. All were injected i.p. at 4C. Cells were washed twice with a staining buffer consisting of 5% FBS
(Gemini Bio-products) in DPBS (Corning). For intracellular staining, the
Cytofix/Cytoperm kit (BD) was used per manufacturers protocols. After
Schedule of CD40/IL-2 and CpG/IL-2 IT treatments. Mice were staining, cells were analyzed using a custom configured Fortessa cytometer,
treated with agonist CD40 antibody and rhIL2 as previously described and by using FACSDiva software (BD). Data were analyzed using FlowJo
(Murphy et al., 2003; Bouchlaka et al., 2013). CD40 or CpG was adminis software vX (Tree Star). Antibodies used to distinguish macrophages were:
tered daily for a total of 5 consecutive days (days: 0, 1, 2, 3, and 4) and IL-2 PB-conjugated antimouse CD45, BV711-conjugated antimouse CD11b,
was administered twice a day for a total of 4 d (days: 1, 4, 8, and 12). Control BV605- and PE-conjugated antimouse CD206, BV785-conjugated anti
mice received rat IgG (rIgG; Jackson ImmunoResearch Laboratories, Inc.) mouse CD19, APC-conjugated antimouse F4/80, and PE-Cy7conjugated
and PBS (Cellgro). Survival was monitored daily. An HD or LD of CD40/ antimouse TNF (BioLegend).
IL-2, or HD CpG/IL-2, was administered as follows on the same scheduling
pattern as previously described (Bouchlaka et al., 2013): HD CD40/IL-2, Serum cytokine bead array. Serum levels of TNF, IFN-, and IL-6 were
mice received 80 g of agonist CD40 and 106 IU of IL-2 in 0.2 ml PBS i.p.; quantified by multiplex measurement using the Cytometric Bead Array
control mice received 80 g rIgG in PBS; LD CD40/IL-2, mice received (CBA; BD) kit as described previously (Bouchlaka et al., 2013). In brief,
40 g of agonist CD40 and 4 105 IU of IL-2 in 0.2 ml PBS i.p; control serum samples and standards were incubated for 1 h at 25C with a bead
mice received 40 g rIgG in PBS; CpG/HD IL-2, mice received 100 g CpG mixture containing bound antibodies to TNF, IFN-, and IL-6 according to
ODN 1826 (InvivoGen) and 106 IU of IL-2 in 0.2 ml PBS i.p. manufacturers instructions. Samples and standards were resuspended in wash
buffer and analyzed by flow cytometry. Data were acquired on a custom-
Macrophage depletion and TNF blockade with etanercept (Enbrel). configured Fortessa cell analyzer using FACSDiva software (BD) and analyzed
In some experiments, mice were in vivo depleted of macrophages using lipo using FlowJo software (Tree Star). Each sample was analyzed in triplicate.
somal clodronate or control-loaded liposomes (Encapsula NanoScience) be Upon analysis of raw data, the mean fluorescent intensities (MFIs) of each
fore and during IT administration, as previously described (Bouchlaka et al., bead cluster were quantified.Where indicated in the figures, protein concen
2013). For experiments where mice were euthanized by day 2 of treatment, trations were extrapolated relative to a standard curve created by serial dilu
0.2 ml per dose was injected i.p. of either clodronate or control liposomes on tion of the mouse positive control cytokine.
days 2 and 0. For all survival experiments, clodronate or control liposomes
were administered on days 2, 0, 2, and 4. Colorimetric liver enzyme assay. Serum ALT was quantified using the
TNF blockade was preformed through in vivo subcutaneous injection ID Labs ALT Enzymatic Assay kit (ID Labs Biotechnology Inc.) as previously
of 1.5 mg/0.1 ml etanercept (Enbrel) on days 1 and 1 for experiments where described (Bouchlaka et al., 2013). Colorimetric determination of ALT levels
mice were euthanized on day 2 of IT therapy. For all survival experiments, was performed by reading the absorbance of each well at 340 nm on a plate
etanercept was administered on days 1, 1, 3, 7, and 9. reader (VERSAmax turntable plate reader).The concentration of ALT (U/liter)

JEM Vol. 211, No. 12 2381


Table 2. Grading of acute intestinal damage
Global assessment Criteria
0: None No inflammation
I: Minimal/indeterminate Scattered infrequent inflammatory infiltrate in a minority of crypts, which is minimal
II: Mild Inflammatory infiltrate in 50% of the crypts, which is generally mild, and confined within the mucosa epithelium with
mild architectural distortion
III: Moderate Inflammatory infiltrate involving most of the crypts, and associated with architectural distortion (villous blunting and
focal mucosal atrophy/erosion) and luminal inflammatory exudate
IV: Severe Extensive inflammatory infiltrate (large lymphoid aggregates) with expanding into the lamina/muscular propria, and
associated with prominent mucosal ulceration/denuding, muscular wall necrosis, and/or perforation

in each sample was then directly determined from the change in absorbance normal body temperature using warm air. Mice were inserted into the scan
within 5 min time. Dilutions of the Pyruvate Control, included in the kit, ner in a head-first prone orientation. Spin-echo T1-weighted images were
were used to construct a standard curve to calibrate the assay. Every serum obtained through the mouse body (neck-to-base of the tail) using a 72-mm
sample was assayed in triplicate. linear volume coil. Scan sequence parameters were the following: TR 1,000 ms,
TE 15 ms, and 2 averages. The field of view was 7.7 3.85 2.0 cm, with a
Histopathology and grading/scoring. Liver and whole intestines were matrix size of 256 128 40. The corresponding voxel size was 0.3 0.3
collected on day 2 of IT, flushed, and fixed in 10% paraformaldehyde, embed 0.5 mm. Images were acquired using ParaVision 5.0 software. After acquisi
ded in paraffin, cut into 5-m sections, and stained with hematoxylin and tion, images were transferred into Invenon Research Workplace 4.0 software
eosin (H&E). All tissues were prepared and stained at Histology Consultation (Siemens Preclinical) allowing fat to be displayed with high intensity (white).
Services, Inc. (Everson, WA). Images were captured with a microscope (BX4;
Olympus) equipped with a Q-color3 camera and 10 numerical aperture Statistical analysis. Statistical analyses were performed using Prism soft
objective lens. Magnification for each captured image is specified in the figure ware (GraphPad Software Inc.). Data were expressed as mean SEM. For
legends. Grading of histopathological inflammation was performed using a analysis of three or more groups, a nonparametric ANOVA test was per
scale from 0 to 4 in a blind fashion by a board-certified pathologist (M. Chen) formed with a Bonferroni post-hoc test. Analysis of differences between two
at the UC Davis Medical Centers Department of Pathology and Laboratory normally distributed test groups was performed using the Students t test.
Medicine. Specifically, the grading score for liver and gastrointestinal tract Non-parametric groups were analyzed with the Mann-Whitney test.Welchs
followed our previously published grading method (Bouchlaka et al., 2013), correction was applied to datasets with significant differences in variance be
summarized in Tables 1 and 2. fore Students t test.

We thank Monja Metcalf and Weihong Ma for technical help.


MRI. To quantify abdominal fat, MR images were acquired in collaboration
This work has been supported by NIH grants CA0905572 and AG034874 and
with the National Institute on Aging. Images were acquired using a Biospec 7
by the Intramural Research Program of the National Institute on Aging, NIH.
Tesla 30-cm MRI scanner (Bruker Biospin) with a 72-mm diameter transmit- The authors declare no competing financial interests.
receive birdcage coil. In each experiment, two mice were inserted into the
scanner, side by side, in feet-first prone orientation immediately after sacrifice. Author contributions: A. Mirsoian, M.N. Bouchlaka, D.D. Taub, and W.J. Murphy
During scanning, the mice were cooled with a stream of cold air from a vortex designed the research; A. Mirsoian, M.N. Bouchlaka, G.D. Sckisel, M. Chen, C.C.S. Pai,
tube (Exair, Inc.) to minimize decomposition. Two different pulse sequences A.M. Monjazeb, and D.D. Taub performed the research; A. Mirsoian, M.N. Bouchlaka,
were used: a heavily T1-weighted fast spin echo (RARE) sequence yielding D.D. Taub, and W.J. Murphy analyzed the data; M. Chen analyzed and scored all
bright-fat (water-suppressed [WS]) images and a fat-suppressed proton density histology data; E. Maverakis provided etanercept (Enbrel) for TNF blockade studies;
weighted fast gradient echo (FLASH) sequence yielding fat-suppressed (FS) R.G. Spencer, K.W. Fishbein, and S. Siddiqui performed and analyzed MRI studies;
images. 3D datasets were acquired in axial orientation with a field of view of B. Martin, C. Hesdorffer, L. Ferrucci, and S. Maudsley provided aged mice and assisted
72 36 80 mm (left-right anterior-posterior head-foot) and matrix size with data interpretation; A. Mirsoian, M.N. Bouchlaka, D.D. Taub, and W.J. Murphy
256 128 128.The voxel size (spatial resolution) was 281 281 625 m. wrote the paper; and G.D. Sckisel, D.L. Longo, B.R. Blazar, and R.H. Wiltrout helped with
The spectral bandwidth in both sequences was 100 kHz (391 Hz/pixel) and the editing of the paper. All authors read and approved the manuscript.
two averages were acquired for improved signal-to-noise ratio. For the RARE
sequence, the RARE factor (i.e., number of spin echoes per shot or number Submitted: 18 January 2014
of k-space lines per segment) was 8, the repetition time (TR) was 125 ms, the Accepted: 6 October 2014
actual echo time (TE) was 11.4 ms, and the effective echo time (TEeff) was
46.3 ms. Each RARE scan took 8 min and 32 s to acquire. For the FLASH REFERENCES
sequence, the repetition time was 25 ms and the echo time was 3.2 ms. The Ailhaud, G. 2000. Adipose tissue as an endocrine organ. Int. J. Obes. Relat.
excitation pulse had a flip angle of 30. Each FLASH scan took 13 min 39 s to Metab. Disord. 24:S1S3. http://dx.doi.org/10.1038/sj.ijo.0801267
acquire. Image processing was performed in ImageJ 1.45 (NIH). The statisti Balducci, L., and W.B. Ershler. 2005. Cancer and ageing: a nexus at several lev
els. Nat. Rev. Cancer. 5:655662. http://dx.doi.org/10.1038/nrc1675
cal analyses were performed using one-way ANOVA and the Newman-Keuls
Bouchlaka, M.N., G.D. Sckisel, M. Chen, A. Mirsoian, A.E. Zamora, E.
post-hoc test. P < 0.05 was considered to be significant.
Maverakis, D.E. Wilkins, K.L. Alderson, H.H. Hsiao, J.M. Weiss, et al.
To visualize fat distribution and content imaging of young ob/ob, DIO,
2013. Aging predisposes to acute inflammatory induced pathology after
and lean age-matched controls were conducted in collaboration with the tumor immunotherapy. J. Exp. Med. 210:22232237. http://dx.doi.org/
Center for Molecular and Genomic Imaging (CMGI) Facility at the Univer 10.1084/jem.20131219
sity of California, Davis. Mice were anesthetized before imaging using 2% Campisi, J. 2013. Aging, cellular senescence, and cancer. Annu. Rev. Physiol.
isoflurane in an induction chamber and then maintained on 12% isoflurane 75:685705. http://dx.doi.org/10.1146/annurev-physiol-030212-183653
throughout imaging via a nose cone fitted for anesthetic inhalation. Mice Franceschi, C. 2007. Inflammaging as a major characteristic of old people: can
were imaged using a BioSpec 70/30 7T (Bruker Biospin) horizontal bore it be prevented or cured? Nutr. Rev. 65:S173S176. http://dx.doi.org/
system designed specifically for small animal imaging. Animals were kept at 10.1111/j.1753-4887.2007.tb00358.x

2382 Obesity induces immunostimulatory therapy toxicity | Mirsoian et al.


Ar ticle

Franceschi, C., M. Bonaf, S. Valensin, F. Olivieri, M. De Luca, E. Ottaviani, anti-tumor responses after administration of agonistic antibodies to CD40
and G. De Benedictis. 2000. Inflamm-aging. An evolutionary perspective and IL-2: coordination of dendritic and CD8+ cell responses. J. Immunol.
on immunosenescence. Ann. N.Y. Acad. Sci. 908:244254. http://dx.doi 170:27272733. http://dx.doi.org/10.4049/jimmunol.170.5.2727
.org/10.1111/j.1749-6632.2000.tb06651.x Ronti, T., G. Lupattelli, and E. Mannarino. 2006. The endocrine function of
Fujisaka, S., I. Usui, A. Bukhari, M. Ikutani, T. Oya, Y. Kanatani, K. Tsuneyama, adipose tissue: an update. Clin. Endocrinol. (Oxf.). 64:355365.
Y. Nagai, K. Takatsu, M. Urakaze, et al. 2009. Regulatory mechanisms Spaulding, C.C., R.L.Walford, and R.B. Effros. 1997. Calorie restriction inhib
for adipose tissue M1 and M2 macrophages in diet-induced obese mice. its the age-related dysregulation of the cytokines TNF-alpha and IL-6 in
Diabetes. 58:25742582. http://dx.doi.org/10.2337/db08-1475 C3B10RF1 mice.Mech.Ageing Dev. 93:8794.http://dx.doi.org/10.1016/
Harris, T.B. 2002. Invited commentary: body composition in studies of aging: S0047-6374(96)01824-6
new opportunities to better understand health risks associated with weight. Turturro, A., W.W. Witt, S. Lewis, B.S. Hass, R.D. Lipman, and R.W. Hart.
Am. J. Epidemiol. 156:122124, discussion :125126. http://dx.doi.org/ 1999. Growth curves and survival characteristics of the animals used
10.1093/aje/kwf024 in the Biomarkers of Aging Program. J. Gerontol. A Biol. Sci. Med. Sci.
Lee, S., and K. Margolin. 2011. Cytokines in cancer immunotherapy. Cancers 54:B492B501. http://dx.doi.org/10.1093/gerona/54.11.B492
(Basel). 3:38563893. http://dx.doi.org/10.3390/cancers3043856 Vzquez-Vela, M.E., N. Torres, and A.R. Tovar. 2008. White adipose tissue
Lumeng, C.N., S.M. Deyoung, J.L. Bodzin, and A.R. Saltiel. 2007. Increased in as endocrine organ and its role in obesity. Arch. Med. Res. 39:715728.
flammatory properties of adipose tissue macrophages recruited during diet- http://dx.doi.org/10.1016/j.arcmed.2008.09.005
induced obesity. Diabetes. 56:1623. http://dx.doi.org/10.2337/db06-1076 Yang, H., Y.H. Youm, B. Vandanmagsar, J. Rood, K.G. Kumar, A.A. Butler,
Murphy, W.J., L. Welniak, T. Back, J. Hixon, J. Subleski, N. Seki, J.M. and V.D. Dixit. 2009. Obesity accelerates thymic aging. Blood. 114:3803
Wigginton, S.E.Wilson, B.R. Blazar,A.M. Malyguine, et al. 2003. Synergistic 3812. http://dx.doi.org/10.1182/blood-2009-03-213595

JEM Vol. 211, No. 12 2383


INSIGHTS

Cr itical role for CX 3 CR1 + mononuclear phagocytes in


intestinal homeostasis
A key challenge at the intestinal barrier is to minimize responses to commensal bacteria, which can lead to
inflammatory bowel disease (IBD) in genetically predisposed individuals, but retain the ability to recognize
and control the growth of infectious pathogens. Group 3 innate lymphoid cells (ILC3) help maintain intes-
tinal homeostasis by producing the cytokine IL-22, which promotes mucosal healing and maintains barrier
integrity. Microbial signals trigger production of IL-23 and IL-1b, which stimulate ILC3s to produce IL-22,
leading to the induction of antibacterial peptides and epithelial cell regeneration.
But the identity of the cell type producing IL-23 in response to microbial signals is unclear and has
Insight from been the subject of much debate; resident mononuclear phagocytes, inflammatory monocytes, and
Giorgio Trinchieri
conventional dendritic cells have all been implicated. In this issue, Longman et al. provide compelling
evidence, both in mouse following Clostridium rodentium infection and in patients with colitis, that
CX3CR1+ mononuclear phagocytes (MNPs) are the most potent producers of IL-23 and IL-1b and are very efficient in
inducing IL-22 production by ILC3.
The authors demonstrated the importance of microbial stimulation in IL-22 induction
in patients with surgical diversion of the fecal streamIL-22 production by ILC3 was lower in
the sites unexposed to the gut microbiota compared with exposed sites. Microbial TLR4 and
TLR9 agonists were particularly efficient at inducing IL-23 and IL-1b production by CX3CR1+
MNPs. The authors also discovered that a gene significantly associated with both ulcerative
colitis and Crohns disease in GWAS, TNF-like ligand 1A (TL1A or TNFSF15), is overexpressed
in mouse CX3CR1+ MNPs and synergizes with IL-23 and IL-1b to induce IL-22 production
in both human and mouse ILC3.
The study does not completely exclude the role of other cell types in the production of IL-23 and
induction of IL-22 production but in the conditions studied (mouse infection with C. rodentium and
human colitis), CX3CR1+ MNPs were crucial in mediating this mucosal protective loop. In basal
conditions or in other types of inflammatory or preneoplastic conditions, and under stimulation by
different microbial TLR agonists (e.g., flagellin), it is quite possible that other cell types, including
conventional dendritic cells, may play an important role, as suggested by published studies. However, Confocal immunofluorescence
the study by Longman et al. greatly contributes to our understanding of the mechanisms of human image of mouse colon shows
IBD and reassures us that when mouse data are carefully combined with experimental human studies juxtaposition (white arrows)
and GWAS, they are powerful in providing mechanistic evidence that explains human pathology. of CX3CR1+ MNPs (green) and
Longman, R.S., et al. 2014. J. Exp. Med. http://dx.doi.org/10.1084/jem.20140678. RORgt+ ILC3 (red).

Giorgio Trinchieri, National Cancer Institute, National Institutes of Health: trinchig@mail.nih.gov

2 INSIGHTS | The Journal of Experimental Medicine


Article

CX3CR1+ mononuclear phagocytes support


colitis-associated innate lymphoid cell
production of IL-22
Randy S. Longman,1,3 Gretchen E. Diehl,1 Daniel A. Victorio,1,3
Jun R. Huh,1 Carolina Galan,1 Emily R. Miraldi,1,5,6 Arun Swaminath,4
Richard Bonneau,5,6 Ellen J. Scherl,3,4 and Dan R. Littman1,2
1The Kimmel Center for Biology and Medicine of the Skirball Institute and 2Howard Hughes Medical Institute, New York
University School of Medicine, New York, NY 10016
3The Jill Roberts Center for IBD, Department of Medicine, Weill-Cornell Medical College, New York, NY 10021
4Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Medical Center, New York, NY 10032
5Center for Genomics and Systems Biology, Department of Biology; and 6Courant Institute of Mathematical Sciences,

Computer Science Department, New York University, New York, NY10003

Interleukin (IL)-22producing group 3 innate lymphoid cells (ILC3) promote mucosal heal-
ing and maintain barrier integrity, but how microbial signals are integrated to regulate
mucosal protection offered by these cells remains unclear. Here, we show that in vivo
depletion of CX3CR1+ mononuclear phagocytes (MNPs) resulted in more severe colitis and
death after infection with Citrobacter rodentium. This phenotype was rescued by exogenous
IL-22, which was endogenously produced by ILC3 in close spatial proximity to CX3CR1+
MNPs that were dependent on MyD88 signaling. CX3CR1+ MNPs from both mouse and
human tissue produced more IL-23 and IL-1 than conventional CD103+ dendritic cells
(cDCs) and were more efficient than cDCs in supporting IL-22 production in ILC3 in vitro
and in vivo. Further, colonic ILC3 from patients with mild to moderate ulcerative colitis or
Crohns disease had increased IL-22 production. IBD-associated SNP gene set analysis
revealed enrichment for genes selectively expressed in human intestinal MNPs. The product
of one of these, TL1A, potently enhanced IL-23 and IL-1-induced production of IL-22
and GM-CSF by ILC3. Collectively, these results reveal a critical role for CX3CR1+ mono-
nuclear phagocytes in integrating microbial signals to regulate colonic ILC3 function in IBD.

CORRESPONDENCE Inflammatory bowel disease (IBD) has been de- characterized in mouse models of colitis as pre-
Randy S. Longman: fined as a dysregulated cellular immune response dominant producers of IL-22 (Satoh-Takayama
ral2006@med.cornell.edu
to environmental triggers in genetically predis- et al., 2008), an IL-10 family member that sig-
OR
Dan R. Littman: posed individuals. Although the initial discovery nals via STAT3 to regulate mucosal healing, a
dan.littman@med.nyu.edu linking single-nucleotide polymorphisms in the critical clinical endpoint in IBD (Pickert et al.,
IL23R locus with susceptibility to IBD (Duerr 2009; Hanash et al., 2012). In light of their ro-
Abbreviations used: CD,
Crohns disease; DT, diphtheria et al., 2006) was consistent with a role for IL-23 bust production of IL-22 and close proximity
toxin; DTR, DT receptor; IBD, responsive T cells, more recent evidence supports to the intestinal epithelial layer (Cella et al., 2009),
inflammatory bowel disease; the importance of IL-23responsive innate lym- ILC3 have been proposed to play an important
ILC3, group 3 innate lymphoid
cell; MAMP, microbe-associated
phoid cells (ILC) in maintaining epithelial ho- role in mucosal healing and maintenance of
molecular pattern; MNP, meostasis (Sonnenberg and Artis, 2012). These barrier integrity, and understanding how they
mononuclear phagocyte; RORt-dependent ILCs (now named group 3 are induced to produce IL-22 has great poten-
PAMP, pathogen-associated
ILCs, or ILC3 (Spits et al., 2013)) were initially tial for therapeutic benefit.
molecular pattern; UC, ulcer-
ative colitis.
R.S. Longman and G.E. Diehl contributed equally
to this paper. 2014 Longman et al. This article is distributed under the terms of an Attribution
J.R. Huhs present address is University of Massachusetts NoncommercialShare AlikeNo Mirror Sites license for the first six months
Medical School, Worcester, MA 01605. after the publication date (see http://www.rupress.org/terms). After six months
it is available under a Creative Commons License (AttributionNoncommercial
A. Swaminaths present address is Lennox Hill Hospital, Share Alike 3.0 Unported license, as described at http://creativecommons.org/
New York, NY 10075. licenses/by-nc-sa/3.0/).

The Rockefeller University Press $30.00


J. Exp. Med. 2014 Vol. 211 No. 8 1571-1583 1571
www.jem.org/cgi/doi/10.1084/jem.20140678
Mononuclear phagocytes (MNPs) are sentinels of the intes- a mouse with the diphtheria toxin receptor (DTR) cDNA in-
tinal lamina propria, capable of responding to microbial prod- serted into the Cx3cr1 locus (Diehl et al., 2013). Analysis of
ucts, and play a crucial role in orchestrating intestinal lymphocyte colonic lamina propria mononuclear cells (LPMCs) after infec-
homeostasis. MNPs can be subdivided based on their expression tion of DT-treated mice revealed a reduction in the percentage
of CD103 or CX3CR1, and each group has been ascribed criti- of CD11c+ MHCII+ LPMCs (Fig. 1 A), which reflected a pref
cal functions in maintaining intestinal homeostasis (Bogunovic erential loss of the CX3CR1+ CD11b+ CD14+ fraction of MNPs
et al., 2009; Merad et al., 2013). CD103+ cells, which can be (Fig. 1 B; Tamoutounour et al., 2012), as well as CX3CR1+
further subdivided based on the expression of CD11b, differen- monocytes in Cx3cr1DTR/+ mice compared with control mice.
tiate from a common DC precursor and are thought to be CD103+ CD11b+ cDCs were not depleted (Fig. 1 C).To induce
the conventional, migratory myeloid DCs (Varol et al., 2010). colitis, mice were infected with C. rodentium, a mouse model
CD103+ CD11b DCs require Irf8, Id2, and Batf3 for their de- for infectious colitis (Zheng et al., 2008; Sonnenberg et al., 2011;
velopment and are thought to play a critical role in cross-priming Qiu et al., 2012). DT-treated infected Cx3cr1DTR/+ mice, but
virus- and tumor-specific CTLs (Hildner et al., 2008; Merad not uninfected or control infected mice, lost more weight
et al., 2013). Loss of these cells, however, does not alter intestinal (Fig. 1 D), displayed more severe intestinal pathology (Fig. 1 E),
T cell homeostasis or lead to spontaneous inflammation (Edelson and ultimately succumbed to infection (Fig. 1 F). Infected
et al., 2010). CD103+CD11b+ DCs, in contrast, require Notch2 Cx3cr1DTR/+ mice also had increased bacterial burden in the
signaling, produce IL-23 in response to flagellin-induced TLR5 spleen, consistent with the loss of barrier integrity (Fig. 1 G).
activation, resulting in IL-22 production by ILC3, and have ad- To examine potential involvement of signaling pathways
ditionally been proposed to support Th17 polarization (Lewis for receptors of pathogen- or microbe-associated molecular
et al., 2011; Kinnebrew et al., 2012). These cells can produce patterns (PAMPs or MAMPs) in mediating this phenotype, mice
retinoic acid, which promotes the expression of the gut-homing with a conditional deletion of MyD88 in CD11c-expressing
receptor CCR9 and synergizes with TGF to induce regulatory MNPs (CD11c-Cre/Myd88fl/fl) were infected with C. roden-
T cells (Sun et al., 2007). One recent study suggests that Notch2- tium. Infection of CD11c-Cre/Myd88fl/fl mice, but not litter-
dependent CD103+ CD11b+ DCs regulate protection from mate controls, was lethal by 15 d after infection (Fig. 2 A),
C. rodentiuminduced colitis (Satpathy et al., 2013). However, implicating PAMP/MAMP signaling as having a critical role
specific depletion of CD103+ CD11b+ intestinal DCs revealed in barrier protection mediated by CD11c-expressing MNPs.
that these cells are not the MNP subset required for protection The C. rodentium colitis model depends on IL-22 for protec-
against C. rodentium or IL-22 production (Welty et al., 2013). tion (Zheng et al., 2008). Thus, to test if exogenous IL-22
In contrast to CD103+ cDCs, CX3CR1+ MNPs differenti- could rescue the susceptibility phenotype described above,
ate from monocyte precursors (Varol et al., 2010). Although CD11c-Cre/Myd88fl/fl (Fig. 2 A) and DT-treated CX3CR1-
these cells were previously thought to be tissue-resident and to DTR (Fig. 2 B) mice were hydrodynamically injected with a
promote local Treg differentiation (Hadis et al., 2011), recent data plasmid encoding IL-22 (Qiu et al., 2012). The exogenous
from our group showed that they can up-regulate CCR7 and IL-22 rescued both lines of mice from colitis-induced death.
migrate to secondary lymphoid organs, suggesting a broader role
in orchestrating immunity (Diehl et al., 2013). Notably, we ob- Colonic CX3CR1+ MNPs regulate ILC3 production of IL-22
served that interaction with microbiota limits the migration of High-dose infection with C. rodentium is controlled by ILC3,
these cells to mesenteric LNs (MLNs; Diehl et al., 2013), and an which represents the large majority of LPMCs producing IL-22
increase in CX3CR1+ cells has been described in the lamina (Sonnenberg et al., 2011). At day 7 after infection, both the
propria during mouse (Zigmond et al., 2012) and human colitis percentage and absolute number of IL-22+ colonic, lineage,
(Kamada et al., 2008). A recent study reported that fractalkine CD90hi, and RORt+ ILCs (Fig. S1 shows gating strategy)
receptor (CX3CR1) expression supports innate celldependent from mice depleted for CX3CR1+ cells were reduced in com-
clearance of C. rodentium infection (Manta et al., 2013), but a parison to ILCs from mice with intact CX3CR1+ cells (Fig. 2,
functional role for CX3CR1+ MNPs in regulating colitis- CE). Depletion of CX3CR1+ cells did not affect the abso-
associated ILC3 remains unclear.To evaluate this question, we lute number of ILC3 (Fig. 2 F). Although T cells can also con-
employed novel mouse models to enable selective depletion of tribute to IL-22 production in low-dose C. rodentium infection
CX3CR1+ MNPs in vivo. Our results reveal a critical role for (Basu et al., 2012), no statistically significant difference in the
CX3CR1+ MNPs from both mouse and human tissue in sup- total IL-22+ (or IL-17+) T cells was noted in mice depleted
porting IL-22 induction in ILC3 in vitro and in vivo. Moreover, for CX3CR1+ cells (Fig. 2 G). To assess the ability of colonic
we identify the ability of TL1A produced by MNPs to potently CX3CR1+ MNPs to interact with ILC3 within the colonic tis-
enhance IL-23 and IL-1induced production of IL-22 and sue, Cx3cr1GFP/+ mice were used to visualize CX3CR1+ MNPs
GM-CSF by ILC3. in situ ( Jung et al., 2000). Consistent with the ability of these
cells to regulate ILC3 function, confocal microscopy revealed
RESULTS the spatial proximity of RORt+ ILC3 cells with CX3CR1+
CX3CR1+ cells protect against C. rodentiuminduced colitis MNPs in the colonic lamina propria (Fig. 2 H).
To investigate the role of the expanded population of CX3CR1+ To evaluate the ability of intestinal CX3CR1+ cells and
cells in the intestinal lamina propria during colitis, we generated cDCs to support ILC3 activation, CX3CR1+ (GFP+) cells and

1572 CX3CR1+ MNPs regulate ILC3 | Longman et al.


Ar ticle

Figure 1. Intestinal CX3CR1+ cells pro-


tect mice from C. rodentium-induced
colitis. (A and B) Depletion efficiency in the
Cx3cr1DTR/GFP mice was assessed by flow
cytometry of small intestinal lamina propria
cells Cx3crDTR/GFP or littermate control mice
after administration of DT to both groups
daily for 2 d. (A) Surface staining for CD11b
versus CX3CR1-GFP. (B) MHCII+ CD11c+ cells
were assessed for expression of CX3CR1,
CD103, and CD14. Results are representative
of five independent experiments with a mini-
mum of three animals per group. (C) Total
number of MHCII+ CD11c+CD103+ (left) and
MHCII+ CD11c+CX3CR1+ cells (right) per intes-
tine as determined by flow cytometry analy-
sis. n.s., P > 0.05; **, P 0.01. Two-tailed
Students t test. Error bars represent the SEM.
Results are representative of five independent
experiments with a minimum of 3 animals per
group. (D) Weight of DT-treated littermate WT
(control) mice or Cx3cr1DTR/+ mice following
infection with C. rodentium (n = 7 mice/
group). DT was administered at days 2, 1,
and 0 and every other day after infection.
Data are representative of two independent
experiments. (E) Representative colonic histol-
ogy from littermate control mice or Cx3cr1DTR/+
mice (analyzed in D) infected with C. rodentium
at day 7 after infection. <, areas of lympho-
cyte infiltration; *, areas of epithelial erosion.
Bar, 100 m. (F) Survival curves of DT-treated
Cx3cr1DTR/+ and littermate control mice in-
fected with C. rodentium or uninfected (n = 7
mice/group). Data are representative of three
independent experiments. Animals were
treated with DT as in D. (G) Bacterial CFUs of
spleens from littermate WT (control, n = 5)
mice or Cx3cr1DTR/+ mice (n = 5) infected with
C. rodentium and treated with DT as above.
*, P 0.05. Two-tailed Students t test. Error
bars represent the SEM. One of two represen-
tative experiments is shown.

CD103+ CD11b+ DCs were sorted from the lamina propria express DTR only upon cre-mediated deletion of a LoxP-Stop
of Cx3cr1GFP/+ mice (Fig. 3 A) and co-cultured with intestinal cassette inserted into the Cx3cr1 locus (Diehl et al., 2013). In-
ILCs. TLR-stimulated CX3CR1+ cells were markedly more jection of DT resulted in a selective loss of Ly6Clo MHCIIhi
efficient than CD103+ CD11b+ DCs in supporting IL-22 pro- MNPs, which express both intermediate and high levels
duction (Fig. 3, B and C). CX3CR1+ cells in the LP include of CX3CR1-GFP, and spared the Ly6Chi monocytes, which
Ly6Chi monocytes and Ly6Clo MHCIIhi MNPs (Fig. 3 D; express intermediate levels of CX3CR1 (Fig. 4 A; Diehl
Zigmond et al., 2012; Diehl et al., 2013).To evaluate the role of et al., 2013). Similar to results with CX3CR1DTR/+ mice, in
these distinct CX3CR1+ populations in supporting ILC3 func- which both monocytes and MNPs were ablated, depletion of
tion, we sorted CX3CR1+ monocytes and CX3CR1+ MNPs CX3CR1+ CD11c+ cells led to reduction in colitis-induced
and co-cultured them with ILCs in the presence of TLR stim- ILC3 production of IL-22 (Fig. 4 B).
uli.TLR-stimulated MNPs were much more potent inducers of Because both IL-23 and IL-1 regulate ILC3, we wished
IL-22 than monocytes (Fig. 3, E and F). In an effort to confirm to determine the contribution of these cytokines to the ob-
the functional potential of MNPs compared with monocytes served regulation of IL-22 production by MNPs. LPS and CpG
in vivo, we selectively ablated CD11c-expressing CX3CR1+ stimulation induced markedly increased IL-23 and IL-1 pro-
MNPs. CD11c-cre mice were bred to mice engineered to duction by CX3CR1+ MNPs compared with CD103+ CD11b+

JEM Vol. 211, No. 8 1573


Figure 2. CX3CR1+ cells support colonic ILC3 pro-
duction of IL-22. (A) Survival curves of C. rodentium
infected Myd88f/f littermate controls (n = 10, open circle)
as compared with CD11c-cre/Myd88f/f mice without
(n = 13, filled circle) or with (n = 5, open triangle) exog-
enous hydrodynamic delivery of an IL-22producing
plasmid. Results are a composite of two independent
experiments. (B) Survival curves of DT-treated Cx3cr1DTR/+
mice infected with C. rodentium after hydrodynamic
delivery of a plasmid expressing IL-22 (n = 8) or control
vector (n = 9). DT was administered at days 2, 1, and 0
and every other day after infection. Results are a com-
posite of three independent experiments. (CE) Percent-
age (C and D) and total number (E) of colonic Lin
CD90.2+ ILCs producing IL-22 from DT-treated Cx-
3cr1DTR/+ (n = 10) and littermate control mice (n = 9) 7 d
after C. rodentium infection and from uninfected mice
(NT; n = 3). Results are a composite of two independent
experiments. DT was administered at days 2, 1, and 0
and every other day after infection. Intracellular IL-22 was
assayed by flow cytometry after 4-h culture. A represen-
tative flow cytometry plot from each group is shown
in C. **, P 0.01. One way ANOVA with Bonferronis
correction. Error bars represent the SEM. (F) Total
number of ILC3 per colon in Cx3cr1DTR/+ (n = 9) or control
(n = 8) mice administered DT. Error bars represent the
SEM. Results are one of three representative experiments.
(G) Total number of IL-17+ or IL-22+ colonic CD4+ T cells
from DT-treated Cx3cr1DTR/+ (n = 10) and littermate con-
trol mice (n = 9) 7 d after C. rodentium infection and
from uninfected mice (n = 3). Intracellular IL-22 and IL-17
was assayed by flow cytometry after 4-h culture.
*, P 0.05. One way ANOVA with Bonferronis correction.
Error bars represent the SEM. Results are a composite of
two independent experiments. (H) Confocal immuno-
fluorescence of colonic samples from Cx3Cr1GFP/+ mice
stained for CD3 and RORt. Bar, 10 m. White arrows
indicate sites of MNP and ILC3 juxtaposition.

DCs in vitro (Fig. 4, C and D). To evaluate the role of MNP- (CD, n = 8) or ulcerative colitis (UC, n = 6;Table S1) as well as
derived IL-23 and IL-1, co-cultures were performed with in- age-matched non-IBD control patients undergoing routine
testinal CD11c+ cells derived from WT or Il23p19/ mice and screening colonoscopy (n = 8). Analysis of intracellular cytokine
ILCs from WT or Il1r/ mice. IL-23deficient MNPs and production revealed significantly increased IL-22 production
IL1R-deficient ILCs yielded significantly reduced production in the CD3 fraction of colonic LPMCs from sites of colonic
of IL-22 (Fig. 4 E), consistent with the importance of MNP- inflammation in both CD and UC compared with the non-IBD
derived IL-1 and IL-23 in supporting IL-22 production. controls (Fig. 5 A and Fig. S2). In contrast, IL-22 production
These data reveal a mechanistic role for IL-23 and IL-1 pro- by T cells was not significantly different between the groups.
duced by colonic MNPs in supporting colitis-associated ILC3 Further characterization of the nonT cells producing IL-22
secretion of IL-22. revealed that a large fraction of these cells expressed c-Kit and
CD56, markers of ILCs (Cella et al., 2009; Fig. 5 B). Consis-
Human intestinal ILC3 production of IL-22 tent with their being ILC3, these cells were lineage negative
is regulated by microbial stimulation of MNPs (Lin; Fig. S3 A); expressed RORt (Fig. 5 C); were CD45int
To evaluate the regulation of intestinal ILC3 in humans with CD127+ (Fig. 5 D); expressed CD161, NKp44, and CCR6
IBD, we prepared LPMCs from descending colon biopsies of (Fig. 5 E), which are phenotypic surface markers of ILC3; and
patients with endoscopically mild to moderate Crohns disease produced IL-22 in response to IL-23 stimulation (Fig. 5 F). As
1574 CX3CR1+ MNPs regulate ILC3 | Longman et al.
Ar ticle

Figure 3. TLR-stimulated CX3CR1+ MNPs


are stronger inducers of ILC3 production
of IL-22 than CD103+ CD11b+ DCs and
monocytes. (AC) CD103 or CX3CR1+ MHCII+
CD11c+ CD11b+ cells were isolated from the
lamina propria of CX3CR1GFP/+ mice (sort strat-
egy shown in A and co-cultured with Lin
RORt-GFP+ ILCs with or without the indi-
cated bacterial TLR ligands or IL-23. IL-22 was
assessed by intracellular staining of CD90.2+
ILCs after 18 h. A representative flow cytom-
etry plot is shown in B. (C) Percent IL-22+ ILCs
is shown from seven independent experi-
ments. **, P 0.01; *, P 0.05. One way
ANOVA with Bonferronis correction. Error
bars represent SEM. (DF) Ly6C+ MHCIIlo
(monocytes) and Ly6C MHCIIhi (MNPs) were
isolated from CX3CR1+ CD11b+ lamina propria
cells (sort strategy is shown in D and co-
cultured with intestinal ILCs with LPS or IL-23
as indicated. Intracellular cytokine staining for
IL-22 is shown after 18 h (E). Supernatants
were harvested after 18 h and IL-22 produc-
tion quantitated by ELISA (F). Results are rep-
resentative of two independent experiments
performed in triplicate. ***, P 0.001. One-
way ANOVA with Bonferroni correction. Error
bars represent the SEM.

Myd88 deficiency abrogated ILC3 production of IL-22, we Parallel subpopulations of CD11c+ MNPs present in the
hypothesized that signals from the microbiota could induce mouse intestine similarly exist in the human intestine (Fig. 6 A;
ILC3 to produce IL-22. To investigate this in human tissue, we Merad et al., 2013). To evaluate whether these distinct sub-
evaluated three patients who had a surgical diversion of the populations of MNPs from human intestinal tissue functioned
fecal stream (i.e., a diverting ostomy) as part of their therapy similarly, we examined the phenotypic properties of CD103+
for IBD. Endoscopic biopsies were taken from a site proximal DCs and CD14+ MNPs (which express CX3CR1; Kamada
to the diversion (afferent limb), where the mucosa was exposed et al., 2008) within the CD11c+ MHCII+ fraction of LPMCs
to intestinal microbiota in the fecal stream, and from mucosa (Fig. 6 B). In contrast to CD103+ DCs, CD14+ MNPs ex-
distal to the diversion (efferent limb), that was unexposed to pressed CD64 as well as higher levels of CD86. Consistent with
the fecal stream. ILCs were present at both mucosal locations, the phenotypic characterization of these subsets, transcriptional
but not in PBMCs from the same donor (Fig. S3 B). In all analysis of these populations by RNA-seq revealed higher
three donors, ILCs from tissue exposed to bacteria in the fecal levels of CLEC9A, XCR1, and CD207 expression in the
stream produced substantially more IL-22 compared with CD103+ cells, whereas MERTK, STAB1, and CX3CR1 were
ILCs from unexposed tissue (postdiversion; Fig. 5 G). higher in the CD14+ cells (Fig. 6 C). We tested the potential

JEM Vol. 211, No. 8 1575


Figure 4. CX3CR1+ MNP-derived IL-23 and IL-1 activate ILC3 to produce IL-22. (A) Phenotype analysis of colonic LPMCs from Cx3cr1STOP-DTR/GFP
mice with or without CD11c-cre after DT injection for 2 d. (top) Selective depletion of CX3CR1hi MNPs. (bottom) Expression of Ly6C and MHCII on
CX3CR1hi and CX3CR1int populations. (B) Expression of IL-22 in Lin CD90.2+ colonic ILCs from Cx3cr1STOP-DTR/+ (Stop-DTR) or CD11c-Cre x Cx3cr1STOP-DTR/+
(Cre-DTR) mice at 7 d after C. rodentium infection. DT was administered at days 2, 1, and 0 and every other day postinfection. One representative
intracellular cytokine flow cytometry plot is shown on the left and a composite graph (n = 6/group) on the right. *, P 0.05, two-tailed Students t test.
Error bars represent the SEM. Results are a composite of two independent experiments. (C) Supernatants from APC-ILC co-cultures (Fig. 3, B and C) were
harvested after 18 h and assayed for IL-23 by ELISA. Results are averages of three independent experiments and the SEM is shown. (D) CX3CR1+ MNPs or
CD103+ CD11b+ DCs were sorted and incubated with media or CpG for 18 h and supernatants were assayed for IL-1 by ELISA. Results are the mean of
two independent experiments performed in duplicate and the SEM is shown. *, P 0.05; **, P 0.01. (E) Lin CD90.2hi ILCs from WT or Il1r/ mice were
co-cultured with sorted intestinal MNPs from WT or Il23p19/ mice, with or without CpG, as indicated. IL-22 production by the ILCs was assessed after
18 h by ELISA. Data are combined from three independent experiments performed in duplicate. *, P 0.05; ***, P 0.001. One-way ANOVA with Bonferroni
correction. Error bars represent the SEM.

of these subsets to induce IL-22 production by co-culturing the expanded population of CD14+ MNPs (Kamada et al.,
TLR-stimulated CD14+ MNPs and CD103+ DCs from human 2008) in supporting colitis-associated IL-22 production by
intestinal resections with intestinal ILCs. Intracellular cyto- ILC3, through their secretion of IL-23 and IL-1.
kine staining at 18 h revealed that the CD14+ MNP were more
effective than the CD103+ DCs at stimulating IL-22 production CX3CR1+ MNP-derived TL1A synergizes
by ILCs (Fig. 6 D). Neither cell population induced signifi- with IL-23 and IL-1 to induce IL-22
cant IL-17 or IFN- production by ILCs (Fig. 6, D and E). We hypothesized that MNP-derived factors in addition to
Consistent with the importance of IL-23 and IL-1 in IL-23 and IL-1 would contribute to the regulation of ILC3
the mouse co-culture experiments, a higher level of IL23A function. The RNA-seq data from CD14+ human MNPs re-
expression was observed by RNA-seq in CD14+ MNPs com- vealed a significant enrichment for genes associated with IBD in
pared with CD103+ DCs (Fig. 6 C). Stimulation of these human GWAS studies, suggesting that IBD-associated pathways are im
MNP subsets with LPS or flagellin revealed increased IL-23p19 portant in these cells (Fig. 7 A and Table S2). Notably, we iden
mRNA and IL-1 protein produced by the CD14+ cells com- tified TNF-like ligand 1A (TL1A, also designated TNFSF15)
pared with the CD103+ cells (Fig. 6 F). In accord with this as a significant contributor to the IBD GWAS-derived gene
finding, in co-cultures of human intestinal ILC3 and CD14+ set enrichment. Evaluation of Tnfsf15 transcript in sorted
MNPs, IL-23 and IL-1 antibody blockade blunted IL-22 mouse colonic APC subsets by qPCR confirmed higher ex-
production (Fig. 6 G).These data reveal a mechanistic role for pression in CX3CR1+ MNPs (Fig. 7 B). DR3/TNFRSF25,

1576 CX3CR1+ MNPs regulate ILC3 | Longman et al.


Ar ticle

Figure 5. Increased ILC3 production of IL-22 in mild to moderate IBD correlates with presence of fecal stream. (A) LPMCs isolated from de-
scending colon biopsies from patients with endoscopically mild to moderate Crohns disease (n = 8, gray) or ulcerative colitis (n = 6, black; Table S1),
as well as age-matched non-IBD control patients undergoing routine screening colonoscopy (n = 8, white), were stimulated ex vivo with PMA/ionomycin
and evaluated by intracellular cytokine staining for expression of IL-17 and IL-22. The percentage of CD3+ or CD3 fraction expressing IL-17 or IL-22 is
indicated. *, P 0.05, two-tailed Students t test. Black bars represent the geometric mean. (B) Expression of c-Kit and CD56 in electronically gated CD3
IL-22+ (black lines) and CD3 IL-22 (gray) LPMCs. (C) Expression of RORt by c-Kit+CD56+ LPMCs. Lin cells (CD14/CD19/CD3/CD11b/CD11c/TCR;
Fig. S3 A) were stained with antibodies to surface markers c-Kit and CD56 and to intracellular RORt. Lin CD56+ c-Kit+ ILC3 (black line) were compared
with Lin CD56+ c-Kit NK cells (gray) for RORt expression. (D) Surface staining of Lin c-Kit+ ILCs for the indicated markers (black line) compared with
isotype control (gray) and all live LPMCs (dotted line) (E) LPMCs stained for intracellular IL-22 after stimulation with IL-23 for 3 h (solid line) or with con-
trol media (dotted line). Cells shown were gated on Lin CD56+ c-Kit+. The isotype control is in gray. (F) CD11c+ MHCII+ human colonic APCs were elec-
tronically gated for expression of CD103 and CD14. One of three donors is shown. (G) Lamina propria cells from biopsy samples of tissue exposed
(prediversion) or not exposed to the fecal stream (post-diversion) were cultured for 3 h and ILC3 production of IL-22 was assessed by flow cytometry.
(left) Result from one representative donor. (right) Percentage of IL-22+ ILCs in afferent (Pre) and efferent (Post) limbs of three diverted patients.
**, P 0.01, two-tailed Students t test. Black bars represent the geometric mean.

the receptor for TL1A, is expressed both on T cells and ILC with LPS-stimulated CX3CR1+ MNPs. These data reveal a
subsets, with the highest expression on ILC2 and ILC3 (Meylan potent ability of TL1A to enhance IL-22 production via DR3/
et al., 2014). Ex vivo stimulation with mouse or human re- TNFRSF25 on ILC3 in both mouse and human.
combinant TL1A significantly enhanced the ability of IL-23
and IL-1 to induce IL-22 by both mouse (Fig. 7 C and Fig. S4) DISCUSSION
and human (Fig. 7 D) intestinal ILC3, respectively. TL1A and Mononuclear phagocytes are spatially and functionally poised
IL-1 also cooperated in inducing GM-CSF production by to integrate microbial signals from the luminal microbiota
mouse ILC3 (Fig. 7 E). To assess the specificity of TL1A for (Niess et al., 2005; Varol et al., 2010). Although MNPs were
DR3/TNFRSF25 on ILCs and its functional role in enhancing previously thought to remain in the tissue, we recently showed
CX3CR1+ support of ILC3 activation, DR3 expression was that CX3CR1+ MNPs can migrate to draining lymph nodes
specifically knocked down using siRNA nucleofection of sorted and initiate immune responses under conditions of dysbiosis
mouse intestinal ILC3. Efficiency of knock-down was con- (Diehl et al., 2013). In the context of inflammation, however,
firmed by surface staining for DR3, comparing with a scram- these CX3CR1+ MNPs expand within the lamina propria dur-
bled control siRNA (Fig. 7 F). Compared with the scramble ing chemical (Zigmond et al., 2012) or infectious colitis and
control, cells targeted with siRNA for Tnfrsf25 had signifi- in IBD patients (Kamada et al., 2008), and their function has
cantly reduced enhancement of IL-22 production upon treat- remained obscure. Conventional DCs (cDCs), rather than the
ment with recombinant TL1A (Fig. 7, G and H).The targeted MNPs, have been postulated to regulate intestinal Th17 cell
ILCs also produced reduced amounts of IL-22 upon co-culture differentiation in response to microbiota (Denning et al., 2011;

JEM Vol. 211, No. 8 1577


Figure 6. Human ILC3 production of IL-22 is supported by IL-23 and IL-1 produced by TLR-stimulated CD14+ and CX3CR1+ MNPs.
(AC) HLA-DR+ CD11c+ cells from intestinal resection tissue were sorted into CD103+ DCs and CD14+ MNPs subpopulations and transcriptional profiles
were assessed by RNA-seq. (A) Sorting strategy. (B) Each subset was examined for expression of the indicated cell surface markers. Isotype controls are
shown in gray. One of three donors is shown. (C) Heatmap of relative expression of relevant MNP-related genes. Values represent the mean of two inde-
pendent donors, and an asterisk denotes individual genes differentially expressed at an FDR = 0.01. (D and E) Induction of IL-22 in human ILCs in co-
culture with CD14+ MNPs or CD103+ DCs in the presence of media alone, LPS, or flagellin, as indicated. c-Kit+ cells were examined for intracellular IL-22
production after 18-h culture. Data are representative of five independent experiments. (F) CD14+ MNPs or CD103+ DCs sorted from human intestine (as
in A) were stimulated with the indicated TLR ligands for 18 h and qPCR or cytometric bead array analysis were used to quantitate IL-23p19 and IL-1,
respectively. Results are averaged from three independent donors, and technical replicates were performed in duplicate or triplicate, respectively. *, P 0.05.
N.D., not detected. Two-tailed Students t test. Error bars represent the SEM. (G) Sorted human intestinal CD14+ MNPs and ILCs were left unstimulated or
were co-cultured in the presence of LPS with or without neutralizing antibodies against IL-1 and IL-23. IL-22 ELISA was performed after 18h. Results
are averaged from two independent donors performed in duplicate. *, P 0.05. Two-tailed Students t test. Error bars represent the SEM.

Lewis et al., 2011; Persson et al., 2013; Schlitzer et al., 2013; of multiple myeloid subsets in the immune response to C. ro-
Goto et al., 2014), but there has been conflicting evidence as dentium (Schreiber et al., 2013), and our data do not exclude a
to which myeloid cell populations regulate IL-22 produc- contribution of other myeloid subsets. Alternatively, Notch2
tion by ILC3 cells (Kinnebrew et al., 2012; Manta et al., 2013; may affect cellular subpopulations in addition to CD103+
Satpathy et al., 2013). CD11b+ cDCs. This latter hypothesis is supported by the re-
Our data reveal that colonic CX3CR1+ MNPs regulate port that an alternate selective depletion strategy for CD103+
ILC3 production of IL-22 and likely play a critical role in pro- CD11b+ cDCs, by expression of DT under the human Lan-
moting mucosal healing during colitis. The requirement for gerin promoter (Welty et al., 2013), did not result in increased
CX3CR1+ MNPs in regulating colitis-induced ILC3 shown susceptibility to C. rodentium. Some of the conflicting data
here appears to contrast with recent work suggesting that may additionally reflect differential roles of inflammatory mono-
Notch2-dependent CD103+ CD11b+ cDCs are required for cytes and MNPs in colitis. Inflammatory monocytes may ex-
the control of C. rodentiuminduced colitis (Satpathy et al., 2013). acerbate pathology, as antibody-mediated depletion of Ly6Chi
These disparate results may reflect a coordinated contribution monocytes during DSS treatment improved pathology in DSS

1578 CX3CR1+ MNPs regulate ILC3 | Longman et al.


Ar ticle

Figure 7. CX3CR1+ MNP-derived TL1A synergizes with IL-23 and IL-1 to induce IL-22 in intestinal ILC3. (A) Gene-set enrichment analysis
was used to determine whether the indicated disease-related SNP were differentially expressed between CD14+ MNPs and CD103+ DCs. Significance was
estimated using the hypergeometric cumulative distribution, with a raw p-value cutoff of 0.05 for differential expression. Data were averaged from two
independent donors. (B) B cells (CD3 CD19+), CX3CR1+ MNPs, CD103+ DCs, and Ly6C+ monocytes were sorted from the intestinal lamina propria of
Cx3cr1GFP/+ and quantitative PCR for TL1A was performed. Relative quantitation was performed by Ct normalized to GAPDH expression. Data are from
two biological replicates performed with two technical replicates. *, P 0.05. Two-tailed Students t test. (CE) Sorted intestinal ILCs from mice (C and E)
or cultured human intestinal ILCs (D) were stimulated with media alone, IL-1, or IL-23 with or without TL1A as indicated for 18 h. Brefeldin was added
to the cultures 4 h before intracellular cytokine staining for IL-22 (C and D) or GM-CSF (E). Data are representative of six independent experiments.
(FH) Sorted intestinal ILCs were transfected with siRNA targeting Tnfrsf25 or a scramble control. (F) Knockdown efficiency was assessed after 24 h by
flow cytometry comparing scramble control (solid line) with Tnfrsf25 siRNA. One of two representative experiments is shown. ILCs were then cultured
with media alone (-) or IL-23 and TL1A or co-cultured with CX3CR1+ MNPs with or without LPS as indicated for an additional 18 h. (G) IL-22 production
was measured by intracellular flow cytometry. Brefeldin was added to the cultures 4 h before intracellular cytokine staining. Data are representative
of two independent experiments. (H) IL-22 secretion by samples from G were assessed by ELISA, performed in duplicate, before addition of Brefeldin.
*, P 0.05; **, P 0.01. Two-tailed Students t test. Error bars represent SEM.

colitis (Zigmond et al., 2012). Selective depletion of such of IL-23 and IL-1. At steady state, commensal-dependent sig-
monocytes may hence explain the intermediate results seen naling may negatively regulate ILC production of IL-22 via in-
in the CCR2-deficient mice exposed to C. rodentium (Satpathy testinal epithelial cell production of IL-25 (Sawa et al., 2011),
et al., 2013). In addition to supporting ILC3, MNPs may also but we and others (Satoh-Takayama et al., 2008) find that
directly promote epithelial barrier repair (Wynn et al., 2013). microbes support colitis-associated ILC production of IL-22,
Although our data suggest a critical role for CX3CR1+ MNPs which promotes mucosal healing. Although intravenous de-
in regulating production of IL-22 by ILC3 during acute coli- livery of flagellin can induce CD103+ CD11b+ cDCs to produce
tis, it will be important to examine the role for these myeloid IL-23 that regulates ILC3 in the small intestine (Kinnebrew
subsets during chronic colitis and their impact on another major et al., 2012), we and others (Kamada et al., 2008) found that
clinical endpoint in IBDtumorigenesis (Huber et al., 2012; in vivo colonic inflammation and other bacterial-derived signals
Kirchberger et al., 2013). induced more robust production of IL-23 and IL-1 by MNPs,
Our results also highlight a beneficial role for microbial sig- suggesting that the type of stimulation and/or colonic inflam-
nals in promoting intestinal homeostasis and driving ILC3 pro- mation may confer specificity. Similar to our results in mice,
duction of IL-22 through TLR/MyD88-dependent induction we found marked reductions in IL-22 production by intestinal

JEM Vol. 211, No. 8 1579


ILC3 from human tissue distal to a surgical diversion of the (Shih et al., 2009).The data shown here reveal a potent ability
fecal stream.These findings may offer clinically relevant mech- of TL1A to enhance ILC3 production of IL-22 and suggest
anistic insight into diverted IBD patients with persistent in- that the increased expression of TL1A by colonic CX3CR1+
flammatory disease or even non-IBD patients with de novo MNPs enables their selective ability to potently support ILC3.
mucosal inflammation after diversion (e.g., diversion colitis; Furthermore, the ability of TL1A to induce GM-CSF pro-
Harig et al., 1989). Although some evidence supports a role duction by ILC3 is consistent with recent data suggesting
for luminal replacement of short chain fatty acids in diversion that, even at steady state, CSF1R-dependent MNPs support
colitis (Harig et al., 1989;Vernia et al., 1995), further work is ILC3 production of GM-CSF that, in turn, regulates oral tol-
required to determine if particular metabolites or intestinal erance (Mortha et al., 2014). Collectively, these data support a
microbes may regulate ILC3 function to promote healing. broader role for colonic CX3CR1+ MNPs in regulating mu-
In light of their robust production of IL-22 and close cosal homeostasis.
proximity to the intestinal epithelial layer (Cella et al., 2009), Mucosal ILC3 play a crucial role in barrier homeostasis,
ILC3 have been proposed to play an important role in muco- mucosal healing, and oral tolerance. CX3CR1+ MNPs are aptly
sal healing and maintenance of barrier integrity, but their char- poised to integrate microbial signals to regulate ILC3 and
acterization in IBD patients has been limited. ILCs producing may serve as important targets for therapeutic manipulation.
IL-17 or IFN- (classified as ILC1) have also been described in Further understanding of the contributions of discrete commen-
mouse models of colitis (Buonocore et al., 2010; Klose et al., sal bacterial species and of the mechanistic interactions be-
2013) and human IBD (Geremia et al., 2011; Bernink et al., tween CX3CR1+ MNPs and ILC3 may provide novel strategies
2013), supporting a potentially important role for ILCs in IBD to promote intestinal healing. In light of the specific and po-
pathogenesis. Here, we observed a specific increase in IL-22 tent regulation of ILC3 by colonic CX3CR1+ MNPs during
production by ILCs from colonic biopsies of patients with mild colitis, diagnostics that correlate susceptible genotypes (Hueber
to moderate CD and UC. Despite characteristically distinct et al., 2012) with clinical disease immunophenotype will pro-
phenotypes, both CD and UC share genetic susceptibility risk vide insight into therapeutic strategies to manipulate colonic
alleles within the IL-23 pathway (Duerr et al., 2006), which MNPs in the clinical management of IBD.
may underlie a common role for ILCs during colonic inflam-
mation in both diseases. In contrast to previous studies, we did MATERIALS AND METHODS
not observe any significant IL-17 production by CD3 cells Antibodies and flow cytometry. Staining of human cells was performed
from patients with CD (Geremia et al., 2011) or a reduction with c-Kit-e450 (104D2), CD56-PECy5.5 (CMSSB), HLA-DR-PE (LN3),
CD11c-FITC (3.9), CD3-e780 (UCHT1), CD19-PerCP5.5 (HIB19), CD103
in ILC3 in CD patients (Bernink et al., 2013). This difference
PECy7 (B-Ly7), CD14 PE (61D3), CD86-PE (IT2.2), CD123 PE (6H6),
may reflect clinical differences in the patient cohorts and CD11b PE (CBRM1/5), and RORt (AFKJS-9) obtained from eBiosci-
samplescolonoscopic biopsies in our cohort were taken from ence; CD161-FITC, CD83-PE, CD11c PE (555392), CD127 FITC (M21),
ambulatory patients with mild to moderate colonic CD and UC, CCR6-biotin (11A9) were purchased from BD; BDCA-1-APC were ob-
in contrast to surgical resection of ileal tissue from medically tained from Miltenyi Biotec; CD45-APC (4505) and CD64 (6404) were ob-
refractory patients in the other studies. As such, mild to mod- tained from Invitrogen; NKp44-APC (p44-8.1) was purchased from R&D
Systems; and BDCA-3 APC (M80) was purchased from BioLegend. Intra
erate colitis may be the ideal human model to evaluate ILC3-
cellular human cytokine staining was performed with IL-22 PE (IC7821P;
producing IL-22 promoting mucosal healing. The association R&D Systems), IL-17A-FITC (eBio64CAP17; eBioscience), or IFN- (45.
of clinical phenotype with ILC phenotype and the potential B3; eBioscience). Staining of mouse cells was performed with CD90.2-e450
plasticity of these ILCs in colitis remain to be evaluated. (532.1), CD3e (145-2C11), NKp46-e710 (29A1.4), RORt-PE (B2D),
Consistent with our results in mouse models, CX3CR1+ CD11c-PECy7 (N418), MHCII A700 (M5/114.15.2), CD103 APC (2E7),
MNPs from human intestinal tissue were more potent than CD14 PerCP5.5 (Sa2-8), CD11b e780 (M1/70), Ly6C PerCP5.5 (AL-21),
and F4/80 PE (BM8) were from eBioscience. DR3-PE (4C12) was obtained
CD103+ DCs in supporting human intestinal ILC3 produc-
from BioLegend. Intracellular mouse cytokine staining was performed with
tion of IL-22. In addition to the increased production of IL-23 IL-22 APC (IL22JOP), IFN- PECy7 (XMG1.2), IL17A FITC (eBio17B7),
and IL-1 by the CX3CR1+ subset described by us and oth- all from eBioscience. Intracellular cytokine staining was performed according
ers (Kamada et al., 2008), RNA seq analysis of these subsets to the manufacturers protocol (Cytofix/Cytoperm buffer set; BD). Intra
revealed enrichment for transcripts from genes having IBD- nuclear staining for RORt was performed according to manufacturers protocol
associated SNPs, notably TL1A/TNFSF15. Polymorphisms (Intracellular Fixation and Permeabilization kit; eBioscience). Flow cytome-
try and analysis were performed with a LSR II (BD) and FlowJo software
in TNFSF15, the gene that encodes TL1A, are strongly asso-
(Tree Star). Dead cells were excluded using the Live/Dead fixable aqua dead
ciated with IBD, and expression of both TL1A and its recep- cell stain kit (Invitrogen).
tors DR3 and DcDR3 is elevated in IBD patients (Kugathasan
et al., 2008). TL1A has been shown to enhance Th17 differen- Mice. C57BL/6, Myd88flox, CD11c-cre (Caton et al., 2007), and Il1r/ mice
tiation and effector function (Pappu et al., 2008; Kamada et al., were purchased from The Jackson Laboratory. Myd88-deficient mice were
2010); promote Treg expansion and ameliorate allergic asthma obtained from S. Akira (Osaka University, Osaka, Japan; Adachi et al., 1998)
and Il23p19/ mice were obtained from D. Cua (Merck Research Labora-
(Schreiber et al., 2010); and promote IL-13 production by
tories, Palo Alto, CA; Cua et al., 2003). RORc(t)GFP/+ (Eberl et al., 2004),
ILC2s (Meylan et al., 2014). Interestingly, microbial stimula- Cx3cr1GFP/+ (Jung et al., 2000) and inducible CX3CR1-DTR mice (Diehl et al.,
tion and Fc-receptor engagement induces TL1A/TNFSF15 2013), were previously described. The latter mice were subsequently bred to
in monocytes and monocyte-derived mononuclear phagocytes a germline-expressing cre mouse to excise the transcriptional stop sequence

1580 CX3CR1+ MNPs regulate ILC3 | Longman et al.


Ar ticle

and produce the Cx3cr1DTR/+ mice. All mice were kept in specific pathogen delivered i.v. at 5 g DNA/mouse diluted in TransIT-EE Hydrodynamic Deliv-
free (SPF) conditions at the animal facility of the Skirball Institute. Mouse ery Solution (Mirus) at 0.1 ml/g body weight. Immune cell functional analysis
experiments were performed with at least three mice per group and multiple and histology were performed at day 7 after exposure. Spleens were harvested on
experiments were combined to assess statistically significant differences as day 21, homogenized, and plated at serial dilutions to determine CFU/spleen.
noted. Littermates of the same genotype were randomly assigned to experi-
mental groups. Animals were used between 8 and 16 wk of age. Males and RNA-seq processing and gene set enrichment analysis. Sequence reads
females were used in approximately equal ratios. All animal experiments were were mapped to the human genome (version hg19) by Tophat (version 2.0.6),
performed in accordance with approved protocols for the NYU Institutional using Bowtie2 (version 2.0.2) and Samtools (version 0.1.18). Reads are depos-
Animal Care and Usage Committee. ited at Bioproject PRJNA219394. Reads mapped per transcript served as input
to DESeq (version 1.12.0), an R package that calculates differential gene ex-
Preparation of LPMCs. Endoscopic biopsies were obtained under IRB- pression. To improve detection of APC lineage-dependent gene-expression
approved protocols at Weill Cornell Medical College (1103011578 and Co- changes and overcome donor-dependent variability, we used the following
lumbia University Medical Center (AAAE5448) including patients >18 yr of strategy: independently for each donor, differential gene expression, comparing
age and able to give informed consent. IBD sample was defined based on en- CD103+ to CD14+ human myeloid cells, was estimated using the negative bi-
doscopic inflammation with history of ulcerative colitis or Crohns disease. nomial distribution (nbinomTest), and then results from biological replicates
Endoscopic score (mild, moderate, or severe) was based on Mayo Endoscopic were combined using Fishers method. Several gene set enrichment techniques
subscore or SES-CD (1, 2, 3, respectively) at the site of biopsy (Pineton de (e.g., hypergeometric test and area under precision-recall curve) were used to
Chambrun et al., 2010). All endoscopic biopsies were taken from the sigmoid test whether specific gene sets (Table S2) were significantly differentially regu-
or descending colon to reduce sampling variation. Study sample sizes for lated between the two lineages.The GWAS gene sets are derived from disease-
human biopsies were based on preliminary data and powered to achieve sta- associated SNPs from the NHGRI GWAS Catalog. False-discovery rates
tistically significant differences in the production of IL-22 or IL-17. Surgical (FDRs) were calculated using the Benjamini-Hochberg procedure. The en-
resections were obtained under an IRB-approved protocol at New York Uni- richment analyses were implemented in Matlab R2013a (8.1.0.604).
versity Langone Medical Center. Mouse and human intestines were washed
in PBS and 1 mM DTT twice with 30 mM EDTA, and then digested in col- qPCR. RNA from primary intestinal APCs stimulated as indicated was pre-
lagenase 8 (Sigma-Aldrich) and DNase-containing media with 10% fetal bo- pared with TRIzol (Invitrogen). RNA was reverse transcribed into cDNA
vine serum. Digested material was passed through a cell strainer and separated (SuperScript III; Invitrogen) and QPCR was performed with a Roche Light-
on a discontinuous 40%/80% Percoll gradient. LPMCs were cultured ex vivo Cycler with SYBR Green Supermix (Bio-Rad Laboratories), 20 pmol forward
in the presence of GolgiPlug (BD) for 4 h or stimulated with phorbol my- and reverse primers, and 0.1 g of cDNA from 5-TGTTCCCCATATC-
ristate acetate (PMA; 20 ng/ml) and ionomycin (1 g/ml) or IL-23 (40 ng/ml; CAGTGTGG-3 and 5-CTGGAGGCTGCGAAGGATTT-3 for human
eBioscience) in the presence of GolgiPlug (BD) for 4 h before staining. IL23p19, 5-ATGCTTCGGGCCATAACAGA-3 and 5-TGAAGGC-
CATCCCTAGGTCA-3 for mouse TL1A; 5-ACCACAGTCCATG
Intestinal ILC cultures. Surgical resections were obtained from the NYU CCATCAC-3 and 5-TCCACCACCCTGTTGCTGTA-3 for human
Biorepository (Rachel Brody). Lin c-Kit+ CD45int ILCs were sorted and GAPDH; and 5-AATGTGTCCGTCGTGGATCT-3 and 5-CATCGA
cultured in tissue culture media (RPMI 1640; Invitrogen) supplemented with AGGTGGAAGAGTGG-3 for mouse GAPDH. The thermocycling pro-
10% (vol/vol) heat-inactivated FBS (HyClone), 50 U penicillin-streptomycin gram was 40 cycles at 95C for 15 s, 60C for 30 s, and 72C for 30 s, with
(Invitrogen), 2 mM glutamine, and 50 M -mercaptoethanol), supplemented an initial cycle of 95C for 2 min. Relative levels of target gene were deter-
with IL-7 (50 ng/ml; PeproTech) and IL-2 (1,000 U/ml; PeproTech) for 810 d mined by using the delta Ct value compared with delta Ct (GAPDH).
before stimulation. Lin, CD90.2+, RORt-GFP mouse ILC3 were sorted
Immunofluorescence. Intestinal tissue was Swiss-rolled before fixing for 4 h
from LPMCs and resuspended in RPMI-based tissue culture media for stim-
in 4% paraformaldehyde. Tissue was incubated overnight in 30% sucrose be-
ulation directly ex vivo. Human and mouse ILCs were stimulated with human
fore freezing in OCT. Tissue was cut into 5-M sections. Tissue was blocked
or mouse IL-23 (eBioscience; 40 ng/ml), IL-1 (eBioscience; 10 ng/ml), or
in PBS-XG (0.1% Triton X-100, 10% goat serum) before incubating over-
TL1A (R&D Systems; 200 ng/ml) as indicated, respectively. After 18 h, super-
night in primary antibody in PBS-XG. Tissue was washed and then incu-
natants were harvested for IL-22 ELISA (eBioscience) and Golgi Plug (BD)
bated with secondary antibody for 1 h before DAPI staining. The following
was added to cells for 4 h for subsequent intracellular cytokine staining.
primary antibodies are from eBioscience: antihuman/mouse RORt (clone
AFKJS-9) and antimouse CD3e (clone 145-2C11). Secondary antibodies
Co-culture assay. ILCs and APC populations were sorted on a FACSAria
are from Jackson ImmunoResearch Laboratories (Cy3-AffiniPure goat antirat
and co-cultured with 5 103 and 2.5 103 cells, respectively, in 96-well
IgG and goat antiArmenian hamster). Tissue was imaged using an LSM 710
round-bottom plates in tissue culture media.TLR stimulation was performed
confocal (Carl Zeiss) and images were processed using ImageJ.
with 1 g/ml LPS (Escherichia coli; Sigma-Aldrich), 1 M CpG 1668 (mouse),
1 M CpG 2216 (human), or 1 g/ml flagellin (Salmonella Typhi; InvivoGen).
Online supplemental material. Fig. S1 shows the gating strategy for co-
Cultures were incubated for 18 h. Supernatants were harvested for ELISA
lonic ILC3. Fig. S2 shows increased production of IL-22 in ILCs from pa-
and remaining cells were incubated with Golgi Plug (BD) for 4 h and sub
tients with IBD. Fig. S3 shows Lin gating and surface phenotype for human
sequently analyzed by flow cytometry.
intestinal ILC3. Fig. S4 shows that TL1A enhances IL-23 and IL-1 induc-
tion of IL-22 by intestinal ILC3s. Online supplemental material is available at
siRNA transfection. Sorted intestinal ILCs were cultured overnight in IL-7
http://www.jem.org/cgi/content/full/jem.20140678/DC1.
(20 ng/ml) and SCF (20 ng/ml). After 24 h, 4 105 ILCs were transfected
using AMAXA T cell nucleofection protocol. 300 pmol of Tnfrsf25 siRNA
We thank Rachel Brody (NYU Biorepository), Fatiha Chabouni, Priyanka Patel, Ryan
pool or scramble control (Thermo Fisher Scientific) was used per transfec- Warren and members of The Roberts Center for IBD for help with sample collection
tion. Cells were rested overnight and harvested at 24 h for experimental use. as well as the patients that participated in this study. We would like to thank
Knockdown efficiency was assessed at 24 h by DR3 surface staining. Michael Cammer for assistance with microscopy.
This work was supported by the American Gastroenterological Association
Colitis models. C. rodentium DBS100 (ATCC 51459; American Type Culture Research Foundation (R.S. Longman), National Institutes of Health K08 DK099381
Collection) was harvested at log phase growth and 1010 CFU were delivered by (R.S. Longman), American Cancer Society (G.E. Diehl), NIH T32 CA009161 (G.E. Diehl),
gavage in PBS. 200 ng of DT was administered i.p. as indicated for depletion. K99DK091508 (J.R. Huh), and the Howard Hughes Medical Institute (D.R. Littman).
Plasmid DNA expressing IL-22 or control plasmid (Qiu et al., 2012) were The authors declare no competing financial interests.

JEM Vol. 211, No. 8 1581


Author contributions: R.S. Longman and G.E. Diehl designed and performed the Goto, Y., C. Panea, G. Nakato, A. Cebula, C. Lee, M.G. Diez, T.M. Laufer,
experiments. R.S. Longman, G.E. Diehl and D.R. Littman planned experiments and L. Ignatowicz, and I.I. Ivanov. 2014. Segmented filamentous bacte-
wrote the manuscript with input from the co-authors. J.R. Huh, C. Galan, and ria antigens presented by intestinal dendritic cells drive mucosal Th17
D.A. Victorio helped plan and perform experiments. E.R. Miraldi and R. Bonneau cell differentiation. Immunity. 40:594607. http://dx.doi.org/10.1016/
performed data analysis. A. Swaminath and E.J. Scherl helped with sample collection j.immuni.2014.03.005
and experimental design. Hadis, U., B. Wahl, O. Schulz, M. Hardtke-Wolenski, A. Schippers, N. Wagner,
W. Mller, T. Sparwasser, R. Frster, and O. Pabst. 2011. Intestinal toler-
Submitted: 9 April 2014 ance requires gut homing and expansion of FoxP3+ regulatory T cells in
Accepted: 18 June 2014 the lamina propria. Immunity. 34:237246. http://dx.doi.org/10.1016/
j.immuni.2011.01.016
REFERENCES Hanash, A.M., J.A. Dudakov, G. Hua, M.H. OConnor, L.F. Young, N.V.
Adachi, O., T. Kawai, K. Takeda, M. Matsumoto, H. Tsutsui, M. Sakagami, K. Singer, M.L. West, R.R. Jenq, A.M. Holland, L.W. Kappel, et al. 2012.
Nakanishi, and S. Akira. 1998. Targeted disruption of the MyD88 gene Interleukin-22 protects intestinal stem cells from immune-mediated tis-
results in loss of IL-1- and IL-18-mediated function. Immunity. 9:143 sue damage and regulates sensitivity to graft versus host disease. Immunity.
150. http://dx.doi.org/10.1016/S1074-7613(00)80596-8 37:339350. http://dx.doi.org/10.1016/j.immuni.2012.05.028
Basu, R., D.B. OQuinn, D.J. Silberger, T.R. Schoeb, L. Fouser, W. Ouyang, Harig, J.M., K.H. Soergel, R.A. Komorowski, and C.M. Wood. 1989. Treatment
R.D. Hatton, and C.T. Weaver. 2012. Th22 cells are an important source of diversion colitis with short-chain-fatty acid irrigation. N. Engl. J. Med.
of IL-22 for host protection against enteropathogenic bacteria. Immunity. 320:2328. http://dx.doi.org/10.1056/NEJM198901053200105
37:10611075. http://dx.doi.org/10.1016/j.immuni.2012.08.024 Hildner, K., B.T. Edelson, W.E. Purtha, M. Diamond, H. Matsushita, M.
Bernink, J.H., C.P. Peters, M. Munneke, A.A. te Velde, S.L. Meijer, K. Weijer, Kohyama, B. Calderon, B.U. Schraml, E.R. Unanue, M.S. Diamond,
H.S. Hreggvidsdottir, S.E. Heinsbroek, N. Legrand, C.J. Buskens, et al. 2013. et al. 2008. Batf3 deficiency reveals a critical role for CD8alpha+ dendritic
Human type 1 innate lymphoid cells accumulate in inflamed mucosal cells in cytotoxic T cell immunity. Science. 322:10971100. http://dx.doi
tissues. Nat. Immunol. 14:221229. http://dx.doi.org/10.1038/ni.2534 .org/10.1126/science.1164206
Bogunovic, M., F. Ginhoux, J. Helft, L. Shang, D. Hashimoto, M. Greter, K. Huber, S., N. Gagliani, L.A. Zenewicz, F.J. Huber, L. Bosurgi, B. Hu, M. Hedl,
Liu, C. Jakubzick, M.A. Ingersoll, M. Leboeuf, et al. 2009. Origin of the W. Zhang, W. OConnor Jr., A.J. Murphy, et al. 2012. IL-22BP is regu-
lamina propria dendritic cell network. Immunity. 31:513525. http://dx lated by the inflammasome and modulates tumorigenesis in the intestine.
.doi.org/10.1016/j.immuni.2009.08.010 Nature. 491:259263. http://dx.doi.org/10.1038/nature11535
Buonocore, S., P.P. Ahern, H.H. Uhlig, I.I. Ivanov, D.R. Littman, K.J. Maloy, and Hueber, W., B.E. Sands, S. Lewitzky, M. Vandemeulebroecke, W. Reinisch,
F. Powrie. 2010. Innate lymphoid cells drive interleukin-23-dependent P.D. Higgins, J. Wehkamp, B.G. Feagan, M.D. Yao, M. Karczewski, et al.
innate intestinal pathology. Nature. 464:13711375. http://dx.doi.org/10 Secukinumab in Crohns Disease Study Group. 2012. Secukinumab, a human
.1038/nature08949 anti-IL-17A monoclonal antibody, for moderate to severe Crohns disease:
Caton, M.L., M.R. Smith-Raska, and B. Reizis. 2007. Notch-RBP-J signal- unexpected results of a randomised, double-blind placebo-controlled trial.
ing controls the homeostasis of CD8- dendritic cells in the spleen. J. Exp. Gut. 61:16931700. http://dx.doi.org/10.1136/gutjnl-2011-301668
Med. 204:16531664. Jung, S., J. Aliberti, P. Graemmel, M.J. Sunshine, G.W. Kreutzberg, A. Sher,
Cella, M., A. Fuchs, W. Vermi, F. Facchetti, K. Otero, J.K. Lennerz, J.M. and D.R. Littman. 2000. Analysis of fractalkine receptor CX(3)CR1
Doherty, J.C. Mills, and M. Colonna. 2009. A human natural killer cell function by targeted deletion and green fluorescent protein reporter gene
subset provides an innate source of IL-22 for mucosal immunity. Nature. insertion. Mol. Cell. Biol. 20:41064114. http://dx.doi.org/10.1128/
457:722725. http://dx.doi.org/10.1038/nature07537 MCB.20.11.4106-4114.2000
Cua, D.J., J. Sherlock,Y. Chen, C.A. Murphy, B. Joyce, B. Seymour, L. Lucian, Kamada, N., T. Hisamatsu, S. Okamoto, H. Chinen, T. Kobayashi, T. Sato, A.
W. To, S. Kwan, T. Churakova, et al. 2003. Interleukin-23 rather than Sakuraba, M.T. Kitazume, A. Sugita, K. Koganei, et al. 2008. Unique
interleukin-12 is the critical cytokine for autoimmune inflammation of CD14 intestinal macrophages contribute to the pathogenesis of Crohn
the brain. Nature. 421:744748. http://dx.doi.org/10.1038/nature01355 disease via IL-23/IFN-gamma axis. J. Clin. Invest. 118:22692280.
Denning, T.L., B.A. Norris, O. Medina-Contreras, S. Manicassamy, D. Geem, Kamada, N., T. Hisamatsu, H. Honda, T. Kobayashi, H. Chinen, T. Takayama,
R. Madan, C.L. Karp, and B. Pulendran. 2011. Functional specializations M.T. Kitazume, S. Okamoto, K. Koganei, A. Sugita, et al. 2010.TL1A pro-
of intestinal dendritic cell and macrophage subsets that control Th17 and duced by lamina propria macrophages induces Th1 and Th17 immune
regulatory T cell responses are dependent on the T cell/APC ratio, source responses in cooperation with IL-23 in patients with Crohns disease.
of mouse strain, and regional localization. J. Immunol. 187:733747. http:// Inflamm. Bowel Dis. 16:568575. http://dx.doi.org/10.1002/ibd.21124
dx.doi.org/10.4049/jimmunol.1002701 Kinnebrew, M.A., C.G. Buffie, G.E. Diehl, L.A. Zenewicz, I. Leiner, T.M.
Diehl, G.E., R.S. Longman, J.X. Zhang, B. Breart, C. Galan, A. Cuesta, S.R. Hohl, R.A. Flavell, D.R. Littman, and E.G. Pamer. 2012. Interleukin 23
Schwab, and D.R. Littman. 2013. Microbiota restricts trafficking of bac- production by intestinal CD103(+)CD11b(+) dendritic cells in response
teria to mesenteric lymph nodes by CX(3)CR1(hi) cells. Nature. 494: to bacterial flagellin enhances mucosal innate immune defense. Immunity.
116120. http://dx.doi.org/10.1038/nature11809 36:276287. http://dx.doi.org/10.1016/j.immuni.2011.12.011
Duerr, R.H., K.D.Taylor, S.R. Brant, J.D. Rioux, M.S. Silverberg, M.J. Daly, A.H. Kirchberger, S., D.J. Royston, O. Boulard, E. Thornton, F. Franchini, R.L.
Steinhart, C. Abraham, M. Regueiro, A. Griffiths, et al. 2006. A genome- Szabady, O. Harrison, and F. Powrie. 2013. Innate lymphoid cells sustain
wide association study identifies IL23R as an inflammatory bowel disease colon cancer through production of interleukin-22 in a mouse model. J.
gene. Science. 314:14611463. http://dx.doi.org/10.1126/science.1135245 Exp. Med. 210:917931. http://dx.doi.org/10.1084/jem.20122308
Eberl, G., S. Marmon, M.J. Sunshine, P.D. Rennert,Y. Choi, and D.R. Littman. Klose, C.S., E.A. Kiss, V. Schwierzeck, K. Ebert, T. Hoyler, Y. dHargues, N.
2004. An essential function for the nuclear receptor RORgamma(t) in Gppert, A.L. Croxford, A.Waisman,Y.Tanriver, and A. Diefenbach. 2013.
the generation of fetal lymphoid tissue inducer cells. Nat. Immunol. 5:64 A T-bet gradient controls the fate and function of CCR6-RORt+
73. http://dx.doi.org/10.1038/ni1022 innate lymphoid cells. Nature. 494:261265. http://dx.doi.org/10.1038/
Edelson, B.T., W. Kc, R. Juang, M. Kohyama, L.A. Benoit, P.A. Klekotka, C. nature11813
Moon, J.C. Albring, W. Ise, D.G. Michael, et al. 2010. Peripheral CD103+ Kugathasan, S., R.N. Baldassano, J.P. Bradfield, P.M. Sleiman, M. Imielinski,
dendritic cells form a unified subset developmentally related to CD8+ S.L. Guthery, S. Cucchiara, C.E. Kim, E.C. Frackelton, K. Annaiah,
conventional dendritic cells. J. Exp. Med. 207:823836. http://dx.doi.org/ et al. 2008. Loci on 20q13 and 21q22 are associated with pediatric-onset
10.1084/jem.20091627 inflammatory bowel disease. Nat. Genet. 40:12111215. http://dx.doi
Geremia, A., C.V. Arancibia-Crcamo, M.P. Fleming, N. Rust, B. Singh, N.J. .org/10.1038/ng.203
Mortensen, S.P.Travis, and F. Powrie. 2011. IL-23responsive innate lymphoid Lewis, K.L., M.L. Caton, M. Bogunovic, M. Greter, L.T. Grajkowska, D. Ng, A.
cells are increased in inflammatory bowel disease. J. Exp. Med. 208:1127 Klinakis, I.F. Charo, S. Jung, J.L. Gommerman, et al. 2011. Notch2 recep
1133. http://dx.doi.org/10.1084/jem.20101712 tor signaling controls functional differentiation of dendritic cells in the

1582 CX3CR1+ MNPs regulate ILC3 | Longman et al.


Ar ticle

spleen and intestine. Immunity. 35:780791. http://dx.doi.org/10.1016/ Schlitzer, A., N. McGovern, P. Teo, T. Zelante, K. Atarashi, D. Low, A.W. Ho, P.
j.immuni.2011.08.013 See, A. Shin, P.S. Wasan, et al. 2013. IRF4 transcription factor-dependent
Manta, C., E. Heupel, K. Radulovic, V. Rossini, N. Garbi, C.U. Riedel, and CD11b+ dendritic cells in human and mouse control mucosal IL-17
J.H. Niess. 2013. CX(3)CR1(+) macrophages support IL-22 production cytokine responses. Immunity. 38:970983. http://dx.doi.org/10.1016/
by innate lymphoid cells during infection with Citrobacter rodentium. j.immuni.2013.04.011
Mucosal Immunol. 6:177188. http://dx.doi.org/10.1038/mi.2012.61 Schreiber, T.H., D. Wolf, M.S. Tsai, J. Chirinos, V.V. Deyev, L. Gonzalez, T.R.
Merad, M., P. Sathe, J. Helft, J. Miller, and A. Mortha. 2013. The dendritic cell Malek, R.B. Levy, and E.R. Podack. 2010.Therapeutic Treg expansion in
lineage: ontogeny and function of dendritic cells and their subsets in the mice by TNFRSF25 prevents allergic lung inflammation. J. Clin. Invest.
steady state and the inflamed setting. Annu. Rev. Immunol. 31:563604. 120:36293640. http://dx.doi.org/10.1172/JCI42933
http://dx.doi.org/10.1146/annurev-immunol-020711-074950 Schreiber, H.A., J. Loschko, R.A. Karssemeijer, A. Escolano, M.M. Meredith,
Meylan, F., E.T. Hawley, L. Barron, J.L. Barlow, P. Penumetcha, M. Pelletier, D. Mucida, P. Guermonprez, and M.C. Nussenzweig. 2013. Intestinal
G. Sciume, A.C. Richard, E.T. Hayes, J. Gomez-Rodriguez, et al. 2014. monocytes and macrophages are required for T cell polarization in re-
The TNF-family cytokine TL1A promotes allergic immunopathology sponse to Citrobacter rodentium. J. Exp. Med. 210:20252039. http://dx.doi
through group 2 innate lymphoid cells. Mucosal Immunol. 731:958968. .org/10.1084/jem.20130903
Mortha, A., A. Chudnovskiy, D. Hashimoto, M. Bogunovic, S.P. Spencer, Shih, D.Q., L.Y. Kwan,V. Chavez, O. Cohavy, R. Gonsky, E.Y. Chang, C. Chang,
Y. Belkaid, and M. Merad. 2014. Microbiota-dependent crosstalk be- C.O. Elson, and S.R. Targan. 2009. Microbial induction of inflammatory
tween macrophages and ILC3 promotes intestinal homeostasis. Science. bowel disease associated gene TL1A (TNFSF15) in antigen presenting cells.
343:1249288. http://dx.doi.org/10.1126/science.1249288 Eur. J. Immunol. 39:32393250. http://dx.doi.org/10.1002/eji.200839087
Niess, J.H., S. Brand, X. Gu, L. Landsman, S. Jung, B.A. McCormick, J.M. Sonnenberg, G.F., and D. Artis. 2012. Innate lymphoid cell interactions with
Vyas, M. Boes, H.L. Ploegh, J.G. Fox, et al. 2005. CX3CR1-mediated microbiota: implications for intestinal health and disease. Immunity. 37:
dendritic cell access to the intestinal lumen and bacterial clearance. 601610. http://dx.doi.org/10.1016/j.immuni.2012.10.003
Science. 307:254258. http://dx.doi.org/10.1126/science.1102901 Sonnenberg, G.F., L.A. Monticelli, M.M. Elloso, L.A. Fouser, and D. Artis. 2011.
Pappu, B.P., A. Borodovsky, T.S. Zheng, X.Yang, P. Wu, X. Dong, S. Weng, B. CD4(+) lymphoid tissue-inducer cells promote innate immunity in the gut.
Browning, M.L. Scott, L. Ma, et al. 2008. TL1A-DR3 interaction regu- Immunity. 34:122134. http://dx.doi.org/10.1016/j.immuni.2010.12.009
lates Th17 cell function and Th17-mediated autoimmune disease. J. Exp. Spits, H., D. Artis, M. Colonna, A. Diefenbach, J.P. Di Santo, G. Eberl, S.
Koyasu, R.M. Locksley, A.N. McKenzie, R.E. Mebius, et al. 2013.
Med. 205:10491062. http://dx.doi.org/10.1084/jem.20071364
Innate lymphoid cellsa proposal for uniform nomenclature. Nat. Rev.
Persson, E.K., H. Uronen-Hansson, M. Semmrich,A. Rivollier, K. Hgerbrand,
Immunol. 13:145149. http://dx.doi.org/10.1038/nri3365
J. Marsal, S. Gudjonsson, U. Hkansson, B. Reizis, K. Kotarsky, and W.W.
Sun, C.M., J.A. Hall, R.B. Blank, N. Bouladoux, M. Oukka, J.R. Mora, and
Agace. 2013. IRF4 transcription-factor-dependent CD103(+)CD11b(+)
Y. Belkaid. 2007. Small intestine lamina propria dendritic cells promote
dendritic cells drive mucosal T helper 17 cell differentiation. Immunity.
de novo generation of Foxp3 T reg cells via retinoic acid. J. Exp. Med.
38:958969. http://dx.doi.org/10.1016/j.immuni.2013.03.009
204:17751785. http://dx.doi.org/10.1084/jem.20070602
Pickert, G., C. Neufert, M. Leppkes, Y. Zheng, N. Wittkopf, M. Warntjen,
Tamoutounour, S., S. Henri, H. Lelouard, B. de Bovis, C. de Haar, C.J. van der
H.A. Lehr, S. Hirth, B. Weigmann, S. Wirtz, et al. 2009. STAT3 links
Woude, A.M. Woltman,Y. Reyal, D. Bonnet, D. Sichien, et al. 2012. CD64
IL-22 signaling in intestinal epithelial cells to mucosal wound healing. distinguishes macrophages from dendritic cells in the gut and reveals the
J. Exp. Med. 206:14651472. http://dx.doi.org/10.1084/jem.20082683 Th1-inducing role of mesenteric lymph node macrophages during colitis.
Pineton de Chambrun,G.,L.Peyrin-Biroulet,M.Lmann,and J.F.Colombel. Eur. J. Immunol. 42:31503166. http://dx.doi.org/10.1002/eji.201242847
2010. Clinical implications of mucosal healing for the management of Varol, C., E. Zigmond, and S. Jung. 2010. Securing the immune tightrope:
IBD. Nat. Rev. Gastroenterol. Hepatol. 7:1529. http://dx.doi.org/10.1038/ mononuclear phagocytes in the intestinal lamina propria. Nat. Rev. Immunol.
nrgastro.2009.203 10:415426. http://dx.doi.org/10.1038/nri2778
Qiu, J., J.J. Heller, X. Guo, Z.M. Chen, K. Fish, Y.X. Fu, and L. Zhou. 2012. Vernia, P., M. Cittadini, R. Caprilli, and A. Torsoli. 1995. Topical treatment of
The aryl hydrocarbon receptor regulates gut immunity through modula- refractory distal ulcerative colitis with 5-ASA and sodium butyrate. Dig.
tion of innate lymphoid cells. Immunity. 36:92104. http://dx.doi.org/10 Dis. Sci. 40:305307. http://dx.doi.org/10.1007/BF02065414
.1016/j.immuni.2011.11.011 Welty, N.E., C. Staley, N. Ghilardi, M.J. Sadowsky, B.Z. Igyrt, and D.H.
Satoh-Takayama, N., C.A.Vosshenrich, S. Lesjean-Pottier, S. Sawa, M. Lochner, Kaplan. 2013. Intestinal lamina propria dendritic cells maintain T cell
F. Rattis, J.J. Mention, K. Thiam, N. Cerf-Bensussan, O. Mandelboim, homeostasis but do not affect commensalism. J. Exp. Med. 210:2011
et al. 2008. Microbial flora drives interleukin 22 production in intestinal 2024. http://dx.doi.org/10.1084/jem.20130728
NKp46+ cells that provide innate mucosal immune defense. Immunity. Wynn, T.A., A. Chawla, and J.W. Pollard. 2013. Macrophage biology in de-
29:958970. http://dx.doi.org/10.1016/j.immuni.2008.11.001 velopment, homeostasis and disease. Nature. 496:445455. http://dx.doi
Satpathy, A.T., C.G. Briseo, J.S. Lee, D. Ng, N.A. Manieri,W. Kc, X.Wu, S.R. .org/10.1038/nature12034
Thomas, W.L. Lee, M. Turkoz, et al. 2013. Notch2-dependent classical Zheng,Y., P.A.Valdez, D.M. Danilenko,Y. Hu, S.M. Sa, Q. Gong, A.R. Abbas, Z.
dendritic cells orchestrate intestinal immunity to attaching-and-effacing Modrusan, N. Ghilardi, F.J. de Sauvage, and W. Ouyang. 2008. Interleukin-
bacterial pathogens. Nat. Immunol. 14:937948. http://dx.doi.org/10.1038/ 22 mediates early host defense against attaching and effacing bacterial
ni.2679 pathogens. Nat. Med. 14:282289. http://dx.doi.org/10.1038/nm1720
Sawa, S., M. Lochner, N. Satoh-Takayama, S. Dulauroy, M. Brard, M. Zigmond, E., C. Varol, J. Farache, E. Elmaliah, A.T. Satpathy, G. Friedlander,
Kleinschek, D. Cua, J.P. Di Santo, and G. Eberl. 2011. RORt+ innate M. Mack, N. Shpigel, I.G. Boneca, K.M. Murphy, et al. 2012. Ly6C hi
lymphoid cells regulate intestinal homeostasis by integrating negative sig- monocytes in the inflamed colon give rise to proinflammatory effector
nals from the symbiotic microbiota. Nat. Immunol. 12:320326. http:// cells and migratory antigen-presenting cells. Immunity. 37:10761090.
dx.doi.org/10.1038/ni.2002 http://dx.doi.org/10.1016/j.immuni.2012.08.026

JEM Vol. 211, No. 8 1583


INSIGHTS

Monocytes find a new place to dwell in the niche of


heartbreak hotel
Monocytes and macrophages belong to the mononuclear phagocyte system (MPS). The con-

Lambrecht Photo courtesy of


cept of the MPS is based on the idea that circulating monocytes represent the immediate
precursors of tissue macrophages. Recent genetic fate mapping of myeloid cells caused a con-
ceptual frameshift, as it was found that many macrophages are derived from embryonic pre-
cursors that seed the tissues before birth and then self-maintain without any significant

www.vib.be
contribution from circulating monocytes. In this new view, monocytes merely represent an
emergency squad that can be rapidly recruited to sites of inflammation to provide a transient
Insight from Bart Lambrecht (left)
supplement of inflammatory macrophages to the local tissue-resident macrophage pool of and Martin Guilliams (right)
embryonic origin.
In this issue, Molawi et al. challenge this new concept by reporting that cardiac macrophages are initially of embryonic
origin but are progressively replaced by monocyte-derived cells. These newcomer macrophages lack the capacity to self-renew
and are slowly but constantly replaced by circulating monocytes. In this regard, the heart resembles the skin and intestine.
There, macrophages are also mainly derived from circulating monocytes. Because the skin and the intestine are barrier sites with
an important influence of the microbiome, a continuous low-grade inflammation could explain continuous recruitment of
circulating monocytes. Molawi et al. report that cardiac macrophages of embryonic origin gradually lose the capacity to self-
renew with age and hypothesize that this allows circulating monocytes to occupy the heart niche and join the cardiac macrophage
pool. Why embryonic macrophages would lose their self-renewal capacity in the heart but not in the liver, the lung, or the
spleen remains to be determined. Another unexplored hypothesis is that continuous microtrauma induced by cardiac contraction
is the driver of monocyte influx to the sterile heart.
Tissue-resident macrophages are astonishingly diverse in terms of gene expression and are adapted to perform specific functions
in their tissue of residence. The unique functions of cardiac macrophages remain largely unknown. Molawi et al. describe
four subsets of cardiac macrophages based on the expression of CX3CR1 and MHC class II (see figure). Embryonic macrophages
preferentially give rise to MHC class IIlow cells, while most monocyte-derived macrophages express high levels of MHC class II.
This implies a different capacity to
interact with lymphocytes and distinct
functional adaptations for embryonic
macrophages. Macrophages play an im-
portant role in wound healing, and many
cardiovascular diseases are associated with
poor or exaggerated tissue repair. It will
therefore be interesting to investigate
whether embryonic macrophages are
better at tissue repair compared with
monocyte-derived macrophages, and
whether the presence of the embryonic
pool explains the superior regeneration
capacity of the heart at a young age.
Manipulation of macrophage self-renewal
might yield important therapeutic appli-
Cardiac macrophages derived from embryonic progenitors gradually lose their capacity to cations to increase macrophage popula-
self-renew and are continually replaced in adulthood by macrophages derived from circulating tions involved in tissue homeostasis and
monocytes, even in the absence of inflammation. repair, while avoiding the recruitment of
macrophages that fuel inflammation.
With these exciting prospects and new concepts, macrophage biology is going through a revival period, and this study
certainly contributes to that. Given the breadth of tissue-specific macrophage heterogeneity, both in terms of functional specialization
and cellular origin, we have many years ahead until we fully understand the cell that initiated immunology research more than
a century ago.
Molawi, K., et al. 2014. J. Exp. Med. http://dx.doi.org/10.1084/jem.20140639.

Bart Lambrecht and Martin Guilliams, VIB Inflammation Research Center, UGent: bart.lambrecht@irc.vib-Ugent.be and martin.guilliams@irc.ugent.be
Brief Definitive Report

Progressive replacement of embryo-derived


cardiac macrophages with age
Kaaweh Molawi,1,2,3,4 Yochai Wolf,5 Prashanth K. Kandalla,1,2,3
Jeremy Favret,1,2,3 Nora Hagemeyer,6 Kathrin Frenzel,6 Alexander R. Pinto,8
Kay Klapproth,9 Sandrine Henri,1,2,3 Bernard Malissen,1,2,3
Hans-Reimer Rodewald,9 Nadia A. Rosenthal,8,10 Marc Bajenoff,1,2,3
Marco Prinz,6,7 Steffen Jung,5 and Michael H. Sieweke1,2,3,4
1Centre dImmunologie de Marseille-Luminy (CIML), Aix-Marseille Universit, UM2, 13288 Marseille, France
2Institut National de la Sant et de la Recherche Mdicale (INSERM), U1104, 13288 Marseille, France
3Centre National de la Recherche Scientifique (CNRS), UMR7280, 13288 Marseille, France
4Max-Delbrck-Centrum fr Molekulare Medizin (MDC), Robert-Rssle-Strasse 10, 13125 Berlin, Germany
5Department of Immunology, The Weizmann Institute of Science, 7610001 Rehovot, Israel
6Institute of Neuropathology and 7BIOSS Centre for Biological Signaling Studies, University of Freiburg, 79106 Freiburg, Germany
8Australian Regenerative Medicine Institute (ARMI), Monash University, Clayton 3800, Victoria, Australia
9Division of Cellular Immunology, German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany
10National Heart and Lung Institute, Imperial College London, London SW7 2AZ, England, UK

Cardiac macrophages (cM) are critical for early postnatal heart regeneration and fibrotic
repair in the adult heart, but their origins and cellular dynamics during postnatal develop-
ment have not been well characterized. Tissue macrophages can be derived from embryonic
progenitors or from monocytes during inflammation. We report that within the first weeks
after birth, the embryo-derived population of resident CX3CR1+ cM diversifies into
MHCII+ and MHCII cells. Genetic fate mapping demonstrated that cM derived from
CX3CR1+ embryonic progenitors persisted into adulthood but the initially high contribution
to resident cM declined after birth. Consistent with this, the early significant prolifera-
tion rate of resident cM decreased with age upon diversification into subpopulations.
Bone marrow (BM) reconstitution experiments showed monocyte-dependent quantitative
replacement of all cM populations. Furthermore, parabiotic mice and BM chimeras of
nonirradiated recipient mice revealed a slow but significant donor contribution to cM.
Together, our observations indicate that in the heart, embryo-derived cM show declining
self-renewal with age and are progressively substituted by monocyte-derived macrophages,
even in the absence of inflammation.

CORRESPONDENCE Cardiovascular disease represents a leading process (Nahrendorf et al., 2007; Swirski et al.,
Michael H. Sieweke: cause of death in the developed world, and 2009). The focus of these studies has been on
sieweke@ciml.univ-mrs.fr
many pathologies result from insufficient repair monocyte-derived cells, consistent with the tra-
Abbreviations used: cM, of cardiac injury. Mammalian wound healing ditional view that macrophages are part of the
cardiac macrophages; HSC, mechanisms in the adult heart involve scar for- mononuclear phagocyte system and monocyte-
hematopoietic stem cell; mation, but for a short time window after birth derived (van Furth and Cohn, 1968; Geissmann
YS, yolk sac.
the neonatal heart maintains full regeneration et al., 2010). Recent evidence, however, sug-
capacity of cardiac tissue (Porrello et al., 2011), gests that monocyte contribution to macro-
a process which requires macrophages (Aurora phages only represents an emergency pathway,
et al., 2014). Studies of cardiac repair after myo- as many tissue macrophage populations get
cardial infarction in the adult have highlighted seeded in the developing embryo and can self-
the critical role of infiltrating monocytes and maintain without major monocyte contribution
monocyte-derived macrophages for the healing
2014 Molawi et al. This article is distributed under the terms of an Attribution
NoncommercialShare AlikeNo Mirror Sites license for the first six months
after the publication date (see http://www.rupress.org/terms). After six months
it is available under a Creative Commons License (AttributionNoncommercial
K. Molawi and Y. Wolf contributed equally to this paper. Share Alike 3.0 Unported license, as described at http://creativecommons.org/
Steffen Jung and M.H. Sieweke contributed equally to this paper. licenses/by-nc-sa/3.0/).

The Rockefeller University Press $30.00


J. Exp. Med. 2014 Vol. 211 No. 11 2151-2158 2151
www.jem.org/cgi/doi/10.1084/jem.20140639
Figure 1. cM develop into 4 subpopulations after birth. (A and B) Cytometry analysis of cM from adult CX3CR1GFP/+ mice. (A) FACS profiles of
cM populations, pregated on living single CD11b+ cells with low CD11c and Ly6C expression. (B) Mean of total and subpopulations of cM in absolute
numbers (left) or as percentage of total (right) as determined by bead-normalized flow cytometry. Error bars represent SEM (n = 4). (C and D) Cytometry
analysis (C) and mean percentage (D) of cM subpopulations from CX3CR1GFP/+ mice of indicated age. Error bars represent SEM (n = 34). (E) MHCII and
MHCII+ cM from adult CX3CR1cre:R26-yfp mice (black line) were analyzed for YFP expression and compared with Cre littermate controls (gray area;
n = 34). Data in all panels are representative of at least two independent experiments.

(Chorro et al., 2009; Ginhoux et al., 2010; Hoeffel et al., et al., 2012; Tamoutounour et al., 2013). Thus, origin and
2012; Schulz et al., 2012; Hashimoto et al., 2013;Yona et al., turnover of tissue macrophages are highly tissue specific and
2013). Macrophages can massively expand in the tissue by need to be assessed individually for different organ systems
local proliferation in response to challenge (Jenkins et al., (Sieweke and Allen, 2013).
2011; Hashimoto et al., 2013; Sieweke and Allen, 2013) and Resident macrophages can be found in all tissues, where
can extensively self-renew without loss of differentiated func- they fulfill a variety of tissue-specific functions contributing
tion in culture upon inactivation of MafB and cMaf tran- to homeostasis, development, and regeneration (Davies et al.,
scription factors (Aziz et al., 2009). This has led to the 2013), making them prominent candidates for therapeutic in-
proposition that tissue macrophages may have a long-term tervention.This holds in particular for the heart, where tissue-
self-renewal capacity akin to that of stem cells (Sieweke and resident cardiac macrophages (cM) have been described as
Allen, 2013). CX3CR1+ cells that can be found throughout the myocar-
Examples of tissue macrophages that are independent dium (Pinto et al., 2012). A recent study identified several
from monocytes are microglia in the brain and epidermal cM populations, including short-lived monocyte-derived
Langerhans cells (Ajami et al., 2007; Chorro et al., 2009; cells that resemble monocyte-derived DCs as well as tissue-
Ginhoux et al., 2010; Hoeffel et al., 2012). In contrast, intesti- resident cM, which were suggested to be primarily of embry-
nal or dermal macrophages have a high turnover rate and are onic origin and to self-maintain under homeostatic conditions
constantly replaced from Ly6C+ blood monocytes (Zigmond (Epelman et al., 2014).

2152 Replacement of embryonic cardiac macrophages with age | Molawi et al.


Br ief Definitive Repor t

Figure 2. Decreased contribution of embryo-


derived macrophages to cM with age.
(A) CX3CR1creER:R26-yfp embryos were treated with
TAM on E9 or E13 and analyzed on the day of delivery
(P0) or 6 wk after delivery (P42). (B) Percentage of
microglia-normalized YFP+ cM and representative
cytometry plots pregated on total cM at P0 and
P42 after E13 labeling. Bars show median (n = 913).
***, P 0.005, Mann-Whitney test. Data were pooled
from two independent experiments. (C) Representa-
tive cytometry plot showing YFP+ within total cM
at P42 and percentage of microglia-normalized YFP+
cM at P0 and P42 after E9 labeling. Bars show
median (n = 7) *, P 0.05. Data were pooled from
three independent experiments. (D) Representative
cytometry plots showing YFP+ cells within MHCII
and MHCII+ cM at P0 and P42 after E13 labeling
(n = 913). (E) Quantification of YFP+ cells within
MHCII and MHCII+ cM at P42 after E13 labeling.
Bars show the median (n = 913). ***, P 0.005,
Mann-Whitney test. Data were pooled from two
independent experiments. (F) Adult TAM-treated
CX3CR1creER:R26-yfp mice were analyzed for YFP+
cM and microglia at 1 and 4 wk after treatment.
Percentage of microglia-normalized YFP+ cM is
shown as median with extreme samples as error
bars (n = 3). Representative cytometry plots
show YFP+ within total cM. Unless otherwise
indicated data in all panels are representative of
two independent experiments.

Here, we examined the origin and turnover of tissue- (Tamoutounour et al., 2013). We found that in 8-wk-old
resident cM during postnatal development. Using genetic mice, the majority of cM was CX3CR1+ (80%) and
lineage tracing, parabiotic mice, and unconditioned BM chi- MHCII+ (70%), allowing the delineation of four distinct
meras, we demonstrate that embryo-derived cM are gradu- cM subpopulations (Fig. 1, A and B). Additional populations
ally replaced by monocyte-derived macrophages with age and of CD11c+MHCII+ or Ly6C+ cells, referred to as cM
in the absence of inflammation or injury. This replacement is by others (Epelman et al., 2014), were excluded from our
mirrored by dynamic changes in the resident cM population analysis (Fig. S1 B), as data from other tissues suggest that
composition and decreasing cM self-renewal over time. these are monocytes and monocyte-derived dendritic cells
(Tamoutounour et al., 2013). Consistent with this, these cells
RESULTS AND DISCUSSION have been shown to be short-lived and monocyte-derived
We focused on the resident cM population and applied a (Epelman et al., 2014).
rigorous flow cytometry gating strategy to exclude potential To investigate the development of the four cM popula-
infiltrating leukocytes (Fig. S1, AC). Tissue-resident macro- tions, we analyzed CX3CR1 and MHCII expression in cM
phages were identified as positive for the core macrophage from newborn to 30-wk-old mice (Fig. 1, C and D). We ob-
signature markers CD14, CD64, and MerTK (Gautier et al., served that almost all embryo-derived cM present at birth
2012; Tamoutounour et al., 2013) and the classical macro- were CX3CR1+MHCII. This homogenous cM compart-
phage marker F4/80 (Fig. 1 A). The chemokine receptor ment diversified with age into four subpopulations with a pro-
CX3CR1 is widely expressed in the mononuclear phagocyte gressive increase of MHCII+ cM and a decrease of CX3CR1+
system (Jung et al., 2000) and cM have been reported to be cM. Importantly, genetic fate mapping analysis using
CX3CR1-positive (Pinto et al., 2012). Therefore, we refined CX3CR1cre mice crossed to R26-yfp reporter mice (CX3CR1cre:
our analysis of the global cM population by testing CX3CR1 R26-yfp; Yona et al., 2013) revealed that all adult cM sub-
expression in CX3CR1GFP/+ knock-in mice (Jung et al., populations must have developed from a CX3CR1+ stage
2000). Additionally, we included MHCII in our analysis, (Fig. 1 E). These results suggested two not mutually exclusive
which can be differentially expressed on tissue macrophages possibilities for cM subpopulation development: persistence

JEM Vol. 211, No. 11 2153


Figure 3. Decreased proliferation rate of embryo-derived cM with age. (A) cM subpopulations of adult CX3CR1GFP/+ mice were analyzed for
BrdU incorporation and expression of Ki67 by flow cytometry four hours after BrdU injection (i.p.). Bars show median (n = 1213). *, P 0.05; **, P 0.01;
***, P 0.005; Wilcoxon test. (B and C) Total (B) or CX3CR1+MHCII (C) cM of CX3CR1GFP/+ mice of indicated age were analyzed for BrdU incorporation
and Ki67 expression by flow cytometry 4 h after BrdU injection (i.p.). Bars show median (n = 415). ***, P 0.005, Mann-Whitney test. Data in all panels
were pooled from 23 independent experiments.

and diversification of embryo-derived cM as suggested else- We further assessed the turnover of CX3CR1+ cM in
where (Epelman et al., 2014), or replacement of embryo- the adult heart by pulse labeling adult CX3CR1creER:R26-yfp
derived cM by adult monocyte-derived macrophages. (Fig. 2 F). 1 wk after treatment, labeling in cM corresponded
To address these alternatives, we first analyzed the persis- to 60% of microglia labeling but drastically decreased to
tence of embryo-derived cM by genetic lineage tracing using 20% after 4 wk. By comparison, microglia labeling was
CX3CR1-driven tamoxifen (TAM)-inducible Cre recom- nearly complete after 1 wk and remained unchanged there-
binase (CreERT2) and R26-yfp reporter mice (CX3CR1creER: after (Goldmann et al., 2013). Collectively, our lineage tracing
R26-yfp; Fig. 2 A; Yona et al., 2013). Tamoxifen-induced experiments argue that a significant proportion of macro-
pulse labeling at E9 permits us to identify cells derived from phages established in early embryonic development is still
yolk sac (YS) CX3CR1+ macrophages before the onset of present at birth but is gradually lost with age.
definitive hematopoiesis. Labeling at E13 cannot distinguish The relatively fast turnover of labeled tissue-resident cM
YS- and hematopoietic stem cell (HSC)derived macro- suggested that local self-renewal was insufficient to maintain
phages, but should increase cM labeling because the heart the resident cM pool. We therefore analyzed the prolifera-
is colonized by CX3CR1+ macrophages at this time (Epelman tive activity of cM subpopulations in adult mice by BrdU
et al., 2014). cM labeling was normalized to YFP+ mi- incorporation after a 4-h pulse labeling period and by Ki67
croglia, which are YS-derived, remain CX3CR1+ throughout staining (Fig. 3 A). The overall number of proliferating mac-
development, self-maintain without contribution of HSC- rophages in adult hearts was low, particularly in MHCII+
derived cells (Ginhoux et al., 2010; Kierdorf et al., 2013), populations, but the proliferative rate of CX3CR1+MHCII
and therefore represent an internal control for maximal cM significantly exceeded that of all other subpopulations.
CX3CR1 labeling efficiency. E13 labeling revealed that rela- Interestingly, this population was also the only cM subset
tive cM labeling declined from 35% in newborn mice present at birth but declined with age at the expense of the other
to 18% in 6-wk-old mice (Fig. 2 B), indicating a loss of cM populations (Fig. 1, C and D). We therefore analyzed the
embryo-derived cM with age. E9 labeling showed that evolution of cM proliferation with age. Both BrdU incor-
embryo-derived cM were at least partially of YS origin and poration and Ki67 staining showed a high proliferative rate
similarly declined from 14% in neonates to 4% in 6-wk- in newborn CX3CR1+MHCII cM, which gradually de-
old mice (Fig. 2 C). Interestingly, all embryo-derived YFP+ creased by 810-fold both in total cM (Fig. 3 B) and in the
cM were MHCII at birth but had partially differentiated CX3CR1+MHCII cM population (Fig. 3 C). Together,
into MHCII+ cM after 6 wk (Fig. 2 D). Although this these data suggest that both a successive loss of the cell popu-
demonstrated that both populations can develop from cells of lation with the highest proliferative rate (CX3CR1+MHCII
embryonic origin, the relative contribution of embryo-derived cM) and a strong decrease of the proliferation rate collectively
YFP+ cM to MHCII+ cM was significantly lower than to result in progressively decreasing self-renewal of the tissue-
MHCII cM, (Fig. 2 E). resident cM pool.

2154 Replacement of embryonic cardiac macrophages with age | Molawi et al.


Br ief Definitive Repor t

Figure 4. Monocytes contribute to four cM subpopulations in adult mice. (A) WT mice were lethally irradiated and reconstituted with BM from
Ubow:CX3CR1GFP/+ mice. cM were analyzed for contribution of dTomato+ cells at indicated time points after reconstitution and graft-derived cells were
analyzed for CX3CR1 and MHCII expression. Data are presented as mean percentage SEM (n = 37) and derived from two independent experiments.
(B) Mixed BM chimeras were generated by reconstituting lethally irradiated WT mice (CD45.1/CD45.2) with BM from CCR2/:CX3CR1GFP/+ (CD45.2) and
WT CX3CR1GFP/+ (CD45.1) mice. cM, circulating CD11b+CD115+ monocytes (Ly6C+ and Ly6C) and B220+ B cells were analyzed for WT (CD45.1) and
CCR2/ (CD45.2) contribution. Host- and graft-derived cM were analyzed for the ratio of MHCII+/MHCII cM. Bars show median (n = 4). *, P 0.05,
Mann-Whitney test. (C) Parabiosis was established between adult CCR2/ (CD45.2) and WT (CD45.1) mice. After 10 wk, CCR2/ mice were analyzed for
contribution of CD45.1+ non-host cells to cM (total, MHCII+, and MHCII), circulating monocytes (Ly6C+ and Ly6C), and B cells. For CD45.1 host and
CD45.1+ non-host cM, the ratio of MHCII+/MHCII cM was calculated and compared. Bars show median (n = 4). **, P 0.01, Mann-Whitney test.
(D) BM chimeras were generated by transfer of LT-HSC isolated from panYFP mice into nonirradiated Rag2/c/KitW/Wv mice. cM (total, MHCII+, and
MHCII) and neutrophils (Gr1hi and SSChi) were analyzed for contribution of grafted YFP+ cells 8 wk after transplantation. Bars show median (n = 6).
**, P 0.01, Mann-Whitney test. Data in all panels are representative of two independent experiments.

We next addressed the question of how the age-dependent However, a small radio-resistant population (10%) persisted
loss of embryo-derived macrophages is compensated. To test for 36 wk without monocyte contribution. To further analyze
the ability of monocytes to contribute to cM populations, whether cM replacement was monocyte-dependent, we
we generated BM chimeras using WT hosts and BM from generated mixed chimeras with BM from WT (CD45.1)
transgenic mice expressing dTomato from a ubiquitously ex- and CCR2/ (CD45.2) CX3CR1GFP/+ mice in CD45.1/
pressed human Ubiquitin-C promoter (Ubow mice; Ghigo CD45.2 double-positive WT hosts (Fig. 4 B). CCR2/ mice
et al., 2013) and GFP from the CX3CR1 locus that makes it have reduced numbers of Ly6C+ monocytes in the blood
possible to monitor grafted cells in all four cM populations. (Serbina and Pamer, 2006) and can therefore be used to ana-
Graft-derived dTomato+ cM contributed to all four cM lyze the contribution of circulating monocytes and CCR2-
populations from 4 wk after transplantation and almost com- dependent progenitors to tissue macrophage populations (Yona
pletely replaced host cM populations within 8 wk (Fig. 4 A). et al., 2013; Tamoutounour et al., 2013). Blood analysis indeed

JEM Vol. 211, No. 11 2155


confirmed that the vast majority of monocytes but not B cells MHCII cells, explaining the relative increase of MHCII+ cM
were of WT origin. Graft-derived cM were almost com- and the reduction of the CX3CR1+MHCII cM subpopu-
pletely derived from WT cells (Fig. 4 B). Collectively these lation with age.The dynamic change of the cM subpopulation
two experiments established that HSC-derived CCR2- distribution therefore appears to reflect the gradual replace-
dependent cells, most likely Ly6C+ monocytes, have the capac- ment of embryo-derived cM by monocyte-derived macro
ity to differentiate into all cM subsets. Residual host-derived phages. The increasing contribution of monocyte-derived
cM included both MHCII+ and MHCII cM but showed macrophages to the cM pool with age may be explained by
a lower proportion of MHCII+ cells than grafted monocyte- the decreasing self-renewal of embryo-derived cM. Although
derived cM (Fig. 4 B). CX3CR1+MHCII cM that contain the majority of em-
To examine monocyte contribution to cM populations bryo-derived macrophages have a higher proliferative rate than
under homeostatic conditions, we generated parabiotic cou- the other subpopulations, overall cM self-renewal is decreas-
ples of WT (CD45.1) and CCR2/ (CD45.2) mice and ing with age and is insufficient to sustain the cM pool with-
analyzed non-host contributions after 10 wk of parabiosis in out input from infiltrating monocytes. It will be interesting to
CCR2/ partners (Fig. 4 C). Circulating B cells were equally determine whether the loss of self-renewal capacity in cM is
distributed between the partner mice. In the CCR2-deficient permanent or might be reactivated. The observed mechanism
host Ly6C+ monocytes had reached 70% non-host chime- of cM homeostasis differs from other macrophage popula-
rism. About 16% of total cM in CCR2-deficient mice were tions such as microglia, Langerhans cells, or alveolar macro-
of non-host origin. Given a 70% chimerism in Ly6C+ mono- phages, which in the absence of tissue damage or inflammation
cytes, the likely cells of origin, and the possibility that de- appear to self-maintain in tissues without replacement from
pending on lifetime and exchange rate cM may not be fully circulating cells. It is also distinct from the high turnover rate of
equilibrated after 10 wk, these observations indicated a signif- dermal tissue macrophages or intestinal lamina propria macro-
icant monocyte contribution to tissue-resident cM. Again, phages that are constantly replenished from circulating mono-
non-host contribution to MHCII+ cM (30%) was signifi- cytes (Zigmond et al., 2012; Tamoutounour et al., 2013). In
cantly higher compared with MHCII cM (5%) as high- contrast to what was proposed elsewhere (Epelman et al., 2014),
lighted by MHCII+/MHCII cM ratios for non-host and our results also show that inflammation or injury are not neces-
host-derived cells (Fig. 4 C). sarily required for replacement of embryo-derived cardiac
As a complimentary experimental approach that does not tissue macrophages by monocytes. It will be interesting to
depend on myeloablative conditioning, we used Rag2/c/ determine whether dynamic age-dependent changes in resi-
KitW/Wv mice, which are universal HSC recipients that accept
dent tissue macrophage populations also occur in other tissues
grafts without prior irradiation (Waskow et al., 2009), and and whether they may be important for the differences in re-
generated BM chimeras by transplantation of HSC from pair mechanisms and regenerative capacity observed between
panYFP mice (Luche et al., 2013). More than 90% of neutrophils young and old individuals.
were YFP+ after 8 wk, demonstrating efficient reconstitution
(Fig. 4 D). Analysis of cM revealed 7% YFP+ cells in the MATERIALS AND METHODS
Mice. CD45.1 and CD45.2 C57BL/6 mice were obtained from Charles
total population. Again, donor cells showed a higher contribu-
River. All transgenic mice used in this study have a C57BL/6 background
tion to MHCII+ (8% YFP+) than to MHCII cM (4% (>7 backcrosses). CX3CR1GFP/+ mice (Jung et al., 2000), CCR2/, Rosa-26-
YFP+) (Fig. 4 D). In the same mice, no donor contribution yfp, CX3CR1cre (JAX Stock No. 25524 B6.C-Cx3cr1<tm1.1(cre)Jung>/J), and
to microglia cells was observed (K. Klapproth and H.R. CX3CR1creER mice (JAX Stock No. 20940 B6J.129-Cx3cr1<tm1.1(cre/
Rodewald, personal communication). Together, these experi- ERT2)Jung>/J; Yona et al., 2013), Ubow mice (Ghigo et al., 2013), Rag2/c/
ments clearly demonstrated a slow but significant replacement KitW/Wv mice (Waskow et al., 2009), and panYFP mice (Luche et al., 2013)
were described elsewhere. Experiments were performed on 610-wk-old
of tissue-resident cM by infiltrating monocyte-derived mac-
mice, if not stated otherwise. Experiments involving CX3CR1cre and
rophages, which preferentially contributed to MHCII+ cM. CX3CR1creER mice included littermate controls. For other experiments
In the present study, we have analyzed homeostasis, origin, C57BL/6 mice served as controls. All mouse experiments were performed
and turnover of resident macrophages in the heart during under specific pathogen-free conditions. Animals were handled according
postnatal development. We have shown that the cM com- to protocols approved by the Weizmann Institute Animal Care Committee
partment is undergoing dynamic changes with age. Embryo- (Israel), the Federal Ministry for Nature, Environment and Consumers Pro-
tection of the state of Baden-Wrttemberg (Germany) or the animal ethics
derived macrophages present in newborn mice are all CX3CR1+
committee of Marseille (France) and were performed in accordance to the
MHCII and at least partially YS-derived. After birth, the respective international, national, and institutional regulations.
cM compartment diversifies into four subpopulations
defined by MHCII and CX3CR1 expression. Our results Parabiosis. 9-wk-old C57BL/6 CD45.1 mice were sutured together with
show that this diversification is due both to differentiation 9-wk-old B6 CCR2/ CD45.2 and subsequently kept under Bactrim for
of persisting embryo-derived CX3CR1+MHCII cM and 10 wk before analysis.
infiltrating monocyte-derived macrophages, which both can
BM chimeras. 710-wk-old host animals were lethally irradiated and were
contribute to all four cM subpopulations. However, mono- reconstituted with donor BM by i.v. injection of minimum 106 BM cells.
cytes preferentially give rise to MHCII+ cM, whereas embryo- Donor BM was isolated from femurs and tibias of donor mice, filtered through
derived or radio-resistant cM had a higher proportion of a 70-m mesh and resuspended in PBS for i.v. injection. Rag2/c/KitW/Wv

2156 Replacement of embryonic cardiac macrophages with age | Molawi et al.


Br ief Definitive Repor t

mice were reconstituted with long-term (LT)HSC from panYFP BM K. Molawi was supported by an HFSP long-term fellowship. The work
without prior conditioning as previously described (Waskow et al., 2009). was supported by grants to M.H. Sieweke from Aviesan ITMO IHP exploratory
LT-HSCs were defined as lineage-negative (B220, CD3e, CD4, Project A12194AS and CNRS PICS program No. 5730. M.H. Sieweke is a
CD8a, CD11b, CD19, Gr1, Ter119, NK1.1), Kit+, Sca1+, CD48, Fondation pour la Recherche Mdicale (DEQ. 20071210559 and DEQ.
and CD150+. 20110421320) and INSERM-Helmholtz group leader. The work was further
supported by the Israel Science Foundation (ISF), the Deutsche Forschungs
gemeinschaft (DFG) Research Unit (FOR) 1336 (S. Jung and M. Prinz), and
Tamoxifen treatment. Tamoxifen (TAM; Sigma-Aldrich) was dissolved in
DFG-SFB 938-project L (H.-R. Rodewald and K. Klapproth).
100% ethanol to get a 1 g/ml solution and was 10-fold diluted in corn oil The authors declare no competing financial interests.
(Sigma-Aldrich) to get the 100 mg/ml final solution for oral or i.p. adminis-
tration. To induce Cre-mediated gene recombination in 5- to 7-wk-old
Submitted: 6 April 2014
CX3CR1creER mice, 5 mg TAM was administrated orally for 5 consecutive
Accepted: 19 August 2014
days (25 mg total). For Cre induction in the embryo, 200 l of 20 mg/ml
TAM and 10 mg/ml Progesterone dissolved in corn oil was injected i.p. into
pregnant females at 9 or 13 days post coitum (dpc). REFERENCES
Ajami, B., J.L. Bennett, C. Krieger, W. Tetzlaff, and F.M.V. Rossi. 2007.
BrdU pulsing. BrdU was purchased from Sigma-Aldrich. Mice were in- Local self-renewal can sustain CNS microglia maintenance and function
jected with 0.1 mg BrdU/g body weight. Newborn mice were injected sub- throughout adult life. Nat. Neurosci. 10:15381543. http://dx.doi.org/10
.1038/nn2014
cutaneously, older mice intraperitoneally. cM isolated from BrdU injected
Aurora, A.B., E.R. Porrello, W. Tan, A.I. Mahmoud, J.A. Hill, R. Bassel-
mice were analyzed 4 h after injection for BrdU incorporation using the
Duby, H.A. Sadek, and E.N. Olson. 2014. Macrophages are required
BrdU Flow kit (BD).
for neonatal heart regeneration. J. Clin. Invest. 124:13821392. http://
dx.doi.org/10.1172/JCI72181
Flow cytometry. Cells were acquired on FACSCanto, LSRII, and LSRFor Aziz, A., E. Soucie, S. Sarrazin, and M.H. Sieweke. 2009. MafB/c-Maf de-
tessa systems (BD) and analyzed with FlowJo software (Tree Star). The fol- ficiency enables self-renewal of differentiated functional macrophages.
lowing antibodies were used for staining cells: anti-CD11b (clone M1/70; Science. 326:867871. http://dx.doi.org/10.1126/science.1176056
eBioscience), anti-CD11c (clone N418; eBioscience), anti-CD14 (clone Chorro, L., A. Sarde, M. Li, K.J. Woollard, P. Chambon, B. Malissen,
Sa2-8; eBioscience), anti-F4/80 (clone BM8; eBioscience), anti-MHCII A. Kissenpfennig, J.-B. Barbaroux, R. Groves, and F. Geissmann.
(clone M5/114.15.2; eBioscience), anti-CD45.1 (clone A20; eBioscience), 2009. Langerhans cell (LC) proliferation mediates neonatal develop-
anti-CD45.2 (clone 104; eBioscience), anti-B220 (clone RA3-6B2; eBiosci- ment, homeostasis, and inflammation-associated expansion of the
ence), anti-CD115 (clone AFS98; eBioscience), anti-CD117 (clone 2B8; epidermal LC network. J. Exp. Med. 206:30893100. http://dx.doi
eBioscience), anti-Sca1 (clone D7; eBioscience), anti-CD48 (clone JM48-1; .org/10.1084/jem.20091586
eBioscience), anti-Gr1 (clone RB6-8C5; BioLegend), anti-CD64 (clone Davies, L.C., S.J. Jenkins, J.E. Allen, and P.R. Taylor. 2013. Tissue-resident
X54-5/7.1; BioLegend), anti-Ly6G (clone 1A8; BioLegend), anti-CD150 macrophages. Nat. Immunol. 14:986995. http://dx.doi.org/10.1038/ni
(clone TC15-12F12.2; BioLegend), anti-Ly6C (clone AL21; BD), anti- .2705
Ki67 (clone B56; BD), anti-CD4 (clone H129.19; BD), anti-CD8 (clone Epelman, S., K.J. Lavine, A.E. Beaudin, D.K. Sojka, J.A. Carrero, B. Calderon,
T. Brija, E.L. Gautier, S. Ivanov, A.T. Satpathy, et al. 2014. Embryonic
536.7; BD), anti-CD19 (clone ID3; BD), Ter119 (clone Ter119; BD),
and adult-derived resident cardiac macrophages are maintained through
NK1.1 (clone PK136; BD), anti-F4/80, anti-MerTK (clone BAF591; R&D
distinct mechanisms at steady state and during inflammation. Immunity.
Systems), anti-BrdU (clone MoBU-1; Life Technologies). Before staining,
40:91104. http://dx.doi.org/10.1016/j.immuni.2013.11.019
cells were preincubated with Fc receptor blocking antibody (clone 2.4 G2; Gautier, E.L., T. Shay, J. Miller, M. Greter, C. Jakubzick, S. Ivanov, J. Helft, A.
BD). LIVE/DEAD Fixable Aqua Dead Cell Stain kit (Life Technologies) Chow, K.G. Elpek, S. Gordonov, et al. Immunological Genome Consortium.
was used to stain dead cells. Absolute cell numbers were determined by 2012. Gene-expression profiles and transcriptional regulatory pathways
using Flow-Count Fluorosperes (Beckman Coulter) according to the manu that underlie the identity and diversity of mouse tissue macrophages. Nat.
facturers instructions. Immunol. 13:11181128. http://dx.doi.org/10.1038/ni.2419
Geissmann, F., M.G. Manz, S. Jung, M.H. Sieweke, M. Merad, and K. Ley.
Preparation of tissue samples. Peripheral blood leukocytes were isolated 2010. Development of monocytes, macrophages, and dendritic cells.
by density separation using Lympholite-M solution (Cedarlane) according to Science. 327:656661. http://dx.doi.org/10.1126/science.1178331
manufacturers instructions. For preparing single cell suspension from heart Ghigo, C., I. Mondor, A. Jorquera, J. Nowak, S. Wienert, S.P. Zahner, B.E.
and brain, the mice were perfused with PBS before collecting the tissue. For Clausen, H. Luche, B. Malissen, F. Klauschen, and M. Bajnoff. 2013.
cM, the heart was macerated and incubated with 1 mg/ml collagenase-2 Multicolor fate mapping of Langerhans cell homeostasis. J. Exp. Med.
(Worthington Biochemical Corporation) and 0.15 mg/ml DNase1 (Sigma- 210:16571664. http://dx.doi.org/10.1084/jem.20130403
Aldrich) at 37C for 30 min during constant agitation. Resulting cell suspen- Ginhoux, F., M. Greter, M. Leboeuf, S. Nandi, P. See, S. Gokhan, M.F.
sion was filtered through a 70-m mesh and erythrocytes were removed by Mehler, S.J. Conway, L.G. Ng, E.R. Stanley, et al. 2010. Fate mapping
ACK lysis. For microglia isolation a gradient of 70, 37, 30% Percoll was used. analysis reveals that adult microglia derive from primitive macrophages.
The brain tissue was homogenized and incubated with HBSS solution con- Science. 330:841845. http://dx.doi.org/10.1126/science.1194637
Goldmann, T., P. Wieghofer, P.F. Mller, Y. Wolf, D. Varol, S. Yona, S.M.
taining 2% BSA (Sigma-Aldrich), 1 mg/ml collagenase d (Roche), and
Brendecke, K. Kierdorf, O. Staszewski, M. Datta, et al. 2013. A new
0.15 mg/ml DNase1, filtered through a 70-m mesh, and resuspended in
type of microglia gene targeting shows TAK1 to be pivotal in CNS
70% Percoll, before density centrifugation (800 g for 30 min at 20C with
autoimmune inflammation. Nat. Neurosci. 16:16181626. http://dx
low acceleration and no brake). .doi.org/10.1038/nn.3531
Hashimoto, D., A. Chow, C. Noizat, P. Teo, M.B. Beasley, M. Leboeuf,
Online supplemental material. Fig. S1 depicts the full gating strategy and C.D. Becker, P. See, J. Price, D. Lucas, et al. 2013. Tissue-resident mac-
population characterization of cM. Online supplemental material is avail- rophages self-maintain locally throughout adult life with minimal contri-
able at http://www.jem.org/cgi/content/full/jem.20140639/DC1. bution from circulating monocytes. Immunity. 38:792804. http://dx.doi
.org/10.1016/j.immuni.2013.04.004
We thank Dr. Sandrine Sarrazin for critical reading of the manuscript and help Hoeffel, G.,Y.Wang, M. Greter, P. See, P.Teo, B. Malleret, M. Leboeuf, D. Low,
with statistical analysis, L. Razafindramanana for animal handling, and M. Barad, G. Oller, F. Almeida, et al. 2012. Adult Langerhans cells derive predomi-
A. Zouine, and S. Bigot for cytometry support. nantly from embryonic fetal liver monocytes with a minor contribution

JEM Vol. 211, No. 11 2157


of yolk sac-derived macrophages. J. Exp. Med. 209:11671181. http:// Schulz, C., E. Gomez Perdiguero, L. Chorro, H. Szabo-Rogers, N. Cagnard,
dx.doi.org/10.1084/jem.20120340 K. Kierdorf, M. Prinz, B. Wu, S.E.W. Jacobsen, J.W. Pollard, et al. 2012.
Jenkins, S.J., D. Ruckerl, P.C. Cook, L.H. Jones, F.D. Finkelman, N. van A lineage of myeloid cells independent of Myb and hematopoietic stem
Rooijen, A.S. MacDonald, and J.E. Allen. 2011. Local macrophage cells. Science. 336:8690. http://dx.doi.org/10.1126/science.1219179
proliferation, rather than recruitment from the blood, is a signature Serbina, N.V., and E.G. Pamer. 2006. Monocyte emigration from bone marrow
of TH2 inflammation. Science. 332:12841288. http://dx.doi.org/10 during bacterial infection requires signals mediated by chemokine recep-
.1126/science.1204351 tor CCR2. Nat. Immunol. 7:311317. http://dx.doi.org/10.1038/ni1309
Jung, S., J. Aliberti, P. Graemmel, M.J. Sunshine, G.W. Kreutzberg, A. Sher, Sieweke, M.H., and J.E. Allen. 2013. Beyond stem cells: self-renewal of differ-
and D.R. Littman. 2000. Analysis of fractalkine receptor CX3CR1 entiated macrophages. Science. 342:1242974. http://dx.doi.org/10.1126/
function by targeted deletion and green fluorescent protein reporter science.1242974
gene insertion. Mol. Cell. Biol. 20:41064114. http://dx.doi.org/10.1128/ Swirski, F.K., M. Nahrendorf, M. Etzrodt, M. Wildgruber, V. Cortez-
MCB.20.11.4106-4114.2000 Retamozo, P. Panizzi, J.L. Figueiredo, R.H. Kohler, A. Chudnovskiy,
Kierdorf, K., D. Erny, T. Goldmann, V. Sander, C. Schulz, E.G. Perdiguero, P. Waterman, et al. 2009. Identification of splenic reservoir monocytes
P. Wieghofer, A. Heinrich, P. Riemke, C. Hlscher, et al. 2013. Microglia and their deployment to inflammatory sites. Science. 325:612616. http://
emerge from erythromyeloid precursors via Pu.1- and Irf8-dependent path- dx.doi.org/10.1126/science.1175202
ways. Nat. Neurosci. 16:273280. http://dx.doi.org/10.1038/nn.3318 Tamoutounour, S., M. Guilliams, F. Montanana Sanchis, H. Liu, D. Terhorst,
Luche, H., T. Nageswara Rao, S. Kumar, A. Tasdogan, F. Beckel, C. Blum,V.C. C. Malosse, E. Pollet, L. Ardouin, H. Luche, C. Sanchez, et al. 2013.
Martins, H.-R. Rodewald, and H.J. Fehling. 2013. In vivo fate mapping Origins and functional specialization of macrophages and of conventional
identifies pre-TCR expression as an intra- and extrathymic, but not and monocyte-derived dendritic cells in mouse skin. Immunity. 39:925
prethymic, marker of T lymphopoiesis. J. Exp. Med. 210:699714. http:// 938. http://dx.doi.org/10.1016/j.immuni.2013.10.004
dx.doi.org/10.1084/jem.20122609 van Furth, R., and Z.A. Cohn. 1968. The origin and kinetics of mononuclear
Nahrendorf, M., F.K. Swirski, E. Aikawa, L. Stangenberg, T. Wurdinger, J.L. phagocytes. J. Exp. Med. 128:415435. http://dx.doi.org/10.1084/jem.128
Figueiredo, P. Libby, R. Weissleder, and M.J. Pittet. 2007. The healing .3.415
myocardium sequentially mobilizes two monocyte subsets with diver- Waskow, C., V. Madan, S. Bartels, C. Costa, R. Blasig, and H.R. Rodewald.
gent and complementary functions. J. Exp. Med. 204:30373047. http:// 2009. Hematopoietic stem cell transplantation without irradiation. Nat.
dx.doi.org/10.1084/jem.20070885 Methods. 6:267269. http://dx.doi.org/10.1038/nmeth.1309
Pinto, A.R., R. Paolicelli, E. Salimova, J. Gospocic, E. Slonimsky, D. Bilbao- Yona, S., K.-W. Kim,Y.Wolf,A. Mildner, D.Varol, M. Breker, D. Strauss-Ayali, S.
Cortes, J.W. Godwin, and N.A. Rosenthal. 2012. An abundant tissue Viukov, M. Guilliams, A. Misharin, et al. 2013. Fate mapping reveals origins
macrophage population in the adult murine heart with a distinct and dynamics of monocytes and tissue macrophages under homeostasis.
alternatively-activated macrophage profile. PLoS ONE. 7:e36814. http:// Immunity. 38:7991. http://dx.doi.org/10.1016/j.immuni.2012.12.001
dx.doi.org/10.1371/journal.pone.0036814 Zigmond, E., C. Varol, J. Farache, E. Elmaliah, A.T. Satpathy, G. Friedlander,
Porrello, E.R., A.I. Mahmoud, E. Simpson, J.A. Hill, J.A. Richardson, M. Mack, N. Shpigel, I.G. Boneca, K.M. Murphy, et al. 2012. Ly6C hi
E.N. Olson, and H.A. Sadek. 2011. Transient regenerative potential of monocytes in the inflamed colon give rise to proinflammatory effector
the neonatal mouse heart. Science. 331:10781080. http://dx.doi.org/10 cells and migratory antigen-presenting cells. Immunity. 37:10761090.
.1126/science.1200708 http://dx.doi.org/10.1016/j.immuni.2012.08.026

2158 Replacement of embryonic cardiac macrophages with age | Molawi et al.


INSIGHTS

M acrophages der ived from infiltrating monocytes mediate


<doi>10.84/jem2nsight1<do>ajem.218insght</ad>uMicelT.Hnka</>AF1UiverstyofBn</AF1>crmihael.nk@ub-ode</cr>haInsigt</doce> piNws</doct>

autoimmune myelin destruction


Macrophages mediate myelin destruction in multiple sclerosis (MS), but the origin of these cells (whether de-
< ID >j e m .2 1 8i n s gh t f j p e < / ID>

rived from tissue-resident microglial cells or infiltrating monocytes) has been widely debated. Now, Yamasaki
and colleagues distinguish these cells in a mouse model of MS and show that monocyte-derived macrophages
(MDMs) mediate myelin destruction, whereas microglia-derived macrophages (MiDMs) clear up the debris.
Previous attempts to decipher the nature and role of cells involved in autoimmune demyelination have
proven challenging. Although ontogenetically distinct, it has not been possible to distinguish macrophages
derived from tissue-resident or -infiltrating cells based on morphological features (by light microscopy) or
surface phenotype. Previous attempts to address this problem include parabiosis and bone marrow trans-
Insight from
plantation after irradiation, both strategies with substantial technical problems and limitations. Michael Heneka
Yamasaki et al. studied double chemokine receptor (CCR2-RFP+; < I D > j em . 2 1 8i ns g h t f p < / I D >

CX3CR1-GFP+) mice in the experimental autoimmune encephalo-


myelitis (EAE) mouse model of MS. Inflammatory lesions were filled with both MDMs and
MiDMs. Confocal immunohistochemistry, serial block-face scanning electron microscopy
(SBF-SEM), and subsequent 3D reconstruction revealed that myelin destruction was initiated
by MDMs, often at the nodes of Ranvier, whereas MiDMs were not detected at this site.
Disruption of MDM infiltration by CCR2 deficiency completely abolished the presence of
macrophages at the nodes of Ranvier. Gene expression profiling of both cell types at disease
onset revealed substantial differences, which correlated well with the observations obtained
by SBF-SEM. MDMs expressed genes attributable to effector functions, including those
involved in phagocytosis and cell clearance. In contrast, MiMD gene expression patterns at
disease onset were characteristic of a repressed metabolic state.
This paper sets a new standard for further studies in the field. For the first time,
MDMs and MiDMs have been clearly differentiated and their morphological relation-
ship to axoglial structures has been analyzed. The finding that MDMs rather than
MiDMs initiate myelin destruction at disease onset should enable this cell population to
be targeted more effectively in future. The next stage is to verify these findings in human
tissue. Future research should also assess further time points over the entire disease
Nodes of Ranvier represent a prime course, in particular to exclude that MiDMs do not join MDMs at the node of Ranvier
site of attack for MDMs at the onset at later stages of disease. A precise distinction between local and infiltrating cell popula-
of EAE. This 3D reconstruction of SBF-
tions may also contribute to a better understanding of pathogenesis in other CNS disor-
SEM images shows a monocyte-derived
macrophage encircling the node of
ders such as stroke and brain trauma and will hopefully lead to the development of new
Ranvier, as shown by the two primary therapeutic strategies.
processes (white and black arrows). Yamasaki, R., et al. 2014. J. Exp. Med. http://dx.doi.org/10.1084/jem.20132477.

Michael T. Heneka, University of Bonn: michael.heneka@ukb.uni-bonn.de


Article

Differential roles of microglia and monocytes


in the inflamed central nervous system
Ryo Yamasaki,1 Haiyan Lu,1 Oleg Butovsky,5 Nobuhiko Ohno,2
Anna M. Rietsch,1 Ron Cialic,5 Pauline M. Wu,2 Camille E. Doykan,2
Jessica Lin,1,6 Anne C. Cotleur,1 Grahame Kidd,2 Musab M. Zorlu,1,7
Nathan Sun,8 Weiwei Hu,2,9 LiPing Liu,1 Jar-Chi Lee,3 Sarah E. Taylor,10
Lindsey Uehlein,1,6 Debra Dixon,1,11 Jinyu Gu,1 Crina M. Floruta,1,12 Min Zhu,1
Israel F. Charo,13 Howard L. Weiner,5 and Richard M. Ransohoff 1,4,11
1Neuroinflammation Research Center and 2Department of Neurosciences, Lerner Research Institute; 3Department
of Quantitative Health Sciences; and 4Mellen Center for Multiple Sclerosis Treatment and Research, Neurological Institute,
Cleveland Clinic, Cleveland, OH 44106
5Center for Neurological Diseases, Brigham and Womens Hospital, Harvard Institutes of Medicine, Boston, MA 02115
6Ohio State University College of Medicine, Columbus, OH 43210
7Hacettepe University Faculty of Medicine, 06100 Ankara, Turkey
8Vanderbilt University, Nashville, TN 37235
9Department of Pharmacology, School of Basic Medical Sciences, Zhejiang University, Hangzhou, 310058 Zhejiang, China
10Case Western Reserve University, School of Medicine, Cleveland, OH 44106
11Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44106
12Baylor University, Waco, TX 77030
13Gladstone Institute of Cardiovascular Disease, University of California, San Francisco, San Francisco, CA 94158

In the human disorder multiple sclerosis (MS) and in the model experimental autoimmune
encephalomyelitis (EAE), macrophages predominate in demyelinated areas and their num-
bers correlate to tissue damage. Macrophages may be derived from infiltrating monocytes
or resident microglia, yet are indistinguishable by light microscopy and surface phenotype.
It is axiomatic that T cellmediated macrophage activation is critical for inflammatory
demyelination in EAE, yet the precise details by which tissue injury takes place remain
poorly understood. In the present study, we addressed the cellular basis of autoimmune
demyelination by discriminating microglial versus monocyte origins of effector macro-
phages. Using serial block-face scanning electron microscopy (SBF-SEM), we show that
monocyte-derived macrophages associate with nodes of Ranvier and initiate demyelination,
whereas microglia appear to clear debris. Gene expression profiles confirm that monocyte-
derived macrophages are highly phagocytic and inflammatory, whereas those arising from
microglia demonstrate an unexpected signature of globally suppressed cellular metabolism
at disease onset. Distinguishing tissue-resident macrophages from infiltrating monocytes
will point toward new strategies to treat disease and promote repair in diverse inflamma-
tory pathologies in varied organs.

CORRESPONDENCE Blood-derived monocytes and resident microglia capacities (Gautier et al., 2012; Chiu et al., 2013;
Richard M. Ransohoff: can both give rise to macrophages in the cen- Butovsky et al., 2014).
ransohr@ccf.org Microglia and monocytes are ontogeneti-
tral nervous system (CNS). In tissue sections,
Abbreviations used: CNS, macrophages derived from these two distinct cally distinct: microglia derive from yolk-sac pro-
central nervous system; EAE, precursors are indistinguishable at the light genitors during embryogenesis (Ginhoux et al.,
experimental autoimmune microscopic level both morphologically and by 2010; Schulz et al., 2012), whereas monocytes
encephalomyelitis; MDM,
monocyte-derived macrophage; surface markers. Using flow cytometry, microglia- continuously differentiate throughout postnatal
MiDM, microglia-derived mac- and monocyte-derived macrophages can be life from bone marrow hematopoietic stem cells
rophage; MS, multiple sclerosis; isolated separately from CNS tissue lysates and 2014 Yamasaki et al. This article is distributed under the terms of an Attribution
SBF-SEM, serial block-face
scanning electron microscopy.
expression profiling suggests distinct functional NoncommercialShare AlikeNo Mirror Sites license for the first six months
after the publication date (see http://www.rupress.org/terms). After six months
it is available under a Creative Commons License (AttributionNoncommercial
R. Yamasaki, H. Lu, and O. Butovsky contributed equally to Share Alike 3.0 Unported license, as described at http://creativecommons.org/
this paper. licenses/by-nc-sa/3.0/).

The Rockefeller University Press $30.00


J. Exp. Med. 2014 Vol. 211 No. 8 1533-1549 1533
www.jem.org/cgi/doi/10.1084/jem.20132477
(HSCs), which require the transcription factor Myb. Microg- were isolated from CNS and analyzed by flow cytometry
lial precursors are Myb independent, and microglia self-renew using cells from double-heterozygous Ccr2rfp::Cx3cr1gfp mice
independently of bone marrow HSCs (Gomez Perdiguero with EAE, GFP was expressed by CD45dim/Ly6C microglia,
et al., 2013). Distinct developmental origin and renewal mech- whereas RFP was restricted to CD45high/Ly6C+ monocytes
anisms imply that monocyte-derived macrophages (MDMs) (Saederup et al., 2010; Mizutani et al., 2012). These findings
and microglia-derived macrophages (MiDMs) might exert dif- suggested an approach to clarifying distinct roles of MDMs
ferent functions in pathological processes. Microglia represent and MiDMs in EAE based on differential expression of GFP
one instance of tissue-resident macrophages, which reside in all and RFP reporters. Here, we use that strategy to extend pre-
organs. Studying the CNS as compared with other organs vious findings and address the hypothesis that MDMs and
may carry advantages for distinguishing tissue-resident my- MiDMs exert different functions in neuroinflammation. We
eloid cells from infiltrating monocytes during disease, as there detected detailed ultrastructural characterization of MDMs
is virtually no background trafficking of monocytes in the and MiDMs at EAE onset.
CNS parenchyma of healthy animals. Unexpectedly, this approach provided insight into the cel-
In EAE, which models inflammatory aspects of MS lular basis for autoimmune demyelination, which has remained
(Williams et al., 1994; Ransohoff, 2012), macrophages dominate obscure despite >80 yr of study in the EAE model. Here we
the inflammatory infiltrates and their numbers correlate to provide evidence that MDMs initiate demyelination, often at
EAE severity (Huitinga et al., 1990, 1993; Ajami et al., 2011). nodes of Ranvier. In contrast, phagocytic microglia appear rela-
However the cellular mechanisms by which macrophages tively inert at disease onset. Results from expression profiling
promote disease progression are uncertain. Whether MiDMs provided insight into mechanisms and signaling pathways un-
or MDMs are functionally distinct and whether the two cell derlying the disparate effector properties of MDMs and MiDMs
types differentially initiate demyelination or promote repair in EAE.The distinct functions of tissue-resident myeloid cells
(Steinman et al., 2002) also remains elusive (Bauer et al., 1995). as compared with infiltrating macrophages broadly underlie
In MS autopsy tissues, prominent macrophage accumulation disease pathogenesis in manifold circumstances and also hold
correlates with active demyelination (Ferguson et al., 1997; promise for innovative treatment strategies.
Trapp et al., 1998). Based on kinetics of cell accumulation and
differential marker expression, its estimated that 3050% of RESULTS
activated macrophages in active MS lesions derive from mi- In the CNS of mice of EAE, MDMs and MiDMs
croglia (Brck et al., 1995; Trebst et al., 2001). Therefore, dif- exhibit different accumulation kinetics
ferential functions of MDMs and MiDMs are relevant for The histological strategy in this study is shown in Table 1. At
human demyelinating disease. onset of EAE, two pools of CD11b+ mononuclear phagocytic
To date, no research techniques have permitted distinction cells (putative red MDMs and green MiDMs) predominated
between monocytes and microglia in CNS tissue without ir- in spinal cord (Fig. 1 A), indicating that fluorochrome mark-
radiation chimerism or parabiosis, techniques that confound ers could be distinguished at this time point. Using cells iso-
interpretation or impose practical limitations (Ajami et al., lated from Ccr2rfp/+::Cx3cr1gfp/+ spinal cords at disease onset,
2007, 2011; Ransohoff, 2007). When F4/80+ macrophages flow cytometry demonstrated distinct expression of RFP and

Table 1. Histology analysis strategy


Method Purpose Finding
Confocal analysis of 0.2 mm optical To distinguish MDMs (RFP+)from MDMs and MiDMs can be distinguished by cell volume and primary
sections (n = 104 cells). MiDMs (GFP+). processes.
SBF-SEM inspection in 0.2 mm sections To detect MDMs and MiDMs in SBF- Using criteria detected in the previous step, it is possible to distinguish
from 14 lesions, 7 mice at EAE onset. SEM using cell volume and process MDMs and MiDMs in SBF-SEM images.
criteria.
SBF-SEM inspection of ultrastructure To detect ultrastructural characteristics MDMs and MiDMs show characteristic ultrastructural differences
of MDMs and MiDMs. of MDMs and MiDMs. regarding their mitochondria, nuclei, osmiophilic granules and
microvilli.
Quantification of relation of MDMs To determine relationship of MDMs Most (55/75; 73%) axoglial units are contacted in limited fashion by
(n = 169) and MiDMs (n = 86) to and MiDMs to axoglial units and MDMs and MiDMs. If one cell type is present (20/75 cells), its nearly
axoglial units (n = 29 intact axons, characterize presence of myelin debris. always (18/20 segments) MDMs.
46 demyelinated axons).
Reconstruction of 3D shape of four To detect relationship of MDMs with In all, 49 MDMs interacting with axoglial units in absence of nearby
representative MDMs at axoglial axoglial units at EAE onset. MiDMs, 2-3 MDMs were attached to each (n = 18) axoglial unit.
units. MDMs have close relationship with nodes of Ranvier (7 MDMs/75
axoglial units). 3D reconstructions showed four representative MDMs
at axoglial units: show one carrying out active demyelination, three at
nodes of Ranvier.
1534 Distinguishing microglia and monocytes in EAE CNS | Yamasaki et al.
Ar ticle

GFP by F4/80+/CD45high MDMs and F4/80+/CD45dim of MDMs and MiDMs could be assayed and early events in
MiDMs, respectively (Fig. 1 B). Enumeration of cells recovered the demyelinating disorder could be explored.
from cell sorting using F4/80+RFP+ as MDMs gate and F4/
80+GFP+ as MiDMs gate indicated that MDMs and MiDMs Morphological features distinguish
showed equal numbers at disease onset when explosive MDM MDMs and MiDMs at EAE onset
accumulation occurred. MiDM expansion began at peak Immunofluorescence staining for RFP and GFP in spinal cord
(Fig. 1 B). At recovery, MiDMs were found near preonset at EAE onset showed that red MDMs exhibited elongated or
numbers as MDM frequency continued to decline, which is spindle shape, whereas green MiDMs showed a process-bearing
compatible with previous studies (Ajami et al., 2011). There- morphology (Fig. 1 C). Quantification in 3D reconstructions
fore, there were unequal numbers of MiDMs/MDMs before from 0.2-m confocal z-stack images showed that MiDMs
and after disease onset (Fig. 1 B). Morphological analyses and exhibited much larger size than MDMs along with multiple
definitions of relations between myeloid cells and axoglial primary processes, which were sparse in MDMs (Fig. 1 D).
units were conducted at disease onset so that equal numbers Several 3D shape parameters also discriminated between MDMs

Figure 1. MDMs and MiDMs exhibit different time courses of accumulation in the CNS of mice with EAE and morphological characteristics
can distinguish them. (A) Immunohistochemistry shows expression of CD11b for red RFP+ MDMs and green GFP+ MiDMs in the spinal cords of
Cx3cr1gfp/+::Ccr2rfp/+ mice at EAE onset. Bars: 25 m. We studied 6 mice at EAE onset from 3 EAE inductions. In each EAE induction, 810 mice were used and
2 mice were selected from each induction. (B) Flow cytometric analysis of CCR2-RFP+ and CX3CR1-GFP+ populations in cells gated for F4/80 expression
(top); CD45 expression of F4/80+RFP+ MDMs and F4/80+GFP+ MiDMs populations (middle); and MDMs and MiDMs numbers at EAE onset, peak, and recovery
(bottom). We studied 3 mice for naive groups; 12 for onset; 15 for peak; 13 for recovery from 5 EAE inductions. For each induction, 810 mice were used
and 23 mice were selected at each time point (onset, peak, and recovery) for analysis. (C) Confocal microscopy assessment of myeloid cell morphology in
lumbar spinal cord from mice at EAE onset. We studied 54 MDMs and 51 MiDMs of 5 EAE onset mice from 3 EAE inductions for (CE); 2 sections/mouse;
46 cells/section; 812 cells/mouse. In each EAE induction, 810 mice were induced and 12 EAE onset mice were selected from each experiment. Bars,
25 m. (D) Cell volumes of 500 m3; surface areas of 1,000 m2; primary process numbers 3 or 5; solidity3D of 0.4; and Formfactor3D of 0.3 discrimi-
nate between MDMs and MiDMs. (E) Model plot of cell volume against primary process number to distinguish MDMs (red symbols and pink area) from
MiDMs (green symbols and green area).

JEM Vol. 211, No. 8 1535


and MiDMs (Fig. 1 D). We observed scant overlap of several in MDMs and MiDMs (unpublished data). MDMs had bi-
values between MDMs and MiDMs (Fig. 1 D), and entirely lobulated or irregular nuclei, whereas MiDMs had round nu-
nonoverlapping distributions for cell volume and primary clei (unpublished data). MDMs, but not MiDMs, frequently
processes (Fig. 1 E). contained osmiophilic granules and microvilli (unpublished
data). Collectively, these ultrastructural features provided con-
MDMs and MiDMs exhibit differentiating firmatory ultrastructural characteristics to distinguish MDMs
ultrastructural characteristics at EAE onset from MiDMs.
We used confocal microscopy in 0.2-m optical sections to
correlate structural features of MDMs and MiDMs with RFP MDMs initiated demyelination at EAE onset
or GFP fluorescence, as a bridge to characterizing cells in 0.2 m Results from confocal and EM analysis yielded a secure basis
SBF-SEM images (Table 1). Using this approach (Fig. 1 E), for examining the relationships of MDMs and MiDMs to ax-
MDMs and MiDMs were identified by estimating volume oglial units at EAE onset (n = 7 mice; 14 lesions) using serial
and counting primary processes. Volume estimations came block-face scanning electron microscopy (SBF-SEM), as pre-
from multiplying the midcell area by the number of sections in sented diagrammatically (Table 1). We quantified contacts
which the cell was identified. In electromagnetic (EM) images, made by MDMs (n = 169) and MiDMs (n = 86) with axoglial
quantitative analysis also demonstrated differentiating ultrastruc- units (n = 75; Fig. 2), and observed that most (55/75; 73%) of
tural characteristics for mitochondria, nuclei, cytoplasmic os- all segments (both intact and demyelinated) contacted both
miophilic granules and microvilli (unpublished data). MDMs MDMs and MiDMs (Fig. 2). Where only one myeloid cell
had shorter, thicker mitochondria than MiDMs (unpublished type was present (20/75; 27%), nearly all axoglial units made
data). Total mitochondrial numbers and volumes were equal contacts to MDMs (Fig. 2). In particular, 8/29 intact and 10/46

Figure 2. SBF-SEM shows MDMs initiating demyelination at EAE onset. Quantitation of MiDMs and MDMs interacting with axoglial units in SBF-
SEM images of CNS at EAE onset. Intact (69%) and demyelinated (76%) segments interacted with MDMs and MiDMs. Red and pink, MDMs; green and
light green, MiDMs; yellow, both MDMs and MiDMs. We studied 29 intact axon segments, 46 demyelinated axon segments, 86 MiDMs, and 169 MDMs in
14 lesions of 7 EAE onset mice from 3 EAE inductions as follows: 810 mice were immunized at each experiment and 23 onset mice were selected from
each induction.

1536 Distinguishing microglia and monocytes in EAE CNS | Yamasaki et al.


Ar ticle

demyelinated axoglial units were contacted solely by MDMs. inside the MDMs (Fig. 3 A). Remaining myelin was undergoing
We found 23 MDMs attached to each of the 18/20 (90%) vesicular breakdown (Fig. 3 A). In contrast, a nearby MiDM
axoglial units where only MDMs were present (Fig. 2). More encompassed a large fragment of myelin debris (Fig. 3 B) and
than half of all analyzed MDM and MiDM cells (n = 255 contacted the nearby MDMs (Fig. 3 B), but made minimal
total) contained myelin debris, regardless of whether axon connection to the axoglial unit (Fig. 3 B). In our SBF-SEM
segments were intact or demyelinated (Fig. 2). Of the MDMs data, only MDMs seemed to be implicated in active damage
found in sole contact with axoglial units, virtually all (>90%) to myelin. These observations suggested that MDMs initiated
MDMs contained myelin when found in sole contact with a demyelination at the onset of EAE.
demyelinated axon (Fig. 2). These findings motivated evalua-
tion of relationships of MDMs to axoglial units by 3D recon- MDMs surrounded apposed and invaded
struction of SBF-SEM image stacks. nodes of Ranvier at EAE onset
MDMs frequently exhibited morphological characteristics We analyzed axoglial units to examine the nature of contacts
suggesting an involvement in active demyelination. Reconstruc- with myeloid cells. Unexpectedly, 7/75 (9%) of axoglial units
tion of one representative image stack shows MDMs with demonstrated MDMs attached to nodes of Ranvier. In each
large intracellular myelin inclusions tightly encircling a partially case, the contact between MDMs and node appeared to be
demyelinated axon (Fig. 3 A). The myelin peeled away from pathogenic. One representative monocyte surrounded a node
the axon remained in continuity with a large myelin inclusion of Ranvier with two microvilli interposed between myelin

Figure 3. SBF-SEM shows example of


MDM-initiating demyelination at EAE
onset. (A) Representative MDMs encircles the
axoglial unit. A myelin ovoid within an intra-
cellular phagolysosome shows physical conti-
nuity with myelin remaining attached to an
axoglial unit which is undergoing active de-
myelination. In serial images, disrupted myelin
shows continuity from outside to inside the
MDM. (B) Rotated view from B demonstrating
MDM-extensive attachment to axoglial unit
and MiDM nearby with limited attachment to
axon. A, axon; m, myelin; c, cytosol; n, nucleus.
Red, MDM cytosol; green, MiDM cytosol;
yellow: nuclei; blue, myelin and myelin debris;
gray, axoplasm; red line, MDM plasma mem-
brane. We studied 14 lesions from 7 EAE on-
set mice from 3 EAE inductions as follows:
810 mice were immunized at each experi-
ment and 23 EAE onset mice were selected
from each induction. Bar, 2 m.

JEM Vol. 211, No. 8 1537


and axolemma near the paranode complex (Fig. 4 A). The ax- address the role of MDMs in demyelination at EAE onset, we
oglial unit appeared otherwise healthy and no myelin debris investigated clinical characteristics in relation to node pathology
was found in the MDM cytosol. This observation suggested and demyelination in Ccr2rfp/rfp::Cx3cr1gfp/+ mice in which
that initial MDMaxoglial contacts might occur at nodes of MDMs were virtually absent from inflamed EAE tissues and
Ranvier. Further, we detected an intratubal (Stoll et al., 1989) replaced in large part by neutrophils (Saederup et al., 2010).We
MDMs with myelin debris interposed between compact my- observed equivalent magnitude of weight loss in Ccr2rfp/+::
elin and axolemma near a node of Ranvier (Fig. 4 B). Addi- Cx3cr1gfp/+ and Ccr2rfp/rfp::Cx3cr1gfp/+ mice at preonset and
tionally we identified an MDM apposed to a node of Ranvier onset stages of EAE, showing that CCR2 deficiency did not
and actively phagocytizing myelin (Fig. 4 C). At this node, affect systemic inflammation in this model (Fig. 5 A). There was
paranode loops were disrupted and surrounded by MDM cy- a moderate delay in disease onset (Fig. 5 B) and slight reduction
tosol (Fig. 4 C), indicating likely involvement in damaging my- in EAE onset severity (Fig. 5 A) in Ccr2rfp/rfp::Cx3cr1gfp/+ mice.
elin near the node. No MiDMs contacted nodes of Ranvier. SBF-SEM was used to evaluate nodal pathology, myeloid
cell relations to axoglial units and demyelination at and before
Nodal pathology without demyelination EAE onset. In three distinct tissues from individual Ccr2rfp/+
at EAE onset in Ccr2rfp/rfp::Cx3cr1gfp/+ mice ::Cx3cr1gfp/+ mice with EAE preonset, we found five MDMs
We interpreted our ultrastructural findings to indicate that attached to disrupted nodes of Ranvier. In an equivalent sam-
MDMs recognized altered nodal structure and initiated demy- ple of EAE tissues from three Ccr2rfp/rfp::Cx3cr1gfp/+ mice, only
elination at EAE onset. CCR2 is essential for monocyte recruit- one MDM was found in contact with a node of Ranvier, despite
ment to CNS tissues during immune-mediated inflammation the presence of disrupted nodes in proximity to neutrophils.
(Fife et al., 2000; Izikson et al., 2000; Savarin et al., 2010). To One representative MDM from Ccr2rfp/+::Cx3cr1gfp/+ tissue

Figure 4. MDMs surrounded, apposed,


and invaded nodes of Ranvier at EAE
onset. (A) SBF-SEM images and 3D reconstruc-
tion of SBF-SEM images of MDMs with a node
of Ranvier. White and black arrow: microvil-
lus. (B) SBF-SEM images and 3D construction
of intratubal MDMs with demyelinated axon
and node of Ranvier. (C) SBF-SEM images and
3D reconstruction of an MDM with intracel-
lular myelin debris apposed to a node of Ran-
vier. Red, MDM cytosol; yellow, nucleus; blue,
myelin; gray, axoplasm. M, myelin; c, cytosol.
red line, MDM plasma membrane. We studied
14 lesions from 7 EAE onset mice collected as
follows: 810 mice were immunized at each
induction and 23 EAE onset mice were
selected from each immunization. Bar, 2 m.

1538 Distinguishing microglia and monocytes in EAE CNS | Yamasaki et al.


Ar ticle

Figure 5. Nodal pathology without demyelination at EAE onset in Ccr2rfp/rfp::Cx3cr1gfp/+ mice. (A) Magnitude of weight loss in Ccr2rfp/+::Cx3cr1gfp/+ and
Ccr2rfp/rfp::Cx3cr1gfp/+ mice at preonset and onset stages of EAE. Clinical score in Ccr2rfp/+::Cx3cr1gfp/+ and Ccr2rfp/rfp::Cx3cr1gfp/+ mice at EAE onset stage. (B) Days
at disease preonset and onset stages of EAE. We studied 28 Ccr2rfp/+::Cx3cr1gfp/+ mice and 26 Ccr2rfp/rfp::Cx3cr1gfp/+ mice for A and B. Data were collected from
12 EAE inductions in Ccr2rfp/+::Cx3cr1gfp/+ mice and 19 EAE inductions in Ccr2rfp/rfp::Cx3cr1gfp/+ mice as follows: 810 mice were immunized at each induction and
13 EAE recovery mice were selected from each immunization. **, P < 0.01; ***, P < 0.001. (C) SBF-SEM imaging of MDMs with nodes of Ranvier phagocytosis in
Ccr2rfp/+::Cx3cr1gfp/+ mice at EAE preonset. Pink, MDM cytosol; red arrow, myelin inclusion of MDM connecting to the node of Ranvier. We studied 3 tissues from 3
Ccr2rfp/+::Cx3cr1gfp/+ EAE mice at preonset stage from 3 EAE inductions: 810 mice were immunized at each experiment and one EAE preonset mouse was selected
from each induction. Bar, 2 m. (D) SBF-SEM of disrupted nodes (black arrows) in preonset spinal cord tissues of Ccr2rfp/rfp::Cx3cr1gfp/+ mice. Bar, 2 m. (E) SBF-
SEM of neutrophil is with myelin phagocytosis from internode at preonset stage of Ccr2rfp/rfp::Cx3cr1gfp/+ mouse. Blue, neutrophil cytosol. For DE, we studied
three tissues from three Ccr2rfp/rfp::Cx3cr1gfp/+ EAE mice at preonset stage from 3 EAE inductions: 810 mice were immunized at each experiment and one EAE
preonset mouse was selected from each induction. Bar: 2 m. (F) Histochemical staining and with aurohalophosphate complexes (black gold staining) and quanti-
fication of demyelinated area of Ccr2rfp/+::Cx3cr1gfp/+ mice and Ccr2rfp/+::Cx3cr1gfp/+ mice. We studied 5 naive Ccr2rfp/+::Cx3cr1gfp/+ mice, 5 naive Ccr2rfp/rfp
::Cx3cr1gfp/+ mice, 5 onset Ccr2rfp/+::Cx3cr1gfp/+ mice, and 5 onset Ccr2rfp/rfp::Cx3cr1gfp/+ mice from 3 EAE inductions as follows: 810 mice were immunized at each
experiment and 12 onset mice were selected from each induction. **, P < 0.01. Bar: 250 m.

JEM Vol. 211, No. 8 1539


Figure 6. Inflammatory signature in MiDMs versus MDMs in the CNS of Cx3cr1gfp/+::Ccr2rfp/+ mice with EAE. (A) Quantitative nCounter expres-
sion profiling of 179 inflammation related genes was performed in CNS-derived GFP+ microglia and RFP+ recruited monocytes from naive and EAE mice
at onset, peak and recovery stages. Each row of the heat map represents an individual gene and each column an individual group from pool of 5 mice at
each time point. The relative abundance of transcripts is indicated by a color (red, higher; green, median; blue, low). For AH, we studied five mice in each
time point (onset, peak, recovery) from 3 EAE inductions; 810 mice were immunized in each induction. (B) Heat map of differentially expressed microglia

1540 Distinguishing microglia and monocytes in EAE CNS | Yamasaki et al.


Ar ticle

having concave nucleus (Fig. 5 C, left) had multiple intracellu- We noted a subset of genes that were expressed in microglia
lar myelin inclusions, one of which (Fig. 5 C, left middle) was and highly regulated in MiDMs during EAE, but not ex-
physically connected to a myelin sheath (Fig. 5 C, right mid- pressed at all in monocytes or MDMs (Fig. 6, A and B). Con-
dle) at a paranode (Fig. 5 C, right), indicating active ongoing versely, a subset of MDM-enriched genes were dynamically
demyelination at a node of Ranvier. By distinct contrast, EAE regulated in monocytes and MDMs but not in microglia
onset tissues of Ccr2rfp/rfp::Cx3cr1gfp/+ mice were characterized (Fig. 6, A and B). MDM-enriched genes were sharply up-
by nodal pathology often without cellular infiltrates (Fig. 5 D). regulated from naive monocytes to onset and peak-stage
In one instance, we detected a neutrophil abstracting myelin MDMs (Fig. 6 B), descending toward naive levels during
from the myelin internode (Fig. 5, left and right) despite a recovery (Fig. 6 B). In contrast, MiDM-enriched genes
nearby disrupted node (Fig. 5, left) in tissues from a Ccr2rfp/rfp:: were strongly expressed in naive cells, almost uniformly si-
Cx3cr1gfp/+ mouse. Importantly, there was no evidence for lenced at onset, and began a return toward naive levels at peak
neutrophil recognition of disrupted nodes of Ranvier. We in- and recovery (Fig. 6 B). Comparing MDM-enriched genes
terpreted these observations to suggest that MDMs specifically with MiDM-enriched genes showed that MDMs were
recognized nodal components to initiate demyelination, and that more likely to express effector functions, including secreted
absence of MDMs at disrupted nodes of Ccr2rfp/rfp::Cx3cr1gfp/+ factors and surface molecules (18/28; 64.3% of MDM-
mice with EAE was caused by the virtual absence of infiltrat- enriched genes encoded effector functions; Fig. 6 C: and
ing monocytes (Saederup et al., 2010). Table S1, purple genes). In contrast, only 18/48 (37.5%) of
To quantify the outcome of these ultrastructural differences, MiDM-enriched genes encoded effector functions (Fig. 6 D,
we monitored demyelination using histochemical staining with Table S1, purple genes). These observations indicated that
aurohalophosphate complexes at disease onset in Ccr2rfp/rfp:: MiDMs and MDMs exhibited markedly distinct expression
Cx3cr1gfp/+ and Ccr2rfp/+::Cx3cr1gfp/+ mice. Demyelination was profiles during EAE.
significantly reduced at EAE onset in CCR2-deficient mice
(Fig. 5 F), indicating the importance of MDM recognition of Differential expression of macrophage
disrupted nodes for efficient inflammatory demyelination. Fur- effector functions by MiDMs and MDMs
thermore, as nodal pathology was equivalent in Ccr2rfp/rfp:: Our ultrastructural analysis of myeloid cells in EAE focused
Cx3cr1gfp/+ (Fig. 5 D) and Ccr2rfp/+::Cx3cr1gfp/+ mice at the on myeloid cell relationships to tissue elements. Expression
preonset stage of EAE, the results suggested that inflammatory profiling also addressed the cytokine and growth factor output
nodal disruption could be reversible if MDMs were prevented of MiDMs and MDMs, potentially providing insight into disease
from initiating demyelination at those sites. pathogenesis.We used k-means clustering to discriminate five
distinct patterns of MiDM gene expression during the course
Expression profiling demonstrates differential of EAE (Fig. 6, E and F). The red, blue and green groups in-
MiDMs and MDMs gene expression creased in MiDMs at onset, peak, and recovery, respectively.
across the time course of an EAE attack Red group genes involved several surface molecules. Green
We reasoned that different phenotypes (Fig. 1) and effector group genes, up-regulated at onset and transiently further
properties (Figs. 24) of MDMs and MiDMs should be re- up-regulated at peak, were comprised mainly of complement-
flected in distinct gene expression profiles in the dynamic CNS system elements (C3aR1; C4a, C1qa, C1qa, C3, and Cfb);
microenvironment during EAE. To address this hypothesis, mononuclear cellspecific chemokines (CCl2, 3, 4, 5, 7, and
nCounter digital multiplexed gene expression analysis (Kulkarni, CXCL9); proliferation related genes ( fos, jun, myc, and CSF1);
2011) was performed using directly ex vivo naive microglia and acute inflammationrelated genes (IL1a, IL1b, TNF,
and splenic F4/80+ macrophages (here termed monocytes and CEBP, STAT1). Cell growthrelated genes expressed at this
considered similar to microglia by expression profiling; Gautier time point correlated to reported patterns of microglial pro-
et al., 2012), as well as flow-sorted MiDMs or MDMs across the liferation during EAE (Ajami et al., 2011). Blue group genes
time course of an EAE attack. Microglia and MiDMs clus- up-regulated at recovery included heterogeneous cytokines
tered together during unsupervised hierarchical clustering, as (IFN-, IFN-, TGFB3, IL2, IL3, IL4, IL12, IL12,
did monocytes and MDMs (Fig. 6, A and B). In both MiDMs PDGFA, CSF2, and CXCL2). Both yellow group and golden
and MDMs, naive and recovery-stage expression profiles were group genes were strongly expressed in naive microglia, re-
more alike than were onset and peak-stage profiles (Fig. 6, duced drastically at onset, and either returned to preEAE lev-
A and B) suggesting a return to homeostasis at EAE recovery. els during recovery (yellow) or failed to do so (golden).These

and monocyte genes. (C) Enriched monocyte genes as compared with resident microglia. Bars represent fold changes of gene expression across naive and
all disease stages versus resident microglia. (D) Enriched microglia genes as compared with recruited monocytes. Bars represent fold changes of gene
expression across naive and all disease stages versus recruited monocytes. (EH) K-means clustering of inflammation genes in resident microglia and
recruited monocytes. K-means clustering was used to generate 5 disease stagerelated clusters in MiDMs. Heat map (E) and expression profile (F) of in-
flammation genes in MiDMs are shown by generated clusters. MDM expression matrix overlaid on microglial based clusters shows (G) heat map and
(H) expression profile.

JEM Vol. 211, No. 8 1541


genes included a large spectrum of intracellular signaling com- other time points as well. However, a substantial minority of
ponents from the MAP-kinase pathways, as well as TGF and genes both for MDMs and MiDMs showed some dissonant
receptor, both of which are implicated in the nave microglial time points, at which a previously down-regulated gene might
phenotype (Butovsky et al., 2014). show up-regulation (unpublished data). We show this subset of
These five gene groups were also analyzed for MDM ex- recovered genes in Fig. 8. In virtually every case (Fig. 8, AD),
pression patterns during EAE (Fig. 6, G and H). None of the these dissonant compensatory changes took place during recov-
gene groups showed coordinate regulation patterns in MDMs, ery and almost always showed an increase in a gene that had
as were observed in MiDMs (Fig. 6 H). This observation un- been down-regulated during onset and peak. Both MiDMs
derscored disparate responses of MiDMs and MDMs to the (Fig. 8 B) and MDMs (Fig. 8 D) demonstrated this pattern of
inflammatory CNS microenvironment of EAE, despite their gene-expression kinetics.
being present in close proximity (Fig. 1 A).
Convergent and divergent responses to upstream
Expression patterns at EAE onset regulatory signaling by MiDMs and MDMs
in relation to MiDM and MDM function Translation of observations made using expression profiles can
To determine whether gene expression patterns could be in- be enabled through identification of upstream regulators. We
formative for understanding the relationships of cells to ax- used Ingenuity IPA software to identify putative upstream reg-
oglial elements in tissues at EAE onset, we interrogated naive ulators of the gene expression alterations demonstrated by
versus onset MDM and MiDM gene expression related to MiDMs and MDMs at disease onset. Putative regulatory ele-
cellular functions (Fig. 7). MiDMs showed highly significant ments were then grouped in signaling modules and subjected
up-regulation of functions associated with cell movement, che- to pathway analysis. Cell motility pathways were clearly differ-
moattraction, and migration (Fig. 7 B). In the Ingenuity IPA ent in MiDMs and MDMs (unpublished data). Core elements
database, the terms cell movement, chemoattraction, and mi- such as RhoA (Xu et al., 2009) were regulated divergently and
gration indicated production of chemokines such as CCL2, associated signaling components were predicted to be enhanced
CCL3, CCL4, CCL5, and CCL7, which are up-regulated at in MDMs but depressed in MiDMs, consistent with our pheno-
onset and further increased at peak (Fig. 6, E and F, green typic characterization using SBF-SEM. Both HIF-1 (Fig. 9 A)
group and genes). In other respects, MiDMs exhibited a re- and TNF pathways (not depicted) were also differentially reg-
pressed metabolic and activation phenotype by comparison ulated in MiDMs and MDMs. By contrast, type I IFN pathway
to naive microglia (Fig. 7 B) including proliferation, RNA (Fig. 9 B) was regulated virtually identically in MiDMs and
metabolism, cytoskeletal organization, microtubule dynamics, MDMs. Collectively, these data suggest that HIF-1 and TNF
extension of processes, phagocytosis and generation of reac- signaling may partly drive pathogenic properties of MDMs.
tive oxygen species. Additionally, these data indicated that the separate ontogeny
MDMs showed up-regulation of functions associated to of microglia and monocytes will lead, probably by epigenetic
macrophages, including phagocytosis, calcium signaling, pro- influences, to divergent responses to some but not all environ-
duction of prostanoids, adhesion, autophagy, and cell clearance mental stimuli, with phenotypic consequences according to
(Fig. 7 B).This pathway analysis corresponded well to effector the CNS microenvironment.
properties displayed by MDMs in our SBF-SEM analysis
(Figs. 24). No functions were reported to be down-regulated DISCUSSION
in MDMs at EAE onset as compared with naive monocytes. In this study, we developed a novel strategy to discriminate
A comprehensive listing (Table S1) of all genes regulated MDMs from MiDMs. We used SBF-SEM to address the de-
by at least twofold in MiDMs or MDMs as compared with tailed relationships of MiDM and MDM to axoglial units in
expression levels in naive mice affirmed and extended these the spinal cords of mice at EAE onset and expression profiling
interpretations. At EAE onset when SBF-SEM analyses were to examine potential mechanisms. Selection of the EAE disease
performed, MiDMs predominantly suppressed the distinctive model ensured that both recruited monocytes and resident mi-
gene expression pattern which correlates to their unique phe- croglia were exposed to the same intensely inflammatory en-
notype (Chiu et al., 2013), reflected by the observation that vironment to increase the likelihood that ambient conditions
MiDMs down-regulated far more genes than were up-regulated could activate these two myeloid cell types toward a convergent
(Table S1). In contrast, MDMs up-regulated far more genes inflammatory phenotype. Instead, we found strikingly diver-
than did MiDMs and up-regulated more transcripts than were gent relationships of MDMs and MiDMs to axoglial units, by
down-regulated. Additionally, the extent of gene up-regulation quantitative and qualitative ultrastructural analysis. Results
in MDMs exceeded that seen in MiDMs. from expression profiling supported this interpretation by show-
ing that MiDM metabolism was severely down-regulated,
MiDM and MDM gene expression kinetics reflected whereas expression profiles of MDMs reflected the activated
return toward homeostasis in recovery stage phagocytic phenotype observed through SBF-SEM.
These expression profiles showed consonant changes for the Several salient new observations emerged from these ex-
vast majority of genes analyzed: if a gene was up-regulated at periments. First, we showed that MDMs initiate demyelination
any time point, then its expression level showed an increase at at EAE onset, as MDMs were the overwhelmingly dominant

1542 Distinguishing microglia and monocytes in EAE CNS | Yamasaki et al.


Ar ticle

cells found in isolation attached to axoglial units and demon- importance of MDMs for this mechanism of demyelination. In
strated destructive interactions with myelinated axons in 3D particular, neutrophils in inflamed CNS of Ccr2rfp/rfp::Cx3cr1gfp/+
reconstructions. Second, MDMs were unexpectedly observed mice did not recognize disrupted nodes. These observations
at nodes of Ranvier in 9% of axoglial units and showed remark- are clinically pertinent: our detection of MDMs at nodes of
ably invasive behavior, including extension of microvilli (Fig. 4 A) Ranvier is consistent with recent reports of nodal pathology
or localization of cell soma (Fig. 4 B) between axolemma and in clinical demyelinated tissues (Fu et al., 2011; Desmazires
myelin sheath. Our observed frequency of MDMnodal inter- et al., 2012).The present observations extend this concept and
action represents a minimum estimate as MDMs found at hemi- provide a cellular basis for nodal pathology at the earliest stages
nodes adjacent to a demyelinated segment (Fig. 3 B) were not of demyelination. Given the presence of potential phagocytic
scored. Comparison of Ccr2rfp/rfp::Cx3cr1gfp/+ and Ccr2rfp/+:: signals at nodes (antibodies to paranodal proteins such as contac-
Cx3cr1gfp/+ mice at and before EAE onset emphasized the tin and neurofascins; Meinl et al., 2011); complement-derived

Figure 7. Affected functions in MiDMs and MDMs at EAE onset. nCounter inflammatory gene expression data were uploaded to IPA. Genes with
fold change (EAE onset vs. Naive) 1.5 or 1.5 were included in downstream effects analysis. (A) MDMs up-regulated functions, sorted by activation
z-score. (B) MiDMs up-regulated (left) and down-regulated (right) functions, sorted by activation z score. The bias term indicates imbalanced numbers of
up- and down-regulated genes associated with a distal function requiring significance at the P < 0.01 level. We studied pooled samples from 5 mice in
each time point (onset, peak, recovery) from 3 EAE inductions; 810 mice were immunized in each induction.

JEM Vol. 211, No. 8 1543


Figure 8. Restoration of affected inflammatory genes in resident microglia and recruited monocytes at recovery stage. For each gene, fold
change of all different disease stages versus naive state were calculated. Genes that contained at least one fold change >2 or < 0.5 and average fold
change for peak and onset >2 or <0.5 were presented. (A) Microglial up-regulated; (B) Microglial down-regulated; (C) monocyte up-regulated; (D) monocyte
down-regulated. We studied pooled samples from five mice in each time point (onset, peak, recovery) from three EAE inductions; 810 mice were immu-
nized in each induction. FC, fold change.

1544 Distinguishing microglia and monocytes in EAE CNS | Yamasaki et al.


Ar ticle

Figure 9. Function networks in MiDMs and MDMs. nCounter inflammatory gene expression data were uploaded to IPA. Genes with fold change
(EAE onset vs. Naive) 1.5 or 1.5 were included in upstream regulators analysis. Predicted upstream regulators were manually curated to form func-
tional clusters. Clusters were uploaded to IPA using Z scores as reference value for each gene. Networks were generated for each cluster consisting of
uploaded genes and additional predicted molecules. (A) Typical example of functions with dissimilar activation pattern in MiDMs and MDMs: HIF1A.
(B) Function with similar activation pattern in MiDMs and MDMs: Type I IFN. Red object denotes positive (>2) z score and green object denotes negative

JEM Vol. 211, No. 8 1545


opsonins (Nauta et al., 2004); and stress-induced eat-me sig- a gradual return toward a homeostatic expression profile, as
nals (Hochreiter-Hufford and Ravichandran, 2013), it may be suggested by MiDM up-regulation of fos, jun, myc, and CSF1
feasible to identify a direct molecular pathway for initiating de- (Wei et al., 2010) at disease onset. By striking contrast, MDMs
myelination in this model. Third, we characterized a molecu- up-regulated a large suite of inflammation-associated genes at
lar signature for resident microglia at EAE onset. Grouping of EAE onset, with subsequent regression to the expression phe-
regulated genes into functional categories demonstrated a notype of circulating monocytes.
remarkable down-regulation of microglial metabolism at the Blood monocytes and resident microglia were exposed to
nuclear, cytoplasmic and cytoskeletal levels. the same inflammatory environment. However, their preEAE
In our initial experiments we found that the presence of states were extremely distinct, with monocytes being gener-
myelin debris at the peak of EAE did not discriminate MDMs ated from a bone marrow progenitor within weeks of entry
from MiDMs. We considered that SBF-SEM would exhibit into CNS, whereas microglia originated during early embryo-
advantages for spatial resolution (Denk and Horstmann, 2004) genesis and had inhabited a serum-free unique environment
required for characterizing relationships of myeloid cells to from midgestation. In a recent study, we characterized resident
axoglial units during the inflammatory demyelinating process microglia by profiling mRNA, miRNA, and protein in com-
at EAE onset.To take advantage of this technique we developed parison with infiltrated brain macrophages, nonmicroglial resi-
methods based on cell volume and process number (Fig. 1 E), dent brain cells, and peripheral macrophages (Butovsky et al.,
to distinguish MDMs from MiDMs in 0.2-m confocal optical 2014). The detailed profiling after separating cells via CD45dim
sections, and translated this approach directly to SBF-SEM status showed distinct mRNA, miRNA, and protein expres-
image sets at 0.2-m intervals.We also noted differential nuclear sion by microglia as compared with infiltrating monocytes or
morphology, mitochondrial shape, and osmiophilic granule neuroepithelial brain cells (Butovsky et al., 2014). The study
content between MDMs and MiDMs. These characteristics of described transcription factors and miRNAs characteristic of
MDMs and MiDMs may not be universally present in other microglia in healthy brain but not in peripheral monocytes.
pathological circumstances but demonstrate an approach These findings partially explain a divergent response of these
to ultrastructural distinction of myeloid cell populations in two cell types to the same stimuli (Butovsky et al., 2014).
tissue sections. The strength of the study is that the dual reporter system
Gene expression profiling across the time course of EAE is sufficient to accurately distinguish monocyte versus mi-
yielded intriguing kinetics as analyzed by k-means clustering. croglial cells and thus to address the general concept that
Five patterns were observed. Red group genes (increased at monocytes and microglia can exert differential functions in
onset) comprised the smallest number and involved several sur- a CNS disease process. At the onset of EAE, the time point
face molecules: CCR1, CCR7, CXCR2, and CD40. Of these, at which our imaging studies were focused, we are able to
CCR7 and CD40 have been reported on activated microglia, make an unequivocal distinction between resident microglia
including those observed in MS tissue sections (Kiviskk et al., (CX3CR1gfp) and infiltrating monocytes (CCR2rfp). Two em-
2004; Serafini et al., 2006). GAPDH was up-regulated in pirical observations underline this discrimination: microglia
MiDMs at onset. Although often regarded as a housekeeping are uniformly CX3CR1+ from early embryonic time points
gene, GAPDH is found in complexes that limit the translation through adulthood (Cardona et al., 2006; Ginhoux et al., 2010;
of inflammatory gene transcripts in activated mouse macro- Schulz et al., 2012), and CCR2+Ly6C+ cells constitute the vast
phages (Mukhopadhyay et al., 2009; Arif et al., 2012). As pre- majority of infiltrating monocytes at EAE onset (Saederup
viously reported (Chiu et al., 2013), MiDM gene expression et al., 2010; Mizutani et al., 2012).
during the course of EAE did not correspond to the M1/M2 There were unavoidable limitations of our research; spe-
pattern of peripheral macrophage responses to infection or tis- cifically, to address how monocytes and microglia respond to
sue injury. Microglial morphological transformation can be a shared microenvironment, we focused on a single, patho-
relatively uniform regardless of the inflammatory process that genically relevant time point: onset of EAE. For this reason, it
provokes it. Despite this apparent uniformity, gene expression was beyond the scope of our study to decipher the phenotypic
by morphologically identical microglia can differ drastically fate of infiltrated monocytes. In peripheral models of inflam-
contingent on context (Perry et al., 2007). mation, Ly6Chi/CCR2rfp monocytes down-regulate the re-
Unsupervised hierarchical clustering provided insight into porter over time and show phenotypic evolution. Furthermore,
gene expression patterns of MDMs and MiDMs. Naive and our conclusions should not be generalized beyond the present
recovery patterns were similar for both cell types. At disease disease paradigm: in other models, such as spinal cord contusion,
onset, microglia showed drastic down-regulation of the expres- the inflammatory infiltrate includes Ly6Clow/CX3CR1gfp
sion profile observed in cells from healthy brain. Brisk mi- monocytes, which are highly pathogenic (Donnelly et al.,
croglial proliferation (Ajami et al., 2011) may have accelerated 2011). Our findings carry biological and medical significance

(<2) z-score. Orange object denotes predicted activation of the network object. Blue object denotes predicted inhibition of the network object. Predicted
relationships (connecting lines): orange, leads to activation; blue, leads to inhibition; yellow, finding inconsistent with state of downstream molecule;
gray, effect not predicted.

1546 Distinguishing microglia and monocytes in EAE CNS | Yamasaki et al.


Ar ticle

by demonstrating and characterizing differential responses of Histological and immunohistochemical analysis. Spinal columns were
infiltrating monocytes and resident microglia in a relevant dis- removed after mice were perfused with 4% paraformaldehyde (PFA). For im-
munofluorescence assay, free floating sections of the lumbar spinal cord were
ease model at a prespecified time point, at which point patho-
prepared as previously described (Huang et al., 2006). For immunofluores-
genic events are taking place.Therefore, we focused our analysis cence assay, sections were blocked with 10% normal serum for 2 h and stained
on the day of EAE onset rather than subsequent events to with primary antibodies at 4C for 2448 h. After washing with PBS-T (PBS
challenge our overall hypothesis that infiltrating monocytes with 0.1% Triton X-100; Sigma-Aldrich) three times, the sections were incu-
versus resident microglia respond very differently to acute in- bated with secondary antibodies at room temperature for 2 h and mounted
flammatory stimuli. in ProLong Gold antifade reagent (Invitrogen). Antibodies used include rat
anti-CD11b (BD), mouse anti-GFP (Abcam), rabbit anti-RFP (Abcam), Alexa
Activated myeloid cells are the proximate effectors of a
Fluor 488 goat antimouse IgG (Invitrogen), Alexa Fluor 594 goat antirabbit
bewildering array of acute and chronic disorders (Wynn et al., IgG (Invitrogen), and Alexa Fluor 647 goat antirat IgG (Invitrogen). Nuclei
2013). The technical and conceptual approach taken in this were labeled by DAPI. Images were collected by confocal laser-scanning mi-
study may be applicable to other tissues and disease processes. croscope (SP5; Leica).
In many pathological conditions, tissues harbor a mixed pop-
ulation of activated resident and recruited monocytes. The Quantitative 3D morphology. Quantitative 3D morphology of MDMs
therapeutic strategy will differ conclusively based on the spe- and MiDMs was analyzed in confocal images from spinal cord of mice at
cific effector properties of each cell type and the stage of dis- EAE onset. Free floating sections of the lumbar spinal cord were stained with
ease. In particular, if monocytes are pathogenic, then their RFP for MDMs, GFP for MiDMs, and DAPI for nuclei. Stack images were
taken at 0.2-m step size along the z-direction with a 63 objective (numer-
trafficking should be blocked using a peripherally active agent. ical aperture [NA] = 1.4) and zoom factor 2. A square (1,024 1,024 pixels)
The optimal application of agents that regulate leukocyte mi- corresponding to 123 123 m2 was used for the analysis. Cells were 3D re-
gration and intracellular signaling will be promoted by de- constructed by ImageJ software and all analyses were performed using ImageJ
tailed examination of each individual myeloid population. with 3D Convex Hull plugin. The parameters analyzed include voxel (volu-
metric pixel), convex voxel, volume, convex volume, surface, and convex sur-
face area. Other calculated parameters were: Solidity3D = volume/convex
MATERIALS AND METHODS
volume; Convexity3D = convex surface area/surface area; Formfactor3D =
Mice. C57BL/6 mice were obtained from the National Cancer Institute. 3 36 volume 2 surface area 3 . The number of primary processes was esti-
Ccr2rfp/+::Cx3cr1gfp/+ mice were generated by crossbreeding Ccr2rfp/rfp::C57BL/6
mated visually. We included 5 mice, 54 MDMs; 51 MiDMs in this assay with
mice (Saederup et al., 2010) with Cx3cr1gfp/gfp::C57BL/6 mice ( Jung et al.,
2 sections/mouse, 46 cells/section and 812 cells/mouse. Those mice came
2000). Ccr2rfp/rfp::Cx3cr1gfp/gfp mice were generated by breeding Ccr2rfp/+
from three EAE inductions.
::Cx3cr1gfp/+ mice. Ccr2rfp/rfp::Cx3cr1gfp/+ mice were generated by crossbreed-
ing Ccr2rfp/rfp::C57BL/6 mice with Ccr2rfp/rfp::Cx3cr1gfp/gfp mice. Animal ex-
periments were performed according to the protocols approved by the SBF-SEM. Spinal cords were removed after mice were perfusion-fixed
Institutional Animal Care and Use Committee at the Cleveland Clinic fol- using 4% PFA with 1% glutaraldehyde. Lumbar spinal cord sections were
lowing the National Institutes of Health guidelines for animal care. made on a vibratome (Leica). Sections were stained with 0.4% OsO4, uranyl
acetate and lead aspartate, then embedded in epon resin (Electronic Micros-
EAE induction and clinical evaluation. EAE was induced in Ccr2rfp/+ copy Sciences). SBF-SEM images were acquired using a Sigma VP SEM
::Cx3cr1gfp/+ mice and Ccr2rfp/rfp::Cx3cr1gfp/+ mice of 2428 wk of age using (Carl Zeiss) with 3View (Gatan). Serial image stacks of images at 100-nm
myelin-oligodendrocyte-glycoprotein peptide 3555 (MOG) as previously steps were obtained by sectioning 48 48 20 m3 tissue blocks (length
described (Huang et al., 2006). All mice were weighed and graded daily for width depth) at a resolution of 8192 8192 pixels. Image stacks were pro-
clinical stages as previously reported (Saederup et al., 2010).We defined clinical cessed for 3D reconstruction by TrakEM2 in FIJI software (National Institutes
stage of EAE as follows: pre-onset was the day sudden weight loss for 810% of Health). Alternating sections from the same stacked images were chosen to
occurred; onset was the day EAE signs appeared; peak was the second day score make stacks for 3D reconstructions which matched the 0.2-m step size used
didnt increase after sustained daily worsening; and recovery was the second for acquiring confocal stacked images. In SBF-SEM images, we discriminated
day score didnt decrease after a period of sustained daily improvement. MDMs and MiDMs using the volume/primary processes model (Fig. 1 E)
To address our research questions, we integrated flow cytometry, immuno generated from analyzing confocal images. Quantifications of myeloid-cell
histochemistry with quantitative morphometry, cell sorting for expression spatial relationships to axoglial units, including myelin incorporation, were
profiling, and serial block-face scanning electronic microscopy. In all, we per- done in SBF-SEM images.
formed 12 EAE immunizations in Ccr2rfp/+::Cx3cr1gfp/+ mice and 19 immu-
nizations in Ccr2rfp/rfp::Cx3cr1gfp/+ mice for this project, with 810 mice in Quantification of nuclei and mitochondria. Characterizations of nu-
each immunization. We selected EAE mice at onset, peak or recovery de- clear shapes were conducted in SBF-SEM images. Nuclei were categorized
pending on the specific studies underway at that time, with the majority of as follows: round, round shape and smooth surface with ratio of length/
mice coming from the onset stage of EAE. Each experiment incorporated width 1.5; elongated, elongated or oval shape with length/width >1.5, and
samples from at least three separate immunizations. Details of mouse numbers may have small indentations; Bilobulated: two connected lobes with single
and how they were selected for each experiment were included in the figure intervening large indentation; Irregular: complicated shape with corrugated
legends as requested. surface, and may have multiple and variable sizable indentations. Blinded ob-
servers (n = 3) scoring the nuclear morphology from SBF-SEM images in-
Cell isolation and flow cytometry. Brains and spinal cords were removed cluded a research student, a research fellow and a neuroscientist. Observers
and homogenized. Mononuclear cells were separated with a 30%/70% Per- were trained on the same nuclear examples in each category and practiced
coll (GE Healthcare) gradient as previously reported (Pino and Cardona, using 20 nuclei comprising all shapes before scoring the nuclei. Kappa test
2011). Single-cell suspensions from CNS were stained with antiF4/80-APC showed good pairwise agreement rates among observers (>0.8) and the data
(BM8; eBioscience) and antiCD45-PerCP (30-F11; BioLegend). Cells were from the neuroscientist are used. Quantifications were done in 3 individual
either analyzed on a LSR-II (BD) or sorted on a FACSAria II (BD) running mice from 3 EAE inductions including 2835 cells from two separate lesions
Diva6. Data were analyzed with FlowJo software (Tree Star). from each mouse in the assay.

JEM Vol. 211, No. 8 1547


Mitochondria of MDMs and MiDMs at EAE onset were reconstructed nonrandom association. The p value of overlap is calculated by the Fishers
from SBF-SEM images to 3D images using TrakEM2. 5 MDM and 5 MiDM Exact Test.
cells from 3 separate mice at EAE onset (total 10 cells) were included in the The activation z-score is a value calculated by the IPA z-score algorithm.
assay.Those mice came from 3 EAE inductions. Mitochondria were quantified The z-score predicts the direction of change for a function or the activation
for length, cross-sectional area, volume and ratio of length/cross-sectional state of the upstream regulator using the uploaded gene expression pattern
area using Fiji software. (upstream to the function and downstream to an upstream regulator). An ab-
solute z-score of 2 is considered significant. A function is increased/upstream
Quantification of demyelination. Black-gold staining was performed ac- regulator is activated if the z-score is 2. A function is decreased/upstream
cording to a protocol described previously (Liu et al., 2010). In brief, 5 free regulator is inhibited if the z-score -2.
floating lumbar spinal cord sections were stained in 0.2% black-gold solution The bias term is the product of the dataset bias and the bias of target
at 65C water bath for 10 min. After staining with black-gold, sections were molecules involved in a particular function annotation or upstream regulator
pictured by 3-CCD video camera interfaced with an Image-Pro Plus Analy- activity. A biased dataset is one where there is more up- than down-regulated
sis System (Version 4.1.0.0; MediaCybernetics) and analyzed with ImageJ genes or vice versa. The dataset bias is constant for any given analysis and the
software. Demyelinated areas are those void of black-gold staining. Mean function/upstream regulator bias is unique for each upstream regulator/function.
percentage of demyelinated areas in white matter were calculated. We in- When the absolute value of this term is 0.25 or higher, then that function/up-
cluded 5 mice from 3 EAE inductions in this assay. stream regulators prediction is considered to be biased and the Fishers exact
p-value must be 0.01 or lower for the analysis to be considered significant.
Statistical analysis of cellular elements. Statistical analyses were per-
formed using SAS (SAS Institute Inc.), PRISM (GraphPad Software) and We thank Dr. Bruce D. Trapp for invaluable suggestions. We thank Flow core
SPSS 17.5 (SPSS Inc.). Flow cytometry data were analyzed by two-way in Cleveland Clinic Foundation for the flow cytometry experiments. We thank
ANOVA test and Wilcoxon matched-pairs signed rank test. Nuclear shape Aishwarya Yenepalli for help with quantification.
quantifications were compared by paired Students t tests and logistic regres- This research was supported by grants from the US National Institutes of
sion. Mitochondrial quantifications were compared by Mann-Whitney U Health, the Charles A. Dana Foundation, the National Multiple Sclerosis Society, and
test and linear mixed model. Quantitative relationships of myeloid cells to the Williams Family Fund for MS Research, as well as a Postdoctoral Fellowship
axoglial units were compared using logistic regression with generalized esti- from National Multiple Sclerosis Society (to N. Ohno).
mating equations (GEE). Clinical characteristics of EAE mice were analyzed The authors have no competing financial interests.
using two-way ANOVA test with Bonferroni post test. Percentage of demye-
Submitted: 28 November 2013
lination was compared by Students t test. Data were shown as mean SEM
Accepted: 9 June 2014
or median (the first quartilethe third quartile) and P < 0.05 was considered
statistically significant. REFERENCES
Ajami, B., J.L. Bennett, C. Krieger, W. Tetzlaff, and F.M. Rossi. 2007. Local
Gene expression. Mononuclear cells were prepared from brains and spinal self-renewal can sustain CNS microglia maintenance and function
cords as described previously. Cells were sorted on a BD FACSAria II by gat- throughout adult life. Nat. Neurosci. 10:15381543. http://dx.doi.org/10
ing on F4/80+GFP+ for MiDMs and F4/80+RFP+ for MDMs. RNA was .1038/nn2014
isolated from FACS-sorted cells mixed from three mice from six EAE induc- Ajami, B., J.L. Bennett, C. Krieger, K.M. McNagny, and F.M. Rossi. 2011.
tions per data point in TRIzol Reagent (Ambion) according to manufactur- Infiltrating monocytes trigger EAE progression, but do not contribute to
ers protocol. RNA samples were analyzed by nCounter gene expression the resident microglia pool. Nat. Neurosci. 14:11421149. http://dx.doi
analysis and quantified with the nCounter Digital Analyzer (NanoString .org/10.1038/nn.2887
Technologies). Expressions of 179 genes were analyzed using nCounter GX Arif, A., P. Chatterjee, R.A. Moodt, and P.L. Fox. 2012. Heterotrimeric
Mouse Inflammation kit. GAIT complex drives transcript-selective translation inhibition in mu-
rine macrophages. Mol. Cell. Biol. 32:50465055. http://dx.doi.org/10
Nanostring data normalization. Normalization was conducted with .1128/MCB.01168-12
nSolver Analysis Software1.1. Data were normalized using positive and nega- Bauer, J., I. Huitinga, W. Zhao, H. Lassmann, W.F. Hickey, and C.D. Dijkstra.
tive controls and housekeeping genes probes. Background level was calcu- 1995. The role of macrophages, perivascular cells, and microglial cells in
lated for each sample as mean of negative control probes + (x2 SD). Calculated the pathogenesis of experimental autoimmune encephalomyelitis. Glia.
background was subtracted from each gene expression value. In cases where 15:437446. http://dx.doi.org/10.1002/glia.440150407
the calculated value was <1, values were set to 1. Brck, W., P. Porada, S. Poser, P. Rieckmann, F. Hanefeld, H.A. Kretzschmar,
and H. Lassmann. 1995. Monocyte/macrophage differentiation in early
Hierarchical and k-means clustering analysis. Hierarchical cluster analy multiple sclerosis lesions. Ann. Neurol. 38:788796. http://dx.doi.org/10
sis was performed using Pearson correlation for distance measure algorithm .1002/ana.410380514
Butovsky, O., M.P. Jedrychowski, C.S. Moore, R. Cialic, A.J. Lanser, G.
to identify samples with similar patterns of gene expression. MiDM samples
Gabriely, T. Koeglsperger, B. Dake, P.M. Wu, C.E. Doykan, et al. 2014.
expression data were used in k-means clustering using Pearson correlation
Identification of a unique TGF--dependent molecular and functional
for distance measures (Multi Experiment Viewer v. 4.8).
signature in microglia. Nat. Neurosci. 17:131143. http://dx.doi.org/10.
1038/nn.3599
IPA (Ingenuity) analysis. Data were analyzed using IPA (Ingenuity Sys-
Cardona, A.E., E.P. Pioro, M.E. Sasse,V. Kostenko, S.M. Cardona, I.M. Dijkstra,
tems). Differentially expressed genes (EAE onset MiDMs versus naive mi- D. Huang, G. Kidd, S. Dombrowski, R. Dutta, et al. 2006. Control of mi-
croglia and EAE onset MDMs versus naive splenic monocytes) were used in croglial neurotoxicity by the fractalkine receptor. Nat. Neurosci. 9:917
downstream effects and upstream regulators analyses. Uploaded dataset for 924. http://dx.doi.org/10.1038/nn1715
analysis were filtered using cutoff definition of 1.5-fold change. Level of con- Chiu, I.M., E.T. Morimoto, H. Goodarzi, J.T. Liao, S. OKeeffe, H.P.
fidence for analysis was set to high-predicted and experimentally observed. Phatnani, M. Muratet, M.C. Carroll, S. Levy, S. Tavazoie, et al. 2013. A
neurodegeneration-specific gene-expression signature of acutely isolated
Terms used in IPA analyses. The p-value is a measure of the likelihood microglia from an amyotrophic lateral sclerosis mouse model. Cell Rep.
that the association between a set of genes in the uploaded dataset and a re- 4:385401. http://dx.doi.org/10.1016/j.celrep.2013.06.018
lated function or upstream regulator is due to random association.The smaller Denk, W., and H. Horstmann. 2004. Serial block-face scanning electron mi-
the p-value, the less likely it is that the association is random and the more croscopy to reconstruct three-dimensional tissue nanostructure. PLoS
significant the association. In general, P < 0.05 indicate a statistically significant, Biol. 2:e329. http://dx.doi.org/10.1371/journal.pbio.0020329

1548 Distinguishing microglia and monocytes in EAE CNS | Yamasaki et al.


Ar ticle

Desmazires, A., N. Sol-Foulon, and C. Lubetzki. 2012. Changes at the nodal Mizutani, M., P.A. Pino, N. Saederup, I.F. Charo, R.M. Ransohoff, and A.E.
and perinodal axonal domains: a basis for multiple sclerosis pathology? Cardona. 2012.The fractalkine receptor but not CCR2 is present on mi-
Mult. Scler. 18:133137. http://dx.doi.org/10.1177/1352458511434370 croglia from embryonic development throughout adulthood. J. Immunol.
Donnelly, D.J., E.E. Longbrake, T.M. Shawler, K.A. Kigerl, W. Lai, C.A. Tovar, 188:2936. http://dx.doi.org/10.4049/jimmunol.1100421
R.M. Ransohoff, and P.G. Popovich. 2011. Deficient CX3CR1 signaling Mukhopadhyay, R., J. Jia, A. Arif, P.S. Ray, and P.L. Fox. 2009. The GAIT sys-
promotes recovery after mouse spinal cord injury by limiting the recruit- tem: a gatekeeper of inflammatory gene expression. Trends Biochem. Sci.
ment and activation of Ly6Clo/iNOS+ macrophages. J. Neurosci. 31:9910 34:324331. http://dx.doi.org/10.1016/j.tibs.2009.03.004
9922. http://dx.doi.org/10.1523/JNEUROSCI.2114-11.2011 Nauta, A.J., G. Castellano, W. Xu, A.M. Woltman, M.C. Borrias, M.R. Daha,
Ferguson, B., M.K. Matyszak, M.M. Esiri, and V.H. Perry. 1997. Axonal dam- C. van Kooten, and A. Roos. 2004. Opsonization with C1q and man-
age in acute multiple sclerosis lesions. Brain. 120:393399. http://dx.doi nose-binding lectin targets apoptotic cells to dendritic cells. J. Immunol.
.org/10.1093/brain/120.3.393 173:30443050. http://dx.doi.org/10.4049/jimmunol.173.5.3044
Fife, B.T., G.B. Huffnagle,W.A. Kuziel, and W.J. Karpus. 2000. CC chemokine re- Perry,V.H., C. Cunningham, and C. Holmes. 2007. Systemic infections and in-
ceptor 2 is critical for induction of experimental autoimmune encephalomy- flammation affect chronic neurodegeneration. Nat. Rev. Immunol. 7:161
elitis. J. Exp. Med. 192:899905. http://dx.doi.org/10.1084/jem.192.6.899 167. http://dx.doi.org/10.1038/nri2015
Fu, Y., T.J. Frederick, T.B. Huff, G.E. Goings, S.D. Miller, and J.X. Cheng. Pino, P.A., and A.E. Cardona. 2011. Isolation of brain and spinal cord mono-
2011. Paranodal myelin retraction in relapsing experimental autoimmune nuclear cells using percoll gradients. J.Vis. Exp. 2348.
encephalomyelitis visualized by coherent anti-Stokes Raman scattering mi- Ransohoff, R.M. 2007. Microgliosis: the questions shape the answers. Nat.
croscopy. J. Biomed. Opt. 16:106006. http://dx.doi.org/10.1117/1.3638180 Neurosci. 10:15071509. http://dx.doi.org/10.1038/nn1207-1507
Gautier, E.L., T. Shay, J. Miller, M. Greter, C. Jakubzick, S. Ivanov, J. Helft, A. Ransohoff, R.M. 2012. Animal models of multiple sclerosis: the good, the bad
Chow, K.G. Elpek, S. Gordonov, et al; Immunological Genome Consortium. and the bottom line. Nat. Neurosci. 15:10741077. http://dx.doi.org/10
2012. Gene-expression profiles and transcriptional regulatory pathways .1038/nn.3168
that underlie the identity and diversity of mouse tissue macrophages. Nat. Saederup, N., A.E. Cardona, K. Croft, M. Mizutani, A.C. Cotleur, C.L. Tsou,
Immunol. 13:11181128. http://dx.doi.org/10.1038/ni.2419 R.M. Ransohoff, and I.F. Charo. 2010. Selective chemokine receptor
Ginhoux, F., M. Greter, M. Leboeuf, S. Nandi, P. See, S. Gokhan, M.F. Mehler, usage by central nervous system myeloid cells in CCR2-red fluorescent
S.J. Conway, L.G. Ng, E.R. Stanley, et al. 2010. Fate mapping analysis protein knock-in mice. PLoS ONE. 5:e13693. http://dx.doi.org/10
reveals that adult microglia derive from primitive macrophages. Science. .1371/journal.pone.0013693
330:841845. http://dx.doi.org/10.1126/science.1194637 Savarin, C., S.A. Stohlman, R. Atkinson, R.M. Ransohoff, and C.C. Bergmann.
Gomez Perdiguero, E., C. Schulz, and F. Geissmann. 2013. Development and 2010. Monocytes regulate T cell migration through the glia limitans
homeostasis of resident myeloid cells: the case of the microglia. Glia. during acute viral encephalitis. J. Virol. 84:48784888. http://dx.doi.org/
61:112120. http://dx.doi.org/10.1002/glia.22393 10.1128/JVI.00051-10
Hochreiter-Hufford, A., and K.S. Ravichandran. 2013. Clearing the dead: apop- Schulz, C., E. Gomez Perdiguero, L. Chorro, H. Szabo-Rogers, N. Cagnard,
totic cell sensing, recognition, engulfment, and digestion. Cold Spring Harb. K. Kierdorf, M. Prinz, B.Wu, S.E. Jacobsen, J.W. Pollard, et al. 2012. A lin-
Perspect. Biol. 5:a008748. http://dx.doi.org/10.1101/cshperspect.a008748 eage of myeloid cells independent of Myb and hematopoietic stem cells.
Huang, D., F.D. Shi, S. Jung, G.C. Pien, J. Wang, T.P. Salazar-Mather, T.T. He, Science. 336:8690. http://dx.doi.org/10.1126/science.1219179
J.T. Weaver, H.G. Ljunggren, C.A. Biron, et al. 2006. The neuronal Serafini, B., B. Rosicarelli, R. Magliozzi, E. Stigliano, E. Capello, G.L.
chemokine CX3CL1/fractalkine selectively recruits NK cells that modify Mancardi, and F. Aloisi. 2006. Dendritic cells in multiple sclerosis le-
experimental autoimmune encephalomyelitis within the central nervous sions: maturation stage, myelin uptake, and interaction with proliferating
system. FASEB J. 20:896905. http://dx.doi.org/10.1096/fj.05-5465com T cells. J. Neuropathol. Exp. Neurol. 65:124141.
Huitinga, I., N. van Rooijen, C.J. de Groot, B.M. Uitdehaag, and C.D. Steinman, L., R. Martin, C. Bernard, P. Conlon, and J.R. Oksenberg. 2002.
Dijkstra. 1990. Suppression of experimental allergic encephalomyelitis in Multiple sclerosis: deeper understanding of its pathogenesis reveals new
Lewis rats after elimination of macrophages. J. Exp. Med. 172:10251033. targets for therapy. Annu. Rev. Neurosci. 25:491505. http://dx.doi.org/
http://dx.doi.org/10.1084/jem.172.4.1025 10.1146/annurev.neuro.25.112701.142913
Huitinga, I., J.G. Damoiseaux, E.A. Dpp, and C.D. Dijkstra. 1993. Treatment Stoll, G., J.W. Griffin, C.Y. Li, and B.D.Trapp. 1989.Wallerian degeneration in
with anti-CR3 antibodies ED7 and ED8 suppresses experimental aller- the peripheral nervous system: participation of both Schwann cells and
gic encephalomyelitis in Lewis rats. Eur. J. Immunol. 23:709715. http:// macrophages in myelin degradation. J. Neurocytol. 18:671683. http://
dx.doi.org/10.1002/eji.1830230321 dx.doi.org/10.1007/BF01187086
Izikson, L., R.S. Klein, I.F. Charo, H.L. Weiner, and A.D. Luster. 2000. Trapp, B.D., J. Peterson, R.M. Ransohoff, R. Rudick, S. Mrk, and L. B.
Resistance to experimental autoimmune encephalomyelitis in mice 1998. Axonal transection in the lesions of multiple sclerosis. N. Engl. J.
lacking the CC chemokine receptor (CCR)2. J. Exp. Med. 192:1075 Med. 338:278285. http://dx.doi.org/10.1056/NEJM199801293380502
1080. http://dx.doi.org/10.1084/jem.192.7.1075 Trebst, C., T.L. Srensen, P. Kiviskk, M.K. Cathcart, J. Hesselgesser, R.
Jung, S., J. Aliberti, P. Graemmel, M.J. Sunshine, G.W. Kreutzberg, A. Sher, Horuk, F. Sellebjerg, H. Lassmann, and R.M. Ransohoff. 2001. CCR1+/
and D.R. Littman. 2000. Analysis of fractalkine receptor CX(3)CR1 CCR5+ mononuclear phagocytes accumulate in the central nervous
function by targeted deletion and green fluorescent protein reporter system of patients with multiple sclerosis. Am. J. Pathol. 159:17011710.
gene insertion. Mol. Cell. Biol. 20:41064114. http://dx.doi.org/10 http://dx.doi.org/10.1016/S0002-9440(10)63017-9
.1128/MCB.20.11.4106-4114.2000 Wei, S., S. Nandi,V. Chitu,Y.G.Yeung,W.Yu, M. Huang, L.T.Williams, H. Lin,
Kiviskk, P., D.J. Mahad, M.K. Callahan, K. Sikora, C.Trebst, B.Tucky, J.Wujek, and E.R. Stanley. 2010. Functional overlap but differential expression
R. Ravid, S.M. Staugaitis, H. Lassmann, and R.M. Ransohoff. 2004. of CSF-1 and IL-34 in their CSF-1 receptor-mediated regulation of
Expression of CCR7 in multiple sclerosis: implications for CNS immu- myeloid cells. J. Leukoc. Biol. 88:495505. http://dx.doi.org/10.1189/
nity. Ann. Neurol. 55:627638. http://dx.doi.org/10.1002/ana.20049 jlb.1209822
Kulkarni, M.M. 2011. Digital multiplexed gene expression analysis using Williams, K.C., E. Ulvestad, and W.F. Hickey. 1994. Immunology of multiple
the NanoString nCounter system. Curr. Protoc. Mol. Biol. Chapter 25, sclerosis. Clin. Neurosci. 2:229245.
Unit25B 10. Wynn,T.A., A. Chawla, and J.W. Pollard. 2013. Macrophage biology in devel-
Liu, L., A. Belkadi, L. Darnall, T. Hu, C. Drescher, A.C. Cotleur, D. Padovani- opment, homeostasis and disease. Nature. 496:445455. http://dx.doi.org/
Claudio,T. He, K. Choi,T.E. Lane, et al. 2010. CXCR2-positive neutrophils 10.1038/nature12034
are essential for cuprizone-induced demyelination: relevance to multiple Xu, Y., J. Li, G.D. Ferguson, F. Mercurio, G. Khambatta, L. Morrison, A.
sclerosis. Nat. Neurosci. 13:319326. http://dx.doi.org/10.1038/nn.2491 Lopez-Girona, L.G. Corral, D.R. Webb, B.L. Bennett, and W. Xie.
Meinl, E., T. Derfuss, M. Krumbholz, A.K. Prbstel, and R. Hohlfeld. 2011. 2009. Immunomodulatory drugs reorganize cytoskeleton by modulating
Humoral autoimmunity in multiple sclerosis. J. Neurol. Sci. 306:180182. Rho GTPases. Blood. 114:338345. http://dx.doi.org/10.1182/blood-
http://dx.doi.org/10.1016/j.jns.2010.08.009 2009-02-200543

JEM Vol. 211, No. 8 1549


INSIGHTS

The real thing: How to make human DC subsets


The developmental relationship between monocytes, dendritic cells (DCs), and macrophages has been
well defined in mice, but human DC development is less well understood and has been hampered by
the lack of a suitable culture system. Now, two papers published in this issue describe a novel in vitro
culture system for human DC progenitors and the use of this system to elucidate the pathway of human
DC development.
The extent to which mouse studies are useful to understand the human immune system is debatable.
Insight from One expectsand observesimportant similarities, but there are also differences in the way the building
Frederic blocks of the immune system are assembled in different species of vertebrates that have evolved in different
Geissmann contexts and milieus, and with different lifespans. Laboratory mice, maintained as inbred strains under
selected housing conditions, are amendable to controlled genetic studies. However, genetic and environmental
variations are difficult to control in human studies, and experimental limitations make it difficult to assess whether "mouse"
immunology "works" in humans.
The two studies in this issue, by Lee et al. and Breton et al., represent an important advance in the DC field. The work
describes, at the population and single cell level, a hierarchy of human myeloid precursors that closely parallel the hierarchy in
the mouse bone marrow and blood. The authors used a combination of technically impressive qualitative and quantitative
approaches, including in vitro differentiation of human hematopoietic precursors cultured on a stromal cell line with defined
sets of cytokines, in vivo "cultures" of human precursors in immunodeficient NOD-scid g mice, and observational studies in
humans. This allowed them to map out the development of monocytes, conventional DCs (cDCs), and plasmacytoid DCs
(pDCs), and the sequential relationship between the different precursor populations. They showed that a granulocyte-
monocyte-DC progenitor (hGMDP) develops into a monocyte-DC progenitor (hMDP), which itself differentiates into mono-
cytes and into a common DC progenitor (hCDP) that produces the three major human DC subsets (CD1c+ cDCs, CD141+ cDCs,
and pDCs). Furthermore, they report the identification of an immediate DC precursor (hpre-cDC) that originates from the
hCDP, circulates in the human blood,
and increases in response to plasma levels
of the cytokine FLT3.
These results are a significant advance
in the field. They illustrate the similari-
ties between mice and humans regarding
the development of monocytes, cDCs,
and pDCs, and provide a guideand
experimental systemsto further in-
terrogate the genetic and molecular
control of human monocyte/DC devel-
opment and their functions in disease.
It is possible to imagine that DC-based
therapy may benefit from knowledge Schematic of the developmental relationship between human monocytes, cDCs, and pDCs.
of the "real thing."
Breton, G., et al. 2015. J. Exp. Med. http://dx.doi.org/10.1084/jem.20141441.
Lee, J., et al. 2015. J. Exp. Med. http://dx.doi.org/10.1084/jem.20141442.

Frederic Geissmann, Centre for Molecular and Cellular Biology of Inflammation, Kings College London: frederic.geissmann@kcl.ac.uk
Ar ticle

Restricted dendritic cell and monocyte


progenitors in human cord blood
and bone marrow
Jaeyop Lee,1* Galle Breton,2* Thiago Yukio Kikuchi Oliveira,2
Yu Jerry Zhou,1 Arafat Aljoufi,1 Sarah Puhr,1 Mark J. Cameron,4
Rafick-Pierre Skaly,4 Michel C. Nussenzweig,2,3** and Kang Liu1**
1Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032
2Laboratoryof Molecular Immunology and 3Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065
4Case Western Reserve University, Cleveland, OH 44106

In mice, two restricted dendritic cell (DC) progenitors, macrophage/dendritic progenitors


(MDPs) and common dendritic progenitors (CDPs), demonstrate increasing commitment to
the DC lineage, as they sequentially lose granulocyte and monocyte potential, respectively.
Identifying these progenitors has enabled us to understand the role of DCs and monocytes in
immunity and tolerance in mice. In humans, however, restricted monocyte and DC progenitors
remain unknown. Progress in studying human DC development has been hampered by lack
of an in vitro culture system that recapitulates in vivo DC hematopoiesis. Here we report a
culture system that supports development of CD34+ hematopoietic stem cell progenitors into
the three major human DC subsets, monocytes, granulocytes, and NK and B cells. Using this
culture system, we defined the pathway for human DC development and revealed the sequential
origin of human DCs from increasingly restricted progenitors: a human granulocyte-monocyte-DC
progenitor (hGMDP) that develops into a human monocyte-dendritic progenitor (hMDP),
which in turn develops into monocytes, and a human CDP (hCDP) that is restricted to produce
the three major DC subsets. The phenotype of the DC progenitors partially overlaps with
granulocyte-macrophage progenitors (GMPs). These progenitors reside in human cord
blood and bone marrow but not in the blood or lymphoid tissues.
CORRESPONDENCE
Kang Liu:
kl2529@columbia.edu
OR DCs, monocytes, and macrophages are closely In the mouse, DC differentiation is depen-
Michel C. Nussenzweig: related cell types whose interrelationship were dent on a hematopoietin, Flt3L, whose recep-
nussen@rockefeller.edu long debated and only recently elucidated in tor, Flt3 (CD135), is expressed throughout DC
Abbreviations used: cDC, con-
the mouse (Geissmann et al., 2010; Merad et al., development (McKenna et al., 2000; Karsunky
ventional DC; CDP, common 2013). In mice, DCs and monocytes arise from et al., 2003; Waskow et al., 2008). In contrast,
dendritic progenitor; CLP, a macrophage/dendritic progenitor (MDP; other hematopoietin receptors such as monocyte
common lymphoid progenitor;
CML, chronic myelogenous
Fogg et al., 2006), which produces monocytes, colony-stimulating factor receptor (M-CSFR
leukemia; CMP, common and a common dendritic progenitor (CDP) or CD115) and granulocyte macrophage colony-
myeloid progenitor; GMP, that is restricted to the DC fate (Shortman and stimulating factor receptor (GM-CSFR or
granulocyte-macrophage Naik, 2007; Liu et al., 2009; Geissmann et al., CD116) are restricted to hematopoietic pro-
progenitor; hGMDP, human
granulocyte-monocyte-DC 2010; Merad et al., 2013). The CDP produces genitors of DCs but not expressed on all ma-
progenitor; hMDP, human preplasmacytoid DCs (pDCs) and pre ture DCs (Kingston et al., 2009).
monocyte-dendritic progenitor; conventional DCs (cDCs), the latter of which DC development in the human is far less
HSC, hematopoietic stem cell;
HSPC, hematopoietic stem
leaves the BM and circulates in the blood be- well understood than in the mouse. Human
and progenitor cell; LMPP, fore entering tissues and developing into the monocytes can be induced to differentiate into
lymphoid-primed multipotent different DCs subsets (Naik et al., 2006, 2007; potent antigen-presenting cells with some
progenitor; MDP, macrophage/
dendritic progenitor; MLP,
Onai et al., 2007b, 2013; Ginhoux et al., 2009;
multilymphoid progenitor; Liu et al., 2009; Onai et al., 2013). 2015 Lee et al. This article is distributed under the terms of an Attribution
MPP, multipotent progenitor; NoncommercialShare AlikeNo Mirror Sites license for the first six months after
NSG, NOD-scid-IL2Rgnull; *J. Lee and G. Breton contributed equally to this paper. the publication date (see http://www.rupress.org/terms). After six months it is
available under a Creative Commons License (AttributionNoncommercialShare
pDC, plasmacytoid DC; SCF, **K. Liu and M.C. Nussenzweig contributed equally to Alike 3.0 Unported license, as described at http://creativecommons.org/licenses/
stem cell factor. this paper. by-nc-sa/3.0/).

The Rockefeller University Press $30.00


J. Exp. Med. 2015 Vol. 212 No. 3 385399 385
www.jem.org/cgi/doi/10.1084/jem.20141442
Figure 1. Stromal culture system for DCs
and other leukocytes. (a) Flow cytometry
plots show DCs obtained from peripheral
blood (PBMC) and DCs obtained from
cultures of cord blood CD34+ cells with
Flt3L+SCF+GM-CSF+IL-4 (FSG-4) or mouse
BM stromal cells (MS5)+Flt3L or MS5+FSG.
pDCs, CD1c+ cDCs, and CD141+ cDCs are
shaded green, blue, and red, respectively. Pie
charts indicate the relative representation of
each DC subset in each group. Plots are repre-
sentative of two Flt3L+SCF+GM-CSF+IL-4 or
more than three MS5+Flt3L and MS5+FSG
experiments. (b) Graphs show output of the
indicated cells derived from CD34+ cells in
MS5 with Flt3L (F) versus FSG. Error bars indi-
cate SEM. (c) Flow cytometry plots show
phenotype of cells developing from 2,000
CD34+ HSPCs purified from human cord blood
and cultured in MS5+FSG for 14 d. Represen-
tative culture of five different donors. Pie
chart shows CD45+ cell composition.

phenotypic features of DCs after in vitro culture with cock- a robust tissue culture system for the in vitro development of
tails of cytokines (Sallusto and Lanzavecchia, 1994). However, all DC subsets (Poulin et al., 2010; Ziegler-Heitbrock et al.,
these monocyte-derived DCs are more closely related to ac- 2010; Proietto et al., 2012).
tivated monocytes than to cDCs (Naik et al., 2006; Xu et al., Here we report a stromal cell culture system that supports
2007; Cheong et al., 2010; Crozat et al., 2010). Progress in the development of CD34+ hematopoietic stem cell (HSC)
defining the human DC lineage has been hampered, in part, progenitors into the three major subsets of human DCs,
by a paucity of reliable markers to distinguish these cells from monocytes, granulocytes, and NK and B cells. Using this cul-
monocytes, limited access to human tissues, the relatively ture system, we have been able to define the sequential origin
small number of circulating DCs in blood, and the lack of of human DCs from a human granulocyte-monocyte-DC

386 Progenitor of human dendritic cells | Lee et al.


Ar ticle

progenitor (hGMDP), which develops into a more restricted CD1c+ cDCs did not, which suggests that blood- and culture-
human monocyte-dendritic progenitor (hMDP), which pro- derived cells are not identical (Fig. 2 a and Fig. S2). Notably,
duces monocytes, and a human CDP (hCDP), which is re- all of the culture-derived CD1c+ and CD141+ cDCs differed
stricted to produce the three major subsets of DCs. from primary bloodderived counterparts in a similar man-
ner in that they were enriched for expression of genes that
RESULTS mediate cell division, as determined by Gene Set Enrichment
Human DC subsets develop in stromal cellcontaining Analysis (GSEA; Fig. 2 e, Fig. S2, and Table S4).This alteration
cultures in vitro in gene expression is likely the result of increased prolifera-
CD34+ hematopoietic stem and progenitor cells (HSPCs) cul- tion in the cultures caused by high levels of Flt3L. Consistent
tured in the presence of cytokines produce CD1c/BDCA1+ with this idea, primary peripheral bloodderived CD141+
and CD141/BDCA3+ cDCs but fail to produce pDCs cDCs phenocopy the culture-derived cells by acquiring
(CD303/BDCA2+; Fig. 1 a; Poulin et al., 2010). Stromal cells CD1c expression when they are placed into the MS5+FSG
have been used to facilitate differentiation of pDCs (Spits et al., (Fig. 2 f) or skin culture (Haniffa et al., 2012), and peripheral
2000; Chicha et al., 2004; Olivier et al., 2006), but their ability blood CD141+ cDCs also coexpress CD1c in individuals that
to support differentiation of all DC subsets as well as other are treated with Flt3L (see accompanying manuscript Breton
hematopoietic lineages has not been evaluated. In an attempt et al. in this issue). Finally, both culture-derived and primary
to develop a method that would support development of all CD1c+ cDCs purified from peripheral blood acquire CD14
three major types of DCs, we used a combination of mouse expression in culture (Fig. 2 f ).
BM stromal cells (MS5; Itoh et al., 1989) and defined human Gene array data were confirmed by flow cytometry using
cytokines. The combination of MS5 and Flt3L was sufficient selected markers. Similar to primary pDCs, culture-derived
to support development of cord blood CD34+ HSPCs into pDCs express high levels of CD123 and CD45RA and low
multiple cell types, including the three DC subsets, in pro- levels of HLA-DR but differ from mature cDCs in that they
portions similar to those found in peripheral blood (Fig. 1 a). do not express CD11c or CD86 (Fig. 3 a). In contrast, culture-
Addition of human stem cell factor (SCF) and human GM- derived CD1c+ and CD141+ cDCs resemble their primary
CSF (MS5+FSG, herein) increased the overall yield of DCs peripheral bloodderived counterparts in their differential
(Fig. 1, a and b). MS5+FSG cultures produced granulocytes expression of CX3CR1 and CD172a and in expressing high
(CD66b+), monocytes (CD14+CD16), NK cells (CD56+), levels of CD11c, HLA-DR, and CD86 but not CD83 or
B cells (CD19+), pDCs, and both subsets of human cDCs CD80 (MacDonald et al., 2002; Lindstedt et al., 2005; Mittag
from human CD34+ cells, as determined by expression of cell et al., 2011). Of note, CD1a and DC-SIGN, which are ex-
surface markers by flow cytometry (Fig. 1 c). pressed by monocyte-derived DCs and absent on primary
To further characterize the culture-derived DCs and CD1c+ cDCs (Chang et al., 2000), are not expressed on
monocytes and compare them with primary cells from blood, culture-derived CD1c+ or CD141+ cDCs (Fig. 3 a). In addition,
we performed whole transcriptome expression analysis on all culture-derived CD141+ cDCs express CLEC9a (DNGR1;
four subsets (Fig. S1). Using sparse hierarchical clustering, we Fig. 3 a), a marker specifically expressed on primary CD141+
showed that all four cell types clustered separately from each cDCs (Poulin et al., 2010, 2012).
other and that cultured and primary monocytes clustered to- To examine the functional properties of cultured-derived
gether, as did pDCs and cDCs (Fig. 2 a, Fig. S2, and Table S5). cDCs, we measured their responses to TLR ligands. Like their
We performed two separate analyses: monocytes versus pDCs primary bloodderived counterparts, only the culture-
(Fig. 2 b) and CD141+ cDCs versus CD1c+ cDCs (Fig. 2 c). derived pDCs produced IFN- in response to CpG (Fig. 3 b;
Sparse hierarchical clustering showed that culture-derived Ito et al., 2005; Liu, 2005). Similarly, culture-derived and pri-
pDCs and monocytes are closely related to their in vivo coun mary CD141+ cDCs produced the highest amount of IFN-
terparts (Fig. 2 b and Table S1), as are culture-derived CD1c+ and IL-12 in response to Poly(I:C) (Fig. 3 b; Kadowaki et al.,
and CD141+ cDCs to theirs (Fig. 2 c and Table S2). 2001; Poulin et al., 2010). We conclude that MS5+FSG sup-
A closer look at the expression of genes that constitute the ports differentiation of multiple hematopoietic lineages from
molecular signature of each subset (e.g., transcription factors their progenitors, including human pDCs and CD1c+ and
and surface receptors; Robbins et al., 2008; Crozat et al., 2010; CD141+ cDCs.
Schmidl et al., 2014) again indicates strong similarity between
cultured-derived cells and their in vivo counterparts (Fig. 2 d DC-restricted progenitors
and Table S3). For example, IRF8, BATF3, Zbtb46, and FLT3 Several different, early, CD34+ hematopoietic progenitors pu-
were similarly and selectively expressed in cultured and pri- rified from human cord blood or BM are reported to give rise
mary CD141+ cDCs; ETS2, ID2, Zbtb46, and FLT3 in cul- to DCs (Chicha et al., 2004; Ishikawa et al., 2007; Doulatov
tured and primary CD1c+ cDCs; FOS, CD14, and CSF1R in et al., 2010; Kohn et al., 2012). These include common lym-
cultured and primary monocytes; and IRF7,TCF4, SPIB, and phoid progenitors (CLPs; Galy et al., 1995; Chicha et al., 2004;
IL3RA in cultured and primary pDCs (Fig. 2 d). Ishikawa et al., 2007), common myeloid progenitors (CMPs;
Within the cDC population, primary and cultured CD141+ Akashi et al., 2000; Manz et al., 2002; Chicha et al., 2004;
cDCs clustered together, whereas primary and cultured Ishikawa et al., 2007), granulocyte-macrophage progenitors

JEM Vol. 212, No. 3 387


Figure 2. Culture-derived DCs resemble primary DCs. (ae) Transcriptional profiling of pDCs, monocytes, and CD1c+ and CD141+ cDCs purified
from primary peripheral blood (blood; six healthy individuals) or from culture of CD34+ cells in MS5+Flt3L for 14 d (culture; four cord blood donors) as
in Fig. S1. (a) Hierarchical clustering dendrogram of cultured versus primary pDCs, monocytes, and CD1c+ and CD141+ cDCs. This dendrogram was gen-
erated using the top 611 differentially expressed genes selected by unsupervised clustering (sparse hierarchical clustering using all genes; Table S5).
(b) Heat map showing the sparse hierarchical clustering of mRNAs expressed by primary and culture-derived pDCs and monocytes. This analysis showed
that a minimal number of 78 genes is enough to distinguish one cell type from another. The normalized expression values for the top 78 differentially
expressed genes (Table S1) are displayed. (c) Heat map showing the sparse hierarchical clustering of mRNAs expressed by primary and culture-derived

388 Progenitor of human dendritic cells | Lee et al.


Ar ticle

Figure 3. Culture-derived cells resemble


their ex vivo counterparts in phenotype
and function. (a) Histograms show cell sur-
face markers of human monocytes and DCs
isolated from blood (top rows) and MS5+FSG
cultures (bottom rows). (b) Cord blood CD34+-
derived DC subsets were cultured for 14 d,
purified by FACs, and exposed to the indicated
TLR stimuli. Graphs indicate concentration of
IFN- and IL-12p70 in the supernatant mea-
sured by ELISA after 48 h from three indepen-
dent experiments. n (number of donors) = 3.
Error bars indicate SEM.

(GMPs; Manz et al., 2002; Chicha et al., 2004; Ishikawa et al., with distinct lineage potential (Fig. 4 a; Doulatov et al., 2010)
2007; Doulatov et al., 2010), a myeloid DC progenitor (Olweus and the other specific against receptors for Flt3L (CD135),
et al., 1997), multilymphoid progenitors (MLPs; Doulatov et al., GM-CSF (CD116), M-CSF (CD115), and IL-3 (CD123),
2010), and lymphoid-primed multipotent progenitors (LMPPs; which are differentially expressed by monocytes and DCs
Kohn et al., 2012). However, none of these cell types is re- (Breton et al., 2015).
stricted to the monocyte or DC lineage (Chicha et al., 2004; We found that among the six populations of cord blood
Ishikawa et al., 2007; Doulatov et al., 2010; Kohn et al., 2012). CD34+ cells, namely HSCs/multipotent progenitors (MPPs),
In mice, monocyte- and DC-restricted progenitors can be MLPs, megakaryocytic and erythroid progenitors, B/NK,
identified by differential expression of CD117 (cKit), CD135 CMPs, and GMPs, only GMPs (CD34+CD38hiCD135+CD
(Flt3), CX3CR1, CD115 (M-CSFR), and Ly6C (Fogg et al., 45RA+CD10) exhibit heterogeneity in CD115+, CD116+,
2006; Naik et al., 2007; Onai et al., 2007a, 2013; Hettinger and CD123hi expression (Fig. 4 b) and contain cells with DC
et al., 2013; Merad et al., 2013). In humans, all myeloid and progenitor activity (Olweus et al., 1997; Chicha et al., 2004;
lymphoid progenitors express CD34, CD117, and CD135 Doulatov et al., 2010). To determine whether GMP can be
(Doulatov et al., 2010). Thus, additional markers are required further fractionated into monocyte- and DC-restricted pro-
to further purify human monocyte and DC progenitors. To genitors, we separated them into five populations on the basis
attempt to distinguish human monocyte and DC progenitors of CD115, CD116, and CD123 expression (Fig. 5 a) and cul-
from earlier and less restricted precursors, we combined two tured 200 cells from each purified population in MS5+FSG
separate sets of antibodies: one set that can separate human cultures for 7 d. We expected that DC progenitors show po-
CD34+ hematopoietic progenitor cells into six populations tential to produce all three DC subsets. CD123hiCD115,

CD1c+ and CD141+ cDCs. This analysis showed that a minimal number of 80 genes is enough to distinguish one cell type from another. The normalized
expression values for the top 80 differentially expressed genes (Table S2) are displayed. (d) Heat map showing the hierarchical clustering of mRNAs for
selected genes (Table S3) expressed by primary and culture-derived pDCs, monocytes, and CD1c+ and CD141+ cDCs. (e) Top 50 enriched KEGG metabolic
pathways (Table S4) for genes shared by both subsets of cultured cDCs but not primary cDCs according to GSEA analysis. (f) Phenotype change of blood
CD141+ and CD1c+ cDCs in culture. Blood CD141+ and CD1c+ cDCs were purified and cultured for 7 d in MS5+FSG. Flow cytometry plots of gated CD45+
cells show cell surface markers of output cells.

JEM Vol. 212, No. 3 389


Figure 4. Fractionation of cord blood progenitors based on cytokine receptor expression. (a and b) Flow cytometry plots show exhaustive sepa-
ration of CD34+ cord blood cells into six populations, HSCs/MPPs, MLPs, B and NK progenitors (B/NK), CMPs, megakaryocytic and erythroid progenitors
(MEP), and GMPs (a), and expression of CD115, CD116, CD135, and CD123 on each of the cord blood CD34+ populations in a (b).

CD123intCD116CD115+, and CD123intCD116CD115 CD123intCD116CD115+ cells produced pDCs, cDCs, and


cells gave rise to pDCs and CD1c+ and CD141+ cDCs CD14+CD1c monocytes (Patterson et al., 2005; Granelli-
(Fig. 5 b). Although CD123intCD116+CD115 or CD123int Piperno et al., 2006), but produced few, if any, granulocytes
CD116+CD115+ cells gave rise to CD1c+ cDCs, they failed and are therefore the human equivalent of the mouse MDP
to produce pDCs or CD141+ cDCs. These two populations (Fogg et al., 2006) and will be referred to as hMDPs. In cord
were therefore excluded as CDPs (Fig. 5 b). blood samples, the hMDPs constitute 0.56% (range of 0.18
CD123hiCD115 cells were restricted to DCs and pro- 0.70%) of CD34+ cells and can be identified using the CD34+
duced no monocytes or granulocytes, reminiscent of the CD45RA+CD123intCD115+ phenotype because of the neg-
CDPs in the mouse (Naik et al., 2007; Onai et al., 2007b), ligible frequency of CD123intCD116+CD115+ (0.04% of
and we will refer to them as hCDPs. These cells constitute CD34+ cells). In contrast, CD123intCD116CD115 cells
0.41% (range of 0.030.87%) of cord blood CD34+ cells. produced DCs, monocytes, and granulocytes and will be

390 Progenitor of human dendritic cells | Lee et al.


Ar ticle

Figure 5. Characterization of cord blood


progenitors. (a) Flow cytometry plots show
gating of cord blood CD34+CD38hiCD10CD45
RA+CD123int/hi GMP cells (Doulatov
et al., 2010) and further separation into
five separate populations based on CD123,
CD115, and CD116 expression: CD123hiCD115
(CD123hi), CD123intCD115+CD116
(CD115+), CD123intCD115+CD116+ (DP),
CD123intCD115CD116+ (CD116+), and
CD123intCD115CD116 (DN). (b) Differen-
tiation potential of 200 purified cells from
each of the five populations indicated in a in
MS5+FSG culture harvested after 7 d. Flow
cytometry plots show CD45+CD56CD19
cells. (c) Graph indicates output/input ratio
of total number of CD45+ cells obtained from
each of the five populations sorted in a.
Bars and error bars are means and SEM,
respectively, from three independent experi-
ments. (d) Histograms show expression of
indicated markers on hGMDPs, hMDPs, and
hCDPs. (e) Morphology of purified cord blood
hGMDPs, hMDPs, and hCDPs by Giemsa
staining of cytospin preparations. Bars, 10 m.
(f) Graph indicates the differentiation poten-
tial of hCDPs, hMDPs, hGMDPs, and CMPs in
methylcellulose colony formation assays
in vitro (as in Materials and methods). Colonies
were enumerated at 14 d after culture. BFU-E,
burst-forming unit erythroid; GEMM, granu-
locyte, erythrocyte, macrophage, megakaryo-
cyte; GM, granulocyte and macrophage; G,
granulocyte; M, macrophage. Bars are means
and error bars are SEM from three independent
experiments. (g) Cross-phenotyping hCDPs,
hMDPs, and hGMDPs with DC-associated pro-
genitors. hCDPs, hMDPs, and hGMDPs were
identified by flow cytometry and overlaid
with previously identified progenitors (gray).
CLPs were gated as CD45+Lin(CD3/19/56/
14)CD34+CD10+CD45RA+ (Galy et al., 1995;
Ishikawa et al., 2007), LMPPs as CD45+
LinCD34+CD10CD62L+CD45RA+ (Kohn
et al., 2012), CMPs as CD45+LinCD34+CD38+
CD10CD45RACD123+, GMPs as CD45+Lin
CD34+CD38+CD10CD45RA+CD123+/hi (Chicha
et al., 2004; Doulatov et al., 2010), MLPs as
CD45+LinCD34+CD38CD45RA+ (Doulatov
et al., 2010), and myeloid DC progenitors as
CD45+LinCD34+CD123hi (Olweus et al., 1997).
n (number of donors) = 3.

referred to as hGMDPs (Fig. 5 b), which constitute 10.08% and hCDPs were very similar and showed a high nucleus/
(range of 6.1513.2%) of cord blood CD34+ cells. As might cytoplasm ratio as well as multilobulated nuclei (Fig. 5 e).
be expected, the less differentiated hGMDPs and hMDPs In view of the more limited outgrowth of granulocytes
have a higher proliferative potential in vitro than hCDPs from the MS5+FSG cultures, we examined the DC pro-
(Fig. 5 c). All of these DC progenitors express HLA-DR but genitors potential to produce monocytes and granulocytes
not CD11c (Fig. 5 d). Morphologically, hGMDPs, hMDPs, in CFU assays. In agreement with the MS5+FSG cultures,

JEM Vol. 212, No. 3 391


efficiency, as measured by their ability to give rise to CD45+
cells, was 22% for hCDPs (89/408 wells), 80% for hMDPs
(152/191 wells), and 58% for hGMDPs (103/179 wells; Fig. 6 a).
In contrast, CD123intCD116+CD115 or CD115+ cells
showed lower clonal efficiency, 11% (13/122 wells) and 10%
(9/95 wells), respectively (Fig. 6 a). All positive wells were fur-
ther evaluated for production of specific cell lineages, and clonal
potential was categorized as multilineage or unilineage. The
multilineage clones yielded combinations of granulocytes,
monocytes, and DCs (G+M+DC), monocytes and DCs
(M+DC), granulocytes and monocytes (G+M), myeloid
(G/M/DC), and lymphoid (B or NK cells; G/M/DC+L), and
the unilineage ones produced only lymphoid cells (B or NK, L),
granulocytes (G), monocytes (M), or DCs (Fig. S3). Of the 89
positive wells produced by hCDPs, 84 contained pDCs and/or
cDCs and no monocytes or granulocytes (Fig. 6 b and Fig. S4).
Importantly, two clones produced both pDCs and cDCs (Fig. S4),
formally proving the existence of a clonal progenitor that pos-
sesses both pDC and cDC potential. Of the 152 positive
wells obtained from hMDPs, 31% contained only monocytes,
41% only DCs, 13% contained both monocytes and DCs, and
rare clones produced granulocytes (3.9%) or lymphocytes
(1.3%; Fig. 6 c and Figs. S5 and S6). Thus, hMDPs show little
granulocyte or lymphocyte potential and are restricted primar-
ily to monocytes and DCs. Among the 103 positive wells pro-
duced by hGMDPs, 22% contained only granulocytes, 4% only
Figure 6. Developmental potential of single progenitor cells. monocytes, 20% only DCs, 17% contained monocytes and
(a) Graph shows the percentage of positive wells obtained from culturing DCs, 6% showed granulocytes, monocytes, and DCs, and 14%
single hCDP, hMDP, hGMDP, CD123intCD115CD116+ (CD116+), and produced lymphoid cells alone or in combination with my-
CD123intCD115+ CD116+ (DP) cells in MS5+FSG culture. Clonal effi-
eloid cells (Fig. 6 d and Figs. S7 and S8). Thus, hGMDPs
ciency calculated based on the number of positive wells is indicated.
Data are pooled from three independent experiments. (bd) Bar graphs
contain clonal progenitors that can produce granulocytes,
summarize the cellular output of all positive single cell cultures of monocytes, and DCs, but this population is heterogeneous and
hCDPs (b), hMDPs (c), and hGMDPs (d) from three independent experi- also contains cells with the potential to give rise to lympho-
ments (n = 3 donors). The number of wells per category is noted on cytes (Fig. 6 d and Figs. S7 and S8). The residual lymphocyte
top of each bar. DC, pDC and/or cDC; G, granulocyte; L, lymphocyte; potential in this population may result in part from contamina-
M, monocyte. tion of MLPs, which are difficult to separate from hGMDPs
using CD38 (Doulatov et al., 2010).
hCDPs produced rare CFU, hMDPs produced mostly mono-
cytes but no granulocytes, and hGMDPs produced both Progenitorprogeny relationships
monocytes and granulocytes but no erythrocytes (Fig. 5 f ). of hGMDPs, hMDPs, and hCDPs
To define the relationship between hCDPs, hMDPs, and The sequential loss of differentiation potential to granulocytes,
hGMDPs and previously reported DC progenitors, we per- and then to monocytes at the hMDP and hCDP stages, sug-
formed cross-phenotyping experiments.We found that, GMPs gests that a differentiation hierarchy exists between hGMDPs,
include hCDPs, hMDPs, and hGMDPs; myeloid DC pro- hMDPs, and hCDPs. To determine whether, in fact, these
genitors overlap with hCDPs; LMPPs partially overlap with progenitors are related in this way, we initially performed
hCDPs, hMDPs, and hGMDPs, whereas CLPs, MLPs, and in vivo transfer experiments. Purified hGMDPs were transferred
CMPs do not overlap with hCDPs, hMDPs, and hGMDPs into the bone cavity of NOD-scid-IL2Rgnull (NSG) mice
(Fig. 5 g). We conclude that populations containing hGMDPs, (Material and methods; Kalscheuer et al., 2012). 7 d after
hMDPs, and hCDPs can be isolated from human cord blood transfer, both hMDPs and hCDPs were detected among the
by using cytokine receptor expression to distinguish them donor-derived cells in the BM, indicating that hMDPs and
from less committed leukocyte precursors. hCDPs descend from hGMDPs (Fig. 7 a).
To further refine this developmental hierarchy, we exam-
Single cell assays ined the development of purified hGMDPs, hMDPs, and
To examine the developmental potential of individual cells in hCDPs in tissue culture over time using flow cytometry.
the hGMDP, hMDP, and hCDP populations, we purified and Whereas hGMDPs and hMDPs retained CD34 expression
cultured single cells from each population. The relative clonal for at least 4 d, hCDPs down-regulated CD34 within 2 d,

392 Progenitor of human dendritic cells | Lee et al.


Ar ticle

Figure 7. Relationship of hGMDPs to hMDPs and to hCDPs. (a) 10,000 hGMDPs were purified from cord blood and adoptively transferred into the
bone cavity of the preconditioned NSG mice (Materials and methods). 7 d later, BM cells were analyzed. Flow cytometry plots show phenotype of BM cells
from NSG mice receiving PBS or hGMDPs (n = 3 mice). (b) 200400 hGMDP, hMDP, and hCDP cells were purified and cultured in MS5+FSG. Cultures were
analyzed on days 1, 4, and 8. Flow cytometry plots gated on live CD45+Lin(CD3/19/56/14)DC(CD1c/141/303)CD45RA+ cells show cell surface markers
and frequency of hCDPs, hMDPs, and hGMDPs. Results are representative of three independent experiments.

indicating that hCDPs are the most differentiated among the BM contained hGMDPs (mean of 5.22%, range of 1.76
three groups. Further phenotypic analysis of the CD34+ cells 7.40% of CD34+ cells), hMDPs (mean of 0.56%, range of
revealed that hGMDPs sequentially produced hMDPs and 0.081.94%), and hCDPs (mean of 1.98%, range of 0.05
then hCDPs and that hMDPs produced hCDPs (Fig. 7 b). In 7.29%; Fig. 8 a). When these populations were purified and
addition, both hGMDPs and hMDPs maintained CD34 ex- cultured in MS5+FSG, they showed differentiation poten-
pression after 4 d in culture (Fig. 7 b), suggesting they may tial similar to those exhibited by their counterparts in cord
have the ability to self-renew. In contrast, hCDPs rapidly lost blood (Fig. 8 b). In contrast, hGMPs, hMDPs, and hCDPs
CD34 and produced all three subsets of DCs, but did not were undetectable in the peripheral blood or tonsils (Fig. 8 a),
produce hMDPs or hGMDPs (Fig. 7 b). We conclude that indicating that in the steady-state, these progenitors are
hGMDPs gave rise to hMDPs, which produced monocytes retained in the BM. Of note, hGMDPs from BM produced
and hCDPs, which are finally restricted to pDCs and cDCs. more monocytes (P < 0.05) and less CD1c+ cDCs (P <
0.01) than their counterparts from cord blood, whereas BM
Distribution in adult hematopoietic organs hCDPs produced less pDCs than their cord blood counter-
To determine whether hGMDPs, hMDPs, and hCDPs parts (P < 0.05; Fig. 8 c). Moreover, CD141+ cDCs derived
also participate in hematopoiesis in the adult, we examined from BM hCDPs did not fully up-regulate CLEC9a expres-
human BM samples, peripheral blood, and tonsils. Adult sion (Fig. 8 b).

JEM Vol. 212, No. 3 393


Figure 8. Distribution of hGMDPs, hMDPs,
and hCDPs in adult hematopoietic organs.
(a) Representative flow cytometry plots of
gated CD45+Lin(CD3/19/56/14)CD34+ cells
show hGMDPs, hMDPs, and hCDPs in human
BM (n = 4), peripheral blood (PBMC; n = 4), and
tonsils (n = 4). (b) hGMDPs, hMDPs, and hCDPs
were purified from human BM, and 2,500 pro-
genitors were cultured in MS5+FSG for 7 d.
Flow cytometry plots of gated live CD45+ cells
show phenotype of output cells, including
granulocytes (brown), CD141+ cDCs (red),
CD1c+ cDCs (blue), monocytes (orange), and
pDCs (green). Data represent three independent
experiments. (c) Graph shows output/input cell
ratio in percentage of the indicated cells de-
rived from BM (n = 3) or cord blood (CB; n = 3)
cultures of hGMDPs, hMDPs, and hCDPs in
MS5+FSG for 7 d. Statistical significance was
determined using unpaired Students t test.
*, P < 0.05; **, P < 0.001.

DISCUSSION Identification of restricted monocyte and DC progenitors


We have delineated the developmental pathway that leads to in human BM facilitates understanding of diseases involving
production of human DCs and enumerated these cells in cord aberrant DC hematopoiesis. For example, patients harboring
blood and BM. This pathway involves sequential loss of devel- GATA2 mutations develop a newly identified form of pri-
opmental potential from a progenitor that can produce granulo- mary immunodeficiency characterized by recurrent infec-
cytes, monocytes, and DCs (hGMDPs), to one that produces tions and disseminated BCG infection after vaccination (Vinh
monocytes and DCs (hMDPs), to a committed DC progenitor et al., 2010). These patients lack B cells, NK cells, mono-
that produces cDCs and pDCs (hCDPs). Others have shown cytes, cDCs, and pDCs in the blood, but have a normal gran-
that human GMPs, CLPs, and LMPPs can produce human DCs ulocyte compartment (Bigley et al., 2011). Analysis of their
(Chicha et al., 2004; Ishikawa et al., 2007; Kohn et al., 2012). CD34+ progenitor compartment revealed an absence of
Our findings are not inconsistent with these observations be- CD45RA+ cells, including MLPs and GMPs (Bigley et al.,
cause GMPs and LMPPs are heterogeneous groups of cells that 2011), which contain hCDPs, hMDPs, and hGMDPs. The
contain subpopulations with the cell surface features of DC pro- normal granulocyte development in these patients suggests
genitors, as revealed by cross-phenotyping (Fig. 5 g). that acquiring CD45RA marks a crucial developmental step

394 Progenitor of human dendritic cells | Lee et al.


Ar ticle

for monocytes and DCs, but not for granulocytes. Addition- Although the cell surface markers that define DC precur-
ally, mutations of IRF8 cause monocyte and DC deficiency sors are not entirely conserved between mouse and human,
without affecting granulocytes (Hambleton et al., 2011), im- there is significant overlap, especially in cytokine receptor
plying an IRF8-dependent developmental block in the tran- expression. Importantly, using cytokine receptor expression
sition from hGMDPs to hMDPs. is crucial for purification of lineage-restricted progenitors.
A second example involves chronic myelogenous leuke- For instance, the originally discovered mouse MDPs
mia (CML). CML is caused by BCR-Abl translocation and is (LinCX3CR1-GFP+cKit+) lack granulocyte potential (Fogg
a myeloproliferative disease of granulocytes. CML patients et al., 2006). However, a subpopulation with 13-fold higher
show a dramatic decrease of blood pDCs and cDCs but rela- clonal efficiency to produce DC-monocyte/macrophage
tively normal numbers of monocytes (Boissel et al., 2004). (Sathe et al., 2014) can be purified using the CD115+CD135+
This is accompanied by a relative deficiency in CD34+CD123hi phenotype from the original MDP population. Moreover,
hCDPs but only slightly reduced CD123intCD45RA+ cells, the sequential loss of granulocyte and monocyte potential in
which include hMDPs and hGMDPs (Diaz-Blanco et al., humans parallels DC differentiation in the mouse. For exam-
2007), suggesting a block in the transition from hMDPs to ple, in both species, MDP is a stage marked with loss of gran-
hCDPs. The ability to purify human DC progenitors and fol- ulocyte potential and expression of CD115 (Fogg et al., 2006;
low their differentiation after transfer into NSG mice (Fig. 7 a; Waskow et al., 2008), the receptor which enables monocyte
Doulatov et al., 2010) should facilitate the study of human and macrophage development (Witmer-Pack et al., 1993;
DC differentiation in vivo. Greter et al., 2012). This degree of conservation is an indica-
Defining human DC progenitors required that we de- tion of the relative importance of this pathway during verte-
velop an efficient tissue culture method that supports devel- brate evolution.
opment of all three major human DC subtypes: CD1c+
cDCs, CD141+ cDCs, and pDCs. Although others have MATERIALS AND METHODS
Cell samples. Human umbilical cord blood and leukophoretic peripheral
shown that human CD141+ cDCs could be obtained by ex-
blood (buffy coat) were purchased from the New York Blood Center.
panding HSPCs in vitro and then subculturing them in SCF, Human BM was obtained from total hip arthroplasty by J. Schreiber at Hos-
Flt3L, GM-CSF, and IL-4 (Poulin et al., 2010) and that stro- pital for Special Surgery (New York). Tonsils were obtained from routine
mal cells could facilitate pDC development (Spits et al., 2000; tonsillectomies performed at the Babies and Childrens Hospital of Columbia-
Chicha et al., 2004; Olivier et al., 2006), defining DC pro- Presbyterian Medical Center. Informed consent was obtained from the
genitors required a culture system that would produce all patients, and/or samples were exempt from informed consent being residual
material after diagnosis and fully de-identified. All samples were collected
three types of DCs and also support development of mono-
according to protocols approved by the Institutional Review Board at Co-
cytes, granulocytes, and lymphoid cells. Addition of stromal lumbia University Medical Center (CUMC) and The Rockefeller Univer-
cells to the cocktail of Flt3L, SCF, and GM-CSF is sufficient sity. The specimens were kept on ice immediately after surgical removal.
to support efficient development of CD34+ human HSPCs Tonsil samples were minced, treated with 400 U/ml collagenase (Roche) at
to lymphoid and myeloid cells, including B cells, NK cells, 37C for 20 min, and proceeded to cell isolation. BM samples were pre-
granulocytes, and all three DC subsets without pre-expansion served in solution containing 1,000 U/ml heparin (National Drug Code
#63323-540-11) and digested in RPMI containing 20 mg/ml collagenase
of stem cells in vitro. Importantly, the DC subsets derived
IV (Sigma-Aldrich) for 15 min at 37C. After density centrifugation using
from the stromal cell cultures closely resemble primary CD1c+ Ficoll-Hypaque (GE Healthcare), aliquots of mononuclear BM cells were
cDCs, CD141+ cDCs, and pDCs obtained from the blood of frozen and stored in liquid nitrogen for future analysis.
normal donors as determined by gene expression, surface
phenotype, and cytokine production. Cell isolation and flow cytometry. Fresh mononuclear cells were iso-
Flt3L, M-CSF, GM-CSF, and IL-3 exert distinct func- lated by density centrifugation using Ficoll-Hypaque (GE Healthcare). Sam-
ples from cord blood, peripheral blood, BM, and tonsil were incubated with
tions on development and homeostasis of monocytes, cDCs,
fluorescent-labeled antibodies for direct analysis on the LSRII flow cytome-
and pDCs (Schmid et al., 2010; Merad et al., 2013). Our ters (BD) or further purification by fluorescence-activated cell sorting on the
experiments show that human CD34+ progenitors are het- Influx or FACSAria (BD), both using HeNe and argon lasers. Sorted popu-
erogeneous for expression of these cytokine receptors, but lation showed >95% purity.
they are all contained in the GMP fraction in cord blood. For purification of differentiated DCs and monocytes from peripheral
GMP can be divided into five populations based on CD115, blood and culture, cells were stained with LIVE/DEAD (Life Technologies),
CD45 (HI30, Alexa Fluor 700 [AF700]; BioLegend), CD66b (G10F5,
CD116, and CD123 expression. Three of these populations
PerCP-Cy5.5; BioLegend), CD56 (B159, Pacific Blue; BD), CD19 (HIB19,
show potential to produce granulocytes, monocytes, and APC-Cy7; BioLegend), CD14 (TuK4, Qdot-655; Invitrogen), CLEC9a
DCs: hGMDPs, hMDPs, and hCDPs. In healthy individu- (8F9, PE; BioLegend), CD1c (L161, PE-Cy7; BioLegend), CD303 (201A,
als, these hGMDPs, hMDPs, and hCDPs are found in cord FITC; BioLegend), CD123 (6H6, Brilliant Violet [BV] 510; BioLegend),
blood and in the BM. They do not circulate and are not and CD141 (AD5-14H12, APC; Miltenyi Biotec) for 40 min on ice. Alter-
found in peripheral lymphoid organs. Most of CD141+ natively, we used CD335 (9E2, BV421; BioLegend), CD11c (3.9, A700;
eBioscience), CD3 (S4.1, PE Texas Red; Invitrogen), and CD19 (SJ25-C1,
cDCs derived from BM progenitors, particularly those de- PE Texas Red; Invitrogen).
rived from hCDPs, did not up-regulate CLEC9a. This may For surface marker analysis, CD11c (3.9; BioLegend), HLA-DR (G46-6;
result from subtle differences between progenitors from adult BD), CD80 (2D10, Biotin; BioLegend), CD83 (HB15e; BioLegend), CD86
BM and cord blood. (IT2.2; BioLegend), and DC-SIGN/CD209 (DCN46; BD) were used in

JEM Vol. 212, No. 3 395


PerCP-Cy5.5, and CD123 (9F5; BD), CX3CR1 (2A9-1; BioLegend), ODN2216 (InvivoGen), 50 g/ml Poly(I:C) (InvivoGen), or 10 g/ml LPS
SIRP/CD172 (SE5A5; BioLegend), CD45RA (HI100; eBioscience), (Sigma-Aldrich). 48 h later, supernatant was collected, and IFN- and IL-
or CD1a (HI149; BD) was used in PE. 12p70 were analyzed using ELISA (Mabtech and eBioscience, respectively).
For single progenitor lineage potential and developmental hierarchy rela-
tionship experiments, CD34+ cells were first enriched from cord blood using In vivo transplantation into NSG mice. NOD.Cg-Prkdcscid-IL2rgtmlWjl/
the CD34 MicroBead kit and LS MACS magnetic columns (Miltenyi Bio- Sz (NSG) mice were developed at the Jackson Laboratory. All experiments
tec). Enriched CD34+ cells (4095% purity) were incubated with antibodies were performed according to the guidelines of the institutional animal care
against CD3 (OKT3, BV650; BioLegend), CD19 (HIB19, BV650; BioLegend), and use committee at CUMC. NSG mice were injected intraperitoneally
CD56 (HCD56, BV650; BioLegend), CD14 (Qdot-655), CD34 (581, with busulfan (30 g/g of body weight; Sigma-Aldrich) to ablate the endog-
AF700; BioLegend), CD38 (HIT2, BV421; BioLegend), CD45RA (HI100, enous hematopoietic system. Human progenitors purified from cord
BV510; BioLegend), CD123 (9F5, PE; BD), CD10 (HI10a, PE-Cy7; Bio- blood were resuspended in 10 l PBS and injected into the bone cavity.
Legend), CD116 (4H1, FITC; BioLegend), and CD115 (9-4D2-1E4, APC; Mice were injected intraperitoneally for five consecutive days with 10 g
BioLegend). From CD34+ HSPCs, hGMDPs were sorted as Lin(CD14/ Flt3L (provided by T. Keller from Celldex Therapeutics), starting at 1 d
19)CD34+CD38hiCD45RA+CD123intCD10CD115CD116, hMDPs after transfer. 7 d after transplantation, BM was harvested from recipient
as LinCD34+CD38hiCD45RA+CD123intCD10CD115+CD116, and mice and analyzed for human CD45+ cells.
hCDPs as LinCD34+CD38hiCD45RA+CD123hiCD115. For surface
phenotype analysis, CD45 (HI30, APC-Cy7; eBioscience), CD123 (6H6, Clonal analysis of progenitors. Progenitors were first sorted as separate
BV421; BioLegend), CD135 (4G8, PE; BD), and CD62L (DREG-56, PB; populations, which were then individually sorted as single cells directly into
BD) were used alternatively. 96-well plates containing mitomycin Ctreated stromal cells. The sorting effi-
For progenitor-progeny experiments, cells from either culture or NSG ciency was determined using CFSE-labeled cells, resulting in 9.5% empty
BM were stained for LIVE/DEAD, CD45 (AF700), CD14 (Qdot-655), wells, 90.5% wells containing one cell, and 0% containing more than one cell.
CD3 (OKT3, BV650; BioLegend), CD19 (HIB19, BV650; BioLegend), CD56 Each well was harvested and stained with LIVE/DEAD, CD45, CD66b,
(HCD56, BV650; BioLegend), CD1c (L161, Biotin; BioLegend), CD141 (M80, CLEC9a, CD14, CD1c, CD303, CD141, CD19, and CD56. Positive clones
PE-Cy7; BioLegend), CD34 (581, APC-Cy7; BioLegend), CD123 (9F5, PE; were determined by the detection of at least 10 human CD45+ cells using flow
BD), CD45RA (BV510), CD303 (FITC), and CD115 (APC) for 40 min on cytometry. We defined each positive clones lineage output potential by posi-
ice. Secondary staining was performed with PerCP-Cy5.5conjugated strep- tively scoring when more than seven cells were found for each cell type.
tavidin on ice for 40 min.
Array analysis of RNA expression. RNA was extracted using RNeasy
Morphological analysis. Purified hGMDPs, hMDPs, and hCDPs were ana- Plus procedures (QIAGEN) according to the manufacturers protocol. Total
lyzed by Giemsa staining of cytospin preparations. As few as 5 104 cells were RNA was checked for quantity and quality using a NanoDrop 2000c spec-
cytospun for 5 min at 800 rpm on a glass slide and then stained with the He- trophotometer (Thermo Fisher Scientific) and Experion automated electro-
macolor stain kit (Harleco) as recommended by the manufacturer. Slides were phoresis system (Bio-Rad Laboratories). Only RNA samples with a 28S/18S
then imaged on an Axioplan 2 microscope (Carl Zeiss) at 100 magnification. ratio >1.5 were processed for array analysis. 500 ng of total RNA was ampli-
fied using the Illumina Total-Prep RNA amplification kits (Applied Biosys-
Cell culture. For cord blood and BM stromal culture, MS5 cells, provided tems), as recommended by the manufacturer. 750 ng of the biotinylated cRNA
by B. Reizis (Columbia University) and originally obtained from Leibniz- was hybridized onto HumanHT-12_V4 Expression BeadChips (Illumina) at
Institut DSMZ, were maintained and passed in complete -MEM medium 58C for 20 h and quantified using an iScan System and GenomeStudio
(Invitrogen) with 10% FCS and penicillin/streptomycin (Invitrogen). After software (both Illumina).
3 h of 10 g/ml mitomycin C (Sigma-Aldrich) treatment and wash, 3.75 For analysis, signature transcripts were selected and clustered using the
104 MS5/ml was seeded per well in 96-well plates or 1.5 105 cells per well sparse hierarchical clustering tool and visualized with the HeatMap Viewer
in 24-well plates 24 h before culturing hematopoietic cells. Bulk CD34+ of the GenePattern genomic analysis platform (Reich et al., 2006). Accord-
cells or purified progenitor populations were seeded in medium containing ing to Witten and Tibshirani (2010), standard hierarchical clustering clusters
100 ng/ml Flt3L (Celldex Therapeutics), 20 ng/ml SCF (PeproTech), and observations using all of the genes, whereas sparse hierarchical clustering will
10 ng/ml GM-CSF (PeproTech). Cells were harvested between days 1 and adaptively choose a subset of the genes to use in the clustering. The goal is
14 for flow cytometry analysis. to identify a small set of genes that is relevant to the clustering and identify a
Stroma cellfree cultures were performed according to previously re- tighter and less noisy clustering of the observations using only the relevant
ported methods (Poulin et al., 2010). In brief, 5 104 purified bulk CD34+ genes. Each gene will be given a nonnegative weight, and depending on the
cells were cultured in 1 ml StemSpan medium (STEMCELL Technologies) tuning parameter used, many of the genes will have zero weights. If a genes
with penicillin/streptomycin and 100 ng/ml SCF, 100 ng/ml Flt3L, 20 ng/ml weight is zero, then it is not involved in the clustering. The weights of the
IL-3, and 20 ng/ml IL-6 for expansion. After 2 wk of culture with one me- genes can be used to rank the genes in terms of importance to the clustering
dium change at day 7, cells were washed and replated at 6.25 104 cells/ml in (the larger the weight, the more important the gene).
RPMI 1640 with glutamine, penicillin/streptomycin, 2-betamercaptoethanol For sparse hierarchical clustering, data were log scaled and standardized
(all from Invitrogen), and 10% FCS in the presence of 20 ng/ml SCF, 20 ng/ml to have mean zero and standard deviation one. The sum of gene weights was
GM-CSF, 20 ng/ml IL-4, and 100 ng/ml Flt3L (Flt3L from Celldex Thera- selected via a permutation approach, and genes and samples were clustered
peutics, all other cytokines from PeproTech) to induce differentiation of by centroid-based method. Selected genes shown in Fig. 2 d were also clus-
CD1c+ and CD141+ cDCs. Cells were cultured for 2 wk with one medium tered by the hierarchical clustering method. GSEA (Subramanian et al.,
change at day 7, before flow cytometry analysis. Absolute CD45+ cell numbers 2005) was conducted using expression profiles from culture and primary
from cultures were calculated relative to a well with a known number of bloodderived CD1c+ and CD141+ cDCs and KEGG metabolic pathways
CD45+ cells (i.e., 10,000 cells). CFU assay was performed using MethoCult gene sets. Microarray data are available in the National Center for Biotech-
(H4434; STEMCELL Technologies), containing SCF, GM-CSF, IL-3, and nology Information GEO DataSets under accession no. GSE65128.
EPO. CFU-cells (CFU-Cs) were counted after 14 d of culture.
Online supplemental material. Fig. S1 shows the gating strategy for iden-
Functional analysis of DC function. DC subsets were sorted from culture, tification and purification of monocytes, pDCs, CD1c+ cDCs, and CD141+
and 40,000 cells/100 l were plated in complete RPMI medium containing cDCs. Fig. S2 shows transcriptional profiling of all primary versus cultured
10% FCS and penicillin/streptomycin. Cells were stimulated with 1 M CpG monocyte and DC subsets. Fig. S3 shows lineage potential analysis of single

396 Progenitor of human dendritic cells | Lee et al.


Ar ticle

progenitor cultures. Figs. S4S8 show hCDP (Fig. S4), hMDP (Figs. S5 and Chicha, L., D. Jarrossay, and M.G. Manz. 2004. Clonal type I interferon-
S6), and hGMDP (Figs. S7 and S8) single cell lineage potential. Tables S1 producing and dendritic cell precursors are contained in both human
S5 are included in a separate Excel file. Table S1 shows the top 78 regulated lymphoid and myeloid progenitor populations. J. Exp. Med. 200:1519
genes in cultured and primary pDCs and monocytes. Table S2 shows the 1524. http://dx.doi.org/10.1084/jem.20040809
top 80 regulated genes in cultured and primary CD1c+ cDCs and CD141+ Crozat, K., R. Guiton, M. Guilliams, S. Henri, T. Baranek, I. Schwartz-
cDCs. Table S3 shows comparison of selected gene expression for pDCs, Cornil, B. Malissen, and M. Dalod. 2010. Comparative genomics as
monocytes, CD1c+ cDCs, and CD141+ cDCs. Table S4 lists all metabolic a tool to reveal functional equivalences between human and mouse
pathways enriched in cultured cDCs when compared with primary cDCs. dendritic cell subsets. Immunol. Rev. 234:177198. http://dx.doi.org/10
Table S5 shows the top 611 regulated genes in primary or cultured pDCs, .1111/j.0105-2896.2009.00868.x
monocytes, CD1c+ cDCs, and CD141+ cDCs. Online supplemental material Diaz-Blanco, E., I. Bruns, F. Neumann, J.C. Fischer, T. Graef, M. Rosskopf,
is available at http://www.jem.org/cgi/content/full/jem.20141442/DC1. B. Brors, S. Pechtel, S. Bork, A. Koch, et al. 2007. Molecular signature
of CD34+ hematopoietic stem and progenitor cells of patients with
CML in chronic phase. Leukemia. 21:494504. http://dx.doi.org/10
This work was inspired by Ralph M. Steinman.
.1038/sj.leu.2404549
We thank Klara Velinzon (Flow Cytometry Core Facility, Laboratory of Molecular
Doulatov, S., F. Notta, K. Eppert, L.T. Nguyen, P.S. Ohashi, and J.E. Dick.
Immunology, The Rockefeller University) for technical support with polychromatic
2010. Revised map of the human progenitor hierarchy shows the origin
flow cytometry sorting and Dr. Chiara Borsotti (Columbia Center for Translational
of macrophages and dendritic cells in early lymphoid development. Nat.
Immunology, Columbia University Medical Center) for teaching intrabone cavity
injection. We thank Peter Wilkinson (Case Western Reserve University) and
Immunol. 11:585593. http://dx.doi.org/10.1038/ni.1889
Stephanie Richards (Collaborative Genomics Center, Vaccine and Gene Therapy
Fogg, D.K., C. Sibon, C. Miled, S. Jung, P. Aucouturier, D.R. Littman,
Institute of Florida) for their input in microarray analysis. We thank Heidi Schreiber A. Cumano, and F. Geissmann. 2006. A clonogenic bone marrow pro-
(Laboratory of Molecular Immunology, The Rockefeller University) for help with genitor specific for macrophages and dendritic cells. Science. 311:8387.
human BM protocol and Joseph Schreiber for providing human BM specimen. We http://dx.doi.org/10.1126/science.1117729
thank Tibor Keller for giving us Flt3L. Galy, A., M. Travis, D. Cen, and B. Chen. 1995. Human T, B, natural
Research reported in this publication was supported by the Empire State Stem killer, and dendritic cells arise from a common bone marrow progeni-
Cell Fund through New York State Department of Health Contract #C029562 tor cell subset. Immunity. 3:459473. http://dx.doi.org/10.1016/1074-
(to K. Liu), Helmsley Foundation (to K. Liu), National Institutes of Health (NIH) grants 7613(95)90175-2
AI101251 (to K. Liu) and NS084776 (to S. Puhr), Iris and Junming Le Foundation Geissmann, F., M.G. Manz, S. Jung, M.H. Sieweke, M. Merad, and K. Ley.
(to G. Breton), NIH grant 1U19AI111825-01 (to M.C. Nussenzweig), Clinical and 2010. Development of monocytes, macrophages, and dendritic cells.
Translational Science Awards, The Rockefeller University Center for Clinical and Science. 327:656661. http://dx.doi.org/10.1126/science.1178331
Translational Science (RUCCTS) grant no. UL1RR024143 from the National Center Ginhoux, F., K. Liu, J. Helft, M. Bogunovic, M. Greter, D. Hashimoto, J.
for Research Resources, NIH, and NIH grant AI13013. The RUCCTS is supported, in Price, N. Yin, J. Bromberg, S.A. Lira, et al. 2009. The origin and devel-
part, by a Clinical and Translational Science Award (CTSA) and the National Center opment of nonlymphoid tissue CD103+ DCs. J. Exp. Med. 206:3115
for Advancing Translational Sciences (NCATS), part of the NIH. M.C. Nussenzweig is 3130. http://dx.doi.org/10.1084/jem.20091756
a Howard Hughes Medical Institute investigator. Granelli-Piperno, A., I. Shimeliovich, M. Pack, C. Trumpfheller, and R.M.
The authors declare no competing financial interests. Steinman. 2006. HIV-1 selectively infects a subset of nonmaturing
BDCA1-positive dendritic cells in human blood. J. Immunol. 176:991
998. http://dx.doi.org/10.4049/jimmunol.176.2.991
Submitted: 30 July 2014
Accepted: 28 January 2015
Greter, M., I. Lelios, P. Pelczar, G. Hoeffel, J. Price, M. Leboeuf, T.M.
Kndig, K. Frei, F. Ginhoux, M. Merad, and B. Becher. 2012. Stroma-
derived interleukin-34 controls the development and maintenance of
REFERENCES Langerhans cells and the maintenance of microglia. Immunity. 37:1050
Akashi, K., D. Traver, T. Miyamoto, and I.L. Weissman. 2000. A clono- 1060. http://dx.doi.org/10.1016/j.immuni.2012.11.001
genic common myeloid progenitor that gives rise to all myeloid lin- Hambleton, S., S. Salem, J. Bustamante, V. Bigley, S. Boisson-Dupuis,
eages. Nature. 404:193197. http://dx.doi.org/10.1038/35004599 J. Azevedo, A. Fortin, M. Haniffa, L. Ceron-Gutierrez, C.M. Bacon, et al.
Bigley,V., M. Haniffa, S. Doulatov, X.N. Wang, R. Dickinson, N. McGovern, 2011. IRF8 mutations and human dendritic-cell immunodeficiency. N. Engl.
L. Jardine, S. Pagan, I. Dimmick, I. Chua, et al. 2011. The human syn- J. Med. 365:127138. http://dx.doi.org/10.1056/NEJMoa1100066
drome of dendritic cell, monocyte, B and NK lymphoid deficiency. Haniffa, M., A. Shin, V. Bigley, N. McGovern, P. Teo, P. See, P.S. Wasan,
J. Exp. Med. 208:227234. http://dx.doi.org/10.1084/jem.20101459 X.N. Wang, F. Malinarich, B. Malleret, et al. 2012. Human tissues con-
Boissel, N., P. Rousselot, E. Raffoux, J.M. Cayuela, O. Maarek, D. Charron, tain CD141hi cross-presenting dendritic cells with functional homology
L. Degos, H. Dombret, A. Toubert, and D. Rea. 2004. Defective blood to mouse CD103+ nonlymphoid dendritic cells. Immunity. 37:6073.
dendritic cells in chronic myeloid leukemia correlate with high plas- http://dx.doi.org/10.1016/j.immuni.2012.04.012
matic VEGF and are not normalized by imatinib mesylate. Leukemia. Hettinger, J., D.M. Richards, J. Hansson, M.M. Barra, A.C. Joschko, J.
18:16561661. http://dx.doi.org/10.1038/sj.leu.2403474 Krijgsveld, and M. Feuerer. 2013. Origin of monocytes and macro-
Breton, G., J. Lee, Y.J. Zhou, J.J. Schreiber, T. Keler, S. Puhr, N. phages in a committed progenitor. Nat. Immunol. 14:821830. http://
Anandasabapathy, S. Schlesinger, M. Caskey, K. Liu, and M.C. dx.doi.org/10.1038/ni.2638
Nussenzweig. 2015. Circulating precursors of human CD1c+ and Ishikawa, F., H. Niiro, T. Iino, S. Yoshida, N. Saito, S. Onohara, T.
CD141+ dendritic cells. J. Exp. Med. 212:401413. http://dx.doi.org/ Miyamoto, H. Minagawa, S. Fujii, L.D. Shultz, et al. 2007. The devel-
10.1084/jem.20141441 opmental program of human dendritic cells is operated independently of
Chang, C.C., A. Wright, and J. Punnonen. 2000. Monocyte-derived CD1a+ conventional myeloid and lymphoid pathways. Blood. 110:35913660.
and CD1a dendritic cell subsets differ in their cytokine production http://dx.doi.org/10.1182/blood-2007-02-071613
profiles, susceptibilities to transfection, and capacities to direct Th Ito, T., Y.H. Wang, and Y.J. Liu. 2005. Plasmacytoid dendritic cell precursors/
cell differentiation. J. Immunol. 165:35843591. http://dx.doi.org/10 type I interferon-producing cells sense viral infection by Toll-like
.4049/jimmunol.165.7.3584 receptor (TLR) 7 and TLR9. Springer Semin. Immunopathol. 26:221
Cheong, C., I. Matos, J.H. Choi, D.B. Dandamudi, E. Shrestha, M.P. Longhi, 229. http://dx.doi.org/10.1007/s00281-004-0180-4
K.L. Jeffrey, R.M. Anthony, C. Kluger, G. Nchinda, et al. 2010. Microbial Itoh, K., H. Tezuka, H. Sakoda, M. Konno, K. Nagata, T. Uchiyama, H.
stimulation fully differentiates monocytes to DC-SIGN/CD209+ den- Uchino, and K.J. Mori. 1989. Reproducible establishment of hemo-
dritic cells for immune T cell areas. Cell. 143:416429. http://dx.doi.org/ poietic supportive stromal cell lines from murine bone marrow. Exp.
10.1016/j.cell.2010.09.039 Hematol. 17:145153.

JEM Vol. 212, No. 3 397


Kadowaki, N., S. Ho, S. Antonenko, R.W. Malefyt, R.A. Kastelein, F. Bazan, Onai, N., A. Obata-Onai, M.A. Schmid, and M.G. Manz. 2007a. Flt3 in
and Y.J. Liu. 2001. Subsets of human dendritic cell precursors express regulation of type I interferon-producing cell and dendritic cell develop-
different toll-like receptors and respond to different microbial antigens. ment. Ann. N. Y. Acad. Sci. 1106:253261. http://dx.doi.org/10.1196/
J. Exp. Med. 194:863870. http://dx.doi.org/10.1084/jem.194.6.863 annals.1392.015
Kalscheuer, H., N. Danzl, T. Onoe, T. Faust, R. Winchester, R. Goland, E. Onai, N., A. Obata-Onai, M.A. Schmid, T. Ohteki, D. Jarrossay, and
Greenberg, T.R. Spitzer, D.G. Savage, H. Tahara, et al. 2012. A model for M.G. Manz. 2007b. Identification of clonogenic common Flt3+M-
personalized in vivo analysis of human immune responsiveness. Sci. Transl. CSFR+ plasmacytoid and conventional dendritic cell progenitors in
Med. 4:125ra30. http://dx.doi.org/10.1126/scitranslmed.3003481 mouse bone marrow. Nat. Immunol. 8:12071216. http://dx.doi.org/
Karsunky, H., M. Merad, A. Cozzio, I.L. Weissman, and M.G. Manz. 2003. 10.1038/ni1518
Flt3 ligand regulates dendritic cell development from Flt3+ lymphoid Onai, N., K. Kurabayashi, M. Hosoi-Amaike, N. Toyama-Sorimachi, K.
and myeloid-committed progenitors to Flt3+ dendritic cells in vivo. Matsushima, K. Inaba, and T. Ohteki. 2013. A clonogenic progenitor with
J. Exp. Med. 198:305313. http://dx.doi.org/10.1084/jem.20030323 prominent plasmacytoid dendritic cell developmental potential. Immunity.
Kingston, D., M.A. Schmid, N. Onai, A. Obata-Onai, D. Baumjohann, 38:943957. http://dx.doi.org/10.1016/j.immuni.2013.04.006
and M.G. Manz. 2009. The concerted action of GM-CSF and Flt3- Patterson, S., H. Donaghy, P. Amjadi, B. Gazzard, F. Gotch, and P.
ligand on in vivo dendritic cell homeostasis. Blood. 114:835843. http:// Kelleher. 2005. Human BDCA-1-positive blood dendritic cells differ-
dx.doi.org/10.1182/blood-2009-02-206318 entiate into phenotypically distinct immature and mature populations
Kohn, L.A., Q.L. Hao, R. Sasidharan, C. Parekh, S. Ge, Y. Zhu, H.K. Mikkola, in the absence of exogenous maturational stimuli: differentiation failure
and G.M. Crooks. 2012. Lymphoid priming in human bone marrow in HIV infection. J. Immunol. 174:82008209. http://dx.doi.org/10
begins before expression of CD10 with upregulation of L-selectin. .4049/jimmunol.174.12.8200
Nat. Immunol. 13:963971. http://dx.doi.org/10.1038/ni.2405 Poulin, L.F., M. Salio, E. Griessinger, F. Anjos-Afonso, L. Craciun,
Lindstedt, M., K. Lundberg, and C.A. Borrebaeck. 2005. Gene family cluster- J.L. Chen, A.M. Keller, O. Joffre, S. Zelenay, E. Nye, et al. 2010.
ing identifies functionally associated subsets of human in vivo blood and Characterization of human DNGR-1+ BDCA3+ leukocytes as putative
tonsillar dendritic cells. J. Immunol. 175:48394846. http://dx.doi.org/10 equivalents of mouse CD8+ dendritic cells. J. Exp. Med. 207:1261
.4049/jimmunol.175.8.4839 1271. http://dx.doi.org/10.1084/jem.20092618
Liu, K., G.D. Victora, T.A. Schwickert, P. Guermonprez, M.M. Meredith, Poulin, L.F., Y. Reyal, H. Uronen-Hansson, B.U. Schraml, D. Sancho,
K. Yao, F.F. Chu, G.J. Randolph, A.Y. Rudensky, and M. Nussenzweig. K.M. Murphy, U.K. Hkansson, L.F. Moita, W.W. Agace, D. Bonnet,
2009. In vivo analysis of dendritic cell development and homeostasis. and C. Reis e Sousa. 2012. DNGR-1 is a specific and universal marker
Science. 324:392397. of mouse and human Batf3-dependent dendritic cells in lymphoid and
Liu, Y.J. 2005. IPC: professional type 1 interferon-producing cells and plas- nonlymphoid tissues. Blood. 119:60526062. http://dx.doi.org/10.1182/
macytoid dendritic cell precursors. Annu. Rev. Immunol. 23:275306. blood-2012-01-406967
http://dx.doi.org/10.1146/annurev.immunol.23.021704.115633 Proietto, A.I., D. Mittag, A.W. Roberts, N. Sprigg, and L. Wu. 2012. The
MacDonald, K.P., D.J. Munster, G.J. Clark, A. Dzionek, J. Schmitz, and D.N. equivalents of human blood and spleen dendritic cell subtypes can be
Hart. 2002. Characterization of human blood dendritic cell subsets. Blood. generated in vitro from human CD34+ stem cells in the presence of fms-
100:45124520. http://dx.doi.org/10.1182/blood-2001-11-0097 like tyrosine kinase 3 ligand and thrombopoietin. Cell. Mol. Immunol.
Manz, M.G., T. Miyamoto, K. Akashi, and I.L. Weissman. 2002. 9:446454. http://dx.doi.org/10.1038/cmi.2012.48
Prospective isolation of human clonogenic common myeloid progeni- Reich, M., T. Liefeld, J. Gould, J. Lerner, P. Tamayo, and J.P. Mesirov.
tors. Proc. Natl. Acad. Sci. USA. 99:1187211877. http://dx.doi.org/10 2006. GenePattern 2.0. Nat. Genet. 38:500501. http://dx.doi.org/
.1073/pnas.172384399 10.1038/ng0506-500
McKenna, H.J., K.L. Stocking, R.E. Miller, K. Brasel, T. De Smedt, E. Robbins, S.H., T. Walzer, D. Dembl, C. Thibault, A. Defays, G. Bessou,
Maraskovsky, C.R. Maliszewski, D.H. Lynch, J. Smith, B. Pulendran, H. Xu, E. Vivier, M. Sellars, P. Pierre, et al. 2008. Novel insights into
et al. 2000. Mice lacking flt3 ligand have deficient hematopoiesis af- the relationships between dendritic cell subsets in human and mouse
fecting hematopoietic progenitor cells, dendritic cells, and natural killer revealed by genome-wide expression profiling. Genome Biol. 9:R17.
cells. Blood. 95:34893497. http://dx.doi.org/10.1186/gb-2008-9-1-r17
Merad, M., P. Sathe, J. Helft, J. Miller, and A. Mortha. 2013. The dendritic Sallusto, F., and A. Lanzavecchia. 1994. Efficient presentation of soluble
cell lineage: ontogeny and function of dendritic cells and their subsets in antigen by cultured human dendritic cells is maintained by granulocyte/
the steady state and the inflamed setting. Annu. Rev. Immunol. 31:563 macrophage colony-stimulating factor plus interleukin 4 and down-
604. http://dx.doi.org/10.1146/annurev-immunol-020711-074950 regulated by tumor necrosis factor alpha. J. Exp. Med. 179:11091118.
Mittag, D., A.I. Proietto, T. Loudovaris, S.I. Mannering, D. Vremec, K. http://dx.doi.org/10.1084/jem.179.4.1109
Shortman, L. Wu, and L.C. Harrison. 2011. Human dendritic cell sub- Sathe, P., D. Metcalf, D. Vremec, S.H. Naik, W.Y. Langdon, N.D.
sets from spleen and blood are similar in phenotype and function but Huntington, L. Wu, and K. Shortman. 2014. Lymphoid tissue and
modified by donor health status. J. Immunol. 186:62076217. http:// plasmacytoid dendritic cells and macrophages do not share a common
dx.doi.org/10.4049/jimmunol.1002632 macrophage-dendritic cell-restricted progenitor. Immunity. 41:104115.
Naik, S.H., D. Metcalf, A. van Nieuwenhuijze, I. Wicks, L. Wu, M. http://dx.doi.org/10.1016/j.immuni.2014.05.020
OKeeffe, and K. Shortman. 2006. Intrasplenic steady-state dendritic Schmid, M.A., D. Kingston, S. Boddupalli, and M.G. Manz. 2010. Instructive
cell precursors that are distinct from monocytes. Nat. Immunol. 7:663 cytokine signals in dendritic cell lineage commitment. Immunol. Rev.
671. http://dx.doi.org/10.1038/ni1340 234:3244. http://dx.doi.org/10.1111/j.0105-2896.2009.00877.x
Naik, S.H., P. Sathe, H.Y. Park, D. Metcalf, A.I. Proietto, A. Dakic, S. Schmidl, C., K. Renner, K. Peter, R. Eder, T. Lassmann, P.J.
Carotta, M. OKeeffe, M. Bahlo, A. Papenfuss, et al. 2007. Development Balwierz, M. Itoh, S. Nagao-Sato, H. Kawaji, P. Carninci, et al.
of plasmacytoid and conventional dendritic cell subtypes from single FANTOM consortium. 2014. Transcription and enhancer profiling
precursor cells derived in vitro and in vivo. Nat. Immunol. 8:12171226. in human monocyte subsets. Blood. 123:e90e99. http://dx.doi.org/
http://dx.doi.org/10.1038/ni1522 10.1182/blood-2013-02-484188
Olivier, A., E. Lauret, P. Gonin, and A. Galy. 2006. The Notch ligand Shortman, K., and S.H. Naik. 2007. Steady-state and inflammatory dendritic-
delta-1 is a hematopoietic development cofactor for plasmacytoid cell development. Nat. Rev. Immunol. 7:1930. http://dx.doi.org/10.1038/
dendritic cells. Blood. 107:26942701. http://dx.doi.org/10.1182/blood- nri1996
2005-03-0970 Spits, H., F. Couwenberg, A.Q. Bakker, K. Weijer, and C.H. Uittenbogaart.
Olweus, J., A. BitMansour, R. Warnke, P.A. Thompson, J. Carballido, L.J. 2000. Id2 and Id3 inhibit development of CD34+ stem cells into preden-
Picker, and F. Lund-Johansen. 1997. Dendritic cell ontogeny: a human dritic cell (pre-DC)2 but not into pre-DC1. Evidence for a lymphoid
dendritic cell lineage of myeloid origin. Proc. Natl. Acad. Sci. USA. origin of pre-DC2. J. Exp. Med. 192:17751784. http://dx.doi.org/10
94:1255112556. http://dx.doi.org/10.1073/pnas.94.23.12551 .1084/jem.192.12.1775

398 Progenitor of human dendritic cells | Lee et al.


Ar ticle

Subramanian, A., P. Tamayo, V.K. Mootha, S. Mukherjee, B.L. Ebert, M.A. Witmer-Pack, M.D., D.A. Hughes, G. Schuler, L. Lawson, A. McWilliam,
Gillette, A. Paulovich, S.L. Pomeroy, T.R. Golub, E.S. Lander, and J.P. K. Inaba, R.M. Steinman, and S. Gordon. 1993. Identification of mac-
Mesirov. 2005. Gene set enrichment analysis: a knowledge-based approach rophages and dendritic cells in the osteopetrotic (op/op) mouse. J. Cell
for interpreting genome-wide expression profiles. Proc. Natl. Acad. Sci. Sci. 104:10211029.
USA. 102:1554515550. http://dx.doi.org/10.1073/pnas.0506580102 Witten, D.M., and R. Tibshirani. 2010. A framework for feature selection in
Vinh, D.C., S.Y. Patel, G. Uzel, V.L. Anderson, A.F. Freeman, K.N. clustering. J. Am. Stat. Assoc. 105:713726. http://dx.doi.org/10.1198/
Olivier, C. Spalding, S. Hughes, S. Pittaluga, M. Raffeld, et al. 2010. jasa.2010.tm09415
Autosomal dominant and sporadic monocytopenia with susceptibility Xu, Y., Y. Zhan, A.M. Lew, S.H. Naik, and M.H. Kershaw. 2007.
to mycobacteria, fungi, papillomaviruses, and myelodysplasia. Blood. Differential development of murine dendritic cells by GM-CSF versus
115:15191529. http://dx.doi.org/10.1182/blood-2009-03-208629 Flt3 ligand has implications for inflammation and trafficking. J. Immunol.
Waskow, C., K. Liu, G. Darrasse-Jze, P. Guermonprez, F. Ginhoux, M. 179:75777584. http://dx.doi.org/10.4049/jimmunol.179.11.7577
Merad, T. Shengelia, K. Yao, and M. Nussenzweig. 2008. The receptor Ziegler-Heitbrock, L., P. Ancuta, S. Crowe, M. Dalod, V. Grau, D.N.
tyrosine kinase Flt3 is required for dendritic cell development in periph- Hart, P.J. Leenen, Y.J. Liu, G. MacPherson, G.J. Randolph, et al.
eral lymphoid tissues. Nat. Immunol. 9:676683. http://dx.doi.org/10 2010. Nomenclature of monocytes and dendritic cells in blood. Blood.
.1038/ni.1615 116:e74e80. http://dx.doi.org/10.1182/blood-2010-02-258558

JEM Vol. 212, No. 3 399


Article

Circulating precursors of human CD1c+


and CD141+ dendritic cells
Galle Breton,2* Jaeyop Lee,1* Yu Jerry Zhou,1 Joseph J. Schreiber,5 Tibor Keler,6
Sarah Puhr,1 Niroshana Anandasabapathy,2,4 Sarah Schlesinger,2
Marina Caskey,2 Kang Liu,1** and Michel C. Nussenzweig2,3**
1Columbia University Medical Center, Department of Microbiology and Immunology, New York, NY 10032
2Laboratory of Molecular Immunology, 3Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065
4Department of Dermatology Brigham and Womens Hospital, Boston, MA 02115
5Hospital for Special Surgery, New York, NY 10021
6Celldex Therapeutics, Hampton, NJ 08827

Two subsets of conventional dendritic cells (cDCs) with distinct cell surface markers and
functions exist in mouse and human. The two subsets of cDCs are specialized antigen-
presenting cells that initiate T cell immunity and tolerance. In the mouse, a migratory cDC
precursor (pre-CDC) originates from defined progenitors in the bone marrow (BM). Small
numbers of short-lived pre-CDCs travel through the blood and replace cDCs in the peripheral
organs, maintaining homeostasis of the highly dynamic cDC pool. However, the identity and
distribution of the immediate precursor to human cDCs has not been defined. Using a tissue
culture system that supports the development of human DCs, we identify a migratory precur-
sor (hpre-CDC) that exists in human cord blood, BM, blood, and peripheral lymphoid organs.
hpre-CDCs differ from premonocytes that are restricted to the BM. In contrast to earlier
progenitors with greater developmental potential, the hpre-CDC is restricted to producing
CD1c+ and CD141+ Clec9a+ cDCs. Studies in human volunteers demonstrate that hpre-CDCs
are a dynamic population that increases in response to levels of circulating Flt3L.

CORRESPONDENCE Conventional DCs (cDCs) induce immunity et al., 2006; Bogunovic et al., 2009; Ginhoux
Kang Liu: or tolerance by capturing, processing, and pre- et al., 2009; Liu et al., 2009;Varol et al., 2009).
kl2529@columbia.edu
senting antigen to T lymphocytes (Banchereau Mouse cDCs can be divided into two major
OR
and Michel C. Nussenzweig: and Steinman, 1998). In the mouse, cDCs are subsets, CD11b+ DCs and CD8+/CD103+ DCs
nussen@rockefeller.edu short-lived cells, whose homeostasis in lymphoid that differ in their microanatomic localization,
and nonlymphoid tissues is critically dependent cell surface antigen expression, antigen-processing
Abbreviation used: cDCs, con-
ventional DCs; CDP, common on continual replenishment from circulating activity, and ability to contribute to immune re-
dendritic progenitor; pre-CDC, pre-CDC (Liu et al., 2007; Liu and Nussenzweig, sponses to specific pathogens (Merad et al., 2013;
cDC precursor; MS5+FSG, 2010). Murine pre-CDCs are BM-derived cells Murphy, 2013). Despite these important differ-
MS5 stromal cells with Flt3L,
SCF, and GM-CSF cytokines.
that are present in very small numbers in the ences, both CD11b+ and CD8+/CD103+ cDC
blood but increase in response to Flt3L injec- subsets of mouse DCs are derived from the same
tion (Liu et al., 2007, 2009). pre-CDCs have a immediate precursor (pre-CDC) that expresses
very short dwell time in the blood, 65% of these CD135 (Flt3), the receptor for Flt3L, a cyto-
cells leave the circulation within 1 min after leav- kine that is critical to DC development in vivo
ing the BM (Liu et al., 2007, 2009). Upon leav- (McKenna et al., 2000; Waskow et al., 2008).
ing the circulation, pre-CDCs seed tissues where Similar to the mouse, humans have two major
they differentiate to cDCs, which divide further subsets of cDCs. CD141 (BDCA3)+Clec9a+ DCs
under the control of Flt3L (Liu et al., 2007, 2009). (CD141+ cDC herein) appear to be the human
Thus, in addition to the BM and blood, mouse counterpart of mouse CD8+/CD103+ DCs,
pre-CDCs are also found in peripheral lym- expressing XCR1, Clec9a, IRF8, and TLR3
phoid organs and nonlymphoid tissues (Naik
2015 Breton et al. This article is distributed under the terms of an Attribution
NoncommercialShare AlikeNo Mirror Sites license for the first six months after
*G. Breton and J. Lee contributed equally to this paper. the publication date (see http://www.rupress.org/terms). After six months it is
available under a Creative Commons License (AttributionNoncommercialShare
**K. Liu and M. Nussenzweig contributed equally to Alike 3.0 Unported license, as described at http://creativecommons.org/licenses/
this paper. by-nc-sa/3.0/).

The Rockefeller University Press $30.00


J. Exp. Med. 2015 Vol. 212 No. 3 401413 401
www.jem.org/cgi/doi/10.1084/jem.20141441
and producing IL-12 (Robbins et al., 2008; Bachem et al., receptor) is expressed only on monocytes, and CD117 (SCF
2010; Crozat et al., 2010; Jongbloed et al., 2010; Poulin et al., receptor) is found on the surface of CD141+ cDCs and most
2010; Haniffa et al., 2012). CD1c (BDCA1)+ cDCs appear to be CD1c+ cDCs, but not on pDCs or monocytes (Fig. 1).
more closely related to mouse CD11b+ DCs, expressing IRF4, CD45RA, a marker expressed by several different hematopo-
inducing Th17 differentiation upon A. fumigatus challenge, etic cell progenitors including granulocyte monocyte pro-
and imprinting intraepithelial homing of T cells (Robbins genitors (GMP) and common lymphoid progenitors (CLP;
et al., 2008; Crozat et al., 2010; Schlitzer et al., 2013;Yu et al., Chicha et al., 2004; Doulatov et al., 2010), is highly expressed
2013). In the mouse, the superior ability of CD8+/CD103+ only on pDCs (Fig. 1). Finally, CD34 is expressed on early
DCs to cross-present exogenous antigens to CD8+ T cells is DC progenitors (Chicha et al., 2004; Doulatov et al., 2010;
attributed to both differential antigen uptake (Kamphorst et al., Lee et al., 2015), but not on fully differentiated DCs or mono-
2010) and to increased expression of proteins and enzymes cytes, suggesting that loss of CD34 would precede cDC de-
that facilitate MHC class I presentation (Dudziak et al., 2007). velopment (Fig. 1). Collectively, the data suggests that if a
Human CD141+ cDCs are more efficient than CD1c+ cDCs hpre-CDC exists in human blood it may be found in the CD
in cross-presentation (Bachem et al., 2010; Crozat et al., 2010; 34CD115CD116+CD117+CD135+ fraction.
Jongbloed et al., 2010; Poulin et al., 2010), but this difference To try to identify hpre-CDCs in cord blood, we ex-
appears to result from differences in antigen uptake and cyto- cluded terminally differentiated lymphoid cells (CD19+CD3+
kine activation rather than a specialized cell-intrinsic program CD56+), granulocytes (CD66b+), monocytes (CD14+), pDCs
( Segura et al., 2012; Cohn et al., 2013; Nizzoli et al., 2013). (CD303/BDCA2+), and cDCs (CD1c+ and CD141+). The
Both CD1c+ cDCs and CD141+ cDCs are present in remaining CD45+ cells were subdivided into 4 groups by ex-
human blood and peripheral tissues. Each subset in the blood pression of CD34, CD117, and CD135, as follows: CD34+
resembles its tissue counterpart in gene expression but appears CD117+ (CD34+), CD34CD117 (CD117), CD34
less differentiated (Haniffa et al., 2012; Segura et al., 2012; CD117+CD135 (CD135), and CD34CD117+CD135+
Schlitzer et al., 2013). These observations are consistent with (CD135+; Fig. 2 a). Pools of 1,000 purified cells were cultured
the idea that less differentiated human cDCs travel through the on MS5 stromal cells with Flt3L, SCF, and GM-CSF cytokines
blood to replenish the cDC pool in the peripheral tissues (MS5+FSG herein) under conditions that support DC devel-
(Collin et al., 2011; Segura et al., 2012; Haniffa et al., 2013). opment in vitro (Lee et al., 2015). Only CD34+CD117+ and
Others have postulated the existence of a less differentiated cir- CD34CD117+CD135+ cells produced identifiable progeny
culating DC progenitor based on absence of CD11c, expres- (Fig. 2 b). As expected, CD34+CD117+ cells, which should
sion of CD123, and response to Flt3L (ODoherty et al., 1994; contain the least committed progenitors because they retain
Pulendran et al., 2000), but the progenitor potential of these CD34 expression, developed into monocytes, granulocytes,
putative precursors that produced large amounts of IFN- was pDCs, CD1c+ and CD141+ cDCs, and B cells, indicating
never tested directly and they appear to correspond at least in that they are indeed multipotent (Fig. 2 b and not depicted).
part to plasmacytoid DCs (Grouard et al., 1997; Siegal et al., In contrast, CD34CD117+CD135+ cells were restricted
1999). Thus, whether there is an immediate circulating pre- to CD1c+ and CD141+ cDCs and monocytes, suggesting
cursor restricted to human immature and mature CD1c+ and that this pool of cells contains the immediate precursor to
CD141+ cDCs is not known. human cDCs and monocytes (Fig. 2 b). To further purify
Here, we report the existence of a migratory pre-CDC in the hpre-CDC, we fractionated the CD34CD117+CD135+
humans (hpre-CDC) that develops from committed DC pro- pool based on expression of CD116, CD115, and CD45RA
genitors (hCDPs) in the BM (Lee et al., 2015) and is the im- (Chicha et al., 2004; Doulatov et al., 2010) and assayed
mediate precursor of both CD1c+ and CD141+ cDCs, but their potential in MS5+FSG cultures (Fig. 2 c). Whereas
not pDCs or monocytes. hpre-CDCs are present in BM, cord, CD116+CD45RA+CD115+ cells produced primarily CD14+
and peripheral blood, as well as peripheral lymphoid tissues. CD1c monocytes, a small number of CD1c+CD14+ cDCs
In studies of human volunteers, Flt3L injection induces ex- (Patterson et al., 2005; Granelli-Piperno et al., 2006), and no
pansion of hpre-CDCs in the circulation. Thus, human cDC CD141+ cDCs; CD116+CD45RA+CD115 cells yielded
precursors constitute a dynamic circulating population whose primarily CD1c+ and CD141+ cDCs and only rare mono-
homeostasis is regulated by Flt3L, a cytokine that is responsive cytes and pDCs (Fig. 2 d). These results were reproducible
to inflammatory and infectious agents. in multiple different human donors (Fig. 2 e). Therefore,
CD116+CD45RA+CD115 cells show the restricted de-
RESULTS velopmental potential expected of hpreDCs and CD116+
Identification of an immediate precursor to human cDCs CD45RA+CD115+ cells of premonocytes. hpre-CDCs ex-
To define cell surface markers that might be expressed by cir- press high HLA-DR, an intermediate level of CD123, and a
culating hpre-CDCs, we examined DCs and monocytes for low level of CD11c (Fig. 2 f). Morphologically, hpre-CDCs
expression of cytokine receptors that are found on BM DC are mononuclear cells that present a distinctive formation of
progenitors (Lee et al., 2015). Whereas CD116 (GM-CSF re- multilobulated nucleus and a few veiled processes but do not
ceptor) is expressed on differentiated monocytes and DCs, possess the membrane extensions typically associated with
CD135 (Flt3L receptor) is restricted to DCs, CD115 (M-CSF cDCs (Fig. 2 g).

402 Circulating precursors of human dendritic cells | Breton et al.


Ar ticle

Figure 1. Expression of cytokine receptors on


DCs and monocytes. Flow cytometry plots show
expression of CD117, CD135, CD115, CD123, CD45RA,
CD34, and CD116 on gated CD3CD19CD56 cells
(gray), differentiated monocytes (CD14+ orange),
pDCs (CD303+ green), CD1c+ cDCs (blue), and CD141+
cDCs (red) from peripheral blood. Table summarizes
results of flow cytometry.

hpre-CDCs in adult blood and lymphoid organs analyze cord blood. LinCD34CD117+CD135+ cells in
In the mouse, pre-CDCs are found in circulation and in tis- the BM resembled those in the cord blood and contained
sues. To determine the physiological distribution of hpre- CD116+CD115 hpre-CDC-like cells and CD116+CD115+
CDCs in adult humans, we examined BM, peripheral blood, premonocyte-like cells (Fig. 3 a). These hpre-CDC-like (Lin
and tonsils using the same cocktail of surface markers used to CD34CD117+CD135+CD116+CD115) cells were also

JEM Vol. 212, No. 3 403


Figure 2. Screening of populations in the cord blood for committed cDC lineage potential. (a) Flow cytometry plots show gating of CD45+CD3
CD19CD56CD14CD66bCD1cCD141CD303 cells in human cord blood can be divided into four populations based on CD117, CD34, and CD135:
CD34+CD117+ (CD34+), CD34CD117 (CD117), CD34CD117+CD135 (CD135), and CD34CD117+CD135+ (CD135+). Numbers indicate the frequency
of respective gates. (b) Differentiation potential of 1,000 purified cells from each of 4 populations indicated in (a) in MS5+FSG cultures for 7 d. Flow cy-
tometry plots show gated live human CD45+ cells. (c) Flow cytometry plots show CD135+ cells indicated as in (a) can be further separated into 4 populations

404 Circulating precursors of human dendritic cells | Breton et al.


Ar ticle

detected in the peripheral blood and the tonsil. In contrast, with this notion, purified blood CD1c+ and CD141+ cDCs also
the premonocyte-like (LinCD34CD117+CD135+CD116 divided 34 times in culture (Fig. 4 c).Thus, hpre-CDCs have
+CD115+) cells are lacking in the peripheral blood and tonsil a more limited proliferative capacity than CD34+ HSPCs
(Fig. 3 a). To determine the developmental potential of the in vitro. Nevertheless, a single hpre-CDC may be able to give
hpre-CDC-like cells in different tissues, we purified them rise to as many as 256 cDCs.
and cultured them under conditions that support DC devel- To determine the clonal efficiency of hpre-CDCs, we
opment. Although occasional hpre-CDCs remained undiffer- compared them to CD34+ HSPCs and progenitors in limit-
entiated, all cultures, irrespective of hpre-CDC origin from ing dilution assays. Whereas 1 in 4.57 CD34+ cells produced
cord blood, BM, blood, or tonsils, predominantly produced CD45+ cells (i.e., granulocytes, monocytes, DCs, or lym-
CD1c+ and CD141+ cDCs and few, if any, CD14+CD1c phoid cells), 1 in 7.84 and 1 in 6.55 hpre-CDCs from cord
monocytes (Fig. 3 b). CD1c+ cDCs produced in our culture sys- blood or peripheral blood, respectively, produced CD45+
tem can express some CD14, but gene array data show that these cells during the culture period (Fig. 5 a). This is in line with
CD1c+ CD14+ cells are cDCs and not monocytes (Lee et al., our observation that some hpre-CDCs remained undifferen-
2015). In contrast, the CD115+CD116+ premonocytes in the tiated in our cultures (Fig. 3 b). Although CD34+ HSPCs had
BM, which are absent from the blood and tonsils, produced higher clonal potential, only 12 out of 43 (28%) were able to
primarily monocytes and no CD141+ cDCs (Fig. 3 c). Cells produce cDCs (Fig. 5 c and Fig. S1), and both cord blood
showing the hpre-CDC phenotype constitute 0.008% (range, and peripheral blood hpre-CDCs generated mainly cDCs
0.0010.016%) of the CD45+ cells in cord blood, 0.117% (Fig. 5 c, Fig. S2, and Fig. S3). Individual hpre-CDCs pro-
(range, 0.0560.188%) in the BM, 0.001% (range of 0.001 duced either CD1c+ cDCs or CD141+ cDCs or both (Fig. 5 b).
0.002%) in the peripheral blood, and 0.001% (range, 0.001 Of 81 cells assayed, 88% of all productive clones of cord
0.002%) in the tonsil (Fig. 3 a). Thus, whereas hpre-CDCs blood hpre-CDCs produced cDCs; 32% yielded only
originate in the BM and travel through the blood to seed lym- CD141+ cDCs, 50% only to CD1c+ cDCs, and 6% produced
phoid tissues, the monocyte precursor is retained in the BM. both CD1c+ and CD141+ cDCs (Fig. 5 c and Fig. S2). Of
105 cells assayed, 90% of hpre-CDCs from the PBMC pro-
Proliferative capacity and clonal potential of hpre-CDC duced only cDCs; 11% gave rise to CD141+ cDCs only, 65%
To examine the proliferative potential of hpre-CDC, we pu- only to CD1c+ cDCs, and 14% produced both CD1c+ and
rified them from cord or peripheral blood and compared CD141+ cDCs (Fig. 5 c and Fig. S3). This demonstrates that
their developmental potential to CD34+ hematopoietic stem hpre-CDCs purified from peripheral or cord blood have
and progenitor cells (HSPCs) purified from cord blood in clonal efficiencies and differentiation potential similar to CD1c+
MS5+FSG culture (Lee et al., 2015). Whereas CD34+ pro- and CD141+ cDCs (Fig. 5 c). Only a very small fraction of
genitors expanded 156-fold during the 7-d culture period, hpre-CDCs produced monocytes alone or monocytes and
hpre-CDCs from cord blood or peripheral blood expanded DCs (Fig. 5 c; 11.5 and 10.5% from cord blood and periph-
only 8- and 6-fold, respectively (Fig. 4 a). The relatively low eral blood, respectively). Thus, the hpre-CDC population in
level of hpre-CDC expansion could result from low prolifer- peripheral and cord blood contains single cells with potential
ative capacity or poor clonal efficiency. To determine their to differentiate into both major subsets of human cDCs.
proliferative capacity, we loaded purified hpre-CDCs or
CD34+ HSPCs with CFSE and documented division by dye Progenitors of hpre-CDCs
dilution at days 2 and 7 of culture. On day 2, HSPCs re- We have identified a common DC progenitor in BM and
mained CD34+ and produced neither CD1c+ nor CD141+ cord blood that produces pDCs and cDCs (hCDP; Lee et al.,
cDCs, whereas hpre-CDCs differentiated into CD1c+ and 2015). To determine whether there exists a progenitorprogeny
CD141+ cDCs (Fig. 4, b and c). On day 7, both CD34+ HSPC relationship between the hCDP and hpre-CDC (Fig. 6 a),
and hpre-CDC cultures contained CD1c+ and CD141+ cDCs we purified hCDPs from cord blood (Fig. 6 b) and tested
(Fig. 4 b). CD34+ HSPCs divided up to 9 times in 7 d, whereas whether they differentiate into hpre-CDCs in MS5+FSG
hpre-CDCs underwent a maximum of 4 divisions (Fig. 4 c). culture. hCDPs down-regulated CD34 as early as 1 d after
Comparison of CFSE dilution revealed that hpre-CDC- culture and diverged into two populations: one group of cells
derived CD1c+ cDCs and CD141+ cDCs underwent at least up-regulated CD303 and assumed a pDC phenotype, the other
one more division than hpre-CDCs in the culture (Fig. 4 c), in- group was CD303 but expressed CD45RA, CD117, and
dicating that cDCs are also capable of cell division. Consistent CD116 resembling hpre-CDCs. The number of hpre-CDCs

based on CD116, CD115, and CD45RA: CD135+CD116 (CD116), CD135+CD116+CD45RA(CD45RA), CD135+CD116+CD45RA+CD115+ (CD115+), and
CD135+CD116+CD45RA+ CD115 (CD115). (d and e) Differentiation potential of 100 purified cells from each of 4 populations indicated in (c) in
MS5+FSG cultures for 7 d. (d) Flow cytometry plots of CD45+ cells gated as in (c), showing expression of CD141, CLEC9a, CD1c, and CD19. (e) Graphs
show mean output of pDC, CD1c+ cDC, CD141+ cDC and monocytes from each population from three independent experiments. (f) Histograms show ex-
pression of CD11c, HLA-DR, CD123, CD135, CD117, CD45RA, CD116, and CD115 on indicated cell-type. (g) Morphology of purified cord blood hpre-CDCs
by Giemsa staining of cytospin preparations (100). Dotted lines indicate cropping out of white space between cells. Bar, 10 m.

JEM Vol. 212, No. 3 405


peaked at day 2, and this was followed by increase of CD1c+
cDCs and CD141+ cDCs (Fig. 6 c). We conclude that hCDPs
lose CD34 expression and give rise to hpre-CDCs and pDCs
in culture.

Flt3L injection induces expansion


of hpre-CDCs in human blood
Flt3L stimulates DC hematopoiesis (Karsunky et al., 2003)
and is an acute phase reactant that can be elevated in infection
in both mice and humans, accounting for DC expansion in
patients with malaria infection and in volunteers injected
with Flt3L (Maraskovsky et al., 2000; Pulendran et al., 2000;
Guermonprez et al., 2013). In humans, Flt3L injection is associ-
ated with a rapid increase in circulating DCs and in CD11c
CD123+ cells that produce large amounts of IFN- when
cultured with influenza virus (Pulendran et al., 2000). The
latter were initially thought to be DC progenitors but were
subsequently shown to be pDCs (Siegal et al., 1999; Liu, 2005).
To determine whether Flt3L mobilizes DC progenitors
into the blood, we made use of samples from three volunteers
subcutaneously injected with 25 g/kg of Flt3L for 10 con-
secutive days. 14-color flow cytometry was used to assess the
number of cDCs, pDCs, and hpre-CDCs (Fig. S4). The
number of circulating cDCs increased on day 5 and peaked
on day 14 after the initial injection (Fig. 7, a and b). The de-
gree of cDC expansion varied with a 32234-fold increase in
CD1c+ cDCs, a 40171-fold increase in CD141+ cDCs, and
1622-fold increase in pDCs in the blood (Fig. 7 b). Similar
to CD141+ cDCs obtained from MS5+FSG culture (Lee
et al., 2015), Flt3L-induced CD141+ cDCs express variable
levels of CD1c, and all DC subtypes in Flt3L-injected indi-
viduals down-regulated expression of the Flt3L receptor,
CD135 (Fig. 7 c; Waskow et al., 2008). Thus, as others have
found, both subtypes of cDCs and pDCs increase in the blood
in response to Flt3L injection (Pulendran et al., 2000).
Like the more differentiated DCs, hpre-CDC also in-
creased in the blood of the same patients, starting on day 5 but
peaking earlier, on days 1112 after injection (Fig. 7, a and b).
Flt3L-expanded hpre-CDCs were similar to their uninduced
counterparts and cord blood derived hpre-CDCs in producing
CD1c+ and CD141+ cDCs in MS5+FSG culture (Fig. 7 d).
In contrast, the immediate precursors of hpre-CDCs, hCDPs,
and more distant progenitors such as hGMDPs or hMDPs
were undetectable in the peripheral blood of individuals in-
jected with Flt3L at any time point examined (Fig. S4). We
conclude that Flt3L expands human DCs and their progenitors
Figure 3. hpre-CDCs in human lymphoid organs and blood. and recruits hpre-CDCs, but not earlier DC progenitors, into
(a) Representative flow cytometry plots of gated Lin(CD3/19/56/14/66b) the circulation.
DC(CD1c/141/303)CD34 cells show gating strategy and composition

of hpre-CDCs (SSCloCD117+CD116+CD135+ CD45RA+CD115, red) and



premonocytes (SSCloCD117+CD116+CD135+ CD45RA+CD115+, green) in
human cord blood (CB), BM, peripheral blood (PB), and tonsil. Numbers (BM), three (PB), and two (tonsil) independent experiments are shown.
indicate frequency of cells of parent gate (CB, n = 5; BM, n = 4, PB, n = 5; (c) Differentiation potential of purified premonocytes from cord blood
Tonsil, n = 3; n, number of donors). (b) Differentiation potential of puri- (CB) and BM in MS5+FSG cultures for 5 d. FACs plots show phenotype
fied hpre-CDCs indicated in (a) in MS5+FSG cultures for 5 d. Flow cytom- of gated live human CD45+ culture-derived cells including monocytes
etry plots of gated live human CD45+ cells show culture output of CD141+ (orange) and CD1c+ cDCs (blue). Representative of three independent
cDC (red) and CD1c+ cDC (blue). Representative results of four (CB), three experiments are shown.

406 Circulating precursors of human dendritic cells | Breton et al.


Ar ticle

Figure 4. Proliferative capacity of hpre-CDCs. (a) Expansion of purified hpre-CDCs or CD34+ HSPCs from peripheral blood (PB) or cord blood (CB) in
MS5+FSG cultures for 7 d. Graph shows the mean fold change of live human CD45+ cells from 100 input cells from three independent experiments. *, P <
0.005, unpaired two-tailed Students t test. (b and c) CD34+ HSPCs and hpre-CDCs were purified from CB, labeled with CFSE, and cultured in MS5+FSG for
2 or 7 d; proliferation was assessed by flow cytometry. FACs plots show (b) gated CD45+ culture-derived cells, including CD34+ cells (purple), CD34CD1c
CD141 cells (orange), CD141+ cDCs (red), and CD1c+ cDCs (blue) and (c) their CFSE dilution. Dotted lines mark last division by hpre-CDCs. Plots are repre-
sentative of three independent experiments.

Other myeloid populations, such as monocytes and gran- Human DC development has been difficult to delineate,
ulocytes also expand in response to Flt3L, starting on day 5 in part because of limited access to human tissues and limited
and peaking on day 14 after the initial injection (Fig. 7, tissue culture methods. Earlier human trials showed increased
e and f). Interestingly, comparison of the fold change at day numbers of DCs in the blood after Flt3L injection, and sug-
14 indicates that Flt3L preferentially increases the number gested that a CD11cCD123+ IFN- producing cell might
of human cDCs and their progenitors rather than the other be a DC progenitor (Pulendran et al., 2000). However, pro-
myeloid subsets, i.e., pDCs, monocytes, and granulocytes in genitor potential was never tested directly and it is likely that
blood (Fig. 7 g). those cells represent pDCs, which are CD123+ and induced
by Flt3L injection (Fig. 7 a). More recently, circulating cDCs
DISCUSSION were shown to be less mature than their tissue-derived coun-
DCs turn over in tissues and must be replaced continuously to terparts and it has been proposed that these less mature cells
maintain homoeostasis (Liu et al., 2007; Liu and Nussenzweig, are the precursors of tissue cDCs (Collin et al., 2011; Segura
2010). In the mouse, lymphoid and nonlymphoid tissue et al., 2012; Ginhoux and Schlitzer, 2014). Our experiments
resident DCs are continually replenished by pre-CDCs, identify a human pre-CDC that resides in the BM, peripheral
which are produced in the BM, enter the circulation and then blood, and peripheral lymphoid organs that is the direct pro-
emigrate into tissues to differentiate into both major subsets genitor of both major subsets of human cDCs. Whether the
of cDCs (Naik et al., 2006; Bogunovic et al., 2009; Ginhoux hpre-CDC differentiates into cDCs in the blood or tissues or
et al., 2009; Liu et al., 2009; Varol et al., 2009). Defining both has yet to be determined.
the mouse pre-CDC was essential to establishing that DCs The cell surface markers used to define mouse DCs and
represent a unique cell lineage because pre-CDCs do not their progenitors are not entirely conserved by their human
produce monocytes, or pDCs; their differentiation potential counterparts. For example, lack of cell surface MHCII expres-
is restricted to cDCs. sion, one of the key features that distinguishes mouse pre-CDCs

JEM Vol. 212, No. 3 407


Figure 5. Clonal efficiency and potential
of hpre-CDC. (a) Limiting dilution outgrowth
assay show clonal efficiency of CD34+ HSPCs
(n = 3) or hpre-CDCs from peripheral (PB, n = 4)
and cord blood (CB, n = 3) in MS5+FSG cul-
tures for 5 d (for hpre-CDCs) or 14 d (for
CD34+ HSPCs; Materials and methods). **, P <
0.05, pairwise test by ELDA. (b) Representative
flow cytometry plots of gated CD45+ cells
derived from single hpre-CDC clones indi-
cated in (a) show output of CD1c+ cDCs (blue)
and CD141+ cDCs (red). (c) Graphs summarize
the lineage output of single clones from CD34+
HSPCS, and hpre-CDCs from cord (CB) or pe-
ripheral blood (PB). G, granulocyte; M, mono-
cyte; L, lymphocytes.

from fully differentiated DCs, cannot be used to define human into specialized subsets in different tissues. We speculate that
DC progenitors because MHCII is expressed on the surface similar tissue specific heterogeneity in cDC subtypes will also
of most early human hematopoietic cells. Similarly, CD11c used be found in human. In contrast, the closely related human
as marker to identify pre-CDCs in the mouse is expressed monocyte progenitor is restricted to the BM (Fig. 3). Similar
at low levels on hpre-CDCs (Fig. 2 f) and should not be used to monocytes, pDCs enter the circulation as fully differenti-
to identify this cell in humans. However, both human and ated effector cells.
mouse pre-CDCs express CD135. This receptor marks the Like all cDCs and their early progenitors, hpre-CDCs
entire DC pathway in both species from the earliest BM pre- express CD135, the receptor for Flt3L. Our experiments
cursor to the fully differentiated cDCs in tissues (Chicha et al., show that like more mature cDCs (Maraskovsky et al., 1997;
2004; Fogg et al., 2006; Naik et al., 2007; Onai et al., 2007; Pulendran et al., 2000), hpre-CDCs are mobilized into the
Waskow et al., 2008; Doulatov et al., 2010; Lee et al., 2015). blood by Flt3L injection. However, the immediate precursor
In the mouse, DC progenitors can be distinguished from of hpre-CDCs, hCDPs, are not. Flt3L levels in serum increase
other cells based on increasingly restricted expression of cyto- in response to DC depletion in mice and are also increased in
kine receptors (Karsunky et al., 2003; Onai et al., 2007; Waskow humans that lack cDCs and other myeloid cells due to GATA2
et al., 2008; Liu et al., 2009). We applied this principle to mutation (Dickinson et al., 2014). In addition, levels of this
identify the hpre-CDC and made use of differential expres- hematopoietin are also increased in the serum of humans in-
sion of CD116 (GM-CSF receptor), CD135 (Flt3L recep- fected with Plasmodium falciparum, and in mice infected with
tor), CD115 (M-CSF receptor), CD117 (SCF receptor), and Plasmodium chaubodii or Plasmodium yoelli or cytomegalovirus
CD45RA (marker for DC progenitors) on monocytes, cDCs, (Eidenschenk et al., 2010; Guermonprez et al., 2013). More-
and pDCs to uncover the hpre-CDC, which is LinCD34 over, in humans infected with P. falciparum, increased serum
CD117+CD135+CD115CD116+CD45RA+. In contrast, Flt3L is associated with an increase in the number of circulat-
otherwise identical cells that are CD115+ appear to be the ing cDCs. Thus, cDC levels in humans are regulated in part by
immediate precursors of monocytes, resembling the mouse hpre-CDC recruitment from the BM in response to Flt3L.
common monocyte progenitor (cMOP), in its differentiation In both humans and mice, pre-CDCs constitute a very
potential as well as in its absence in the circulation (Hettinger small population of cells in the peripheral blood (0.001 and
et al., 2013). 0.02%, respectively). Although present in only small num-
hpre-CDCs enter the circulation and emigrate into tissues bers, the flux of pre-CDCs through the blood stream is remark-
as incompletely differentiated progenitors. In the mouse, this ably high due to their short residence time in circulation. In the
feature of the cDC lineage enables pre-CDC differentiation mouse, 65% of pre-CDCs are cleared from the circulation

408 Circulating precursors of human dendritic cells | Breton et al.


Ar ticle

Figure 6. hpre-CDCs descend from hCDPs. (a) Flow cytometry plots show comparison of hCDP (purple) and hpre-CDC (orange) in cord blood. (b) Flow
cytometry plots of gated CD45+Lin(CD3/19/56/14)CD34+ show phenotype and purity of magnetically enriched cord blood (CB) CD34+ cells (presorting,
upper) and sorted cells (post-sorting, bottom). hCDPs were gated as CD34+CD38hiCD45RA+CD10CD123hi. (c) Differentiation kinetics of hCDPs purified
from CB in MS5+FSG cultures for 1, 2, or 4 d. FACs plots show culture output of live human CD45+ cells, including CD34+ cells (purple), pDC (green),
CD1c+ cDC (blue), CD141+ cDCs (red), and hpre-CDCs (orange). Representative of four independent experiments are shown. Graphs summarize composi-
tion of indicated populations among total hCD45+ cells. Bars are mean values from four independent experiments, and error bars are SEM.

within 1 min after entering the blood, indicating a t1/2 < 1 min MATERIALS AND METHODS
(Liu et al., 2007, 2009). Thus, a cell with a half-life in circu Cell samples. Human umbilical cord blood and leukophoretic peripheral
blood (buffy coat) were purchased from New York Blood Center. Human
lation of 24 h would have to be present in circulation at a
BM was obtained from total hip arthroplasty at Hospital for Special Surgery
1,440-fold higher concentration than the pre-CDC to achieve (New York, NY). Tonsils were obtained from routine tonsillectomies per-
the same overall flux. Similar behavior has been demonstrated formed at the Babies and Childrens Hospital of Columbia-Presbyterian
for HSCs in the circulation (Wright et al., 2001). Like their Medical Center (New York, NY). Informed consent was obtained from the
mouse counterparts, hpre-CDCs have limited expansion po- patients and/or samples were exempt from informed consent being residual
tential (Fig. 4 a; Liu et al., 2009), but their immediate prog- material after diagnosis and fully de-identified. All samples were collected
according to protocols approved by the Institutional Review Board at Co-
eny, CD1c+ and CD141+ cDCs, can proliferate to further
lumbia University Medical Center and The Rockefeller University (New
expand the DC pool in the periphery (Fig. 4 c; Kabashima York, NY). The specimens were kept on ice immediately after surgical re-
et al., 2005; Liu et al., 2009; KC et al., 2014). We speculate that moval. Tonsil samples were minced, treated with 400 U/ml collagenase
this highly dynamic pool of specialized antigen-presenting (Roche) at 37C for 20 min, and put into cell isolation. BM samples were
cells allows rapid adaptation to acute antigenic challenges. preserved in solution containing 1000 U/ml heparin (National Drug Code

JEM Vol. 212, No. 3 409


Figure 7. Flt3L administration increases
circulating DC subsets and hpre-CDCs in
humans. PBMCs from Flt3L-treated volun-
teers (n = 3; 25 g/kg for 10 consecutive
days) were analyzed by flow cytometry over a
21-d period to assess the expansion of DC
subsets (CD1c+ cDCs [blue], CD141+ cDCs
[red], and pDCs [green]; hpre-CDCs (gray);
monocytes (orange); and granulocytes
(brown). (a) Representative flow cytometry
dot plots show DC subsets and hpre-CDCs in
blood (gating strategy in Fig. S4). (b) Graphs
show the kinetics of cell number of cDC sub-
sets, pDCs, and hpre-CDCs over 21 d in
3 Flt3L-treated volunteers. The absolute num-
bers per milliliter of blood were obtained by
multiplying the number of cells (obtained by
flow cytometry) by the total number of
PBMCs per milliliter of blood. (c) Representa-
tive histograms show CD135 expression on
CD141+ cDCs, CD1c+ cDCs, pDCs and hpre-
CDCs over 21 d in one Flt3L-treated volunteer.
(d) Differentiation potential of purified hpre-
CDCs from blood of Flt3L-injected volunteers
in MS5+FSG cultures after 7 d. Flow cytom-
etry plots of gated CD45+ cells from culture
show output of CD141+ cDCs (red), CD1c+
cDCs (blue), and lack of pDCs (green gate).
(e) Representative flow cytometry dot plots
show CD14+ monocytes and CD66b+ granulo-
cytes in blood (gating strategy in Fig. S4).
(f) Graphs show the kinetics of cell number of
monocytes and granulocytes over 21 d in
3 Flt3L-treated volunteers. The absolute num-
bers per milliliter of blood were obtained by
multiplying the number of cells (obtained by
flow cytometry) by the total number of
PBMCs per milliliter of blood. (g) Graph show-
ing the mean fold change increase from the
three patients (d1 vs. d14) of hpre-CDCs
(gray), CD1c+ cDCs (blue), CD141+ cDCs (red),
pDCs (green), monocytes (orange), and granu-
locytes (brown) per milliliter of blood. Error
bars are SDs.

#63323-540-11) until digestion. The samples were then digested in RPMI from cord blood, peripheral blood, BM, and tonsil were incubated with
containing 20 mg/ml collagenase IV (#C-5138; Sigma-Aldrich) for 15 min fluorescent-labeled antibodies for direct analysis on the BDLSR II flow
at 37C. After density centrifugation using Ficoll-Hypaque (GE Healthcare), cytometers (BD) or further purification by fluorescence-activated cell sort-
aliquots of mononuclear BM cells were frozen and stored in liquid nitrogen ing on the BD FACSAria or Influx, both using HeNe and argon lasers.
for future analysis. For isolation of rare hpre-CDCs from cord blood and peripheral blood,
an enrichment step was performed before sorting. In brief, mononuclear cells
Cell isolation and flow cytometry. Fresh mononuclear cells were isolated were incubated with antibodies against CD135 (4G8; PE; BD) and CD117
by density centrifugation using Ficoll-Hypaque (GE Healthcare). Samples (A3C6E2; Biotin; BioLegend) for 40 min at 4C. After washing, antibody

410 Circulating precursors of human dendritic cells | Breton et al.


Ar ticle

against PE (PE001; Biotin; BioLegend) was added and incubated for another cells per well for CD34+ HSPCs, and at 1, 5 or 10 cells per well for hpre-
10 min at 4C. After washing, CD117+ and CD135+ cells were positively se- CDCs. The percentage of detection failure for cDCs was calculated as 100%
lected using anti-biotin MicroBeads and LS MACS magnetic columns (Mil (1-k/n), where k is the number of positive wells and n the total number of wells
tenyi Biotec). For sorting hpre-CDCs, enriched cells (from CB or PB) or total combined from three or four independent experiments (CB and PB, respec-
mononuclear cells (from CB, PB, BM, or tonsils) were stained for CD14 tively). We classified each well as positive if more than 5 cells were detected for
(TuK4; Qdot-655; Invitrogen), CD3 (OKT3; Brilliant Violet [BV] 650; cDCs or other lineages among live human CD45+ cells.Then, for every posi-
BioLegend), CD19 (HIB19; BV650; BioLegend), CD56 (HCD56; BV650; tive clone, we scored positive for cDCs or other lineages based on their re-
BioLegend), CD66b (G10F5; PerCP-Cy5.5; BioLegend), CD303 (201A; spective gates to determine the clonal lineage potential. The frequency of
PerCP-Cy5.5; BioLegend), CD1c (L161; APC-Cy7; BioLegend), CD141 DC-producing precursors was determined by Loi de Poisson using the Ex-
(M80; PE-Cy7; BioLegend), CD34 (581; Alexa Fluor 700; BioLegend), treme Limiting Dilution Analysis software provided by the Walter and Eliza Hall
CD117 (104D2; BV421; BioLegend), CD135 (4G8; PE; BD), CD45RA Institute of Medical Research Bioinformatics (Parkville,Victoria, Australia.
(HI100; BV510; BioLegend), CD116 (4H1; FITC; BioLegend), and CD115
(9-4D2-1E4; APC; BioLegend) for 40 min on ice. hpre-CDCs were isolated Flt3L injection in volunteers. 3 healthy volunteers received a 25 g/kg/
as Lin(CD3/19/56/14)Granulocyte(CD66b)pDC(CD303)cDC(CD1c/ day subcutaneous injection of CDX-301 (a clinical formulation of recombi-
CD141)CD34CD117+CD135+SSClo CD116+CD115CD45RA+. nant human Flt3L; Celldex) for 10 d. Blood samples were collected before
In addition to these antibodies, for surface marker phenotype charac- and at 5, 8, 10, 11, 12, 14, and 21 d after initial administration. PBMCs were
terization of fully differentiated cells, CD45 (HI30; Alexa Fluor 700; Bio- isolated from heparinized blood using Ficoll-Hypaque and stored in liquid
Legend), CD34 (581; APC-Cy7; BioLegend), CD3 (UCHT1; Biotin; BD), nitrogen for further analysis.
CD19 (HIB19; Biotin; BD), CD56 (B159; Biotin; BD), CD115 (12-3A3- PBMCs were stained with CD116 (4H1; FITC; BioLegend), CD135
1B10; PE; eBioscience), CD123 (9F5; PE; BD) and CD45RA (120458-42; (4G8; PE; BD), CD66b (G10F5; PerCP-Cy5.5; BioLegend), CD335 (9E2,
eBioscience; PE) were used alternatively, followed by a secondary stain with PerCP-Cy5.5, BioLegend), CD303 (201A; PerCP Cy5.5; BioLegend),
streptavidin-conjugated PerCP-Cy5.5 for 40 min on ice. CD141 (M80; PE-Cy7; BioLegend), CD117 (104D2; BV421; BioLegend),
Cultured cells were harvested and stained with LIVE/DEAD (Life CD123 (6H6; BV510; BioLegend), CD20 (2H7; BV605; BioLegend), CD3
Technologies), CD45 (AF700), CD56 (B159; Pacific Blue; BD), CD66b (OKT3; BV605; BioLegend), CD45RA (MEM-56; Qdot 655; Life Tech-
(G10F5; PerCP-Cy5.5; BioLegend), CD19 (HIB19; APC-Cy7; BioLeg- nologies), LIVE/DEAD Fixable Blue Dead Cell Stain (Life Technologies),
end), CLEC9a (8F9; PE; BioLegend), CD14 (Qdot-655), CD1c (L161; PE- CD14 (Tk4; Qdot 800; Life Technologies), CD115 APC (9-4D2-1E4;
Cy7; BioLegend), CD303 (201A; FITC; BioLegend), CD123 (6H6; BV510; APC; BioLegend), CD34 (581; Alexa Fluor 700; BioLegend), CD1c (L161;
BioLegend), CD141 (AD5-14H12; APC; Miltenyi Biotec) for 40 min on APC-Cy7; BioLegend). For surface staining, titrated antibodies were added
ice and analyzed for lineage potential by flow cytometry. Absolute cell counts to 2 million cells in 50 l PBS for 20 min at 4C.
have been calculated relative to a well containing a known number of CD45+ Washed cells were fixed in 2% formaldehyde and stored at 4C until
cells (i.e., 10,000 cells), which were added on the day of harvest. analysis, which was performed using an LSR II flow cytometer (BD). The
For CFSE assays, cultured cells were stained with LIVE/DEAD (Life whole sample was acquired and analysis was performed using Flow Jo 9.1
Technologies), CD45 (AF700), CD34 (APC-Cy7), CLEC9a (PE), CD14 software (Tree Star). hpre-CDCs were isolated as Lin(CD3/20/335/14)
(Qdot-655), CD1c (PE-Cy7), and CD141 (APC) for 40 min on ice. Granulocyte(CD66b)DC(CD303CD1c/CD141)CD34CD117+CD135+
For progenitorprogeny relationship experiments, CD34+ cells were first CD116+ CD115CD45RA+CD123+.
enriched from cord blood using CD34 MicroBead kit and LS MACS magnetic
columns (Miltenyi Biotec). Enriched CD34+ cells (4095% purity) were then
Online supplemental material. Fig. S1 shows clonal output of cord blood
incubated with Lin (CD3/19/56/14; BV650), CD34 (AF700), CD38 (HIT2;
CD34+ HSPCs. Fig. S2 shows clonal output of cord blood hpre-CDCs. Fig. S3
BV421; BioLegend), CD10 (HI10a; PE-Cy7; BioLegend), CD45RA (BV510),
shows clonal output of peripheral blood hpre-CDCs. Fig. S4 shows 14-color
CD123 (PE), CD116 (FITC), and CD115 (APC). hCDPs were sorted as Lin
gating strategy for identification of human hpre-CDCs in blood and that
CD34+CD38hiCD10CD45RA+CD123hiCD115. Cultured cells were har-
hCDPs, hMDPs, and hGMDPs are undetectable in the blood before and after
vested at specific time points and stained with LIVE/DEAD, CD45 (AF700),
Flt3L administration in healthy volunteers. Online supplemental material is
CD14 (Qdot-655), CD3 (BV650), CD19 (BV650), CD56 (BV650), CD303
available at http://www.jem.org/cgi/content/full/jem.20141441/DC1.
(FITC), CD1c (PE-Cy7), CD141 (APC), CD34 (APC-Cy7), CD117 (BV421),
CD123 (PE), CD45RA (BV510), and CD116 (FITC) for 40 min on ice.
This work was inspired by Dr. Ralph M. Steinman. We thank Klara Velinzon (Flow
Cytometry Core Facility, Laboratory of Molecular Immunology, The Rockefeller
Morphological analysis. Purified hpreDCs were analyzed by Giemsa stain- University) for technical support with polychromatic flow cytometry sorting. We thank
ing of cytospin preparations. As few as 5 104 cells were cytospun for 5 min Heidi Schreiber for help with human BM protocol (Laboratory of Molecular Immunology,
at 800 rpm on a glass slide and then stained with the Hemacolor stain kit as The Rockefeller University). We acknowledge the assistance of Arlene Hurley and the
recommended by the manufacturer (HARLECO). Slides were then imaged clinical team at the Rockefeller University Hospital. We also thank James Pring, Popi
on an Axioplan 2 microscope at 100 magnification (Carl Zeiss). Sarma and Christine Trumpfheller for the management of the Flt3L patient samples
(Laboratory of Cellular Physiology and Immunology, The Rockefeller University).
Cell culture. MS5 BM stromal cells were maintained and passed in com- Research reported in this publication was supported by the Empire State Stem
plete -MEM medium (Invitrogen) with 10% FCS and penicillin/strepto- Cell Fund through New York State Department of Health Contract #C029562
mycin (Invitrogen). 24 h before hpre-CDC culture, MS5 stromal cells were (to K.Liu), Helmsley Foundation (to K. Liu), National Institute of Allergy and Infectious
Diseases of National Institutes of Health under award numbers of AI101251 (to K. Liu)
treated with 10 g/ml of mitomycin C (Sigma-Aldrich) for 3 h, washed, and
and U19AI111825 (to M.C. Nussenzweig), National Institute of Neurological Disorder
reseeded at 3.75 104 MS5 cells per well in 96-well plates. Purified popula-
and Stroke of NIH under award number of NS084776 (to S. Puhr), Iris and Junming Le
tions were seeded in medium containing 100 ng of Flt3L/ml (Celldex),
Foundation (to G.Breton), the Dermatology Foundation (to N. Anandasabapathy),
20 ng/ml SCF (PeproTech), and 10 ng/ml GM-CSF (PeproTech). Cells
National Institute of Arthritis and Musculoskeletal and Skin Diseases AR063461-
were harvested between day 1 and 14 for flow cytometry analysis. 01A1 (to N. Anandasabapathy), and CTSA, RUCCTS grant no. UL1RR024143 from
To determine cellular divisions in culture, input populations were la- the National Center for Research Resources, NIH, and by NIH grant AI13013. The
beled for 15 min with 5 M CFSE (Molecular Probes) at 37C. Rockefeller University Center for Clinical and Translational Science is supported, in
part, by a Clinical and Translational Science Award (CTSA), and the National Center for
Limiting dilution assay. For limiting dilution experiments, cells were Advancing Translational Sciences (NCATS), part of the NIH. The content is solely the
sorted directly into 96-well plates containing MS5+FSG at 1, 2, 4, 8, or 16 responsibility of the authors and does not necessarily represent the official views of

JEM Vol. 212, No. 3 411


the National Institutes of Health or the Empire State Stem Cell Board, the NY State BDCA1-positive dendritic cells in human blood. J. Immunol. 176:991
Department of Health or the State of New York or any other applicable federal 998. http://dx.doi.org/10.4049/jimmunol.176.2.991
funding agency. M.C. Nussenzweig is an HHMI Investigator. Grouard, G., M.C. Rissoan, L. Filgueira, I. Durand, J. Banchereau, and Y.J.
T. Keler works for Celldex Therapeutics. The authors declare no additional Liu. 1997. The enigmatic plasmacytoid T cells develop into dendritic
financial interests. cells with interleukin (IL)-3 and CD40-ligand. J. Exp. Med. 185:1101
1111. http://dx.doi.org/10.1084/jem.185.6.1101
Submitted: 30 July 2014 Guermonprez, P., J. Helft, C. Claser, S. Deroubaix, H. Karanje, A.
Accepted: 30 January 2015 Gazumyan, G. Darasse-Jze, S.B. Telerman, G. Breton, H.A. Schreiber,
et al. 2013. Inflammatory Flt3l is essential to mobilize dendritic cells and
REFERENCES for T cell responses during Plasmodium infection. Nat. Med. 19:730
Bachem, A., S. Gttler, E. Hartung, F. Ebstein, M. Schaefer, A. Tannert, 738. http://dx.doi.org/10.1038/nm.3197
A. Salama, K. Movassaghi, C. Opitz, H.W. Mages, et al. 2010. Superior Haniffa, M., A. Shin, V. Bigley, N. McGovern, P. Teo, P. See, P.S. Wasan,
antigen cross-presentation and XCR1 expression define human CD11c+ X.N. Wang, F. Malinarich, B. Malleret, et al. 2012. Human tissues con-
CD141+ cells as homologues of mouse CD8+ dendritic cells. J. Exp. tain CD141hi cross-presenting dendritic cells with functional homology
Med. 207:12731281. http://dx.doi.org/10.1084/jem.20100348 to mouse CD103+ nonlymphoid dendritic cells. Immunity. 37:6073.
Banchereau, J., and R.M. Steinman. 1998. Dendritic cells and the control of http://dx.doi.org/10.1016/j.immuni.2012.04.012
immunity. Nature. 392:245252. http://dx.doi.org/10.1038/32588 Haniffa, M., M. Collin, and F. Ginhoux. 2013. Ontogeny and functional
Bogunovic, M., F. Ginhoux, J. Helft, L. Shang, D. Hashimoto, M. Greter, specialization of dendritic cells in human and mouse. Adv. Immunol.
K. Liu, C. Jakubzick, M.A. Ingersoll, M. Leboeuf, et al. 2009. Origin 120:149. http://dx.doi.org/10.1016/B978-0-12-417028-5.00001-6
of the lamina propria dendritic cell network. Immunity. 31:513525. Hettinger, J., D.M. Richards, J. Hansson, M.M. Barra, A.C. Joschko, J.
http://dx.doi.org/10.1016/j.immuni.2009.08.010 Krijgsveld, and M. Feuerer. 2013. Origin of monocytes and macro-
Chicha, L., D. Jarrossay, and M.G. Manz. 2004. Clonal type I interferon- phages in a committed progenitor. Nat. Immunol. 14:821830. http://
producing and dendritic cell precursors are contained in both human dx.doi.org/10.1038/ni.2638
lymphoid and myeloid progenitor populations. J. Exp. Med. 200:1519 Jongbloed, S.L., A.J. Kassianos, K.J. McDonald, G.J. Clark, X. Ju, C.E.
1524. http://dx.doi.org/10.1084/jem.20040809 Angel, C.J. Chen, P.R. Dunbar, R.B. Wadley, V. Jeet, et al. 2010.
Cohn, L., B. Chatterjee, F. Esselborn, A. Smed-Srensen, N. Nakamura, Human CD141+ (BDCA-3)+ dendritic cells (DCs) represent a unique
C. Chalouni, B.C. Lee, R. Vandlen, T. Keler, P. Lauer, et al. 2013. myeloid DC subset that cross-presents necrotic cell antigens. J. Exp.
Antigen delivery to early endosomes eliminates the superiority of human Med. 207:12471260. http://dx.doi.org/10.1084/jem.20092140
blood BDCA3+ dendritic cells at cross presentation. J. Exp. Med. 210: Kabashima, K., T.A. Banks, K.M. Ansel, T.T. Lu, C.F. Ware, and J.G.
10491063. http://dx.doi.org/10.1084/jem.20121251 Cyster. 2005. Intrinsic lymphotoxin-beta receptor requirement for
Collin, M., V. Bigley, M. Haniffa, and S. Hambleton. 2011. Human den- homeostasis of lymphoid tissue dendritic cells. Immunity. 22:439450.
dritic cell deficiency: the missing ID? Nat. Rev. Immunol. 11:575583. http://dx.doi.org/10.1016/j.immuni.2005.02.007
http://dx.doi.org/10.1038/nri3046 Kamphorst, A.O., P. Guermonprez, D. Dudziak, and M.C. Nussenzweig.
Crozat, K., R. Guiton, V. Contreras, V. Feuillet, C.A. Dutertre, E. Ventre, T.P. 2010. Route of antigen uptake differentially impacts presentation by
Vu Manh, T. Baranek, A.K. Storset, J. Marvel, et al. 2010. The XC che- dendritic cells and activated monocytes. J. Immunol. 185:34263435.
mokine receptor 1 is a conserved selective marker of mammalian cells http://dx.doi.org/10.4049/jimmunol.1001205
homologous to mouse CD8alpha+ dendritic cells. J. Exp. Med. 207: Karsunky, H., M. Merad, A. Cozzio, I.L. Weissman, and M.G. Manz. 2003.
12831292. http://dx.doi.org/10.1084/jem.20100223 Flt3 ligand regulates dendritic cell development from Flt3+ lymphoid
Dickinson, R.E., P. Milne, L. Jardine, S. Zandi, S.I. Swierczek, N. McGovern, and myeloid-committed progenitors to Flt3+ dendritic cells in vivo.
S. Cookson, Z. Ferozepurwalla, A. Langridge, S. Pagan, et al. 2014. The J. Exp. Med. 198:305313. http://dx.doi.org/10.1084/jem.20030323
evolution of cellular deficiency in GATA2 mutation. Blood. 123:863 KC, W., A.T. Satpathy, A.S. Rapaport, C.G. Briseo, X. Wu, J.C. Albring,
874. http://dx.doi.org/10.1182/blood-2013-07-517151 E.V. Russler-Germain, N.M. Kretzer, V. Durai, S.P. Persaud, et al. 2014.
Doulatov, S., F. Notta, K. Eppert, L.T. Nguyen, P.S. Ohashi, and J.E. L-Myc expression by dendritic cells is required for optimal T-cell prim-
Dick. 2010. Revised map of the human progenitor hierarchy shows ing. Nature. 507:243247. http://dx.doi.org/10.1038/nature12967
the origin of macrophages and dendritic cells in early lymphoid de- Lee, J., G. Breton, T.Y.K. Oliveira, Y.J. Zhou, A. Aljoufi, S. Puhr,
velopment. Nat. Immunol. 11:585593. http://dx.doi.org/10.1038/ M.J. Cameron, R.-P. Skaly, M.C. Nussenzweig, and K. Liu. 2015.
ni.1889 Restricted dendritic cell and monocyte progenitors in human cord blood
Dudziak, D., A.O. Kamphorst, G.F. Heidkamp, V.R. Buchholz, C. Trumpfheller, and bone marrow. J. Exp. Med. 212:385399. http://dx.doi.org/10.1084/
S. Yamazaki, C. Cheong, K. Liu, H.W. Lee, C.G. Park, et al. 2007. Dif jem.20141441
ferential antigen processing by dendritic cell subsets in vivo. Science. 315: Liu, Y.J. 2005. IPC: professional type 1 interferon-producing cells and plas-
107111. http://dx.doi.org/10.1126/science.1136080 macytoid dendritic cell precursors. Annu. Rev. Immunol. 23:275306.
Eidenschenk, C., K. Crozat, P. Krebs, R. Arens, D. Popkin, C.N. Arnold, http://dx.doi.org/10.1146/annurev.immunol.23.021704.115633
A.L. Blasius, C.A. Benedict, E.M. Moresco, Y. Xia, and B. Beutler. Liu, K., and M.C. Nussenzweig. 2010. Development and homeostasis of den-
2010. Flt3 permits survival during infection by rendering dendritic cells dritic cells. Eur. J. Immunol. 40:20992102. http://dx.doi.org/10.1002/
competent to activate NK cells. Proc. Natl. Acad. Sci. USA. 107:9759 eji.201040501
9764. http://dx.doi.org/10.1073/pnas.1005186107 Liu, K., C. Waskow, X. Liu, K. Yao, J. Hoh, and M. Nussenzweig. 2007.
Fogg, D.K., C. Sibon, C. Miled, S. Jung, P. Aucouturier, D.R. Littman, Origin of dendritic cells in peripheral lymphoid organs of mice. Nat.
A. Cumano, and F. Geissmann. 2006. A clonogenic bone marrow pro- Immunol. 8:578583. http://dx.doi.org/10.1038/ni1462
genitor specific for macrophages and dendritic cells. Science. 311:8387. Liu, K., G.D. Victora, T.A. Schwickert, P. Guermonprez, M.M. Meredith,
http://dx.doi.org/10.1126/science.1117729 K. Yao, F.F. Chu, G.J. Randolph, A.Y. Rudensky, and M. Nussenzweig.
Ginhoux, F., and A. Schlitzer. 2014. CD11b+ DCs rediscovered: implica- 2009. In vivo analysis of dendritic cell development and homeostasis.
tions for vaccination. Expert Rev. Vaccines. 13:445447. http://dx.doi.org/ Science. 324:392397. http://dx.doi.org/10.1126/science.1170540
10.1586/14760584.2014.893196 Maraskovsky, E., B. Pulendran, K. Brasel, M. Teepe, E.R. Roux, K. Shortman,
Ginhoux, F., K. Liu, J. Helft, M. Bogunovic, M. Greter, D. Hashimoto, J. S.D.Lyman,and H.J.McKenna.1997.Dramatic numerical increase of func-
Price, N. Yin, J. Bromberg, S.A. Lira, et al. 2009. The origin and devel- tionally mature dendritic cells in FLT3 ligand-treated mice. Adv. Exp. Med.
opment of nonlymphoid tissue CD103+ DCs. J. Exp. Med. 206:3115 Biol. 417:3340. http://dx.doi.org/10.1007/978-1-4757-9966-8_6
3130. http://dx.doi.org/10.1084/jem.20091756 Maraskovsky, E., E. Daro, E. Roux, M. Teepe, C.R. Maliszewski, J. Hoek,
Granelli-Piperno, A., I. Shimeliovich, M. Pack, C. Trumpfheller, and R.M. D. Caron, M.E. Lebsack, and H.J. McKenna. 2000. In vivo generation
Steinman. 2006. HIV-1 selectively infects a subset of nonmaturing of human dendritic cell subsets by Flt3 ligand. Blood. 96:878884.

412 Circulating precursors of human dendritic cells | Breton et al.


Ar ticle

McKenna, H.J., K.L. Stocking, R.E. Miller, K. Brasel,T. De Smedt, E. Maraskovsky, tative equivalents of mouse CD8alpha+ dendritic cells. J. Exp. Med.
C.R. Maliszewski, D.H. Lynch, J. Smith, B. Pulendran, et al. 2000. Mice lack- 207:12611271. http://dx.doi.org/10.1084/jem.20092618
ing flt3 ligand have deficient hematopoiesis affecting hematopoietic pro- Pulendran, B., J. Banchereau, S. Burkeholder, E. Kraus, E. Guinet, C.
genitor cells, dendritic cells, and natural killer cells. Blood. 95:34893497. Chalouni, D. Caron, C. Maliszewski, J. Davoust, J. Fay, and K. Palucka.
Merad, M., P. Sathe, J. Helft, J. Miller, and A. Mortha. 2013. The dendritic 2000. Flt3-ligand and granulocyte colony-stimulating factor mobilize
cell lineage: ontogeny and function of dendritic cells and their subsets in distinct human dendritic cell subsets in vivo. J. Immunol. 165:566572.
the steady state and the inflamed setting. Annu. Rev. Immunol. 31:563 http://dx.doi.org/10.4049/jimmunol.165.1.566
604. http://dx.doi.org/10.1146/annurev-immunol-020711-074950 Robbins, S.H., T. Walzer, D. Dembl, C. Thibault, A. Defays, G. Bessou,
Murphy, K.M. 2013. Transcriptional control of dendritic cell development. H. Xu, E. Vivier, M. Sellars, P. Pierre, et al. 2008. Novel insights into
Adv. Immunol. 120:239267. http://dx.doi.org/10.1016/B978-0-12- the relationships between dendritic cell subsets in human and mouse
417028-5.00009-0 revealed by genome-wide expression profiling. Genome Biol. 9:R17.
Naik, S.H., D. Metcalf, A. van Nieuwenhuijze, I. Wicks, L. Wu, M. http://dx.doi.org/10.1186/gb-2008-9-1-r17
OKeeffe, and K. Shortman. 2006. Intrasplenic steady-state dendritic Schlitzer, A., N. McGovern, P. Teo, T. Zelante, K. Atarashi, D. Low, A.W.
cell precursors that are distinct from monocytes. Nat. Immunol. 7:663 Ho, P. See, A. Shin, P.S. Wasan, et al. 2013. IRF4 transcription fac-
671. http://dx.doi.org/10.1038/ni1340 tor-dependent CD11b+ dendritic cells in human and mouse control
Naik, S.H., P. Sathe, H.Y. Park, D. Metcalf, A.I. Proietto, A. Dakic, S. mucosal IL-17 cytokine responses. Immunity. 38:970983. http://dx.doi
Carotta, M. OKeeffe, M. Bahlo, A. Papenfuss, et al. 2007. Development .org/10.1016/j.immuni.2013.04.011
of plasmacytoid and conventional dendritic cell subtypes from single Segura, E., J. Valladeau-Guilemond, M.H. Donnadieu, X. Sastre-Garau,
precursor cells derived in vitro and in vivo. Nat. Immunol. 8:12171226. V. Soumelis, and S. Amigorena. 2012. Characterization of resident and
http://dx.doi.org/10.1038/ni1522 migratory dendritic cells in human lymph nodes. J. Exp. Med. 209:653
Nizzoli, G., J. Krietsch, A. Weick, S. Steinfelder, F. Facciotti, P. Gruarin, A. 660. http://dx.doi.org/10.1084/jem.20111457
Bianco, B. Steckel, M. Moro, M. Crosti, et al. 2013. Human CD1c+ Siegal, F.P., N. Kadowaki, M. Shodell, P.A. Fitzgerald-Bocarsly, K. Shah, S.
dendritic cells secrete high levels of IL-12 and potently prime cytotoxic Ho, S. Antonenko, and Y.J. Liu. 1999. The nature of the principal type
T-cell responses. Blood. 122:932942. http://dx.doi.org/10.1182/blood- 1 interferon-producing cells in human blood. Science. 284:18351837.
2013-04-495424 http://dx.doi.org/10.1126/science.284.5421.1835
ODoherty, U., M. Peng, S. Gezelter, W.J. Swiggard, M. Betjes, N. Varol, C., A. Vallon-Eberhard, E. Elinav, T. Aychek, Y. Shapira, H. Luche,
Bhardwaj, and R.M. Steinman. 1994. Human blood contains two sub- H.J. Fehling, W.D. Hardt, G. Shakhar, and S. Jung. 2009. Intestinal lamina
sets of dendritic cells, one immunologically mature and the other im- propria dendritic cell subsets have different origin and functions. Immunity.
mature. Immunology. 82:487493. 31:502512. http://dx.doi.org/10.1016/j.immuni.2009.06.025
Onai, N.,A. Obata-Onai, M.A. Schmid,T. Ohteki, D. Jarrossay, and M.G. Manz. Waskow, C., K. Liu, G. Darrasse-Jze, P. Guermonprez, F. Ginhoux, M.
2007. Identification of clonogenic common Flt3+M-CSFR+ plasmacy- Merad, T. Shengelia, K. Yao, and M. Nussenzweig. 2008. The recep-
toid and conventional dendritic cell progenitors in mouse bone marrow. tor tyrosine kinase Flt3 is required for dendritic cell development in
Nat. Immunol. 8:12071216. http://dx.doi.org/10.1038/ni1518 peripheral lymphoid tissues. Nat. Immunol. 9:676683. http://dx.doi
Patterson, S., H. Donaghy, P. Amjadi, B. Gazzard, F. Gotch, and P. .org/10.1038/ni.1615
Kelleher. 2005. Human BDCA-1-positive blood dendritic cells differ- Wright, D.E., A.J. Wagers, A.P. Gulati, F.L. Johnson, and I.L. Weissman.
entiate into phenotypically distinct immature and mature populations 2001. Physiological migration of hematopoietic stem and progenitor cells.
in the absence of exogenous maturational stimuli: differentiation fail- Science. 294:19331936. http://dx.doi.org/10.1126/science.1064081
ure in HIV infection. J. Immunol. 174:82008209. http://dx.doi.org/ Yu, C.I., C. Becker, Y. Wang, F. Marches, J. Helft, M. Leboeuf, E.
10.4049/jimmunol.174.12.8200 Anguiano, S. Pourpe, K. Goller, V. Pascual, et al. 2013. Human CD1c+
Poulin, L.F., M. Salio, E. Griessinger, F. Anjos-Afonso, L. Craciun, dendritic cells drive the differentiation of CD103+ CD8+ mucosal ef-
J.L. Chen, A.M. Keller, O. Joffre, S. Zelenay, E. Nye, et al. 2010. fector T cells via the cytokine TGF-. Immunity. 38:818830. http://
Characterization of human DNGR-1+ BDCA3+ leukocytes as pu- dx.doi.org/10.1016/j.immuni.2013.03.004

JEM Vol. 212, No. 3 413


NEW ADJUVANTS FROM AVANTI 3D-PHAD

Monophosphoryl 3-Deacyl Lipid A (Synthetic)


Avanti Number 699852

PHOSPHORYLATED HEXAACYL DISACCHARIDE


(PHAD)
Monophosphoryl
Lipid A (Synthetic)
Avanti Number 699800

Vaccination is well-accepted as an effective method to


prevent infections by mounting pathogen-specific im-
mune responses prior to the infection. Usually, immuni-
zation with vaccine antigens alone is not able to induce
robust or long-lasting immune responses resulting in
failure of protective immunity against infections. Thus,
adjuvants are required to enhance cellular or humoral im-
mune responses upon immunization. Because vaccine
adjuvants using Lipid A have proven to be safe and effec-
tive in inducing Th-1 type immune responses to heterolo-
gous proteins in animal and human vaccines, Avanti devel-
Stimulatory effect of PHAD and 3D-PHAD on
macrophages. Macrophage cell line J774 cells oped Phosphorylated HexaAcyl Disaccharide (PHAD),
were cultured with Avanti PHAD or 3D-PHAD for the first fully synthetic monophosphoryl Lipid A available
24hrs. IL-12 levels in supernatants were measured for use as an adjuvant in human vaccines.
by sandwich ELISA.
For details of these and other adjuvants visit
www.avantilipids.com

THERE IS ONLY ONE AVANTI


AVANTI FOR THE MS STANDARDS YOU CAN TRUST

You might also like