Sunteți pe pagina 1din 14

© 2019. Published by The Company of Biologists Ltd | Development (2019) 146, dev156000. doi:10.1242/dev.

156000

REVIEW

The origins and non-canonical functions of macrophages


in development and regeneration
Marine Theret1,2, Remi Mounier3 and Fabio Rossi1,2,*

ABSTRACT tools have provided strong evidence for their ability to execute tissue-
The discovery of new non-canonical (i.e. non-innate immune) functions specific tasks beyond the response to pathogens (Davies et al., 2013).
of macrophages has been a recurring theme over the past 20 years. In this Review, we describe some of these non-canonical and organ-
Indeed, it has emerged that macrophages can influence the specific functions of MPs. We begin by providing an overview of the
development, homeostasis, maintenance and regeneration of many populations of MPs that exist and discussing their developmental
tissues and organs, including skeletal muscle, cardiac muscle, the origins. We then highlight how such MPs function in the context of
brain and the liver, in part by acting directly on tissue-resident stem tissue development, homeostasis and repair.
cells. In addition, macrophages play crucial roles in diseases such as
obesity-associated diabetes or cancers. Increased knowledge of their Macrophage origins during development and adulthood
regulatory roles within each tissue will therefore help us to better In summary, at least three developmentally distinct types of MPs
understand the full extent of their functions and could highlight exist in the body: yolk sac (YS)-derived tissue-resident MPs, fetal
new mechanisms modulating disease pathogenesis. In this Review, liver-derived tissue-resident MPs, and infiltrating bone marrow-
we discuss recent studies that have elucidated the developmental derived MPs. All of these MPs emerge at different points during
origins of various macrophage populations and summarize our development and adulthood (Fig. 1) and play key roles in tissue
knowledge of the non-canonical functions of macrophages in development, growth, homeostatic maintenance and remodeling
development, regeneration and tissue repair. (reviewed by Epelman et al., 2014b).

KEY WORDS: Macrophage, Homeostasis, Stem cells The emergence of MPs: from embryonic to adult hematopoiesis
During embryogenesis, MPs are detected in most organs and tissues
Introduction including the brain, heart, liver and skeletal muscle. The progeny of
The immune functions of macrophages (MPs) were first highlighted these first tissue-resident MPs persist for the life of the organism.
by Elie Metchnikoff, who was awarded the Nobel Prize in Physiology The precise ontogeny of embryonic MPs is still debated, but it has
or Medicine in 1908. He developed the now commonly accepted been elegantly demonstrated that at least one subset arises before the
theory that phagocytic cells, such as MPs, monocytes (MOs) and appearance of the first multipotent hematopoietic stem cells (HSCs)
neutrophils, fight against pathogens (reviewed by Cavaillon, 2011). (Ginhoux and Guilliams, 2016). For example, during early
Subsequently, during the 20th century, a lineage relationship between embryogenesis in mice, ‘primitive’ hematopoiesis takes place in
circulating blood MOs and the MPs found in inflamed tissues was the extra-embryonic YS [embryonic day (E)6.5 to E11] and gives
discovered, first in 1930 in the amphibian larvae model and later in rise to MPs (Fig. 1), which are the only ‘white’ hematopoietic cells
1939 for mammals (Cavaillon, 2011). This was soon followed by found in the embryo at this time point. Later, from E8.5 to E12,
myriad studies of macrophage activation and functions in immature HSCs emerge from the aorta-gonad-mesonephros (AGM)
inflammation. and give rise to other early immune lineages (which do not include
In its most reductionist definition, inflammation is described as a lymphoid cells) (Orkin and Zon, 2008). These immature HSCs
process that is triggered in response to viral or bacterial infections. eventually colonize the fetal liver (between E10-P1). Soon after
However, it is now commonly accepted that sterile inflammation birth (E19-P1), fetal liver HSCs migrate to the bone marrow and
(non-pathogen-induced inflammation) also occurs in multiple give rise to mature HSCs that are capable of generating all blood
settings, including during development and in afflictions such as lineages, including MPs, in a process called ‘definitive’
autoimmune disease and cancers, and even in aging (Jackaman et al., hematopoiesis (E19-P1) (Fig. 1) (Palis and Yoder, 2001; Orkin
2017). Moreover, it is clear that any form of tissue damage invariably and Zon, 2008). In addition to the fetal liver, the placenta has been
triggers sterile inflammation, influencing outcomes spanning from revealed as a site of active hematopoiesis (between E10-E13)
functional restoration to fibrosis depending on the efficiency of the (Dzierzak and Robin, 2010). At each of these stages, MP precursors
regenerative process. Importantly, MPs are now thought to play key are generated and seed a specific subset of organs, often establishing
roles in modulating such processes, and recently developed molecular local lineages that will last the organism’s lifetime.
DEVELOPMENT

During adulthood, MOs/MPs originate in the bone marrow via


1
Department of Medical Genetics, The Biomedical Research Centre, University of
a differentiation cascade that comprises multiple cellular
British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada. intermediates. According to a classical model, HSCs give rise to
2
Faculty of Medicine, The University of British Columbia, 317-2194 Health Sciences common myeloid progenitors (CMPs) that generate additional
Mall, Vancouver, BC V6T 1Z3, Canada. 3Institut Neuromyogène, CNRS UMR 5310,
INSERM U1217, Université de Lyon, 69008 Lyon, France. progenitors that are further restricted in their developmental
potential, i.e. granulocyte-monocyte progenitors (GMPs) and
*Author for correspondence (fabio@brc.ubc.ca) megakaryocyte-erythrocyte progenitors (MEPs) (Fig. 2A).
M.T., 0000-0002-8059-8756; R.M., 0000-0003-3464-3322; F.R., 0000-0002- However, this classical model has been challenged for years, and
0368-2620 alternative models in which all other myeloid cells (neutrophils,

1
REVIEW Development (2019) 146, dev156000. doi:10.1242/dev.156000

Yolk Sac AGM Liver Bone marrow Fig. 1. Waves of hematopoiesis and MP
emergence. From E6.5 to E11, primitive
hematopoiesis takes place in the YS and tissue-
resident MPs such as microglia and Langerhans cells
appear in their respective tissues. Later, from E8.5 to
E12, immature HSCs appear from the AGM and give
rise to Kupffer cells and alveolar MPs. From E10 to
E19/P1 (birth), the fetal liver starts generating all
immune cells, and tissue-resident MPs within the
cardiac system, skeletal muscle, dermis and gut
E6.5 E8.5 E10 E11 E12 E19 P1 colonize their respective tissues. Soon after birth,
hematopoiesis shifts to the bone marrow and gives rise
to circulating blood monocytes that replace a
percentage of tissue-resident macrophages over time.

Heart-resident MPs
Microglia Langerhans cells Skeletal muscle-resident MPs
Kupffer cells Dermis-resident MPs
Alveolar MPs Gut-resident MPs

eosinophils, mast cells), in addition to MOs/MPs, also come from required for maintaining lung-, spleen- and peritoneum-resident MP
GMPs has been suggested (Kierdorf et al., 2015; Ginhoux and homeostasis (Hashimoto et al., 2013). Other lineage tracing models,
Guilliams, 2016; Yamamoto et al., 2018) (Fig. 2A). Even today, the such as Kit-Mer-Cre-Mer, Tek-Mer-Cre-Mer, Csfr1-Mer-Cre-Mer or
debate is not settled. For example, based on single cell RNA Runx1-Mer-Cre-Mer, specifically target YS-derived hematopoietic
sequencing, Drissen and colleagues recently proposed that mast cells. In these models, activation of Cre recombinase at E7 targets
cells, basophils and eosinophils arise from a different, earlier the YS hematopoietic system but at E8.5 and beyond targets the
progenitor (Gata1+) than do neutrophils and MOs (Flt3+) (Drissen ‘definitive’ hematopoietic system. This allowed independent
et al., 2016; Sarrazin and Sieweke, 2016) (Fig. 2B). In parallel, the validation of the existence of two waves of erythro-myeloid
existence of MEPs has been questioned by reports demonstrating progenitors contributing to tissue-resident MOs (Ginhoux et al.,
that erythrocytes and megakaryocytes can directly derive from 2010; Hoeffel et al., 2012; Hoeffel and Ginhoux, 2015).
HSCs, both during fetal development and during adulthood Additional studies have clarified that progenitors derived from
(Adolfsson et al., 2005; Notta et al., 2016) (Fig. 2B). the YS give rise to MPs in the brain (microglia), and to a fraction of
In conclusion, our understanding of the hematopoietic lineage is tissue-resident MPs in other organs such as the heart, lung (alveolar
evolving fast, thanks to rapid progress in single cell-based MPs), liver (Kupffer cells), spleen, bone (osteoclasts) and skin
techniques, and has highlighted that the MP lineage can arise at (Langerhans cells) (Merad et al., 2002; Guilliams et al., 2013;
multiple points during development and adulthood (Figs 1 and 2). Hashimoto et al., 2013; Jakubzick et al., 2013; Epelman et al.,
2014b; Molawi et al., 2014; Gomez Perdiguero et al., 2015). Thus,
The cellular origins and maintenance of tissue-resident macrophages with the exclusion of microglia, tissue-resident MP populations in
Tissue-resident MPs are the most abundant immune cell present in other tissues comprise cells of multiple developmental origins
most tissues, yet at resting state they are still rare compared with (Fig. 1), at least some of which are maintained locally from early
parenchymal cells. In 1968, Van Furth and Cohn proposed that development onwards.
tissue-resident MPs originate from circulating blood MOs (van A correct number of tissue-resident MPs needs to be maintained
Furth and Cohn, 1968). This hypothesis has been accepted for many during adulthood. Although the self-renewal of tissue-resident MPs
years without more thorough investigation. More recently, however, is complex, tissue specific and not yet fully understood, we know
the development of new murine models allowing the specific that the long-term maintenance of tissue-resident MPs is essential
labeling of MO/MP populations in vivo (Box 1) has allowed (Aziz et al., 2009; Jenkins and Hume, 2014). Indeed, the disruption
researchers to explore the ontogeny and roles of the different subsets of MP numbers or their inflammatory state can lead to severe defects
of MOs and MPs found in tissues. in organ function during life, leading to early aging (Linehan and
For example, mice that express GFP from the CX3CR1 locus, Fitzgerald, 2015).
which encodes the receptor for the chemokine CX3CL1, have During adult life, a percentage of tissue-resident MPs, variable
DEVELOPMENT

provided a tool for following MOs and MPs within blood and from tissue to tissue, is continuously renewed by blood-derived
tissues (Jung et al., 2000; Geissmann et al., 2003). The subsequent MPs. However, whether these cells should be considered as ‘tissue-
development of CX3CR1-creER mice has allowed lineage tracing resident’ MPs is a matter for debate. In addition, this process does
studies, confirming that at least a subset of tissue-resident MPs is not occur to the same extent in all tissues. For example, microglia
established during embryogenesis and self-renews in peripheral are capable of self-renewal in their tissue of residence and
tissues during adulthood (Yona et al., 2013; Salter and Beggs, are completely independent of definitive hematopoietic
2014). Furthermore, by using CCR2-KO mice, in which MOs lose progenitors for their maintenance, and circulating bone marrow-
their responsiveness to CCL2 and therefore fail to infiltrate tissues, derived myelomonocytic cells only enter the CNS in the context of
Hashimoto and colleagues showed that blood-derived MPs are not disease (Ajami et al., 2007).

2
REVIEW Development (2019) 146, dev156000. doi:10.1242/dev.156000

Fig. 2. Models of adult hematopoiesis.


A Original model LT-HSC
(A) Original model of adult hematopoiesis. In this
simple model, basophils, eosinophils, neutrophils
and monocytes derive from a common progenitor
called a granulocyte-monocyte progenitor (GMP).
ST-HSC (B) Revised model of adult hematopoiesis. The
revised version of hematopoiesis adds layers of
CMP LMPP complexity. For example, neutrophils and
monocytes come from an independent Flt3+
MEP
GMP neutrophil-monocyte progenitor (NMP), unlike
CLP basophils and eosinophils, which arise from a
Gata1+ progenitor. In addition, erythrocytes and
megakaryocytes hail from distinct intermediate
progenitors that arise downstream of
NK cell megakaryocyte-erythrocyte progenitors (MEPs),
Basophil i.e. E-MEPs and MK-MEPs. CLP, common
Monocyte lymphoid progenitor; CMP, common myeloid
Platelet progenitor; E-MEP, erythroid-primed MEP; EMP,
Erythrocyte Eosinophil Neutrophil Lymphocyte eosinophil-mast cell progenitor; LMPP, lymphoid-
primed multipotent progenitor; LT-HSC, long-term
hematopoietic stem cell; MK-MEP, megakaryocyte-
primed MEP; NK, natural killer; NMP, neutrophil-
B Revised model monocyte progenitor; ST-HSC, short-term
LT-HSC
hematopoietic stem cell.

ST-HSC

E-MEP pre-MEP Flt3+


Gata1+

CLP

MK-MEP NMP
EMP
NK cell
Erythrocyte

Lymphocyte
Platelet Monocyte
Basophil Eosinophil Neutrophil

Tissue-resident MPs also display distinct molecular In summary, the ontogeny and behavior of tissue-resident MPs
characteristics depending on their tissue of residence, but the links has started to be unlocked over the past decade, revealing that many
between them are unclear (Ayata et al., 2018). For example, do of these cells originate during embryonic development. Although
resident MPs in different tissues come from the same type of many questions remain, the combination of single cell-based
progenitor during development, suggesting that their specialization technologies and new specific markers for adult tissue-resident cells
takes place due to environmental signals present in the target tissue? should rapidly lead to their answers.
Or do specific progenitors exist, with different predispositions for
infiltrating specific organs, suggesting that at least some of the Infiltrating bone marrow-derived MPs
reported differences are cell autonomous? In this context the In addition to the tissue-resident MPs discussed above, MPs can arise
existence of a niche, composed of tissue-specific cells, which could from MOs that are present within the blood and that infiltrate into
induce MP specialization has been proposed (Gautier and Yvan- tissues. Two types of MOs, classified according to their expression of
Charvet, 2014; Guilliams and Scott, 2017). The fate of resident MPs the marker Ly6C, are found in the circulation in both mice and humans.
following catastrophic damage also appears to vary depending on Their ratio in the blood is dynamic and changes in response to various
the tissue. For example, following ablation in the CNS, microglia stimuli (e.g. bacterial or virus infection, tissue damage and sterile
are restored from a small number of surviving YS-derived cells inflammation). Ly6C+ MOs originate directly from bone-marrow
progenitors, whereas Ly6C− MOs likely arise from circulating Ly6C+
DEVELOPMENT

without contribution from the bone marrow (Elmore et al., 2014;


Huang et al., 2018). In contrast, YS-derived cells in the heart, which MOs (Yona et al., 2013). Functionally, these two populations are
form a sizable subset of tissue-resident MPs, are replaced by blood- different. The Ly6C+ MO subset responds to damage by infiltrating the
derived MOs (Epelman et al., 2014b). It is unclear whether these tissue and then differentiating into MPs. In contrast, Ly6C− MOs do
‘new’ tissue-resident MPs have the same function as the embryo- not infiltrate tissues, but rather patrol the vascular system to detect and
derived ones (Blériot et al., 2015; Machiels et al., 2017). For further likely remove damaged endothelial cells within vessels (Geissmann
consideration about tissue-resident MP ontogeny, fate and self- et al., 2003; Carlin et al., 2013; Jakubzick et al., 2013).
renewal, we refer the reader to recent reviews (Sieweke and Allen, After a long debate on the origins of blood-derived MPs found in
2013; Ginhoux and Guilliams, 2016; Kierdorf and Dionne, 2016). damaged tissue, it is now commonly accepted that infiltrating MPs

3
REVIEW Development (2019) 146, dev156000. doi:10.1242/dev.156000

apoptotic debris but also modulate tissue-specific stem cells through


Box 1. Mouse models for studying MPs the secretion of multiple factors (Freire-de-Lima et al., 2000; Johann
Csf1op/Csf1op: These mice carry a spontaneous null mutation in the et al., 2006; Arnold et al., 2007). Next, these MPs transition to a
Csf1 gene, leading to loss of circulating MOs and tissue-resident MPs pro-restorative status (or the ‘M2’ state, see Box 2 and Table 1).
(but not microglia), with significant effects on the development of many These Ly6C−CCR2−CX3CR1highF4/80high cells are associated
tissues and organs.
Csf1r−/Csf1−: Deletion of both Csf1 and its receptor Csf1r leads to
with fibrosis formation, wound healing and tissue regeneration.
severe defects in hematopoiesis and MPs. Most of the phenotypes Historically, these phenotypes and related polarization states have
observed in these mice are similar to those seen in the Csf1op/Csf1op been defined mostly based on in vitro results. It is therefore unclear
model, but these mice lack additional populations such as microglia (Dai how similar the cells found in damaged tissues are to the MPs that
et al., 2002) and thus exhibit a stronger phenotype. are generated in vitro by exposing bone-marrow progenitors to
CCR2-KO: This model targets CCR2, which is a receptor expressed by specific cytokine cocktails (Martinez et al., 2008). For this reason,
MOs and involved in their chemotaxis. This model is widely used to
and also reflecting a recent rejection of the M1/M2 terminology in
abrogate the infiltration of circulating blood MOs in tissues after damage,
and to analyze MP involvement in this process (Boring et al., 1997). The the literature (Varga et al., 2013; Varga et al., 2016a,b), pro-
CCL2-KO model, in which the ligand for CCR2 is targeted, is a inflammatory M1 MPs will be referred to hereafter as Ly6C+ MPs,
complementary model (Lu et al., 1998). and pro-restorative M2 MPs will be called Ly6C− MPs (see Box 2).
CX3CR1-GFP: In this strain, GFP labels all MOs and MPs. This model is
very useful for FACS-based analysis and in situ visualization of MOs and The function of tissue-resident macrophages in organ
MPs (Jung et al., 2000; Geissmann et al., 2003).
development and homeostasis
CX3CR1creER: This model allows long-lived or self-renewing MP subsets
to be specifically labeled (Yona et al., 2013).
Although significant differences are maintained between tissue-
CD11b-DTR: In this model, CD11b+ cells undergo apoptosis after resident MPs and MO-derived infiltrating MPs, these are not
diphtheria toxin injection. This model thus efficiently depletes circulating obvious, and phenotypically the cells are very similar. This raises
CD11b+ cells but not tissue-resident CD11b+ cells. However, CD11b is questions about the overlap in biological functions between these
also expressed in various leukocytes such as eosinophils and two populations (reviewed by Epelman et al., 2014b). As an
neutrophils, thus the CD11b-DTR model, like the GM-CSF-KO model example, in the bone marrow itself, MPs are required for correct
(below) is not specific to MPs.
GM-CSF-KO (Csf2−/−): This model targets GM-CSF, which is secreted
hematopoiesis. Their depletion affects the HSC niche and induces
by a variety of cells and stimulates granulocyte and MP differentiation. HSC mobilization in the blood (Winkler et al., 2010; Gustafsson and
Although most GM-CSF-KO mice appear to be superficially healthy, they Welsh, 2016). Moreover, MPs are also required to be in direct
exhibit altered hematopoiesis. contact with erythroblasts in the bone marrow for proper
PU.1-KO: These mice are deficient for Pu.1, which encodes an ETS family erythropoiesis (Chow et al., 2013). These two examples
transcription factor that is exclusively expressed by the hematopoietic demonstrate that tissue-resident MPs are crucial even for their
lineage. Pu.1-deficient mice die in the 48 h following birth owing to the
own homeostasis. However, animals congenitally deficient for
absence of a range of immune cells (McKercher et al., 1996).
tissue-resident MPs have reduced survival, making the study of MP
function during embryogenesis or postnatal growth a difficult task.
For example, mice carrying a spontaneous mutation in Csf1 (op/op
(in contrast to tissue-resident MPs) only arise from Ly6C+ MOs mice), a factor that is essential for the maintenance of mature MPs,
recruited from the bloodstream, and are associated with the present many abnormalities (Dai et al., 2002). This is owing to a
scavenging of damaged tissue components, wound healing and lack of osteoclasts, which are multinucleated cells that are formed by
tissue remodeling (Arnold et al., 2007; Mounier et al., 2013; Juban fusion of MPs with each other and are required for bone resorption.
et al., 2018). As a result, bone deposition is excessive in these mice, cavities
After tissue injury, MO infiltration is dramatic, and essentially for bone marrow are not formed and hematopoiesis is deeply
equivalent in the majority of tissues, with the noticeable exception altered (Dai et al., 2002), making it hard to distinguish direct and
of the CNS, in which infiltration following sterile damage is limited indirect effects of the lack of MPs. Likewise, deletion of the
because of the blood-brain barrier (Engelhardt and Ransohoff,
2012). Infiltration involves CCR2, the receptor for the tissue-
damage induced chemoattractant CCL2. CCR2 is highly expressed Box 2. The M1-M2 paradigm for macrophages
in Ly6C+ MOs and is required for them to infiltrate tissues. The M1 and M2 terminology was initially used to parallel the Th1 and Th2
Importantly, studies of CCR2-KO mice (Box 1), in which MOs fail nomenclature used to describe T cells (Mills et al., 2000). Indeed, Th1-
to infiltrate in response to damage, have revealed that blood-derived skewed mice (C56BL/6) are more prone to produce M1 macrophages,
MPs are required for the efficient regeneration of many tissues whereas Th2-skewed mice (BALB/c) are more biased towards M2
(Boring et al., 1997). macrophage production (Mills et al., 2000; Ley, 2017). However, these
phenotypes were defined based on in vitro results using INFγ (M1), IL4
Functionally, blood-derived MPs are plastic cells that can adopt
(M2a), IL10 (M2c) and opsonized bacteria/lipopolysaccharide (M2b) in
different transcriptional programs depending on the cytokines that different combinations (Martinez et al., 2008). Although this
are present in the microenvironment, assuming a range of functions nomenclature has been helpful, and is still used because of its
DEVELOPMENT

spanning from the promotion of inflammatory and adaptive immune simplicity, it is unclear how similar the cells found in vivo during sterile
responses to their inhibition in favor of tissue regeneration (Epelman damage are to the MPs generated in vitro starting from bone-marrow
et al., 2014b). In sterile damage, these ‘polarization’ states are often progenitors. In this Review, we therefore have opted to use a more
descriptive nomenclature for MPs based on Ly6C expression, i.e. Ly6C+/
not the result of classical, irreversible binary cell fate decisions but
Ly6C−. We understand that this terminology is still not perfect, as more
rather are sequentially acquired according to a well-defined than two ‘flavors’ of MPs exist in vivo. However, Ly6C is still the main
temporal succession. Early after damage, Ly6C+ MOs enter the marker used to differentiate the wide categories of inflammatory and
tissue and differentiate into pro-inflammatory MPs (classically restorative MPs and thus, we believe, is one of the best ways to describe
called ‘M1’ MPs, see Box 2). These cells, which are them.
Ly6C+CCR2+CX3CR1midF4/80 (Adgre1)mid (Table 1), scavenge

4
REVIEW Development (2019) 146, dev156000. doi:10.1242/dev.156000

Table 1. MO and MP types and features


Localization Name Phenotype Characteristics
+ + + low low low
Blood Ly6C MO Ly6C CCR2 CX3CR1 F4/80 CD11b Infiltrate tissues when required.
Ly6C− MO Ly6C−CCR2−CX3CR1hiF4/80lowCD11blow Patrol the vascular system.
Resting state tissue Embryonic-derived resident MPs Ly6C−CCR2−F4/80hiCD11blow Independent self-renewal.
Bone marrow-derived resident MPs Ly6C−CCR2−F4/80lowCD11bhigh Continuous self-renewal by circulating MOs.
Exist in all tissues except CNS.
Damaged tissue Infiltrating MPs Ly6C+CCR2+CX3CR1lowF4/80lowCD11blow Associated with early phase of sterile inflammation.
Ly6C− MPs Ly6C−CCR2−CX3CR1hiF4/80hiCD11bhi Come from Ly6C+ MPs. Associated with tissue
remodeling.

hematopoietic-specific transcription factor PU.1 (Spi1) causes number of microglia increases significantly during brain
lethality in the late embryo or soon after birth. However, by using development, owing to both local proliferation and additional
a conditional LysM-Cre PU.1 floxed model, it has been demonstrated waves of infiltration (Ginhoux et al., 2010; Erblich et al., 2011). In
that PU.1 is dispensable for HSC development, allowing the addition to playing a role in embryonic and fetal brain development,
community to investigate hematopoiesis more thoroughly microglia are required for normal postnatal development, likely by
(McKercher et al., 1996; DeKoter et al., 1998; Dakic et al., 2005). modulating interneuronal connections through synaptic pruning
Similarly, other conditional knockout mouse models (see Box 1) have (Graeber and Streit, 2010; Tremblay et al., 2010; Paolicelli et al.,
aided the study of tissue-resident MPs, revealing how these cells arise 2011; Fig. 3). This phenomenon is thought to be modulated by
and are maintained, and highlighting organ-specific roles for these CX3CR1, which is expressed at high levels in microglia. Indeed
cells during embryogenesis and early development. CX3CR1-KO animals have a higher number of unconnected
synapses, which is reminiscent of the immature brain and similar to
Central nervous systems what can be seen in neurodevelopmental disorders (Graeber and
Microglia, first described in 1932 (del Rio-Hortega, 1932), are the Streit, 2010; Tremblay et al., 2010; Paolicelli et al., 2011). Moreover,
resident MPs of the CNS. Microglia are phenotypically identical to microglia are able to recognize CD47, which appears to modulate
other MPs and share the ability to respond to damage and to pruning of neuronal projections (Lehrman et al., 2018; Fig. 3). CD47-
phagocyte cell debris. However, as mentioned above, microglia KO mice exhibit increased engulfment of neurons by microglia,
have distinct developmental origins: although the timing of their resulting in a decrease in synaptic connections (Lehrman et al., 2018).
colonization of the brain is still debated, and is likely to happen at Interestingly, the phagocytic activity of microglia is different
multiple stages of embryonic life, it is now accepted that microglia depending on their location in the CNS, and depends on epigenetic
derive mainly from the YS at E8.5-E9 (Alliot et al., 1999; Herbomel changes likely acquired locally (Ayata et al., 2018).
et al., 2001; Ajami et al., 2007; Chan et al., 2007; Ginhoux et al., Microglia are also involved in angiogenesis and vessel sprouting
2010; Hashimoto et al., 2013; Salter and Beggs, 2014), before the during brain development, acting via the secretion of VEGF (Fig. 3).
origins of the first HSCs and of most adult MPs. Interestingly, This well characterized pro-angiogenic function is shared with other
microglia formation is independent of the CSF1/CSF1R axis, but non-CNS resident MPs, and represents an important pro-cancer
rather depends on IL34, an alternative ligand for CSF1R (Erblich function of tumor-associated MPs (TAMs) (Fantin et al., 2010).
et al., 2011; Schulz et al., 2012). Finally, by analogy with other tissue-resident MPs, microglia are
Microglia have important functions during development and adult thought to acquire different functions depending on the local
life (reviewed in detail by Hammond et al., 2018). As an example, a microenvironment, a process often called polarization. Such
lack of microglia in Csf1r−/− mice results in functional deficiencies, proposed functions range from inflammatory roles, involving the
for example in olfaction processing, pointing to a requirement for release of reactive oxygen species (ROS), nitric oxide (NO) or
these cells during development (Erblich et al., 2011). Indeed, the TNFα, to pro-neurogenic roles. It has also been proposed that, in

Neuron pruning Fig. 3. The functions of microglia. Microglia are YS-


(CX3CR1/CX3CL1) derived tissue-resident MPs that are able to self-renew
throughout adulthood. They actively participate in neuron
Microglia and synapse pruning activity via CX3CR1/CX3CL1 and
CD42 recognition, and in angiogenesis via the secretion of
VEGF during brain development.

Self-renewal
DEVELOPMENT

Vessel sprouting
Synapse pruning (VEGF)
(CD47 recognition)

5
REVIEW Development (2019) 146, dev156000. doi:10.1242/dev.156000

some cases, microglia activation can have a direct neurotoxic effect follicles, and the hypodermis, which contains sweat glands, fat and
(Hellwig et al., 2013). However, the ability of microglia to polarize, blood vessels. Various populations of MPs and dendritic cells are
and in particular to polarize to functional states that resemble those known to be present in the skin but, owing to their phenotypic
described for MPs in other tissues, is still controversial and based overlap, the distinction between them is ambiguous (Henri et al.,
more on analogy than solid data (Wang et al., 2016). 2010; Tamoutounour et al., 2013). Aside from dendritic cells, at least
two types of tissue-resident MPs are present in the skin: Langerhans
The cardiovascular system cells are mainly present in the epidermis, whereas cutaneous (or
Tissue-resident MPs in the heart comprise a mixture of cells from dermal) MPs are more present in the dermis (Di Meglio et al., 2011;
both YS and fetal liver origins (Epelman et al., 2014b). The self- Tamoutounour et al., 2013; Mojumdar et al., 2014). Of note,
renewal capacity of these cardiac-resident MPs is unclear and Langerhans cells share properties with both MPs and dendritic cells
appears to depend on their anatomical location. Indeed, whereas (e.g. self-renewal and T cell stimulation), making them an
some publications report a slow but progressive replacement of unclassified cell type (i.e. neither MP nor dendritic cell) (Doebel
heart-resident MPs by blood-derived MPs with age (Epelman et al., et al., 2017). In terms of their developmental origins, both
2014a; Molawi et al., 2014), others suggest that less than 1% of the Langerhans cells and cutaneous MPs are highly heterogeneous.
MPs localized in the atrioventricular bundle are replaced (Hulsmans Indeed, Langerhans cells are first derived from YS progenitors but are
et al., 2017) (Fig. 4). More recently, it has been demonstrated that later gradually replaced by precursors from fetal liver hematopoiesis
cardiac TIMD4+ MPs are self-renewing with little input from (Fig. 1) (Hoeffel et al., 2012). In contrast, cutaneous MPs appear to
infiltrating MPs, whereas cardiac TIMD4− MPs are almost fully originate from the fetal liver and are then gradually replaced by
replaced by infiltrating MPs (Dick et al., 2019). Whether cardiac circulating Ly6C+ MOs (Varol et al., 2009) (Fig. 1). Langerhans cells
MPs are required during normal development is still unclear. In are also able to self-renew, but Ly6C+ MOs can be recruited to replace
contrast to the adult heart, the neonatal heart is able to fully these cells after damage (Tamoutounour et al., 2013).
regenerate, and the depletion of circulating MOs (using clodronate Until after birth, tissue-resident MPs (both Langerhans cells and
liposomes) blocks this regenerative capacity (Porrello et al., 2011; cutaneous MPs) are the only immune cells to appear in the skin
Aurora et al., 2014). This could be because of the absence of (Tamoutounour et al., 2013). So far, the function of tissue-resident
angiogenesis in MP-depleted mice (Aurora et al., 2014). Although MPs during development of the skin is unknown, however we
the specific molecular mechanism underlying this observation is not hypothesize that they are required for extracellular matrix formation
known, the authors’ suggestion that a lack of MP-derived VEGF may and/or deposition and skin layer differentiation. The number of
be crucial appears to be reasonable (Fig. 4). This function matches these tissue-resident MPs in the skin also changes during normal
the pro-angiogenic function of microglia described above and, by hair cycles, suggesting roles for them during hair follicle
analogy, also suggests that cardiac MPs are required for the development. For example, the number of Langerhans cells and
establishment of a full blood coronary tree during development cutaneous MPs increases in anagen (the active hair growth phase)
(Fantin et al., 2010). Lastly, it has been described that, at resting state, and decreases in telogen (the resting phase), because of MP
heart-resident MPs are particularly abundant in the atrioventricular apoptosis (Osaka et al., 2007; Wang et al., 2017). Concomitantly, an
node, leading to the proposal that they actively communicate with increase in Wnts expressed by MPs is found locally, activating
cardiomyocytes via GAP junctions to regulate cardiac rhythm epithelial cells to initiate the anagen phase (Paus, 1998; Castellana
(Hulsmans et al., 2017) (Fig. 4), again demonstrating the et al., 2014) and suggesting that MPs modulate the activity of hair
importance of tissue-resident MPs. follicle stem cells. Consistent with this notion, it has been shown
that Ly6C− MPs secrete TGFβ1 to activate follicular stem cells, and
The skin thus hair regrowth, in the context of wound healing (Rahmani et al.,
The skin is composed of three layers: the epidermis, which directly 2018). Interestingly, this phenomenon is dependent on CX3CR1
faces the external environment, the dermis, which contains hair but not CCR2 (Rahmani et al., 2018), suggesting it may not involve

Fig. 4. The regulation and function of tissue-resident MPs in


the heart. MPs present in the atrioventricular (AV) node of the
AV node-resident MPs heart autonomously self-renew and actively participate in the
(TIMD4+) establishment of cardiac rhythm, acting via GAP junctions. By
contrast, tissue-resident macrophages in the myocardium are
replenished via circulating MOs and facilitate angiogenesis via
the secretion of VEGF.
Self-
renewal

Maintain electrical
contraction
(GAP junctions)
DEVELOPMENT

Angiogenesis
(VEGF)

Ly6C+ MOs Replenishment


Blood
Heart-resident MPs
(TIMD4−)

6
REVIEW Development (2019) 146, dev156000. doi:10.1242/dev.156000

blood-derived, infiltrating cells. In addition, MPs can modulate hair (Schulz et al., 2012; Mass et al., 2016). After liver colonization, a
cycle and hair growth after plucking via the secretion of TNFα distinct transcriptional program (i.e. different to that in YS-derived
(Chen et al., 2015). cells) is activated, specifically involving the transcription factor
inhibitor of DNA binding 3 (ID3) and leading to the development of
The spleen and liver Kupffer cells that are endowed with self-renewal activity (Mass
Beyond their important functions in immunity, tissue-resident MPs et al., 2016).
in the spleen play crucial roles related to the spleen’s function as the One important non-immune function of Kupffer cells is the
filter of the bloodstream (Fig. 5). Thus, it is perhaps not surprising modulation of metabolism in hepatocytes via PPARγ, preventing the
that various subsets of these cells exist, segregated in the different pathogenic accumulation of lipids and thus hepatic steatosis
functional areas of this organ. Splenic-resident MPs originate from (Desvergne, 2008; Kang et al., 2008; Odegaard et al., 2008). After
the fetal liver, are Spi1 (PU.1-related transcription factor)- injury, the main function of Kupffer cells is to clear dead cells and
dependent and self-renew mostly independently of blood-derived pathogens in order to protect the liver (Nguyen-Lefebvre and
cells (Kohyama et al., 2009; Hashimoto et al., 2013; Yona et al., Horuzsko, 2015). Following acute damage (e.g. carbon tetrachloride
2013). Tissue-resident MPs within the red-pulp region of the spleen injection or bile duct ligation), Kupffer cells (CD11blowF4/80+) are
are CD11b (Itgam)low, F4/80+ and CD206 (Mrc1)+ and have activated by damage-associated molecular patterns (DAMPs),
important functions in iron processing connected with their main upregulate Ly6C, an event that may be indicative of activation
function: the clearance of damaged red blood cells (Kohyama et al., rather than polarization into M1, and contribute to the attraction of
2009). In contrast, MPs in the white-pulp region express different circulating MOs (by secretion of CX3CL1, CCL2 and CCL5) and
markers (e.g. CD68 and MERTK) and mainly phagocytose other cells such as neutrophils, basophils or natural killer cells (via
lymphocytes (Kohyama et al., 2009). Defects in the function of CXCL16) (Nguyen-Lefebvre and Horuzsko, 2015; Wynn and
these white-pulp MPs lead to B cell accumulation and auto antibody Vannella, 2016) (Fig. 5). Basophils are also of importance for liver
development, providing a link between their scavenger and immune repair, as they are a source of IL4, a pro-proliferative cytokine for
roles (Khan et al., 2013). hepatocytes (Blériot et al., 2015). Thus, appropriate coordination
The liver contains a specialized population of resident MPs called between resident Kupffer cells and cells infiltrating from the
Kupffer cells, named after Karl Wilhelm von Kupffer who bloodstream is required for damage resolution and maintenance of
discovered them in 1876 (von Kupffer, 1876). Until recently, it liver function.
was commonly accepted that, between E10.5 and E12, AGM-
derived HSCs give rise to fetal liver MOs, which later differentiate The role of infiltrating blood-derived macrophages in tissue
into Kupffer cells (Gomez Perdiguero et al., 2015). However, this homeostasis and repair
model was recently disputed. Indeed, it was shown that YS-derived After completion of postnatal growth, organs and tissues need to
erythro-myeloid progenitors are also able to generate a common maintain homeostasis for a prolonged period of time to preserve
circulating precursor, which colonizes the embryo from E9.5 and function and integrity. The involvement of blood-derived MPs in
directly gives rise to Kupffer cells in a CX3CR1-dependent way organ and tissue homeostasis is paramount. A detailed discussion of

Liver Blood Spleen

CCL2/CCL3/Csf1 Ly6C+ MOs Phagocytosis

Fe+

Ly6C+ MPs Fe+


Ly6C+ MOs
Fe+
(iron charged
from spleen) Dead Fe+
erythrocytes

Fe+ Tissue-resident MPs


Fe+ (CD206)
Fe+
Kupffer Fe+ Dead
cells lymphocytes
DEVELOPMENT

Ly6C− MOs

Phagocytosis

Tissue-resident MPs
(CD68/MERTK)

Fig. 5. The functions of tissue-resident MPs in the liver and spleen. In the spleen, circulating Ly6C+ MOs and tissue-resident MPs (marked by CD206 or
CD68/MERTK expression) phagocytose dead erythrocytes and lymphocytes. Iron-charged Ly6C+ MOs then migrate to the liver, guided by the secretion
of CCL2, CCL3 and Csf1 from Kupffer cells, and locally liberate the iron for hepatocytes to take up. In the liver, Kupffer cells continuously phagocytose dead cells
and debris (not shown) in order to protect the liver against pathogenicity.

7
REVIEW Development (2019) 146, dev156000. doi:10.1242/dev.156000

the specific roles of these MPs in every tissue is beyond the scope of CCR2 delays the infiltration of myelomonocytic cells after damage,
this review. We therefore focus below on the roles of blood-derived causing a strong impairment of skeletal muscle regeneration and the
MPs in the homeostasis, as well as the regeneration, of four well- appearance of diffused fibrosis, demonstrating that these cells are
studied tissues: skeletal muscle, cardiac muscle, the liver and skin crucial for efficient healing (Warren et al., 2005). These findings
(Fig. 6). were validated by depleting CD11b+ cells using the CD11b-DTR
mice model (Box 1; Jung et al., 2000; Arnold et al., 2007).
Skeletal muscle regeneration Early after skeletal muscle damage, Ly6C+ MPs express various
Despite the fact that skeletal muscle was one of the first tissues in inflammatory markers (see below) but lose Ly6C within 48-72 h
which the crucial roles played by MPs in damage-induced (Arnold et al., 2007; Mounier et al., 2013; Saclier et al., 2013a,b;
regeneration were revealed, a developmental role for macrophages Varga et al., 2016a,b). This phenotype switch, often called skewing,
in this tissue has not been investigated. MPs are present within also occurs in multiple other tissues (e.g. heart, liver or kidney) (Sica
muscle at steady state and are found in the perimysium (connective et al., 2015) and is regulated at the DNA, RNA and protein levels
tissue surrounding muscle fascicles) and the epimysium (fascia throughout the regenerative process (Ruffell et al., 2009; Mounier
surrounding the muscle) (Saclier et al., 2013b). However, a lack of et al., 2013; Varga et al., 2016a,b). Soon after infiltration, Ly6C+ MPs
tools to discriminate tissue-resident versus infiltrating MPs in also express molecules such as TNFα (at high levels), IL6 and IL1β.
skeletal muscle has made it difficult to ascertain the precise role of Beyond their well-known roles as inflammatory mediators, these
tissue-resident MPs in development. In contrast, it is well factors also act as mitogens, stimulating myogenic cell proliferation
established that infiltrating MOs are required for efficient skeletal and inhibiting their differentiation/fusion into damaged fibers
muscle regeneration following acute damage (Fig. 6A). Deletion of (Sonnet et al., 2006; Arnold et al., 2007; Saclier et al., 2013a,b).

A Skeletal muscle B Heart


Skeletal muscle tissue Blood Cardiac tissue Blood

Ly6C+ MP Ly6C+ MP
Myogenic Ly6C+ MO
cell IL6, IL1β,
IL13

TNFα Ly6C+ MO

FAP Promote scar


formation
TGFβ
,
TGFβ, IL10
GDF3 Ly6C− MP

Ly6C− MO
Ly6C− MP Ly6C− MO
FAPs/fibroblasts

Myofiber Replace resident MPs

C Skin D Liver
Liver Blood
Hair follicle stem cell
Langerhans activation and growth TGFβ1 CXCL16 NK and neutrophil
cells recruitment
Kupffer cell
(Ly6C+)
CX3CR1+ Debris
Anagen Telogen clearance
Apoptosis (hair growth) (resting state)

Ly6C+ MO

Cutaneous MPs
PPARγ
Ly6C+
CX3CL1, CCL2, MO recruitment
Lipid CCL5
Infiltration accumulation
Blood
Hepatocyte
DEVELOPMENT

Fig. 6. Tissue-resident and infiltrating MPs act together to orchestrate tissue repair. (A) After damage, Ly6C+ MOs infiltrate skeletal muscle, differentiate into
Ly6C+ MPs and secrete cytokines such as IL6, IL1β, IL13 or TNFα to support myogenic cell proliferation and to ablate excess fibro/adipogenic progenitors
(FAPs, orange cell). Next, these Ly6C+ MPs transition into Ly6C− MPs, which then activate an anabolic program via TGFβ, IL10 or GD3, resulting in differentiation
of myogenic cells and remaining FAPs (green cells). (B) In the heart, circulating MOs infiltrate the tissue following myocardial infarction, differentiate into Ly6C−
MPs and quickly provide trophic support to FAPs in order to promote fibroblast differentiation and scar formation. (C) The number of Langerhans cells and
cutaneous MPs in the skin changes with the hair growth cycle. During anagen (hair growth), the number of resident MPs increases, favoring hair stem cell
expansion through TGFβ. They then decrease in telogen (the resting phase), because of MP apoptosis, and are replenished by circulating Ly6C+ MOs. (D) After
liver damage, Kupffer cells become activated, upregulate Ly6C and secrete CXCL16, CXC3CL1, CCL2/3 and CCL5 to attract natural killer (NK) cells, neutrophils
and Ly6C+ monocytes. These cells then clear the site of damage and block lipid metabolism in hepatocytes to avoid lipid accumulation and pathogenicity.

8
REVIEW Development (2019) 146, dev156000. doi:10.1242/dev.156000

It is conceivable that, in the context of sterile damage, the non-


inflammatory functions of these factors may be prevalent. Box 3. Macrophage functions in degenerative disease –
Another key function of Ly6C+ MPs, more in line with their DMD as a case study
classical role as scavengers, is the removal of debris via their In some pathologies, tissue regeneration is impaired. For example,
phagocytic capacities. Phagocytosis also plays a role in triggering degenerative myopathies, such as Duchenne Muscular Dystrophy
the transition of MPs to a pro-restorative phenotype through the (DMD), are muscle diseases associated with a continuous succession
of degeneration/regeneration cycles, eventually leading to disruption of
activation of signaling pathways linked to metabolic control such as the regenerative process and alterations to the fate and function of
AMP-activated kinase (AMPK) (Mounier et al., 2013). As a result multiple cell types (Dadgar et al., 2014). DMD is associated with
of skewing, the cytokines secreted by MPs change dramatically: increases in inflammatory cells, cytokines, fibrosis and lipid droplets
inflammatory factors such as TNFα are downregulated whereas pro- (Petrof et al., 1993; Barton, 2006; Desguerre et al., 2012). In this context,
regenerative factors such as IL10, IGF1, VEGF, and TGFβ are a clear role for inflammatory cells in modulating fibrosis has been shown.
produced and stimulate muscle cell survival, differentiation, fusion For example, whereas MPs act to clear stromal progenitors in the context
of normal skeletal muscle regeneration, MPs found in chronically
and fiber growth (Fig. 6A) (Arnold et al., 2007; Mounier et al.,
damaged muscle, despite being similar to early MPs in terms of
2013; Saclier et al., 2013a; Tonkin et al., 2015). More recently, it has surface phenotype, promote fibro/adipocytic progenitor survival and
been shown that the transcription factor PPARγ is required for such fibrogenic differentiation (Lemos et al., 2015; Juban et al., 2018).
pro-restorative effects and, in particular, for secretion of the TGFβ Interestingly, this phenotype may be reversed by treating mice with
family member growth differentiation factor 3 (GDF3). Indeed, metformin, an activator of AMPK (an enzyme involved in MP-skewing
suppression of either PPARγ or GDF3 leads to a delay in the following acute damage; Mounier et al., 2013). Thus, the failure of
skeletal muscle regeneration in muscular dystrophies may be due to a
anabolic processes taking place in the late phase of regeneration
disruption of the carefully timed switch from a pro-inflammatory
(myogenic cell fusion and myofiber growth) (Fig. 6A) (Varga et al., environment to a pro-regenerative environment normally observed in
2016a,b). Altogether these results demonstrate that the metabolic response to acute damage. Indeed, in chronic disease, the
and functional states of MPs are highly correlated (Mounier et al., asynchronous nature of the damage leads to contrasting signals being
2013; Varga et al., 2016a,b). present within the muscle at the same time. This is likely to impair the
During the later stages of skeletal muscle regeneration, vessel functions of both myogenic and stromal progenitors, resulting in
remodeling is required to restore muscle function, which also disrupted regeneration and fibrofatty infiltration (Dadgar et al., 2014;
Tidball et al., 2014; Lemos et al., 2015; Tidball, 2017; Juban et al., 2018).
appears to involve MPs (Zordan et al., 2014; Latroche et al., 2017). In the future, a better understanding of MP functions and gene
Ly6C− MPs express matrix-remodeling enzymes such as matrix expression profiles in chronic disease is likely to lead to novel
metallopeptidases (MMP) 2, 13 and 14, and VEGF, and are therapeutic approaches for chronic disease.
therefore good candidates to mediate vessel remodeling (Zordan
et al., 2014). However, the precise roles of Ly6C+ and Ly6C− MPs
or even of tissue-resident MPs in damage-induced angiogenesis
have yet to be addressed in depth. MPs mainly promote matrix deposition and support scarring
Ly6C+ MPs also play a role in regulating fibrosis and fibrogenic (Fig. 6B) (Aurora et al., 2014). These differences in MP function
differentiation during muscle regeneration. During early regeneration, in neonatal versus adult heart repair could be because of distinct
stromal progenitors called fibro/adipocytic progenitors (FAPs) phenotypes of MPs. Indeed, at least some tissue-resident MPs in the
expand dramatically to establish a trophic environment but are then heart are of embryonic origin, and are phenotypically more similar
cleared, thus preventing their differentiation and the formation of to Ly6C− MPs than to Ly6C+ MPs (Aurora et al., 2014). Again,
fibrofatty infiltration. The clearance of these cells is mediated by depletion of tissue-resident or infiltrating MPs before or following
Ly6C+ MPs through the secretion of TNFα (Lemos et al., 2015). In cardiac injury has been performed in many ways. Whereas depletion
contrast, TGFβ, an anti-inflammatory cytokine mainly secreted by of circulating MOs at an early point in repair negatively affects
Ly6C− MPs at later stages of regeneration, provides protection to cardiac repair, depleting them late after myocardial infarction in fact
FAPs from apoptosis (Lemos et al., 2015), highlighting how crucial improves cardiac recovery, highlighting the changing roles that
the timing of the switch from Ly6C+ to Ly6C− MPs is for the these cells play at different stages of the response to damage (Van
restoration of tissue function. This is also the case in the heart, where Amerongen et al., 2007; Heidt et al., 2014; Sager et al., 2016).
FAPs and MPs collaborate in the response to damage (Wang et al., Moreover, depletion of tissue-resident MPs using CX3CR1-DTR
2015). Notably, this switch may be perturbed in the context of mice affects cardiac repair after myocardial infarction in the adult.
regenerative diseases such as Duchenne Muscular Dystrophy (DMD; This appears to be because of the massive infiltration of new MPs
see Box 3). and the proliferation of the remaining tissue-resident MPs (Dick
et al., 2019). To conclude, it appears that both tissue-resident MPs
Cardiac muscle repair and infiltrating MPs have important functions in cardiac repair, and
Cardiac damage, such as myocardial infarction, triggers massive boosting one or the other systems at a precise time could be a
inflammation. However, in this context, tissue-resident MPs die at potentially useful therapeutic tool.
the site of damage and the infiltration of new MPs that give rise to
DEVELOPMENT

tissue-resident MPs is required for cardiac tissue repair (Heidt et al., Skin and hair growth/repair
2014). As in other tissues, it is now accepted that damage recruits a Following skin damage, MPs are recruited and actively participate
unique wave of Ly6C+ MOs, followed by their skewing into a in wound healing (Fig. 6C). Ly6C+ MOs infiltrate the skin,
Ly6C− profile (Fig. 6B; Hilgendorf et al., 2014, Pinto et al., 2014; providing immune protection against external pathogens, but also
Frangogiannis, 2015). act on stromal cells, keratinocytes and endothelial cells (Wang et al.,
Consistent with the role of MPs in supporting cardiac 2017). Again, depletion of circulating MOs using GM-CSF-KO,
angiogenesis during neonatal development, infiltrated MPs are CD11b-DTR or CCR2-KO mice highlights their importance in this
capable of stimulating cardiac angiogenesis following perinatal process (Fang et al., 2007; Mirza et al., 2009). Indeed, angiogenesis
ischemic damage (Aurora et al., 2014). However, in adult infarction, is strongly decreased owing to CD31+ cell apoptosis (likely because

9
REVIEW Development (2019) 146, dev156000. doi:10.1242/dev.156000

of decreases in TGFβ and VEGF secretion), strongly delaying Chronic inflammation and disease
wound healing (Lucas et al., 2010). Infiltration of circulating MOs The roles of MPs in disease are so varied that it is not possible to
(and other immune cells) to the site of healing is driven by activated provide a full and comprehensive review here. However, as these
keratinocytes and the secretion of several chemokines such as cells are crucially important in many human afflictions, we have
CXCL10, CXCL11, CCL5 and CCL20 (Banno et al., 2004). In selected below a few examples, illustrating their non-immune
return, MPs activate keratinocyte proliferation and reepithelization functions in pathological conditions, and their interactions with
via the secretion of members of the epidermal growth factor (EGF) tissue-resident stem cells in these contexts.
family (Hancock et al., 1988; Shirakata, 2005). Although this direct Type II diabetes, which is associated with obesity, is on the rise and
interaction has been demonstrated in vitro, it is not as clear-cut in accounts for a significant number of deaths every year. Obesity is
vivo (Grose and Werner, 2004). associated with inflammation, and an accumulation of MPs in
Interestingly, wound-induced macrophages acquire a profile that adipose tissue is thought to predispose to insulin resistance (Weisberg
differs from the aforementioned Ly6C+/Ly6C− progression that is et al., 2003). In this context, it is of interest that high fat diet-induced
observed in other tissues. Indeed, from day 1 to day 7 after insulin resistance is reduced in mice lacking the myelomonocytic
wounding, MPs express CD206, arginase 1 and Ym1 (Chil3), integrin CD11b, and this correlates with an increase in adipose tissue-
which are markers typical of pro-regenerative cells that appear at resident MP proliferation and an inhibition of MO recruitment from
later stages of regeneration in other tissues (Mounier et al., 2013). the circulation (Zheng et al., 2015). This suggests that Ly6C+ MPs
However, IL4 and IL13, which are cytokines described as being freshly immigrated from the bloodstream may play a pathogenic role
required for an alternative MP activation pathway, are not present in in Type II diabetes. Consistent with the idea that they modulate
the wound, which suggest that other types of cytokines must be at adipose metabolism, MPs have been proposed to contribute to
play. Ly6C and TNFα are downregulated, whereas the secretion of thermogenesis regulation in brown adipose tissue by secreting
TGFβ and VEGF is increased to accelerate the process of wound catecholamine, although this controversial notion has recently been
healing, as described above for muscle (Daley et al., 2010). TNFα questioned (Nguyen et al., 2011; Fischer et al., 2017).
secreted by MPs is important for the activation of hair follicle stem Obesity also causes adipocyte apoptosis and thus damages the
cells after damage (Wang et al., 2017). Indeed, whereas TNFα- signals that attract Ly6C+ MOs, leading to their significant increase
deficient mice show delays in hair growth, TNFα actually stimulates in white adipose tissue. This may further contribute to adipocyte
the telogen-anagen transition required for hair growth (Wang et al., enlargement and apoptosis, perpetuating a vicious circle (Sun et al.,
2017). To conclude, during wound healing, MPs are the source of 2011). As Ly6C+ MPs engulf dead adipocytes, they become
multiple cytokines and growth factors acting on hair follicle stem engorged by fatty acids, resulting in the adoption of a highly
cells, endothelial cells and keratinocytes. inflammatory phenotype including secretion of TNFα and/or IL6.
This increase in inflammatory cytokines in the blood stream results
Liver regeneration in chronic systemic inflammation and contributes to insulin
Infiltrating MPs play unique functional roles in both the resting and resistance (Lumeng et al., 2007). Thus, increasing the skewing of
regenerating liver (Fig. 6D). For example, Ly6C+ MOs engulf stressed MPs into a more mature phenotype (Ly6C−) may represent a
red blood cells in the bloodstream or the spleen, migrate to the liver and promising therapy to treat obesity-induced chronic inflammation
differentiate into Ly6C+ MPs in a process supported by CCL2, CCL3, and its effects.
and Csf1 released from Kupffer cells. These iron-charged MPs express Tissue-resident MPs in the liver, Kupffer cells, are also involved
high levels of ferroportin1, allowing them to dispense iron to in metabolic homeostasis and related pathologies. Kupffer cells
hepatocytes (Gammella et al., 2014; Theurl et al., 2016). secrete high levels of IL4 and IL13, possibly because of their low
As in most other tissues, MPs play key roles during regeneration levels of PPARγ, a transcription factor that promotes the acquisition
in the liver, and undergo a skewing process essentially identical to of an anti-inflammatory phenotype and the loss of Ly6C. The
that described for skeletal muscle, and likely just as important. acquisition of this pro-inflammatory phenotype contributes to
CD11b-DTR-mediated myelomonocytic cell depletion at the early activating fatty acid oxidation by hepatocytes and eventually leads
stages (the acute damage phase) of regeneration leads to reduced to liver failure due to lipid accumulation (Kang et al., 2008;
fibrosis, whereas depletion at late stages (the recovery phase) leads Odegaard et al., 2008).
to the impairment of matrix degradation, inducing more fibrosis MPs also play a key role in the pathogenesis of autoimmune
(Duffield et al., 2005). It has also been shown that blood-derived metabolic diseases. For example, pancreatic islets of Type II diabetic
Ly6C− MPs are present in large numbers during fibrosis resolution, patients and rodent models are massively infiltrated by Ly6C+ MOs
which they likely promote through their secretion of MMP enzymes and MPs, likely attracted by cytokines produced locally by
(Ramachandran et al., 2012). In CCR2-deficient mice, in which MO autoimmune T cells. In turn, IL1β, CCL2 and CX3CL1 released
infiltration is reduced, damage-induced liver fibrosis is increased, as from these Ly6C+ MPs contribute to pancreatic dysfunction by
occurs in the case of cardiac and skeletal muscle, suggesting that recruiting new inflammatory TNFα-secreting Ly6C+ MOs, and
loss of the matrix remodeling function of Ly6C− cells has a possibly starting a vicious cycle (Ehses et al., 2007).
dominant effect (Mitchell et al., 2009). As in most other tissues that In summary, it appears likely that MPs mediate a number of the
DEVELOPMENT

have been analyzed, liver MPs also directly influence the function of pathogenic effects that inflammation has on metabolism, and
tissue-specific stem/progenitor cells. For example, it was patients may benefit from treatments modulating the functional state
demonstrated that, in a model of biliary and hepatocyte of these cells.
regeneration, blood-derived MPs secrete Wnt ligands and remodel
extracellular matrix to induce hepatocyte differentiation of Conclusions and perspectives
progenitor cells (Boulter et al., 2012). After the resolution of The more we study MPs, the more we realize that the early simplistic
inflammation, it is possible that blood-derived MPs replace the views of these cells as ‘bug gobblers’ only describes a small portion
missing Kupffer cells, thereby maintaining a pool of tissue-resident of their core functions. Indeed, we now know that these cells act as a
MPs ready for further needs (Blériot et al., 2015). distinct disseminated tissue that is capable of influencing the local

10
REVIEW Development (2019) 146, dev156000. doi:10.1242/dev.156000

microenvironment as well as priming the systemic response to Nr4a1-dependent Ly6C(low) monocytes monitor endothelial cells and orchestrate
their disposal. Cell 153, 362-375. doi:10.1016/j.cell.2013.03.010
various types of damage. Acting via the carefully timed secretion of Castellana, D., Paus, R. and Perez-Moreno, M. (2014). Macrophages contribute to
a variety of molecular signals that modulate the activity of tissue- the cyclic activation of adult hair follicle stem cells. PLoS Biol. 12, e1002002.
resident cells, notably stem cells, MPs are involved in a large doi:10.1371/journal.pbio.1002002
number of developmental processes. Importantly, they also play key Cavaillon, J.-M. (2011). The historical milestones in the understanding of leukocyte
biology initiated by Elie Metchnikoff. J. Leukoc. Biol. 90, 413-424. doi:10.1189/jlb.
roles in most events that lead to the reactivation of such processes, 0211094
including during adult regeneration and in the context of disease. In Chan, W. Y., Kohsaka, S. and Rezaie, P. (2007). The origin and cell lineage of
line with these diverse functions, it is becoming apparent that MPs microglia: new concepts. Brain Res. Rev. 53, 344-354. doi:10.1016/j.brainresrev.
are far more heterogeneous and significantly more complex than can 2006.11.002
Chen, C.-C., Wang, L., Plikus, M. V., Jiang, T. X., Murray, P. J., Ramos, R.,
be captured by current classifications. The deployment of emerging Guerrero-Juarez, C. F., Hughes, M. W., Lee, O. K., Shi, S. et al. (2015). Organ-
technologies allowing high-throughput single cell analysis will level quorum sensing directs regeneration in hair stem cell populations. Cell 161,
be required to match phenotype to function and to identify 277-290. doi:10.1016/j.cell.2015.02.016
interventions that may modulate them specifically. Chow, A., Huggins, M., Ahmed, J., Hashimoto, D., Lucas, D., Kunisaki, Y.,
Pinho, S., Leboeuf, M., Noizat, C., Van Rooijen, N. et al. (2013). CD169(+)
macrophages provide a niche promoting erythropoiesis under homeostasis and
Acknowledgements
stress. Nat. Med. 19, 429-436. doi:10.1038/nm.3057
We thank Andrew Hagner for inspiring discussions. Some images of cell types,
Dadgar, S., Wang, Z., Johnston, H., Kesari, A., Nagaraju, K., Chen, Y.-W., Hill,
organs and tissues were obtained from smart.servier.com and modified. D. A., Partridge, T. A., Giri, M., Freishtat, R. J. et al. (2014). Asynchronous
remodeling is a driver of failed regeneration in Duchenne muscular dystrophy.
Competing interests J. Cell Biol. 207, 139-158. doi:10.1083/jcb.201402079
The authors declare no competing or financial interests. Dai, X.-M., Ryan, G. R., Hapel, A. J., Dominguez, M. G., Russell, R. G., Kapp, S.,
Sylvestre, V. and Stanley, E. R. (2002). Targeted disruption of the mouse colony-
Funding stimulating factor 1 receptor gene results in osteopetrosis, mononuclear
This work was supported by the Fondation pour la Recherche Mé dicale (FRM; 40248 phagocyte deficiency, increased primitive progenitor cell frequencies, and
to M.T.); European Molecular Biology Organization (EMBO; ALTF 115-2016 to reproductive defects. Blood 99, 111-120. doi:10.1182/blood.V99.1.111
M.T.); and the Canadian Institutes of Health Research (CIHR-FDN-159908 to F.R.). Dakic, A., Metcalf, D., Di Rago, L., Mifsud, S., Wu, L. and Nutt, S. L. (2005). PU.1
regulates the commitment of adult hematopoietic progenitors and restricts
References granulopoiesis. J. Exp. Med. 201, 1487-1502. doi:10.1084/jem.20050075
Adolfsson, J., Månsson, R., Buza-Vidas, N., Hultquist, A., Liuba, K., Jensen, Daley, J. M., Brancato, S. K., Thomay, A. A., Reichner, J. S. and Albina, J. E.
(2010). The phenotype of murine wound macrophages. J. Leukoc. Biol. 87, 59-67.
C. T., Bryder, D., Yang, L., Borge, O.-J., Thoren, L. A. M. et al. (2005).
doi:10.1189/jlb.0409236
Identification of Flt3+ lympho-myeloid stem cells lacking erythro-megakaryocytic
Davies, L. C., Jenkins, S. J., Allen, J. E. and Taylor, P. R. (2013). Tissue-resident
potential: a revised road map for adult blood lineage commitment. Cell 121,
macrophages. Nat. Immunol. 14, 986-995. doi:10.1038/ni.2705
295-306. doi:10.1016/j.cell.2005.02.013
Dekoter, R. P., Walsh, J. C. and Singh, H. (1998). PU.1 regulates both cytokine-
Ajami, B., Bennett, J. L., Krieger, C., Tetzlaff, W. and Rossi, F. M. V. (2007). Local
dependent proliferation and differentiation of granulocyte/macrophage
self-renewal can sustain CNS microglia maintenance and function throughout
progenitors. EMBO J. 17, 4456-4468. doi:10.1093/emboj/17.15.4456
adult life. Nat. Neurosci. 10, 1538-1543. doi:10.1038/nn2014
Del Rio-Hortega, P. (1932). Microglia. In Cytology and cellular pathology of the
Alliot, F., Godin, I. and Pessac, B. (1999). Microglia derive from progenitors,
nervous system (ed. W. Penfield), pp. 483-534. New York: P.B. Hoeber.
originating from the yolk sac, and which proliferate in the brain. Brain Res. Dev.
Desguerre, I., Arnold, L., Vignaud, A., Cuvellier, S., Yacoub-Youssef, H.,
Brain Res. 117, 145-152. doi:10.1016/S0165-3806(99)00113-3 Gherardi, R. K., Chelly, J., Chretien, F., Mounier, R., Ferry, A. et al. (2012). A
Arnold, L., Henry, A., Poron, F., Baba-Amer, Y., Van Rooijen, N., Plonquet, A.,
new model of experimental fibrosis in hindlimb skeletal muscle of adult mdx
Gherardi, R. K. and Chazaud, B. (2007). Inflammatory monocytes recruited after
mouse mimicking muscular dystrophy. Muscle Nerve 45, 803-814. doi:10.1002/
skeletal muscle injury switch into antiinflammatory macrophages to support
mus.23341
myogenesis. J. Exp. Med. 204, 1057-1069. doi:10.1084/jem.20070075 Desvergne, B. (2008). PPARdelta/beta: the lobbyist switching macrophage
Aurora, A. B., Porrello, E. R., Tan, W., Mahmoud, A. I., Hill, J. A., Bassel-Duby, allegiance in favor of metabolism. Cell Metab. 7, 467-469. doi:10.1016/j.cmet.
R., Sadek, H. A. and Olson, E. N. (2014). Macrophages are required for neonatal 2008.05.002
heart regeneration. J. Clin. Invest. 124, 1382-1392. doi:10.1172/JCI72181 Di Meglio, P., Perera, G. K. and Nestle, F. O. (2011). The multitasking organ: recent
Ayata, P., Badimon, A., Strasburger, H. J., Duff, M. K., Montgomery, S. E., Loh, insights into skin immune function. Immunity 35, 857-869. doi:10.1016/j.immuni.
Y.-H. E., Ebert, A., Pimenova, A. A., Ramirez, B. R., Chan, A. T. et al. (2018). 2011.12.003
Epigenetic regulation of brain region-specific microglia clearance activity. Nat. Dick, S. A., Macklin, J. A., Nejat, S., Momen, A., Clemente-Casares, X.,
Neurosci. 21, 1049-1060. doi:10.1038/s41593-018-0192-3 Althagafi, M. G., Chen, J., Kantores, C., Hosseinzadeh, S., Aronoff, L. et al.
Aziz, A., Soucie, E., Sarrazin, S. and Sieweke, M. H. (2009). MafB/c-Maf (2019). Self-renewing resident cardiac macrophages limit adverse remodeling
deficiency enables self-renewal of differentiated functional macrophages. Science following myocardial infarction. Nat. Immunol. 20, 29-39. doi:10.1038/s41590-
326, 867-871. doi:10.1126/science.1176056 018-0272-2
Banno, T., Gazel, A. and Blumenberg, M. (2004). Effects of tumor necrosis factor-α Doebel, T., Voisin, B. and Nagao, K. (2017). Langerhans cells – the macrophage in
(TNFα) in epidermal keratinocytes revealed using global transcriptional profiling. dendritic cell clothing. Trends Immunol. 38, 817-828. doi:10.1016/j.it.2017.06.008
J. Biol. Chem. 279, 32633-32642. doi:10.1074/jbc.M400642200 Drissen, R., Buza-Vidas, N., Woll, P., Thongjuea, S., Gambardella, A.,
Barton, E. R. (2006). Impact of sarcoglycan complex on mechanical signal Giustacchini, A., Mancini, E., Zriwil, A., Lutteropp, M., Grover, A. et al.
transduction in murine skeletal muscle. Am. J. Physiol. Cell Physiol. 290, (2016). Distinct myeloid progenitor-differentiation pathways identified through
C411-C419. doi:10.1152/ajpcell.00192.2005 single-cell RNA sequencing. Nat. Immunol. 17, 666-676. doi:10.1038/ni.3412
Blé riot, C., Dupuis, T., Jouvion, G., Eberl, G., Disson, O. and Lecuit, M. (2015). Duffield, J. S., Forbes, S. J., Constandinou, C. M., Clay, S., Partolina, M.,
Liver-resident macrophage necroptosis orchestrates type 1 microbicidal Vuthoori, S., Wu, S., Lang, R. and Iredale, J. P. (2005). Selective depletion of
inflammation and type-2-mediated tissue repair during bacterial infection. macrophages reveals distinct, opposing roles during liver injury and repair. J. Clin.
Immunity 42, 145-158. doi:10.1016/j.immuni.2014.12.020 Invest. 115, 56-65. doi:10.1172/JCI200522675
DEVELOPMENT

Boring, L., Gosling, J., Chensue, S. W., Kunkel, S. L., Farese, R. V., Broxmeyer, Dzierzak, E. and Robin, C. (2010). Placenta as a source of hematopoietic stem
H. E. and Charo, I. F. (1997). Impaired monocyte migration and reduced type 1 cells. Trends Mol. Med. 16, 361-367. doi:10.1016/j.molmed.2010.05.005
(Th1) cytokine responses in C-C chemokine receptor 2 knockout mice. J. Clin. Ehses, J. A., Perren, A., Eppler, E., Ribaux, P., Pospisilik, J. A., Maor-Cahn, R.,
Invest. 100, 2552-2561. doi:10.1172/JCI119798 Gueripel, X., Ellingsgaard, H., Schneider, M. K. J., Biollaz, G. et al. (2007).
Boulter, L., Govaere, O., Bird, T. G., Radulescu, S., Ramachandran, P., Increased number of islet-associated macrophages in type 2 diabetes. Diabetes
Pellicoro, A., Ridgway, R. A., Seo, S. S., Spee, B., Van Rooijen, N. et al. 56, 2356-2370. doi:10.2337/db06-1650
(2012). Macrophage-derived Wnt opposes Notch signaling to specify hepatic Elmore, M. R. P., Najafi, A. R., Koike, M. A., Dagher, N. N., Spangenberg, E. E.,
progenitor cell fate in chronic liver disease. Nat. Med. 18, 572-579. doi:10.1038/ Rice, R. A., Kitazawa, M., Matusow, B., Nguyen, H., West, B. L. et al. (2014).
nm.2667 Colony-stimulating factor 1 receptor signaling is necessary for microglia viability,
Carlin, L. M., Stamatiades, E. G., Auffray, C., Hanna, R. N., Glover, L., Vizcay- unmasking a microglia progenitor cell in the adult brain. Neuron 82, 380-397.
Barrena, G., Hedrick, C. C., Cook, H. T., Diebold, S. and Geissmann, F. (2013). doi:10.1016/j.neuron.2014.02.040

11
REVIEW Development (2019) 146, dev156000. doi:10.1242/dev.156000

Engelhardt, B. and Ransohoff, R. M. (2012). Capture, crawl, cross: The T cell code of monocytes to heart macrophages in steady-state and after myocardial
to breach the blood-brain barriers. Trends Immunol. 33, 579-589. doi:10.1016/j.it. infarction. Circ. Res. 115, 284-295. doi:10.1161/CIRCRESAHA.115.303567
2012.07.004 Hellwig, S., Heinrich, A. and Biber, K. (2013). The brain’s best friend: microglial
Epelman, S., Lavine, K. J., Beaudin, A. E., Sojka, D. K., Carrero, J. A., Calderon, neurotoxicity revisited. Front. Cell. Neurosci. 7, 71. doi:10.3389/fncel.2013.00071
B., Brija, T., Gautier, E. L., Ivanov, S., Satpathy, A. T. et al. (2014a). Embryonic Henri, S., Poulin, L. F., Tamoutounour, S., Ardouin, L., Guilliams, M., De Bovis,
and adult-derived resident cardiac macrophages are maintained through distinct B., Devilard, E., Viret, C., Azukizawa, H., Kissenpfennig, A. et al. (2010).
mechanisms at steady state and during inflammation. Immunity 40, 91-104. CD207+ CD103+ dermal dendritic cells cross-present keratinocyte-derived
doi:10.1016/j.immuni.2013.11.019 antigens irrespective of the presence of Langerhans cells. J. Exp. Med. 207,
Epelman, S., Lavine, K. J. and Randolph, G. J. (2014b). Origin and functions of 189-206. doi:10.1084/jem.20091964
tissue macrophages. Immunity 41, 21-35. doi:10.1016/j.immuni.2014.06.013 Herbomel, P., Thisse, B. and Thisse, C. (2001). Zebrafish early macrophages
Erblich, B., Zhu, L., Etgen, A. M., Dobrenis, K. and Pollard, J. W. (2011). Absence colonize cephalic mesenchyme and developing brain, retina, and epidermis
of colony stimulation factor-1 receptor results in loss of microglia, disrupted brain through a M-CSF receptor-dependent invasive process. Dev. Biol. 238, 274-288.
development and olfactory deficits. PloS one 6, e26317. doi:10.1371/journal. doi:10.1006/dbio.2001.0393
pone.0026317 Hilgendorf, I., Gerhardt, L. M. S., Tan, T. C., Winter, C., Holderried, T. A. W.,
Fang, Y., Gong, S.-J., Xu, Y.-H., Hambly, B. D. and Bao, S. (2007). Impaired Chousterman, B. G., Iwamoto, Y., Liao, R., Zirlik, A., Scherer-Crosbie, M.
cutaneous wound healing in granulocyte/ macrophage colony-stimulating factor et al. (2014). Ly-6Chigh monocytes depend on Nr4a1 to balance both
knockout mice. Br. J. Dermatol. 157, 458-465. doi:10.1111/j.1365-2133.2007. inflammatory and reparative phases in the infarcted myocardium. Circ. Res.
07979.x 114, 1611-1622. doi:10.1161/CIRCRESAHA.114.303204
Fantin, A., Vieira, J. M., Gestri, G., Denti, L., Schwarz, Q., Prykhozhij, S., Peri, F., Hoeffel, G. and Ginhoux, F. (2015). Ontogeny of tissue-resident macrophages.
Wilson, S. W. and Ruhrberg, C. (2010). Tissue macrophages act as cellular Front. Immunol. 6, 486. doi:10.3389/fimmu.2015.00486
chaperones for vascular anastomosis downstream of VEGF-mediated endothelial Hoeffel, G., Wang, Y., Greter, M., See, P., Teo, P., Malleret, B., Leboeuf, M., Low,
tip cell induction. Blood 116, 829-840. doi:10.1182/blood-2009-12-257832 D., Oller, G., Almeida, F. et al. (2012). Adult Langerhans cells derive
Fischer, K., Ruiz, H. H., Jhun, K., Finan, B., Oberlin, D. J., Van Der Heide, V., predominantly from embryonic fetal liver monocytes with a minor contribution of
Kalinovich, A. V., Petrovic, N., Wolf, Y., Clemmensen, C. et al. (2017). yolk sac-derived macrophages. J. Exp. Med. 209, 1167-1181. doi:10.1084/jem.
Alternatively activated macrophages do not synthesize catecholamines or 20120340
contribute to adipose tissue adaptive thermogenesis. Nat. Med. 23, 623-630. Huang, Y., Xu, Z., Xiong, S., Sun, F., Qin, G., Hu, G., Wang, J., Zhao, L., Liang, Y.-
doi:10.1038/nm.4316 X., Wu, T. et al. (2018). Repopulated microglia are solely derived from the
Frangogiannis, N. G. (2015). Inflammation in cardiac injury, repair and regeneration. proliferation of residual microglia after acute depletion. Nat. Neurosci. 21,
Curr. Opin. Cardiol. 30, 240-245. doi:10.1097/HCO.0000000000000158 530-540. doi:10.1038/s41593-018-0090-8
Freire-De-Lima, C. G., Nascimento, D. O., Soares, M. B. P., Bozza, P. T., Castro- Hulsmans, M., Clauss, S., Xiao, L., Aguirre, A. D., King, K. R., Hanley, A.,
Faria-Neto, H. C., De Mello, F. G., Dosreis, G. A. and Lopes, M. F. (2000). Hucker, W. J., Wü lfers, E. M., Seemann, G., Courties, G. et al. (2017).
Uptake of apoptotic cells drives the growth of a pathogenic trypanosome in Macrophages facilitate electrical conduction in the heart. Cell 169, 510-522.e20.
macrophages. Nature 403, 199-203. doi:10.1038/35003208 doi:10.1016/j.cell.2017.03.050
Gammella, E., Buratti, P., Cairo, G. and Recalcati, S. (2014). Macrophages: Jackaman, C., Tomay, F., Duong, L., Abdol Razak, N. B., Pixley, F. J., Metharom,
central regulators of iron balance. Metallomics 6, 1336-1345. doi:10.1039/
P. and Nelson, D. J. (2017). Aging and cancer: the role of macrophages and
C4MT00104D
neutrophils. Ageing Res. Rev. 36, 105-116. doi:10.1016/j.arr.2017.03.008
Gautier, E. L. and Yvan-Charvet, L. (2014). Understanding macrophage diversity
Jakubzick, C., Gautier, E. L., Gibbings, S. L., Sojka, D. K., Schlitzer, A.,
at the ontogenic and transcriptomic levels. Immunol. Rev. 262, 85-95. doi:10.
Johnson, T. E., Ivanov, S., Duan, Q., Bala, S., Condon, T. et al. (2013). Minimal
1111/imr.12231
differentiation of classical monocytes as they survey steady-state tissues and
Geissmann, F., Jung, S. and Littman, D. R. (2003). Blood monocytes consist of
transport antigen to lymph nodes. Immunity 39, 599-610. doi:10.1016/j.immuni.
two principal subsets with distinct migratory properties. Immunity 19, 71-82.
2013.08.007
doi:10.1016/S1074-7613(03)00174-2
Jenkins, S. J. and Hume, D. A. (2014). Homeostasis in the mononuclear
Ginhoux, F. and Guilliams, M. (2016). Tissue-resident macrophage ontogeny and
phagocyte system. Trends Immunol. 35, 358-367. doi:10.1016/j.it.2014.06.006
homeostasis. Immunity 44, 439-449. doi:10.1016/j.immuni.2016.02.024
Johann, A. M., Von Knethen, A., Lindemann, D. and Brü ne, B. (2006).
Ginhoux, F., Greter, M., Leboeuf, M., Nandi, S., See, P., Gokhan, S., Mehler,
Recognition of apoptotic cells by macrophages activates the peroxisome
M. F., Conway, S. J., Ng, L. G., Stanley, E. R. et al. (2010). Fate mapping
proliferator-activated receptor-gamma and attenuates the oxidative burst. Cell
analysis reveals that adult microglia derive from primitive macrophages. Science
Death Differ. 13, 1533-1540. doi:10.1038/sj.cdd.4401832
330, 841-845. doi:10.1126/science.1194637
Juban, G., Saclier, M., Yacoub-Youssef, H., Kernou, A., Arnold, L., Boisson, C.,
Gomez Perdiguero, E., Klapproth, K., Schulz, C., Busch, K., Azzoni, E., Crozet,
L., Garner, H., Trouillet, C., De Bruijn, M. F., Geissmann, F. et al. (2015). Ben Larbi, S., Magnan, M., Cuvellier, S., Thé ret, M. et al. (2018). AMPK
Tissue-resident macrophages originate from yolk-sac-derived erythro-myeloid activation regulates LTBP4-dependent TGF-beta1 secretion by pro-inflammatory
progenitors. Nature 518, 547-551. doi:10.1038/nature13989 macrophages and controls fibrosis in Duchenne muscular dystrophy. Cell Rep. 25,
Graeber, M. B. and Streit, W. J. (2010). Microglia: biology and pathology. Acta 2163-2176.e6. doi:10.1016/j.celrep.2018.10.077
Neuropathol. 119, 89-105. doi:10.1007/s00401-009-0622-0 Jung, S., Aliberti, J., Graemmel, P., Sunshine, M. J., Kreutzberg, G. W., Sher, A.
Grose, R. and Werner, S. (2004). Wound-healing studies in transgenic and and Littman, D. R. (2000). Analysis of fractalkine receptor CX(3)CR1 function by
knockout mice. Appl. Biochem. Biotechnol. B Mol. Biotechnol. 28, 147-166. targeted deletion and green fluorescent protein reporter gene insertion. Mol. Cell.
doi:10.1385/MB:28:2:147 Biol. 20, 4106-4114. doi:10.1128/MCB.20.11.4106-4114.2000
Guilliams, M. and Scott, C. L. (2017). Does niche competition determine the origin Kang, K., Reilly, S. M., Karabacak, V., Gangl, M. R., Fitzgerald, K., Hatano, B.
of tissue-resident macrophages? Nat. Rev. Immunol. 17, 451-460. doi:10.1038/ and Lee, C.-H. (2008). Adipocyte-derived Th2 cytokines and myeloid PPARdelta
nri.2017.42 regulate macrophage polarization and insulin sensitivity. Cell Metab. 7, 485-495.
Guilliams, M., De Kleer, I., Henri, S., Post, S., Vanhoutte, L., De Prijck, S., doi:10.1016/j.cmet.2008.04.002
Deswarte, K., Malissen, B., Hammad, H. and Lambrecht, B. N. (2013). Alveolar Khan, T. N., Wong, E. B., Soni, C. and Rahman, Z. S. M. (2013). Prolonged
macrophages develop from fetal monocytes that differentiate into long-lived cells apoptotic cell accumulation in germinal centers of Mer-deficient mice causes
in the first week of life via GM-CSF. J. Exp. Med. 210, 1977-1992. doi:10.1084/ elevated B cell and CD4+ Th cell responses leading to autoantibody production.
jem.20131199 J. Immunol. 190, 1433-1446. doi:10.4049/jimmunol.1200824
Gustafsson, K. and Welsh, M. (2016). Maintenance of hematopoietic stem cell Kierdorf, K. and Dionne, M. S. (2016). The software and hardware of
dormancy: yet another role for the macrophage. Stem Cell Investig. 3, 46. doi:10. macrophages: a diversity of options. Dev. Cell 38, 122-125. doi:10.1016/j.
21037/sci.2016.08.10 devcel.2016.07.008
Hammond, T. R., Robinton, D. and Stevens, B. (2018). Microglia and the brain: Kierdorf, K., Prinz, M., Geissmann, F. and Gomez Perdiguero, E. (2015).
DEVELOPMENT

complementary partners in development and disease. Annu. Rev. Cell Dev. Biol. Development and function of tissue resident macrophages in mice. Semin.
34, 523-544. doi:10.1146/annurev-cellbio-100616-060509 Immunol. 27, 369-378. doi:10.1016/j.smim.2016.03.017
Hancock, G. E., Kaplan, G. and Cohn, Z. (1988). Keratinocyte growth regulation by Kohyama, M., Ise, W., Edelson, B. T., Wilker, P. R., Hildner, K., Mejia, C., Frazier,
the products of immune cells. J. Exp. Med. 168, 1395-1402. doi:10.1084/jem.168. W. A., Murphy, T. L. and Murphy, K. M. (2009). Role for Spi-C in the development
4.1395 of red pulp macrophages and splenic iron homeostasis. Nature 457, 318-321.
Hashimoto, D., Chow, A., Noizat, C., Teo, P., Beasley, M. B., Leboeuf, M., Becker, doi:10.1038/nature07472
C. D., See, P., Price, J., Lucas, D. et al. (2013). Tissue-resident macrophages self- Latroche, C., Weiss-Gayet, M., Muller, L., Gitiaux, C., Leblanc, P., Liot, S., Ben-
maintain locally throughout adult life with minimal contribution from circulating Larbi, S., Abou-Khalil, R., Verger, N., Bardot, P. et al. (2017). Coupling between
monocytes. Immunity 38, 792-804. doi:10.1016/j.immuni.2013.04.004 myogenesis and angiogenesis during skeletal muscle regeneration is stimulated
Heidt, T., Courties, G., Dutta, P., Sager, H. B., Sebas, M., Iwamoto, Y., Sun, Y., Da by restorative macrophages. Stem Cell Rep. 9, 2018-2033. doi:10.1016/j.stemcr.
Silva, N., Panizzi, P., Van Der Laan, A. M. et al. (2014). Differential contribution 2017.10.027

12
REVIEW Development (2019) 146, dev156000. doi:10.1242/dev.156000

Lehrman, E. K., Wilton, D. K., Litvina, E. Y., Welsh, C. A., Chang, S. T., Frouin, obesity-induced insulin resistance. Cell Metab. 7, 496-507. doi:10.1016/j.cmet.
A., Walker, A. J., Heller, M. D., Umemori, H., Chen, C. et al. (2018). CD47 2008.04.003
protects synapses from excess microglia-mediated pruning during development. Orkin, S. H. and Zon, L. I. (2008). Hematopoiesis: an evolving paradigm for stem
Neuron 100, 120-134.e6. doi:10.1016/j.neuron.2018.09.017 cell biology. Cell 132, 631-644. doi:10.1016/j.cell.2008.01.025
Lemos, D. R., Babaeijandaghi, F., Low, M., Chang, C.-K., Lee, S. T., Fiore, D., Osaka, N., Takahashi, T., Murakami, S., Matsuzawa, A., Noguchi, T., Fujiwara,
Zhang, R.-H., Natarajan, A., Nedospasov, S. A. and Rossi, F. M. V. (2015). T., Aburatani, H., Moriyama, K., Takeda, K. and Ichijo, H. (2007). ASK1-
Nilotinib reduces muscle fibrosis in chronic muscle injury by promoting TNF- dependent recruitment and activation of macrophages induce hair growth in skin
mediated apoptosis of fibro/adipogenic progenitors. Nat. Med. 21, 786-794. wounds. J. Cell Biol. 176, 903-909. doi:10.1083/jcb.200611015
doi:10.1038/nm.3869 Palis, J. and Yoder, M. C. (2001). Yolk-sac hematopoiesis: the first blood cells of
Ley, K. (2017). M1 means kill; M2 means heal. J. Immunol. 199, 2191-2193. doi:10. mouse and man. Exp. Hematol. 29, 927-936. doi:10.1016/S0301-
4049/jimmunol.1701135 472X(01)00669-5
Linehan, E. and Fitzgerald, D. (2015). Ageing and the immune system: focus on Paolicelli, R. C., Bolasco, G., Pagani, F., Maggi, L., Scianni, M., Panzanelli, P.,
macrophages. Eur. J. Microbiol. Immunol. 5, 14-24. doi:10.1556/EuJMI-D-14- Giustetto, M., Ferreira, T. A., Guiducci, E., Dumas, L. et al. (2011). Synaptic
00035 pruning by microglia is necessary for normal brain development. Science 333,
Lu, B., Rutledge, B. J., Gu, L., Fiorillo, J., Lukacs, N. W., Kunkel, S. L., North, R., 1456-1458. doi:10.1126/science.1202529
Gerard, C. and Rollins, B. J. (1998). Abnormalities in monocyte recruitment and Paus, R. (1998). Principles of hair cycle control. J. Dermatol. 25, 793-802. doi:10.
cytokine expression in monocyte chemoattractant protein 1-deficient mice. J. Exp. 1111/j.1346-8138.1998.tb02507.x
Med. 187, 601-608. doi:10.1084/jem.187.4.601 Petrof, B. J., Shrager, J. B., Stedman, H. H., Kelly, A. M. and Sweeney, H. L.
Lucas, T., Waisman, A., Ranjan, R., Roes, J., Krieg, T., Muller, W., Roers, A. and (1993). Dystrophin protects the sarcolemma from stresses developed during
Eming, S. A. (2010). Differential roles of macrophages in diverse phases of skin muscle contraction. Proc. Natl. Acad. Sci. USA 90, 3710-3714. doi:10.1073/pnas.
repair. J. Immunol. 184, 3964-3977. doi:10.4049/jimmunol.0903356 90.8.3710
Lumeng, C. N., Bodzin, J. L. and Saltiel, A. R. (2007). Obesity induces a Pinto, A. R., Godwin, J. W. and Rosenthal, N. A. (2014). Macrophages in cardiac
phenotypic switch in adipose tissue macrophage polarization. J. Clin. Invest. 117, homeostasis, injury responses and progenitor cell mobilisation. Stem Cell Res.
175-184. doi:10.1172/JCI29881 13, 705-714. doi:10.1016/j.scr.2014.06.004
Machiels, B., Dourcy, M., Xiao, X., Javaux, J., Mesnil, C., Sabatel, C., Desmecht, Porrello, E. R., Mahmoud, A. I., Simpson, E., Hill, J. A., Richardson, J. A., Olson,
D., Lallemand, F., Martinive, P., Hammad, H. et al. (2017). A gammaherpesvirus E. N. and Sadek, H. A. (2011). Transient regenerative potential of the neonatal
provides protection against allergic asthma by inducing the replacement of mouse heart. Science 331, 1078-1080. doi:10.1126/science.1200708
resident alveolar macrophages with regulatory monocytes. Nat. Immunol. 18, Rahmani, W., Liu, Y., Rosin, N. L., Kline, A., Raharjo, E., Yoon, J., Stratton, J. A.,
1310-1320. doi:10.1038/ni.3857 Sinha, S. and Biernaskie, J. (2018). Macrophages promote wound-induced hair
Martinez, F. O., Sica, A., Mantovani, A. and Locati, M. (2008). Macrophage follicle regeneration in a CX3CR1- and TGF-beta1-dependent manner. J. Invest.
activation and polarization. Front. Biosci. 13, 453-461. doi:10.2741/2692 Dermatol. 138, 2111-2122. doi:10.1016/j.jid.2018.04.010
Mass, E., Ballesteros, I., Farlik, M., Halbritter, F., Gunther, P., Crozet, L., Ramachandran, P., Pellicoro, A., Vernon, M. A., Boulter, L., Aucott, R. L., Ali, A.,
Jacome-Galarza, C. E., Handler, K., Klughammer, J., Kobayashi, Y. et al. Hartland, S. N., Snowdon, V. K., Cappon, A., Gordon-Walker, T. T. et al.
(2016). Specification of tissue-resident macrophages during organogenesis.
(2012). Differential Ly-6C expression identifies the recruited macrophage
Science 353, aaf4238. doi:10.1126/science.aaf4238
phenotype, which orchestrates the regression of murine liver fibrosis. Proc. Natl.
Mckercher, S. R., Torbett, B. E., Anderson, K. L., Henkel, G. W., Vestal, D. J.,
Acad. Sci. USA 109, E3186-E3195. doi:10.1073/pnas.1119964109
Baribault, H., Klemsz, M., Feeney, A. J., Wu, G. E., Paige, C. J. et al. (1996).
Ruffell, D., Mourkioti, F., Gambardella, A., Kirstetter, P., Lopez, R. G.,
Targeted disruption of the PU.1 gene results in multiple hematopoietic
Rosenthal, N. and Nerlov, C. (2009). A CREB-C/EBPbeta cascade induces
abnormalities. EMBO J. 15, 5647-5658. doi:10.1002/j.1460-2075.1996.tb00949.x
M2 macrophage-specific gene expression and promotes muscle injury repair.
Merad, M., Manz, M. G., Karsunky, H., Wagers, A., Peters, W., Charo, I.,
Proc. Natl. Acad. Sci. USA 106, 17475-17480. doi:10.1073/pnas.0908641106
Weissman, I. L., Cyster, J. G. and Engleman, E. G. (2002). Langerhans cells
Saclier, M., Yacoub-Youssef, H., Mackey, A. L., Arnold, L., Ardjoune, H.,
renew in the skin throughout life under steady-state conditions. Nat. Immunol. 3,
Magnan, M., Sailhan, F., Chelly, J., Pavlath, G. K., Mounier, R. et al. (2013a).
1135-1141. doi:10.1038/ni852
Differentially activated macrophages orchestrate myogenic precursor cell fate
Mills, C. D., Kincaid, K., Alt, J. M., Heilman, M. J. and Hill, A. M. (2000). M-1/M-2
during human skeletal muscle regeneration. Stem Cells 31, 384-396. doi:10.1002/
macrophages and the Th1/Th2 paradigm. J. Immunol. 164, 6166-6173. doi:10.
stem.1288
1177/000271622310900111
Saclier, M., Cuvellier, S., Magnan, M., Mounier, R. and Chazaud, B. (2013b).
Mirza, R., Dipietro, L. A. and Koh, T. J. (2009). Selective and specific macrophage
ablation is detrimental to wound healing in mice. Am. J. Pathol. 175, 2454-2462. Monocyte/macrophage interactions with myogenic precursor cells during skeletal
doi:10.2353/ajpath.2009.090248 muscle regeneration. FEBS J. 280, 4118-4130. doi:10.1111/febs.12166
Mitchell, C., Couton, D., Couty, J.-P., Anson, M., Crain, A.-M., Bizet, V., Ré nia, L., Sager, H. B., Hulsmans, M., Lavine, K. J., Moreira, M. B., Heidt, T., Courties, G.,
Pol, S., Mallet, V. and Gilgenkrantz, H. (2009). Dual role of CCR2 in the Sun, Y., Iwamoto, Y., Tricot, B., Khan, O. F. et al. (2016). Proliferation and
constitution and the resolution of liver fibrosis in mice. Am. J. Pathol. 174, recruitment contribute to myocardial macrophage expansion in chronic heart
1766-1775. doi:10.2353/ajpath.2009.080632 failure. Circ. Res. 119, 853-864. doi:10.1161/CIRCRESAHA.116.309001
Mojumdar, K., Liang, F., Giordano, C., Lemaire, C., Danialou, G., Okazaki, T., Salter, M. W. and Beggs, S. (2014). Sublime microglia: expanding roles for the
Bourdon, J., Rafei, M., Galipeau, J., Divangahi, M. et al. (2014). Inflammatory guardians of the CNS. Cell 158, 15-24. doi:10.1016/j.cell.2014.06.008
monocytes promote progression of Duchenne muscular dystrophy and can be Sarrazin, S. and Sieweke, M. H. (2016). Eosinophils and mast cells: a lineage apart.
therapeutically targeted via CCR2. EMBO Mol. Med. 6, 1476-1492. doi:10.15252/ Nat. Immunol. 17, 609-611. doi:10.1038/ni.3446
emmm.201403967 Schulz, C., Perdiguero, E. G., Chorro, L., Szabo-Rogers, H., Cagnard, N.,
Molawi, K., Wolf, Y., Kandalla, P. K., Favret, J., Hagemeyer, N., Frenzel, K., Kierdorf, K., Prinz, M., Wu, B., Jacobsen, S. E. W., Pollard, J. W. et al. (2012). A
Pinto, A. R., Klapproth, K., Henri, S., Malissen, B. et al. (2014). Progressive lineage of myeloid cells independent of Myb and hematopoietic stem cells.
replacement of embryo-derived cardiac macrophages with age. J. Exp. Med. 211, Science 336, 86-90. doi:10.1126/science.1219179
2151-2158. doi:10.1084/jem.20140639 Shirakata, Y. (2005). Heparin-binding EGF-like growth factor accelerates
Mounier, R., Thé ret, M., Arnold, L., Cuvellier, S., Bultot, L., Gö ransson, O., keratinocyte migration and skin wound healing. J. Cell Sci. 118, 2363-2370.
Sanz, N., Ferry, A., Sakamoto, K., Foretz, M. et al. (2013). AMPKalpha1 doi:10.1242/jcs.02346
regulates macrophage skewing at the time of resolution of inflammation during Sica, A., Erreni, M., Allavena, P. and Porta, C. (2015). Macrophage polarization in
skeletal muscle regeneration. Cell Metab. 18, 251-264. doi:10.1016/j.cmet.2013. pathology. Cell. Mol. Life Sci. 72, 4111-4126. doi:10.1007/s00018-015-1995-y
06.017 Sieweke, M. H. and Allen, J. E. (2013). Beyond stem cells: self-renewal of
Nguyen, K. D., Qiu, Y., Cui, X., Goh, Y. P. S., Mwangi, J., David, T., Mukundan, L., differentiated macrophages. Science 342, 1242974. doi:10.1126/science.
Brombacher, F., Locksley, R. M. and Chawla, A. (2011). Alternatively activated 1242974
DEVELOPMENT

macrophages produce catecholamines to sustain adaptive thermogenesis. Sonnet, C., Lafuste, P., Arnold, L., Brigitte, M., Poron, F., Authier, F. J., Chré tien,
Nature 480, 104-108. doi:10.1038/nature10653 F., Gherardi, R. K. and Chazaud, B. (2006). Human macrophages rescue
Nguyen-Lefebvre, A. T. and Horuzsko, A. (2015). Kupffer cell metabolism and myoblasts and myotubes from apoptosis through a set of adhesion molecular
function. J. Enzymol. Metab. 1, 101. systems. J. Cell Sci. 119, 2497-2507. doi:10.1242/jcs.02988
Notta, F., Zandi, S., Takayama, N., Dobson, S., Gan, O. I., Wilson, G., Kaufmann, Sun, K., Kusminski, C. M. and Scherer, P. E. (2011). Adipose tissue remodeling
K. B., Mcleod, J., Laurenti, E., Dunant, C. F. et al. (2016). Distinct routes of and obesity. J. Clin. Invest. 121, 2094-2101. doi:10.1172/JCI45887
lineage development reshape the human blood hierarchy across ontogeny. Tamoutounour, S., Guilliams, M., Montanana Sanchis, F., Liu, H., Terhorst, D.,
Science 351, aab2116. doi:10.1126/science.aab2116 Malosse, C., Pollet, E., Ardouin, L., Luche, H., Sanchez, C. et al. (2013).
Odegaard, J. I., Ricardo-Gonzalez, R. R., Red Eagle, A., Vats, D., Morel, C. R., Origins and functional specialization of macrophages and of conventional and
Goforth, M. H., Subramanian, V., Mukundan, L., Ferrante, A. W. and Chawla, monocyte-derived dendritic cells in mouse skin. Immunity 39, 925-938. doi:10.
A. (2008). Alternative M2 activation of Kupffer cells by PPARdelta ameliorates 1016/j.immuni.2013.10.004

13
REVIEW Development (2019) 146, dev156000. doi:10.1242/dev.156000

Theurl, I., Hilgendorf, I., Nairz, M., Tymoszuk, P., Haschka, D., Asshoff, M., He, Wang, M., Zhang, G., Wang, Y., Liu, T., Zhang, Y., An, Y. and Li, Y. (2015).
S., Gerhardt, L. M. S., Holderried, T. A. W., Seifert, M. et al. (2016). On-demand Crosstalk of mesenchymal stem cells and macrophages promotes cardiac muscle
erythrocyte disposal and iron recycling requires transient macrophages in the repair. Int. J. Biochem. Cell Biol. 58, 53-61. doi:10.1016/j.biocel.2014.11.003
liver. Nat. Med. 22, 945-951. doi:10.1038/nm.4146 Wang, X., Zhao, W., Ransohoff, R. M. and Zhou, L. (2016). Identification and
Tidball, J. G. (2017). Regulation of muscle growth and regeneration by the immune function of fibrocytes in skeletal muscle injury repair and muscular dystrophy.
system. Nat. Rev. Immunol. 17, 165-178. doi:10.1038/nri.2016.150 J. Immunol. 197, 4750-4761. doi:10.4049/jimmunol.1601308
Tidball, J. G., Dorshkind, K. and Wehling-Henricks, M. (2014). Shared signaling Wang, X., Chen, H., Tian, R., Zhang, Y., Drutskaya, M. S., Wang, C., Ge, J., Fan,
systems in myeloid cell-mediated muscle regeneration. Development 141, Z., Kong, D., Wang, X. et al. (2017). Macrophages induce AKT/β-catenin-
1184-1196. doi:10.1242/dev.098285
dependent Lgr5+ stem cell activation and hair follicle regeneration through TNF.
Tonkin, J., Temmerman, L., Sampson, R. D., Gallego-Colon, E., Barberi, L.,
Nat. Commun. 8, 14091. doi:10.1038/ncomms14091
Bilbao, D., Schneider, M. D., Musarò, A. and Rosenthal, N. (2015). Monocyte/
Warren, G. L., Hulderman, T., Mishra, D., Gao, X., Millecchia, L., O’Farrell, L.,
macrophage-derived IGF-1 orchestrates murine skeletal muscle regeneration and
Kuziel, W. A. and Simeonova, P. P. (2005). Chemokine receptor CCR2
modulates autocrine polarization. Mol. Ther. 23, 1189-1200. doi:10.1038/mt.2015.66
Tremblay, M.-E., Lowery, R. L. and Majewska, A. K. (2010). Microglial interactions involvement in skeletal muscle regeneration. FASEB J. 19, 413-415. doi:10.1096/
with synapses are modulated by visual experience. PLoS Biol. 8, e1000527. fj.04-2421fje
doi:10.1371/journal.pbio.1000527 Weisberg, S. P., Mccann, D., Desai, M., Rosenbaum, M., Leibel, R. L. and
Van Amerongen, M. J., Harmsen, M. C., Van Rooijen, N., Petersen, A. H. and Ferrante, A. W. (2003). Obesity is associated with macrophage accumulation in
Van Luyn, M. J. A. (2007). Macrophage depletion impairs wound healing and adipose tissue. J. Clin. Invest. 112, 1796-1808. doi:10.1172/JCI200319246
increases left ventricular remodeling after myocardial injury in mice. Am. J. Pathol. Winkler, I. G., Sims, N. A., Pettit, A. R., Barbier, V., Nowlan, B., Helwani, F.,
170, 818-829. doi:10.2353/ajpath.2007.060547 Poulton, I. J., Van Rooijen, N., Alexander, K. A., Raggatt, L. J. et al. (2010).
Van Furth, R. and Cohn, Z. A. (1968). The origin and kinetics of mononuclear Bone marrow macrophages maintain hematopoietic stem cell (HSC) niches and
phagocytes. J. Exp. Med. 128, 415-435. doi:10.1084/jem.128.3.415 their depletion mobilizes HSCs. Blood 116, 4815-4828. doi:10.1182/blood-2009-
Varga, T., Mounier, R., Gogolak, P., Poliska, S., Chazaud, B. and Nagy, L. 11-253534
(2013). Tissue LyC6- macrophages are generated in the absence of circulating Wynn, T. A. and Vannella, K. M. (2016). Macrophages in tissue repair, regeneration,
LyC6- monocytes and Nur77 in a model of muscle regeneration. J. Immunol. 191, and fibrosis. Immunity 44, 450-462. doi:10.1016/j.immuni.2016.02.015
5695-5701. doi:10.4049/jimmunol.1301445 Yamamoto, R., Wilkinson, A. C. and Nakauchi, H. (2018). Changing concepts in
Varga, T., Mounier, R., Horvath, A., Cuvellier, S., Dumont, F., Poliska, S., hematopoietic stem cells. Science 362, 895-896. doi:10.1126/science.aat7873
Ardjoune, H., Juban, G., Nagy, L. and Chazaud, B. (2016a). Highly dynamic Yona, S., Kim, K.-W., Wolf, Y., Mildner, A., Varol, D., Breker, M., Strauss-Ayali,
transcriptional signature of distinct macrophage subsets during sterile D., Viukov, S., Guilliams, M., Misharin, A. et al. (2013). Fate mapping reveals
inflammation, resolution, and tissue repair. J. Immunol. 196, 4771-4782. doi:10.
origins and dynamics of monocytes and tissue macrophages under homeostasis.
4049/jimmunol.1502490
Immunity 38, 79-91. doi:10.1016/j.immuni.2012.12.001
Varga, T., Mounier, R., Patsalos, A., Gogolá k, P., Peloquin, M., Horvath, A., Pap,
Zheng, C., Yang, Q., Xu, C., Shou, P., Cao, J., Jiang, M., Chen, Q., Cao, G., Han,
A., Daniel, B., Nagy, G., Pintye, E. et al. (2016b). Macrophage PPARgamma, a
Y., Li, F. et al. (2015). CD11b regulates obesity-induced insulin resistance via
lipid activated transcription factor controls the growth factor GDF3 and skeletal
muscle regeneration. Immunity 45, 1038-1051. doi:10.1016/j.immuni.2016.10.016 limiting alternative activation and proliferation of adipose tissue macrophages.
Varol, C., Vallon-Eberhard, A., Elinav, E., Aychek, T., Shapira, Y., Luche, H., Proc. Natl. Acad. Sci. USA 112, E7239-E7248. doi:10.1073/pnas.1500396113
Fehling, H. J., Hardt, W.-D., Shakhar, G. and Jung, S. (2009). Intestinal lamina Zordan, P., Rigamonti, E., Freudenberg, K., Conti, V., Azzoni, E., Rovere-
propria dendritic cell subsets have different origin and functions. Immunity 31, Querini, P. and Brunelli, S. (2014). Macrophages commit postnatal endothelium-
502-512. doi:10.1016/j.immuni.2009.06.025 derived progenitors to angiogenesis and restrict endothelial to mesenchymal
Von Kupffer, K. W. (1876). Ueber Sternzellen der Leber. Arch Mikroskop Anat. 12, transition during muscle regeneration. Cell Death Dis. 5, e1031. doi:10.1038/
353-358. doi:10.1007/BF02933897 cddis.2013.558

DEVELOPMENT

14

S-ar putea să vă placă și