Grupo 1

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

REVIEW MACROPHAGES

The development and maintenance of


resident macrophages
Elisa Gomez Perdiguero1 & Frederic Geissmann2

The molecular and cellular mechanisms that underlie the many roles of macrophages in health and disease states in vivo remain
poorly understood. The purpose of this Review is to present and discuss current knowledge on the developmental biology of
macrophages, as it underlies the concept of a layered myeloid system composed of ‘resident’ macrophages that originate mainly
© 2016 Nature America, Inc. All rights reserved.

from progenitor cells generated in the yolk sac and of ‘passenger’ or ‘transitory’ myeloid cells that originate and renew from bone
marrow hematopoietic stem cells, and to provide a framework for investigating the functions of macrophages in vivo.

The current ‘mononuclear phagocyte system’ model1 has guided many include spleen red-pulp macrophages, lung alveolar macrophages, epider-
investigators studying the functions of macrophages over the past 30 years. mal Langerhans cells, brain microglia, liver Kupffer cells, large peritoneal
It postulates that tissue macrophages are maintained from a constant sup- macrophages, and F4/80bright pancreatic, kidney and cardiac macrophages.
ply of bone marrow–derived circulating blood monocytes that extrava- Many tissue-resident macrophages are long lived in mice and can prolifer-
sate into tissues2. This model was initially based on the observation that ate within their tissue of residence, a mechanism involved in their mainte-
blood leukocytes recruited into the inflamed peritoneum differentiate nance in adults20–27. Nevertheless, bone marrow–derived progenitor cells
into macrophages. Subsequent experiments of transplantation into irra- also contribute to subsets that reside in the lamina propria, spleen, brain,
diated recipients demonstrated that bone marrow hematopoietic stem cells skin, heart, liver and kidneys in a proportion that varies by tissue, mouse
(HSCs) and HSC-derived myeloid progenitors can indeed give rise to tissue age and pathological processes12–18,28,29.
macrophages or dendritic cells in mice and humans3–7. Therefore, investi- Therefore, the purpose of this Review is to present and discuss current
gations designed to study the molecular and cellular basis of macrophage knowledge of the developmental biology of resident macrophages, as it
phenotypic diversity and function have focused largely on the activation underlies the concept of a layered myeloid system composed of tissue-
or polarization of monocyte-derived macrophages by external cues, such resident macrophages distinct from passenger macrophages and myeloid
as cytokines and microbe-associated signals8–10. Nevertheless, it remains cells and provides a new framework and experimental tools with which to
unclear whether the polarization stages of monocytes are stable or represent characterize the functions of macrophages in vivo, in health and in disease
npg

transient states of activation, whether they account for the phenotypic and settings.
functional heterogeneity of macrophages in vivo in health or disease states,
and how many such polarized states exist10,11. The original concept of ‘primitive macrophages’
A new model of macrophage development is emerging that is based on During ontogeny, macrophages are found in the mouse yolk sac and the
many independent observations that two distinct populations of macro- embryo before the initiation of HSC-derived hematopoiesis and before
phages, which can be distinguished by their progenitors, developmental monocytes can be detected in the fetal blood. These ‘primitive macro-
history, turnover and mechanisms of maintenance, coexist within the tis- phages’ were described in a series of seminal works20,30,31 that reported
sues of an adult mouse. The macrophage system of a mouse can now be the presence of large cells with macrophage morphology in blood islands
described as a ‘layered’ system in which ‘resident’ macrophages that develop of the yolk sac at embryonic day 9 (E9) and in the fetal liver at E10. These
in embryos independently of HSCs12 persist in adult tissues independently were described as ‘immature’ because of their lack of detectable phagocytic
of adult HSCs12–18 and coexist with ‘passenger’ leukocytes, such as mono- activity and weak immunoreactivity for the macrophage marker F4/80,
cytes and classic dendritic cells, that originate and renew from bone mar- as detected by electron microscopy20,30. In addition to reporting those
row HSCs and myeloid progenitors4,6,19. Tissue-resident macrophages ‘immature macrophages’, these early studies also reported the existence
of ‘fetal macrophages’ from E10 to E17 as slightly smaller cells with pseu-
1Macrophages and Endothelial Cells group, Department of Developmental dopodia, phagocytic activity and frequent mitotic figures31. At E17, the
and Stem Cell Biology, CNRS URA 2578, Institut Pasteur, Paris, France. ‘fetal macrophages’ were reported as more differentiated, as they acquired
2Immunology Program, Memorial Sloan Kettering Cancer Center, New York, ultrastructural features similar to those of adult tissue macrophages30,32.
New York, USA. Correspondence should be addressed to F.G. (geissmaf@ These early studies noted that the remaining difference between fetal tissue
mskcc.org). macrophages and adult tissue macrophages was the presence of numer-
ous mitotic figures in fetal macrophages throughout development. It was
Received 24 September; accepted 3 November; published online also noted that poorly differentiated myeloid cells were detected in the
17 December 2015; doi:10.1038/ni.3341 blood vessels of the yolk sac at E10 (ref. 20), and immature stages of the

2 VOLUME 17 NUMBER 1 JANUARY 2016 NATURE IMMUNOLOGY


REVIEW

monocyte and neutrophil differentiation series could be detected from E12 region) in which they are generated, expand and differentiate, their differ-
onward, and finally true neutrophils and monocytes, identified on the basis entiation potential and their dependence on transcription factors (Table 1
of ultrastructural features and endogenous peroxidase-cytochemistry, were and Fig. 1). Here we will focus on the myeloid lineages; erythroid lineages
present in the fetal liver at E15 and E16, respectively30. These researchers have already been reviewed37.
therefore proposed that fetal macrophages develop without a monocytic
intermediate and in parallel with the monocyte and granulocyte lineage. Primitive hematopoiesis as a source of macrophages in embryos
In subsequent literature, that early work was sometimes reported as evi- Primitive hematopoiesis is the source of the first hematopoietic cells
dence of the early colonization of the fetus by ‘yolk-sac macrophages’ before detected in the developing embryo. It is characterized by the emergence of
their replacement by monocyte-derived macrophages. However, this body progenitor cells with limited potential for development into erythrocytes
of work described the identification of proliferating non-phagocytic pre- and macrophages and that can be found in the extra-embryonic yolk-sac
cursors of macrophages (described as ‘immature macrophages’) that origi- blood islands in mice36. These progenitor cells are thought to arise directly
nate in the yolk sac, colonize the embryo and finish their differentiation in from the posterior plate mesoderm, and clonal analysis of the blood islands
situ within the fetal tissues20,30,31. This description did not correspond to has revealed that the endothelial cells lining the blood islands and the
a particular wave of hematopoietic progenitors (‘primitive’ or ‘definitive’), hematopoietic cells they contain do not share a common origin38. This
as discovered subsequently. However, the description of erythro-myeloid distinguishes them from the subsequent ‘definitive’ hematopoietic waves,
progenitors (EMPs)33–36 (discussed below) and additional fate-mapping which are thought to arise from a hemogenic endothelium. Accordingly,
data are in accordance with the original interpretation of the morphological primitive progenitors and their daughter cells are not affected when tran-
data. Indeed, genetic pulse labeling of Csf1r-positive c-Kit+CD45loAA4.1+ scription factors required for the endothelial-hematopoietic transition,
EMPs present in the yolk sac at E8.5 is followed by the detection of labeled such as Runx1, are absent (Table 1).
CD11b+ cells with negative or faint immunoreactivity for F4/80 at E9.5 in In zebrafish embryos, the ‘primitive’ wave can give rise to erythrocytes
both the yolk sac and embryo and then by the detection of F4/80bright cells and macrophages, as well as neutrophils, in the absence of the gene encod-
© 2016 Nature America, Inc. All rights reserved.

starting from E10.5 (ref. 18). ing Runx1 (refs. 39–41). In the mouse embryo, in vivo characterization of
More recent work has identified three successive but overlapping waves the macrophage progeny of the primitive wave is still hampered by experi-
of hematopoietic progenitors during development, all of which have the mental constrains. However, (rare) progenitors with restricted macrophage
potential to give rise to fetal macrophages34. These can be distinguished potential in vitro can be found in the yolk sac at the neural-plate stage
by the anatomical site (extra-embryonic yolk sac versus intra-embryonic between E7.5 and E8 (refs. 34,36).

Table 1 Primitive and definitive waves of hematopoiesis


Primitive Definitive
Progenitor name MP EMP HSC
Origin Posterior plate mesoderm Hemogenic endothelium Arterial hemogenic endothelium
Progenitor surface phenotype c-KitloCD41lo c-Kit+CD41+VE- Lin–c-Kit+Sca-1+CD41+VE-
Cad+CD16/32+AA4.1+Tie2+CD45loSca-1– Cad+Tie2+CD45+
Time window of generation E7.0–? E8.25–E10.5 E9.5–E12.5
Anatomical Generation YS (blood islands) YS AGM (and large arteries)
location of: Expansion YS FL FL
Differentiation-commitment YS YS and FL FL, BM
Differentiation potential Myeloid Erythroid Erythroid
npg

Myeloid Myeloid
Lymphoid
Macrophage lineage PU.1 dependent PU.1 dependent PU.1 dependent
Runx1 independent Runx1 dependent Runx1 dependent
c-Myb independent c-Myb independent c-Myb dependent
Notch1 independent Notch1 dependent
Macrophage self-renewal ? Yes ?
Progenitor long-term self-renewal No (transient) No (transient) Yes
TF expression Pu.1 (Spi1) NA + +
c-Myb NA + +
GATA-2 NA + +
GATA-3 NA – +
Lmo2 NA – +
TF requirement PU.1 (Spi1) Yes Yes No**
c-Myb No No* Yes
Runx1 No Yes Yes
CBFb No Yes Yes
Scl–Tal-1 Yes Yes Yes
Notch1 No No Yes
The surface phenotypes, ability to self-renew, anatomical sites of origin, expansion and differentiation, expression and dependence on transcription factors of various
progenitor cells. MP, myeloid progenitor; YS, yolk sac; FL, fetal liver; BM, bone marrow; TF, transcription factor; NA, not applicable.

*Red blood cell differentiation is compromised. **Macrophage differentiation is compromised.

NATURE IMMUNOLOGY VOLUME 17 NUMBER 1 JANUARY 2016 3


REVIEW

Figure 1 Time scale for the development of Blood


hematopoietic progenitor cells in the mouse islands
Fetal liver
embryo. There are three successive but Hematopoietic Yolk
organs sac
overlapping waves of hematopoietic progenitor Bone marrow
cells during development, each of which has
the potential to give rise to fetal macrophages. “Primitive”
progenitors
These can be distinguished by the hematopoietic EMPs
niches in which the progenitor cells expand Hematopoietic
and differentiate during development (top): progenitors Fetal HSCs Adult HSCs
the yolk sac (primitive progenitors (green) and
EMPs (blue)), the fetal liver (EMPs and HSCs),
Erythrocytes and myeloid cells Resident macrophages
and, finally, the bone marrow (HSCs (orange)). Hematopoietic Erythrocytes
While primitive progenitor cells have been found cells (myeloid cells) Erythrocytes, passenger myeloid cells, lymphoid cells
only in a small time window (middle), definitive
progenitor cells (including EMPs and HSCs)
Timeline
co-exist during most fetal development in the
E7.5 E10.5 E12.5 E14.5 E16.5 E18.5 Birth Weaning Adulthood
fetal liver. Only HSC-derived hematopoiesis
then shifts to the bone marrow niche. The three
waves of progenitor cells can also be distinguished by their differentiation potential in vivo (bottom). Primitive progenitor cells are restricted to the erythroid
or myeloid lineage, while EMPs have both erythroid and myeloid potential. EMP-derived hematopoiesis gives rise to erythrocytes, macrophages, monocytes,
granulocytes and mast cells and is sufficient to support survival of HSC-deficient embryos until birth. EMP-derived macrophages persist after birth in most
tissues as resident macrophages, except in the intestinal lamina propria. HSCs are distinguished from other progenitor cells by their ability to perform long-
term repopulation of all leukocyte lineages when transplanted into a conventional irradiated recipient mouse.
© 2016 Nature America, Inc. All rights reserved.

EMPs give rise to most tissue-resident macrophages has been described elsewhere52,53. HSCs and EMPs arise from distinct
The first ‘definitive’ progenitor cells emerge in the yolk sac of mouse hemogenic endothelial cells43. HSCs emerge from the hemogenic endothe-
embryos at E8.25 (refs. 34,36). These progenitors (EMPs) are phenotypi- lium of the aorto-gonado-mesonephros (AGM) region (starting at E10.5 in
cally defined as c-Kit+, AA4.1+ (CD93+), CD41+, positive for the endothelial mice)39,54,55 and of the umbilical and vitelline arteries56 and migrate to the
marker VE-cadherin, positive for CD16/32 (FCgII and FCgIII recep- fetal liver, where they expand and differentiate from E12.5 until definitive
tors), and CD45lo (refs. 33,35) (Table 1). Examination of their erythroid hematopoiesis begins to shift to the bone marrow. HSCs colonize the embry-
progeny led to their classification as ‘definitive’ progenitors42. However, onic bone marrow at E15, and active hematopoiesis starts there at E17 (refs.
they can be distinguished from HSCs by their lack of lymphoid poten- 47,57). Fetal and adult HSCs require c-Myb for their self-renewal and main-
tial, both in vitro and in vivo, their lack of long-term repopulating poten- tenance, and loss of c-Myb expression leads to rapid HSC-derived hemato-
tial35 and lack of surface expression of the lineage marker Sca-1 (ref. 35). poiesis failure12,58–60. In addition, HSCs also require the transmembrane
EMP-derived hematopoiesis is sufficient to support survival of HSC- receptor Notch1 for their emergence, in contrast to EMPs, as Notch1–/–
deficient embryos until birth43. EMP emerge from the yolk-sac hemogenic embryos have normal numbers of hematopoietic progenitor cells in the
endothelium in a Runx1-dependant endothelial-to-hematopoietic transi- yolk sac but very few in the body of the embryo61. Several groups have
tion44. The emergence of EMPs from the yolk sac does not require blood attempted to pinpoint the specific site and time at which HSCs can be first
flow, as indicated by results obtained by the study of embryos deficient detected within the embryo proper. Pioneering work demonstrated that
in the sodium-calcium exchanger NCX1 or VE-cadherin35,45,46. They are the AGM region contains the progenitor cells endowed with long-term–
generated from Tie2+ yolk-sac ancestors from E7.5 to E10.5 (ref. 18). The repopulating ability at E10.5 (ref. 62). However, immature HSCs have been
number of EMPs in the yolk sac peaks between E9.5 and E10.5, and they detected at E9–E9.5 in the para-aortic splanchnopleura, a structure that
npg

seed the fetal liver as soon as E9 (ref. 47). Circulating EMPs are still detected later evolves into the AGM region. Such immature HSCs migrate into the
in peripheral blood at E14.5 (ref. 35), and EMP-derived c-Kit+ cells are fetal liver at E10, mature into HSCs and contribute to the adult HSC pool47.
found in the fetal liver at 16.5 (ref. 18). EMPs expand in the fetal liver and
differentiate into erythrocytes, megakaryocytes, macrophages, monocytes, Genetic models distinguish the three progenitor waves
granulocytes and mast cells18. EMPs isolated from the yolk sac at E8.5 and As described above, the emergence of different waves of hematopoietic
the fetal liver at E12.5 have the same differentiation potential in vitro, but precursor cells partially overlaps in a short time frame, which has made
erythroid, monocyte and granulocyte and mast-cell potential is observed fate-mapping difficult. However, these precursor cells can be distin-
in vivo only after seeding of the fetal liver18. Thus, the fetal liver niche pro- guished genetically, and overall, the layering of the hematopoietic system
vides critical cues or an environment for EMPs to reach their full potential. and in particular of macrophage development seems to stand on a robust
Yolk-sac EMPs express the gene encoding the transcription factor c-Myb genetic basis. Primitive hematopoiesis occurs in the absence of Runx1 and
(Myb)48, but their commitment and differentiation into the myeloid fate is c-Myb63,64, while EMPs are dependent on Runx1 for their emergence64 but
unaltered in c-Myb-deficient embryos, although their erythroid potential are independent of c-Myb for their myeloid differentiation12,18,49 and are
is blocked12,18,49,50 (Table 1). Therefore, c-Myb is required for the commit- Notch1 independent61. In contrast, fetal and adult HSCs require Runx1,
ment and differentiation of EMPs into the erythroid fate50 but is dispens- c-Myb and Notch1 (refs. 39,40,58,59,61) (Table 1). On the basis of these
able for myeloid differentiation and is possibly redundant for this, because data, several reports have investigated the lineage of origin of fetal (primi-
EMPs express the gene encoding b-Myb (Mybl2), a transcription factor that tive) and adult macrophages and the mechanisms that might account for
can restore hematopoiesis in c-Myb-deficient Drosophila melanogaster51. their persistence in adults.

HSCs are developmentally distinct from EMPs Resident macrophages originate mainly from yolk-sac EMPs
HSCs are distinguished from other progenitor cells by their ability to per- The development of fetal F4/80bright macrophages is unaltered in c-Myb-
form long-term repopulation of all leukocyte lineages when transplanted deficient mice, and adult F4/80bright macrophages are largely maintained
into a conventional irradiated recipient mouse. HSC-derived hematopoiesis in adult mice in the absence of c-Myb and independently of bone marrow

4 VOLUME 17 NUMBER 1 JANUARY 2016 NATURE IMMUNOLOGY


REVIEW

progenitor cells12. In addition, adult tissue-resident F4/80bright macro- Mesoderm


phages have been traced back to c-Myb-independent precursor cells
expressing the cytokine receptor CSF1R and present in the early mouse
Runx1 independent
embryo at E8.5, as assessed through the use of Cre recombinase–mediated, YS hemogenic
E7.5
hydroxytamoxifen-induced pulse labeling65,66 of transgenic mice express- endothelium

ing a tamoxifen-inducible fusion protein of ‘improved’ Cre recombinase


(iCre) and the mouse estrogen receptor (Mer-iCre-Mer) under control of E8.5 Primitive YS-derived EMPs
hematopoiesis
the macrophage-specific mouse gene encoding CSF1R12 (discussed below). Runx1 c-Myb-
dependent independent
The data noted above would suggest that F4/80bright macrophages origi-
nate not from HSCs but from yolk-sac progenitor cells such as either primi- E9.5 Intraembyonic
tive progenitor cells or EMPs48. In accordance with that, brain microglia hemogenic
endothelium
develop from yolk-sac progenitor cells14 that have been functionally identi-
fied as EMPs48. Following pulse labeling of CSF1R-expressing cells with E10.5
c-Myb-dependent Fetal macrophages
hydroxytamoxifen at E8.5 (in the mouse model described above), the label-
ing efficiency of F4/80bright macrophages is 30% at day 10.5 and remains E12.5
HSCs
high (~20%) in microglia but progressively decreases from E12.5 onward
in other organs to reach a plateau at birth that is maintained even in 1-year- Erythrocytes,
old mice (5% for the liver Kupffer cells; 1% for epidermal Langerhans E16.5 Flt3+ progenitors megakaryocytes
cells)12,18,67 (data not shown). This might be explained by the observation granulocytes
monocytes
that pulse labeling with hydroxytamoxifen at E8.5 labels only the very first mast cells
Birth
EMPs that emerge in the yolk sac, while EMPs continue to emerge in the
© 2016 Nature America, Inc. All rights reserved.

yolk sac until at least E10.5 (ref. 18), and therefore the frequency with which
the macrophages are labeled in embryonic tissues might reflect the time
at which their colonization by fetal macrophages is completed. However,
it is also possible that this ‘dilution’ reflects a contribution by another pro- Tissue-resident
Erythrocytes,
Aging macrophages
genitor cells, such as fetal HSCs or HSC-derived monocytes, to several megakaryocytes
(Kupffer cells,
lymphoid cells
fetal macrophage populations17,67–69. A contribution by fetal monocytes myeloid cells
microglia,
Langerhans cells)
is unlikely, as they also derive from EMP until E16.5 (refs. 18,35). However,
a contribution by fetal HSCs is still under debate17,68–70. Figure 2 Myeloid development and maintenance in the mouse embryo
The main argument against the possibility of a substantial contribu- and adult mouse. Primitive progenitor cells do not share a common origin
tion by fetal HSCs has been provided by experiments with a fate-mapping with endothelial cells lining the blood islands in the yolk sac (YS), and they
emerge the absence of Runx1 and c-Myb. During definitive hematopoiesis,
mouse model in which the expression of Cre recombinase (as the Mer-
HSCs and EMPs arise from distinct hemogenic endothelial cells through
iCre-Mer fusion) is inducible in cells expressing the tyrosine kinase Tie2 a Runx1-dependent endothelial-to-hematopoietic transition. Both EMPs
(encoded by Tek)18,71. Tie2 is expressed in the hemogenic endothelium, and HSCs express c-Myb, and although no fetal or adult HSC-derived
endothelial cells and hematopoietic progenitor cells in the yolk sac72, hematopoiesis can occur in the absence of c-Myb, EMPs are c-Myb
AGM region, fetal liver HSCs and adult HSCs52. Pulse labeling of these independent for their myeloid differentiation. EMP-derived hematopoiesis
mice with tamoxifen at E7.5, E8.5, E9.5 and E10.5 results in the labeling gives rises to erythrocytes and short-lived myeloid cells (monocytes,
granulocytes and mast cells) that are replaced by HSC-derived cells late
of adult HSCs, lymphocytes, monocytes and neutrophils with the same
during gestation. EMP-derived macrophages colonize all tissues during
efficiency, as expected. In contrast, fetal macrophages and adult tissue-
npg

fetal development, where they specialize to their tissue of residency after


resident F4/80bright macrophages in all organs are labeled with a decreasing birth and can persist throughout adult life by local proliferation (circular
efficiency (high at E7.5 to very low at E10.5), which indicates that fetal and red arrows indicate self-renewal potential). Depending on the age of the
adult F4/80bright macrophages originate mainly from a progenitor cell that organism and environmental challenges, HSC-derived cells can contribute to
does not express Tie2 after E10.5, with a minor contribution of progenitor adult tissue-resident populations.
cells (such as HSCs) that express Tie2 after E10.5.
Research with fate-mapping models in which the expression of Cre embryo18, while EMP-derived macrophages persist all through fetal and
recombinase is either inducible (as the Mer-iCre-Mer fusion) or constitu- post-natal development as tissue-resident macrophages12,18 (Fig. 2). It has
tive (iCre alone) in CSF1R-expressing cells has identified the macrophage also been speculated that a first wave of c-Myb-independent EMPs gives
progenitor itself as a CD45loKit+AA4.1+ EMP that appears in the yolk sac rise to microglia and a second wave of c-Myb-dependent EMPs gives rise
from E8.5 (ref. 18). These EMPs migrate into the fetal liver, where they to other macrophages67. However, there are no data available to support
expand and persist as c-Kit+ progenitors until at least E16.5 (ref. 18). These the proposal of the existence of two genetically distinct populations of
EMPs give rise to erythrocytes, monocytes, granulocytes and mast cells EMPs, and this hypothesis is not in accordance with reports that c-Myb
until late in fetal development18,35, in accordance with the observation that is expressed in early yolk-sac EMPs36,48 or with the persistence of fetal
HSC-independent hematopoiesis is not only necessary but also sufficient to resident macrophages in all c-Myb-deficient embryonic tissues12.
support the survival of HSC-deficient embryos until birth43. These EMPs
also give rise to F4/80bright fetal macrophages in all embryonic tissues from The contribution of ‘primitive’ hematopoiesis to microglia
E10.5 onward18. An EMP origin of the majority of tissue-resident macro- Primitive hematopoiesis could also contribute to ‘primitive’ and fetal mac-
phages has also been favored in an independent report67. rophages. Early pulse labeling of progenitor cells (at E7.5) with several dif-
Together, the data reported above support the proposal of a major ferent reporters, such as Runx1, CSF1R, Tie2 and c-Kit12,14,18,70, results in
contribution of yolk-sac EMPs to fetal and adult resident macrophages more efficient labeling of adult microglia than of other adult tissue-resident
(Figs. 1 and 2). EMP-derived erythrocytes, monocytes, granulocytes and macrophages. As mentioned above, this might relate either to earlier ter-
mast cells are ultimately replaced by HSC-derived myeloid cells in the late mination of colonization of the brain by macrophages than that of other

NATURE IMMUNOLOGY VOLUME 17 NUMBER 1 JANUARY 2016 5


REVIEW

organs or to the existence of several subsets of EMPs. However, it might originate from a macrophage-restricted, self-renewing progenitor cell in
also reflect the contribution of primitive hematopoiesis to microglia. Future bone marrow that does not give rise to any other lineage73. These find-
studies should determine whether primitive progenitor cells are labeled ings, if confirmed, would indicate that myelopoiesis in birds is markedly
in these models and should investigate whether macrophages produced different from its counterparts in mice, zebrafish and human, because a
by primitive hematopoiesis can persist in adult mice. Independence of macrophage-restricted, self-renewing progenitor cell in the bone marrow
c-Myb does not allow genetic distinction of primitive hematopoiesis from or its equivalent has not been identified in these species.
EMP-derived hematopoiesis, and it is not yet known whether Runx1 is
required for the differentiation of macrophages in mice. Together, the Maintenance of macrophages by local proliferation or recruitment
lack of genetic delineation and the low temporal resolution of classic fate- As indicated above, a proportion of alveolar macrophages, kidney mac-
mapping approaches in the mouse have not yet allowed independent track- rophages and heart macrophages seem to be replaced during the normal
ing of the respective progeny of the two progenitor populations. Better aging process12,18,75. Partial replacement of tissue-resident macrophages
understanding of the molecular control of primitive progenitor cells and is also observed following g-irradiation, bone marrow transplantation or
the commitment and differentiation of EMPs is needed for the develop- adoptive-transfer experiments3,4,77 and in macrophage-depletion studies,
ment of better tools for characterization of the possible contribution of such as intravenous injection of clodronate liposomes. Liver Kupffer cells
‘primitive’ hematopoiesis to the development of fetal macrophages and, are also reported to be replaced by bone marrow–derived progenitor cells
possibly, adult macrophages in mice. following, for example, the death of Kupffer cells in severe experimental
infection with Listeria monocytogenes29. However, resident macrophages
Contribution of HSCs depends on organ and time such as Langerhans cells, Kupffer cells, microglia, alveolar macrophages,
Analysis of the contribution of HSCs to adult resident macrophages in peritoneal macrophages and splenic macrophages all have the potential
the absence of bone marrow irradiation has been performed by several to proliferate15,22,23,27,78–82. In some cases, tissue-resident macrophages
groups studying mice expressing Cre recombinase from the gene encod- can immediately replenish themselves following severe depletion15,23,82.
© 2016 Nature America, Inc. All rights reserved.

ing the receptor tyrosine kinase Flt3 (Flk2) (refs. 12,17,18,73), on the A growing body of evidence now clearly shows that fully differentiated
basis of the observation that Flt3 is expressed by HSC-derived progeni- resident macrophages can re-enter the cell cycle and maintain their num-
tor cells71,74 but not by fetal macrophages12. The contribution of HSCs bers by local proliferation23–27. Macrophage proliferation increases sub-
has also been studied in several independent models of bone marrow stantially during inflammatory conditions, such as skin inflammation,
transplantation without irradiation, such as Mybfl/flMx1-Cre mice (which during which up to 30% of Langerhans cells express the proliferation
undergo deletion of loxP-flanked Myb alleles (Mybfl/fl) via Cre expressed marker Ki67 (ref. 22). During an acute inflammatory response, the tissue-
from the interferon-inducible gene Mx1) and KitW/WvRag2–/–Il2rg–/– resident peritoneal macrophages that survive undergo a transient and
mice (which lack mast cells and have hypoplastic bone marrow (KitW/Wv) intense proliferative burst in situ to repopulate the tissue23. In response
and lack lymphocytes (Rag2–/–Il2rg–/–))12,18 and in parabiotic mice15,75. to helminth infection, macrophages residing in the pleural and peritoneal
Overall, these analyses have revealed that the contribution of HSCs cavities expand by local proliferation81. In glucan-induced formation of
to tissue-resident macrophages differs among organs and frequently granulomas in monocytopenic mice, Kupffer cells proliferate actively
increases with age. The contribution of HSCs to adult tissue-resident and contribute to granuloma formation78. After conditional ablation of
macrophages is minor (<5%) in the brain, liver and epidermis12,14,48, microglia in adult mice, the microglial compartment is reconstituted
is small but increases with age in the lungs, heart and spleen18,75, and within 1 week by cells residing in the central nervous system, indepen-
sometimes predominates after weaning in gut lamina propria28,76 (Figs. dently of bone marrow–derived precursor cells82. However, it remains
1 and 2). It is not yet known whether the precursor(s) of the HSC-derived unclear whether the tissue-resident macrophage populations contain a
resident macrophages is (are) the monocyte or earlier myeloid progeni- local ‘progenitor compartment’ or whether all macrophages have the
tor, except in the gut, in which Ly6C+ monocytes are proposed to be the ability to undergo mitosis. The mechanisms that underlie this ability of
npg

immediate precursor of gut lamina propria macrophages28,76. tissue-resident macrophages to self-maintain and to proliferate are still
Of note, a published report has investigated the origin of tissue-resident unclear. An important role for the Maf family of transcription factors is
macrophages through the use of a fate-mapping model in which the expres- likely, as combined deficiency in MafB and c-Maf enables the KLF4- and
sion of Cre recombinase (as Mer-iCre-Mer) is inducible in cells expressing c-Myc–dependent proliferation of differentiated macrophages in culture
the gene encoding the stem-cell-factor receptor c-Kit (CD117) (ref. 70). in response to the cytokine CSF1 (M-CSF)83. In epidermal Langerhans
Notably, this study represents independent confirmation of the finding that cell, proliferation in vivo has been shown to be driven by an unidentified
most adult tissue-resident macrophages originate from embryonic and fetal keratinocyte-derived signal22. During nematode infection, macrophage
progenitor cells and persist in adults independently of adult HSCs, as c-Kit+ proliferation is impaired in the absence of interleukin 4 (refs. 81,84).
progenitor cells do not seem to contribute to tissue-resident macrophages The cellular and molecular mechanisms that control macrophage main-
after E16.5 (ref. 70). The authors also conclude that most tissue-resident tenance and expansion in adult mice, and the consequences of aging on
macrophages, except microglia, are descendants of fetal HSCs, not EMPs, these processes, are an area of active investigation, as their elucidation
on the basis of the observation that labeling of c-Kit+Sca-1+ EMPs at E7.5 will shed light on the pathophysiology of inflammation.
results in the labeling of microglia only, while labeling of c-Kit+ progenitor
cells at E8.5 or E9.5, in contrast, results in the labeling of all adult resident Conclusions and perspectives on macrophage function
macrophages and HSC-derived cells70. However, EMPs do not express A revised model for macrophage development and function is therefore
Sca-1 (ref. 35) (Table 1), while EMPs and fetal HSCs both express c-Kit emerging that distinguishes the tissue-resident macrophages that develop
and co-exist in the fetal liver until E16.5 (refs. 18,35). Thus, c-Kit expres- during embryogenesis from the passenger myeloid cells that originate
sion and c-Kit-based fate mapping might not allow the distinction of EMPs and undergo renewal from bone marrow HSCs. This distinction between
versus fetal HSCs. passenger macrophages and tissue-resident macrophages is in line with
Studies of chickens have suggested that in contrast to results obtained early kinetic studies of the splenic-myeloid compartment in adult mice,
for mice, yolk sac–derived macrophages are not retained in chickens after which proposed a dual origin for myeloid cells, with half of myeloid
hatching and, surprisingly, that tissue macrophages in the adult birds cells renewing from blood and the other half being produced within the

6 VOLUME 17 NUMBER 1 JANUARY 2016 NATURE IMMUNOLOGY


REVIEW

spleen85. It also explains the observations that Langerhans cells do not 1. van Furth, R. et al. The mononuclear phagocyte system: a new classification of macro-
phages, monocytes, and their precursor cells. Bull. World Health Organ. 46, 845–852
undergo exchange between parabiotic mice13 and that fetal microglia (1972).
persist in adult mice14. 2. van Furth, R. & Cohn, Z.A. The origin and kinetics of mononuclear phagocytes. J. Exp.
That distinction is relevant to disease pathogenesis, as it is apparent Med. 128, 415–435 (1968).
3. Katz, S.I., Tamaki, K. & Sachs, D.H. Epidermal Langerhans cells are derived from cells
that the functions of HSC-derived monocytes can be very different from originating in bone marrow. Nature 282, 324–326 (1979).
those of tissue-resident macrophages in the same inflammatory environ- 4. Fogg, D.K. et al. A clonogenic bone marrow progenitor specific for macrophages and
dendritic cells. Science 311, 83–87 (2006).
ment. The two cell types have distinct expansion mechanisms and distinct 5. Liu, K. et al. Origin of dendritic cells in peripheral lymphoid organs of mice. Nat.
functions in the brain during experimental autoimmune encephalitis86. Immunol. 8, 578–583 (2007).
Infiltrating monocytes are recruited via extravasation from blood vessels 6. Liu, K. et al. In vivo analysis of dendritic cell development and homeostasis. Science
324, 392–397 (2009).
and produce inflammatory mediators important for disease progression 7. Auffray, C. et al. CX3CR1+ CD115+ CD135+ common macrophage/DC precursors and
but do not persist after the resolution of inflammation, while, in contrast, the role of CX3CR1 in their response to inflammation. J. Exp. Med. 206, 595–606
(2009).
activated resident microglia proliferate locally, persist and return to quies- 8. Gordon, S. & Martinez, F.O. Alternative activation of macrophages: mechanism and
cence following remission86. Following parasitic helminth infection, pleural functions. Immunity 32, 593–604 (2010).
resident macrophages adopt anti-inflammatory phenotypes, while pas- 9. Gordon, S. Alternative activation of macrophages. Nat. Rev. Immunol. 3, 23–35
(2003).
senger macrophages exhibit pro-inflammatory responses81. However, in 10. Murray, P.J. et al. Macrophage activation and polarization: nomenclature and experi-
a model of chronic neurodegeneration, proliferating resident microglial mental guidelines. Immunity 41, 14–20 (2014).
cells contribute to neuronal damage during development of the disease87. 11. Schultze, J.L., Freeman, T., Hume, D.A. & Latz, E. A transcriptional perspective on
human macrophage biology. Semin. Immunol. 27, 44–50 (2015).
Thus, it might be inaccurate to describe tissue-resident macrophages as 12. Schulz, C. et al. A lineage of myeloid cells independent of Myb and hematopoietic
always being ‘anti-inflammatory’ in contrast to passenger macrophages stem cells. Science 336, 86–90 (2012).
13. Merad, M. et al. Langerhans cells renew in the skin throughout life under steady-state
that are always ‘pro-inflammatory’. The respective roles of tissue-resident conditions. Nat. Immunol. 3, 1135–1141 (2002).
macrophages and passenger macrophages remain to be studied in detail,
© 2016 Nature America, Inc. All rights reserved.

14. Ginhoux, F. et al. Fate mapping analysis reveals that adult microglia derive from primi-
and most experimental models available at present do not allow easy elu- tive macrophages. Science 330, 841–845 (2010).
15. Hashimoto, D. et al. Tissue-resident macrophages self-maintain locally throughout adult
cidation of the roles of resident and recruited macrophages in adult mouse life with minimal contribution from circulating monocytes. Immunity 38, 792–804
tissues. Future work should help characterize the roles of (EMP-derived) (2013).
16. Yona, S. et al. Fate mapping reveals origins and dynamics of monocytes and tissue
tissue-resident macrophages, separate them from the functions of HSC- macrophages under homeostasis. Immunity 38, 79–91 (2013).
derived passenger macrophages and characterize the underlying molecular 17. Epelman, S. et al. Embryonic and adult-derived resident cardiac macrophages are
mechanisms involved. maintained through distinct mechanisms at steady state and during inflammation.
Immunity 40, 91–104 (2014).
It is also important to recognize that the distinct subsets of resident mac- 18. Gomez Perdiguero, E. et al. Tissue-resident macrophages originate from yolk-sac-
rophages associated with different tissues, such as microglia, Langerhans derived erythro-myeloid progenitors. Nature 518, 547–551 (2015).
19. Geissmann, F. et al. Development of monocytes, macrophages, and dendritic cells.
cells, Kupffer cells, peritoneal macrophages, red-pulp macrophages and
Science 327, 656–661 (2010).
alveolar macrophages, have different phenotypes, transcriptional profiles 20. Takahashi, K., Yamamura, F. & Naito, M. Differentiation, maturation, and proliferation
and chromatin ‘landscapes’88,89, require specific growth factors, and rely on of macrophages in the mouse yolk sac: a light-microscopic, enzyme-cytochemical,
immunohistochemical, and ultrastructural study. J. Leukoc. Biol. 45, 87–96 (1989).
distinct transcription factors for their differentiation or maintenance. For 21. Alliot, F., Godin, I. & Pessac, B. Microglia derive from progenitors, originating from the
example, CSF1 is essential for Kupffer cells90, the growth factor TGF-b1 and yolk sac, and which proliferate in the brain. Brain Res. Dev. Brain Res. 117, 145–152
interleukin 34 are essential for microglia and Langerhans cells88,91–93, CSF2 (1999).
22. Chorro, L. et al. Langerhans cell (LC) proliferation mediates neonatal development,
(GM-CSF) is essential for alveolar macrophages in mice and humans94–99, homeostasis, and inflammation-associated expansion of the epidermal LC network.
and retinoic acid and the transpiration factor GATA-6 are essential for J. Exp. Med. 206, 3089–3100 (2009).
large peritoneal macrophages88,100,101. The transcription factor Spi-C is 23. Davies, L.C. et al. A quantifiable proliferative burst of tissue macrophages restores
homeostatic macrophage populations after acute inflammation. Eur. J. Immunol. 41,
important for the development of red-pulp macrophages102, while mice
npg

2155–2164 (2011).
deficient in the nuclear receptor LXR are defective in the generation of 24. Lawson, L.J., Perry, V.H. & Gordon, S. Turnover of resident microglia in the normal
adult mouse brain. Neuroscience 48, 405–415 (1992).
marginal-zone and metallophilic macrophages103. 25. Ghigo, C. et al. Multicolor fate mapping of Langerhans cell homeostasis. J. Exp. Med.
Tissue-specific environments are important for the maintenance of mac- 210, 1657–1664 (2013).
rophage identity88,89. Several studies also suggest that individual resident 26. Kanitakis, J., Morelon, E., Petruzzo, P., Badet, L. & Dubernard, J.M. Self-renewal
capacity of human epidermal Langerhans cells: observations made on a composite
macrophage subsets follow a differentiation program during embryogene- tissue allograft. Exp. Dermatol. 20, 145–146 (2011).
sis and early post-natal life that results in the establishment of stable pheno- 27. Bouwens, L., Baekeland, M., De Zanger, R. & Wisse, E. Quantitation, tissue distribution
and proliferation kinetics of Kupffer cells in normal rat liver. Hepatology 6, 718–722
typic and functional subsets22,48. Future work investigating the respective
(1986).
contributions and interplay of lineage-specific ‘hard-wired’ differentiation 28. Bain, C.C. et al. Constant replenishment from circulating monocytes maintains the
programs and of the local tissue micro-environment to the differentiation, macrophage pool in the intestine of adult mice. Nat. Immunol. 15, 929–937 (2014).
29. Bllériot, C. et al. Liver-resident macrophage necroptosis orchestrates type 1 micro-
maintenance and activation of tissue-specific resident macrophages will bicidal inflammation and type-2-mediated tissue repair during bacterial infection.
contribute to the understanding of their functions. Immunity 42, 145–158 (2015).
30. Naito, M., Takahashi, K. & Nishikawa, S. Development, differentiation, and maturation
ACKNOWLEDGMENTS of macrophages in the fetal mouse liver. J. Leukoc. Biol. 48, 27–37 (1990).
Supported by the National Cancer Institute of the US National Institutes of Health 31. Takahashi, K., Takahashi, H., Naito, M., Sato, T. & Kojima, M. Ultrastructural and
(P30CA008748 to F.G.), the Wellcome Trust (WT101853MA to F.G.), the European functional development of macrophages in the dermal tissue of rat fetuses. Cell Tissue
Res. 232, 539–552 (1983).
Research Council (2010-StG-261299 to F.G.), the Institut Pasteur (E.G.P.) and
32. Deimann, W. & Fahimi, H.D. Peroxidase cytochemistry and ultrastructure of resident
Agence Nationale de la Recherche (Laboratoire d'Excellence Revive, Investissement macrophages in fetal rat liver. A developmental study. Dev. Biol. 66, 43–56 (1978).
d'Avenir; ANR-10-LABX-73 to E.G.P.). 33. Bertrand, J.Y. et al. Characterization of purified intraembryonic hematopoietic stem
cells as a tool to define their site of origin. Proc. Natl. Acad. Sci. USA 102, 134–139
COMPETING FINANCIAL INTERESTS (2005).
The authors declare no competing financial interests. 34. Bertrand, J.Y. et al. Three pathways to mature macrophages in the early mouse yolk
sac. Blood 106, 3004–3011 (2005).
Reprints and permissions information is available online at http://www.nature.com/ 35. McGrath, K.E. et al. Distinct sources of hematopoietic progenitors emerge before
HSCs and provide functional blood cells in the mammalian embryo. Cell Rep. 11,
reprints/index.html.
1892–1904 (2015).

NATURE IMMUNOLOGY VOLUME 17 NUMBER 1 JANUARY 2016 7


REVIEW

36. Palis, J., Robertson, S., Kennedy, M., Wall, C. & Keller, G. Development of erythroid 70. Sheng, J., Ruedl, C. & Karjalainen, K. Most tissue-resident macrophages except microg-
and myeloid progenitors in the yolk sac and embryo proper of the mouse. Development lia are derived from fetal hematopoietic stem cells. Immunity 43, 382–393 (2015).
126, 5073–5084 (1999). 71. Busch, K. et al. Fundamental properties of unperturbed haematopoiesis from stem
37. Palis J. Primitive and definitive erythropoiesis in mammals. Front. Physiol. 5, 3 cells in vivo. Nature 518, 542–546 (2015).
(2014). 72. Ema, M. et al. Primitive erythropoiesis from mesodermal precursors expressing
38. Ueno, H. & Weissman, I.L. Clonal analysis of mouse development reveals a polyclonal VE-cadherin, PECAM-1, Tie2, endoglin, and CD34 in the mouse embryo. Blood 108,
origin for yolk sac blood islands. Dev. Cell 11, 519–533 (2006). 4018–4024 (2006).
39. Kissa, K. & Herbomel, P. Blood stem cells emerge from aortic endothelium by a novel 73. Garceau, V. et al. The development and maintenance of the mononuclear phagocyte
type of cell transition. Nature 464, 112–115 (2010). system of the chick is controlled by signals from the macrophage colony-stimulating
40. Okuda, T., van Deursen, J., Hiebert, S.W., Grosveld, G. & Downing, J.R. AML1, the factor receptor. BMC Biol. 13, 12 (2015).
target of multiple chromosomal translocations in human leukemia, is essential for 74. Buza-Vidas, N. et al. FLT3 expression initiates in fully multipotent mouse hematopoietic
normal fetal liver hematopoiesis. Cell 84, 321–330 (1996). progenitor cells. Blood 118, 1544–1548 (2011).
41. Le Guyader, D. et al. Origins and unconventional behavior of neutrophils in developing 75. Molawi, K. et al. Progressive replacement of embryo-derived cardiac macrophages with
zebrafish. Blood 111, 132–141 (2008). age. J. Exp. Med. 211, 2151–2158 (2014).
42. England, S.J., McGrath, K.E., Frame, J.M. & Palis, J. Immature erythroblasts with 76. Varol, C. et al. Intestinal lamina propria dendritic cell subsets have different origin and
extensive ex vivo self-renewal capacity emerge from the early mammalian fetus. Blood functions. Immunity 31, 502–512 (2009).
117, 2708–2717 (2011). 77. Hickey, W.F., Vass, K. & Lassmann, H. Bone marrow-derived elements in the central
43. Chen, M.J. et al. Erythroid/myeloid progenitors and hematopoietic stem cells originate nervous system: an immunohistochemical and ultrastructural survey of rat chimeras.
from distinct populations of endothelial cells. Cell Stem Cell 9, 541–552 (2011). J. Neuropathol. Exp. Neurol. 51, 246–256 (1992).
44. Chen, M.J., Yokomizo, T., Zeigler, B.M., Dzierzak, E. & Speck, N.A. Runx1 is required 78. Yamada, M., Naito, M. & Takahashi, K. Kupffer cell proliferation and glucan-
for the endothelial to haematopoietic cell transition but not thereafter. Nature 457, induced granuloma formation in mice depleted of blood monocytes by strontium-89.
887–891 (2009). J. Leukoc. Biol. 47, 195–205 (1990).
45. Lux, C.T. et al. All primitive and definitive hematopoietic progenitor cells emerging 79. Golde, D.W., Byers, L.A. & Finley, T.N. Proliferative capacity of human alveolar mac-
before E10 in the mouse embryo are products of the yolk sac. Blood 111, 3435–3438 rophage. Nature 247, 373–375 (1974).
(2008). 80. Sawyer, R.T., Strausbauch, P.H. & Volkman, A. Resident macrophage proliferation in
46. Rampon, C. & Huber, P. Multilineage hematopoietic progenitor activity generated mice depleted of blood monocytes by strontium-89. Lab. Invest. 46, 165–170 (1982).
autonomously in the mouse yolk sac: analysis using angiogenesis-defective embryos. 81. Jenkins, S.J. et al. Local macrophage proliferation, rather than recruitment from the
Int. J. Dev. Biol. 47, 273–280 (2003). blood, is a signature of TH2 inflammation. Science 332, 1284–1288 (2011).
© 2016 Nature America, Inc. All rights reserved.

47. Kieusseian, A., Brunet de la Grange, P., Burlen-Defranoux, O., Godin, I. & Cumano, A. 82. Bruttger, J. et al. Genetic cell ablation reveals clusters of local self-renewing microglia
Immature hematopoietic stem cells undergo maturation in the fetal liver. Development in the mammalian central nervous system. Immunity 43, 92–106 (2015).
139, 3521–3530 (2012). 83. Aziz, A., Soucie, E., Sarrazin, S. & Sieweke, M.H. MafB/c-Maf deficiency enables self-
48. Kierdorf, K. et al. Microglia emerge from erythromyeloid precursors via Pu.1- and Irf8- renewal of differentiated functional macrophages. Science 326, 867–871 (2009).
dependent pathways. Nat. Neurosci. 16, 273–280 (2013). 84. Jenkins, S.J. et al. IL-4 directly signals tissue-resident macrophages to proliferate
49. Sumner, R., Crawford, A., Mucenski, M. & Frampton, J. Initiation of adult myelopoi- beyond homeostatic levels controlled by CSF-1. J. Exp. Med. 210, 2477–2491 (2013).
esis can occur in the absence of c-Myb whereas subsequent development is strictly 85. van Furth, R. & Diesselhoff-den Dulk, M.M. Dual origin of mouse spleen macrophages.
dependent on the transcription factor. Oncogene 19, 3335–3342 (2000). J. Exp. Med. 160, 1273–1283 (1984).
50. Bartůnek, P., Kralova, J., Blendinger, G., Dvorak, M. & Zenke, M. GATA-1 and c-myb 86. Ajami, B., Bennett, J.L., Krieger, C., McNagny, K.M. & Rossi, F.M. Infiltrating mono-
crosstalk during red blood cell differentiation through GATA-1 binding sites in the c-myb cytes trigger EAE progression, but do not contribute to the resident microglia pool.
promoter. Oncogene 22, 1927–1935 (2003). Nat. Neurosci. 14, 1142–1149 (2011).
51. Davidson, C.J., Tirouvanziam, R., Herzenberg, L.A. & Lipsick, J.S. Functional evolution 87. Gomez-Nicola, D., Fransen, N.L., Suzzi, S. & Perry, V.H. Regulation of microglial
of the vertebrate Myb gene family: B-Myb, but neither A-Myb nor C-Myb, complements proliferation during chronic neurodegeneration. J. Neurosci. 33, 2481–2493 (2013).
Drosophila Myb in hemocytes. Genetics 169, 215–229 (2005). 88. Gosselin, D. et al. Environment drives selection and function of enhancers controlling
52. Cumano, A. & Godin, I. Ontogeny of the hematopoietic system. Annu. Rev. Immunol. tissue-specific macrophage identities. Cell 159, 1327–1340 (2014).
25, 745–785 (2007). 89. Lavin, Y. et al. Tissue-resident macrophage enhancer landscapes are shaped by the
53. Orkin, S.H. & Zon, L.I. Hematopoiesis: an evolving paradigm for stem cell biology. Cell local microenvironment. Cell 159, 1312–1326 (2014).
132, 631–644 (2008). 90. Yamamoto, T. et al. Macrophage colony-stimulating factor is indispensable for repopula-
54. Bertrand, J.Y. et al. Haematopoietic stem cells derive directly from aortic endothelium tion and differentiation of Kupffer cells but not for splenic red pulp macrophages in
during development. Nature 464, 108–111 (2010). osteopetrotic (op/op) mice after macrophage depletion. Cell Tissue Res. 332, 245–256
55. Boisset, J.C. et al. In vivo imaging of haematopoietic cells emerging from the mouse (2008).
aortic endothelium. Nature 464, 116–120 (2010). 91. Butovsky, O. et al. Identification of a unique TGF-b-dependent molecular and functional
56. Zovein, A.C. et al. Vascular remodeling of the vitelline artery initiates extravascular signature in microglia. Nat. Neurosci. 17, 131–143 (2014).
emergence of hematopoietic clusters. Blood 116, 3435–3444 (2010). 92. Wang, Y. et al. IL-34 is a tissue-restricted ligand of CSF1R required for the development
57. Delassus, S. & Cumano, A. Circulation of hematopoietic progenitors in the mouse of Langerhans cells and microglia. Nat. Immunol. 13, 753–760 (2012).
npg

embryo. Immunity 4, 97–106 (1996). 93. Borkowski, T.A., Letterio, J.J., Farr, A.G. & Udey, M.C. A role for endogenous transform-
58. Mucenski, M.L. et al. A functional c-myb gene is required for normal murine fetal ing growth factor beta 1 in Langerhans cell biology: the skin of transforming growth
hepatic hematopoiesis. Cell 65, 677–689 (1991). factor beta 1 null mice is devoid of epidermal Langerhans cells. J. Exp. Med. 184,
59. Mukouyama, Y. et al. Hematopoietic cells in cultures of the murine embryonic aorta- 2417–2422 (1996).
gonad-mesonephros region are induced by c-Myb. Curr. Biol. 9, 833–836 (1999). 94. Lieschke, G.J. et al. Mice lacking both macrophage- and granulocyte-macrophage
60. Soza-Ried, C., Hess, I., Netuschil, N., Schorpp, M. & Boehm, T. Essential role of c-myb colony-stimulating factor have macrophages and coexistent osteopetrosis and severe
in definitive hematopoiesis is evolutionarily conserved. Proc. Natl. Acad. Sci. USA 107, lung disease. Blood 84, 27–35 (1994).
17304–17308 (2010). 95. Shibata, Y. et al. GM-CSF regulates alveolar macrophage differentiation and innate
61. Kumano, K. et al. Notch1 but not Notch2 is essential for generating hematopoietic immunity in the lung through PU.1. Immunity 15, 557–567 (2001).
stem cells from endothelial cells. Immunity 18, 699–711 (2003). 96. Happle C. et al. Pulmonary transplantation of macrophage progenitors as effective and
62. Müller, A.M., Medvinsky, A., Strouboulis, J., Grosveld, F. & Dzierzak, E. Development of long-lasting therapy for hereditary pulmonary alveolar proteinosis. Sci. Transl. Med. 6,
hematopoietic stem cell activity in the mouse embryo. Immunity 1, 291–301 (1994). 250ra113 (2014).
63. Tober, J., McGrath, K.E. & Palis, J. Primitive erythropoiesis and megakaryopoiesis in 97. Willinger, T. et al. Human IL-3/GM-CSF knock-in mice support human alveolar macro-
the yolk sac are independent of c-myb. Blood 111, 2636–2639 (2008). phage development and human immune responses in the lung. Proc. Natl. Acad. Sci.
64. Yokomizo, T. et al. Requirement of Runx1/AML1/PEBP2aB for the generation of hae- USA 108, 2390–2395 (2011).
matopoietic cells from endothelial cells. Genes Cells 6, 13–23 (2001). 98. Akagawa, K.S., Kamoshita, K. & Tokunaga, T. Effects of granulocyte-macrophage
65. Qian, B.Z. et al. CCL2 recruits inflammatory monocytes to facilitate breast-tumour colony-stimulating factor and colony-stimulating factor-1 on the proliferation and dif-
metastasis. Nature 475, 222–225 (2011). ferentiation of murine alveolar macrophages. J. Immunol. 141, 3383–3390 (1988).
66. Metzger, D., Clifford, J., Chiba, H. & Chambon, P. Conditional site-specific recombina- 99. Chen, B.D., Mueller, M. & Chou, T.H. Role of granulocyte/macrophage colony-stim-
tion in mammalian cells using a ligand-dependent chimeric Cre recombinase. Proc. ulating factor in the regulation of murine alveolar macrophage proliferation and dif-
Natl. Acad. Sci. USA 92, 6991–6995 (1995). ferentiation. J. Immunol. 141, 139–144 (1988).
67. Hoeffel, G. et al. C-Myb+ erythro-myeloid progenitor-derived fetal monocytes give rise 100. Okabe, Y. & Medzhitov, R. Tissue-specific signals control reversible program of localiza-
to adult tissue-resident macrophages. Immunity 42, 665–678 (2015). tion and functional polarization of macrophages. Cell 157, 832–844 (2014).
68. Hoeffel, G. et al. Adult Langerhans cells derive predominantly from embryonic fetal 101. Rosas, M. et al. The transcription factor Gata6 links tissue macrophage phenotype and
liver monocytes with a minor contribution of yolk sac–derived macrophages. J. Exp. proliferative renewal. Science 344, 645–648 (2014).
Med. 6, 1167–1181 (2012). 102. Kohyama, M. et al. Role for Spi-C in the development of red pulp macrophages and
69. Guilliams, M. et al. Alveolar macrophages develop from fetal monocytes that dif- splenic iron homeostasis. Nature 457, 318–321 (2009).
ferentiate into long-lived cells in the first week of life via GM-CSF. J. Exp. Med. 210, 103. A-Gonzalez, N. et al. The nuclear receptor LXRa controls the functional specialization
1977–1992 (2013). of splenic macrophages. Nat. Immunol. 14, 831–839 (2013).

8 VOLUME 17 NUMBER 1 JANUARY 2016 NATURE IMMUNOLOGY

You might also like