You are on page 1of 5

Cellular Immunology xxx (xxxx) xxx–xxx

Contents lists available at ScienceDirect

Cellular Immunology
journal homepage: www.elsevier.com/locate/ycimm

Research paper

Alveolar Macrophages

Nikita Joshi, James M. Walter, Alexander V. Misharin
Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, IL, USA

A R T I C L E I N F O A B S T R A C T

Keywords: Alveolar macrophages are the most abundant innate immune cells in the distal lung parenchyma, located on the
Alveolar macrophages luminal surface of the alveolar space. They are the first to encounter incoming pathogens and pollutants and to
Macrophages help orchestrate the initiation and resolution of the immune response in the lung. Similar to other tissue-resident
macrophages, alveolar macrophages also perform non-immune, tissue-specific, homeostatic functions, most
notably clearance of surfactant. In this review we will discuss how ontogeny and local lung environment shape
the role of alveolar macrophages in health and disease.

1. Introduction [5–8]. Instead, they express high levels of integrin CD11c, and sialic
acid-binding lectin Siglec F [5–12]. These markers allow separation of
Alveolar macrophages are the most abundant innate immune cells true long-living alveolar macrophages (i.e. cells that have adapted to
in the distal lung, located on the luminal surface of the alveolar space. the local microenvironment) from transient monocyte-derived cells that
They are the first to encounter incoming pathogens and pollutants and were recruited to the alveolar space during injury [7,10,13–16].
help orchestrate the initiation and resolution of the immune response in Normal human alveolar macrophages express CD206, CD169, CD11c,
the lung. Similar to other tissue-resident macrophages, alveolar mac- CD163 and MARCO [17–21] which are also expressed by alveolar
rophages also perform non-immune, tissue-specific, homeostatic func- macrophages in non-human primates [22]. Alveolar macrophages are
tions, most notably clearance of surfactant. The immune and homeo- known to be highly autofluorescent. Although high autofluorescence
static functions of alveolar macrophages have been recently reviewed may complicate flow cytometric analysis of alveolar macrophages,
[1–4]. Considerable debate has surrounded the origin of these cells: making them to appear falsely positive for a given marker, this feature
while some studies indicated that alveolar macrophages were capable can aid in their identification [23–25].
of proliferation, others suggested that circulating monocytes help Numerous genetic systems exist for tracking and targeting macro-
maintain the alveolar macrophage pool. Recent scientific and technical phages, including alveolar macrophages. These include: simple trans-
advances have substantially improved our understanding of alveolar gene systems [26–28], Cre/lox systems (in particular Cx3cr1, LysM,
macrophage ontogeny. Here we will discuss how ontogeny and the lung Cd11c, Runx1) [13,14,26,28,29], tetracycline-inducible Tet-on systems
microenvironment shape the role of alveolar macrophages in health and [16], bone marrow chimeras including chimeras with thoracic shielding
disease. [5,12,30–32], adoptive transfer of macrophages and their precursors
[8,33,34], and parabiosis and depletion of alveolar macrophages
2. Identification and tracking of alveolar macrophages in the [5,12]. Use of Cre/lox-based systems for targeting macrophages and
laboratory tools for depletion of macrophages, including alveolar macrophages,
has been extensively discussed [35–37]. Importantly, none of these
Exposure to high partial oxygen pressure, surfactant and signals systems are specific for alveolar macrophages and often target other cell
provided by alveolar type I and II cells produce a distinct alveolar types, particularly other myeloid cells (such as monocytes and dendritic
macrophage phenotype which can be identified using multiparameter cells) and in some cases non-immune cells, for example, the CreLysM
flow cytometry. The phenotype of mouse alveolar macrophages is well- system, targets myeloid cells in addition to alveolar type II cells [38].
described: similar to macrophages in other tissues they express MerTK,
CD64, CD68, CD206 and F4/80. However, because of their adaptation
to the unique lung environment, steady state mouse alveolar macro-
phages do not express fractalkine receptor CX3CR1 or integrin CD11b


Corresponding author.
E-mail address: a-misharin@northwestern.edu (A.V. Misharin).

https://doi.org/10.1016/j.cellimm.2018.01.005
Received 10 December 2017; Received in revised form 7 January 2018; Accepted 11 January 2018
0008-8749/ © 2018 Elsevier Inc. All rights reserved.

Please cite this article as: Joshi, N., Cellular Immunology (2018), https://doi.org/10.1016/j.cellimm.2018.01.005
N. Joshi et al. Cellular Immunology xxx (xxxx) xxx–xxx

3. Ontogeny and maintenance of alveolar macrophages in the relatively sessile cells, [27,61] it is possible that during the period of
steady state active postnatal lung growth, alveolar macrophages that populated the
lung soon after birth cannot migrate into newly formed alveoli. Hence,
The origin of alveolar macrophages has long been debated. Studies recruitment of monocytes from the circulation may be necessary in
performed by van Furth over a century ago suggested that circulating order to fully populate the niche [62]. Supporting this hypothesis,
monocytes give rise to all tissue-resident macrophages, including al- Gomez-Pedriguero and colleagues, using a Flt3-Cre reporter system
veolar macrophages at steady state [39,40]. This concept has been re- which labels all cells derived from definitive hematopoiesis, found that
vised during the past decade. Multiple studies have unambiguously at 4 weeks of age, 10–20% of alveolar macrophages were positive for
demonstrated that some prototypical tissue-resident macrophages, such the fluorescent reporter and that the proportion of these cells increased
as microglia in the brain, Kupffer cells in the liver and large peritoneal up to 40% by one year of life [63]. Perhaps stereological techniques
macrophages in the peritoneal cavity, originate from various progeni- combined with new “Microfetti” fate mapping models [64] which can
tors, populate tissues during different stages of embryogenesis and track the clonal expansion of individual macrophages, might help fur-
maintain their population throughout their lifetime with minimal to no ther address the question of alveolar macrophage ontogeny and main-
contribution from circulating monocytes [35,41]. Although several tenance during physiological lung growth.
waves of macrophages populate the lung during embryogenesis and While it is certain that alveolar macrophages are long-lived, current
participate in organogenesis, these do not give rise to the definitive fate mapping studies do not cover the entire lifespan of the animal. It
alveolar macrophage population, since the alveolar niche does not exist was recently demonstrated that normal unperturbed aging is associated
until birth [8,11,28,42]. Thus, the first breath of a newborn may be with decreased number of alveolar macrophages and downregulation of
viewed as the lung’s first “wound”: it creates a niche, which gets rapidly genes involved in cell cycle pathways [65]. Thus, it is possible that in
populated by circulating fetal monocytes. the context of aging, increased environmental stress (such as exposure
The differentiation of recruited monocytes into alveolar macro- to airborne particulate matter over the long period of time) may lead to
phages and subsequent engraftment in the alveolar niche depends on the recruitment of monocyte-derived alveolar macrophages in adults.
the production of granulocyte-macrophage colony-stimulating factor This raises the question: does the origin of alveolar macrophages
(GM-CSF) by alveolar type II cells [8,11]. Loss of GM-CSF signaling, due matter? Several investigators have leveraged powerful genomic tools to
to genetic mutation or development of autoantibodies against GM-CSF help find an answer. Building on work from the ImmGen consortium
or its receptor, results in the loss of mature alveolar macrophages, ac- [6], Lavin and colleagues demonstrated that the transcriptome and
cumulation of surfactant and the development of alveolar proteinosis epigenetic landscape of tissue macrophages are determined by the
[43]. Rescue of GM-CSF signaling by adoptive transfer of alveolar tissue-specific microenvironment [66]. Using bone marrow transfer
macrophages or their precursors with functional GM-CSF receptor or into whole body-irradiated hosts, they found that the lung micro-
administration of exogenous GM-CSF can reverse this condition in environment shapes the epigenetic landscape of bone marrow-derived
mouse models [8,44–48]. In addition, there are reports of successful alveolar macrophages, inducing a convergence with the epigenetic
resolution of alveolar proteinosis after bone marrow transplantation in landscape of tissue-resident alveolar macrophages from naïve mice.
humans [49–51]. In contrast, M-CSF signaling is dispensable for sur- Moreover, upon adoptive transfer into the alveolar space, mature
vival of mature alveolar macrophages [52]. GM-CSF signaling is re- peritoneal macrophages adapt to the niche, rapidly reshape their
quired for the expression of peroxisome proliferator-activated receptor transcriptome and become somewhat alveolar macrophage-like cells. In
gamma (PPARG) – a key transcription factor necessary for realization of another study, Gibbings and colleagues used congenic (CD45.1/
the transcriptional program driving macrophage adaptation to the lung CD45.2) bone marrow chimeras with thoracic shielding to protect the
environment and acquisition of a high lipid load [6,8,53–55]. PPARG recipient’s tissue-resident alveolar macrophages (a system originally
deletion in myeloid lineage prevented formation of mature alveolar introduced by Janssen and colleagues [32]), followed by partial de-
macrophages, which resulted in delayed bacterial clearance after in- pletion of these cells using clodronate-loaded liposomes, to generate a
fection with S. pneumoniae and resulted in alveolar proteinosis. Other mouse that harbors alveolar macrophages of embryonic and adult
transcriptional factors, such as Bach2 and C/EBPβ are also required for origin simultaneously [12]. Upon opening the niche, monocytes entered
development and maintenance of alveolar macrophages [56,57]. L- the uninjured alveolar space and differentiated into cells which were
plastin is another protein that is required for the transmigration and virtually indistinguishable from tissue-resident alveolar macrophages
retention of alveolar macrophage precursors into the alveolar niche: both immunophenotypically and functionally. They further isolated
mice deficient for the Lcp1 gene in myeloid cells fail to develop func- tissue-resident and monocyte-derived alveolar macrophages via FAC-
tional alveolar macrophages and have impaired clearance of pulmonary Sorting and subjected them to transcriptomic profiling via microarray.
pathogens [42]. Recently, a crucial role of autocrine TGFβ signaling in Only 35 probes were differentially expressed between tissue-resident
development and maintenance of alveolar macrophages has been also and monocyte-derived cells. Similarly, van de Laar and colleagues de-
demonstrated [58]. monstrated that upon adoptive transfer into a permissive niche, yolk
Using various fate-mapping techniques, several groups have de- sac macrophages, fetal monocytes and adult bone marrow monocytes
monstrated that in naïve, unchallenged adult mice housed in clean fa- all gave rise to cells that were functionally and transcriptionally in-
cilities, alveolar macrophages, microglia in the brain, Kupffer cells in distinguishable from tissue-resident alveolar macrophages derived from
the liver and large peritoneal macrophages maintain their population fetal monocytes [33]. Together, these studies clearly demonstrate that
via proliferation in situ for months without contribution from circu- in the unperturbed lung, at steady state, the origin of alveolar macro-
lating monocytes [5,8,14,26,29,32]. Supporting these observations phages does not matter, as cells of fetal and adult origin as well as
made in mice, two recent clinical reports demonstrated that following differentiated alveolar macrophages can occupy the permissive lung
lung transplant, the population of alveolar macrophages is remarkably niche and give rise to fully functional, long-living, self-maintaining al-
stable with donor cells comprising the bulk of the alveolar macrophage veolar macrophages, which are virtually identical at the phenotypic,
pool even 5 years after transplant [59,60]. However, it is not known functional, genomic and epigenomic levels.
whether the initial seeding of alveolar macrophages into the lung
during the postnatal period occurs as a single wave with subsequent 4. Origin and function of alveolar macrophages during aging,
expansion via local proliferation, or if additional alveolar macrophages stress and lung injury
derived from adult bone marrow-derived monocytes contribute to the
alveolar macrophage pool during the rapid period of physiological body The aforementioned studies show that lung environment, and not
growth and lung niche expansion. Since alveolar macrophages are cell ontogeny (embryonic versus adult), is a major driver of alveolar

2
N. Joshi et al. Cellular Immunology xxx (xxxx) xxx–xxx

macrophage development. However, it should be noted, that all studies the alveolar niche and differentiated into alveolar macrophages. No-
discussed above were performed under steady state conditions in the tably, the deletion of monocyte-derived alveolar macrophages led to the
unperturbed lung. This raises two questions: 1) does the environment of amelioration of pulmonary fibrosis, highlighting a crucial role of these
an injured lung alter differentiation of newly recruited monocytes into cells in disease pathogenesis. Combined deletion of Casp8 and Ripk3
alveolar macrophages and 2) does the environment reshape tissue-re- prevented necroptosis of monocyte-derived alveolar macrophages and
sident alveolar macrophages that were present in the lung before lung “rescued” the fibrotic phenotype. Gene expression profiling of FAC-
injury? Several studies have attempted to answer this question using Sorted monocytes, monocyte-derived interstitial macrophages, mono-
various models of lung injury coupled with fate mapping systems to cyte-derived alveolar macrophages and tissue-resident alveolar mac-
track tissue-resident and monocyte-derived alveolar macrophages. rophages demonstrated distinct transcriptomes for all cell types despite
Janssen and colleagues used bone marrow chimeras with thoracic their prolonged coexistence in the same environment. Moreover, only
shielding and adoptive transfer of bone marrow from pan-GFP mice monocyte-derived alveolar macrophages were enriched for genes pre-
[32]. They were one of the first groups to demonstrate the remarkable viously associated with development of fibrosis in various organs. Fi-
longevity and stability of tissue-resident alveolar macrophages: 80% of nally, since bleomycin-induced pulmonary fibrosis resolves sponta-
alveolar macrophages were found to be of host origin up to 240 days neously over time, the authors asked whether these recruited
after the generation of chimeras. Using intratracheal instillation of monocyte-derived alveolar macrophages persist in the lung after injury
bacterial lipopolysaccharide (LPS) – a popular model of lung injury – resolution. Using bone marrow chimeras with thoracic shielding to
they demonstrated that the numbers of tissue-resident alveolar mac- unambiguously track tissue-resident and monocyte-derived cells in two
rophages did not change over the course of injury. Thus, the alveolar models of lung injury (bleomycin- and influenza A virus-mediated),
niche remained occupied by tissue-resident alveolar macrophages and they demonstrated that monocyte-derived alveolar macrophages persist
was not permissive for engraftment of recruited cells. They also found for at least 10 months after the resolution of initial injury at which time
that while most of the recruited macrophages underwent apoptosis they are phenotypically and transcriptionally indistinguishable from
during the resolution phase, some of the recruited monocyte-derived tissue-resident alveolar macrophages.
macrophages persisted for up to 12 days after injury. Moreover, some of Similarly, McCubbrey and colleagues used a previously validated
these cells obtained a surface phenotype similar to tissue-resident al- tetracycline-inducible hCD68rtTA system [16] to delete c-FLIP from
veolar macrophages, characterized by the absence of CD11b expression monocyte-derived alveolar macrophages making them susceptible to
and increased expression of CD11c. Recently, this group performed an apoptosis [13]. They demonstrated that deletion of monocyte-derived
in-depth characterization of tissue-resident alveolar macrophages and alveolar macrophages ameliorated the development of pulmonary fi-
recruited monocyte-derived macrophages using gene expression pro- brosis. Analysis of gene expression profiles of FACSorted tissue-resident
filing via RNA-seq during the course of LPS-induced lung injury [15]. and monocyte-derived alveolar macrophages showed that these cells
They found that tissue-resident alveolar macrophages and recruited exhibit markedly different gene expression profiles with only mono-
monocyte-derived macrophages exhibited strikingly different gene ex- cyte-derived alveolar macrophages exhibiting a profibrotic gene sig-
pression profiles during the acute phase of lung injury and concluded nature. Together, these studies demonstrate that tissue-resident and
that cell origin dictates macrophage programming in this setting. It monocyte-derived alveolar macrophages differ in their capacity to be
must be noted, however, that the focus of this study was on acute lung programmed by their environment and that this programming has a
injury, and that while recruited macrophages were recovered from the distinct impact on their function, resulting in clinically apparent phe-
alveolar space, they were not bona fide alveolar macrophages (i.e. cells notypes.
adapted for long-term persistence in the niche), as evidenced by a lack The recruitment of Siglec Flow monocyte-derived alveolar macro-
of an alveolar macrophage-specific phenotype (CD169, CD206, Siglec phages has also been observed in mouse models of asthma [69,70].
F). Importantly, since LPS is a spontaneously resolving model of lung Sanfilippo and colleagues demonstrated that the recruitment and per-
injury, the physiological significance of the differences in expression sistence of monocyte-derived Siglec Flow alveolar macrophages during
between the two cell types is unclear. allergic inflammation protects against pulmonary pneumococcal in-
Long-term models of lung injury may provide more informative fection [69]. Allergic inflammation was associated with enhanced early
platforms to investigate the differential contribution of tissue-resident clearance of S. pneumoniae from the lung, decreased bacterial invasion
and recruited monocyte-derived alveolar macrophages. Recently, two from the airway into the lung tissue, a blunted inflammatory cytokine
independent studies utilized a bleomycin model of lung injury and fi- and neutrophil response to infection, and enhanced expression of TGF-
brosis to identify distinct contributions of tissue-resident and monocyte- β1. Non-selective depletion of alveolar macrophages using clodronate-
derived alveolar macrophages to fibrosis pathogenesis [13,14]. loaded liposomes abrogated this effect.
Bleomycin-induced models of lung fibrosis are characterized by an In a reverse system, Machiels and colleagues subjected mice to
early inflammatory phase, followed by a fibrotic response [67]. During gammaherpesvirus infection and then induced allergic airway in-
the early phase of injury, there is an influx of monocytes and monocyte- flammation using a house dust mite model [71]. They found that pre-
derived interstitial macrophages while the fibrotic phase is character- vious infection with murid herpesvirus 4 inhibits subsequent house dust
ized by an expansion of alveolar macrophages with low expression of mite-induced experimental asthma. In a series of experiments, they
the surface marker Siglec F [7]. Using bone marrow chimeras with attributed this protective effect to alveolar macrophages. Using bone
thoracic shielding the same group has later demonstrated that during marrow chimeras with thoracic shielding, they demonstrated that
the first weeks following lung injury, tissue-resident and monocyte- murid herpesvirus 4 infection depletes alveolar macrophages and in-
derived alveolar macrophages can be distinguished based on the dif- duces monocyte recruitment. Over time these recruited monocytes
ferential expression of Siglec F: tissue-resident alveolar macrophages differentiated into long-lived alveolar macrophages. In agreement with
maintained high expression of Siglec F, whereas monocyte-derived al- previous reports, these recruited alveolar macrophages also expressed
veolar macrophages expressed lower levels of this molecule [14]. They low levels of Siglec F. The authors also performed gene expression
then used two overlapping Cre-drivers, namely LysMCre and CD11cCre, to profiling of the bulk alveolar macrophage population and found that
delete pulmonary macrophages by removing caspase 8 and inducing herpesvirus 4 and house dust mite challenge, alone or in combination,
necroptosis via the unchecked RIPK3 pathway [68]. Since it takes time produced significant changes in the gene expression profiles of alveolar
for the effects of Cre-mediated deletion to take place, circulating macrophages.
monocytes or monocyte-derived interstitial macrophages were not af- Collectively, these studies demonstrate that the environment of the
fected in this system. Using bone marrow transfer, the authors found injured lung can reshape the transcriptome of both monocyte-derived
that only cells that escaped Cre-mediated recombination reconstituted and tissue-resident alveolar macrophages. It is not yet known whether

3
N. Joshi et al. Cellular Immunology xxx (xxxx) xxx–xxx

less acutely injurious, but chronic stimuli produce a similar effect. For type II cells.
example, the lung is constantly exposed to air pollutants and ambient
particular matter – a serious public health concern in both the devel- Acknowledgments
oping and developed world [72,73]. Exposure to ambient particular
matter does not result in acute lung injury and recruitment of mono- James M Walter is supported by Northwestern University’s Lung
cyte-derived alveolar macrophages. However, in response to particulate Sciences Training Program 5T32HL076139-14 and Northwestern
matter exposure, alveolar macrophages produce IL-6, which leads to University’s Dixon Young Investigator Translational Research Grant.
activation of the coagulation cascade in the liver and increased risk of Alexander V Misharin is supported by NIH NHLBI 1R56HL135124-01,
thrombosis [74,75]. There is evidence that exposure to particulate BD Bioscience Immunology Research Grant and by the Office of the
matter can modify the epigenetic landscape of macrophages in vitro Assistant Secretary of Defense for Health Affairs, through the Peer
[76,77]. While detailed epigenetic profiling of alveolar macrophages Reviewed Medical Research Program under Award W81XWH-15-1-
after long-term exposure has not yet been reported, it is tempting to 0215. Opinions, interpretations, conclusions and recommendations are
speculate that epigenetic changes may also modulate macrophage re- those of the author and are not necessarily endorsed by the Department
sponse, IL-6 production and subsequent risk of thrombosis. of Defense.
As discussed above, both tissue-resident and monocyte-derived al-
veolar macrophages respond to changes in the local environment References
during lung injury. However, quantitative and qualitative changes in
the transcriptome of monocyte-derived alveolar macrophages are more [1] T. Hussell, T.J. Bell, Alveolar macrophages: plasticity in a tissue-specific context,
prominent than in tissue-resident alveolar macrophages. Nevertheless, Nat. Rev. Immunol. 14 (2) (2014) 81–93.
[2] E. Gwyer Findlay, T. Hussell, Macrophage-mediated inflammation and disease: a
upon resolution of lung injury, alveolar macrophages, irrespective of focus on the lung, Mediators Inflam. 2012 (2012) 140937.
their origin, become increasingly similar, as evident by convergence of [3] A.J. Byrne, S.A. Mathie, L.G. Gregory, C.M. Lloyd, Pulmonary macrophages: key
their transcriptomes. Despite these similarities, tissue-resident and players in the innate defence of the airways, Thorax 70 (12) (2015) 1189–1196.
[4] N. Garbi, B.N. Lambrecht, Location, function, and ontogeny of pulmonary macro-
monocyte-derived macrophages may respond differently to a sub- phages during the steady state, Pflugers Arch. 469 (3–4) (2017) 561–572.
sequent stimulus. Recent studies have shown that monocyte to macro- [5] S.L. Gibbings, S.M. Thomas, S.M. Atif, et al., Three unique interstitial macrophages
phage differentiation is associated with a global reshaping of the epi- in the murine lung at steady state, Am. J. Respir. Cell Mol.Biol. (2017).
[6] E.L. Gautier, T. Shay, J. Miller, et al., Gene-expression profiles and transcriptional
genetic landscape [78,79]. Moreover, previous exposure of monocytes
regulatory pathways that underlie the identity and diversity of mouse tissue mac-
to stimulus can leave long-lasting epigenetic marks, which can be re- rophages, NatureImmunology 13 (11) (2012) 1118–1128.
tained upon their differentiation into macrophages. Depending on the [7] A.V. Misharin, L. Morales-Nebreda, G.M. Mutlu, G.R. Budinger, H. Perlman, Flow
cytometric analysis of macrophages and dendritic cell subsets in the mouse lung,
type of stimulus, this may result in the development of a trained im-
Am. J. Respir. Cell Mol.Biol. 49 (4) (2013) 503–510.
munity, characterized by enhanced inflammatory status, or tolerance, [8] M. Guilliams, I. De Kleer, S. Henri, et al., Alveolar macrophages develop from fetal
characterized by a blunted inflammatory response [78]. Recently, monocytes that differentiate into long-lived cells in the first week of life via GM-
Schmidt and colleagues demonstrated that in vitro differentiated in- CSF, J. Exp. Med. 210 (10) (2013) 1977–1992.
[9] M. Guilliams, C.-A. Dutertre, L. Scott Charlotte, et al., Unsupervised high-dimen-
flammatory macrophages are characterized by a unique network of sional analysis aligns dendritic cells across tissues and species, Immunity (2016).
transcriptional and epigenetic regulators, which respond to activation [10] Y.R. Yu, E.G. O'Koren, D.F. Hotten, et al., A protocol for the comprehensive flow
in a stimulus-specific manner [80]. This “inflammation specific” net- cytometric analysis of immune cells in normal and inflamed murine non-lymphoid
tissues, PloS One 11 (3) (2016) e0150606.
work was characterized by globally permissive histone modifications. [11] C. Schneider, S.P. Nobs, M. Kurrer, H. Rehrauer, C. Thiele, M. Kopf, Induction of the
While the authors did not investigate the response of tissue-resident nuclear receptor PPAR-gamma by the cytokine GM-CSF is critical for the differ-
macrophages to stimulation directly, they obtained contrasting data entiation of fetal monocytes into alveolar macrophages, NatureImmunology 15 (11)
(2014) 1026–1037.
from a re-analysis of a previously published dataset [66]. Tissue-re- [12] S.L. Gibbings, R. Goyal, A.N. Desch, et al., Transcriptome analysis highlights the
sident macrophages from naïve animals, including alveolar macro- conserved difference between embryonic and postnatal-derived alveolar macro-
phages, were characterized not only by the absence of expression of phages, Blood 126 (11) (2015) 1357–1366.
[13] A.L. McCubbrey, L. Barthel, M.P. Mohning, et al., Deletion of c-FLIP from CD11bhi
inflammatory genes, but also by the presence of repressive chromatin macrophages prevents development of bleomycin-induced lung fibrosis, Am. J.
marks. However, it is still possible that inflammation can reshape the Respir. Cell Mol. Biol. (2017).
epigenetic landscape of differentiated macrophages and activate latent [14] A.V. Misharin, L. Morales-Nebreda, P.A. Reyfman, et al., Monocyte-derived alveolar
macrophages drive lung fibrosis and persist in the lung over the life span, J. Exp.
enhancers in differentiated cells or reshape epigenetic changes caused
Med. (2017).
by a previous stimulation [81,82]. Unfortunately, all these experiments [15] K.J. Mould, L. Barthel, M.P. Mohning, et al., Cell origin dictates programming of
were performed in vitro in very simplistic and short-term models. Ulti- resident versus recruited macrophages during acute lung injury, Am. J. Respir. Cell
mately, the validity and significance of “the second hit” hypothesis Mol. Biol. (2017).
[16] A.L. McCubbrey, L. Barthel, K.J. Mould, M.P. Mohning, E.F. Redente, W.J. Janssen,
needs to be tested in long-term animal models with measurable and Selective and Inducible Targeting of CD11b+ Mononuclear Phagocytes in the
clinically relevant physiological outcome. Murine Lung with hCD68-rtTA Transgenic Systems, Am. J. Physiol. Lung Cellular
Mol. Physiol., 2016:ajplung 00141 02016.
[17] A. Bharat, S.M. Bhorade, L. Morales-Nebreda, et al., Flow cytometry reveals simi-
5. Conclusions and open questions larities between lung macrophages in humans and mice, Am. J. Respir. Cell Mol.
Biol. 54 (1) (2016) 147–149.
Our understanding of macrophage biology has improved dramati- [18] Y.R. Yu, D.F. Hotten, Y. Malakhau, et al., Flow cytometric analysis of myeloid cells
in human blood, bronchoalveolar lavage, and lung tissues, Am. J. Respir. Cell Mol.
cally in the past decade. Irrespective of their origin, macrophages, in- Biol. 54 (1) (2016) 13–24.
cluding alveolar macrophages, demonstrate remarkable plasticity to [19] A.N. Desch, S.L. Gibbings, R. Goyal, et al., Flow cytometric analysis of mononuclear
their surrounding environment. The environment of the injured lung phagocytes in nondiseased human lung and lung-draining lymph nodes, Am. J.
Respir. Crit. Care Med. 193 (6) (2016) 614–626.
can reshape alveolar macrophages from protective and homeostatic [20] V.I. Patel, J.L. Booth, E.S. Duggan, et al., Transcriptional classification and func-
cells into pathogenic cells. Better understanding of the mechanisms tional characterization of human airway macrophage and dendritic cell subsets, J.
through which inflammatory and environmental insults affect alveolar Immunol. (2016).
[21] F. Baharom, S. Thomas, G. Rankin, et al., Dendritic cells and monocytes with dis-
macrophage function may guide development of novel therapies for
tinct inflammatory responses reside in lung mucosa of healthy humans, J. Immunol.
pulmonary diseases with macrophage involvement. The importance of 196 (11) (2016) 4498–4509.
the environment in guiding alveolar macrophage development and [22] Y. Cai, C. Sugimoto, M. Arainga, X. Alvarez, E.S. Didier, M.J. Kuroda, In vivo
function suggest that future research should take a systems biology characterization of alveolar and interstitial lung macrophages in rhesus macaques:
implications for understanding lung disease in humans, J. Immunol. 192 (6) (2014)
approach and investigate the function of alveolar macrophages in the 2821–2829.
context of their partnering cell types, in particular alveolar type I and

4
N. Joshi et al. Cellular Immunology xxx (xxxx) xxx–xxx

[23] A.J. Mitchell, L.C. Pradel, L. Chasson, et al., Technical advance: autofluorescence as macrophages, Biochem. Biophys. Res. Commun. 393 (4) (2010) 682–687.
a tool for myeloid cell analysis, J. Leukocyte Biol. 88 (3) (2010) 597–603. [55] H. Dalrymple, B.P. Barna, A. Malur, A.G. Malur, M.S. Kavuru, M.J. Thomassen,
[24] M. Duan, W.C. Li, R. Vlahos, M.J. Maxwell, G.P. Anderson, M.L. Hibbs, Distinct Alveolar macrophages of GM-CSF knockout mice exhibit mixed M1 and M2 phe-
macrophage subpopulations characterize acute infection and chronic inflammatory notypes, BMC Immunol. 14 (2013) 41.
lung disease, J. Immunol. 189 (2) (2012) 946–955. [56] A. Nakamura, R. Ebina-Shibuya, A. Itoh-Nakadai, et al., Transcription repressor
[25] U. Maus, S. Rosseau, W. Seeger, J. Lohmeyer, Separation of human alveolar mac- Bach2 is required for pulmonary surfactant homeostasis and alveolar macrophage
rophages by flow cytometry, Am. J. Physiol. 272 (3 Pt 1) (1997) L566–571. function, J. Exp. Med. 210 (11) (2013) 2191–2204.
[26] S. Yona, K.W. Kim, Y. Wolf, et al., Fate mapping reveals origins and dynamics of [57] D.W. Cain, E.G. O'Koren, M.J. Kan, et al., Identification of a tissue-specific, C/
monocytes and tissue macrophages under homeostasis, Immunity 38 (1) (2013) EBPbeta-dependent pathway of differentiation for murine peritoneal macrophages,
79–91. J. Immunol. 191 (9) (2013) 4665–4675.
[27] M.P. Rodero, L. Poupel, P.L. Loyher, et al., Immune surveillance of the lung by [58] X. Yu, A. Buttgereit, I. Lelios, et al., The cytokine TGF-β promotes the development
migrating tissue monocytes, Elife 4 (2015) e07847. and homeostasis of alveolar macrophages, Immunity (2017).
[28] S.Y. Tan, M.A. Krasnow, Developmental origin of lung macrophage diversity, [59] D.K. Nayak, F. Zhou, M. Xu, et al., Long-term persistence of donor alveolar mac-
Development 143 (8) (2016) 1318–1327. rophages in human lung transplant recipients that influences donor specific im-
[29] D. Hashimoto, A. Chow, C. Noizat, et al., Tissue-resident macrophages self-maintain mune responses, Am. J. Transpl. Official J. Am. Soc. Transpl. Am. Soc. Transpl.
locally throughout adult life with minimal contribution from circulating monocytes, Surg. (2016).
Immunity 38 (4) (2013) 792–804. [60] I. Eguiluz-Gracia, H.H. Schultz, L.I. Sikkeland, et al., Long-term persistence of
[30] L. Landsman, S. Jung, Lung macrophages serve as obligatory intermediate between human donor alveolar macrophages in lung transplant recipients, Thorax (2016).
blood monocytes and alveolar macrophages, J. Immunol. 179 (6) (2007) [61] K. Westphalen, G.A. Gusarova, M.N. Islam, et al., Sessile alveolar macrophages
3488–3494. communicate with alveolar epithelium to modulate immunity, Nature 506 (7489)
[31] L. Landsman, C. Varol, S. Jung, Distinct differentiation potential of blood monocyte (2014) 503–506.
subsets in the lung, J Immunol. 178 (4) (2007) 2000–2007. [62] M. Guilliams, C.L. Scott, Does niche competition determine the origin of tissue-
[32] W.J. Janssen, L. Barthel, A. Muldrow, et al., Fas determines differential fates of resident macrophages? Nat. Rev. Immunol. 17 (7) (2017) 451–460.
resident and recruited macrophages during resolution of acute lung injury, Am. J. [63] E. Gomez Perdiguero, K. Klapproth, C. Schulz, et al., Tissue-resident macrophages
Resp. Crit. Care Med. 184 (5) (2011) 547–560. originate from yolk-sac-derived erythro-myeloid progenitors, Nature 518 (7540)
[33] L. van de Laar, W. Saelens, S. De Prijck, et al., Yolk Sac macrophages, fetal liver, and (2015) 547–551.
adult monocytes can colonize an empty niche and develop into functional tissue- [64] T.L. Tay, D. Mai, J. Dautzenberg, et al., A new fate mapping system reveals context-
resident macrophages, Immunity 44 (4) (2016) 755–768. dependent random or clonal expansion of microglia, Nat. Neurosci. 20 (6) (2017)
[34] M.A. Gibbons, A.C. MacKinnon, P. Ramachandran, et al., Ly6Chi monocytes direct 793–803.
alternatively activated profibrotic macrophage regulation of lung fibrosis, Am. J. [65] C.K. Wong, C.A. Smith, K. Sakamoto, N. Kaminski, J.L. Koff, D.R. Goldstein, Aging
Respir. Crit. Care Med. 184 (5) (2011) 569–581. impairs alveolar macrophage phagocytosis and increases influenza-induced mor-
[35] S. Epelman, K.J. Lavine, G.J. Randolph, Origin and functions of tissue macrophages, tality in mice, J. Immunol. (2017).
Immunity 41 (1) (2014) 21–35. [66] Y. Lavin, D. Winter, R. Blecher-Gonen, et al., Tissue-resident macrophage enhancer
[36] D. Hashimoto, J. Miller, M. Merad, Dendritic cell and macrophage heterogeneity in landscapes are shaped by the local microenvironment, Cell 159 (6) (2014)
vivo, Immunity 35 (3) (2011) 323–335. 1312–1326.
[37] A.L. McCubbrey, K.C. Allison, A.B. Lee-Sherick, C.V. Jakubzick, W.J. Janssen, [67] B. Moore, W.E. Lawson, T.D. Oury, T.H. Sisson, K. Raghavendran, C.M. Hogaboam,
Promoter specificity and efficacy in conditional and inducible transgenic targeting Animal models of fibrotic lung disease, Am. J. Respir. Cell Mol. Biol. 49 (2) (2013)
of lung macrophages, Front. Immunol. 8 (2017) 1618. 167–179.
[38] T.J. Desai, D.G. Brownfield, M.A. Krasnow, Alveolar progenitor and stem cells in [68] C.M. Cuda, A.V. Misharin, S. Khare, et al., Conditional deletion of caspase-8 in
lung development, renewal and cancer, Nature 507 (7491) (2014) 190–194. macrophages alters macrophage activation in a RIPK-dependent manner, Arthritis
[39] R. van Furth, Z.A. Cohn, The origin and kinetics of mononuclear phagocytes, J. Exp. Res. Ther. 17 (2015) 291.
Med. 128 (3) (1968) 415–435. [69] A.M. Sanfilippo, Y. Furuya, S. Roberts, S.L. Salmon, D.W. Metzger, Allergic lung
[40] R. van Furth, Macrophage activity and clinical immunology. Origin and kinetics of inflammation reduces tissue invasion and enhances survival from pulmonary
mononuclear phagocytes, Ann. N. Y. Acad. Sci. 278 (1976) 161–175. pneumococcal infection in mice, which correlates with increased expression of
[41] F. Ginhoux, S. Jung, Monocytes and macrophages: developmental pathways and transforming growth factor beta1 and SiglecF(low) alveolar macrophages, Infection
tissue homeostasis, Nat. Rev. Immunol. 14 (6) (2014) 392–404. Immun. 83 (7) (2015) 2976–2983.
[42] E.M. Todd, J.Y. Zhou, T.P. Szasz, et al., Alveolar macrophage development in mice [70] H. Abdala Valencia, L.F. Loffredo, A.V. Misharin, S. Berdnikovs, Phenotypic plas-
requires L-plastin for cellular localization and retention within alveoli, Blood ticity and targeting of Siglec-F(high) CD11c(low) eosinophils to the airway in a
(2016). murine model of asthma, Allergy 71 (2) (2016) 267–271.
[43] R. Borie, C. Danel, M.P. Debray, et al., Pulmonary alveolar proteinosis, Eur Respir [71] B. Machiels, M. Dourcy, X. Xiao, et al., A gammaherpesvirus provides protection
Rev. 20 (120) (2011) 98–107. against allergic asthma by inducing the replacement of resident alveolar macro-
[44] C. Happle, N. Lachmann, J. Skuljec, et al., Pulmonary transplantation of macro- phages with regulatory monocytes, Nat. Immunol. (2017).
phage progenitors as effective and long-lasting therapy for hereditary pulmonary [72] C.A. Pope 3rd, M. Ezzati, D.W. Dockery, Fine-particulate air pollution and life ex-
alveolar proteinosis, Sci. Transl. Med. 6 (250) (2014) 250ra113. pectancy in the United States, New England J. Med. 360 (4) (2009) 376–386.
[45] T. Suzuki, P. Arumugam, T. Sakagami, et al., Pulmonary macrophage transplanta- [73] Q. Di, Y. Wang, A. Zanobetti, et al., Air pollution and mortality in the medicare
tion therapy, Nature 514 (7523) (2014) 450–454. population, New England J. Med. 376 (26) (2017) 2513–2522.
[46] K. Takata, T. Kozaki, C.Z.W. Lee, et al., Induced-pluripotent-stem-cell-derived pri- [74] D.Q. Rich, H.M. Kipen, W. Huang, et al., Association between changes in air pol-
mitive macrophages provide a platform for modeling tissue-resident macrophage lution levels during the Beijing Olympics and biomarkers of inflammation and
differentiation and function, Immunity 47 (1) (2017) 183–198 e186. thrombosis in healthy young adults, JAMA: J. Am. Med. Assoc. 307 (19) (2012)
[47] A. Rongvaux, T. Willinger, J. Martinek, et al., Development and function of human 2068–2078.
innate immune cells in a humanized mouse model, Nat. Biotechnol. 32 (4) (2014) [75] S.E. Chiarella, S. Soberanes, D. Urich, et al., beta(2)-Adrenergic agonists augment
364–372. air pollution-induced IL-6 release and thrombosis, J. Clin. Investig. 124 (7) (2014)
[48] T. Willinger, A. Rongvaux, H. Takizawa, et al., Human IL-3/GM-CSF knock-in mice 2935–2946.
support human alveolar macrophage development and human immune responses in [76] I.R. Miousse, M.C. Chalbot, N. Aykin-Burns, et al., Epigenetic alterations induced by
the lung, PNAS 108 (6) (2011) 2390–2395. ambient particulate matter in mouse macrophages, Environ. Mol. Mutagen. 55 (5)
[49] S. Tabata, S. Shimoji, K. Murase, et al., Successful allogeneic bone marrow trans- (2014) 428–435.
plantation for myelodysplastic syndrome complicated by severe pulmonary alveolar [77] I.R. Miousse, M.C. Chalbot, R. Pathak, et al., In vitro toxicity and epigenotoxicity of
proteinosis, Int. J. Hematol. 90 (3) (2009) 407–412. different types of ambient particulate matter, Toxicol Sci. 148 (2) (2015) 473–487.
[50] K. Fukuno, A. Tomonari, N. Tsukada, et al., Successful cord blood transplantation [78] S. Saeed, J. Quintin, H.H. Kerstens, et al., Epigenetic programming of monocyte-to-
for myelodysplastic syndrome resulting in resolution of pulmonary alveolar pro- macrophage differentiation and trained innate immunity, Science 345 (6204)
teinosis, Bone Marrow Transplant. 38 (8) (2006) 581–582. (2014) 1251086.
[51] A. Numata, E. Matsuishi, K. Koyanagi, et al., Successful therapy with whole-lung [79] T.H. Pham, C. Benner, M. Lichtinger, et al., Dynamic epigenetic enhancer signatures
lavage and autologous peripheral blood stem cell transplantation for pulmonary reveal key transcription factors associated with monocytic differentiation states,
alveolar proteinosis complicating acute myelogenous leukemia, Am. J. Hematol. 81 Blood 119 (24) (2012) e161–171.
(2) (2006) 107–109. [80] S.V. Schmidt, W. Krebs, T. Ulas, et al., The transcriptional regulator network of
[52] K.A. Sauter, C. Pridans, A. Sehgal, et al., Pleiotropic effects of extended blockade of human inflammatory macrophages is defined by open chromatin, Cell Res. 26 (2)
CSF1R signaling in adult mice, J. Leukocyte Biol. 96 (2) (2014) 265–274. (2016) 151–170.
[53] E.L. Gautier, A. Chow, R. Spanbroek, et al., Systemic analysis of PPARgamma in [81] R. Ostuni, V. Piccolo, I. Barozzi, et al., Latent enhancers activated by stimulation in
mouse macrophage populations reveals marked diversity in expression with critical differentiated cells, Cell 152 (1–2) (2013) 157–171.
roles in resolution of inflammation and airway immunity, J. Immunol. 189 (5) [82] V. Piccolo, A. Curina, M. Genua, et al., Opposing macrophage polarization programs
(2012) 2614–2624. show extensive epigenomic and transcriptional cross-talk, Nat. Immunol. 18 (5)
[54] A.D. Baker, A. Malur, B.P. Barna, M.S. Kavuru, A.G. Malur, M.J. Thomassen, (2017) 530–540.
PPARgamma regulates the expression of cholesterol metabolism genes in alveolar

You might also like