Summary
Monocytes are circulating myeloid cells that are derived from dedicated progenitors in the bone marrow. Originally thought of as mere precursors for the replacement of tissue macrophages, it is increasingly clear that monocytes execute distinct effector functions and may give rise to monocyte-derived cells with unique properties from tissue-resident macrophages. Recently, the advent of novel experimental approaches such as single-cell analysis and fate-mapping tools has uncovered an astonishing display of monocyte plasticity and heterogeneity, which we believe has emerged as a key theme in the field of monocyte biology in the last decade. Monocyte heterogeneity is now recognized to develop as early as the progenitor stage through specific imprinting mechanisms, giving rise to specialized effector cells in the tissue. At the same time, monocytes must overcome their susceptibility towards cellular death to persist as monocyte-derived cells in the tissues. Environmental signals that preserve their heterogenic phenotypes and govern their eventual fates remain incompletely understood. In this review, we will summarize recent advances on the developmental trajectory of monocytes and discuss emerging concepts that contributes to the burgeoning field of monocyte plasticity and heterogeneity.
Keywords: monocyte heterogeneity, monocyte-derived cells, macrophages, bone marrow progenitors and precursors, monopoiesis
Graphical Abstract
Graphical Abstract.
Introduction
Monocytes were first discovered in human white blood cells as large mononucleated leukocytes with kidney-shaped nuclei by Paul Ehrlich in 1880 [1]. Originally thought of as phagocytic cells [2] or mere precursors for macrophages [3], these monocyte concepts have been significantly transformed in distinct settings in the last few decades. In particular, it is now well established that tissue-resident macrophages (TRMs) and conventional dendritic cells (cDCs) have defined precursors with the former originating in the embryo before the onset of bone marrow (BM) haematopoiesis [4–6]. Consequently, these cells were found to maintain themselves independently from incoming monocytes and are only replaced by the latter through specific tissue cues [7]. Monocytes constitute ~10% and ~4% of peripheral leukocytes in humans and mice respectively [7]. They can be categorized broadly as ‘classical’ monocytes, exemplified by CD14+CD16+ expression in humans and Ly6ChiCX3CR1intCCR2+CD43lo in mice; and ‘non-classical’ monocytes, depicted as CD14loCD16+ expression in humans and Ly6CloCX3CR1hiCCR2loCD43+ in mice [7, 8]. In humans, CD88 and CD89 further delineate monocytes from circulating inflammatory DCs in the blood [9]. Importantly, the advent of novel technologies has uncovered an astonishing display of monocyte plasticity and heterogeneity, which we believe has surfaced as one of the essential themes in the field of monocyte biology in recent years. Emerging evidence suggests that monocytes can be imprinted as early as in the progenitor or precursor stage by the tissue microenvironment to adopt specialized roles that alter the fate of immune responses, revealing a previously under-appreciated complexity in their plasticity [10]. At the same time, monocytes are also recognized as short-lived cells [10]. Consequently, how the immune system ensures a continuity of imprinted or ‘trained’ functional responses beyond the monocyte stage will rely on tissue/niche-specific signals that prolong their viability and function upon their differentiation. In this article, we will present an overview of the developmental trajectory of monocytes (Fig. 1) and focus our discussion on the latest findings of how tissue and cellular signals imprint and alter their eventual fates. Specifically, we will review current evidence of monocyte phenotypes, revisit competing dogmas, and discuss how the ontogeny of these phenotypes is unique to distinct disease settings. Finally, we will explore how these concepts contribute together towards monocyte-derived heterogeneity in tissues and offer perspectives on their transitional states.
Figure 1:
Emerging complexity in the expanding monocyte universe. Monocyte heterogeneity is now recognized to occur at distinct stages of development and locations. During the steady-state, classical Ly6Chi monocytes may originate from two distinct BM progenitors (GMP or MDP) and differentiate along a continuum to give rise to either non-classical Ly6Clo monocytes in the endothelium or TRMs in organs that require TRM replenishment. During inflammatory states, GMPs or MDPs may be primed in the BM to give rise to unique monocyte subsets with different functional states. Monocytes and their precursors/progenitors may also undergo additional modifications upon entering the circulation or tissues, resulting in further complexity in their heterogeneity. The list of monocyte subsets and monocyte-derived cells is not exhaustive but is illustrated to the best of our knowledge to depict the emerging complexity of monocyte heterogeneity.
Monocyte development: Different fates before birth
Fetal versus Adult monocytes
The earliest form of monocytes begins its development in the embryo and are derived from erythro-myeloid progenitors that migrate and seed the fetal liver at E9.5 of gestation in mice [11, 12]. These fetal monocytes subsequently emerge in the fetal liver at around E12.5 and upon entering the blood circulation at E13.5 [13, 14], colonize the open niches of all the organs apart from those in the brain [15] to give rise to steady-state TRMs. In contrast to these fetal monocytes that are found in the developing embryo, adult monocytes that are found in the circulation are derived from hematopoietic stem cells (HSCs) located within the BM. These HSCs first generate a population of common myeloid progenitors that subsequently differentiate into granulocyte-monocyte progenitors (GMPs) and the monocyte and dendritic cell (DC) progenitors (MDPs) [10]. While MDPs were first believed to be the only monocyte progenitor, it is now recognized that GMPs and MDPs both give rise to monocytes [10]. Current findings indicate a disparity between the developmental lineage among GMPs, MDPs, and common monocyte progenitors (cMoPs) [16–18]. In particular, cMoPs, which represent the first stage of monocyte commitment, can be derived from both GMPs and MDPs albeit disparities in their transcriptomes and function [19, 20]. From the cMoPs, monocyte-committed development then commences, first differentiating into transitional premonocytes (TpMos) (CXCR4hiLy6ChiCCR2+) before downregulating CXCR4 into mature classical Ly6Chi monocytes (CXCR4loLy6ChiCCR2+) that exit into the circulation [21]. Alternatively, Ly6Chi monocytes were also found to convert into non-classical Ly6Clo monocytes within the BM in mice [17, 22] although these Ly6Clo monocytes cells were not found in BM biopsies in humans [23].
While studies have revealed that fetal monocytes share many similarities with adult monocytes, fetal monocytes appear to be functionally immature as they poorly express pathogen recognition or antigen presentation genes and do not rely on CCR2-signalling to migrate into tissues [13]. More importantly, unlike adult monocytes that are terminally differentiated and do not proliferate actively in the blood, fetal monocytes retain a high capacity to proliferate in tissues even in the absence of the CSF-1 receptor [14], thereby displaying a more progenitor-like phenotype compared to their adult counterparts. It is likely that these disparities are needed for monocytes to carry out their roles in different stages of life—fetal monocytes require the proliferative capacity as their main function is to colonize tissues and give rise to TRMs efficiently while the higher expression levels of effector function genes in adult monocytes are needed for immunosurveillance in the periphery. Consequently, genes required for differentiation in adult monocytes are only activated when they enter the tissues during times of need or when inflammation occurs.
Emergency monopoiesis
During inflammatory states, an increase in monocyte demand is significantly required, resulting in a process known as emergency monopoiesis that may also occur for other myeloid cell types [24]. Emergency monopoiesis occurs when an increase in committed monocyte progenitors in the BM occurs at the expense of expansion of other lineage-committed progenitors [25]. For example, DC-lineage progenitors were found to be reduced in order to enhance monocyte production for pathogen control during systemic bacterial infection [26]. Importantly, these severe inflammation states that result in emergency monopoiesis may generate monocytes with alternative phenotypes. Recently, the existence of distinct monocyte trajectories was demonstrated by Weinreb et al. using a combination of lineage tracing and single-cell RNA-seq [19]. Evidence of imprinting in the early stages of monocyte development in these studies is in line with earlier work by Yáñez et al., where they demonstrated that GMPs and MDPs expressed contrasting transcription factors (TFs) typically seen in neutrophils (Gfi1, Cebpa) and monocytes/DCs (Irf8, Klf4) respectively [20]. Upon exposure to CSF-1 in vitro, novel differentiated BM monocyte subsets were seen to emerge from GMPs and MDPs. These GMP- and MDP-derived mature monocytes were subsequently reported to be functionally unique and responded distinctly to independent microbial agents. In particular, Yanez et al. demonstrated that in vivo activation of IFN-γ signalling via CpG triggered the emergence of MDP-derived monocytes labelled as monocyte-derived DC (moDC)-producing Ly6Chi monocytes (DCMos) [20]. DCMos were found to express Cd74, Cd209, Flt3, H2-Aa, and H2-Ab1 genes involved in antigen presentation [10, 20]. Furthermore, DCMo-like cells have been described by Askenase et al. in an infection model of T. gondii [27]. These monocytes with DCMo gene signatures were primed by IFN-γ producing natural killer cells in the BM. Furthermore, these cells expressed Sca-1 and MHCII, as well as inflammatory mediators such as PGE2 and IL-10 that contribute to the inflammatory resolution process. Recently, Barman et al. also identified DCMos and described its expansion from MDPs within the BM of old aging mice [28]. It was outlined that this observation was not representative of improved antigen presentation functions with aging but instead an inflammatory state coupled with underlying deficiencies.
On the other hand, Yáñez et al. demonstrated that in vivo LPS predominantly stimulated the differentiation of GMPs into monocytes termed as ‘neutrophil-like’ Ly6Chi monocytes (NeuMos), characterized by their expression of genes such as Elane, Ctsg, Chil3, Mpo, and Lcn2 [20]. A study by Ikeda et al. later identified a subset of Ym1+(Chil3) Ly6Chi monocytes (Ym1+ Mos) that displayed NeuMo properties and these cells were derived from GMP but not MDP-differentiated cMoPs under LPS challenge [29]. Ym1+ Mos were deemed crucial for inflammation resolution, simultaneously producing higher levels of anti-inflammatory IL-10 and lower pro-inflammatory cytokines (IL-6, IL-12) than Ym1− monocytes. NeuMos were also discovered to be present in the steady-state BM and blood [30, 31], perhaps suggesting the presence of a background signal that maintains a basal level differentiation of GMPs into NeuMos in the BM. On the other hand, a deviation of GMP differentiation into cMoPs into a unique progenitor state could also occur in particular diseases such as bleomycin-induced fibrosis, whereby cells termed segregated-nucleus-containing atypical monocytes (SatMs) were discovered [32]. These SatMs were derived from a progenitor downstream of GMPs but distinct from cMoPs and MDPs [32]. Notably, these cells also displayed neutrophil-like properties such as expression of lipocalin, cathepsin G, neutrophil elastase, and S100a8. Interestingly, both DCMos (Cd74, H2-Aa, and H2-Ab1) [33] and NeuMos (S100a9, Retnlg, Mmp8, Saa3) [30] were reported separately in two studies using an experimental autoimmune encephalomyelitis (EAE) model albeit with different protocols, suggesting that the generation of these cells might involve an intricate play of mechanisms for their appearance.
Trained immunity
While it is clear that distinct BM monocyte subsets are generated through the conditioning of upstream progenitors, the duration and mechanism that these unique subsets are maintained have often been debated. This question has now been addressed through the concept of ‘trained immunity’, whereby innate immune cells undergo long-term functional reprogramming through epigenetic modifications in their progenitors, resulting in an altered response towards a secondary challenge [33]. These findings were observed as early as in the 1970s, whereby glucan pre-treated mice were protected against subsequent septic S. aureus infection [34]. Trained immunity was also observed in the study conducted by Askenase et al., whereby DCMos isolated from IFN-γ stimulated mice were reportedly heightened, producing more PGE2 and IL-10 upon secondary infection compared to naïve mice [27]. Recently, it has also been shown that exposure to LPS at low versus high doses results in distinct epigenomic landscapes, with key implications for pathology in chronic diseases [35]. Furthermore, Bacillus Calmette-Guerin (BCG) resulted in trained immunity in both mice and human studies, leading to improved monocyte functions [36–38]. Interestingly, BCG-vaccinated individuals displayed protective monocyte responses against non-related infections such as an experimental infection with an attenuated yellow fever virus vaccine strain through upregulation of IL-1β [37]. Studies by Cirovic et al. have also shown that hepatic nuclear factor family members 1α and 1β are crucial regulators that underlie the transcriptional shift, resulting in an activated transcriptional signature of peripheral CD14+ monocytes 3 months after BCG vaccination [38]. In addition, Katzmarski et al. have demonstrated that trained immunity can be retained across generations by showing that prior parental infection imparts resistance towards future infections in their offspring in mice [39]. Observations of early life perturbations that result in life-long diseases later in adulthood have also attributed these effects to possible imprinting of EMP-derived cells such as fetal monocytes [40]. However, how these epigenetic modifications in embryonic or BM progenitors can be translated into TRM and monocyte-derived cells upon differentiation remains to be explored. Future studies exploring the manipulation of epigenetic modifications resulting in such persistent trained memory might be beneficial for combating chronic pathologies.
Monocyte reservoirs in the periphery: Strategically positioned to await cues
Upon leaving the BM, monocytes spend the majority of their lifetime in circulation in the blood. Classical or Ly6Chi monocytes are the first monocytic cells that leave the BM and have a half-life of approximately 22 h before they constitutively differentiate into an intermediate state that expresses intermediate levels of Ly6C or CD14 before giving rise to Ly6Clo or non-classical monocytes that have a longer half-life of about 48 h in mice [22, 41]. These patterns were similarly observed in humans albeit with longer circulating lifespans for intermediate and non-classical monocytes [23]. Their differentiation process is governed by Nr4a1 and C/EBPβ signalling [42–44] while CSF-1 signalling [22] and the long non-coding RNA Morrbid [45] regulate their half-lives. Briefly, Ly6Chi monocytes play a critical role in surveying the circulation for inflammatory sites and are the main cells that extravasate into these inflamed tissues to give rise to monocyte-derived cells. In contrast, Ly6Clo monocytes serve as gatekeepers of the blood endothelium by patrolling blood vessels through an LFA-1/ICAM interaction with endothelial cells, which allows them to scavenge luminal microparticles and maintain the endothelial integrity. They are thought to be terminally differentiated and have been proposed to represent blood macrophages [10, 41].
Spleen monocytes
Besides the BM and circulation, monocytes have also been discovered to reside in other peripheral tissues (Fig. 2). In the spleen, a population of monocytes was identified by Swirski et al. within the subcapsular red-pulp region [46] and their numbers and function were recently found to be regulated by gut microbial products [47]. Based on morphology and transcriptome profiles, these splenic monocytes originated from the migration of their blood analogues first produced in the BM [46] and have been shown to be sequestered within the spleen where they clustered mostly in the reticular fibre-rich pulp cords. In the event of certain pathological events such as myocardium ischaemia, the increase of angiotensin II (Ang II) serum levels triggered splenic monocyte egress into the inflamed heart via angiotensin type 1 (AT-1) receptor binding where they differentiated specifically into cardiac macrophages [48, 49]. Mobilization of splenic monocytes into peripheral tissues has also been described in Alzheimer’s disease and in muscular dystrophy [50]. These studies mainly incorporated the use of Ccr2−/− mice (whereby BM monocytes are unable to egress into the periphery), as well as splenectomy, CD45.1 to CD45.2 splenic transplantation and intravital imaging experiments to distinguish splenic from BM-derived circulating monocytes in these mobilization events. While it is unclear why splenic monocytes are preferentially mobilized in these pathological conditions, it is evident that the splenic reservoir of monocytes may serve as an important source of cells for immediate immune defense within the spleen. Specifically, a study done by Hoffman et al. on Salmonella enterica serovar Typhimurium (S. Tm) infection demonstrated that S. Tm pathogens characteristically invade the spleen and activate the differentiation of Ly6Chi and Ly6Clo monocytes into distinct macrophage populations that highly express iNOS and CD9 respectively with the latter replacing red-pulp macrophages [47]. Through the course of infection, these splenic monocyte populations are further maintained alongside S. Tm pathogens in their local environment, suggesting that the monocyte reservoir in the spleen may serve as a ready source of precursor cells that can be rapidly differentiated to control the invasion of pathogens in the spleen [47]. Given the appearance of new monocyte-derived macrophage populations, it is also probable that splenic monocyte–pathogen interactions could also prime monocytes towards atypical developmental fates, and further studies would be needed to determine these outcomes.
Figure 2:
Monocyte reservoirs serve as rapid responders during inflammation. [Top] During MI, splenic monocytes enter the circulation in response to increased serum Angiotensin II (Ang II) levels. They subsequently infiltrate the heart and differentiate into cardiac macrophages. Exposure to Salmonella typhimurium (S. Tm) triggers splenic monocyte differentiation into iNOS-expressing macrophages. [Bottom] Monocytes in the lung differentiate into interstitial and alveolar macrophages with distinct phenotypes according to the environmental signals they encounter. CXCR4-signalling regulates monocyte retention in lung microvessels. Upon sensing endotoxins, monocytes adhere to the endothelium and prolong their lung transit time, possibly resembling PIM-like monocyte-derived macrophages over chronic exposure.
Besides the spleen, blood Ly6Chi monocytes have also been identified to settle in other peripheral organs such as the skin, lungs, and lymph nodes. Specifically, Jakubzick et al. demonstrated that these Ly6Chi monocytes remain nominally differentiated in comparison to their blood counterparts after entering these tissues, but undoubtedly retain their capability to differentiate into macrophages [51], perhaps in response to appropriate stimuli.
Lung marginal pool of monocytes
We have previously shown that the lung consists of a marginal pool of monocytes that are regulated by CXCR4-signalling [21]. This marginal pool was formed due to the multiple small-calibre microvessels (<5 μm in diameter) specifically in the lung, which necessitate larger leukocytes such as monocytes (6–8 μm) to slow down and deform from a spherical into an elongated shape for their transit [52]. Consequently, the lung harbours more monocytes within its vasculature than any other organ [53, 54] and while this marginal pool of monocytes is sequestered from the circulating blood, they can still be rapidly mobilized within the lung parenchyma in times of need [55]. Upon endotoxin sensing, Ly6Chi monocytes increased their lung transit time by adhering to the endothelium [23, 56], resulting in an increased predisposition towards lung injury that can be reversed with CXCR4 inhibition [21]. In particular, livestock animals such as sheep are known to be highly susceptible to acute lung injury induced by such endotoxin exposure [57, 58]. It was postulated that these animals possess a resident lung macrophage population known as pulmonary intravascular macrophages (PIMs) [59, 60], which are seemingly absent in humans and mice. However, O’Dea et al [56]. have discussed that the accumulation of monocytes in the lungs under chronic systematic inflammation may eventually give rise to a population with a PIM-like phenotype [61, 62]. Future studies that involve time kinetics and single-cell sequencing analysis of the lung marginal pool of monocytes after endotoxin exposure would be needed to clarify these intriguing questions. Considering the significant damage that these marginal pools of monocytes exert towards acute lung injury, its physiological relevance remains enigmatic and further research would be required to understand its protective role in various conditions. Future studies exploring specific markers to distinguish monocytes from the spleen versus the lung reservoir and BM-derived circulating monocytes would also be beneficial to provide further insight into their different roles in disease settings.
Timely waves of monocytes with alternative functions
As earlier discussed, monocyte progenitors can be primed in the BM to give rise to monocyte phenotypes with unique functions. Since systemic exposure to pathogens and inflammation may occur before these signals reach the BM, it is conceived that circulatory monocytes may adopt their own unique trajectory after such stimuli. Using single-cell transcriptomic analysis, Lawlor et al. demonstrated that upon exposure to LPS, classical blood monocytes displayed an early activation state characterized by the expression of chemoattractant such as CXCL7, CCL7, and CCL8, followed by a late activation state characterized by the expression of effector molecules such as IL-6, IL-8, and IL-1β [63]. These data suggest that the first wave of monocytes may function mainly as an emergency squad to recruit other immune cells during inflammation, whereas the second wave of monocytes was primed for effector functions such as pathogen clearance. However, many questions still remain: How do these circulating phenotypes differ from monocytes that arise through the priming of BM progenitors such as NeuMos and DCMos? In a recent study, Rigamonti et al. have attempted to address this question by demonstrating nine distinct functional populations of human circulating monocytes in healthy donors [31]. Specifically, there were five populations related either to inflammatory, neutrophil-like, interferon-related, or platelet-related pathways among the classical subset of monocytes. On the other hand, non-classical monocytes consisted of two populations, one of which had elevated expression of complement components. In particular, they identified one cluster of cells in both classical and non-classical subsets that had a strong cytotoxic signature that was markedly increased during COVID-19 infection. Furthermore, patients with advanced gastrointestinal cancer displayed a selective increase of monocytes with platelet-associated pathways while immunotherapy increased the population of interferon-related monocytes. These findings hence suggest that circulating monocytes may indeed derive from distinct developmental trajectories. However, it is unclear if these circulating monocyte phenotypes described in the human settings may represent the same kind of cells that have been primed in the BM or are a unique phenotype that has been primed in the blood. It is also uncertain what happens to NeuMos and DcMos when they are further exposed to circulatory inflammatory signals. Do these cells continue to perform their dedicated functions that were conveyed to them in the BM or do they eventually develop into a tolerance state [64, 65]? Finally, do NeuMos and DcMos eventually differentiate into Ly6Clo monocytes that carry distinct functions from that of Ly6Clo monocytes that were differentiated from circulating unprimed Ly6Chi monocytes? Taken together, these studies highlight numerous ontogeny possibilities that can contribute towards monocyte heterogeneity in the circulation and future studies that address these questions will provide fascinating insights into their plasticity and trajectories.
Monocyte differentiation in tissues: Shapeshifting according to environmental signals
As monocytes infiltrate into the tissue, they gradually acquire unique signatures under the influence of distinct tissue local cues, resulting in TFs that direct tissue-specific differentiation of macrophages [66, 67]. Nevertheless, monocyte differentiation into macrophages occurs in a step-wise process through specific cues as summarized below.
Phenotypic and functional changes of monocytes during differentiation
As Ly6Chi monocytes begin their differentiation, they have been commonly described to undergo a loss of Ly6C while increasing their expression of CD11c, CX3CR1, and CD64 [68–70], which complicates the discrimination of these cells from Ly6Clo monocytes that might have potentially migrated into the tissue. Consequently, it remains a challenge to truly differentiate these two cell types considering the derivation of Ly6Clo monocytes from Ly6Chi monocytes. While it has been proposed that Ly6Clo monocytes are generally constrained to residency in the vasculature where they represent vasculature-resident macrophages [22, 71], their ability to differentiate into tissue-resident macrophages have been reported in some settings through adoptive transfer experiments [72–74] and in the Nr4a1−/− mice [75]. Therefore, developing improved tools to fate-map and track the development of Ly6Chi separately from Ly6Clo monocytes would be needed in the future to resolve this challenge.
Besides the downregulation of Ly6C expression, the differentiation process of Ly6Chi monocytes into macrophages relies on the entry of these non-proliferating cells into the cell cycle [76, 77]. In a mouse model of skeletal muscle injury, inflammatory Ly6Chi monocytes were discovered to lose their expression of Ly6C and adopted a proliferative state before giving rise to macrophages that assist in resolution and regeneration [78]. A similar developmental monocytic fate was described in retinal pigment epithelial cell injury where Ly6Chi monocytes infiltrated the retina and upregulated their proliferative capacity to replenish the loss of endogenous microglia and thereafter acquired microglia-like morphologies [79]. These findings were similarly described in other inflamed tissues such as during cutaneous wound healing [80], urinary tract infection [81], and after helminth infection in the liver [82], where monocytes were found to become proliferative after a few days in the tissue. Deficiency in DNAX activating protein of 12 kD (DAP12) resulted in reduced proliferation of monocytes upon CSF-1 stimulation, suggesting that CSF-1 may potentially be important for inducing their proliferative state [77]. Nevertheless, Ly6Chi monocytes in the blood are non-proliferative yet constitutively absorb CSF-1 [22]. Therefore, further research would be required to decipher the exact signals required for entry and exit of Ly6Chi monocytes in the cell cycle. Specific blockage of proliferation only in monocyte-derived cells and not TRMs would also provide further insight into the specific reasons for the proliferative activity of monocytes upon their differentiation.
Niche signals that influence the differentiation outcome of monocytes
Since tissue-derived signals are critical for the subsequent fate of monocyte-derived cells, it is conceived that the absence or presence of inflammatory signals in the tissue would also alter their differentiation outcome. Furthermore, basic homeostatic cues such as neuronal, circadian rhythms, sleep, diet, and the microbiome have also been recently found to have a significant impact on monocyte fate [83, 84]. In particular, the gut is constantly exposed to a low-grade inflammation caused by commensal microbiota and their products, resulting in monocytes undergoing a loss of Ly6C and concomitant gain in MHCII expression, which has been described as the ‘monocyte waterfall’ [68, 85] that have also been described in other inflammatory settings [86]. In particular, major transcriptomic changes were already found between Ly6Chi monocytes and TRMs early after engraftment [87]. Using an adoptive monocyte transfer model with transcriptional profiling in the small intestine, Desalegn and Pabst further showed that donor monocytes altered their transcriptional profile as early as day 1 after transfer in a Triggering Receptor Expressed On Myeloid Cells 1 (TREM1)-dependent manner, indicating that monocytes are highly sensitive to tissue cues and modify their gene expression as soon as they enter the tissue [88]. Interestingly, monocytes infiltrating the inflamed small intestine adopted an alternative differentiation program from the earliest time point, characterized by higher expression of pro-inflammatory mediators as compared to their counterparts in the normal gut [88]. These distinct transcriptional profiles were documented despite monocytes displaying a superficially similar Ly6C+MHCII− surface phenotype in both homeostasis and inflamed conditions, indicating that improved surface phenotyping would be needed in future studies to correlate their functional profiles. These studies also emphasize the importance of the timing and tissue-specific cues in determining whether monocytes differentiate into functionally similar or different cells from their TRM counterparts. Interestingly, monocytes also give rise to distinct macrophage subtypes in the same tissue depending on their interaction with the micro-niche [89, 90]. For example, Chakarov et al. demonstrated two distinct monocyte-derived interstitial macrophage populations expressing differential levels of Lyve1, MHCII, and CX3CR1 that coexist in specific subtissular niches across tissues [91]. Monocyte-derived macrophages were also found to mimic the transcriptomic signatures of embryo-derived TRMs in the steady-state or after resolution of inflammation, suggesting that tissue signals are highly critical in providing instructions for TRM identities, be it from monocytes or from embryonic progenitors [7, 15]. For example, Hoeffel et al. demonstrated a role for sensory neurons in influencing monocyte recruitment, resulting in the differentiation of monocytes into dermal-resident macrophages that are functionally similar to the embryo-derived Tim4+ macrophages with healing properties [92].
Persistence of imprinted monocytes into monocyte-derived cells
While it is evident that the tissue provides critical signals to modulate the outcome of monocyte-derived cells, Ly6Chi monocytes often adopt distinct phenotypic and effector functions that may be imprinted as early as in the BM [20, 29, 33]. Therefore, whether these phenotypes may be further modified or continue to persist in the tissue remains unclear. During EAE, Ly6Chi monocytes are mobilized into the inflamed tissue where they utilize CSF-2 from T cells to generate inflammatory cytokines and chemokines for pathology [30]. Interestingly, these CNS infiltrating monocytes did not integrate into the tissue-resident microglia pool even after resolution of clinical symptoms [93] although monocytes to MHCII+ microglia transformation was reported after experimental bacterial meningitis [94]. While it is unclear why these findings may occur, it is important to highlight the consideration of heterogeneity among brain macrophages that include border-associated macrophages (BAMs), which unlike microglia in the brain parenchyma, continue to harbour monocyte and inflammatory signatures after resolution [95]. Similarly, monocytes that are recruited into the liver in a model of viral hepatitis [96] and as part of a VEGF-initiated angiogenic program [97] did not differentiate into macrophages. However, on the other hand, infection with gammaherpesvirus generated IL-10-producing MHCIIhiSca-1hi monocytes within the BM, which resulted in engraftment of these regulatory monocytes in the alveolar macrophage pool that self-maintained for months after [98]. Therefore, it seems that differing models of inflammation may provide varying levels of survival factors to retain monocyte-derived cells in distinct anatomical sites of the tissue. Studies have also shown that BM-imprinted monocytes such as NeuMos and DcMos are distinct from mature Ly6Chi-derived monocytes [20, 29, 33] but how these cells behave in the periphery depends on distinct models of inflammation. In particular, Giladi et al. have shown that CXCL10+ monocytes that appear during EAE [33] were exclusively derived from BM monocyte progenitors while mature unprimed Ly6Chi monocytes that enter the CNS gave rise to another subset of cells characterized by iNOS+Arg1+ expression, which seemed similar to TNF/iNOS-expressing (Tip)-DCs that were originally described by Pamer et al. [99]. These findings support a previous study [100] that depicted two distinct CCR2-dependent Ly6Chi monocyte subsets: classical Ly6C+MHCII−CD209a− monocytes that gave rise to Tip-DCs and Ly6C+MHCII+CD209a+ cells that could be primed in the BM. Intriguing, IFN-γ has also been shown to be a key modulator for the appearance of Tip-DCs [101–103], similar to the IFN-γ-driven mechanism of CXCL10+ monocytes [33]. Therefore, further fate-mapping studies would be required to confirm the origin of Ly6C+MHCII+CD209a+ cells and tissue outcome of DcMos that are primed in the BM. It is also intriguing to note that NeuMo-like populations may possibly give rise to distinct monocyte-derived cells in the tissue. In Cohen et al.’s investigation of the effects of copper metabolism MURR1 domain 10 (COMMD10) protein deficiency during liver injury-induced inflammation, the developmental fate of COMMD10-deficient Ly6Chi monocytes in the BM was skewed towards a NeuMo phenotype [104]. This NeuMo-like monocyte population was then recruited to the inflamed liver and further found to mirror the gene expression profile of lipid-associated macrophages (LAMs) [104]. NeuMo-like monocyte populations have also been recently described in the tumour, where they adopt a Ym1+Ly6Chi or Chil3+ phenotype that drives a pro-tumoural response [105, 106]. However, how these cells may integrate into the tumour-associated macrophage (TAM) population remains unclear. Finally, while it seems that BM-imprinted monocytes may have little ability to be further modified in peripheral tissues, Desalegn and Pabst showed that an adoptive transfer of BM monocytes from donors with established small intestinal inflammation developed into a phenotype characteristic of the homeostatic condition when transferred into healthy host tissues [88]. These findings hence indicate that local environmental cues may surpass initial priming phenotypes of monocytes to alter their functional outcome, similar to studies conducted by Lavin et al [107]. whereby macrophages were able to alter their phenotypes when transferred to other tissue environments. Nevertheless, an in-depth understanding of precise tissue- and context-specific factors that regulate monocyte plasticity would be required to clarify these observations.
Mobilization of monocyte progenitors into the periphery: Rising up to the occasion
Monocytes and TRMs require specific growth factors for their differentiation and survival [7, 10, 15]. As a result, only a dedicated number of residential cells can be supported by the tissue at any one point in time. Indeed, Guilliams et al. have proposed a paradigm whereby macrophages and monocyte-derived cells occupy individual ‘niches’ in the tissue and these niche sites are limited by the amount of trophic factors required for survival [108, 109]. In particular, monocyte-derived cells and TRMs rely heavily on stromal cell-derived CSF-1 for their proliferation and survival [110], and therefore, it has been proposed that the levels of CSF-1 in the tissue dictate the number of niches and residential cells in the tissue. Interestingly, this phenomenon closely resembles a model of ‘quorum sensing’ [111, 112], which is the ability to detect and respond to cell population density similarly to that described in bacteria. According to this model, monocytes and macrophages readily consume tissue-derived CSF-1, which allows them to regulate the number of TRMs or monocyte-derived cells in the tissue by functioning as a CSF-1 sink [22, 113]. It is likely that the ‘quorum sensing’ model is partially responsible for allowing a pool of undifferentiated monocytes to reside in tissues, although more studies would be needed to determine if there are other factors that allow monocytes to remain in an undifferentiated state.
Death of TRMs and the monocyte conundrum
TRMs are long-lived cells that have the ability to self-renew in the absence of monocyte input during the steady-state [5]. Since CSF-1 levels govern the availability of niche spaces in the tissue [114–116], it is conceived that monocytes are only able to begin their differentiation process when there is an excess of niche spaces that cannot be fulfilled by the proliferation of remaining TRMs. For example, this might occur when tissues such as the uterus [117, 118] and mammary gland [119] undergo an expansion in stromal network and size as seen during pregnancy; or in the event of TRM cellular death in inflammatory settings [120–124] that impedes the expansion of neighbouring TRMs. In particular, TRMs are well established to carry out homeostatic functions in the tissue but poorly adapt to inflammatory stimuli, which leads to a replacement of TRMs by monocyte-derived cells. Indeed, Ginhoux et al. have proposed that the death of TRMs is an altruistic and necessary process to counteract infections [125]. However, this process may be inappropriately executed in non-infectious settings whereby the death of TRMs resulted in exacerbated liver damage during non-alcoholic steatohepatitis due to less efficient hepatic triglyceride storage by monocyte-derived cells [126]. While TRM death necessitates the infiltration of monocyte-derived cells, the urgency for monocyte replacement in these distinct settings differs significantly. Specifically, the temporary absence of TRMs would likely result in minimal consequences in the steady-state or non-pathogenic inflammatory settings while the lack of TRMs in the presence of pathogens leads to dire consequences associated with increased morbidity if these empty niches are not replaced rapidly to reinstate the peripheral immune defense [127–129]. To complicate matters, active monocyte differentiation is incompatible with simultaneous anti-pathogen effector functions as monocytes are highly susceptible to cellular death after exhaustion of their effector functions. Monocytes from COVID-19-infected patients had activated NLRP3 and AIM2 inflammasomes leading to increased pyroptosis cell death, especially in severe cases [130]. Furthermore, in response to bacterial pathogens or toll-like receptor (TLR) ligand stimulation, monocytes produce pro-inflammatory cytokines and their activation triggers phagocytosis-induced cell death [131]. Similarly, inflammasome activation occurs and monocytes are subjected to various cell death pathways [132]. Consequently, whether the host has evolved strategies to balance the need of monocytes for immunosurveillance versus the replenishment of the TRM niche to sustain immune defense against invading pathogens remains unclear.
Mobilization of monocyte precursors for TRM replenishment
Recently, we have shown that the host is able to overcome the monocyte effector versus replenishment conundrum in an acute bacterial infection model and sepsis setting by mobilizing TpMos, a constitutive proliferating immediate precursor of mature Ly6Chi monocytes (MatMos) located in the BM, into the periphery in a CCR2-independent manner [133]. Upon migrating into the periphery, TpMos were found to serve as an important source of proliferative monocytes that can readily replenish the macrophage pool. This contrasts with MatMos, which are terminally differentiated cells, which were earlier described to enter the cell cycle only after a few days in the presence of local proliferative cues in the tissue site. Indeed, we discovered that TpMos not only gave rise to more macrophages at a much faster rate than MatMos, TpMo-derived macrophages were also more resistant to cellular death. These findings are also reminiscent of the proliferative and progenitor-like features of fetal monocytes that give rise to TRMs rapidly in the embryo [13, 14], suggesting that TpMos may be recruited due to the similar qualities that they possess with fetal monocytes for TRM replenishment.
More importantly, the mobilization of TpMos into the periphery may represent a unique form of ‘quorum-sensing’, whereby the host is able to sense an urgent need for macrophage replenishment at the peripheral site during an infectious setting. This is because TpMos were mobilized from the BM into the periphery only when TRMs were depleted in the presence of an ongoing bacterial infection and their numbers in the periphery correlated with an increasing bacteria burden. In contrast, TpMos were not found in the circulation in the steady state or when TRMs were depleted in the absence of infection. Consequently, these findings highlight an evolutionary mechanism that enables the host to balance the competing demands of monocytes specifically under a pathogen threat, such that the bactericidal and macrophage replacement tasks can be specialized by MatMos and TpMos in the early stages of infection, respectively.
The mobilization of BM TpMos into the periphery suggests that TpMos and other progenitors might provide critical effector functions beyond their roles in the BM. Indeed, while hematopoietic stem and progenitor cells (HSPCs) reside mostly in specialized niches in the BM, these cells are not entirely sessile even in adulthood [134, 135]. It is well established that some HSPCs may recirculate between BM and blood and it is thought that their recirculation fosters the local production of tissue-resident innate immune cells in both steady-state and inflammatory settings [136]. A dysregulation in their circulatory numbers has also been associated with disease states [137]. Besides HSPCs, committed progenitors such as common DC progenitors (CDPs) [138], as well as GMPs and MDPs [139], have been described to migrate into the circulation during infectious settings, with the latter study demonstrating their role in suppressing inflammation. In support of these findings, our research revealed that TpMos gave rise to macrophages that were less inflammatory and transcriptionally distinct from MatMo-derived macrophages, leading to improved sepsis survival [133]. Since TpMos are the direct precursors of MatMos and the differentiation of these cells occurs along a continuum, it remains challenging to delineate TpMo-derived macrophages from MatMo-derived macrophages through specific markers and future studies would be needed to identify and determine their functions in other inflammatory settings. Nonetheless, our results highlight the distinct outcomes of monocyte-derived cells when priming occurs in monocyte precursors/progenitors versus terminally differentiated monocytes, reminiscence of how NeuMos and DcMos are exclusive phenotypes from priming of monocyte progenitors and not mature Ly6Chi monocytes themselves [20, 29, 33]. In further support of these notions, Losslein et al. have also highlighted monocyte progenitors that have been mobilized into the periphery as key contributors towards multinucleated giant cells (MGCs), a unique macrophage type found in granulomas towards mycobacteria [140]. They showed that Mycobacterium tuberculosis (M. tb) programmed cMoPs to accumulate cholesterol and lipids, which are prerequisites for giant cell transformation but may also favour intracellular mycobacterial survival since cholesterol serves as an energy source for M. tb [140]. Their findings also suggest that the adaption towards MGC formation requires progenitor traits, which include high proliferative activity and commitment towards the monocyte lineage [140]. Taken together, these findings indicate a deep level of plasticity in the monocyte hierarchal system and that the host has evolved strategies to exploit the traits of monocyte progenitors for effector functions in the periphery (Fig. 3).
Figure 3:
Mobilization of monocyte progenitors from the BM into the periphery during inflammation. Although hematopoietic stem and progenitor cells (HSPCs) and their progenies are usually immobilized in the BM to generate mature monocytes, they have also been found to have distinct roles in the periphery. HSPCs recirculate between the BM and blood to foster local production of tissue-resident immune cells. GMPs and MDPs migrate into the circulation during infections to suppress inflammation. cMoPs exposed to mycobacteria accumulate cholesterol and lipids, inducing the formation of MGCs against mycobacteria. TpMos are mobilized into circulation and infiltrate tissues during bacterial infections to generate proliferating macrophages with attenuated inflammatory response and apoptotic resistance, improving sepsis survival.
Monocyte-derived cells in tissues: Revisiting transitional inflammatory states
In response to an injury or infectious insult, the tissue often undergoes two distinct phases: an inflammatory phase followed by a resolution phase, which coincides with macrophages or monocyte-derived cells exhibiting either an inflammatory or anti-inflammatory phenotype, respectively [7, 10, 84, 141].
Debunking the M1/M2 classification of TRMs
The concept that macrophages or monocyte-derived cells can be grouped into two such phenotypes was initiated in the early 1980s and 1990s, whereby a paradigm of classically or alternatively activated macrophage subset was established [142–144]. Classically activated macrophages had an overall pro-inflammatory phenotype while alternatively activated displayed immunosuppression activities [142, 145]. Subsequently, Mills et al. described a similar dogma with the M1/M2 nomenclature whereby M1 macrophages were induced by IFN-γ or LPS that primed Th1/Th17 responses while M2 macrophages were polarized through IL-4 and generated Th2 responses [146].
Gradually, these concepts were employed in various settings, which led to substantial confusion among the field of myeloid heterogeneity. Besides the fusion and interchangeable use of the M1/M2 nomenclature with the classical and alternative macrophage polarization concept over time, markers linked to inflammatory processes and resolution of inflammation were beginning to be considered M1 and M2 markers respectively [147]. However, the M1/M2 concept is based on an in vitro construction, and it has been shown that TRMs lose their tissue-specific gene expressions after being incubated in culture for a period of time [148]. Furthermore, a comparison of in vitro gene lists to in vivo M1 and M2 macrophages by Orecchioni et al. revealed little similarity in these two settings [142]. Consequently, it seems that the macrophage polarization dogma is the result of an IL-12/arginase ratio with M1 and M2 signatures at the extreme ends and forcing macrophage and monocyte-related data into M1/M2 groups limits potential data discovery. More importantly, these studies often highlight the ability of M1 and M2 phenotypes to be highly dynamic, suggesting that in vitro macrophages have the potential to switch from an M1 to M2 phenotype interchangeably [149–151].
While the M1/M2 concept has provided some functional insight under specific conditions, it is increasingly clear that these theories do not provide a comprehensive understanding of the actual dynamics of macrophages and monocyte-derived cells that occur in vivo. A pertinent question of the M1-M2 phenotype switch remains: is it the same cell that switches from a pro-inflammatory M1 to an anti-inflammatory M2 phenotype through the course of inflammation? In response to this question, it is increasingly clear that recent work combining fate-mapping, single-cell transcriptomics and epigenetics have revealed that macrophages are less plastic than we thought [152]. In particular, Guilliams and Svedberg have proposed that prolonged tissue residency restricts the plasticity of macrophages in order to allow steady-state tissues to imprint a consistent transcriptomic identity on different types of progenitors [152]. Consequently, this process includes silencing of inflammatory signatures on incoming monocytes, in order to safeguard tissue homeostasis [152]. Indeed, it is now evident that TRMs often undergo cellular death upon exposure to inflammatory signals [120–124] and even if they do survive, these cells clearly carry out non-inflammatory roles compared to infiltrating monocyte-derived cells [15], suggesting that an M1 to M2 switch in TRMs would rarely occur in vivo.
Switching of monocyte-derived cell phenotypes in vivo
In contrast to TRMs, monocytes that infiltrate during inflammation are imprinted with both tissue and inflammation signatures and may disappear upon cellular death or convert into cells that represent TRMs upon tissue resolution [152]. Studies that have utilized fate-mapping and adoptive transfer experiments have indicated that the same monocyte-derived cell may undergo a phenotypic switch under certain circumstances [153]. Specifically, Chen et al. utilized a Ccr2-driven fate-mapping strategy with tamoxifen-inducible Cre-loxP recombination in a stroke model and found that monocyte-derived cells gradually lost their inflammatory signature and increase their restorative phenotype [154]. While the mechanism for this is unclear, Arnold et al. have labelled circulating monocytes with fluorescent latex beads in a model of skeletal muscle injury and discovered that these monocytes differentiated into inflammatory macrophages that switched into a restorative phenotype only upon phagocytosis of apoptotic cells from muscle cell debris [78]. These findings suggest that monocyte-derived cells may require certain specific cues for them to switch their phenotypes. Furthermore, in a recent study by Kratofil et al. that utilized a similar Ccr2-driven fate-mapping approach to Chen et al., monocytes were not found to contribute towards bacterial clearance but instead converted to ghrelin-producing monocyte-derived macrophages that were important for would healing and persisted for weeks after infection [155]. Since the expression of ghrelin was only detected 14 days post-infection in their study, whether ghrelin expression is restricted only to monocytes that have differentiated into a restorative macrophage phenotype and if its expression is absent in inflammatory monocytes would be an intriguing avenue for further research. Furthermore, it is likely that the avoidance of anti-bacterial functions by these monocytes preserves their viability by avoiding phagocytosis-induced cell death [131], which allows these cells to differentiate and persist in the environment. Since neutrophils were the main immune cells involved in bacterial clearance [155], it would be interesting to investigate the impact of neutrophil depletion on monocyte differentiation and its subsequent phenotypes. In light of recent literature that documents distinct BM-imprinted monocytes upon inflammation and mobilization of BM monocyte precursors [20, 29, 33, 133, 140], it also remains unclear if these cell types may give rise to different macrophages with inflammatory and anti-inflammatory properties over time. Taken together, the persistence of monocyte-derived cells and their ability to undergo a phenotypic switch over the course of disease relies on a combination of tissue signals (Fig. 4) and future studies combining spatiotemporal methods, fate-mapping, transcriptomic, and epigenetic analysis will provide further insight into these mechanisms.
Figure 4:
Monocyte-derived cells undergo distinct transitional states during tissue inflammation. During the steady state, Ly6Chi monocytes infiltrate tissues like the heart, pancreas, intestines, and dermis, producing monocyte-derived macrophages (Mo-dMΦs) that resemble TRMs. CSF-1 regulates Mo-dMΦ and TRM proliferation, survival and numbers in the tissue niche. During inflammation, pathogens and inflammatory mediators trigger the death of TRMs. Ly6Chi monocytes, and Ly6Clo monocytes to some extent, infiltrate inflamed tissues and proliferate through CSF-1. They either execute effector functions and undergo phagocytosis-induced cell death, or differentiate into pro-inflammatory Mo-dMΦs. Mo-dMΦs that manage to survive inflammatory signals and phagocytose apoptotic debris consequently adopt a restorative phenotype. Restorative Mo-dMΦs could persist weeks after inflammation and gradually transform into TRMs.
Conclusion
Monocytes are unique cells that can function both as a precursor and effector cell in the periphery. It is now increasingly clear that these cells embark on different fates based on cues from distinct environmental signals. In particular, recent advances and tools that allow us to tease out nuance signals have highlighted how basic environmental factors such as neuronal signals and diet have a much larger impact on monocyte development and function than we would have expected. It is anticipated that the advent of improved technological methods in the future would allow us to converge all these datasets into one unity in order to appreciate the immense depth of plasticity they are capable of and identify coordinating mechanisms between these signals under disease conditions. While emerging evidence suggests that monocyte plasticity begins way earlier than expected, many questions remain for future investigation. In particular, the signals that allow monocyte phenotypes to persist or change with differentiation in the tissue remain unclear. Furthermore, what factors allow some monocyte-derived cells to persist for long periods after tissue resolution while some conditions prevent the engraftment of these cells into the macrophage pool? Given the rapid advancement of research tools that are increasingly available for answering these questions, a systems immunology and in-depth comprehension of monocyte biology should provide important insight for the future development of novel therapeutic strategies.
Acknowledgements
We would like to thank Dr Immanuel Kwok for his valuable feedback and suggestions for the manuscript.
Glossary
Abbreviations:
- Ang II
angiotensin II
- AT-1
angiotensin type 1
- BAM
border-associated macrophage
- BCG
Bacillus Calmette-Guerin
- BM
bone marrow
- cDC
conventional dendritic cell
- CDP
common dendritic cell progenitor
- cMoP
common monocyte progenitor
- CMP
common myeloid progenitor
- COMMD10
copper metabolism MURR1 domain 10
- DAP12
DNAX activating protein of 12 kD
- DC
dendritic cell
- DCMo
moDC-producing Ly6Chi monocyte
- EAE
experimental autoimmune encephalomyelitis
- EMP
erythro-myeloid progenitor
- GMP
granulocyte-monocyte progenitor
- HNF
hepatic nuclear factor
- HSC
haematopoietic stem cell
- HSPC
haematopoietic stem and progenitor cell
- LAM
lipid-associated macrophage
- LPS
lipopolysaccharide
- MatMo
mature Ly6Chi monocyte
- MDP
monocyte and dendritic cell progenitor
- MGC
multinucleated giant cell
- MI
myocardium ischaemia
- Mo-dMΦ
monocyte-derived macrophage
- moDC
monocyte-derived dendritic cell
- NeuMo
neutrophil-like Ly6Chi monocyte
- PIM
pulmonary intravascular macrophage
- SatMs
segregated-nucleus-containing atypical monocyte
- TAM
tumour-associated macrophage
- TF
transcription factor
- TIP-DC
TNF/iNOS-expressing-dendritic cell
- TLR
toll-like receptor
- TpMo
transitional premonocyte
- TREM1
Triggering Receptor Expressed On Myeloid Cells 1
- TRM
tissue-resident macrophages
Contributor Information
Ye Chean Teh, Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore.
Ming Yao Chooi, Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore.
Shu Zhen Chong, Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore.
Ethical approval
Ethical approval is not applicable to this article as no human or animal research was performed for data generation in this paper.
Conflicts of interest
The authors declare no conflict of interest.
Funding
M.Y. Chooi is a graduate scholar supported by the SIgN-NTU-NUS Scholarship, Singapore. This work was supported by the Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR); A*STAR (202D800011) and National Medical Research Council (NMRC) (MOH-001219-00).
Data availability
Data sharing is not applicable to this article as no datasets were generated or analysed in this paper.
Author contributions
Writing, Y.C.T, M.Y.C, and S.Z.C; Visualization, Y.C.T., and S.Z.C.; Funding Acquisition, S.Z.C.; Supervision, S.Z.C.
Permission to reproduce
Figures were created with BioRender.com.
Clinical trial registration
The clinical trial registration is not applicable to this article as no clinical trial was carried out in this paper.
References
- 1.Ehrlich P. Methodologische Beiträge zur Physiologie und Pathologie der verschiedenen Formen der Leukocyten. Z Klin Med 1880, 1, 553–560. doi: 10.1016/b978-0-08-009054-2.50013-4 [DOI] [Google Scholar]
- 2.Metchnikoff E. Über den Kampf der Zellen gegen Erysipel Kokken, ein Beitrag zur Phagocytenlehre. Virchows Arch 1887, 107, 209–249. [Google Scholar]
- 3.van Furth R, Cohn ZA, Hirsch JG, Humphrey JH, Spector WG, Langevoort HL.. The mononuclear phagocyte system: a new classification of macrophages, monocytes, and their precursor cells. Bull World Health Organ 1972, 46, 845–52. [PMC free article] [PubMed] [Google Scholar]
- 4.Guilliams M, Ginhoux F, Jakubzick C, Naik SH, Onai N, Schraml BU, et al. Dendritic cells, monocytes and macrophages: a unified nomenclature based on ontogeny. Nat Rev Immunol 2014, 14, 571–8. doi: 10.1038/nri3712 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Ginhoux F, Guilliams M.. Tissue-resident macrophage ontogeny and homeostasis. Immunity 2016, 44, 439–49. doi: 10.1016/j.immuni.2016.02.024 [DOI] [PubMed] [Google Scholar]
- 6.Merad M, Sathe P, Helft J, Miller J, Mortha A.. The dendritic cell lineage: ontogeny and function of dendritic cells and their subsets in the steady state and the inflamed setting. Annu Rev Immunol 2013, 31, 563–604. doi: 10.1146/annurev-immunol-020711-074950 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Guilliams M, Mildner A, Yona S.. Developmental and functional heterogeneity of monocytes. Immunity 2018, 49, 595–613. doi: 10.1016/j.immuni.2018.10.005 [DOI] [PubMed] [Google Scholar]
- 8.Geissmann F, Jung S, Littman DR.. Blood monocytes consist of two principal subsets with distinct migratory properties. Immunity 2003, 19, 71–82. doi: 10.1016/s1074-7613(03)00174-2 [DOI] [PubMed] [Google Scholar]
- 9.Dutertre CA, Becht E, Irac SE, Khalilnezhad A, Narang V, Khalilnezhad S. et al. Single-cell analysis of human mononuclear phagocytes reveals subset-defining markers and identifies circulating inflammatory dendritic cells. Immunity 2019, 51, 573–589.e8. [DOI] [PubMed] [Google Scholar]
- 10.Trzebanski S, Jung S.. Plasticity of monocyte development and monocyte fates. Immunol Lett 2020, 227, 66–78. doi: 10.1016/j.imlet.2020.07.007 [DOI] [PubMed] [Google Scholar]
- 11.Palis J, Robertson SM, Kennedy M, Wall C, Keller G.. Development of erythroid and myeloid progenitors in the yolk sac and embryo proper of the mouse. Development 1999, 126, 5073–84. [DOI] [PubMed] [Google Scholar]
- 12.Bertrand JY, Jalil A, Klaine M, Jung S, Cumano A, Godin I.. Three pathways to mature macrophages in the early mouse yolk sac. Blood 2005, 106, 3004–11. doi: 10.1182/blood-2005-02-0461 [DOI] [PubMed] [Google Scholar]
- 13.Hoeffel G, Chen J, Lavin Y, Low D, Almeida FF, See P, et al. C-Myb(+) erythro-myeloid progenitor-derived fetal monocytes give rise to adult tissue-resident macrophages. Immunity 2015, 42, 665–78. doi: 10.1016/j.immuni.2015.03.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Hoeffel G, Wang Y, Greter M, See P, Teo P, Malleret B, et al. Adult Langerhans cells derive predominantly from embryonic fetal liver monocytes with a minor contribution of yolk sac–derived macrophages. J Exp Med 2012, 209, 1167–81. doi: 10.1084/jem.20120340 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Park MD, Silvin A, Ginhoux F, Merad M.. Macrophages in health and disease. Cell 2022, 185, 4259–79. doi: 10.1016/j.cell.2022.10.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Liu Z, Gu Y, Chakarov S, Bleriot C, Kwok I, Chen X, et al. Fate mapping via Ms4a3-expression history traces monocyte-derived cells. Cell 2019, 178, 1509–1525.e19. doi: 10.1016/j.cell.2019.08.009 [DOI] [PubMed] [Google Scholar]
- 17.Hettinger J, Richards DM, Hansson J, Barra MM, Joschko A-C, Krijgsveld J, et al. Origin of monocytes and macrophages in a committed progenitor. Nat Immunol 2013, 14, 821–30. doi: 10.1038/ni.2638 [DOI] [PubMed] [Google Scholar]
- 18.Kwok I, Becht E, Xia Y, Ng M, Teh YC, Tan L, et al. Combinatorial single-cell analyses of granulocyte-monocyte progenitor heterogeneity reveals an early uni-potent neutrophil progenitor. Immunity 2020, 53, 303–318.e5. doi: 10.1016/j.immuni.2020.06.005 [DOI] [PubMed] [Google Scholar]
- 19.Weinreb C, Rodriguez-Fraticelli A, Camargo F, Klein AM.. Lineage tracing on transcriptional landscapes links state to fate during differentiation. Science 2020, 367, eaaw3381. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Yáñez A, Coetzee SG, Olsson A, Muench DE, Berman BP, Hazelett DJ, et al. Granulocyte-monocyte progenitors and monocyte-dendritic cell progenitors independently produce functionally distinct monocytes. Immunity 2017, 47, 890–902.e4. doi: 10.1016/j.immuni.2017.10.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Chong SZ, Evrard M, Devi S, Chen J, Lim JY, See P, et al. CXCR4 identifies transitional bone marrow premonocytes that replenish the mature monocyte pool for peripheral responses. J Exp Med 2016, 213, 2293–314. doi: 10.1084/jem.20160800 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Yona S, Kim K-W, Wolf Y, Mildner A, Varol D, Breker M, et al. Fate mapping reveals origins and dynamics of monocytes and tissue macrophages under homeostasis. Immunity 2013, 38, 79–91. doi: 10.1016/j.immuni.2012.12.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Patel AA, Zhang Y, Fullerton JN, Boelen L, Rongvaux A, Maini AA, et al. The fate and lifespan of human monocyte subsets in steady state and systemic inflammation. J Exp Med 2017, 214, 1913–23. doi: 10.1084/jem.20170355 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Schultze JL, Mass E, Schlitzer A.. Emerging principles in myelopoiesis at homeostasis and during infection and inflammation. Immunity 2019, 50, 288–301. doi: 10.1016/j.immuni.2019.01.019 [DOI] [PubMed] [Google Scholar]
- 25.Takizawa H, Boettcher S, Manz MG.. Demand-adapted regulation of early hematopoiesis in infection and inflammation. Blood 2012, 119, 2991–3002. doi: 10.1182/blood-2011-12-380113 [DOI] [PubMed] [Google Scholar]
- 26.Pasquevich KA, Bieber K, Günter M, Grauer M, Pötz O, Schleicher U, et al. Innate immune system favors emergency monopoiesis at the expense of DC-differentiation to control systemic bacterial infection in mice. Eur J Immunol 2015, 45, 2821–33. doi: 10.1002/eji.201545530 [DOI] [PubMed] [Google Scholar]
- 27.Askenase MH, Han S-J, Byrd Allyson L, Morais da Fonseca D, Bouladoux N, Wilhelm C, et al. Bone-marrow-resident NK cells prime monocytes for regulatory function during infection. Immunity 2015, 42, 1130–42. doi: 10.1016/j.immuni.2015.05.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Barman PK, Shin JE, Lewis SA, Kang S, Wu D, Wang Y, et al. Production of MHCII-expressing classical monocytes increases during aging in mice and humans. Aging Cell 2022, 21, e13701. doi: 10.1111/acel.13701 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Ikeda N, Asano K, Kikuchi K, Uchida Y, Ikegami H, Takagi R, et al. Emergence of immunoregulatory Ym1 + Ly6C hi monocytes during recovery phase of tissue injury. Sci Immunol 2018, 3, eaat0207. doi: 10.1126/sciimmunol.aat0207 [DOI] [PubMed] [Google Scholar]
- 30.Amorim A, De Feo D, Friebel E, Ingelfinger F, Anderfuhren CD, Krishnarajah S, et al. IFNγ and GM-CSF control complementary differentiation programs in the monocyte-to-phagocyte transition during neuroinflammation. Nat Immunol 2022, 23, 217–28. doi: 10.1038/s41590-021-01117-7 [DOI] [PubMed] [Google Scholar]
- 31.Rigamonti A, Castagna A, Viatore M, Colombo FS, Terzoli S, Peano C, et al. Distinct responses of newly identified monocyte subsets to advanced gastrointestinal cancer and COVID-19. Front Immunol 2022, 13, 967737. doi: 10.3389/fimmu.2022.967737 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Satoh T, Nakagawa K, Sugihara F, Kuwahara R, Ashihara M, Yamane F, et al. Identification of an atypical monocyte and committed progenitor involved in fibrosis. Nature 2017, 541, 96–101. doi: 10.1038/nature20611 [DOI] [PubMed] [Google Scholar]
- 33.Giladi A, Wagner LK, Li H, Dörr D, Medaglia C, Paul F, et al. Cxcl10+ monocytes define a pathogenic subset in the central nervous system during autoimmune neuroinflammation. Nat Immunol 2020, 21, 525–34. doi: 10.1038/s41590-020-0661-1 [DOI] [PubMed] [Google Scholar]
- 34.Di Luzio NR, Williams DL.. Protective effect of glucan against systemic Staphylococcus aureus septicemia in normal and leukemic mice. Infect Immun 1978, 20, 804–10. doi: 10.1128/iai.20.3.804-810.1978 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Naler LB, Hsieh Y-P, Geng S, Zhou Z, Li L, Lu C.. Epigenomic and transcriptomic analyses reveal differences between low-grade inflammation and severe exhaustion in LPS-challenged murine monocytes. Commun Biol 2022, 5, 1–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Kaufmann E, Sanz J, Dunn Jonathan L, Khan N, Mendonça LE, Pacis A, et al. BCG educates hematopoietic stem cells to generate protective innate immunity against tuberculosis. Cell 2018, 172, 176–190.e19. doi: 10.1016/j.cell.2017.12.031 [DOI] [PubMed] [Google Scholar]
- 37.Arts RJW, Moorlag Simone JCFM, Novakovic B, Li Y, Wang S-Y, Oosting M, et al. BCG vaccination protects against experimental viral infection in humans through the induction of cytokines associated with trained immunity. Cell Host Microbe 2018, 23, 89–100.e5. doi: 10.1016/j.chom.2017.12.010 [DOI] [PubMed] [Google Scholar]
- 38.Cirovic B, de Bree L, Charlotte J, Groh L, Blok BA, Chan J, van der Velden WJFM, et al. BCG vaccination in humans elicits trained immunity via the hematopoietic progenitor compartment. Cell Host Microbe 2020, 28, 322–334.e5. doi: 10.1016/j.chom.2020.05.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Katzmarski N, Domínguez-Andrés J, Cirovic B, Renieris G, Ciarlo E, Le Roy D, et al. Transmission of trained immunity and heterologous resistance to infections across generations. Nat Immunol 2021, 22, 1382–90. doi: 10.1038/s41590-021-01052-7 [DOI] [PubMed] [Google Scholar]
- 40.Mass E, Gentek R, Gentek R.. Fetal-derived immune cells at the roots of lifelong pathophysiology. Front Cell Dev Biol 2021, 9: 648313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Patel A, Patel AA, Ginhoux F, Yona S.. Monocytes, macrophages, dendritic cells and neutrophils: an update on lifespan kinetics in health and disease. Immunology 2021, 163, 250–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Mildner A, Schönheit J, Giladi A, David E, Lara-Astiaso D, Lorenzo-Vivas E, et al. Genomic characterization of murine monocytes reveals C/EBPβ transcription factor dependence of Ly6C(-) cells. Immunity 2017, 46, 849–862.e7. doi: 10.1016/j.immuni.2017.04.018 [DOI] [PubMed] [Google Scholar]
- 43.Hanna RN, Carlin LM, Hubbeling HG, Nackiewicz D, Green AM, Punt JA, et al. The transcription factor NR4A1 (Nur77) controls bone marrow differentiation and the survival of Ly6C- monocytes. Nat Immunol 2011, 12, 778–85. doi: 10.1038/ni.2063 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Thomas GD, Hanna RN, Vasudevan NT, Hamers AA, Romanoski CE, McArdle S, et al. Deleting an Nr4a1 super-enhancer subdomain ablates Ly6C low monocytes while preserving macrophage gene function. Immunity 2016, 45, 975–87. doi: 10.1016/j.immuni.2016.10.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Kotzin JJ, Spencer SP, McCright SJ, Kumar DBU, Collet MA, Mowel WK, et al. The long non-coding RNA Morrbid regulates Bim and short-lived myeloid cell lifespan. Nature 2016, 537, 239–43. doi: 10.1038/nature19346 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Swirski FK, Nahrendorf M, Etzrodt M, Wildgruber M, Cortez-Retamozo V, Panizzi P, et al. Identification of splenic reservoir monocytes and their deployment to inflammatory sites. Science 2009, 325, 612–616. doi: 10.1126/science.1175202 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Hoffman D, Tevet Y, Trzebanski S, Rosenberg G, Vainman L, Solomon A, et al. A non-classical monocyte-derived macrophage subset provides a splenic replication niche for intracellular Salmonella. Immunity 2021, 54, 2712–2723.e6. doi: 10.1016/j.immuni.2021.10.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Peet C, Ivetic A, Bromage DI, Shah AM.. Cardiac monocytes and macrophages after myocardial infarction. Cardiovasc Res 2020, 116, 1101–12. doi: 10.1093/cvr/cvz336 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Bronte V, Pittet MJ.. The spleen in local and systemic regulation of immunity. Immunity 2013, 39, 806–18. doi: 10.1016/j.immuni.2013.10.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Yan P, Kim K-W, Xiao Q, Ma X, Czerniewski LR, Liu H, et al. Peripheral monocyte–derived cells counter amyloid plaque pathogenesis in a mouse model of Alzheimer’s disease. J Clin Invest 2022, 132: e152565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Jakubzick C, Gautier Emmanuel L, Gibbings Sophie L, Sojka DK, Schlitzer A, Johnson TE, et al. Minimal differentiation of classical monocytes as they survey steady state tissues and transport antigen to lymph nodes. Immunity 2013, 39, 599–610. doi: 10.1016/j.immuni.2013.08.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Kuebler WM, Kuebler WM, Goetz AE.. The marginated pool. Eur Surg Res 2002, 34, 92–100. [DOI] [PubMed] [Google Scholar]
- 53.Pabst R, Binns RM, Licence ST, Peter M.. Evidence of a selective major vascular marginal pool of lymphocytes in the lung. Am Rev Respir Dis 1987, 136, 1213–8. doi: 10.1164/ajrccm/136.5.1213 [DOI] [PubMed] [Google Scholar]
- 54.Van Furth R, Sluiter W.. Distribution of blood monocytes between a marginating and a circulating pool. J Exp Med 1986, 163, 474–9. doi: 10.1084/jem.163.2.474 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Dhabhar FS, Dhabhar FS, Malarkey WB, Neri E, McEwen BS.. Stress-induced redistribution of immune cells--from barracks to boulevards to battlefields: a tale of three hormones—Curt Richter Award winner. Psychoneuroendocrinology 2012, 37, 1345–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.O’Dea KP, Wilson MR, Dokpesi JO, Wakabayashi K, Tatton L, van Rooijen N, et al. Mobilization and Margination of Bone Marrow Gr-1(high) monocytes during subclinical endotoxemia predisposes the lungs toward acute injury. J Immunol 2009, 182, 1155–66. doi: 10.4049/jimmunol.182.2.1155 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Sone Y, Nicolaysen A, Staub NC.. Effect of particles on sheep lung hemodynamics parallels depletion and recovery of intravascular macrophages. J Appl Physiol 1997, 83, 1499–507. doi: 10.1152/jappl.1997.83.5.1499 [DOI] [PubMed] [Google Scholar]
- 58.Warner AE, Angeline EW.. Pulmonary intravascular macrophages: role in acute lung injury. Clin Chest Med 1996, 17, 125–35. [DOI] [PubMed] [Google Scholar]
- 59.Warner AE, Brain JD.. The cell biology and pathogenic role of pulmonary intravascular macrophages. Am J Physiol Lung Cell Mol Physiol 1990, 258, L1–12. doi: 10.1152/ajplung.1990.258.2.L1 [DOI] [PubMed] [Google Scholar]
- 60.Staub NC. Pulmonary intravascular macrophages. Annu Rev Physiol 1994, 56, 47–67. doi: 10.1146/annurev.ph.56.030194.000403 [DOI] [PubMed] [Google Scholar]
- 61.Charavaryamath C, Janardhan KS, Caldwell S, Singh B.. Pulmonary intravascular monocytes/macrophages in a rat model of sepsis. Anat Record Adv Integr Anat Evolut Biol 2006, 288, 1259–71. doi: 10.1002/ar.a.20401 [DOI] [PubMed] [Google Scholar]
- 62.Chang S-W, Ohara N.. Chronic biliary obstruction induces pulmonary intravascular phagocytosis and endotoxin sensitivity in rats. J Clin Investig 1994, 94, 2009–19. doi: 10.1172/JCI117554 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Lawlor N, Nehar-Belaid D, Grassmann JDS, Stoeckius M, Smibert P, Stitzel ML, et al. Single cell analysis of blood mononuclear cells stimulated through either LPS or anti-CD3 and anti-CD28. Front Immunol 2021, 12: 636720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Wiśnik E, Pikus E, Duchnowicz P, Koter-Michalak M.. Tolerance of monocytes and macrophages in response to bacterial endotoxin. Postepy Hig Med Dos 2017, 71, 176–85. doi: 10.5604/01.3001.0010.3802 [DOI] [PubMed] [Google Scholar]
- 65.Widdrington J, et al. Exposure of monocytic cells to lipopolysaccharide induces coordinated endotoxin tolerance, mitochondrial biogenesis, mitophagy, and antioxidant defenses. Front Immunol 2018, 9, 2217–2217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Varol C, Mildner A, Jung S.. Macrophages: development and tissue specialization. Annu Rev Immunol 2015, 33, 643–75. doi: 10.1146/annurev-immunol-032414-112220 [DOI] [PubMed] [Google Scholar]
- 67.Blériot C, Chakarov S, Ginhoux F.. Determinants of resident tissue macrophage identity and function. Immunity 2020, 52, 957–70. doi: 10.1016/j.immuni.2020.05.014 [DOI] [PubMed] [Google Scholar]
- 68.Tamoutounour S, Henri S, Lelouard H, de Bovis B, de Haar C, van der Woude CJ, et al. CD64 distinguishes macrophages from dendritic cells in the gut and reveals the Th1-inducing role of mesenteric lymph node macrophages during colitis. Eur J Immunol 2012, 42, 3150–66. doi: 10.1002/eji.201242847 [DOI] [PubMed] [Google Scholar]
- 69.Bain CC, Bravo-Blas A, Scott Charlotte L, Perdiguero EG, Geissmann F, Henri S, et al. Constant replenishment from circulating monocytes maintains the macrophage pool in the intestine of adult mice. Nat Immunol 2014, 15, 929–37. doi: 10.1038/ni.2967 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Schridde A, Bain CC, Mayer JU, Montgomery J, Pollet E, Denecke B, et al. Tissue-specific differentiation of colonic macrophages requires TGFβ receptor-mediated signaling. Mucos Immunol 2017, 10, 1387–99. doi: 10.1038/mi.2016.142 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Auffray C, Fogg D, Garfa M, Elain G, Join-Lambert O, Kayal S, et al. Monitoring of blood vessels and tissues by a population of monocytes with patrolling behavior. Science 2007, 317, 666–70. doi: 10.1126/science.1142883 [DOI] [PubMed] [Google Scholar]
- 72.Misharin AV, Cuda CM, Saber R, Turner JD, Gierut AK, Haines GK, et al. Nonclassical Ly6C–monocytes drive the development of inflammatory arthritis in mice. Cell Rep 2014, 9, 591–604. doi: 10.1016/j.celrep.2014.09.032 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Nahrendorf M, Swirski FK, Aikawa E, Stangenberg L, Wurdinger T, Figueiredo J-L, et al. The healing myocardium sequentially mobilizes two monocyte subsets with divergent and complementary functions. J Exp Med 2007, 204, 3037–47. doi: 10.1084/jem.20070885 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Hou X, Chen G, Bracamonte-Baran W, Choi HS, Diny NL, Sung J, et al. The cardiac microenvironment instructs divergent monocyte fates and functions in myocarditis. Cell Rep 2019, 28, 172–189.e7. doi: 10.1016/j.celrep.2019.06.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Schyns J, Bai Q, Ruscitti C, Radermecker C, De Schepper S, Chakarov S, et al. Non-classical tissue monocytes and two functionally distinct populations of interstitial macrophages populate the mouse lung. Nat Commun 2019, 10, 3964. doi: 10.1038/s41467-019-11843-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Jenkins SJ, Knipper JA, Zaiss DMW.. Local proliferation of monocytes. J Leukoc Biol 2020, 107, 547–9. doi: 10.1002/JLB.1CE0220-534RR [DOI] [PubMed] [Google Scholar]
- 77.Otero K, Turnbull IR, Poliani PL, Vermi W, Cerutti E, Aoshi T, et al. Macrophage colony-stimulating factor induces the proliferation and survival of macrophages via a pathway involving DAP12 and beta-catenin. Nat Immunol 2009, 10, 734–43. doi: 10.1038/ni.1744 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Arnold L, Henry A, Poron F, Baba-Amer Y, van Rooijen N, Plonquet A, et al. Inflammatory monocytes recruited after skeletal muscle injury switch into antiinflammatory macrophages to support myogenesis. J Exp Med 2007, 204, 1057–69. doi: 10.1084/jem.20070075 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Ma W, Zhang Y, Gao C, Fariss RN, Tam J, Wong WT.. Monocyte infiltration and proliferation reestablish myeloid cell homeostasis in the mouse retina following retinal pigment epithelial cell injury. Sci Rep 2017, 7, 8433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Pang J, Urao N, Koh TJ, Koh TJ, Timothy JK.. Proliferation of Ly6C+ monocytes/macrophages contributes to their accumulation in mouse skin wounds. J Leukoc Biol 2020, 107, 551–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Dixit A, Bottek J, Beerlage A-L, Schuettpelz J, Thiebes S, Brenzel A, et al. Frontline science: proliferation of Ly6C+ monocytes during urinary tract infections is regulated by IL-6 trans-signaling. J Leukoc Biol 2017, 103, 13–22. doi: 10.1189/jlb.3HI0517-198R [DOI] [PubMed] [Google Scholar]
- 82.Rolot M, Dougall A, Javaux J, Lallemand F, Machiels B, Martinive P, et al. Recruitment of hepatic macrophages from monocytes is independent of IL-4Rα but is associated with ablation of resident macrophages in schistosomiasis. Eur J Immunol 2019, 49, 1067–81. doi: 10.1002/eji.201847796 [DOI] [PubMed] [Google Scholar]
- 83.Patel AA, Patel A, Simon Y, Yona S.. Inherited and environmental factors influence human monocyte heterogeneity. Front Immunol 2019, 10, 2581–2581 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Mildner A, Yona S, Jung S.. A close encounter of the third kind: monocyte-derived cells. Adv Immunol 2013, 120, 69–103. doi: 10.1016/B978-0-12-417028-5.00003-X [DOI] [PubMed] [Google Scholar]
- 85.Bsat M, Chapuy L, Rubio M, Sarfati M.. A two-step human culture system replicates intestinal monocyte maturation cascade: conversion of tissue-like inflammatory monocytes into macrophages. Eur J Immunol 2020, 50, 1676–90. doi: 10.1002/eji.202048555 [DOI] [PubMed] [Google Scholar]
- 86.Goh CC, Evrard M, Chong SZ, Tan Y, Tan Leonard De L, Teng KWW, et al. The impact of ischemia-reperfusion injuries on skin resident murine dendritic cells. Eur J Immunol 2018, 48, 1014–9. doi: 10.1002/eji.201747347 [DOI] [PubMed] [Google Scholar]
- 87.Gross-Vered M, Trzebanski S, Shemer A, Bernshtein B, Curato C, Stelzer G, et al. Defining murine monocyte differentiation into colonic and ileal macrophages. eLife 2020, 9, e49998. doi: 10.7554/eLife.49998 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Desalegn G, Desalegn G, Pabst O.. Inflammation triggers immediate rather than progressive changes in monocyte differentiation in the small intestine. Nat Commun 2019, 10, 3229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Mass E, Nimmerjahn F, Katrin K, Schlitzer A.. Tissue-specific macrophages: how they develop and choreograph tissue biology. Nat Rev Immunol 2023, 15, 1-17. doi: 10.1038/s41577-023-00848-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Chong SZ, Evrard M, Goh CC, Gascoigne NRJ, Ng LG.. Illuminating the covert mission of mononuclear phagocytes in their regional niches. Curr Opin Immunol 2018, 50, 94–101. [DOI] [PubMed] [Google Scholar]
- 91.Chakarov S, Lim HY, Tan L, Lim SY, See P, Lum J, et al. Two distinct interstitial macrophage populations coexist across tissues in specific subtissular niches. Science 2019, 363, eaau0964. doi: 10.1126/science.aau0964 [DOI] [PubMed] [Google Scholar]
- 92.Hoeffel G, Debroas G, Roger A, Rossignol R, Gouilly J, Laprie C, et al. Sensory neuron-derived TAFA4 promotes macrophage tissue repair functions. Nature 2021, 594, 94–9. doi: 10.1038/s41586-021-03563-7 [DOI] [PubMed] [Google Scholar]
- 93.Djukic M, Mildner A, Schmidt H, Czesnik D, Brück W, Priller J, et al. Circulating monocytes engraft in the brain, differentiate into microglia and contribute to the pathology following meningitis in mice. Brain 2006, 129, 2394–403. doi: 10.1093/brain/awl206 [DOI] [PubMed] [Google Scholar]
- 94.Ajami B, Bennett JL, Krieger C, McNagny KM, Rossi FMV.. Infiltrating monocytes trigger EAE progression, but do not contribute to the resident microglia pool. Nat Neurosci 2011, 14, 1142–9. [DOI] [PubMed] [Google Scholar]
- 95.De Vlaminck K, et al. Differential plasticity and fate of brain-resident and recruited macrophages during the onset and resolution of neuroinflammation. Immunity 2022, 55, 2085–2102.e9. [DOI] [PubMed] [Google Scholar]
- 96.Borst K, Frenz T, Spanier J, Tegtmeyer P-K, Chhatbar C, Skerra J, et al. Type I interferon receptor signaling delays Kupffer cell replenishment during acute fulminant viral hepatitis. J Hepatol 2017, 68, 682–90. doi: 10.1016/j.jhep.2017.11.029 [DOI] [PubMed] [Google Scholar]
- 97.Avraham-Davidi I, Yona S, Grunewald M, Landsman L, Cochain C, Silvestre JS, et al. On-site education of VEGF-recruited monocytes improves their performance as angiogenic and arteriogenic accessory cells. J Exp Med 2013, 210, 2611–25. doi: 10.1084/jem.20120690 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Machiels B, Dourcy M, Xiao X, Javaux J, Mesnil C, Sabatel C, et al. A gammaherpesvirus provides protection against allergic asthma by inducing the replacement of resident alveolar macrophages with regulatory monocytes. Nat Immunol 2017, 18, 1310–20. doi: 10.1038/ni.3857 [DOI] [PubMed] [Google Scholar]
- 99.Serbina NV, Salazar-Mather TP, Biron CA, Kuziel WA, Pamer EG.. TNF/iNOS-producing dendritic cells mediate innate immune defense against bacterial infection. Immunity 2003, 19, 59–70. doi: 10.1016/s1074-7613(03)00171-7 [DOI] [PubMed] [Google Scholar]
- 100.Menezes S, Melandri D, Anselmi G, Perchet T, Loschko J, Dubrot J, et al. The heterogeneity of Ly6Chi monocytes controls their differentiation into iNOS+ macrophages or monocyte-derived dendritic cells. Immunity 2016, 45, 1205–18. doi: 10.1016/j.immuni.2016.12.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Bosschaerts T, Guilliams M, Stijlemans B, Morias Y, Engel D, Tacke F, et al. Tip-DC development during parasitic infection is regulated by IL-10 and requires CCL2/CCR2, IFN-γ and MyD88 signaling. PLoS Pathog 2010, 6, e1001045–36. doi: 10.1371/journal.ppat.1001045 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Chong SZ, Wong KL, Lin G, Yang CM, Wong S-C, Angeli V, et al. Human CD8+ T cells drive Th1 responses through the differentiation of TNF/iNOS‐producing dendritic cells. Eur J Immunol 2011, 41, 1639–51. doi: 10.1002/eji.201041022 [DOI] [PubMed] [Google Scholar]
- 103.Chong SZ, Tan KW, Wong FHS, Chua YL, Tang Y, Ng LG, et al. CD8 T cells regulate allergic contact dermatitis by modulating CCR2–dependent TNF/iNOS–expressing Ly6C+CD11b+ monocytic cells. J Investig Dermatol 2014, 134, 666–76. doi: 10.1038/jid.2013.403 [DOI] [PubMed] [Google Scholar]
- 104.Cohen K, Mouhadeb O, Ben Shlomo S, Langer M, Neumann A, Erez N, et al. COMMD10 is critical for Kupffer cell survival and controls Ly6Chi monocyte differentiation and inflammation in the injured liver. Cell Rep 2021, 37, 110026. doi: 10.1016/j.celrep.2021.110026 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Shibuya T, Kamiyama A, Sawada H, Kikuchi K, Maruyama M, Sawado R, et al. Immunoregulatory monocyte subset promotes metastasis associated with therapeutic intervention for primary tumor. Front Immunol 2021, 12, 663115. doi: 10.3389/fimmu.2021.663115 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Baharom F, Ramirez-Valdez RA, Khalilnezhad A, Khalilnezhad S, Dillon M, Hermans D, et al. Systemic vaccination induces CD8+ T cells and remodels the tumor microenvironment. Cell 2022, 185, 4317–4332.e15. doi: 10.1016/j.cell.2022.10.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Lavin Y, Winter D, Blecher-Gonen R, David E, Keren-Shaul H, Merad M, et al. Tissue-resident macrophage enhancer landscapes are shaped by the local microenvironment. Cell 2014, 159, 1312–26. doi: 10.1016/j.cell.2014.11.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Guilliams M, Scott CL.. Does niche competition determine the origin of tissue-resident macrophages? Nat Rev Immunol 2017, 17, 451–60. doi: 10.1038/nri.2017.42 [DOI] [PubMed] [Google Scholar]
- 109.Guilliams M, Thierry GR, Bonnardel J, Bajénoff M, Bajénoff M.. Establishment and maintenance of the macrophage niche. Immunity 2020, 52, 434–51. [DOI] [PubMed] [Google Scholar]
- 110.Dai X, Ryan GR, Hapel AJ, Dominguez MG, Russell RG, Kapp S, et al. Targeted disruption of the mouse colony-stimulating factor 1 receptor gene results in osteopetrosis, mononuclear phagocyte deficiency, increased primitive progenitor cell frequencies, and reproductive defects. Blood 2002, 99, 111–20. doi: 10.1182/blood.v99.1.111 [DOI] [PubMed] [Google Scholar]
- 111.Miller MB, Bassler BL.. Quorum sensing in bacteria. Annu Rev Microbiol 2001, 55, 165–99. doi: 10.1146/annurev.micro.55.1.165 [DOI] [PubMed] [Google Scholar]
- 112.Eickhoff MJ, Bassler BL.. SnapShot: bacterial quorum sensing. Cell 2018, 174, 1328–1328.e1. doi: 10.1016/j.cell.2018.08.003 [DOI] [PubMed] [Google Scholar]
- 113.Postat J, Bousso P.. Quorum sensing by monocyte-derived populations. Front Immunol 2019, 10, 2140–2140. doi: 10.3389/fimmu.2019.02140 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Bellomo A, Mondor I, Spinelli L, Lagueyrie M, Stewart BJ, Brouilly N, et al. Reticular fibroblasts expressing the transcription factor WT1 define a stromal niche that maintains and replenishes splenic red pulp macrophages. Immunity 2020, 53, 127–142.e7. doi: 10.1016/j.immuni.2020.06.008 [DOI] [PubMed] [Google Scholar]
- 115.Zhou X, Franklin RA, Adler M, Jacox JB, Bailis W, Shyer JA, et al. Circuit design features of a stable two-cell system. Cell 2018, 172, 744–757.e17. doi: 10.1016/j.cell.2018.01.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Adler M, Mayo A, Zhou X, Franklin RA, Jacox JB, Medzhitov R, et al. Endocytosis as a stabilizing mechanism for tissue homeostasis. Proc Natl Acad Sci USA 2018, 115, E1926–1935. doi: 10.1073/pnas.1714377115 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Elisa T, et al. Coordinate regulation of tissue macrophage and dendritic cell population dynamics by CSF-1. J Exp Med 2011, 208, 1901–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Bartocci A, Pollard JW, Stanley ER, Stanley ER.. Regulation of colony-stimulating factor 1 during pregnancy. J Exp Med 1986, 164, 956–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Dawson CA, Pal B, Vaillant F, Gandolfo Luke C, Liu Z, Bleriot C, et al. Tissue-resident ductal macrophages survey the mammary epithelium and facilitate tissue remodelling. Nat Cell Biol 2020, 22, 546–58. doi: 10.1038/s41556-020-0505-0 [DOI] [PubMed] [Google Scholar]
- 120.Zychlinsky A, Prevost MC, Sansonetti PJ.. Shigella flexneri induces apoptosis in infected macrophages. Nature 1992, 358, 167–9. doi: 10.1038/358167a0 [DOI] [PubMed] [Google Scholar]
- 121.Robinson N, McComb S, Mulligan R, Dudani R, Krishnan L, Sad S.. Type I interferon induces necroptosis in macrophages during infection with Salmonella enterica serovar Typhimurium. Nat Immunol 2012, 13, 954–62. doi: 10.1038/ni.2397 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Blériot C, Dupuis T, Jouvion G, Eberl G, Disson O, Lecuit M.. Liver-resident macrophage necroptosis orchestrates type 1 microbicidal inflammation and type-2-mediated tissue repair during bacterial infection. Immunity 2015, 42, 145–58. doi: 10.1016/j.immuni.2014.12.020 [DOI] [PubMed] [Google Scholar]
- 123.Lai SM, Sheng J, Gupta P, Renia L, Duan K, Zolezzi F, et al. Organ-specific fate, recruitment, and refilling dynamics of tissue-resident macrophages during blood-stage malaria. Cell Rep 2018, 25, 3099–3109.e3. doi: 10.1016/j.celrep.2018.11.059 [DOI] [PubMed] [Google Scholar]
- 124.Remmerie A, Martens L, Thoné T, Castoldi A, Seurinck R, Pavie B, et al. Osteopontin expression identifies a subset of recruited macrophages distinct from kupffer cells in the fatty liver. Immunity 2020, 53, 641–657.e14. doi: 10.1016/j.immuni.2020.08.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Ginhoux F, Bleriot C, Lecuit M.. Dying for a cause: regulated necrosis of tissue-resident macrophages upon infection. Trends Immunol 2017, 38, 693–5. doi: 10.1016/j.it.2017.05.009 [DOI] [PubMed] [Google Scholar]
- 126.Tran S, Baba I, Poupel L, Dussaud S, Moreau M, Gélineau A, et al. Impaired Kupffer cell self-renewal alters the liver response to lipid overload during non-alcoholic steatohepatitis. Immunity 2020, 53, 627–640.e5. doi: 10.1016/j.immuni.2020.06.003 [DOI] [PubMed] [Google Scholar]
- 127.Ferrer MF, Scharrig E, Charo N, Rípodas Ana L, Drut R, Carrera Silva EA, et al. Macrophages and galectin 3 control bacterial burden in acute and subacute murine leptospirosis that determines chronic kidney fibrosis. Front Cell Infect Microbiol 2018, 8, 384–384. doi: 10.3389/fcimb.2018.00384 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Lee HH, Aslanyan L, Vidyasagar A, Brennan MB, Tauber MS, Carrillo-Sepulveda MAet al. Depletion of alveolar macrophages increases pulmonary neutrophil infiltration, tissue damage, and sepsis in a murine model of acinetobacter baumannii pneumonia. Infect Immun 2020, 88, e00128-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Nancy Z, et al. Expression of factor V by resident macrophages boosts host defense in the peritoneal cavity. J Exp Med 2019, 216, 1291–300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Junqueira C, Crespo A, Ranjbar S, de Lacerda LB, Lewandrowski M, Ingber J, et al. FcγR-mediated SARS-CoV-2 infection of monocytes activates inflammation. Nature 2022, 606, 576–84. doi: 10.1038/s41586-022-04702-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Webster SJ, Daigneault M, Bewley MA, Preston JA, Marriott HM, Walmsley SR, et al. Distinct cell death programs in monocytes regulate innate responses following challenge with common causes of invasive bacterial disease. J Immunol 2010, 185, 2968–79. doi: 10.4049/jimmunol.1000805 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Gaidt MM, Ebert TS, Chauhan D, Ramshorn K, Pinci F, Zuber S, et al. The DNA inflammasome in human myeloid cells is initiated by a STING-cell death program upstream of NLRP3. Cell 2017, 171, 1110–1124.e18. doi: 10.1016/j.cell.2017.09.039 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Teh YC, Chooi MY, Liu D, Kwok I, Lai GC, Ayub Ow Yong L, et al. Transitional premonocytes emerge in the periphery for host defense against bacterial infections. Sci Adv 2022, 8, eabj4641. doi: 10.1126/sciadv.abj4641 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Massberg S, Schaerli P, Knezevic-Maramica I, Köllnberger M, Tubo N, Moseman EA, et al. Immunosurveillance by hematopoietic progenitor cells trafficking through blood, lymph, and peripheral tissues. Cell 2007, 131, 994–1008. doi: 10.1016/j.cell.2007.09.047 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Gold R, Denburg JA, Van Eeden SF.. Bone marrow progenitors in inflammation and repair: new vistas in respiratory biology and pathophysiology. Eur Respir J 2006, 27, 441–5. [DOI] [PubMed] [Google Scholar]
- 136.Schulz C, von Andrian UH, Massberg S.. Hematopoietic stem and progenitor cells: their mobilization and homing to bone marrow and peripheral tissue. Immunol Res 2009, 44, 160–8. doi: 10.1007/s12026-009-8109-6 [DOI] [PubMed] [Google Scholar]
- 137.Bozdag-Turan I, Turan RG, Turan CH, Ludovicy S, Akin I, Kische S, et al. Relation between the frequency of CD34+ bone marrow derived circulating progenitor cells and the number of diseased coronary arteries in patients with myocardial ischemia and diabetes. Cardiovasc Diabetol 2011, 10, 107–107. doi: 10.1186/1475-2840-10-107 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Schmid M, et al. Bone marrow dendritic cell progenitors sense pathogens via toll-like receptors and subsequently migrate to inflamed lymph nodes. Blood 2011, 118, 4829–4840. [DOI] [PubMed] [Google Scholar]
- 139.Serrano-Lopez J, Hegde S, Kumar S, Serrano J, Fang J, Wellendorf AM, et al. Inflammation rapidly recruits mammalian GMP and MDP from bone marrow into regional lymphatics. eLife 2021, 10, e66190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Lösslein AK, Lohrmann F, Scheuermann L, Gharun K, Neuber J, Kolter J, et al. Monocyte progenitors give rise to multinucleated giant cells. Nat Commun 2021, 12, 2027–2027. doi: 10.1038/s41467-021-22103-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Kratofil RM, Kubes P, Deniset JF.. Conversion during inflammation and injury. ATVB 2017, 37, 35–42. [DOI] [PubMed] [Google Scholar]
- 142.Marco Orecchioni et al.;. Macrophage polarization: different gene signatures in M1(LPS+) vs. classically and M2(LPS-) vs. alternatively activated macrophages. Front Immunol 2019, 10, 1084–1084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Nathan C, Murray HW, Wiebe ME, Berish YR, Rubin BY.. Identification of interferon-gamma as the lymphokine that activates human macrophage oxidative metabolism and antimicrobial activity. J Exp Med 1983, 158, 670–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Stein MB, et al. Interleukin 4 potently enhances murine macrophage mannose receptor activity: a marker of alternative immunologic macrophage activation. J Exp Med 1992, 176, 287–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Mosser DM. The many faces of macrophage activation. J Leukoc Biol 2003, 73, 209–12. doi: 10.1189/jlb.0602325 [DOI] [PubMed] [Google Scholar]
- 146.Mills CD, Kincaid K, Alt JM, Heilman MJ, Hill AM.. M-1/M-2 macrophages and the Th1/Th2 paradigm. J Immunol 2000, 164, 6166–73. doi: 10.4049/jimmunol.164.12.6166 [DOI] [PubMed] [Google Scholar]
- 147.Nahrendorf M, Swirski FK.. Abandoning M1/M2 for a network model of macrophage function. Circ Res 2016, 119, 414–7. doi: 10.1161/CIRCRESAHA.116.309194 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Gosselin D, Link VM, Romanoski CE, Fonseca GJ, Eichenfield DZ, Spann NJ, et al. Environment drives selection and function of enhancers controlling tissue-specific macrophage identities. Cell 2014, 159, 1327–40. doi: 10.1016/j.cell.2014.11.023 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Kim H-S, Wang SY, Kwak G, Yang Y, Kwon IC, Kim SH.. Exosome-guided phenotypic switch of M1 to M2 macrophages for cutaneous wound healing. Adv Sci 2019, 6, 1900513. doi: 10.1002/advs.201900513 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Alhamdi JR, Peng T, Al-Naggar IM, Hawley Kelly L, Spiller Kara L, Kuhn LT.. Controlled M1-to-M2 transition of aged macrophages by calcium phosphate coatings. Biomaterials 2019, 196, 90–9. doi: 10.1016/j.biomaterials.2018.07.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Zhang F, Parayath NN, Ene CI, Stephan SB, Koehne AL, Coon ME, et al. Genetic programming of macrophages to perform anti-tumor functions using targeted mRNA nanocarriers. Nat Commun 2019, 10, 3974–3974. doi: 10.1038/s41467-019-11911-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Guilliams M, Svedberg FR.. Does tissue imprinting restrict macrophage plasticity. Nat Immunol 2021, 22, 118–27. doi: 10.1038/s41590-020-00849-2 [DOI] [PubMed] [Google Scholar]
- 153.Hilgendorf I, Gerhardt LMS, Tan Timothy C, Winter C, Holderried TAW, Chousterman BG, et al. Ly-6chigh monocytes depend on Nr4a1 to balance both inflammatory and reparative phases in the infarcted myocardium. Circ Res 2014, 114, 1611–22. doi: 10.1161/CIRCRESAHA.114.303204 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Chen H-R, Sun Y-Y, Chen C-W, Kuo Y-M, Kuan IS, Tiger Li Z-R, et al. Fate mapping via CCR2-CreER mice reveals monocyte-to-microglia transition in development and neonatal stroke. Sci Adv 2020, 6, eabb2119. doi: 10.1126/sciadv.abb2119 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Kratofil RM, Shim HB, Shim R, Lee WY, Labit E, Sinha S, et al. A monocyte–leptin–angiogenesis pathway critical for repair post-infection. Nature 2022, 609, 166–73. doi: 10.1038/s41586-022-05044-x [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Data sharing is not applicable to this article as no datasets were generated or analysed in this paper.





