Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Jan 5.
Published in final edited form as: Circ Res. 2018 Jan 5;122(1):113–127. doi: 10.1161/CIRCRESAHA.117.311071

Resident and monocyte-derived macrophages in cardiovascular disease

Lisa Honold 1, Matthias Nahrendorf 1,2
PMCID: PMC5777215  NIHMSID: NIHMS920176  PMID: 29301844

Abstract

Macrophages are ubiquitous cells that reside in all major tissues. Counter to long-held beliefs, we now know that resident macrophages in many organs are seeded during embryonic development and self-renew independently from blood monocytes. Under inflammatory conditions, those tissue macrophages are joined and sometimes replaced by recruited monocyte-derived macrophages. Macrophage function in steady state and disease depends on not only their developmental origin but also the tissue environment. Here, we discuss the ontogeny, function and interplay of tissue-resident and monocyte-derived macrophages in various organs contributing to the development and progression of cardiovascular disease.

Keywords: Macrophage ontogeny, tissue-resident macrophages, cardiovascular disease, hematopoietic system, inflammation

Introduction

Over the last decade, sophisticated fate mapping, adoptive transfer and parabiosis techniques enabled experiments that challenge the hierarchical “mononuclear phagocyte system” proposed by Van Furth et al. in 19721. Immature cells in the bone marrow were thought to give rise to circulating monocytes that continuously migrated to peripheral tissues, where they differentiated into macrophages. We now know that many tissue macrophage populations arise during embryonic development, are seeded well before birth and are thereafter maintained by local proliferation rather than monocyte recruitment. Consequently, resident macrophages in many organs are completely or at least partly independent from blood monocytes generated by medullary or extramedullary hematopoiesis24.

Embryonic macrophage development occurs in several waves (reviewed in detail in5,6): In the first primitive hematopoiesis, myeloid progenitors arise in the yolk sac posterior plate mesoderm around embryonic day 7. A few days later, the yolk sac hemogenic endothelium gives rise to erythromyeloid precursors (EMPs) that can differentiate into fetal macrophages. As circulation establishes, EMPs subsequently migrate to the fetal liver where they differentiate into erythrocytes, megakaryocytes, granulocyte, mast cell and monocytes. At the same time, the first hematopoietic progenitors arise in the aorta-gonad-mesonephros, the major vitelline and umbilical arteries and the placenta and seed the fetal liver around day 10. Here, definite hematopoietic stem cells (HSCs) differentiate and thus make the fetal liver the main site of embryonic hematopoiesis from embryonic day 11 onwards. Since tissue macrophage populations arise from different waves of macrophage development, we describe their ontogeny – if known – at the beginning of each section.

In the adult, new monocytes are generated by hematopoiesis, a hierarchical differentiation program emanating from HSCs. In the mouse, two main monocyte subsets can be identified by their Ly6C expression levels. “Classical” Ly6Chigh monocytes differentiate from Ly6Chigh progenitors, while the “non-classical” or “alternative” Ly6Clow monocytes transdifferentiate from Ly6Chigh monocytes in a process that depends on the transcription factor Nr4a17. Under healthy conditions, Ly6Clow monocyte function closely associates with the vasculature, where they patrol along vessel walls and dispose of damaged endothelial cells8,9. “Classical” Ly6Chigh monocytes are recruited to sites of inflammation, where they differentiate into macrophages. Ly6Chigh monocytes can also infiltrate peripheral tissues under homeostatic conditions and have been shown to transport antigens to draining lymph nodes10. Human monocyte subsets can be identified by their expression of CD16 (Fc receptor) and CD14 (a coreceptor for LPS). The current working framework of monocyte subsets will likely become more complex, as emerging data suggest a continuum – rather than two or three defined states – between the subsets. The use of single cell RNA sequencing and mass cytometry (CyTOF) in conjunction with high-dimensional antibody panels has improved subset identification resolution and thus allowed unbiased detection of previously unknown monocyte and macrophage phenotypes in different tissues under homeostatic conditions11,12. Most of these phenotypes are still awaiting functional characterization, which is ultimately what makes a newly-described cell subset significant.

Both heterogeneous origins and distinct tissue microenvironments strongly shape resident macrophages’ expression profiles and functions. We see this clearly in splenic red pulp macrophages, which express the haem-induced transcription factor Spi-C that allows them to manage large amounts of iron13,14. Transplanting macrophages to a foreign environment can drastically change the gene expression profile and thus the function of the transplanted cells, thereby indicating that the tissue environment powerfully controls macrophage function. Transplanting peritoneal macrophages into the lung, for example, will induce a distinct alveolar macrophage expression profile in formerly peritoneal macrophages15. Similarly, monocyte-derived macrophages can take over the functions of embryonic tissue macrophages if the latter are lost e.g. due to irradiation or after myocardial infarction. Those newly-derived macrophages are able to self-maintain over long periods of time, thereby indicating that the bone marrow origin does not preclude macrophages from self-maintenance through local proliferation1618.

The cardiovascular system is constantly in intimate contact with the immune system, including circulating monocytes and tissue-resident immune cells in different organs. It has become clear that cardiovascular disorders involve and affect a variety of organ systems beyond the cardiovascular tissues, including the immune, hematopoietic and nervous systems. As such, risk factors of cardiovascular disorders are associated with changes in inflammatory response or in hematopoiesis. Obesity, for example, causes a chronic low-grade inflammation that procures the development of metabolic and cardiovascular co-morbidities19. Aging is associated with clonal hematopoiesis and a marked myeloid bias that may similarly increase the risk of cardiovascular disease20,21. In this manuscript, we discuss the multiple roles of and interplay between tissue macrophages and blood-derived monocytes/macrophages in organ systems relevant to cardiovascular disease. We also review macrophages that do not reside in the heart or vasculature to provide a wider context and highlight potential organ system interactions.

1) Heart and arteries

Myeloid cells in the murine heart constitute about 7-8% of non-cardiomyocytes16,22. Murine cardiac macrophages are commonly classified by their surface expression of C-C chemokine receptor type 2 (CCR2) and major histocompatibility complex II (MHC-II). Most cardiac macrophages are MHC-IIlow CCR2 and are derived from yolk sac progenitors23,24. The adult heart contains a separate MHC-IIhigh CCR2 macrophage population with predominantly embryonic origin. As cardiac MHC-IIhigh CCR2 macrophages increase after birth, they may arise from MHC-IIlow CCR2 cells23. The third and smallest macrophage population, which is CCR2+, derives from definitive hematopoiesis, i.e. from HSCs in the fetal liver24,23. Resident macrophages in the adult heart retain expression of the fractalkine receptor CX3CR12,25. Cardiac tissue macrophages constantly renew, with a turnover rate of approximately five weeks in steady state16. Parabiosis experiments performed by both Epelman et al. and Heidt et al. reported tissue macrophage renewal in the healthy heart to be mostly independent from blood monocytes16,23.

The homeostatic function of cardiac tissue macrophages remains somewhat unclear. Gene expression analysis suggests these macrophages for example play a role in angiogenesis and immune quiescence25. However, we still lack functional in vivo data that clearly define the role of cardiac macrophages in those processes. During development, CCR2+ and CCR2 macrophage populations occupy different cardiac locations associated with distinct functions24. CCR2 cells with embryonic ontogeny reside predominantly in the myocardial wall. They are important for normal development and, more specifically, development of the coronary vasculature. In contrast, CCR2+ macrophages, which are mostly found in the trabecular projections of the endocardium, are dispensable for cardiac development.

A recent study described macrophages in the heart’s conduction system, particularly the adult atrioventricular and sinus node26. These macrophages form direct connections to cardiomyocytes via connexin 43 (Cx43) gap junctions and depolarize in synchrony with the cardiomyocytes to which they connect. Interestingly, both Cx43-deficiency and macrophage depletion by diphtheria toxin injection in Cd11bDTR mice significantly impairs atrioventricular conduction. Macrophages enter a source-sink relationship with conducting cells and influence their action potential shape and duration. Taken together with reports that monocyte-derived macrophages possess functional ion channels2729, these data invite exploring macrophages’ role in conduction disorders and whether cells recruited to the myocardium under inflammatory conditions impact cardiac conduction. Considering that microglia are essential for synaptic pruning, it will be interesting to investigate if cardiac macrophages participate in the development of the cardiac conduction system.

Arterial macrophages likely originate from yolk sac-derived EMPs and one additional wave of blood-derived monocytes shortly after birth. Thereafter, arterial macrophages self-maintain both in steady state and after exposure to bacteria30. Like cardiac macrophages, arterial macrophages retain continuous CX3CR1 expression, which likely plays a role in their continuous survival and self-maintenance30.

While murine cardiac and arterial macrophages have been extensively studied, data on human cardiac macrophages, their phenotypes, subsets and function during homeostasis and in disease are still very sparse. Emerging technologies like CyTOF and subset-specific non-invasive clinical imaging approaches will hopefully aid in addressing those knowledge gaps.

Myocardial infarction

The temporal dynamics of different leukocyte populations were analyzed by flow cytometry of single cell suspensions at different time points after surgically-induced myocardial infarction in mice31. Neutrophils are among the first leukocytes to accumulate in ischemic cardiac tissue. Recently, Li et al. proposed a role for resident CCR2+ macrophages in initial neutrophil extravasation using a model of cardiac ischemia reperfusion and syngeneic heart transplantation32. In their study, chemoattractants C-X-C motif chemokine ligand 2 and 5 (CXCL2 and CXCL5) produced by CCR2+ resident macrophages contribute to transendothelial leukocyte migration into the ischemic area. Monocyte recruitment (Figure 1) may occur as early as 30 minutes after ischemia onset and depends on CCR2 signaling, predominantly via C-C chemokine ligand 2 (CCL2)31,3335. Within a few days, the infiltrating Ly6Chigh cells, which have a life span of only 20 hours, begin to differentiate into reparative Ly6Clow macrophages3638. Those reparative macrophages can persist for several weeks and renew partially by local proliferation36. Ly6Clow monocytes are also recruited during this reparative phase, albeit in much lower numbers and with unclear functional consequences36. In addition to the bone marrow, the spleen acts as a reservoir for monocytes after MI. Adrenergic signaling stimulates bone marrow HSC egress, consecutive seeding in the spleen and extramedullary hematopoiesis38,39. In the initial 24 hours post MI, the spleen releases its monocyte reservoir; consequently, as much as 40% of infiltrating Ly6Chigh monocytes in the infarct originate from the spleen40. Embryonic cardiac macrophages die locally shortly after MI and thus disappear from ischemic heart tissue38. In the chronic phase post MI, the majority of macrophages is again maintained by local proliferation, with a smaller contribution from circulating monocytes41.

Figure 1. Macrophages in cardiovascular disease.

Figure 1

During MI, atherosclerosis and stroke, monocytes are supplied by medullary and extramedullary hematopoiesis in bone marrow and spleen. Monocytes infiltrate diseased tissues, differentiate into macrophages and proliferate locally. In MI, resident cardiac macrophages are lost, while arterial macrophages in atherosclerosis persist. Cerebral microglia are activated after stroke and contribute to disease progression and healing. In obesity, macrophage accumulation in adipose tissue stems from local proliferation of resident and recruitment of monocyte-derived macrophages.

In addition to pursuing phagocytosis and efferocytosis, macrophages produce important mediators post MI that establish crosstalk with other cardiac cell types (Fig. 2). TNF-α, released by macrophages but also by cardiomyocytes and endothelial cells, induces cardiomyocyte hypertrophy and thereby increases the chance of heart failure post MI. In patients, high TNF-α levels predict impaired cardiac function and increased mortality42. Of note, in a genetic heart failure model TNF-α also protects cardiomyocytes from apoptosis43. Macrophages influence fibroblasts via TGF-β, that, together with additional mediators, induces their conversion to myofibroblasts. Myofibroblasts thereupon express α-smooth muscle actin and produce essential collagen44. Additionally, secreted proteolytic enzymes like matrix metalloproteinases (MMPs) degrade extracellular matrix and contribute to tissue remodeling and scar formation. Macrophage-derived VEGF acts on endothelial cells and stimulates angiogenesis31.

Figure 2. Macrophage mediators and crosstalk after myocardial infarction.

Figure 2

TNF-α acts on cardiomyocytes and can induce hypertrophy and cell death. TGF-β induces conversion of fibroblasts to myofibroblasts that produce collagen necessary for scar formation. Proteolytic enzymes like matrix metalloproteinases (MMPs) contribute to tissue remodeling while VEGF acts on endothelial cells and stimulates angiogenesis.

In total, monocytes/macrophages promote infarct healing, as macrophage depletion drastically impairs healing and worsens disease outcome45,46. However, systemically increased monocyte/macrophage numbers, as in ApolipoproteinE-deficient (ApoE−/−) or chemokine Decoy Receptor D6-deficient mice, can be detrimental to infarct healing and remodeling47,48. Further, reducing the number of infiltrating monocytes, e.g. by limiting B-cell mediated mobilization or therapeutically reducing recruitment via CCR2 blockage, decreases infarct size and supports post-MI recovery in animal models34,49. We hypothesize that therapeutic approaches limiting the inflammatory monocyte supply to the heart can improve post MI recovery in patients with high systemic inflammatory activity.

Post-MI heart failure

While the initial fibrosis post MI is necessary to aid stable scar formation and prevent ventricular rupture, extensive fibrosis can result in pathological cardiac tissue remodeling and long-term heart failure50,51. The failing heart contains increased number of macrophages that expand by both local proliferation and further recruitment16,41. The recruited monocytes originate from medullary and extramedullary hematopoiesis and have a net detrimental effect41,52. Consequently, splenectomy reduces inflammatory infiltrates and improves cardiac geometry and function52. The expanded cardiac macrophage population plays an important role in the progression of heart failure by secreting factors that directly or indirectly stimulate continued fibrosis (reviewed in53): TGF-β directly induces expression of pro-fibrotic genes in myofibroblasts, while indirect factors like angiotensin-II, PDGF or endothelin-1, produced by macrophages, endothelial cells and fibroblasts, further stimulate the release of TGF-β from myofibroblasts and cardiomyocytes.

Cardiac regeneration

Certain fish and amphibians can regenerate cardiac tissue following injury throughout adult life54. In zebrafish, for example, the heart is able to fully regenerate after resection of approximately 20% of the left ventricle55. The de novo generated tissue is produced by cardiomyocyte proliferation56,57.

In rodents, the neonatal heart can regenerate in response to multiple injuries, including partial surgical resection, myocardial infarction and cryoinjury5860. This regenerative ability depends on the presence of macrophages and, more specifically, on expansion of the embryo-derived CCR2 macrophage population61,62. Inflammatory response and regenerative potential are regulated via both parasympathetic and sympathetic nerve activity. Either pharmacological inhibition of cholinergic nerve activity or surgical left vagus nerve ablation impairs cardiac regeneration in neonatal mice63. Likely influential factors are, on the one hand, direct cell cycle regulators like Ccnd2 and Cdk4 and, on the other hand, inflammatory genes that are downregulated upon vagotomy. A study by White et al. implies involvement of sympathetic innervation as chemical sympathectomy by 6-OHDA treatment similarly impairs regeneration64.

The adult murine heart lacks this regenerative ability due to a reduced capacity of adult cardiomyocytes to re-enter the cell cycle65. Cardiac regenerative capacity may be modified by sympathetic and parasympathetic nerve activity, proper vascular regeneration65 and in part also due to an inflammatory monocyte/macrophage response that changes in the first two weeks after birth62. While macrophage expansion in neonates results from CCR2 subset proliferation, this embryonic population is lost in adult hearts after cardiac injury and is instead replaced by CCR2+ blood monocytes. Consequently, limiting CCR2+ influx by administering a CCR2-inhibitor can preserve embryonic tissue macrophages in the adult heart and thus improve cardiac repair62.

Hypertension

Following angiotensin II-induced hypertension, the cardiac Ly6Chigh monocyte/macrophage population expands by both blood monocyte recruitment and local tissue macrophage proliferation23. Additionally, angiotensin II-induced hypertension results in pathogenic macrophage accumulation in the adventitia and perivascular adipose tissue, mediated by CCR26668. Reducing macrophage numbers by inhibiting CCR2 may provide a strategy to lessen vascular remodeling and, consequently, hypertension69. At the same time, the inflammatory response might be influenced by CD11b+ Gr1+ myeloid-derived suppressor cells (MDSC), as depleting this cell population was reported to elevate the inflammatory hypertensive response and increases organ damage70. More precise cellular analysis is warranted to reveal which myeloid cells or subsets are protective in hypertension, as MDSC is an overly broad term that even includes neutrophils. Overall, the role of tissue-resident and monocyte-derived macrophages in the genesis and complications of hypertension is incompletely understood.

Atherosclerosis

Macrophages are critically involved in all stages of atherosclerosis, a chronic inflammatory disorder, affecting large- and medium-sized arteries, that results in acute myocardial infarction and stroke. Atherosclerosis is characterized by the formation of atherosclerotic plaques, which are vessel wall accumulations of lipids and immune cells covered by a fibrous cap composed of collagen. Macrophage contribution to atherosclerotic lesion formation and progression is extensively described elsewhere7174. In short, lesions typically start out with subendothelial lipid deposition and leukocyte recruitment. Macrophages in the lesion take up high amounts of lipoproteins and become large foam cells (Fig. 1). Those “fatty streak” lesions subsequently progress to larger plaques containing a necrotic core and a fibrous cap of varying stability. Continuous plaque growth can lead to vessel lumen narrowing and surrounding tissue ischemia. Organ ischemia occurs if the fibrous cap ruptures and the necrotic core is exposed, resulting in thrombosis at the site of the plaque or, in case of detachment, a traveling embolus that obstructs blood flow into peripheral tissues. Plaque erosion is a third process that results in thrombus formation, vessel occlusion and organ ischemia. Erosion is characterized by a loss of endothelial cells and thrombus formation in the absence of plaque rupture75,76. In humans, eroding plaques contain high amounts of fibrotic tissue and smooth muscle cells and can often be found in regions of disturbed flow. Recently, Franck et al. elicited superficial plaque erosion in mice by endothelial injury and subsequent local flow perturbation77. As in human eroding plaques, blood flow mediation in this setup disturbs the endothelium and causes neutrophil accumulation that is mediated by arterial TLR2 expression.

Monocytes recruited to atherosclerotic lesions are Ly6Chigh cells that originate from both medullary and extramedullary hematopoiesis78. The major chemokine receptors mediating monocyte transmigration are CCR2, CX3CR1 and CCR5; and deleting all three drastically reduces atherosclerosis in ApoE−/− mice79. It was recently reported that smooth muscle cell-derived cells in atherosclerotic lesions express a number of macrophage markers, including CD11b, F4/80 and Mac38082. These markers likely arise due to a phenotype switch from smooth muscle cells to macrophages or foam cells. That such a switch in cell identity contributes substantially to the macrophage population in atherosclerotic lesions is difficult to reconcile with parabiosis data demonstrating that all plaque macrophages derive from circulating monocytes78. Curiously, the smooth muscle cell-derived “macrophage-like” cells do not express the leukocyte marker CD4580. We believe the relative contribution of phenotype transition from smooth muscle cells to macrophage-like cells is currently unresolved (and quite possibly minor). We further posit that the pathophysiological importance of this putative lineage transition requires clarification.

Parabiosis studies have shown that the monocyte expansion mechanism in atherosclerosis depends on plaque characteristics and state. While newly developed lesions mostly recruit monocytes from the blood, more advanced lesions rely primarily on local macrophage proliferation, with a smaller contribution from new monocyte recruitment83. However, even the locally proliferating macrophages derive from cells recruited from the circulation. How exactly tissue-resident arterial macrophages, of embryonic or neonatal origin, that continuously proliferate in steady state interact with the monocyte population in the atherosclerotic lesion remains to be seen.

Even advanced atherosclerotic lesions can regress. In mice, this phenomenon has been investigated in a variety of settings. Infusion of the human ApoA-I variant improves cholesterol efflux and decreases foam cell formation and macrophage content in ApoE−/− mice84. Likewise, introducing ApoA-I via adenoviral gene therapy causes fast regression of atherosclerosis85,86. In the so-called Reversa mouse, physiological blood LDL levels can be reestablished by Cre-mediated inhibition of liver lipoprotein production87,88. Alternatively, ApoE−/− aortas have been transplanted into wild type mice8991. In those settings of plaque regression, the number of plaque macrophages declines and the remaining cells assume reparative, less inflammatory phenotypes. Rahman et al. found that, after aortic transplantation, the mostly reparative macrophages in regressing lesions originate from the circulation, thereby indicating that even in the context of plaque regression new monocytes infiltrate the arterial wall. Those newly recruited macrophages arise from Ly6Chigh monocytes that infiltrate in a CCR2-dependent manner and subsequently differentiate into macrophages in a STAT6-regulated differentiation program91. The confounding setting of arterial transplantation, the surgically-induced inflammation and the sudden, steep decrease in LDL cholesterol will require careful clinical studies to evaluate if this scenario translates to patients with regressing atherosclerosis.

While not directly targeting macrophages but rather one of their inflammatory products, the recent Canakinumab Antiinflammatory Thrombosis Outcome Study (CANTOS) confirmed that modifying inflammation in patients with atherosclerosis is a viable strategy. CANTOS investigated the effect of Interleukin-1β inhibition in patients with previous myocardial infarction and increased high-sensitivity C-reactive protein levels. The treatment reduced inflammatory blood parameters in a dose-dependent manner and lowered circulating leukocyte numbers92,93. Notably, canakinumab treatment decreased the incidence of cardiovascular events as compared to placebo by 15%, thus emphasizing the immune system as a highly promising therapeutic intervention point in cardiovascular disease.

2) Bone marrow

Distinct macrophage populations, with so far unresolved ontogeny, reside in specific tissue niches in the bone marrow. In steady state conditions, these macrophages most likely self-renew; however, if they are depleted by irradiation, the lost cells can be replenished by bone marrow myelopoiesis17. Macrophages can be found within the bone marrow hematopoietic stem cell endosteal and perivascular niches. These niches are characterized by the presence of long term HSCs as well as stromal cells, including perivascular Nestin+ mesenchymal stromal cells (MSCs), osteoblasts, CXCL12-abundant reticular cells, endothelial cells and the aforementioned macrophages94,95. In addition, macrophages are located in erythroblast islands.

Bone marrow macrophages anchor hematopoietic stem and progenitor cells (HSPCs) in the hematopoietic niche. Consequently, macrophage depletion results in HSPC mobilization from the bone marrow and increased numbers of circulating HSPCs. This was observed after genetic macrophage depletion, clodronate-liposome depletion and DT treatment of CD169DTR mice96,97. Consistent with this framework, macrophage depletion downregulates the expression of HSPC retention factors such as CXCL12, angiopoietin-1, Kit ligand and VCAM-1 in the bone marrow and specifically in Nestin+ MSCs97. As those depletion strategies act systemically, rather than tissue-specifically, and macrophages have been reported to similarly anchor HSPCs in the spleen, the increase in circulating HSPCs may in part result from depletion in extramedullary tissues98. Surprisingly, macrophage depletion may also result in accumulation of quiescent and proliferative HSCs in the BM, which is inconsistent with the idea of macrophages acting as an anchor99.

Bone marrow macrophages are also important for regulating granulopoiesis. Circulating neutrophils have very short lifetimes of only 13 hours and are thus constantly cleared and produced in large quantities100. Elimination of aged neutrophils in the bone marrow follows a circadian rhythm and is mediated by macrophages that phagocytose the aged cells and in turn induce release of hematopoietic progenitor cells via liver X receptor (LXR) signaling101,102. Macrophages are thus an active part of a circadian feed-back-loop that maintains steady state neutrophil numbers.

Bone marrow in cardiovascular disease

Following myocardial ischemia, the hematopoietic system is activated, and HSC and HSPC proliferation in the BM increases, as does their migration into the blood stream. The elevated proliferation rate is attributed to CCR2+ HSPCs and their progeny and depends on expression of the transcriptional co-repressor myeloid translocation gene on chromosome 16 (Mtg16)103. Interestingly, after MI CCR2+ HSPCs also raise the expression of toll-like receptors 2 and 4 (TLR2, TLR4), which may sense danger associated molecular patterns (DAMPs) released from the damaged myocardium.

While the actual endogenous ligand signaling via TLRs post MI remains undefined, there are several factors that could mediate the communication between ischemic cardiac tissue and the BM, including the DAMPs High-mobility group box 1 (HMBG1), alarmins S100A8/9 and IL-1β. If and how stromal cell-like resident macrophages contribute to DAMP detection and TLR signaling in the bone marrow remains to be determined. HMBG1 increases in the circulation of MI patients and inversely correlates with disease outcome and survival104,105. HMBG1 is secreted by macrophages as a delayed mediator of inflammation and signals through the receptor for advanced glycation end-products (RAGE), TLR2 and TLR4106. Injecting HMBG1 in mice enhances CCR2+ HSPC proliferation103. Alarmins S100A8/A9, which similarly signal through RAGE, TLR2 and TLR4, also have higher concentrations in MI patient myocardium and serum107109. Further, elevated S100A8/A9 levels associate with higher blood leukocyte numbers108. The inflammatory cytokine IL-1β similarly increases in MI patient circulation110. After coronary ligation in mice, antibody-mediated inhibition of IL-1β suppresses HSPC proliferation in the bone marrow. In this case, not only direct signaling via the IL-1 receptor expressed by HSPCs but also indirect effects via bone marrow stromal cells, possibly including macrophages, are critical to the effect of IL-1β111.

3) Spleen

The spleen is home to at least four distinct resident macrophage populations that all fulfill specific functions in their anatomical niches: marginal, metallophilic, red pulp and white pulp macrophages.

The marginal zone, surrounding the white pulp, contains both marginal and metallophilic macrophages with so far undefined ontogeny. Because it is difficult to distinguish between marginal and metallophilic macrophages, many of the described functions are attributed to both cell types. In general, both populations are responsible for filtering pathogens from the circulation and subsequently producing cytokines to stimulate immune responses. To identify pathogens, those macrophages express a variety of pattern recognition receptors, including SIGNR1112. Resident macrophages regulate B-cell development, retention and function within the marginal zone113,114; relatedly, proper macrophage function depends on B-cells, as macrophages do not express SIGNR1 in the absence of B-cells115.

Red pulp macrophages seed prenatally from the fetal liver and self-maintain independently from blood monocytes2,17. Critical for HSPC retention in the spleen via adhesion molecule VCAM-198, red pulp macrophages are also key to regulating red blood cell homeostasis as they phagocytose aged erythrocytes retained on their way through the red pulp116.

Spleen in cardiovascular disease

In addition to the macrophages described above, the splenic red pulp contains a population of monocytes found in subcapsular clusters40. Upon myocardial ischemia, those monocytes undergo angiotensin II-dependent mobilization and migrate to the ischemic tissue. Indeed, a major portion of monocytes in the heart within the first few days after myocardial infarction originate from the spleen38,40. Consequently, the splenic monocyte reservoir is rapidly exhausted and subsequently refilled by differentiation of splenic macrophage/dendritic cell precursors (MDP)38. Similar to acute myocardial ischemia, extramedullary myelopoiesis and monocyte recruitment from the spleen have been reported in chronic ischemic heart failure, atherosclerosis and stroke38,52,117119.

4) Adipose tissue

Adipose tissue is generally divided into white (WAT), beige and brown adipose tissue (BAT). WAT and BAT consist of specific adipocytes (white, beige and brown) and fulfill distinct tasks at certain locations in the body (reviewed in120122). One specific depot is the perivascular adipose tissue (PVAT), which can have characteristics of either white or brown adipose tissue depending on its location. While WAT locates throughout the body in humans, BAT is solely localized in the neck, above the claviculae and around the spine. PVAT surrounds middle- and large-sized blood vessels.

In non-obese animals, WAT contains a variety of immune cells, including T-cells (natural killer cells, Th2 cells and regulatory cells), eosinophils and macrophages. Adipose tissue macrophages are at least in part of embryonic origin and are maintained by local proliferation in addition to recruitment, which occurs, for example, in obesity123126. In steady state, the immune cells within WAT create an anti-inflammatory environment crucial to homeostatic metabolism127. Resident macrophages’ role in regulating insulin sensitivity requires alternative polarization by activation of Peroxisome proliferator-associated receptor gamma (PPARγ) and is mediated by signal transducer and activators of transcription 6 (STAT6)128131. In a paracrine loop, Th2 cytokines, including interleukins IL-4 and IL-13, released by adipocytes and eosinophils act on tissue macrophages128,132. Macrophage-specific PPARγ-deficiency results in insulin resistance and tissue inflammation128. Complementarily, administering IL-4 protects mice from insulin resistance and obesity131,133. In obese mice, the number of macrophages in WAT drastically increases, and the macrophage population switches from an alternative to a more classical inflammatory activation state124,134. The resulting chronic low-grade inflammation is believed to play an essential role in metabolic disorders127,135.

Adipose tissue in cardiovascular disease

Obesity is a major risk factor of cardiovascular disease. More specifically, obesity can include systemic inflammation, hyperlipidaemia and inflammatory adipokine production, all of which can initiate atherosclerosis and are thus considered risk factors136,137. PVAT has been particularly implicated in the development and progression of atherosclerosis. PVAT, like other adipose tissues, is considered an endocrine organ and produces a number of molecules that can act on vessels in a paracrine fashion138. As in WAT, the number of immune cells in PVAT increases in obesity. Transplanting PVAT – but not WAT – into ApoE−/− mice leads to increased formation of atherosclerotic plaques at the site of implantation, thereby suggesting inflamed PVAT has pro-atherosclerotic properties139. On the other hand, brown adipose tissue characteristics of PVAT may protect from atherosclerosis. Lack of PVAT in ApoE−/− mice with conditional knock-out of peroxisome proliferator-activated receptor-c in vascular smooth muscle cells (SMPG-KO) causes reduced prostacyclin production in response to cold, which in turn might stimulate endothelial dysfunction and promote atherosclerosis140.

In human samples, PVAT close to atherosclerotic plaques contains high numbers of macrophages141, specifically at the interface between adventitia and PVAT142. The leukocyte infiltration likely results from increased expression of chemoattractants, including IL-8 and MCP-1142,143. Additionally, the volume of human pericoronary adipose tissue, covering the larger branches of the coronary arteries, increases with age. Independent of the volume of visceral adipose tissue, pericoronary adipose tissue correlates with coronary artery calcification, giving rise to the view that PVAT may locally promote atherogenesis144,145.

5) Brain

The best-characterized tissue resident macrophages in the brain are parenchymal microglia, that originate pre-birth, likely from RUNX1+ yolk sac-derived progenitors around embryonic day 7.253,4. Microglia are independent from blood monocytes as they self-renew in a process requiring colony-stimulating factor 1 receptor (CSF1R) signaling. If they are depleted by irradiation, microglia are partly replenished by infiltrating blood monocytes3,146,147. Likewise, depletion of microglia by intracerebroventricular administration of ganciclovir in mice expressing the herpes-simplex virus suicide-gene thymidine kinase under the CD11b-promoter (CD11b-HSVTK) causes replenishment of microglia by monocyte-derived cells148,149. On the contrary, partial depletion of microglia in tamoxifen and DT treated CX3CR1CreERRosa26DTR mice or by application of CSF1R-inhibitor is followed by rapid proliferation of remaining microglia, rather than by recruitment of peripheral myeloid cells150,151.

Under homeostatic conditions, microglia in the brain are ramified cells with a small stationary cell body. Their cell processes constantly scan the surrounding area and communicate with neighboring cells including neurons, astrocytes and endothelial cells. During neuronal development, microglia are essential for the survival and differentiation of neurons due to production of different neurotrophic factors, including IGF-1152. Microglia further mediate controlled neuronal cell death by non-inflammatory phagocytosis, synaptic pruning and synapse function153156.

In addition to microglia, the brain hosts at least three more resident macrophage populations, all with distinct expression profiles. Despite their differences, parenchymal microglia and non-parenchymal macrophages in the brain are closely related, and all are distinct from peripheral macrophages in terms of expression profiles157. Surface markers of brain macrophage populations are closely related, which poses a considerable hurdle to separating these populations. Non-parenchymal macrophages are best characterized by their localization in the brain and their closest neighbors. Perivascular macrophages can be found next to endothelial cells, between laminin-positive endothelial and glial basement membranes. Leptomeningeal and choroid plexus macrophages, as the names suggest, are found in the meningeal space and the choroid plexus, respectively158. Perivascular and leptomeningeal macrophages are long-lived and self-renewing cells replenished only by blood monocytes after irradiation159. In contrast, choroid plexus macrophages have a slower turnover that at least partly depends on blood monocytes. Perivascular and leptomeningeal macrophages are antigen-presenting cells activated in response to inflammation and neuronal injury160162. Their choroid plexus brethren are believed to play a role in immunosurveillance via phagocytosis of cellular debris and antigens in the cerebral spinal fluid158.

Brain macrophages in cardiovascular disease

Upon cerebral ischemia, microglia react to DAMPs released by dying cells and subsequently produce cytokines, chemokines and other signaling molecules that recruit peripheral immune cells to the brain. Following microglia activation, peripheral blood monocytes, dendritic cells and neutrophils start infiltrating the brain within one day and stay for several days post stroke163. The temporal dynamics of immune cell infiltration as well as the role of recruited monocyte-derived macrophages in the progression of stroke are still discussed controversially and likely vary depending on the employed animal model and experimental set up.

Preventing monocyte infiltration by depleting peripheral monocytes/macrophages before middle cerebral artery occlusion (MCAO) was reported to be beneficial164,165. In those studies, clodronate liposomes or CCR2 knockout decreased microglial activation and brain atrophy and improved overall recovery, indicating a negative role for monocyte-derived macrophages in the progression of stroke. However, preventing monocyte influx by clodronate treatment one and two days after induction of stroke by MCAO or photothrombosis increased cerebral hemorrhage and severely impaired neurological recovery166. Yet another study detected no therapeutic effects of either clodronate treatment or prevention of monocyte infiltration by anti-CCR2 antibody application starting 24 hours before induction of stroke by photothrombosis167.

In contrast, studies selectively depleting microglia in the context of stroke have mostly reported detrimental effects. Microglia depletion in CD11b-HSVTK mice in the context of stroke caused larger infarcts with higher excitotoxicity and altered neuronal activity168. Similarly detrimental, microglia depletion by CSF1R-inhibitor treatment three weeks prior to MCAO increased the size of the ischemic area, resulted in infiltration of more myeloid cells and exacerbated the pro-inflammatory astrocyte response169. These studies indicate that microglia, despite their potentially inflammatory profile, might protect neurons from excitatory stress and death by limiting inflammation following ischemia. Overall, microglia and macrophage response to stroke is likely a dynamic process, similar to the one following MI, that thus depends on the stroke model, mouse strain, timing, response amplitude and cell phenotype.

Little is known about other brain-resident macrophages in the context of CVD. Perivascular macrophages may play a role in hypertension, specifically the neurological symptoms and cognitive impairment associated with chronic hypertension. Depleting perivascular macrophages by injecting clodronate-liposomes into the cerebral ventricles decreases oxidative stress and restores cognitive functions in angiotensin II-induced hypertension170.

6) Tools for studying macrophages

Lineage tracing

Lineage tracing has been used to investigate the developmental origins of tissue macrophages. For those studies, genetically modified mice carrying a fluorescent reporter gene downstream of a loxP-flanked Stop-cassette and under a ubiquitously active promotor (e.g. Rosa-loxP-STOP-loxP-YFP mice) were crossed with inducible Cre-lines. A frequently-used model is the tamoxifen-inducible CX3CR1CreER line. If crossed with the reporter, tamoxifen injection in pregnant mice activates fluorescence in all CX3CR1 expressing embryonic cells and their progeny2,4,159. Consequently, if the induction is stopped before the onset of hematopoiesis, blood-derived monocytes will not express the reporter and can thus be distinguished from fluorescent tissue macrophages that arise early during embryonic development. In addition to CX3CR1CreER, CSF1RCreER and RUNX1CreER have been used to label macrophages originating in the embryonic yolk sac3,23,171. All monocytes and macrophages that arise specifically from adult hematopoiesis can similarly be labeled, for example with an inducible FLT3CreER line17,172,23. The limitations of lineage tracing experiments mirror the limitations of the respective Cre-line, i.e. the specific activity and leakiness in other cells. For instance, LysMCre, frequently used to delete genes in macrophages, monocytes and neutrophils, expresses Cre in the interventricular septum during development. Hence, a targeted gene may be excised in septal cardiomyocytes also173. Care must be taken to consider that Cre reports on the gene expression history and not necessarily on the precise lineage.

Depletion strategies

Depleting macrophages and distinct sub-populations allows researchers to investigate cell function by analyzing the consequences of cell absence. Clodronate-liposomes, which are devoured by macrophages and induce cell death, are perhaps the easiest way to deplete macrophages and have been used for many years45,174. Using this approach, however, it is nearly impossible to selectively target specific macrophage populations, as the systemic clodronate injection eliminates macrophages regardless of origin throughout the body. An exception might be locally injecting clodronate-liposomes into the cerebral ventricles, an approach that has been applied to specifically eliminate perivascular macrophages in the brain170. The technique’s limitations, including limited efficiency in some tissues such as the steady state myocardium, can be at least partly overcome by applying inducible genetic depletion strategies. In genetically-modified mice that express the diphtheria toxin receptor (DTR) under a macrophage-specific promotor, DT injection results in death of the targeted cell population. For example, CD11bDTR mice have been used to study the effects of cardiac macrophage depletion and CD169DTR mice to deplete bone marrow macrophages26,96,97. While this technique has a higher specificity than clodronate injection, DTR knock-in mice are limited by the specificity of the chosen genetic marker. As only very few markers are exclusive for distinct macrophage populations, DT injection in most studies depletes mixed macrophage populations in many organs. An organ-specific macrophage depletion strategy is sorely needed and would be extremely useful for studying distinct tissue macrophage functions.

Therapeutic targeting

Reducing the blood-derived monocyte infiltration to limit inflammation or atherosclerotic plaque progression has proven beneficial in mice after myocardial infarction and atherosclerosis. Pre-clinical models showed promising results by targeting different adhesion molecules, including intercellular adhesion molecules (ICAMs), vascular cell adhesion molecules (VCAMs), selectins and C-C chemokine receptors49,175,176. In addition to recruitment, studies have also targeted macrophages’ inflammatory status. Silencing transcription factor Interferon Regulatory Factor 5 (IRF5) in cardiac macrophages post MI generated encouraging outcomes37. Nanoparticle-based delivery of siRNA targeting IRF5 reduced inflammatory macrophage markers in the infarct, supported infarct healing and prevented heart failure. In atherosclerotic plaques, inflammatory status and macrophage proliferation have successfully been modified by targeted administration of statins incorporated into nanoparticles177,178. Similarly, local anti-inflammatory treatment with glucocorticoids incorporated into small liposomes that are taken up by plaque macrophages showed positive effects on plaque size in a rabbit atherosclerosis model179. To date these preclinical results have not been successfully translated into the clinic, as prednisolone-liposomes accumulated in human atherosclerotic plaque macrophages but did not elicit any significant anti-inflammatory effects180.

Monitoring

Monitoring macrophages is necessary to track the success of macrophage-targeting therapies. In addition, macrophage monitoring answers fundamental questions about the development, location and spatio-temporal dynamics of macrophage populations in steady state and disease. Intravital microscopy (IVM) is a powerful tool for distinguishing and following individual cells in vivo. Using fluorescent reporter lines, individual macrophages can be imaged over time to follow their interactions with other cells. The CX3CR1GFP mouse has been used to visualize CX3C31+ macrophages by IVM in the heart, spleen and atherosclerotic plaques40,103,181,182. Alternatively, cells can be labeled by injecting fluorophore-labeled antibodies, as is routinely done to visualize different cell types in the bone marrow183,184. Macrophages’ high phagocytic activity also allows macrophage imaging with fluorescently-labeled nanoparticles181.

In human atherosclerotic patients, nuclear imaging can allow non-invasive assessment of plaque development and progression and can ideally distinguish between stable and high-risk lesions. Radioactively-labeled glucose (18F-Fluoro-Deoxy-Glucose (FDG)) is the most widely applied radiotracer to image atherosclerosis by positron emission tomography (PET) in clinical studies185.18F-FDG-PET imaging of macrophages in atherosclerotic lesions has limitations, including uptake by other metabolically active cells and the possible influence of plaque core hypoxia on18F-FDG signals185187. Therefore, while detecting atherosclerotic lesions with18F-FDG-PET is possible, the resulting images do not specifically show macrophages.

Quite similar to optical imaging, macrophages in atherosclerotic lesions or after MI can be targeted by nanoparticles that are labeled with radioisotopes and can therefore be detected by PET188. In fact, the same nanoparticle is often labeled with radioisotopes and fluorophores, thus allowing non-invasive PET imaging and its validation on a cellular level using the same particle181,189. Macrophages have also been targeted more specifically, e.g. via radioactively-labeled small molecules or nanoparticles binding to CCR2 and CCR5, which have been successfully applied to visualize plaque macrophages190,191. We do however still lack a clinical imaging probe that offers high prognostic value by reliably distinguishing among different plaque characteristics (i.e. stable, rupture-prone, regressing).

Overall, future advances will need to focus on developing imaging probes that monitor of macrophage subsets as well as dynamic changes in cell phenotype. While there have been attempts to develop probes that specifically bind to pro-inflammatory macrophages in atherosclerotic lesions (e.g. 68Ga-DOTATATE binding to somatostatin receptor 2), the specificity of those probes needs further validation192,193.

In the brain, activated microglia have been monitored using radiotracers targeting the translocator protein TSPO194,195. One of the PET tracers targeting TSPO is 18F-DPA-714, which has been used to image neuroinflammation in mice with cerebral ischemia, with signals peaking two weeks post stroke196. While there is very low TSPO expression in the healthy brain, TSPO is expressed not only by activated microglia but also by astrocytes and infiltrated peripheral immune cells in a variety of neurological disorders197. Therefore, specific non-invasive monitoring of brain microglia remains a challenge.

7) Conclusions and future directions

This is an exciting time: increased availability of avant-garde technologies transforms research and accelerates knowledge gain that hopefully will lead to new cures soon. Enabled by interdisciplinary collaborations, we begin to understand the interactions among the innate immune, hematopoietic and cardiovascular systems. Of particular interest is myelopoiesis, i.e. the production of monocytes, which can give rise to inflammatory disease-promoting macrophages. We study how cardiovascular risk factors and comorbidities shape hematopoiesis, and these insights will become a springboard for therapeutic development. New tools now allow relatively easy genetic manipulation of the hematopoietic system. CRISPR/Cas9 mediated gene editing to modify human and murine HSPCs creates new opportunities for basic research and, in the near future, cardiovascular therapeutics198201. Single cell sequencing and CyTOF provide unbiased discovery tools that can be applied to discover many new cell subsets, all of which will have to be tested for functional diversity. Intravital microscopy and non-invasive macrophage imaging are on the brink of broader adoption in basic and clinical research, respectively. In general, a more nuanced, well-resolved and higher definition understanding of macrophage functions will enable targeting of disease-promoting cellular functions while sparing macrophage activities or subsets that are essential for defending homeostasis. The results of the first successful large scale clinical study on IL-1β neutralization are encouraging and hopefully only the beginning of a new era in cardiovascular immunotherapy93.

Acknowledgments

We thank Kaley Joyes, PhD, for editing the manuscript. Figures were designed using Servier Medical Art (www.servier.com).

Sources of Funding

This work was funded in part by grants from the German Research Foundation (HO 5953/1-1), the National Heart, Lung, and Blood Institute (HL096576, HL117829, HL128264) and the MGH Research Scholar Award.

Footnotes

Nonstandard Abbreviations and Acronyms

None.

Disclosures

None.

References

  • 1.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]
  • 2.Yona S, Kim KW, Wolf Y, Mildner A, Varol D, Breker M, Strauss-Ayali D, Viukov S, Guilliams M, Misharin A, Hume DA, Perlman H, Malissen B, Zelzer E, Jung S. 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]
  • 3.Ginhoux F, Greter M, Leboeuf M, Nandi S, See P, Gokhan S, Mehler MF, Conway SJ, Ng LG, Stanley ER, Samokhvalov IM, Merad M. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science. 2010;330:841–5. doi: 10.1126/science.1194637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Sheng J, Ruedl C, Karjalainen K. Most Tissue-Resident Macrophages Except Microglia Are Derived from Fetal Hematopoietic Stem Cells. Immunity. 2015;43:382–393. doi: 10.1016/j.immuni.2015.07.016. [DOI] [PubMed] [Google Scholar]
  • 5.Ginhoux F, Guilliams M. Tissue-Resident Macrophage Ontogeny and Homeostasis. Immunity. 2016;44:439–449. doi: 10.1016/j.immuni.2016.02.024. [DOI] [PubMed] [Google Scholar]
  • 6.Hoeffel G, Ginhoux F. Ontogeny of tissue-resident macrophages. Front Immunol. 2015;6 doi: 10.3389/fimmu.2015.00486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Hanna RN, Carlin LM, Hubbeling HG, Nackiewicz D, Green AM, Punt JA, Geissmann F, Hedrick CC. 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]
  • 8.Auffray C, Fogg D, Garfa M, Elain G, Join-Lambert O, Kayal S, Sarnacki S, Cumano A, Lauvau G, Geissmann F. 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]
  • 9.Carlin LM, Stamatiades EG, Auffray C, Hanna RN, Glover L, Vizcay-Barrena G, Hedrick CC, Cook HT, Diebold S, Geissmann F. Nr4a1-dependent Ly6C(low) monocytes monitor endothelial cells and orchestrate their disposal. Cell. 2013;153:362–75. doi: 10.1016/j.cell.2013.03.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Jakubzick C, Gautier EL, Gibbings SL, Sojka DK, Schlitzer A, Johnson TE, Ivanov S, Duan Q, Bala S, Condon T, vanRooijen N, Grainger JR, Belkaid Y, Ma’ayan A, Riches DWH, Yokoyama WM, Ginhoux F, Henson PM, Randolph GJ. 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]
  • 11.Becher B, Schlitzer A, Chen J, Mair F, Sumatoh HR, Teng KWW, Low D, Ruedl C, Riccardi-Castagnoli P, Poidinger M, Greter M, Ginhoux F, Newell EW. High-dimensional analysis of the murine myeloid cell system. Nat Immunol. 2014;15:1181–9. doi: 10.1038/ni.3006. [DOI] [PubMed] [Google Scholar]
  • 12.Thomas GD, Hamers AAJ, Nakao C, Marcovecchio P, Taylor AM, McSkimming C, Nguyen AT, McNamara CA, Hedrick CC. Human Blood Monocyte Subsets: A New Gating Strategy Defined Using Cell Surface Markers Identified by Mass Cytometry. Arterioscler Thromb Vasc Biol. 2017 doi: 10.1161/ATVBAHA.117.309145. ATVBAHA.117.309145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Kohyama M, Ise W, Edelson BT, Wilker PR, Hildner K, Mejia C, Frazier WA, Murphy TL, Murphy KM. Role for Spi-C in the development of red pulp macrophages and splenic iron homeostasis. Nature. 2009;457:318–321. doi: 10.1038/nature07472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Haldar M, Kohyama M, So AYL, Kc W, Wu X, Briseño CG, Satpathy AT, Kretzer NM, Arase H, Rajasekaran NS, Wang L, Egawa T, Igarashi K, Baltimore D, Murphy TL, Murphy KM. Heme-mediated SPI-C induction promotes monocyte differentiation into iron-recycling macrophages. Cell. 2014;156:1323–1334. doi: 10.1016/j.cell.2014.01.069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Lavin Y, Winter D, Blecher-Gonen R, David E, Keren-Shaul H, Merad M, Jung S, Amit I. Tissue-resident macrophage enhancer landscapes are shaped by the local microenvironment. Cell. 2014;159:1312–1326. doi: 10.1016/j.cell.2014.11.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Heidt T, Courties G, Dutta P, Sager HB, Sebas M, Iwamoto Y, Sun Y, Da Silva N, Panizzi P, van der Laan AM, van der Lahn AM, Swirski FK, Weissleder R, Nahrendorf M. Differential contribution of monocytes to heart macrophages in steady-state and after myocardial infarction. Circ Res. 2014;115:284–95. doi: 10.1161/CIRCRESAHA.115.303567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Hashimoto D, Chow A, Noizat C, Teo P, Beasley MB, Leboeuf M, Becker CD, See P, Price J, Lucas D, Greter M, Mortha A, Boyer SW, Forsberg EC, Tanaka M, van Rooijen N, García-Sastre A, Stanley ER, Ginhoux F, Frenette PS, Merad M. Tissue-resident macrophages self-maintain locally throughout adult life with minimal contribution from circulating monocytes. Immunity. 2013;38:792–804. doi: 10.1016/j.immuni.2013.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Scott CL, Zheng F, De Baetselier P, Martens L, Saeys Y, De Prijck S, Lippens S, Abels C, Schoonooghe S, Raes G, Devoogdt N, Lambrecht BN, Beschin A, Guilliams M. Bone marrow-derived monocytes give rise to self-renewing and fully differentiated Kupffer cells. Nat Commun. 2016;7:10321. doi: 10.1038/ncomms10321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Fuster JJ, Ouchi N, Gokce N, Walsh K. Obesity-induced changes in adipose tissue microenvironment and their impact on cardiovascular disease. Circ Res. 2016;118:1786–1807. doi: 10.1161/CIRCRESAHA.115.306885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Jaiswal S, Natarajan P, Silver AJ, Gibson CJ, Bick AG, Shvartz E, McConkey M, Gupta N, Gabriel S, Ardissino D, Baber U, Mehran R, Fuster V, Danesh J, Frossard P, Saleheen D, Melander O, Sukhova GK, Neuberg D, Libby P, Kathiresan S, Ebert BL. Clonal Hematopoiesis and Risk of Atherosclerotic Cardiovascular Disease. N Engl J Med. 2017;377:111–121. doi: 10.1056/NEJMoa1701719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Pang WW, Schrier SL, Weissman IL. Age-associated changes in human hematopoietic stem cells. Semin Hematol. 2017;54:39–42. doi: 10.1053/j.seminhematol.2016.10.004. [DOI] [PubMed] [Google Scholar]
  • 22.Pinto AR, Ilinykh A, Ivey MJ, Kuwabara JT, D’antoni ML, Debuque R, Chandran A, Wang L, Arora K, Rosenthal NA, Tallquist MD. Revisiting cardiac cellular composition. Circ Res. 2016;118:400–409. doi: 10.1161/CIRCRESAHA.115.307778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Epelman S, Lavine KJ, Beaudin AE, Sojka DK, Carrero JA, Calderon B, Brija T, Gautier EL, Ivanov S, Satpathy AT, Schilling JD, Schwendener R, Sergin I, Razani B, Forsberg EC, Yokoyama WM, Unanue ER, Colonna M, Randolph GJ, Mann DL. Embryonic and adult-derived resident cardiac macrophages are maintained through distinct mechanisms at steady state and during inflammation. Immunity. 2014;40:91–104. doi: 10.1016/j.immuni.2013.11.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Leid J, Carrelha J, Boukarabila H, Epelman S, Jacobsen SEW, Lavine KJ. Primitive Embryonic Macrophages are Required for Coronary Development and Maturation. Circ Res. 2016;118:1498–1511. doi: 10.1161/CIRCRESAHA.115.308270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Pinto AR, Paolicelli R, Salimova E, Gospocic J, Slonimsky E, Bilbao-Cortes D, Godwin JW, Rosenthal NA. An abundant tissue macrophage population in the adult murine heart with a distinct alternatively-activated macrophage profile. PLoS One. 2012;7:e36814. doi: 10.1371/journal.pone.0036814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Hulsmans M, Clauss S, Xiao L, Aguirre AD, King KR, Hanley A, Hucker WJ, Wülfers EM, Seemann G, Courties G, Iwamoto Y, Sun Y, Savol AJ, Sager HB, Lavine KJ, Fishbein GA, Capen DE, Da Silva N, Miquerol L, Wakimoto H, Seidman CE, Seidman JG, Sadreyev RI, Naxerova K, Mitchell RN, Brown D, Libby P, Weissleder R, Swirski FK, Kohl P, Vinegoni C, Milan DJ, Ellinor PT, Nahrendorf M. Macrophages Facilitate Electrical Conduction in the Heart. Cell. 2017;169:510–522.e20. doi: 10.1016/j.cell.2017.03.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Woehlck HJ, McCann FV. Action potentials in human macrophages are calcium spikes. Cell Biol Int Rep. 1986;10:517–25. doi: 10.1016/0309-1651(86)90026-3. [DOI] [PubMed] [Google Scholar]
  • 28.McCann FV, Cole JJ, Guyre PM, Russell JA. Action potentials in macrophages derived from human monocytes. Science. 1983;219:991–3. doi: 10.1126/science.6823563. [DOI] [PubMed] [Google Scholar]
  • 29.Gallin EK, Livengood DR. Nonlinear current-voltage relationships in cultured macrophages. J Cell Biol. 1980;85:160–5. doi: 10.1083/jcb.85.1.160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Ensan S, Li A, Besla R, Degousee N, Cosme J, Roufaiel M, Shikatani EA, El-Maklizi M, Williams JW, Robins L, Li C, Lewis B, Yun TJ, Lee JS, Wieghofer P, Khattar R, Farrokhi K, Byrne J, Ouzounian M, Zavitz CCJ, Levy GA, Bauer CMT, Libby P, Husain M, Swirski FK, Cheong C, Prinz M, Hilgendorf I, Randolph GJ, Epelman S, Gramolini AO, Cybulsky MI, Rubin BB, Robbins CS. Self-renewing resident arterial macrophages arise from embryonic CX3CR1(+) precursors and circulating monocytes immediately after birth. Nat Immunol. 2016;17:159–68. doi: 10.1038/ni.3343. [DOI] [PubMed] [Google Scholar]
  • 31.Nahrendorf M, Swirski FK, Aikawa E, Stangenberg L, Wurdinger T, Figueiredo J-L, Libby P, Weissleder R, Pittet MJ. The healing myocardium sequentially mobilizes two monocyte subsets with divergent and complementary functions. J Exp Med. 2007;204:3037–3047. doi: 10.1084/jem.20070885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Li W, Hsiao H-M, Higashikubo R, Saunders BT, Bharat A, Goldstein DR, Krupnick AS, Gelman AE, Lavine KJ, Kreisel D. Heart-resident CCR2+ macrophages promote neutrophil extravasation through TLR9/MyD88/CXCL5 signaling. JCI Insight. 2016;1:1–14. doi: 10.1172/jci.insight.87315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Dewald O, Zymek P, Winkelmann K, Koerting A, Ren G, Abou-Khamis T, Michael LH, Rollins BJ, Entman ML, Frangogiannis NG. CCL2/monocyte chemoattractant protein-1 regulates inflammatory responses critical to healing myocardial infarcts. Circ Res. 2005;96:881–889. doi: 10.1161/01.RES.0000163017.13772.3a. [DOI] [PubMed] [Google Scholar]
  • 34.Zouggari Y, Ait-Oufella H, Bonnin P, Simon T, Sage AP, Guérin C, Vilar J, Caligiuri G, Tsiantoulas D, Laurans L, Dumeau E, Kotti S, Bruneval P, Charo IF, Binder CJ, Danchin N, Tedgui A, Tedder TF, Silvestre J-S, Mallat Z. B lymphocytes trigger monocyte mobilization and impair heart function after acute myocardial infarction. Nat Med. 2013;19:1273–80. doi: 10.1038/nm.3284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Jung K, Kim P, Leuschner F, Gorbatov R, Kim JK, Ueno T, Nahrendorf M, Yun SH. Endoscopic time-lapse imaging of immune cells in infarcted mouse hearts. Circ Res. 2013;112:891–899. doi: 10.1161/CIRCRESAHA.111.300484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Hilgendorf I, Gerhardt LMS, Tan TC, Winter C, Holderried TAW, Chousterman BG, Iwamoto Y, Liao R, Zirlik A, Scherer-Crosbie M, Hedrick CC, Libby P, Nahrendorf M, Weissleder R, Swirski FK. 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]
  • 37.Courties G, Heidt T, Sebas M, Iwamoto Y, Jeon D, Truelove J, Tricot B, Wojtkiewicz G, Dutta P, Sager HB, Borodovsky A, Novobrantseva T, Klebanov B, Fitzgerald K, Anderson DG, Libby P, Swirski FK, Weissleder R, Nahrendorf M. In vivo silencing of the transcription factor IRF5 reprograms the macrophage phenotype and improves infarct healing. J Am Coll Cardiol. 2014;63:1556–1566. doi: 10.1016/j.jacc.2013.11.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Leuschner F, Rauch PJ, Ueno T, Gorbatov R, Marinelli B, Lee WW, Dutta P, Wei Y, Robbins C, Iwamoto Y, Sena B, Chudnovskiy A, Panizzi P, Keliher E, Higgins JM, Libby P, Moskowitz MA, Pittet MJ, Swirski FK, Weissleder R, Nahrendorf M. Rapid monocyte kinetics in acute myocardial infarction are sustained by extramedullary monocytopoiesis. J Exp Med. 2012;209:123–37. doi: 10.1084/jem.20111009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Dutta P, Courties G, Wei Y, Leuschner F, Gorbatov R, Robbins CS, Iwamoto Y, Thompson B, Carlson AL, Heidt T, Majmudar MD, Lasitschka F, Etzrodt M, Waterman P, Waring MT, Chicoine AT, van der Laan AM, Niessen HWM, Piek JJ, Rubin BB, Butany J, Stone JR, Katus HA, Murphy SA, Morrow DA, Sabatine MS, Vinegoni C, Moskowitz MA, Pittet MJ, Libby P, Lin CP, Swirski FK, Weissleder R, Nahrendorf M. Myocardial infarction accelerates atherosclerosis. Nature. 2012;487:325–9. doi: 10.1038/nature11260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Swirski FK, Nahrendorf M, Etzrodt M, Wildgruber M, Cortez-Retamozo V, Panizzi P, Figueiredo J-L, Kohler RH, Chudnovskiy A, Waterman P, Aikawa E, Mempel TR, Libby P, Weissleder R, Pittet MJ. Identification of splenic reservoir monocytes and their deployment to inflammatory sites. Science. 2009;325:612–6. doi: 10.1126/science.1175202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Sager HB, Hulsmans M, Lavine KJ, Moreira MB, Heidt T, Courties G, Sun Y, Iwamoto Y, Tricot B, Khan OF, Dahlman JE, Borodovsky A, Fitzgerald K, Anderson DG, Weissleder R, Libby P, Swirski FK, Nahrendorf M. Proliferation and Recruitment Contribute to Myocardial Macrophage Expansion in Chronic Heart Failure. Circ Res. 2016;119:853–864. doi: 10.1161/CIRCRESAHA.116.309001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Dunlay SM, Weston SA, Redfield MM, Killian JM, Roger VL. Tumor necrosis factor-alpha and mortality in heart failure: a community study. Circulation. 2008;118:625–31. doi: 10.1161/CIRCULATIONAHA.107.759191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Papathanasiou S, Rickelt S, Soriano ME, Schips TG, Maier HJ, Davos CH, Varela A, Kaklamanis L, Mann DL, Capetanaki Y. Tumor necrosis factor-α confers cardioprotection through ectopic expression of keratins K8 and K18. Nat Med. 2015;21:1076–1084. doi: 10.1038/nm.3925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Frangogiannis NG. The inflammatory response in myocardial injury, repair, and remodelling. Nat Rev Cardiol. 2014;11:255–265. doi: 10.1038/nrcardio.2014.28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.van Amerongen MJ, Harmsen MC, van Rooijen N, Petersen AH, van Luyn MJA. Macrophage Depletion Impairs Wound Healing and Increases Left Ventricular Remodeling after Myocardial Injury in Mice. Am J Pathol. 2007;170:818–829. doi: 10.2353/ajpath.2007.060547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Leblond AL, Klinkert K, Martin K, Turner EC, Kumar AH, Browne T, Caplice NM. Systemic and cardiac depletion of M2 macrophage through CSF-1R signaling inhibition alters cardiac function post myocardial infarction. PLoS One. 2015;10:1–13. doi: 10.1371/journal.pone.0137515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Panizzi P, Swirski FK, Figueiredo J-L, Waterman P, Sosnovik DE, Aikawa E, Libby P, Pittet M, Weissleder R, Nahrendorf M. Impaired infarct healing in atherosclerotic mice with Ly-6C(hi) monocytosis. J Am Coll Cardiol. 2010;55:1629–38. doi: 10.1016/j.jacc.2009.08.089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Cochain C, Auvynet C, Poupel L, Vilar J, Dumeau E, Richart A, Recalde A, Zouggari Y, Yin KYHW, Bruneval P, Renault G, Marchiol C, Bonnin P, Levy B, Bonecchi R, Locati M, Combadiere C, Silvestre J-S. The Chemokine Decoy Receptor D6 Prevents Excessive Inflammation and Adverse Ventricular Remodeling After Myocardial Infarction. Arterioscler Thromb Vasc Biol. 2012;32:2206–2213. doi: 10.1161/ATVBAHA.112.254409. [DOI] [PubMed] [Google Scholar]
  • 49.Leuschner F, Dutta P, Gorbatov R, Novobrantseva TI, Donahoe JS, Courties G, Lee KM, Kim JI, Markmann JF, Marinelli B, Panizzi P, Lee WW, Iwamoto Y, Milstein S, Epstein-Barash H, Cantley W, Wong J, Cortez-Retamozo V, Newton A, Love K, Libby P, Pittet MJ, Swirski FK, Koteliansky V, Langer R, Weissleder R, Anderson DG, Nahrendorf M. Therapeutic siRNA silencing in inflammatory monocytes in mice. Nat Biotechnol. 2011;29:1005–1010. doi: 10.1038/nbt.1989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Yancy CW, Jessup M, Bozkurt B, Butler J, Casey DE, Drazner MH, Fonarow GC, Geraci SA, Horwich T, Januzzi JL, Johnson MR, Kasper EK, Levy WC, Masoudi FA, McBride PE, McMurray JJV, Mitchell JE, Peterson PN, Riegel B, Sam F, Stevenson LW, Tang WHW, Tsai EJ, Wilkoff BL. 2013 ACCF/AHA guideline for the management of heart failure: Executive summary: A report of the American college of cardiology foundation/American Heart Association task force on practice guidelines. Circulation. 2013;128:1810–1852. doi: 10.1161/CIR.0b013e31829e8807. [DOI] [PubMed] [Google Scholar]
  • 51.McMurray JJV, Adamopoulos S, Anker SD, Auricchio A, Böhm M, Dickstein K, Falk V, Filippatos G, Fonseca C, Gomez-Sanchez MA, Jaarsma T, Køber L, Lip GYH, Pietro Maggioni A, Parkhomenko A, Pieske BM, Popescu BA, Rønnevik PK, Rutten FH, Schwitter J, Seferovic P, Stepinska J, Trindade PT, Voors AA, Zannad F, Zeiher A, ESC Committee for Practice Guidelines ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure 2012: The Task Force for the Diagnosis and Treatment of Acute and Chronic Heart Failure 2012 of the European Society of Cardiology. Developed in collaboration with the Heart. Eur Heart J. 2012;33:1787–847. doi: 10.1093/eurheartj/ehs104. [DOI] [PubMed] [Google Scholar]
  • 52.Ismahil MA, Hamid T, Bansal SS, Patel B, Kingery JR, Prabhu SD. Remodeling of the mononuclear phagocyte network underlies chronic inflammation and disease progression in heart failure critical importance of the cardiosplenic axis. Circ Res. 2014;114:266–282. doi: 10.1161/CIRCRESAHA.113.301720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Leask A. Getting to the heart of the matter: new insights into cardiac fibrosis. Circ Res. 2015;116:1269–76. doi: 10.1161/CIRCRESAHA.116.305381. [DOI] [PubMed] [Google Scholar]
  • 54.Laflamme MA, Murry CE. Heart regeneration. Nature. 2011;473:326–335. doi: 10.1038/nature10147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Poss KD, Wilson LG, Keating MT. Heart regeneration in zebrafish. Science. 2002;298:2188–90. doi: 10.1126/science.1077857. [DOI] [PubMed] [Google Scholar]
  • 56.Kikuchi K, Holdway JE, Werdich AA, Anderson RM, Fang Y, Egnaczyk GF, Evans T, MacRae CA, Stainier DYR, Poss KD. Primary contribution to zebrafish heart regeneration by gata4+ cardiomyocytes. Nature. 2010;464:601–605. doi: 10.1038/nature08804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Jopling C, Sleep E, Raya M, Martí M, Raya A, Belmonte JCI. Zebrafish heart regeneration occurs by cardiomyocyte dedifferentiation and proliferation. Nature. 2010;464:606–609. doi: 10.1038/nature08899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Porrello ER, Mahmoud AI, Simpson E, Hill JA, Richardson JA, Olson EN, Sadek HA. Transient regenerative potential of the neonatal mouse heart. Science. 2011;331:1078–80. doi: 10.1126/science.1200708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Haubner BJ, Adamowicz-Brice M, Khadayate S, Tiefenthaler V, Metzler B, Aitman T, Penninger JM. Complete cardiac regeneration in a mouse model of myocardial infarction. Aging (Albany NY) 2012;4:966–977. doi: 10.18632/aging.100526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Jesty SA, Steffey MA, Lee FK, Breitbach M, Hesse M, Reining S, Lee JC, Doran RM, Nikitin AY, Fleischmann BK, Kotlikoff MI. c-kit+ precursors support postinfarction myogenesis in the neonatal, but not adult, heart. Proc Natl Acad Sci U S A. 2012;109:13380–5. doi: 10.1073/pnas.1208114109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Aurora AB, Porrello ER, Tan W, Mahmoud AI, Hill JA, Bassel-Duby R, Sadek HA, Olson EN. Macrophages are required for neonatal heart regeneration. J Clin Invest. 2014;124:1382–92. doi: 10.1172/JCI72181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Lavine KJ, Epelman S, Uchida K, Weber KJ, Nichols CG, Schilling JD, Ornitz DM, Randolph GJ, Mann DL. Distinct macrophage lineages contribute to disparate patterns of cardiac recovery and remodeling in the neonatal and adult heart. Proc Natl Acad Sci. 2014;111:16029–16034. doi: 10.1073/pnas.1406508111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Mahmoud AI, O’Meara CC, Gemberling M, Zhao L, Bryant DM, Zheng R, Gannon JB, Cai L, Choi WY, Egnaczyk GF, Burns CE, Burns CG, MacRae CA, Poss KD, Lee RT. Nerves Regulate Cardiomyocyte Proliferation and Heart Regeneration. Dev Cell. 2015;34:387–399. doi: 10.1016/j.devcel.2015.06.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.White IA, Gordon J, Balkan W, Hare JM. Sympathetic reinnervation is required for mammalian cardiac regeneration. Circ Res. 2015;117:990–994. doi: 10.1161/CIRCRESAHA.115.307465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Uygur A, Lee RT. Mechanisms of Cardiac Regeneration. Dev Cell. 2016;36:362–374. doi: 10.1016/j.devcel.2016.01.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Shirai T, Hilhorst M, Harrison DG, Goronzy JJ, Weyand CM. Macrophages in vascular inflammation – From atherosclerosis to vasculitis. Autoimmunity. 2015;48:139–151. doi: 10.3109/08916934.2015.1027815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Bush E, Maeda N, Kuziel WA, Dawson TC, Wilcox JN, DeLeon H, Taylor WR. CC chemokine receptor 2 is required for macrophage infiltration and vascular hypertrophy in angiotensin II-induced hypertension. Hypertens (Dallas, Tex 1979) 2000;36:360–3. doi: 10.1161/01.hyp.36.3.360. [DOI] [PubMed] [Google Scholar]
  • 68.Wenzel P, Knorr M, Kossmann S, Stratmann J, Hausding M, Schuhmacher S, Karbach SH, Schwenk M, Yogev N, Schulz E, Oelze M, Grabbe S, Jonuleit H, Becker C, Daiber A, Waisman A, Münzel T. Lysozyme M-positive monocytes mediate angiotensin ii-induced arterial hypertension and vascular dysfunction. Circulation. 2011;124:1370–1381. doi: 10.1161/CIRCULATIONAHA.111.034470. [DOI] [PubMed] [Google Scholar]
  • 69.Moore JP, Vinh A, Tuck KL, Sakkal S, Krishnan SM, Chan CT, Lieu M, Samuel CS, Diep H, Kemp-Harper BK, Tare M, Ricardo SD, Guzik TJ, Sobey CG, Drummond GR. M2 macrophage accumulation in the aortic wall during angiotensin II infusion in mice is associated with fibrosis, elastin loss, and elevated blood pressure. Am J Physiol Heart Circ Physiol. 2015;309:H906–17. doi: 10.1152/ajpheart.00821.2014. [DOI] [PubMed] [Google Scholar]
  • 70.Shah KH, Shi P, Giani JF, Janjulia T, Bernstein EA, Li Y, Zhao T, Harrison DG, Bernstein KE, Shen XZ. Myeloid Suppressor Cells Accumulate and Regulate Blood Pressure in Hypertension. Circ Res. 2015;117:858–869. doi: 10.1161/CIRCRESAHA.115.306539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Moore KJ, Tabas I. Macrophages in the pathogenesis of atherosclerosis. Cell. 2011;145:341–355. doi: 10.1016/j.cell.2011.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Hansson GK, Hermansson A. The immune system in atherosclerosis. Nat Immunol. 2011;12:204–212. doi: 10.1038/ni.2001. [DOI] [PubMed] [Google Scholar]
  • 73.Tall AR, Yvan-Charvet L. Cholesterol, inflammation and innate immunity. Nat Rev Immunol. 2015;15:104–16. doi: 10.1038/nri3793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Moore KJ, Sheedy FJ, Fisher EA. Macrophages in atherosclerosis: a dynamic balance. Nat Rev Immunol. 2013;13:709–721. doi: 10.1038/nri3520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Virmani R, Kolodgie FD, Burke AP, Farb A, Schwartz SM. Lessons from sudden coronary death: a comprehensive morphological classification scheme for atherosclerotic lesions. Arterioscler Thromb Vasc Biol. 2000;20:1262–75. doi: 10.1161/01.atv.20.5.1262. [DOI] [PubMed] [Google Scholar]
  • 76.Lafont A. Basic aspects of plaque vulnerability. Heart. 2003;89:1262–1267. doi: 10.1136/heart.89.10.1262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Franck G, Mawson T, Sausen G, Salinas M, Masson GS, Cole A, Beltrami-Moreira M, Chatzizisis Y, Quillard T, Tesmenitsky Y, Shvartz E, Sukhova GK, Swirski FK, Nahrendorf M, Aikawa E, Croce KJ, Libby P. Flow Perturbation Mediates Neutrophil Recruitment and Potentiates Endothelial Injury via TLR2 in Mice: Implications for Superficial Erosion. Circ Res. 2017;121:31–42. doi: 10.1161/CIRCRESAHA.117.310694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Robbins CS, Hilgendorf I, Weber GF, Theurl I, Iwamoto Y, Figueiredo J-L, Gorbatov R, Sukhova GK, Gerhardt LMS, Smyth D, Zavitz CCJ, Shikatani EA, Parsons M, van Rooijen N, Lin HY, Husain M, Libby P, Nahrendorf M, Weissleder R, Swirski FK. Local proliferation dominates lesional macrophage accumulation in atherosclerosis. Nat Med. 2013;19:1166–1172. doi: 10.1038/nm.3258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Combadière C, Potteaux S, Rodero M, Simon T, Pezard A, Esposito B, Merval R, Proudfoot A, Tedgui A, Mallat Z. Combined inhibition of CCL2, CX3CR1, and CCR5 abrogates Ly6Chi and Ly6Clo monocytosis and almost abolishes atherosclerosis in hypercholesterolemic mice. Circulation. 2008;117:1649–1657. doi: 10.1161/CIRCULATIONAHA.107.745091. [DOI] [PubMed] [Google Scholar]
  • 80.Shankman LS, Gomez D, Cherepanova OA, Salmon M, Alencar GF, Haskins RM, Swiatlowska P, Newman AAC, Greene ES, Straub AC, Isakson B, Randolph GJ, Owens GK. KLF4-dependent phenotypic modulation of smooth muscle cells has a key role in atherosclerotic plaque pathogenesis. Nat Med. 2015;21:628–637. doi: 10.1038/nm.3866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Chappell J, Harman JL, Narasimhan VM, Yu H, Foote K, Simons BD, Bennett MR, Jørgensen HF. Extensive Proliferation of a Subset of Differentiated, yet Plastic, Medial Vascular Smooth Muscle Cells Contributes to Neointimal Formation in Mouse Injury and Atherosclerosis Models. Circ Res. 2016;119:1313–1323. doi: 10.1161/CIRCRESAHA.116.309799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Sandison ME, Dempster J, McCarron JG. The transition of smooth muscle cells from a contractile to a migratory, phagocytic phenotype: direct demonstration of phenotypic modulation. J Physiol. 2016;594:6189–6209. doi: 10.1113/JP272729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Robbins CS, Hilgendorf I, Weber GF, Theurl I, Iwamoto Y, Figueiredo J, Gorbatov R, Sukhova GK, Gerhardt LMS, Smyth D, Zavitz CCJ, Shikatani EA, Parsons M, van Rooijen N, Lin HY, Husain M, Libby P, Nahrendorf M, Weissleder R, Swirski FK. Local proliferation dominates lesional macrophage accumulation in atherosclerosis. Nat Med. 2013;19:1166–72. doi: 10.1038/nm.3258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Shah PK, Yano J, Reyes O, Chyu KY, Kaul S, Bisgaier CL, Drake S, Cercek B. High-dose recombinant apolipoprotein A-I(milano) mobilizes tissue cholesterol and rapidly reduces plaque lipid and macrophage content in apolipoprotein e-deficient mice. Potential implications for acute plaque stabilization. Circulation. 2001;103:3047–3050. doi: 10.1161/hc2501.092494. [DOI] [PubMed] [Google Scholar]
  • 85.Tian F, Wang L, Arias A, Yang M, Sharifi BG, Shah PK. Comparative antiatherogenic effects of intravenous AAV8- and AAV2-mediated ApoA-IMilano gene transfer in hypercholesterolemic mice. J Cardiovasc Pharmacol Ther. 2015;20:66–75. doi: 10.1177/1074248414530041. [DOI] [PubMed] [Google Scholar]
  • 86.Wang L, Tian F, Arias A, Yang M, Sharifi BG, Shah PK. Comparative Effects of Diet-Induced Lipid Lowering Versus Lipid Lowering Along With Apo A-I Milano Gene Therapy on Regression of Atherosclerosis. J Cardiovasc Pharmacol Ther. 2016;21:320–8. doi: 10.1177/1074248415610216. [DOI] [PubMed] [Google Scholar]
  • 87.Lieu HD, Withycombe SK, Walker Q, Rong JX, Walzem RL, Wong JS, Hamilton RL, Fisher EA, Young SG. Eliminating atherogenesis in mice by switching off hepatic lipoprotein secretion. Circulation. 2003;107:1315–1321. doi: 10.1161/01.cir.0000054781.50889.0c. [DOI] [PubMed] [Google Scholar]
  • 88.Feig JE, Parathath S, Rong JX, Mick SL, Vengrenyuk Y, Grauer L, Young SG, Fisher EA. Reversal of hyperlipidemia with a genetic switch favorably affects the content and inflammatory state of macrophages in atherosclerotic plaques. Circulation. 2011;123:989–998. doi: 10.1161/CIRCULATIONAHA.110.984146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Llodrá J, Angeli V, Liu J, Trogan E, Fisher EA, Randolph GJ. Emigration of monocyte-derived cells from atherosclerotic lesions characterizes regressive, but not progressive, plaques. Proc Natl Acad Sci U S A. 2004;101:11779–84. doi: 10.1073/pnas.0403259101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Feig JE, Rong JX, Shamir R, Sanson M, Vengrenyuk Y, Liu J, Rayner K, Moore K, Garabedian M, Fisher EA. HDL promotes rapid atherosclerosis regression in mice and alters inflammatory properties of plaque monocyte-derived cells. Proc Natl Acad Sci. 2011;108:7166–7171. doi: 10.1073/pnas.1016086108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Rahman K, Vengrenyuk Y, Ramsey SA, Vila NR, Girgis NM, Liu J, Gusarova V, Gromada J, Weinstock A, Moore KJ, Loke P, Fisher EA. Inflammatory Ly6Chi monocytes and their conversion to M2 macrophages drive atherosclerosis regression. J Clin Invest. 2017:1–12. doi: 10.1172/JCI75005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Ridker PM, MacFadyen JG, Thuren T, Everett BM, Libby P, Glynn RJ, Ridker P, Lorenzatti A, Krum H, Varigos J, Siostrzonek P, Sinnaeve P, Fonseca F, Nicolau J, Gotcheva N, Genest J, Yong H, Urina-Triana M, Milicic D, Cifkova R, Vettus R, Koenig W, Anker SD, Manolis AJ, Wyss F, Forster T, Sigurdsson A, Pais P, Fucili A, Ogawa H, Shimokawa H, Veze I, Petrauskiene B, Salvador L, Kastelein J, Cornel JH, Klemsdal TO, Medina F, Budaj A, Vida-Simiti L, Kobalava Z, Otasevic P, Pella D, Lainscak M, Seung K-B, Commerford P, Dellborg M, Donath M, Hwang J-J, Kultursay H, Flather M, Ballantyne C, Bilazarian S, Chang W, East C, Everett B, Forgosh L, Glynn R, Harris B, Libby P, Ligueros M, Thuren T, Bohula E, Charmarthi B, Cheng S, Chou S, Danik J, McMahon G, Maron B, Ning M, Olenchock B, Pande R, Perlstein T, Pradhan A, Rost N, Singhal A, Taqueti V, Wei N, Burris H, Cioffi A, Dalseg AM, Ghosh N, Gralow J, Mayer T, Rugo H, Fowler V, Limaye AP, Cosgrove S, Levine D, Lopes R, Scott J, Thuren T, Ligueros M, Hilkert R, Tamesby G, Mickel C, Manning B, Woelcke J, et al. Effect of interleukin-1β inhibition with canakinumab on incident lung cancer in patients with atherosclerosis: exploratory results from a randomised, double-blind, placebo-controlled trial. Lancet. 2017;6736:1–10. doi: 10.1016/S0140-6736(17)32247-X. [DOI] [PubMed] [Google Scholar]
  • 93.Ridker PM, Everett BM, Thuren T, MacFadyen JG, Chang WH, Ballantyne C, Fonseca F, Nicolau J, Koenig W, Anker SD, Kastelein JJP, Cornel JH, Pais P, Pella D, Genest J, Cifkova R, Lorenzatti A, Forster T, Kobalava Z, Vida-Simiti L, Flather M, Shimokawa H, Ogawa H, Dellborg M, Rossi PRF, Troquay RPT, Libby P, Glynn RJ. Antiinflammatory Therapy with Canakinumab for Atherosclerotic Disease. N Engl J Med. 2017 doi: 10.1056/NEJMoa1707914. NEJMoa1707914. [DOI] [PubMed] [Google Scholar]
  • 94.Ehninger A, Trumpp A. The bone marrow stem cell niche grows up: mesenchymal stem cells and macrophages move in. J Exp Med. 2011;208:421–428. doi: 10.1084/jem.20110132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Heideveld E, van den Akker E. Digesting the role of bone marrow macrophages on hematopoiesis. Immunobiology. 2017;222:814–822. doi: 10.1016/j.imbio.2016.11.007. [DOI] [PubMed] [Google Scholar]
  • 96.Winkler IG, Sims NA, Pettit AR, Barbier V, Nowlan B, Helwani F, Ingrid J. Bone marrow macrophages maintain hematopoietic stem cell (HSC) niches and their depletion mobilizes HSC Bone marrow macrophages maintain hematopoietic stem cell (HSC) niches and their depletion mobilizes HSC. Blood. 2010;116:4815–4829. doi: 10.1182/blood-2009-11-253534. [DOI] [PubMed] [Google Scholar]
  • 97.Chow A, Lucas D, Hidalgo A, Méndez-Ferrer S, Hashimoto D, Scheiermann C, Battista M, Leboeuf M, Prophete C, van Rooijen N, Tanaka M, Merad M, Frenette PS. Bone marrow CD169+ macrophages promote the retention of hematopoietic stem and progenitor cells in the mesenchymal stem cell niche. J Exp Med. 2011;208:261–271. doi: 10.1084/jem.20101688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Dutta P, Hoyer FF, Grigoryeva LS, Sager HB, Leuschner F, Courties G, Borodovsky A, Novobrantseva T, Ruda VM, Fitzgerald K, Iwamoto Y, Wojtkiewicz G, Sun Y, Da Silva N, Libby P, Anderson DG, Swirski FK, Weissleder R, Nahrendorf M. Macrophages retain hematopoietic stem cells in the spleen via VCAM-1. J Exp Med. 2015;212:497–512. doi: 10.1084/jem.20141642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.McCabe A, Zhang Y, Thai V, Jones M, Jordan MB, MacNamara KC. Macrophage-lineage cells negatively regulate the hematopoietic stem cell pool in response to interferon gamma at steady state and during infection. Stem Cells. 2015;33:2294–2305. doi: 10.1002/stem.2040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Basu S, Hodgson G, Katz M, Dunn AR. Evaluation of role of G-CSF in the production, survival, and release of neutrophils from bone marrow into circulation. Blood. 2002;100:854–861. doi: 10.1182/blood.v100.3.854. [DOI] [PubMed] [Google Scholar]
  • 101.Stark MA, Huo Y, Burcin TL, Morris MA, Olson TS, Ley K. Phagocytosis of apoptotic neutrophils regulates granulopoiesis via IL-23 and IL-17. Immunity. 2005;22:285–294. doi: 10.1016/j.immuni.2005.01.011. [DOI] [PubMed] [Google Scholar]
  • 102.Casanova-Acebes M, Pitaval C, Weiss LA, Nombela-Arrieta C, Chèvre R, A-González N, Kunisaki Y, Zhang D, van Rooijen N, Silberstein LE, Weber C, Nagasawa T, Frenette PS, Castrillo A, Hidalgo A. Rhythmic modulation of the hematopoietic niche through neutrophil clearance. Cell. 2013;153:1025–35. doi: 10.1016/j.cell.2013.04.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Dutta P, Sager HB, Stengel KR, Naxerova K, Courties G, Saez B, Silberstein L, Heidt T, Sebas M, Sun Y, Wojtkiewicz G, Feruglio PF, King K, Baker JN, van der Laan AM, Borodovsky A, Fitzgerald K, Hulsmans M, Hoyer F, Iwamoto Y, Vinegoni C, Brown D, Di Carli M, Libby P, Hiebert SW, Scadden DT, Swirski FK, Weissleder R, Nahrendorf M. Myocardial Infarction Activates CCR2(+) Hematopoietic Stem and Progenitor Cells. Cell Stem Cell. 2015;16:477–87. doi: 10.1016/j.stem.2015.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Sørensen MV, Pedersen S, Møgelvang R, Skov-Jensen J, Flyvbjerg A. Plasma high-mobility group box 1 levels predict mortality after ST-segment elevation myocardial infarction. JACC Cardiovasc Interv. 2011;4:281–6. doi: 10.1016/j.jcin.2010.10.015. [DOI] [PubMed] [Google Scholar]
  • 105.Andrassy M, Volz HC, Riedle N, Gitsioudis G, Seidel C, Laohachewin D, Zankl AR, Kaya Z, Bierhaus A, Giannitsis E, Katus HA, Korosoglou G. HMGB1 as a predictor of infarct transmurality and functional recovery in patients with myocardial infarction. J Intern Med. 2011;270:245–53. doi: 10.1111/j.1365-2796.2011.02369.x. [DOI] [PubMed] [Google Scholar]
  • 106.Lotze MT, Tracey KJ. High-mobility group box 1 protein (HMGB1): nuclear weapon in the immune arsenal. Nat Rev Immunol. 2005;5:331–342. doi: 10.1038/nri1594. [DOI] [PubMed] [Google Scholar]
  • 107.Morrow DA, Wang Y, Croce K, Sakuma M, Sabatine MS, Gao H, Pradhan AD, Healy AM, Buros J, McCabe CH, Libby P, Cannon CP, Braunwald E, Simon DI. Myeloid-related protein 8/14 and the risk of cardiovascular death or myocardial infarction after an acute coronary syndrome in the Pravastatin or Atorvastatin Evaluation and Infection Theraphy: Thrombolysis in Myocardial Infarction (PROVE IT-TIMI 22) tria. Am Heart J. 2008;155:49–55. doi: 10.1016/j.ahj.2007.08.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Katashima T, Naruko T, Terasaki F, Fujita M, Otsuka K, Murakami S, Sato A, Hiroe M, Ikura Y, Ueda M, Ikemoto M, Kitaura Y. Enhanced expression of the S100A8/A9 complex in acute myocardial infarction patients. CircJ. 2010;74:741–748. doi: 10.1253/circj.cj-09-0564. [DOI] [PubMed] [Google Scholar]
  • 109.Schiopu A, Cotoi OS. S100A8 and S100A9: DAMPs at the Crossroads between Innate Immunity, Traditional Risk Factors, and Cardiovascular Disease. Mediators Inflamm. 2013;2013:1–10. doi: 10.1155/2013/828354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Guillén I, Blanes M, Gómez-Lechón MJ, Castell JV. Cytokine signaling during myocardial infarction: sequential appearance of IL-1 beta and IL-6. Am J Physiol. 1995;269:R229–35. doi: 10.1152/ajpregu.1995.269.2.R229. [DOI] [PubMed] [Google Scholar]
  • 111.Sager HB, Heidt T, Hulsmans M, Dutta P, Courties G, Sebas M, Wojtkiewicz GR, Tricot B, Iwamoto Y, Sun Y, Weissleder R, Libby P, Swirski FK, Nahrendorf M. Targeting Interleukin-1β Reduces Leukocyte Production After Acute Myocardial Infarction. Circulation. 2015;132:1880–90. doi: 10.1161/CIRCULATIONAHA.115.016160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Geijtenbeek TBH, Groot PC, Nolte MA, Van Vliet SJ, Gangaram-Panday ST, Van Duijnhoven GCF, Kraal G, Van Oosterhout AJM, Van Kooyk Y. Marginal zone macrophages express a murine homologue of DC-SIGN that captures blood-borne antigens in vivo. Blood. 2002;100:2908–2916. doi: 10.1182/blood-2002-04-1044. [DOI] [PubMed] [Google Scholar]
  • 113.Karlsson MCI, Guinamard R, Bolland S, Sankala M, Steinman RM, Ravetch JV. Macrophages control the retention and trafficking of B lymphocytes in the splenic marginal zone. J Exp Med. 2003;198:333–340. doi: 10.1084/jem.20030684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Nolte MA, Arens R, Kraus M, van Oers MH, Kraal G, van Lier RA, Mebius RE. B cells are crucial for both development and maintenance of the splenic marginal zone. J Immunol. 2004;172:3620–3627. doi: 10.4049/jimmunol.172.6.3620. [DOI] [PubMed] [Google Scholar]
  • 115.You Y, Myers RC, Freeberg L, Foote J, Kearney JF, Justement LB, Carter RH. Marginal Zone B Cells Regulate Antigen Capture by Marginal Zone Macrophages. J Immunol. 2011;186:2172–2181. doi: 10.4049/jimmunol.1002106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Mebius RE, Kraal G. Structure and function of the spleen. Nat Rev Immunol. 2005;5:606–616. doi: 10.1038/nri1669. [DOI] [PubMed] [Google Scholar]
  • 117.Dutta P, Hoyer FF, Sun Y, Iwamoto Y, Tricot B, Weissleder R, Magnani JL, Swirski FK, Nahrendorf M. E-selectin inhibition mitigates splenic HSC activation and myelopoiesis in hypercholesterolemic mice with myocardial infarction. Arterioscler Thromb Vasc Biol. 2016;36:1802–1808. doi: 10.1161/ATVBAHA.116.307519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Kim E, Yang J, Beltran CD, Cho S. Role of spleen-derived monocytes/macrophages in acute ischemic brain injury. J Cereb Blood Flow Metab. 2014;34:1411–9. doi: 10.1038/jcbfm.2014.101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Robbins CS, Chudnovskiy A, Rauch PJ, Figueiredo JL, Iwamoto Y, Gorbatov R, Etzrodt M, Weber GF, Ueno T, Van Rooijen N, Mulligan-Kehoe MJ, Libby P, Nahrendorf M, Pittet MJ, Weissleder R, Swirski FK. Extramedullary hematopoiesis generates Ly-6C high monocytes that infiltrate atherosclerotic lesions. Circulation. 2012;125:364–374. doi: 10.1161/CIRCULATIONAHA.111.061986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Harms M, Seale P. Brown and beige fat: development, function and therapeutic potential. Nat Med. 2013;19:1252–1263. doi: 10.1038/nm.3361. [DOI] [PubMed] [Google Scholar]
  • 121.Giralt M, Villarroya F. White, brown, beige/brite: Different adipose cells for different functions? Endocrinology. 2013;154:2992–3000. doi: 10.1210/en.2013-1403. [DOI] [PubMed] [Google Scholar]
  • 122.Rosenwald M, Wolfrum C. The origin and definition of brite versus white and classical brown adipocytes. Adipocyte. 2014;3:4–9. doi: 10.4161/adip.26232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Amano SU, Cohen JL, Vangala P, Tencerova M, Nicoloro SM, Yawe JC, Shen Y, Czech MP, Aouadi M. Local proliferation of macrophages contributes to obesity-associated adipose tissue inflammation. Cell Metab. 2014;19:162–171. doi: 10.1016/j.cmet.2013.11.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW. Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest. 2003;112:1796–1808. doi: 10.1172/JCI19246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Kanda H, Tateya S, Tamori Y, Kotani K, Hiasa K, Kitazawa R, Kitazawa S, Miyachi H, Maeda S, Egashira K, Kasuga M. MCP-1 contributes to macrophage infiltration into adipose tissue, insulin resistance, and hepatic steatosis in obesity. J Clin Invest. 2006;116:1494–505. doi: 10.1172/JCI26498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Hassnain Waqas SF, Noble A, Hoang AC, Ampem G, Popp M, Strauß S, Guille M, Röszer T. Adipose tissue macrophages develop from bone marrow-independent progenitors in Xenopus laevis and mouse. J Leukoc Biol. 2017 doi: 10.1189/jlb.1A0317-082RR. Epub. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Brestoff JR, Artis D. Immune regulation of metabolic homeostasis in health and disease. Cell. 2015;161:146–160. doi: 10.1016/j.cell.2015.02.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Odegaard JI, Ricardo-Gonzalez RR, Goforth MH, Morel CR, Subramanian V, Mukundan L, Eagle AR, Vats D, Brombacher F, Ferrante AW, Chawla A. Macrophage-specific PPARγ controls alternative activation and improves insulin resistance. Nature. 2007;447:1116–1120. doi: 10.1038/nature05894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Kang K, Reilly SM, Karabacak V, Gangl MR, Fitzgerald K, Hatano B, Lee C-H. Adipocyte-derived Th2 cytokines and myeloid PPARdelta regulate macrophage polarization and insulin sensitivity. Cell Metab. 2008;7:485–95. doi: 10.1016/j.cmet.2008.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Szanto A, Balint BL, Nagy ZS, Barta E, Dezso B, Pap A, Szeles L, Poliska S, Oros M, Evans RM, Barak Y, Schwabe J, Nagy L. STAT6 transcription factor is a facilitator of the nuclear receptor PPARγ-regulated gene expression in macrophages and dendritic cells. Immunity. 2010;33:699–712. doi: 10.1016/j.immuni.2010.11.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Ricardo-Gonzalez RR, Red Eagle A, Odegaard JI, Jouihan H, Morel CR, Heredia JE, Mukundan L, Wu D, Locksley RM, Chawla A. IL-4/STAT6 immune axis regulates peripheral nutrient metabolism and insulin sensitivity. Proc Natl Acad Sci U S A. 2010;107:22617–22622. doi: 10.1073/pnas.1009152108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Wu D, Molofsky AB, Liang H-E, Ricardo-Gonzalez RR, Jouihan HA, Bando JK, Chawla A, Locksley RM. Eosinophils sustain adipose alternatively activated macrophages associated with glucose homeostasis. Science. 2011;332:243–7. doi: 10.1126/science.1201475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Chang Y-H, Ho K-T, Lu S-H, Huang C-N, Shiau M-Y. Regulation of glucose/lipid metabolism and insulin sensitivity by interleukin-4. Int J Obes. 2012;36:993–998. doi: 10.1038/ijo.2011.168. [DOI] [PubMed] [Google Scholar]
  • 134.Lumeng CN, Bodzin JL, Saltiel AR. Obesity induces a phenotipic switch in adipose tissue macrophage polarization. J Clin Invest. 2007;117:175–184. doi: 10.1172/JCI29881. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.McNelis JC, Olefsky JM. Macrophages, Immunity, and Metabolic Disease. Immunity. 2014;41:36–48. doi: 10.1016/j.immuni.2014.05.010. [DOI] [PubMed] [Google Scholar]
  • 136.Shoelson SE, Lee J, Goldfine AB. Inflammation and insulin resistance. J Clin Invest. 2006;116:1793–801. doi: 10.1172/JCI29069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Nordestgaard BG. Triglyceride-Rich Lipoproteins and Atherosclerotic Cardiovascular Disease: New Insights From Epidemiology, Genetics, and Biology. Circ Res. 2016;118:547–63. doi: 10.1161/CIRCRESAHA.115.306249. [DOI] [PubMed] [Google Scholar]
  • 138.Brown NK, Zhou Z, Zhang J, Zeng R, Wu J, Eitzman DT, Chen YE, Chang L. Perivascular adipose tissue in vascular function and disease: A review of current research and animal models. Arterioscler Thromb Vasc Biol. 2014;34:1621–1630. doi: 10.1161/ATVBAHA.114.303029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Öhman MK, Luo W, Wang H, Guo C, Abdallah W, Russo HM, Eitzman DT. Perivascular visceral adipose tissue induces atherosclerosis in apolipoprotein E deficient mice. Atherosclerosis. 2011;219:33–39. doi: 10.1016/j.atherosclerosis.2011.07.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Chang L, Villacorta L, Li R, Hamblin M, Xu W, Dou C, Zhang J, Wu J, Zeng R, Chen YE. Loss of perivascular adipose tissue on peroxisome proliferator-activated receptor-γ deletion in smooth muscle cells impairs intravascular thermoregulation and enhances atherosclerosis. Circulation. 2012;126:1067–1078. doi: 10.1161/CIRCULATIONAHA.112.104489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Poledne R, Králová Lesná I, Čejková S. Adipose tissue and atherosclerosis. Physiol Res. 2015;64(Suppl 3):S395–402. doi: 10.33549/physiolres.933152. [DOI] [PubMed] [Google Scholar]
  • 142.Henrichot E, Juge-Aubry CE, Pernin A, Pache JC, Velebit V, Dayer JM, Meda P, Chizzolini C, Meier CA. Production of chemokines by perivascular adipose tissue: A role in the pathogenesis of atherosclerosis? Arterioscler Thromb Vasc Biol. 2005;25:2594–2599. doi: 10.1161/01.ATV.0000188508.40052.35. [DOI] [PubMed] [Google Scholar]
  • 143.Rittig K, Dolderer JH, Balletshofer B, MacHann J, Schick F, Meile T, Küper M, Stock UA, Staiger H, MacHicao F, Schaller HE, Königsrainer A, Häring HU, Siegel-Axel DI. The secretion pattern of perivascular fat cells is different from that of subcutaneous and visceral fat cells. Diabetologia. 2012;55:1514–1525. doi: 10.1007/s00125-012-2481-9. [DOI] [PubMed] [Google Scholar]
  • 144.Rosito GA, Massaro JM, Hoffmann U, Ruberg FL, Mahabadi AA, Vasan RS, O’Donnell CJ, Fox CS. Pericardial fat, visceral abdominal fat, cardiovascular disease risk factors, and vascular calcification in a community-based sample the framingham heart study. Circulation. 2008;117:605–613. doi: 10.1161/CIRCULATIONAHA.107.743062. [DOI] [PubMed] [Google Scholar]
  • 145.Liu J, Fox CS, Hickson D, Sarpong D, Ekunwe L, May WD, Hundley GW, Carr JJ, Taylor HA. Pericardial adipose tissue, atherosclerosis, and cardiovascular disease risk factors: the Jackson heart study. Diabetes Care. 2010;33:1635–1639. doi: 10.2337/dc10-0245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Ajami B, Bennett JL, Krieger C, Tetzlaff W, Rossi FMV. Local self-renewal can sustain CNS microglia maintenance and function throughout adult life. Nat Neurosci. 2007;10:1538–1543. doi: 10.1038/nn2014. [DOI] [PubMed] [Google Scholar]
  • 147.Erblich B, Zhu L, Etgen AM, Dobrenis K, Pollard JW. Absence of Colony Stimulation Factor-1 Receptor Results in Loss of Microglia, Disrupted Brain Development and Olfactory Deficits. PLoS One. 2011;6:e26317. doi: 10.1371/journal.pone.0026317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Varvel NH, Grathwohl SA, Baumann F, Liebig C, Bosch A, Brawek B, Thal DR, Charo IF, Heppner FL, Aguzzi A, Garaschuk O, Ransohoff RM, Jucker M. Microglial repopulation model reveals a robust homeostatic process for replacing CNS myeloid cells. Proc Natl Acad Sci. 2012;109:18150–18155. doi: 10.1073/pnas.1210150109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Prokop S, Miller KR, Drost N, Handrick S, Mathur V, Luo J, Wegner A, Wyss-Coray T, Heppner FL. Impact of peripheral myeloid cells on amyloid-β pathology in Alzheimer’s disease–like mice. J Exp Med. 2015;212:1811–1818. doi: 10.1084/jem.20150479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Bruttger J, Karram K, Wörtge S, Regen T, Marini F, Hoppmann N, Klein M, Blank T, Yona S, Wolf Y, Mack M, Pinteaux E, Müller W, Zipp F, Binder H, Bopp T, Prinz M, Jung S, Waisman A. Genetic Cell Ablation Reveals Clusters of Local Self-Renewing Microglia in the Mammalian Central Nervous System. Immunity. 2015;43:92–107. doi: 10.1016/j.immuni.2015.06.012. [DOI] [PubMed] [Google Scholar]
  • 151.Elmore MRP, Najafi AR, Koike MA, Dagher NN, Spangenberg EE, Rice RA, Kitazawa M, Matusow B, Nguyen H, West BL, Green KN. Colony-stimulating factor 1 receptor signaling is necessary for microglia viability, unmasking a microglia progenitor cell in the adult brain. Neuron. 2014;82:380–397. doi: 10.1016/j.neuron.2014.02.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Ueno M, Fujita Y, Tanaka T, Nakamura Y, Kikuta J, Ishii M, Yamashita T. Layer V cortical neurons require microglial support for survival during postnatal development. Nat Neurosci. 2013;16:543–51. doi: 10.1038/nn.3358. [DOI] [PubMed] [Google Scholar]
  • 153.Marín-Teva JL, Dusart I, Colin C, Gervais A, Van Rooijen N, Mallat M. Microglia Promote the Death of Developing Purkinje Cells. Neuron. 2004;41:535–547. doi: 10.1016/s0896-6273(04)00069-8. [DOI] [PubMed] [Google Scholar]
  • 154.Takahashi K, Rochford CDP, Neumann H. Clearance of apoptotic neurons without inflammation by microglial triggering receptor expressed on myeloid cells-2. J Exp Med. 2005;201:647–657. doi: 10.1084/jem.20041611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Roumier A, Béchade C, Poncer J-C, Smalla K-H, Tomasello E, Vivier E, Gundelfinger ED, Triller A, Bessis A. Impaired synaptic function in the microglial KARAP/DAP12-deficient mouse. J Neurosci. 2004;24:11421–8. doi: 10.1523/JNEUROSCI.2251-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Paolicelli RC, Bolasco G, Pagani F, Maggi L, Scianni M, Panzanelli P, Giustetto M, Ferreira TA, Guiducci E, Dumas L, Ragozzino D, Gross CT. Synaptic pruning by microglia is necessary for normal brain development. Science. 2011;333:1456–8. doi: 10.1126/science.1202529. [DOI] [PubMed] [Google Scholar]
  • 157.Zeisel A, Muñoz-Manchado AB, Codeluppi S, Lönnerberg P, La Manno G, Juréus A, Marques S, Munguba H, He L, Betsholtz C, Rolny C, Castelo-Branco G, Hjerling-Leffler J, Linnarsson S. Brain structure. Cell types in the mouse cortex and hippocampus revealed by single-cell RNA-seq. Science. 2015;347:1138–42. doi: 10.1126/science.aaa1934. [DOI] [PubMed] [Google Scholar]
  • 158.Prinz M, Erny D, Hagemeyer N. Ontogeny and homeostasis of CNS myeloid cells. Nat Immunol. 2017;18:385–392. doi: 10.1038/ni.3703. [DOI] [PubMed] [Google Scholar]
  • 159.Goldmann T, Wieghofer P, Jordão MJC, Prutek F, Hagemeyer N, Frenzel K, Amann L, Staszewski O, Kierdorf K, Krueger M, Locatelli G, Hochgerner H, Zeiser R, Epelman S, Geissmann F, Priller J, Rossi FMV, Bechmann I, Kerschensteiner M, Linnarsson S, Jung S, Prinz M. Origin, fate and dynamics of macrophages at central nervous system interfaces. Nat Immunol. 2016;17:797–805. doi: 10.1038/ni.3423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Streit WJ, Graeber MB, Kreutzberg GW. Expression of Ia antigen on perivascular and microglial cells after sublethal and lethal motor neuron injury. Exp Neurol. 1989;105:115–126. doi: 10.1016/0014-4886(89)90111-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Kida S, Steart PV, Zhang ET, Weller RO. Perivascular cells act as scavengers in the cerebral perivascular spaces and remain distinct from pericytes, microglia and macrophages. Acta Neuropathol. 1993;85:646–52. doi: 10.1007/BF00334675. [DOI] [PubMed] [Google Scholar]
  • 162.Elmquist JK, Breder CD, Sherin JE, Scammell TE, Hickey WF, Dewitt D, Saper CB. Intravenous lipopolysaccharide induces cyclooxygenase 2-like immunoreactivity in rat brain perivascular microglia and meningeal macrophages. J Comp Neurol. 1997;381:119–29. doi: 10.1002/(sici)1096-9861(19970505)381:2<119::aid-cne1>3.0.co;2-6. [DOI] [PubMed] [Google Scholar]
  • 163.Gelderblom M, Leypoldt F, Steinbach K, Behrens D, Choe CU, Siler DA, Arumugam TV, Orthey E, Gerloff C, Tolosa E, Magnus T. Temporal and spatial dynamics of cerebral immune cell accumulation in stroke. Stroke. 2009;40:1849–1857. doi: 10.1161/STROKEAHA.108.534503. [DOI] [PubMed] [Google Scholar]
  • 164.Hammond MD, Taylor RA, Mullen MT, Ai Y, Aguila HL, Mack M, Kasner SE, McCullough LD, Sansing LH. CCR2+Ly6Chi Inflammatory Monocyte Recruitment Exacerbates Acute Disability Following Intracerebral Hemorrhage. J Neurosci. 2014;34:3901–3909. doi: 10.1523/JNEUROSCI.4070-13.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Ma Y, Li Y, Jiang L, Wang L, Jiang Z, Wang Y, Zhang Z, Yang G-Y. Macrophage depletion reduced brain injury following middle cerebral artery occlusion in mice. J Neuroinflammation. 2016;13:38. doi: 10.1186/s12974-016-0504-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Gliem M, Mausberg AK, Lee JI, Simiantonakis I, Van Rooijen N, Hartung HP, Jander S. Macrophages prevent hemorrhagic infarct transformation in murine stroke models. Ann Neurol. 2012;71:743–752. doi: 10.1002/ana.23529. [DOI] [PubMed] [Google Scholar]
  • 167.Schmidt A, Strecker JK, Hucke S, Bruckmann NM, Herold M, MacK M, Diederich K, Schäbitz WR, Wiendl H, Klotz L, Minnerup J. Targeting Different Monocyte/Macrophage Subsets Has No Impact on Outcome in Experimental Stroke. Stroke. 2017;48:1061–1069. doi: 10.1161/STROKEAHA.116.015577. [DOI] [PubMed] [Google Scholar]
  • 168.Szalay G, Martinecz B, Lénárt N, Környei Z, Orsolits B, Judák L, Császár E, Fekete R, West BL, Katona G, Rózsa B, Dénes Á. Microglia protect against brain injury and their selective elimination dysregulates neuronal network activity after stroke. Nat Commun. 2016;7:11499. doi: 10.1038/ncomms11499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Jin W-N, Shi SX-Y, Li Z, Li M, Wood K, Gonzales RJ, Liu Q. Depletion of microglia exacerbates postischemic inflammation and brain injury. J Cereb Blood Flow Metab. 2017;37:2224–2236. doi: 10.1177/0271678X17694185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Faraco G, Sugiyama Y, Lane D, Garcia-Bonilla L, Chang H, Santisteban MM, Racchumi G, Murphy M, Van Rooijen N, Anrather J, Iadecola C. Perivascular macrophages mediate the neurovascular and cognitive dysfunction associated with hypertension. J Clin Invest. 2016;126:4674–4689. doi: 10.1172/JCI86950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Hoeffel G, Wang Y, Greter M, See P, Teo P, Malleret B, Leboeuf M, Low D, Oller G, Almeida F, Choy SHY, Grisotto M, Renia L, Conway SJ, Stanley ER, Chan JKY, Ng LG, Samokhvalov IM, Merad M, Ginhoux F. Adult Langerhans cells derive predominantly from embryonic fetal liver monocytes with a minor contribution of yolk sac–derived macrophages. J Exp Med. 2012;209:1167–1181. doi: 10.1084/jem.20120340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Schulz C, Gomez Perdiguero E, Chorro L, Szabo-Rogers H, Cagnard N, Kierdorf K, Prinz M, Wu B, Jacobsen SEW, Pollard JW, Frampton J, Liu KJ, Geissmann F. A lineage of myeloid cells independent of Myb and hematopoietic stem cells. Science. 2012;336:86–90. doi: 10.1126/science.1219179. [DOI] [PubMed] [Google Scholar]
  • 173.Stadtfeld M, Ye M, Graf T. Identification of interventricular septum precursor cells in the mouse embryo. Dev Biol. 2007;302:195–207. doi: 10.1016/j.ydbio.2006.09.025. [DOI] [PubMed] [Google Scholar]
  • 174.van Rooijen N, van Kesteren-Hendrikx E. Clodronate liposomes: perspectives in research and therapeutics. J Liposome Res. 2002;12:81–94. doi: 10.1081/lpr-120004780. [DOI] [PubMed] [Google Scholar]
  • 175.Majmudar MD, Keliher EJ, Heidt T, Leuschner F, Truelove J, Sena BF, Gorbatov R, Iwamoto Y, Dutta P, Wojtkiewicz G, Courties G, Sebas M, Borodovsky A, Fitzgerald K, Nolte MW, Dickneite G, Chen JW, Anderson DG, Swirski FK, Weissleder R, Nahrendorf M. Monocyte-directed RNAi targeting CCR2 improves infarct healing in atherosclerosis-prone mice. Circulation. 2013;127:2038–2046. doi: 10.1161/CIRCULATIONAHA.112.000116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Sager HB, Dutta P, Dahlman JE, Hulsmans M, Courties G, Sun Y, Heidt T, Vinegoni C, Borodovsky A, Fitzgerald K, Wojtkiewicz GR, Iwamoto Y, Tricot B, Khan OF, Kauffman KJ, Xing Y, Shaw TE, Libby P, Langer R, Weissleder R, Swirski FK, Anderson DG, Nahrendorf M. RNAi targeting multiple cell adhesion molecules reduces immune cell recruitment and vascular inflammation after myocardial infarction. Sci Transl Med. 2016;8:342ra80. doi: 10.1126/scitranslmed.aaf1435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Tang J, Lobatto ME, Hassing L, van der Staay S, van Rijs SM, Calcagno C, Braza MS, Baxter S, Fay F, Sanchez-Gaytan BL, Duivenvoorden R, Sager H, Astudillo YM, Leong W, Ramachandran S, Storm G, Pérez-Medina C, Reiner T, Cormode DP, Strijkers GJ, Stroes ESG, Swirski FK, Nahrendorf M, Fisher EA, Fayad ZA, Mulder WJM. Inhibiting macrophage proliferation suppresses atherosclerotic plaque inflammation. Sci Adv. 2015;1:e1400223–e1400223. doi: 10.1126/sciadv.1400223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Duivenvoorden R, Tang J, Cormode DP, Mieszawska AJ, Izquierdo-Garcia D, Ozcan C, Otten MJ, Zaidi N, Lobatto ME, van Rijs SM, Priem B, Kuan EL, Martel C, Hewing B, Sager H, Nahrendorf M, Randolph GJ, Stroes ESG, Fuster V, Fisher EA, Fayad ZA, Mulder WJM. A statin-loaded reconstituted high-density lipoprotein nanoparticle inhibits atherosclerotic plaque inflammation. Nat Commun. 2014;5:3065. doi: 10.1038/ncomms4065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Lobatto ME, Fayad ZA, Silvera S, Vucic E, Calcagno C, Mani V, Dickson SD, Nicolay K, Banciu M, Schiffelers RM, Metselaar JM, Van Bloois L, Wu HS, Fallon JT, Rudd JH, Fuster V, Fisher EA, Storm G, Mulder WJM. Multimodal clinical imaging to longitudinally assess a nanomedical anti-inflammatory treatment in experimental atherosclerosis. Mol Pharm. 2010;7:2020–2029. doi: 10.1021/mp100309y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.van der Valk FM, van Wijk DF, Lobatto ME, Verberne HJ, Storm G, Willems MCM, Legemate DA, Nederveen AJ, Calcagno C, Mani V, Ramachandran S, Paridaans MPM, Otten MJ, Dallinga-Thie GM, Fayad ZA, Nieuwdorp M, Schulte DM, Metselaar JM, Mulder WJM, Stroes ES. Prednisolone-containing liposomes accumulate in human atherosclerotic macrophages upon intravenous administration. Nanomedicine. 2015;11:1039–46. doi: 10.1016/j.nano.2015.02.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Keliher EJ, Ye Y-X, Wojtkiewicz GR, Aguirre AD, Tricot B, Senders ML, Groenen H, Fay F, Perez-Medina C, Calcagno C, Carlucci G, Reiner T, Sun Y, Courties G, Iwamoto Y, Kim H-Y, Wang C, Chen JW, Swirski FK, Wey H-Y, Hooker J, Fayad ZA, Mulder WJM, Weissleder R, Nahrendorf M. Polyglucose nanoparticles with renal elimination and macrophage avidity facilitate PET imaging in ischaemic heart disease. Nat Commun. 2017;8:14064. doi: 10.1038/ncomms14064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.McArdle S, Chodaczek G, Ray N, Ley K. Intravital live cell triggered imaging system reveals monocyte patrolling and macrophage migration in atherosclerotic arteries. J Biomed Opt. 2015;20:26005. doi: 10.1117/1.JBO.20.2.026005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Lo Celso C, Fleming HE, Wu JW, Zhao CX, Miake-Lye S, Fujisaki J, Côté D, Rowe DW, Lin CP, Scadden DT. Live-animal tracking of individual haematopoietic stem/progenitor cells in their niche. Nature. 2009;457:92–6. doi: 10.1038/nature07434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Courties G, Herisson F, Sager HB, Heidt T, Ye Y, Wei Y, Sun Y, Severe N, Dutta P, Scharff J, Scadden DT, Weissleder R, Swirski FK, Moskowitz MA, Nahrendorf M. Ischemic stroke activates hematopoietic bone marrow stem cells. Circ Res. 2015;116:407–417. doi: 10.1161/CIRCRESAHA.116.305207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Tarkin JM, Joshi FR, Rudd JHF. PET imaging of inflammation in atherosclerosis. Nat Rev Cardiol. 2014;11:443–57. doi: 10.1038/nrcardio.2014.80. [DOI] [PubMed] [Google Scholar]
  • 186.Folco EJ, Sheikine Y, Rocha VZ, Christen T, Shvartz E, Sukhova GK, Di Carli MF, Libby P. Hypoxia but not inflammation augments glucose uptake in human macrophages: Implications for imaging atherosclerosis with 18fluorine-labeled 2-deoxy-D-glucose positron emission tomography. J Am Coll Cardiol. 2011;58:603–14. doi: 10.1016/j.jacc.2011.03.044. [DOI] [PubMed] [Google Scholar]
  • 187.Pedersen SF, Græbe M, Hag AMF, Højgaard L, Sillesen H, Kjær A. (18)F-FDG imaging of human atherosclerotic carotid plaques reflects gene expression of the key hypoxia marker HIF-1α. Am J Nucl Med Mol Imaging. 2013;3:384–92. [PMC free article] [PubMed] [Google Scholar]
  • 188.Nahrendorf M, Zhang H, Hembrador S, Panizzi P, Sosnovik DE, Aikawa E, Libby P, Swirski FK, Weissleder R. Nanoparticle PET-CT imaging of macrophages in inflammatory atherosclerosis. Circulation. 2008;117:379–387. doi: 10.1161/CIRCULATIONAHA.107.741181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Majmudar MD, Yoo J, Keliher EJ, Truelove JJ, Iwamoto Y, Sena B, Dutta P, Borodovsky A, Fitzgerald K, Di Carli MF, Libby P, Anderson DG, Swirski FK, Weissleder R, Nahrendorf M. Polymeric nanoparticle PET/MR imaging allows macrophage detection in atherosclerotic plaques. Circ Res. 2013;112:755–761. doi: 10.1161/CIRCRESAHA.111.300576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Junker A, Kokornaczyk AK, Zweemer AJM, Frehland B, Schepmann D, Yamaguchi J, Itami K, Faust A, Hermann S, Wagner S, Schäfers M, Koch M, Weiss C, Heitman LH, Kopka K, Wünsch B. Synthesis, binding affinity and structure-activity relationships of novel, selective and dual targeting CCR2 and CCR5 receptor antagonists. Org Biomol Chem. 2015;13:2407–22. doi: 10.1039/c4ob02397h. [DOI] [PubMed] [Google Scholar]
  • 191.Luehmann HP, Detering L, Fors BP, Pressly ED, Woodard PK, Randolph GJ, Gropler RJ, Hawker CJ, Liu Y. PET/CT Imaging of Chemokine Receptors in Inflammatory Atherosclerosis Using Targeted Nanoparticles. J Nucl Med. 2016;57:1124–1129. doi: 10.2967/jnumed.115.166751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Wan MYS, Endozo R, Michopoulou S, Shortman R, Rodriguez-Justo M, Menezes L, Yusuf S, Richards T, Wild D, Waser B, Reubi JC, Groves A. PET/CT Imaging of Unstable Carotid Plaque with (68)Ga-Labeled Somatostatin Receptor Ligand. J Nucl Med. 2017;58:774–780. doi: 10.2967/jnumed.116.181438. [DOI] [PubMed] [Google Scholar]
  • 193.Tarkin JM, Joshi FR, Evans NR, Chowdhury MM, Figg NL, Shah AV, Starks LT, Martin-Garrido A, Manavaki R, Yu E, Kuc RE, Grassi L, Kreuzhuber R, Kostadima MA, Frontini M, Kirkpatrick PJ, Coughlin PA, Gopalan D, Fryer TD, Buscombe JR, Groves AM, Ouwehand WH, Bennett MR, Warburton EA, Davenport AP, Rudd JHF. Detection of Atherosclerotic Inflammation by 68Ga-DOTATATE PET Compared to [18F]FDG PET Imaging. J Am Coll Cardiol. 2017;69:1774–1791. doi: 10.1016/j.jacc.2017.01.060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Cerami C, Perani D. Imaging neuroinflammation in ischemic stroke and in the atherosclerotic vascular disease. Curr Vasc Pharmacol. 2015;13:218–22. doi: 10.2174/15701611113116660168. [DOI] [PubMed] [Google Scholar]
  • 195.Dollé F, Luus C, Reynolds A, Kassiou M. Radiolabelled molecules for imaging the translocator protein (18 kDa) using positron emission tomography. Curr Med Chem. 2009;16:2899–923. doi: 10.2174/092986709788803150. [DOI] [PubMed] [Google Scholar]
  • 196.Zinnhardt B, Viel T, Wachsmuth L, Vrachimis A, Wagner S, Breyholz H-J, Faust A, Hermann S, Kopka K, Faber C, Dollé F, Pappata S, Planas AM, Tavitian B, Schäfers M, Sorokin LM, Kuhlmann MT, Jacobs AH. Multimodal imaging reveals temporal and spatial microglia and matrix metalloproteinase activity after experimental stroke. J Cereb Blood Flow Metab. 2015;35:1711–21. doi: 10.1038/jcbfm.2015.149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Cosenza-Nashat M, Zhao M-L, Suh H-S, Morgan J, Natividad R, Morgello S, Lee SC. Expression of the translocator protein of 18 kDa by microglia, macrophages and astrocytes based on immunohistochemical localization in abnormal human brain. Neuropathol Appl Neurobiol. 2009;35:306–28. doi: 10.1111/j.1365-2990.2008.01006.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Mandal PK, Ferreira LMR, Collins R, Meissner TB, Boutwell CL, Friesen M, Vrbanac V, Garrison BS, Stortchevoi A, Bryder D, Musunuru K, Brand H, Tager AM, Allen TM, Talkowski ME, Rossi DJ, Cowan CA. Efficient ablation of genes in human hematopoietic stem and effector cells using CRISPR/Cas9. Cell Stem Cell. 2014;15:643–652. doi: 10.1016/j.stem.2014.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Heckl D, Kowalczyk MS, Yudovich D, Belizaire R, Puram RV, McConkey ME, Thielke A, Aster JC, Regev A, Ebert BL. Generation of mouse models of myeloid malignancy with combinatorial genetic lesions using CRISPR-Cas9 genome editing. Nat Biotechnol. 2014;32:941–946. doi: 10.1038/nbt.2951. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Gundry MC, Brunetti L, Lin A, Mayle AE, Kitano A, Wagner D, Hsu JI, Hoegenauer KA, Rooney CM, Goodell MA, Nakada D. Highly Efficient Genome Editing of Murine and Human Hematopoietic Progenitor Cells by CRISPR/Cas9. Cell Rep. 2016;17:1453–1461. doi: 10.1016/j.celrep.2016.09.092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Dever DP, Bak RO, Reinisch A, Camarena J, Washington G, Nicolas CE, Pavel-Dinu M, Saxena N, Wilkens AB, Mantri S, Uchida N, Hendel A, Narla A, Majeti R, Weinberg KI, Porteus MH. CRISPR/Cas9 β-globin gene targeting in human haematopoietic stem cells. Nature. 2016;539:384–389. doi: 10.1038/nature20134. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES