Abstract
In the absence of an immune challenge, healthy, aged individuals have a significantly higher basal inflammatory state where circulating levels of cytokines, including IL-6, TNF-α and IL-1β, are elevated[1]. This progressive pro-inflammatory state, termed “inflamm-aging,” affects the phenotype/function of cells present in the aged as well as renders the older individuals more susceptible to a poor prognosis after systemic insults. Although it is important to understand the mechanisms that underlie the progression of disease, most preclinical analyses of disease therapies are performed in young adult mice that have an intact, functional immune system. Oftentimes, this is not necessarily representative of the immune disposition in the aged, let alone diseased, aged. Herein, two distinct responses that are not only commonly associated with aging but that also have dendritic cells and/or monocytes and macrophages as key players are discussed: pulmonary infection and myocardial infarction. Although studies of pulmonary infection in the aged have progressed significantly, studies of monocytes and macrophages in inflammation and cardiac injury following ischemia in the aged have not been as forthcoming. Nonetheless, several elegant studies have established the dynamic role of monocytes and macrophages post infarction. These will be discussed in light of what is known with aging.
Keywords: aging, monocytes, macrophage, dendritic cell, lung, heart
1. Introduction
Macrophages and dendritic cells (DCs)1 play pivotal roles in modulating immune function following infection and tissue injury [2-4]. As professional antigen presenting cells, these cells help shape the innate and adaptive immune responses. In response to insult, DCs are activated to induce adaptive immunity, while monocytes and macrophages initiate an inflammatory response. Monocytes and macrophages respond through the production of pro-inflammatory cytokines and anti-microbial mediators. Macrophages are not only key players in the initiation of inflammation, but also orchestrate its resolution, and in the case of sterile injury, wound healing. Plasticity is characteristic of macrophages, whose phenotype and function evolves in response to changing conditions within their microenvironment.
Macrophages can be broadly categorized into two subsets. The classically activated or M1 macrophages are induced by the bacterial cell wall component lipopolysaccharide (LPS) or a combination of Th1 cytokines, IFN-γ and TNF-α. The alternatively activated or M2 macrophages are induced by Th2 cytokines such as IL-4 and IL-13[5]. M1 macrophages are characterized by the production of reactive oxygen species (ROS), reactive nitrogen intermediates, IL-1, IL-12, and TNF-α[3,6]. In addition, M1 macrophages drive Th1 responses and kill intracellular foreign pathogens[4]. Anti-inflammatory, i.e., M2 macrophages, express arginase-1, scavenger and mannose receptors, and the intracellular proteins Found In Inflammatory Zone 1 (FIZZ1) and Ym1[7,8]. M2 macrophages have elevated levels of IL-10 and predominately function in immunoregulation, neovascularization and tissue remodeling[4,9].
Aging is characterized by the elevation in baseline inflammation (called “inflamm-aging”) with a refractory response to immune challenge (known as “immunosenescence”). Importantly, the chronic exposure to systemic low levels of pro-inflammatory cytokines modulates phagocytic mononuclear cell activity, and skews the M1/M2 populations present in the aged independent of injury or infection[10-13]. Superimposed on these changes in the ‘milieu’ and steady state populations, is the aged response. Whether responding to tissue injury or infection, a state of diminished and prolonged inflammation ensues. In addition to affecting pathogen clearance, age-related changes in nitric oxide production and phagocytic activity lead to impaired removal of damaged tissue and promote adverse remodeling following injury. Thus, a disruption in the balance between inflammation and immune activation may contribute to a variety of co-morbidities and increased mortalities following local and systemic insults[14-19].
With a focus on monocytes, macrophages and DCs, this review will cover age-associated changes in the development and function of these cells in the context of two distinct responses, a respiratory infection and a sterile insult, i.e., myocardial infarction. These responses were chosen due to their high incidence of morbidity and mortality in the aged population. Recent studies have elucidated the controversy regarding the lineage of monocytes and macrophages with an emerging picture that some macrophages derive from a hematopoietic stem cell lineage that involves specific progenitor intermediates and monocytes[20], whereas other macrophages derive from primitive macrophages that have colonized tissue prior to hematopoiesis[21-24]. The observations that macrophages can polarize to various functional states[25,26] as well as the emerging identification of distinct monocyte subsets[27] has fostered the idea that macrophages are fated for specific functions, suggesting that harmful subsets can be therapeutically targeted while those that are beneficial can be spared. Although in some cases the findings are so new that potential changes with age have yet to be determined, this review will cover macrophage lineage and heterogeneity in the young noting areas of potential differences in the aged.
2. Pulmonary Response
2.1. Mononuclear phagocytes in the lung
Constant exposure to airborne pollutants and microbial challenges renders the respiratory tract especially vulnerable to infection and damage. Highlighting this state of permanent alert, the lung, comprised of parenchyma and alveolar space, contains numerous mononuclear phagocytes, including DCs and macrophages. In the steady state, macrophages are the major cell component of the alveolar space and are key for removal of pathogens and debris[28]. Like in other epithelia and skin, throughout the lung airways and interstitium is an elaborate network of DCs that performs unique sentinel function for pulmonary immune responses. At least five phenotypically distinct DC subsets have been identified, a minority population of the plasmacytoid type, and several distinct myeloid “conventional” subsets[29]. The definition of tissue DCs and macrophages has relied upon phenotypic determinations, which are not definitive, leading to some confusion surrounding the exact contribution of DCs vs. macrophages to tissue immunity. More exacting analyses, such as transcriptional profiling should shed more light on this issue[30].
Alveolar macrophages and interstitial macrophages are distinct phenotypically and functionally[31,32]. Alveolar macrophages are unique amongst tissue-resident macrophages in both phenotype and function. Their phenotype is similar to DCs, e.g. high levels of CD11c and CD205 expression. Their ability to cross-present antigen is higher than other macrophage populations[31] and they are able to clear particulates and pathogens without the induction of inflammation or recruitment of neutrophils or monocytes[33-35]. The lung microenvironment that is particularly higher in oxygen tension and levels of GM-CSF and M-CSF, are critical for the induction and maintenance of the alveolar macrophage population[31,36]. Alveolar macrophages differ from the pulmonary interstitial macrophages in origin and lifespan as well. They populate the lung during embryogenesis and renew largely from tissue resident populations, with little contribution from the bone marrow[24,37]. Blood-borne precursors are thought to undergo an obligate intermediate differentiation step within the parenchyma transitioning to macrophages that subsequently migrate into the alveolar space[28]. Under steady-state, alveolar macrophages turn over very slowly with a half-life of 30-60 days[38] and a population half-life of 40%/year[39]. In contrast, pulmonary interstitial macrophages originate from bone marrow-derived monocytes and have a shorter lifespan[28,40]. Upon activation they actively recruit other inflammatory cells[33,35,41,42]. Viral and bacterial infection results in the appearance of other monocytic and macrophage-like cells within the lungs. Some of these cells likely differentiate into DCs[43,44] and participate in pathogen clearance; however, an overabundance of these cells may result in pathologic changes in the lung.
DCs are classified into conventional and plasmacytoid DCs (PDC) subsets that differ in phenotype and ontogeny. PDC have a limited ability to populate non-lymphoid tissues in steady state or to phagocytose and present antigen. Within the lung, in the absence of inflammation, two populations of conventional DCs can be identified, the CD103+ and the CD11b+ subsets. The CD103+ population forms a network within the epithelial layer of the airways and extends protrusions between the basolateral spaces. The CD11b+ DC subsets reside underneath the basement membrane within the lamina propria[43]. Pulmonary DCs turnover rapidly, with a half-life of 1.5-2 days in the airway and 3-4 days in the parenchyma[45]. The majority of DCs in the lung are derived from the circulating blood monocyte pool, while the PDC population arises from Flt3+ conventional DC (cDC)[29]. The pulmonary CD103+CD11b− DCs are potent at production of IL-12, cross-presentation of antigens to CD8 T cells, and induction of CD8 T cell differentiation[46-49].
DCs sample their environment through a collection of receptors that bind various pathogens or damage associated molecular patterns (PAMPS and DAMPS)[29,45]. Reduced expression of these receptors or alterations in their signaling pathways can increase susceptibility to infection[50-52]. Upon ingestion of antigen, DCs become activated, up-regulating surface expression of activation markers, mobilizing to local lymph nodes and secreting cytokines. The production of Type I and III interferons (IFNs) by the PDC subset is critical for anti-viral responses, while the profile of cytokine production by the cDC subsets directs Th cell polarization. Similarly, lung resident macrophages recognize PAMPS and DAMPS by expressing anti-microbial reactive oxygen and nitrogen species; thus, shaping the innate and adaptive immune responses through the release of a variety of pro-inflammatory mediators at early time points post injury or infection. These cells also dampen the inflammatory response through phagocytosis of apoptotic cells and secretion of soluble factors, e.g., IL-10, and can participate in tissue remodeling and repair[4,9].
Advancing age affects the respiratory system in a variety of ways including alterations in macrophage and DC function that impact innate and adaptive immunity. Among the earlier observed alterations in DCs were changes in the overall numbers and subset distribution present in various tissues, activation-induced expression of co-stimulatory molecules, cytokine synthesis, migration, antigen-presentation and T cell activation (reviewed in[53-56]. Variable results and conflicting conclusions are abundant, likely reflecting differing methods used in the identification of cell populations, different cell sources, reagents, and health status, but consensus is emerging. The preponderance of evidence indicates that aging does not result in a change in the number of DCs in the lungs, or most other lymphoid tissues, although there may be changes in subset frequency[56]. The basal expression levels of most markers on DCs, including Toll-like receptor (TLR) are also unchanged across the lifespan. However, the TLR expression profile in macrophages may change with age. It has been reported that all TLRs are decreased in peritoneal and splenic macrophages of aged C57BL/6 mice[57,58] whereas no differences were reported in TLR2 and TLR4 in aged BALB/c mice[59-61]. Basal levels of pro-inflammatory cytokine production is elevated in aged DCs and macrophages derived from several tissue sources[62-65]. Indeed, elevated inflammatory cytokine levels in the lung and other evidence of chronic inflammation are commonly identified in aged humans[63,66,67] and mice[68]. However, TLR-mediated pro-inflammatory cytokine production by aged DCs and macrophages is often reduced relative to young cells[18,55,62-64,69]. In vivo stimulation with live or attenuated virus or bacteria bolster this conclusion, indicating that the capacity of aged DCs and macrophages for cytokine production is both diminished and delayed[63,70-77]. TLR ligand stimulus or viral infection-induced Type I and III IFN synthesis is universally found to be lower in aged DCs[53,56,78]. Similarly, age-associated defects in the signaling of other pattern recognition receptors, i.e., RIG-1 and NLRP-3, have led to impaired IFN or IL-1β production after stimulation with West Nile virus and influenza virus, respectively[74,79].
2.2. Molecular Mechanisms: GSK-3, miRNA, histone modifications, and oxidative stress
At the center of the signal transduction network regulating inflammatory cytokine gene expression is the glycogen synthase kinase-3 (GSK-3) family of serine/threonine kinases[80-83]. GSK-3 is a constitutively active protein kinase that has broad regulatory influence due to the multiplicity of substrates that it can phosphorylate. These substrates include metabolic enzymes, signaling molecules, structural proteins and transcription factors, typically involved in regulating cell proliferation and differentiation, cellular metabolism, cell survival and cell cycle regulation[82,84]. Evidence suggests that GSK-3 is a novel regulator of aging that retards age-related pathologies in a wide variety of tissues[85]. De-regulation of GSK-3 on the other hand, has been associated with the initiation or progression of many diseases[86-88] and the induction of cellular senescence[89].
GSK-3 plays a pivotal role in regulating the production of pro- and anti-inflammatory cytokines in DCs and macrophages. This enzyme is comprised of two isoforms both of which are constitutively active under basal conditions and whose activity can be differentially regulated by phosphorylation[83], intracellular localization, and protein complex formation[86]. In general, GSK inactivation by N-terminal serine phosphorylation in DCs and macrophages augments anti-inflammatory cytokine production while concurrently suppressing the production of pro-inflammatory cytokines[82], although the outcome of GSK-3 inactivation is complex and context specific.
Recent findings implicate the de-regulation of GSK-3 activity in age-related changes in pro-inflammatory cytokine production, particularly by means of altered phosphoinositide-3 kinase (PI3K) activity in aged macrophages and DCs. PI3Ks are a family of lipid kinases that phosphorylate the hydroxyl group of the inositol ring of phosphoinositides. The resulting phosphorylated products regulate a multitude of cellular events including cytokine production[90]. PI3K recruits and activates the serine-threonine kinase Akt that subsequently phospho-inactivates GSK-3, thus shifting the balance of pro-and anti-inflammatory cytokine production[91]. Fallah, et al.[71] reported that in aged murine splenic macrophages gene expression for both the catalytic and regulatory subunits of the Class IA PI3K was higher than in young macrophages. This increased mRNA for PI3K was associated with heightened phosphorylation of Akt and GSK-3 in the aged cells. Interestingly, the age-associated decline in pro-inflammatory cytokine production by aged macrophages to Streptococcus pneumoniae activation was corrected by inhibition of PI3K. Also implicating GSK-3 associated signaling pathway alterations mediating aging changes were the results of Boyd et al.[70]. This group reported alveolar macrophages taken from aged mice displayed lower phosphorylation of p65 NF-κB, JNK and p38 MAPK and an increase in ERK phosphorylation, consistent with an up-stream inhibition through GSK. These data suggest that alterations in signal transduction pathway activity participate in the reduced innate immune inflammatory responses of the elderly, but clearly other mechanisms are active as well.
Xu, et al.[92] report that Langerhans cells in the skin of aged mice display a unique microRNA (miRNA) pattern. MiRNAs are small, non-coding RNA molecules that are key regulators of gene expression by means of their capacity to limit translation of specific RNAs. Although a comparative analysis of miRNA expression in young and aged whole lungs did not reveal age-associated changes[93], Jiang, et al.[94] found high levels of miR-146a in murine macrophages in aged animals. miR-146a negatively regulates the expression of IL-1β and IL-6. These authors further found that histone modifications play a role regulating miR-146a expression, as suppression of histone deacetylase activity improved the inflammatory response of aged macrophages. Histone modification, such as by methylation or acetylation can have a dramatic impact on chromatin structure and thereby play a critical role in gene activation and induction of expression through controlling DNA accessibility to polymerases and transcription factors. An analysis of chromatin structure of human monocyte-derived DCs revealed that the promoter regions for genes encoding IL-29 and IFN-A2 are more highly associated with the repressor histone in aged populations than in young[78]. This was accompanied by a decreased association of these promoters with the activator histone after activation.
ROS are physiologically produced by all cells and mostly derived from leakage of the electron transport chain in mitochondria[95,96]. In inflammatory cells a second important source of ROS production is the “oxidative burst”, where the NADPH oxidase complex catalyzes the formation of hypochlorite, hypochlorous, and hypobromous acid to eradicate pathogens[97,98]. In aging cells an imbalance due to increased ROS production and a decrease in the levels of activity of the ROS-converting enzymes, leads to the non-enzymatic oxidation of proteins, carbohydrates, lipids and nucleic acids, a process generally known as oxidative stress[99-105]. Mildly oxidized cytosolic proteins are degraded by the proteasome system or by chaperone-mediated autophagy[106-108]. Extensively oxidized proteins become irreversibly aggregated and are degraded through aggrephagy, a selective form of macroautophagy. In aged cells of the immune system, there is an increased level of free radicals[109], a decreased level/function of enzymes involved in clearing free radicals[110] which participate in compromising phagocytosis, proteasomal activity and TLR signaling[19,69,111]. Recently, Cannizo et al.[112] showed that DCs isolated from the spleen and lymph nodes as well as the CD34+ bone marrow precursors of old mice accumulate oxidatively modified proteins with side chain carbonylation, advanced glycation end products and lipid peroxidation, and that endosomal accumulation of oxidatively modified proteins interferes with the efficient processing of exogenous antigens and degradation of macroautophagy delivered proteins. Further, this group [112] demonstrated that in vivo treatment with antioxidant improves aged DC antigen processing and presentation. It is likely pulmonary DCs also accumulate oxidatively modified proteins which negatively impacts their activity, contributing to immunosenescence in this tissue.
2.3. Senescence associated secretory profile (SASP) and the aged microenvironment
Macrophage phenotypic and functional profiles are highly plastic and dependent on external cues, and DC function is also strongly influenced by the microenvironmental milieu, as previously mentioned. It has recently become apparent that the development of age-associated chronic inflammation has significant impacts on these cell types, modifying innate immune reactions in the aged. The source of the inflammatory cytokines and the mechanisms that maintain the chronic inflammatory state are somewhat enigmatic, but it has been hypothesized that cellular senescence underlies the phenomenon. Cellular senescence is the state of permanent inhibition of cell division. It is associated with a unique secretory phenotype, termed the senescence-associated secretory phenotype (SASP), characterized by production of pro-inflammatory factors[113-115]. It is likely the SASP phenomenon explains the paradoxical situation of increased basal inflammation accompanied by reduced pathogen-induced inflammatory responses. The consequences of SASP on disease susceptibility have been clearly demonstrated, affecting both the incidence and severity of inflammatory diseases such as COPD and IDF [116] as well as infectious disease.
The aging lung displays elevated basal inflammation[63,66-68,117] that contributes to increased susceptibility to infectious disease in a number of ways. The pulmonary SASP includes increased level of the markers, keratin 10, laminin receptor and platelet activation factor receptor, all of which act as adhesion receptors for bacteria, including S. pneumoniae [117]. Elevated circulating levels of TNF-α, another pro-inflammatory factor produced by senescent cells, strongly affects the function of aged pulmonary macrophages, as demonstrated in an elegant series of papers by the Orihuela group[70,118-120]. These investigators identified age-dependent changes in the activity of alveolar macrophages that included reduced phagocytosis and induced cytokine production, a complex of changes that they termed age-dependent macrophage dysfunction (ADMD). They demonstrated that after infection with S. pneumoniae, the reduction in pro-inflammatory cytokine production by aged alveolar macrophages was not due to lowered expression levels of TLR, but to reduced NF-κB activation. This signaling defect could be replicated in young alveolar macrophages by pre-treatment of the cells with physiologic levels of TNF-α, linking the response profile of the aged macrophages to the changes in the milieu that occurs as the epithelial cells undergo cellular senescence and express the SASP phenotype. Further work dissecting the molecular signaling pathways indicated that reduced NF-κB activation was in part due to increased expression in the aged cells of A20 whose activity reduces TRAF6 polyubiquitination and thus lowers NF-κB activation, and consequently the expression of pro-inflammatory cytokines. Enhanced A20 expression is also linked to exposure to TNF-α, whose basal levels is elevated as a sequela of epithelial cell senescence.
Respiratory DC migration upon viral infection also is reduced in aged mice due to elevated levels of prostaglandin D2 expression in the lungs[121], another characteristic of senescent cells. Taken together these data suggest that senescence-associated changes in the lung parenchymal cells, particularly the epithelial compartment, create a microenvironment that depresses normal, immunoprotective activities of the resident macrophages and DCs[6]. Of relevance to improving immune function in the elderly, alveolar macrophage and DC activity can be improved both by dietary manipulations[119] as well as administration of mTOR inhibitory drugs, reversing the negative effects of pulmonary cellular senescence[122].
3. Cardiac Ischemia
3.1. Role of monocytes/macrophages in myocardial ischemia
Atherosclerotic lesions that rupture in coronary arteries cause myocardial infarction. This ischemic event kills cardiomyocytes and triggers the influx of myeloid cells (see Table I). Neutrophils are the first leukocyte population to infiltrate the ischemic site[123]. Shortly thereafter, monocytes and macrophages can be found in the infarct at numbers up to a million cells. Cells with an inflammatory phenotype (Ly-6Chigh monocytes/M1 type macrophages) dominate initially, followed by cells with a lesser inflammatory phenotype promoting tissue repair (Ly-6Clow/int monocytes/M2 type macrophages). Other leukocytes invading the infarct in lower cell numbers include DCs[124], lymphocytes[125,126] and mast cells[127]. During the wound healing process, a deficiency in the influx of CD4 T cells delays the transition from an inflammatory to reparatory phase and DC depletion affects the resolution of inflammation[124]; however the mechanism underlying these changes are unclear. When inflammation resolves, non-leukocyte cells join the rebuilding activities in the infarct. Angiogenic factors induce the growth of numerous vessels into newly forming granulation tissue. Myofibroblasts produce collagen that strengthens the emerging infarct scar.
Table I.
INFLAMMATORY PHASE | REPARATIVE PHASE | ||
---|---|---|---|
Neutrophil influx -phagocytose dead cells/debris -release inflammatory mediators |
Influx of Ly6-Chi mono to elevated MCP-1 -phagocytosis -produce pro-inflammatory mediators and proteases --tissue destabilization |
Increase Ly6-Clo/int mono to elevated CX3CL1 -phagocytosis -produce VEGF, TGFβ --support angiogenesis and collagen synthesis |
|
AGED RESPONSE | Reduced neutrophil influx; Timely neutrophil clearance | Fewer early macrophages; Reduced levels of MCP-1, IL-1β, TNF-α, IL-6, M-CSF; Increased levels of IL-10; Persistence of dead cardiomyocytes in infarcted area | Macrophages peak later; Defective scar formation (reduced myofibroblast density and decreased collagen deposition in infarcted area); Comparable TGFβ levels |
Monocytes accumulating in the infarcted myocardium arrive from the bone marrow and spleen[128] in two sequential phases (i.e., inflammatory and reparative): Ly-6Chigh monocytes arrive during the first days post myocardial infarction in response to MCP-1, whereas days later Ly-6Clow/int monocytes arrive second in response to fractalkine (CX3CL1)[123]. This time course corresponds to expression of M1-type markers in tissue early after injury and M2-type macrophage markers later[129]. In the infarct, myeloid cells turnover very rapidly, and monocytes are recruited at a high rate[130]. Thereafter, most cells become apoptotic, and a fraction of the accumulated CD11b+ population exits the infarct within 24hrs and accumulates in the liver, lymph node and spleen[130]. During days 1 to 4 after ischemia, the milieu is highly inflammatory with Ly-6Chigh cells secreting TNF-α and proteases[123]. Dead cells, extracellular matrix and debris surrounding the infarct are cleared by phagocytosis.
At certain levels, inflammation may promote adverse effects, e.g., the tissue-destabilizing function of proteases may lead to infarct expansion, rupture, and dilation of the left ventricle[131]. However, inflammation is necessary to clear dead cardiac tissue and begin the active process of resolving inflammation, as well as to promote scar formation. Marginated leukocytes clear the dying and necrotic cardiomyocytes and support fibrogenic and angiogenic responses[131,132]. Thus, modulation of the inflammatory response post myocardial infarction contributes to the quality of heart repair[123,133,134]. Harel-Adar et al.[135] showed that modulation of cardiac macrophages to a reparative state at a predetermined time after myocardial infarction promoted angiogenesis, the preservation of small scars, and prevented ventricular dilatation and remodeling.
During this rebuilding phase, the inflammatory activity resolves and gives way to Ly-6Clow/int monocytes/macrophages. These cells release vascular endothelial growth factor (VEGF) and TGFβ, supporting angiogenesis and collagen production. In the aged (Table I), studies have reported a prolonged course of wound repair, associated with a prolonged inflammatory state, and delayed neovascularization and restoration of the extracellular matrix[16,136]. Specifically, in older mice post myocardial infarct, there is impaired inflammation with decreased and delayed neutrophil and macrophage infiltration, reduced cytokine and chemokine expression and impaired phagocytosis of dead cardiomyocytes[16,136]. The impaired inflammation and suboptimal clearance of dead cardiomyocytes that may lead to maladaptive vascular remodeling and tissue repair in the healing heart and therefore accelerate transition into heart failure.
3.2. Monocyte/macrophage lineage and heterogeneity at steady state and in the infarct
3.2.1. Steady State
Although evidence for lineage relationship in the infarcted myocardium is rather sparse, the developmental relationship between monocyte subsets and macrophages is emerging. Using complementary in vivo cell tracking, parabiosis, bone marrow transplants and fate-mapping studies, Epelman et al.[21] found the majority of cardiac macrophages were established during embryogenesis and persist into adulthood in substantial numbers. Unlike the lung, there are very few macrophages present in the uninjured heart at steady state. The heart contains two separate and discrete cardiac macrophage pools[21]. The first pool (CCR2−, CD11clow) includes the majority of MHC-IIhigh, MHC-IIlow, and Ly-6C+ macrophages. These macrophage subsets are separate from the blood monocyte pool and represent an embryonically established lineage made up of progeny from yolk sac macrophages and fetal monocytes. The second pool of macrophages is smaller in number and is derived from blood CCR2+ Ly-6Chigh monocytes. Both embryonic and adult derived macrophages are maintained through local proliferation and replacement by blood monocytes, respectively.
In the mouse, circulating monocytes are phenotypically and functionally heterogeneous and can be separated based on Ly-6C expression[123,137]. In the steady state 50-60% of monocytes belong to the Ly-6Chigh CCR2high CX3CR1low CD62L+ subset. These inflammatory or classical monocytes have a relatively short circulating life span and accumulate preferentially in inflammatory sites where they give rise to macrophages. The remaining Ly-6Clow CCR2low CX3CR1high CD62L− subset, referred to as nonclassical, patrols the vasculature and accumulates at low numbers in the steady state[138,139]. In the steady state, monocyte conversion from Ly-6Chigh to Ly-6Clow may occur in blood and bone marrow[24].
3.2.2. Post Infarct
Soon after coronary artery ligation[123] or after angiotensin II (AngII) infusion[21], Ly-6Chigh monocytes infiltrate the heart in large numbers [The AngII signaling cascade is induced in virtually all forms of cardiovascular disease[140] and has been shown to mobilize splenic monocytes to the infarcted myocardium[128]]. Many of these monocytes may not differentiate to macrophages but either exit or die in the tissue. Those that differentiate acquire M1-like properties, continue to express Ly-6C, CCR2, MHC-IIhigh and contribute to inflammation[21,130]. Over time, as inflammation gives way to resolution, a second Ly-6Clow/int population emerges. Whether macrophages dominating this regenerative wound-healing phase arise via the differentiation of Ly-6Chigh monocytes and less via differentiation of Ly-6Clow monocytes or local M1 to M2 macrophage conversion needs to be elucidated. As inflammation completely subsides, a population of F4/80high macrophages resembling steady state macrophages returns. Studies by Epelman et al.[21] suggest that despite the autonomy between resident and monocyte-derived macrophages, after depletion and in a setting of competitive resident macrophage proliferation, blood monocyte-derived macrophages have the ability to take up residence within tissue and become the dominant macrophage population. After repopulation is complete, tissue macrophage autonomy is restored, albeit with a large complement of adult monocyte-derived macrophages as the new resident macrophage population.
Studies in ischemic myocardium indicated that recruitment, rather than local proliferation, is the primary mechanism regulating monocyte and macrophage numbers[130]. Within the first 24hrs after coronary artery ligation in mice, approximately half of all monocytes recruited to the heart are derived from the spleen[128]. These monocytes reside in the subcapsular red pulp of the spleen and resemble their circulating blood counterparts. The bone marrow and blood also substantially contribute to the monocytes recruited to the infarct. Because the residence time of monocytes and macrophages is short, and because large numbers of cells are continuously recruited to the heart post infarction, the splenic monocyte pool is exhausted in a short time. However, upon myocardial infarction the bone marrow outsources the production of monocytes to extramedullary sites through the export and relocation of hematopoietic stem cells and progenitor cells to the spleen[141,142]. In mice, the splenic reservoir of monocytes is replenished by day 6 after coronary artery ligation and continues to supply the infarct with a significant number of cells[130].
The findings of Epelman[21] in which various macrophage subsets may have different functional roles might explain why, in models of cardiac injury, blocking monocyte influx is protective, whereas broad macrophage depletion strategies that also target resident cardiac macrophages abolish protection[132,143]. These data suggest that preserving resident cardiac macrophage expansion via proliferation, while targeting peripheral monocyte recruitment, might lead to improved myocardial recovery after injury. Moreover, the data underscore the importance in dissecting the roles of various monocyte/macrophage populations during the course of an ischemic response. Given these findings in young mice and the known findings of prolonged inflammation and delayed recovery in the aged, raises many questions. How does “inflamm-aging” affect resident macrophage populations and the Ly-6Chigh monocyte population from the spleen, blood and bone marrow? Is there skewing in the representation of the various monocyte/macrophage populations with age? Do cells in each of the subsets respond as well as their young counterpart, and to what extent are differences driven by the aged microenvironment or by intrinsic changes (e.g., oxidative stress)? Can we restore responsiveness in the aged by targeting specific monocyte/macrophage populations?
3.3. Clinical Relevance
A number of clinical studies support concepts of cardiac monocyte and macrophage heterogeneity and dynamics that have been recently described in mice. A bi-phasic monocyte response has been observed in the blood of patients after myocardial infarction[144]. A number of clinical studies confirm the correlation of blood leukocyte levels in heart failure with prognosis[145,146]. Other observations in patients after myocardial infarction include an increased level of hematopoietic progenitors in the blood[147], increased metabolic activity of the bone marrow[141], and evidence for an increased progenitor activity in the spleen[142]. Clinical signs of inflammation in heart failure patients support an active role of macrophages in heart disease.
Still lacking is a clear understanding of the potentially dynamic changes in monocyte/macrophage heterogeneity that occur over a lifespan, clarification of alterations in functional monocyte/macrophage responses due to microenvironmental vs. intrinsic influences, and a determination of these changes in steady state vs. post infarction. Understanding these changes in the aged will aid in the design of efficacious interventions that may ameliorate maladaptive remodeling and improve outcome post myocardial infarction. This becomes especially significant given that the majority of cardiovascular deaths occur in the elderly despite improved therapies[148]. Although several factors may contribute to this, it is clear that more studies are needed using aged animals.
4.Conclusion/Overview
Phagocytic leukocytes, macrophages and DCs, are amongst the initial responders to tissue injury and infection. Their response may include the synthesis of microbicidal mediators, chemokines and cytokines, up-regulation of the expression of cell surface activation markers and mobilization. Unlike the lung where sufficient cell numbers are present to confront pathogens, the number of phagocytic leukocytes present at steady state in the heart is low and upon injury, a large number of monocytes/macrophages are mobilized to the infarcted area. In both models, an efficacious outcome requires initiation of an adequate inflammatory response with a timely resolution. Both macrophages and DCs are diverse populations and the macrophage lineage in particular displays extreme functional and phenotypic heterogeneity. Distinct subpopulations can be found in circulation and resident in various tissues. Tissue distribution of these subpopulations frequently changes over the course of response to injury or pathogenic challenge, in answer to microenvironmental cues.
Advancing age erodes the capacity for appropriate disease and injury management and is characterized by an increased level of chronic inflammation, but diminished and protracted acute inflammatory responsiveness upon challenge. Although several factors may contribute to inflamm-aging and immunosenescence, it seems increasingly likely that the development of the senescence associated secretory phenotype creates a unique “senescent” tissue microenvironment, dramatically altering macrophage and DC function. The chronic presence of pro-inflammatory mediators alters the molecular processes and signal transduction pathways triggered in response to tissue damage or infectious challenge. An increased understanding of these changes will provide unique opportunities for therapeutic intervention to restore appropriate innate immune activity in the elderly.
Highlights.
Age erodes the capacity for appropriate disease and injury management
Age is associated with elevated chronic inflammation and diminished acute inflammatory response
Distribution of populations change over the course of response in answer to microenvironment cues
Inflamm-aging alters molecular processes triggered in response to tissue damage or infection
ACKNOWLEDGEMENTS
This work was supported by grants HL112725 and AG026727 (PJL). The funding sources had no involvement in the study design, collection, analysis and interpretation of data, the writing of the report or in the decision to submit the article for publication.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Abbreviations: angiotensin II – AngII; conventional dendritic cells – cDC; damage associated molecular protein – DAMP; Found In Inflammatory Zone 1 – FIZZ1; glycogen synthase kinase-3 – GSK-3; inducible nitric oxide synthase – iNOS; microRNA – miRNA; pathogen associated molecular protein – PAMP; reactive oxygen species – ROS; senescence-associated secretory phenotype – SASP
The authors have no other conflicting financial interest.
References
- 1.Franceschi C, Bonafe M, Valensin S, Olivieri F, De Luca M, Ottaviani E, De Benedictis G. Inflamm-aging. An evolutionary perspective on immunosenescence. Annals of the New York Academy of Sciences. 2000;908:244–254. doi: 10.1111/j.1749-6632.2000.tb06651.x. [DOI] [PubMed] [Google Scholar]
- 2.Adamson R. Role of macrophages in normal wound healing: an overview. Journal of wound care. 2009;18:349–351. doi: 10.12968/jowc.2009.18.8.43636. [DOI] [PubMed] [Google Scholar]
- 3.Gordon S. The macrophage: past, present and future. European journal of immunology. 2007;37(Suppl 1):S9–17. doi: 10.1002/eji.200737638. [DOI] [PubMed] [Google Scholar]
- 4.Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A, Locati M. The chemokine system in diverse forms of macrophage activation and polarization. Trends in immunology. 2004;25:677–686. doi: 10.1016/j.it.2004.09.015. [DOI] [PubMed] [Google Scholar]
- 5.Gordon S. Alternative activation of macrophages. Nature reviews. 2003;3:23–35. doi: 10.1038/nri978. [DOI] [PubMed] [Google Scholar]
- 6.Stout RD, Jiang C, Matta B, Tietzel I, Watkins SK, Suttles J. Macrophages sequentially change their functional phenotype in response to changes in microenvironmental influences. J Immunol. 2005;175:342–349. doi: 10.4049/jimmunol.175.1.342. [DOI] [PubMed] [Google Scholar]
- 7.Murray PJ, Wynn TA. Obstacles and opportunities for understanding macrophage polarization. Journal of leukocyte biology. 2011;89:557–563. doi: 10.1189/jlb.0710409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Nair MG, Du Y, Perrigoue JG, Zaph C, Taylor JJ, Goldschmidt M, Swain GP, Yancopoulos GD, Valenzuela DM, Murphy A, Karow M, Stevens S, Pearce EJ, Artis D. Alternatively activated macrophage-derived RELM-{alpha} is a negative regulator of type 2 inflammation in the lung. The Journal of experimental medicine. 2009;206:937–952. doi: 10.1084/jem.20082048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Mantovani A, Sozzani S, Locati M, Allavena P, Sica A. Macrophage polarization: tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends in immunology. 2002;23:549–555. doi: 10.1016/s1471-4906(02)02302-5. [DOI] [PubMed] [Google Scholar]
- 10.Dace DS, Apte RS. Effect of senescence on macrophage polarization and angiogenesis. Rejuvenation research. 2008;11:177–185. doi: 10.1089/rej.2007.0614. [DOI] [PubMed] [Google Scholar]
- 11.Jackaman C, Radley-Crabb HG, Soffe Z, Shavlakadze T, Grounds MD, Nelson DJ. Targeting macrophages rescues age-related immune deficiencies in C57BL/6J geriatric mice. Aging cell. 2013;12:345–357. doi: 10.1111/acel.12062. [DOI] [PubMed] [Google Scholar]
- 12.Mahbub S, Deburghgraeve CR, Kovacs EJ. Advanced age impairs macrophage polarization. J Interferon Cytokine Res. 2012;32:18–26. doi: 10.1089/jir.2011.0058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Pelegrin P, Surprenant A. Dynamics of macrophage polarization reveal new mechanism to inhibit IL-1beta release through pyrophosphates. The EMBO journal. 2009;28:2114–2127. doi: 10.1038/emboj.2009.163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Bruunsgaard H, Andersen-Ranberg K, Hjelmborg J, Pedersen BK, Jeune B. Elevated levels of tumor necrosis factor alpha and mortality in centenarians. The American journal of medicine. 2003;115:278–283. doi: 10.1016/s0002-9343(03)00329-2. [DOI] [PubMed] [Google Scholar]
- 15.Bruunsgaard H, Pedersen BK. Age-related inflammatory cytokines and disease. Immunology and allergy clinics of North America. 2003;23:15–39. doi: 10.1016/s0889-8561(02)00056-5. [DOI] [PubMed] [Google Scholar]
- 16.Chen W, Frangogiannis NG. The role of inflammatory and fibrogenic pathways in heart failure associated with aging. Heart failure reviews. 2011;15:415–422. doi: 10.1007/s10741-010-9161-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Gabay C, Lamacchia C, Palmer G. IL-1 pathways in inflammation and human diseases. Nature reviews. 2010;6:232–241. doi: 10.1038/nrrheum.2010.4. [DOI] [PubMed] [Google Scholar]
- 18.Gomez CR, Karavitis J, Palmer JL, Faunce DE, Ramirez L, Nomellini V, Kovacs EJ. Interleukin-6 contributes to age-related alteration of cytokine production by macrophages. Mediators of inflammation. 2010;2010:475139. doi: 10.1155/2010/475139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Nomellini V, Gomez CR, Kovacs EJ. Aging and impairment of innate immunity. Contributions to microbiology. 2008;15:188–205. doi: 10.1159/000136358. [DOI] [PubMed] [Google Scholar]
- 20.Fogg DK, Sibon C, Miled C, Jung S, Aucouturier P, Littman DR, Cumano A, Geissmann F. A clonogenic bone marrow progenitor specific for macrophages and dendritic cells. Science (New York, NY. 2006;311:83–87. doi: 10.1126/science.1117729. [DOI] [PubMed] [Google Scholar]
- 21.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]
- 22.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 (New York, NY. 2010;330:841–845. doi: 10.1126/science.1194637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Schulz C, Gomez Perdiguero E, Chorro L, Szabo-Rogers H, Cagnard N, Kierdorf K, Prinz M, Wu B, Jacobsen SE, Pollard JW, Frampton J, Liu KJ, Geissmann F. A lineage of myeloid cells independent of Myb and hematopoietic stem cells. Science (New York, NY. 2012;336:86–90. doi: 10.1126/science.1219179. [DOI] [PubMed] [Google Scholar]
- 24.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]
- 25.Kadl A, Meher AK, Sharma PR, Lee MY, Doran AC, Johnstone SR, Elliott MR, Gruber F, Han J, Chen W, Kensler T, Ravichandran KS, Isakson BE, Wamhoff BR, Leitinger N. Identification of a novel macrophage phenotype that develops in response to atherogenic phospholipids via Nrf2. Circulation research. 2010;107:737–746. doi: 10.1161/CIRCRESAHA.109.215715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Sica A, Mantovani A. Macrophage plasticity and polarization: in vivo veritas. The Journal of clinical investigation. 2012;122:787–795. doi: 10.1172/JCI59643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Auffray C, Sieweke MH, Geissmann F. Blood monocytes: development, heterogeneity, and relationship with dendritic cells. Annual review of immunology. 2009;27:669–692. doi: 10.1146/annurev.immunol.021908.132557. [DOI] [PubMed] [Google Scholar]
- 28.Landsman L, Jung S. Lung macrophages serve as obligatory intermediate between blood monocytes and alveolar macrophages. J Immunol. 2007;179:3488–3494. doi: 10.4049/jimmunol.179.6.3488. [DOI] [PubMed] [Google Scholar]
- 29.Lambrecht BN, Hammad H. Biology of lung dendritic cells at the origin of asthma. Immunity. 2009;31:412–424. doi: 10.1016/j.immuni.2009.08.008. [DOI] [PubMed] [Google Scholar]
- 30.Hashimoto D, Miller J, Merad M. Dendritic cell and macrophage heterogeneity in vivo. Immunity. 2011;35:323–335. doi: 10.1016/j.immuni.2011.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Guth AM, Janssen WJ, Bosio CM, Crouch EC, Henson PM, Dow SW. Lung environment determines unique phenotype of alveolar macrophages. American journal of physiology. 2009;296:L936–946. doi: 10.1152/ajplung.90625.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Misharin AV, Morales-Nebreda L, Mutlu GM, Budinger GR, Perlman H. Flow cytometric analysis of macrophages and dendritic cell subsets in the mouse lung. American journal of respiratory cell and molecular biology. 2013;49:503–510. doi: 10.1165/rcmb.2013-0086MA. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Balhara J, Gounni AS. The alveolar macrophages in asthma: a double-edged sword. Mucosal immunology. 2012;5:605–609. doi: 10.1038/mi.2012.74. [DOI] [PubMed] [Google Scholar]
- 34.Beck-Schimmer B, Schwendener R, Pasch T, Reyes L, Booy C, Schimmer RC. Alveolar macrophages regulate neutrophil recruitment in endotoxin-induced lung injury. Respiratory research. 2005;6:61. doi: 10.1186/1465-9921-6-61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Lee JS. Heterogeneity of lung mononuclear phagocytes in chronic obstructive pulmonary disease. Journal of innate immunity. 2012;4:489–497. doi: 10.1159/000337434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Chen BD, Mueller M, Chou TH. Role of granulocyte/macrophage colony-stimulating factor in the regulation of murine alveolar macrophage proliferation and differentiation. J Immunol. 1988;141:139–144. [PubMed] [Google Scholar]
- 37.Janssen WJ, Barthel L, Muldrow A, Oberley-Deegan RE, Kearns MT, Jakubzick C, Henson PM. Fas determines differential fates of resident and recruited macrophages during resolution of acute lung injury. American journal of respiratory and critical care medicine. 2011;184:547–560. doi: 10.1164/rccm.201011-1891OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Blusse van Oud Alblas A, Mattie H, van Furth R. A quantitative evaluation of pulmonary macrophage kinetics. Cell and tissue kinetics. 1983;16:211–219. [PubMed] [Google Scholar]
- 39.Maus UA, Janzen S, Wall G, Srivastava M, Blackwell TS, Christman JW, Seeger W, Welte T, Lohmeyer J. Resident alveolar macrophages are replaced by recruited monocytes in response to endotoxin-induced lung inflammation. American journal of respiratory cell and molecular biology. 2006;35:227–235. doi: 10.1165/rcmb.2005-0241OC. [DOI] [PubMed] [Google Scholar]
- 40.Landsman L, Varol C, Jung S. Distinct differentiation potential of blood monocyte subsets in the lung. J Immunol. 2007;178:2000–2007. doi: 10.4049/jimmunol.178.4.2000. [DOI] [PubMed] [Google Scholar]
- 41.Dhaliwal K, Scholefield E, Ferenbach D, Gibbons M, Duffin R, Dorward DA, Morris AC, Humphries D, MacKinnon A, Wilkinson TS, Wallace WA, van Rooijen N, Mack M, Rossi AG, Davidson DJ, Hirani N, Hughes J, Haslett C, Simpson AJ. Monocytes control second-phase neutrophil emigration in established lipopolysaccharide-induced murine lung injury. American journal of respiratory and critical care medicine. 2012;186:514–524. doi: 10.1164/rccm.201112-2132OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Maus UA, Waelsch K, Kuziel WA, Delbeck T, Mack M, Blackwell TS, Christman JW, Schlondorff D, Seeger W, Lohmeyer J. Monocytes are potent facilitators of alveolar neutrophil emigration during lung inflammation: role of the CCL2-CCR2 axis. J Immunol. 2003;170:3273–3278. doi: 10.4049/jimmunol.170.6.3273. [DOI] [PubMed] [Google Scholar]
- 43.Guilliams M, Lambrecht BN, Hammad H. Division of labor between lung dendritic cells and macrophages in the defense against pulmonary infections. Mucosal immunology. 2013;6:464–473. doi: 10.1038/mi.2013.14. [DOI] [PubMed] [Google Scholar]
- 44.Lin KL, Suzuki Y, Nakano H, Ramsburg E, Gunn MD. CCR2+ monocyte-derived dendritic cells and exudate macrophages produce influenza-induced pulmonary immune pathology and mortality. J Immunol. 2008;180:2562–2572. doi: 10.4049/jimmunol.180.4.2562. [DOI] [PubMed] [Google Scholar]
- 45.Grayson MH. Lung dendritic cells and the inflammatory response. Ann Allergy Asthma Immunol. 2006;96:643–651. doi: 10.1016/S1081-1206(10)61061-7. quiz 652-643, 678. [DOI] [PubMed] [Google Scholar]
- 46.Bedoret D, Wallemacq H, Marichal T, Desmet C, Quesada Calvo F, Henry E, Closset R, Dewals B, Thielen C, Gustin P, de Leval L, Van Rooijen N, Le Moine A, Vanderplasschen A, Cataldo D, Drion PV, Moser M, Lekeux P, Bureau F. Lung interstitial macrophages alter dendritic cell functions to prevent airway allergy in mice. The Journal of clinical investigation. 2009;119:3723–3738. doi: 10.1172/JCI39717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.GeurtsvanKessel CH, Willart MA, van Rijt LS, Muskens F, Kool M, Baas C, Thielemans K, Bennett C, Clausen BE, Hoogsteden HC, Osterhaus AD, Rimmelzwaan GF, Lambrecht BN. Clearance of influenza virus from the lung depends on migratory langerin+CD11b- but not plasmacytoid dendritic cells. The Journal of experimental medicine. 2008;205:1621–1634. doi: 10.1084/jem.20071365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Kim TS, Braciale TJ. Respiratory dendritic cell subsets differ in their capacity to support the induction of virus-specific cytotoxic CD8+ T cell responses. PloS one. 2009;4:e4204. doi: 10.1371/journal.pone.0004204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Sung SS, Fu SM, Rose CE, Jr., Gaskin F, Ju ST, Beaty SR. A major lung CD103 (alphaE)-beta7 integrin-positive epithelial dendritic cell population expressing Langerin and tight junction proteins. J Immunol. 2006;176:2161–2172. doi: 10.4049/jimmunol.176.4.2161. [DOI] [PubMed] [Google Scholar]
- 50.Kawai T, Akira S. TLR signaling. Cell death and differentiation. 2006;13:816–825. doi: 10.1038/sj.cdd.4401850. [DOI] [PubMed] [Google Scholar]
- 51.Kong KF, Delroux K, Wang X, Qian F, Arjona A, Malawista SE, Fikrig E, Montgomery RR. Dysregulation of TLR3 impairs the innate immune response to West Nile virus in the elderly. Journal of virology. 2008;82:7613–7623. doi: 10.1128/JVI.00618-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Takeuchi O, Hoshino K, Akira S. Cutting edge: TLR2-deficient and MyD88-deficient mice are highly susceptible to Staphylococcus aureus infection. J Immunol. 2000;165:5392–5396. doi: 10.4049/jimmunol.165.10.5392. [DOI] [PubMed] [Google Scholar]
- 53.Agrawal A, Gupta S. Impact of aging on dendritic cell functions in humans. Ageing research reviews. 2011;10:336–345. doi: 10.1016/j.arr.2010.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Ciaramella A, Spalletta G, Bizzoni F, Salani F, Caltagirone C, Bossu P. Effect of age on surface molecules and cytokine expression in human dendritic cells. Cellular immunology. 2011;269:82–89. doi: 10.1016/j.cellimm.2011.04.010. [DOI] [PubMed] [Google Scholar]
- 55.Panda A, Arjona A, Sapey E, Bai F, Fikrig E, Montgomery RR, Lord JM, Shaw AC. Human innate immunosenescence: causes and consequences for immunity in old age. Trends in immunology. 2009;30:325–333. doi: 10.1016/j.it.2009.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Wong C, Goldstein DR. Impact of aging on antigen presentation cell function of dendritic cells. Current opinion in immunology. 2013;25:535–541. doi: 10.1016/j.coi.2013.05.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Murciano C, Yanez A, O'Connor JE, Gozalbo D, Gil ML. Influence of aging on murine neutrophil and macrophage function against Candida albicans. FEMS immunology and medical microbiology. 2008;53:214–221. doi: 10.1111/j.1574-695X.2008.00418.x. [DOI] [PubMed] [Google Scholar]
- 58.Renshaw M, Rockwell J, Engleman C, Gewirtz A, Katz J, Sambhara S. Cutting edge: impaired Toll-like receptor expression and function in aging. J Immunol. 2002;169:4697–4701. doi: 10.4049/jimmunol.169.9.4697. [DOI] [PubMed] [Google Scholar]
- 59.Boehmer ED, Goral J, Faunce DE, Kovacs EJ. Age-dependent decrease in Toll-like receptor 4-mediated proinflammatory cytokine production and mitogen-activated protein kinase expression. Journal of leukocyte biology. 2004;75:342–349. doi: 10.1189/jlb.0803389. [DOI] [PubMed] [Google Scholar]
- 60.Boehmer ED, Meehan MJ, Cutro BT, Kovacs EJ. Aging negatively skews macrophage TLR2- and TLR4-mediated pro-inflammatory responses without affecting the IL-2-stimulated pathway. Mechanisms of ageing and development. 2005;126:1305–1313. doi: 10.1016/j.mad.2005.07.009. [DOI] [PubMed] [Google Scholar]
- 61.Liang S, Domon H, Hosur KB, Wang M, Hajishengallis G. Age-related alterations in innate immune receptor expression and ability of macrophages to respond to pathogen challenge in vitro. Mechanisms of ageing and development. 2009;130:538–546. doi: 10.1016/j.mad.2009.06.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Agrawal S, Gollapudi S, Gupta S, Agrawal A. Dendritic cells from the elderly display an intrinsic defect in the production of IL-10 in response to lithium chloride. Experimental gerontology. 2013;48:1285–1292. doi: 10.1016/j.exger.2013.08.006. [DOI] [PubMed] [Google Scholar]
- 63.Lowery EM, Brubaker AL, Kuhlmann E, Kovacs EJ. The aging lung. Clinical interventions in aging. 2013;8:1489–1496. doi: 10.2147/CIA.S51152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Panda A, Qian F, Mohanty S, van Duin D, Newman FK, Zhang L, Chen S, Towle V, Belshe RB, Fikrig E, Allore HG, Montgomery RR, Shaw AC. Age-associated decrease in TLR function in primary human dendritic cells predicts influenza vaccine response. J Immunol. 2010;184:2518–2527. doi: 10.4049/jimmunol.0901022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Paula C, Motta A, Schmitz C, Nunes CP, Souza AP, Bonorino C. Alterations in dendritic cell function in aged mice: potential implications for immunotherapy design. Biogerontology. 2009;10:13–25. doi: 10.1007/s10522-008-9150-x. [DOI] [PubMed] [Google Scholar]
- 66.Li Z, Li J, Bu X, Liu X, Tankersley CG, Wang C, Huang K. Age-induced augmentation of p38 MAPK phosphorylation in mouse lung. Experimental gerontology. 2011;46:694–702. doi: 10.1016/j.exger.2011.04.005. [DOI] [PubMed] [Google Scholar]
- 67.Meyer KC, Rosenthal NS, Soergel P, Peterson K. Neutrophils and low-grade inflammation in the seemingly normal aging human lung. Mechanisms of ageing and development. 1998;104:169–181. doi: 10.1016/s0047-6374(98)00065-7. [DOI] [PubMed] [Google Scholar]
- 68.Aoshiba K, Nagai A. Chronic lung inflammation in aging mice. FEBS letters. 2007;581:3512–3516. doi: 10.1016/j.febslet.2007.06.075. [DOI] [PubMed] [Google Scholar]
- 69.Shaw AC, Panda A, Joshi SR, Qian F, Allore HG, Montgomery RR. Dysregulation of human Toll-like receptor function in aging. Ageing research reviews. 2011;10:346–353. doi: 10.1016/j.arr.2010.10.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Boyd AR, Shivshankar P, Jiang S, Berton MT, Orihuela CJ. Age-related defects in TLR2 signaling diminish the cytokine response by alveolar macrophages during murine pneumococcal pneumonia. Experimental gerontology. 2012;47:507–518. doi: 10.1016/j.exger.2012.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Fallah MP, Chelvarajan RL, Garvy BA, Bondada S. Role of phosphoinositide 3-kinase-Akt signaling pathway in the age-related cytokine dysregulation in splenic macrophages stimulated via TLR-2 or TLR-4 receptors. Mechanisms of ageing and development. 2011;132:274–286. doi: 10.1016/j.mad.2011.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Hajishengallis G. Too old to fight? Aging and its toll on innate immunity. Molecular oral microbiology. 2010;25:25–37. doi: 10.1111/j.2041-1014.2009.00562.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Mares CA, Ojeda SS, Li Q, Morris EG, Coalson JJ, Teale JM. Aged mice display an altered pulmonary host response to Francisella tularensis live vaccine strain (LVS) infections. Experimental gerontology. 2010;45:91–96. doi: 10.1016/j.exger.2009.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Qian F, Wang X, Zhang L, Lin A, Zhao H, Fikrig E, Montgomery RR. Impaired interferon signaling in dendritic cells from older donors infected in vitro with West Nile virus. The Journal of infectious diseases. 2011;203:1415–1424. doi: 10.1093/infdis/jir048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Toapanta FR, Ross TM. Impaired immune responses in the lungs of aged mice following influenza infection. Respiratory research. 2009;10:112. doi: 10.1186/1465-9921-10-112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Wen J, Li CM, Gu L, Yin SJ, Li W, Yang R. Aging Reduces the Expression of Lung CINC and MCP-1 mRNA in a P. aeruginosa Rat Model of Infection. Inflammation. 2014 doi: 10.1007/s10753-014-9813-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Wong CP, Magnusson KR, Ho E. Aging is associated with altered dendritic cells subset distribution and impaired proinflammatory cytokine production. Experimental gerontology. 2010;45:163–169. doi: 10.1016/j.exger.2009.11.005. [DOI] [PubMed] [Google Scholar]
- 78.Prakash S, Agrawal S, Cao JN, Gupta S, Agrawal A. Impaired secretion of interferons by dendritic cells from aged subjects to influenza : role of histone modifications. Age (Dordrecht, Netherlands) 2013;35:1785–1797. doi: 10.1007/s11357-012-9477-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Stout-Delgado HW, Vaughan SE, Shirali AC, Jaramillo RJ, Harrod KS. Impaired NLRP3 inflammasome function in elderly mice during influenza infection is rescued by treatment with nigericin. J Immunol. 2012;188:2815–2824. doi: 10.4049/jimmunol.1103051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Beurel E, Michalek SM, Jope RS. Innate and adaptive immune responses regulated by glycogen synthase kinase-3 (GSK3). Trends in immunology. 2009;31:24–31. doi: 10.1016/j.it.2009.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Lin YC, Kuo HC, Wang JS, Lin WW. Regulation of inflammatory response by 3-methyladenine involves the coordinative actions on Akt and glycogen synthase kinase 3beta rather than autophagy. J Immunol. 2012;189:4154–4164. doi: 10.4049/jimmunol.1102739. [DOI] [PubMed] [Google Scholar]
- 82.Wang H, Brown J, Martin M. Glycogen synthase kinase 3: a point of convergence for the host inflammatory response. Cytokine. 2011;53:130–140. doi: 10.1016/j.cyto.2010.10.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Wang H, Kumar A, Lamont RJ, Scott DA. GSK3beta and the control of infectious bacterial diseases. Trends in microbiology. 2014 doi: 10.1016/j.tim.2014.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Zhou J, Force T. Focusing the spotlight on GSK-3 in aging. Aging. 2013;5:388–389. doi: 10.18632/aging.100568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Zhou J, Freeman TA, Ahmad F, Shang X, Mangano E, Gao E, Farber J, Wang Y, Ma XL, Woodgett J, Vagnozzi RJ, Lal H, Force T. GSK-3alpha is a central regulator of age-related pathologies in mice. The Journal of clinical investigation. 2013;123:1821–1832. doi: 10.1172/JCI64398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Jope RS, Johnson GV. The glamour and gloom of glycogen synthase kinase-3. Trends in biochemical sciences. 2004;29:95–102. doi: 10.1016/j.tibs.2003.12.004. [DOI] [PubMed] [Google Scholar]
- 87.Jope RS, Yuskaitis CJ, Beurel E. Glycogen synthase kinase-3 (GSK3): inflammation, diseases, and therapeutics. Neurochemical research. 2007;32:577–595. doi: 10.1007/s11064-006-9128-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Patel S, Woodgett J. Glycogen synthase kinase-3 and cancer: good cop, bad cop? Cancer cell. 2008;14:351–353. doi: 10.1016/j.ccr.2008.10.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Kim YM, Song I, Seo YH, Yoon G. Glycogen Synthase Kinase 3 Inactivation Induces Cell Senescence through Sterol Regulatory Element Binding Protein 1-Mediated Lipogenesis in Chang Cells. Endocrinology and metabolism (Seoul, Korea) 2013;28:297–308. doi: 10.3803/EnM.2013.28.4.297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Okkenhaug K, Vanhaesebroeck B. PI3K in lymphocyte development, differentiation and activation. Nature reviews. 2003;3:317–330. doi: 10.1038/nri1056. [DOI] [PubMed] [Google Scholar]
- 91.Nandan D, Camargo de Oliveira C, Moeenrezakhanlou A, Lopez M, Silverman JM, Subek J, Reiner NE. Myeloid cell IL-10 production in response to leishmania involves inactivation of glycogen synthase kinase-3beta downstream of phosphatidylinositol-3 kinase. J Immunol. 2012;188:367–378. doi: 10.4049/jimmunol.1100076. [DOI] [PubMed] [Google Scholar]
- 92.Xu YP, Qi RQ, Chen W, Shi Y, Cui ZZ, Gao XH, Chen HD, Zhou L, Mi QS. Aging affects epidermal Langerhans cell development and function and alters their miRNA gene expression profile. Aging. 2012;4:742–754. doi: 10.18632/aging.100501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Williams AE, Perry MM, Moschos SA, Lindsay MA. microRNA expression in the aging mouse lung. BMC genomics. 2007;8:172. doi: 10.1186/1471-2164-8-172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Jiang M, Xiang Y, Wang D, Gao J, Liu D, Liu Y, Liu S, Zheng D. Dysregulated expression of miR-146a contributes to age-related dysfunction of macrophages. Aging cell. 2012;11:29–40. doi: 10.1111/j.1474-9726.2011.00757.x. [DOI] [PubMed] [Google Scholar]
- 95.Barja G, Herrero A. Oxidative damage to mitochondrial DNA is inversely related to maximum life span in the heart and brain of mammals. Faseb J. 2000;14:312–318. doi: 10.1096/fasebj.14.2.312. [DOI] [PubMed] [Google Scholar]
- 96.Dufour E, Boulay J, Rincheval V, Sainsard-Chanet A. A causal link between respiration and senescence in Podospora anserina. Proceedings of the National Academy of Sciences of the United States of America. 2000;97:4138–4143. doi: 10.1073/pnas.070501997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Cathcart MK. Regulation of superoxide anion production by NADPH oxidase in monocytes/macrophages: contributions to atherosclerosis. Arteriosclerosis, thrombosis, and vascular biology. 2004;24:23–28. doi: 10.1161/01.ATV.0000097769.47306.12. [DOI] [PubMed] [Google Scholar]
- 98.Park JB. Phagocytosis induces superoxide formation and apoptosis in macrophages. Experimental & molecular medicine. 2003;35:325–335. doi: 10.1038/emm.2003.44. [DOI] [PubMed] [Google Scholar]
- 99.Alexeyev MF. Is there more to aging than mitochondrial DNA and reactive oxygen species? The FEBS journal. 2009;276:5768–5787. doi: 10.1111/j.1742-4658.2009.07269.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Broadley SA, Hartl FU. Mitochondrial stress signaling: a pathway unfolds. Trends in cell biology. 2008;18:1–4. doi: 10.1016/j.tcb.2007.11.003. [DOI] [PubMed] [Google Scholar]
- 101.Durieux J, Wolff S, Dillin A. The cell-non-autonomous nature of electron transport chain-mediated longevity. Cell. 2011;144:79–91. doi: 10.1016/j.cell.2010.12.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Halliwell B. The wanderings of a free radical. Free radical biology & medicine. 2009;46:531–542. doi: 10.1016/j.freeradbiomed.2008.11.008. [DOI] [PubMed] [Google Scholar]
- 103.Hamanaka RB, Chandel NS. Mitochondrial reactive oxygen species regulate cellular signaling and dictate biological outcomes. Trends in biochemical sciences. 2010;35:505–513. doi: 10.1016/j.tibs.2010.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Roberts CK, Sindhu KK. Oxidative stress and metabolic syndrome. Life sciences. 2009;84:705–712. doi: 10.1016/j.lfs.2009.02.026. [DOI] [PubMed] [Google Scholar]
- 105.Wellen KE, Thompson CB. Cellular metabolic stress: considering how cells respond to nutrient excess. Molecular cell. 2010;40:323–332. doi: 10.1016/j.molcel.2010.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Cuervo AM. Chaperone-mediated autophagy: selectivity pays off. Trends in endocrinology and metabolism: TEM. 2010;21:142–150. doi: 10.1016/j.tem.2009.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Dunlop RA, Brunk UT, Rodgers KJ. Oxidized proteins: mechanisms of removal and consequences of accumulation. IUBMB life. 2009;61:522–527. doi: 10.1002/iub.189. [DOI] [PubMed] [Google Scholar]
- 108.Tyedmers J, Mogk A, Bukau B. Cellular strategies for controlling protein aggregation. Nat Rev Mol Cell Biol. 2010;11:777–788. doi: 10.1038/nrm2993. [DOI] [PubMed] [Google Scholar]
- 109.de la Fuente M, Hernanz A, Guayerbas N, Alvarez P, Alvarado C. Changes with age in peritoneal macrophage functions. Implication of leukocytes in the oxidative stress of senescence. Cellular and molecular biology (Noisy-le-Grand, France) 2004:50. Online Pub:OL683-690. [PubMed] [Google Scholar]
- 110.Fujimoto H, Kobayashi H, Ohno M. Age-induced reduction in mitochondrial manganese superoxide dismutase activity and tolerance of macrophages against apoptosis induced by oxidized low density lipoprotein. Circ J. 2010;74:353–360. doi: 10.1253/circj.cj-09-0491. [DOI] [PubMed] [Google Scholar]
- 111.Ponnappan S, Ovaa H, Ponnappan U. Lower expression of catalytic and structural subunits of the proteasome contributes to decreased proteolysis in peripheral blood T lymphocytes during aging. The international journal of biochemistry & cell biology. 2007;39:799–809. doi: 10.1016/j.biocel.2007.01.002. [DOI] [PubMed] [Google Scholar]
- 112.Cannizzo ES, Clement CC, Morozova K, Valdor R, Kaushik S, Almeida LN, Follo C, Sahu R, Cuervo AM, Macian F, Santambrogio L. Age-related oxidative stress compromises endosomal proteostasis. Cell reports. 2012;2:136–149. doi: 10.1016/j.celrep.2012.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Freund A, Orjalo AV, Desprez PY, Campisi J. Inflammatory networks during cellular senescence: causes and consequences. Trends in molecular medicine. 2010;16:238–246. doi: 10.1016/j.molmed.2010.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Salminen A, Kauppinen A, Kaarniranta K. Emerging role of NF-kappaB signaling in the induction of senescence-associated secretory phenotype (SASP). Cellular signalling. 2012;24:835–845. doi: 10.1016/j.cellsig.2011.12.006. [DOI] [PubMed] [Google Scholar]
- 115.Sikora E, Arendt T, Bennett M, Narita M. Impact of cellular senescence signature on ageing research. Ageing research reviews. 2011;10:146–152. doi: 10.1016/j.arr.2010.10.002. [DOI] [PubMed] [Google Scholar]
- 116.Chilosi M, Carloni A, Rossi A, Poletti V. Premature lung aging and cellular senescence in the pathogenesis of idiopathic pulmonary fibrosis and COPD/emphysema. Transl Res. 162:156–173. doi: 10.1016/j.trsl.2013.06.004. [DOI] [PubMed] [Google Scholar]
- 117.Shivshankar P, Boyd AR, Le Saux CJ, Yeh IT, Orihuela CJ. Cellular senescence increases expression of bacterial ligands in the lungs and is positively correlated with increased susceptibility to pneumococcal pneumonia. Aging cell. 2011;10:798–806. doi: 10.1111/j.1474-9726.2011.00720.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Boyd AR, Orihuela CJ. Dysregulated inflammation as a risk factor for pneumonia in the elderly. Aging and disease. 2012;2:487–500. [PMC free article] [PubMed] [Google Scholar]
- 119.Hinojosa CA, Akula Suresh Babu R, Rahman MM, Fernandes G, Boyd AR, Orihuela CJ. Elevated A20 contributes to age-dependent macrophage dysfunction in the lungs. Experimental gerontology. 2014 doi: 10.1016/j.exger.2014.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Hinojosa E, Boyd AR, Orihuela CJ. Age-associated inflammation and toll-like receptor dysfunction prime the lungs for pneumococcal pneumonia. The Journal of infectious diseases. 2009;200:546–554. doi: 10.1086/600870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Zhao J, Zhao J, Legge K, Perlman S. Age-related increases in PGD(2) expression impair respiratory DC migration, resulting in diminished T cell responses upon respiratory virus infection in mice. The Journal of clinical investigation. 2011;121:4921–4930. doi: 10.1172/JCI59777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Hinojosa CA, Mgbemena V, Van Roekel S, Austad SN, Miller RA, Bose S, Orihuela CJ. Enteric- delivered rapamycin enhances resistance of aged mice to pneumococcal pneumonia through reduced cellular senescence. Experimental gerontology. 2012;47:958–965. doi: 10.1016/j.exger.2012.08.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Nahrendorf M, Swirski FK, Aikawa E, Stangenberg L, Wurdinger T, Figueiredo JL, Libby P, Weissleder R, Pittet MJ. The healing myocardium sequentially mobilizes two monocyte subsets with divergent and complementary functions. The Journal of experimental medicine. 2007;204:3037–3047. doi: 10.1084/jem.20070885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Anzai A, Anzai T, Nagai S, Maekawa Y, Naito K, Kaneko H, Sugano Y, Takahashi T, Abe H, Mochizuki S, Sano M, Yoshikawa T, Okada Y, Koyasu S, Ogawa S, Fukuda K. Regulatory role of dendritic cells in postinfarction healing and left ventricular remodeling. Circulation. 2012;125:1234–1245. doi: 10.1161/CIRCULATIONAHA.111.052126. [DOI] [PubMed] [Google Scholar]
- 125.Dobaczewski M, Xia Y, Bujak M, Gonzalez-Quesada C, Frangogiannis NG. CCR5 signaling suppresses inflammation and reduces adverse remodeling of the infarcted heart, mediating recruitment of regulatory T cells. The American journal of pathology. 2010;176:2177–2187. doi: 10.2353/ajpath.2010.090759. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Hofmann U, Beyersdorf N, Weirather J, Podolskaya A, Bauersachs J, Ertl G, Kerkau T, Frantz S. Activation of CD4+ T lymphocytes improves wound healing and survival after experimental myocardial infarction in mice. Circulation. 2012;125:1652–1663. doi: 10.1161/CIRCULATIONAHA.111.044164. [DOI] [PubMed] [Google Scholar]
- 127.Frangogiannis NG, Lindsey ML, Michael LH, Youker KA, Bressler RB, Mendoza LH, Spengler RN, Smith CW, Entman ML. Resident cardiac mast cells degranulate and release preformed TNF-alpha, initiating the cytokine cascade in experimental canine myocardial ischemia/reperfusion. Circulation. 1998;98:699–710. doi: 10.1161/01.cir.98.7.699. [DOI] [PubMed] [Google Scholar]
- 128.Swirski FK, Nahrendorf M, Etzrodt M, Wildgruber M, Cortez-Retamozo V, Panizzi P, Figueiredo JL, 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 (New York, NY. 2009;325:612–616. doi: 10.1126/science.1175202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Troidl C, Mollmann H, Nef H, Masseli F, Voss S, Szardien S, Willmer M, Rolf A, Rixe J, Troidl K, Kostin S, Hamm C, Elsasser A. Classically and alternatively activated macrophages contribute to tissue remodelling after myocardial infarction. Journal of cellular and molecular medicine. 2009;13:3485–3496. doi: 10.1111/j.1582-4934.2009.00707.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.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. The Journal of experimental medicine. 2012;209:123–137. doi: 10.1084/jem.20111009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Spinale FG. Myocardial matrix remodeling and the matrix metalloproteinases: influence on cardiac form and function. Physiological reviews. 2007;87:1285–1342. doi: 10.1152/physrev.00012.2007. [DOI] [PubMed] [Google Scholar]
- 132.van Amerongen MJ, Harmsen MC, van Rooijen N, Petersen AH, van Luyn MJ. Macrophage depletion impairs wound healing and increases left ventricular remodeling after myocardial injury in mice. The American journal of pathology. 2007;170:818–829. doi: 10.2353/ajpath.2007.060547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Frangogiannis NG, Smith CW, Entman ML. The inflammatory response in myocardial infarction. Cardiovascular research. 2002;53:31–47. doi: 10.1016/s0008-6363(01)00434-5. [DOI] [PubMed] [Google Scholar]
- 134.Roberts R, DeMello V, Sobel BE. Deleterious effects of methylprednisolone in patients with myocardial infarction. Circulation. 1976;53:I204–206. [PubMed] [Google Scholar]
- 135.Harel-Adar T, Ben Mordechai T, Amsalem Y, Feinberg MS, Leor J, Cohen S. Modulation of cardiac macrophages by phosphatidylserine-presenting liposomes improves infarct repair. Proceedings of the National Academy of Sciences of the United States of America. 2011;108:1827–1832. doi: 10.1073/pnas.1015623108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Bujak M, Kweon HJ, Chatila K, Li N, Taffet G, Frangogiannis NG. Aging-related defects are associated with adverse cardiac remodeling in a mouse model of reperfused myocardial infarction. Journal of the American College of Cardiology. 2008;51:1384–1392. doi: 10.1016/j.jacc.2008.01.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Nahrendorf M, Swirski FK. Monocyte and macrophage heterogeneity in the heart. Circulation research. 2013;112:1624–1633. doi: 10.1161/CIRCRESAHA.113.300890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.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;153:362–375. doi: 10.1016/j.cell.2013.03.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Geissmann F, Manz MG, Jung S, Sieweke MH, Merad M, Ley K. Development of monocytes, macrophages, and dendritic cells. Science (New York, NY. 2010;327:656–661. doi: 10.1126/science.1178331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Francis GS. Neurohormonal control of heart failure. Cleveland Clinic journal of medicine. 2011;78(Suppl 1):S75–79. doi: 10.3949/ccjm.78.s1.13. [DOI] [PubMed] [Google Scholar]
- 141.Assmus B, Iwasaki M, Schachinger V, Roexe T, Koyanagi M, Iekushi K, Xu Q, Tonn T, Seifried E, Liebner S, Kranert WT, Grunwald F, Dimmeler S, Zeiher AM. Acute myocardial infarction activates progenitor cells and increases Wnt signalling in the bone marrow. European heart journal. 2012;33:1911–1919. doi: 10.1093/eurheartj/ehr388. [DOI] [PubMed] [Google Scholar]
- 142.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 HW, 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;487:325–329. doi: 10.1038/nature11260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Kaikita K, Hayasaki T, Okuma T, Kuziel WA, Ogawa H, Takeya M. Targeted deletion of CC chemokine receptor 2 attenuates left ventricular remodeling after experimental myocardial infarction. The American journal of pathology. 2004;165:439–447. doi: 10.1016/S0002-9440(10)63309-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Tsujioka H, Imanishi T, Ikejima H, Kuroi A, Takarada S, Tanimoto T, Kitabata H, Okochi K, Arita Y, Ishibashi K, Komukai K, Kataiwa H, Nakamura N, Hirata K, Tanaka A, Akasaka T. Impact of heterogeneity of human peripheral blood monocyte subsets on myocardial salvage in patients with primary acute myocardial infarction. Journal of the American College of Cardiology. 2009;54:130–138. doi: 10.1016/j.jacc.2009.04.021. [DOI] [PubMed] [Google Scholar]
- 145.Engstrom G, Melander O, Hedblad B. Leukocyte count and incidence of hospitalizations due to heart failure. Circ Heart Fail. 2009;2:217–222. doi: 10.1161/CIRCHEARTFAILURE.108.827071. [DOI] [PubMed] [Google Scholar]
- 146.Maekawa Y, Anzai T, Yoshikawa T, Asakura Y, Takahashi T, Ishikawa S, Mitamura H, Ogawa S. Prognostic significance of peripheral monocytosis after reperfused acute myocardial infarction:a possible role for left ventricular remodeling. Journal of the American College of Cardiology. 2002;39:241–246. doi: 10.1016/s0735-1097(01)01721-1. [DOI] [PubMed] [Google Scholar]
- 147.Massa M, Rosti V, Ferrario M, Campanelli R, Ramajoli I, Rosso R, De Ferrari GM, Ferlini M, Goffredo L, Bertoletti A, Klersy C, Pecci A, Moratti R, Tavazzi L. Increased circulating hematopoietic and endothelial progenitor cells in the early phase of acute myocardial infarction. Blood. 2005;105:199–206. doi: 10.1182/blood-2004-05-1831. [DOI] [PubMed] [Google Scholar]
- 148.Thom T, Haase N, Rosamond W, Howard VJ, Rumsfeld J, Manolio T, Zheng ZJ, Flegal K, O'Donnell C, Kittner S, Lloyd-Jones D, Goff DC, Jr., Hong Y, Adams R, Friday G, Furie K, Gorelick P, Kissela B, Marler J, Meigs J, Roger V, Sidney S, Sorlie P, Steinberger J, Wasserthiel-Smoller S, Wilson M, Wolf P. Heart disease and stroke statistics--2006 update: a report from the American Heart Association Statistics Committee and Stroke Statistics Subcommittee. Circulation. 2006;113:e85–151. doi: 10.1161/CIRCULATIONAHA.105.171600. [DOI] [PubMed] [Google Scholar]
- 149.Swirski FK, Nahrendorf M. Leukocyte behavior in atherosclerosis, myocardial infarction, and heart failure. Science (New York, NY. 2013;339:161–166. doi: 10.1126/science.1230719. [DOI] [PMC free article] [PubMed] [Google Scholar]