Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.833
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 23(2): 229-236, 2022 02.
Article in English | MEDLINE | ID: mdl-34949832

ABSTRACT

Aging is characterized by an increased vulnerability to infection and the development of inflammatory diseases, such as atherosclerosis, frailty, cancer and neurodegeneration. Here, we find that aging is associated with the loss of diurnally rhythmic innate immune responses, including monocyte trafficking from bone marrow to blood, response to lipopolysaccharide and phagocytosis. This decline in homeostatic immune responses was associated with a striking disappearance of circadian gene transcription in aged compared to young tissue macrophages. Chromatin accessibility was significantly greater in young macrophages than in aged macrophages; however, this difference did not explain the loss of rhythmic gene transcription in aged macrophages. Rather, diurnal expression of Kruppel-like factor 4 (Klf4), a transcription factor (TF) well established in regulating cell differentiation and reprogramming, was selectively diminished in aged macrophages. Ablation of Klf4 expression abolished diurnal rhythms in phagocytic activity, recapitulating the effect of aging on macrophage phagocytosis. Examination of individuals harboring genetic variants of KLF4 revealed an association with age-dependent susceptibility to death caused by bacterial infection. Our results indicate that loss of rhythmic Klf4 expression in aged macrophages is associated with disruption of circadian innate immune homeostasis, a mechanism that may underlie age-associated loss of protective immune responses.


Subject(s)
Circadian Clocks/genetics , Macrophages/physiology , Aging , Animals , Atherosclerosis/genetics , Cell Differentiation/genetics , Gene Expression Regulation/genetics , Immunity, Innate/genetics , Inflammation/genetics , Kruppel-Like Factor 4/genetics , Kruppel-Like Transcription Factors/genetics , Male , Mice , Mice, Inbred C57BL , Monocytes/physiology , Phagocytosis/genetics
2.
Nat Immunol ; 23(2): 217-228, 2022 02.
Article in English | MEDLINE | ID: mdl-35102344

ABSTRACT

During inflammation, Ly6Chi monocytes are rapidly mobilized from the bone marrow (BM) and are recruited into inflamed tissues, where they undergo monocyte-to-phagocyte transition (MTPT). The in vivo developmental trajectories of the MTPT and the contribution of individual cytokines to this process remain unclear. Here, we used a murine model of neuroinflammation to investigate how granulocyte-macrophage colony-stimulating factor (GM-CSF) and interferon-γ (IFNγ), two type 1 cytokines, controlled MTPT. Using genetic fate mapping, gene targeting and high-dimensional single-cell multiomics analyses, we found that IFNγ was essential for the gradual acquisition of a mature inflammatory phagocyte phenotype in Ly6Chi monocytes, while GM-CSF was required to license interleukin-1ß (IL-1ß) production, phagocytosis and oxidative burst. These results suggest that the proinflammatory cytokine environment guided MTPT trajectories in the inflamed central nervous system (CNS) and indicated that GM-CSF was the most prominent target for the disarming of monocyte progenies during neuroinflammation.


Subject(s)
Cell Differentiation/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Interferon-gamma/metabolism , Monocytes/metabolism , Neuroinflammatory Diseases/metabolism , Phagocytes/metabolism , Animals , Cytokines/metabolism , Female , Macrophages/metabolism , Macrophages/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Monocytes/physiology , Neuroinflammatory Diseases/physiopathology , Phagocytes/physiology
3.
Nat Immunol ; 21(5): 525-534, 2020 05.
Article in English | MEDLINE | ID: mdl-32313246

ABSTRACT

Multiple sclerosis (MS) is characterized by pathological inflammation that results from the recruitment of lymphoid and myeloid immune cells from the blood into the brain. Due to subset heterogeneity, defining the functional roles of the various cell subsets in acute and chronic stages of MS has been challenging. Here, we used index and transcriptional single-cell sorting to characterize the mononuclear phagocytes that infiltrate the central nervous system from the periphery in mice with experimentally induced autoimmune encephalomyelitis, a model of MS. We identified eight monocyte and three dendritic cell subsets at acute and chronic disease stages in which the defined transcriptional programs pointed toward distinct functions. Monocyte-specific cell ablation identified Cxcl10+ and Saa3+ monocytic subsets with a pathogenic potential. Transfer experiments with different monocyte and precursor subsets indicated that these Cxcl10+ and Saa3+ pathogenic cells were not derived from Ly6C+ monocytes but from early myeloid cell progenitors. These results suggest that blocking specific pathogenic monocytic subsets, including Cxcl10+ and Saa3+ monocytes, could be used for targeted therapeutic interventions.


Subject(s)
Dendritic Cells/physiology , Encephalomyelitis, Autoimmune, Experimental/immunology , Monocytes/physiology , Multiple Sclerosis/immunology , Phagocytes/physiology , Animals , Autoimmunity , Cell Differentiation , Cells, Cultured , Central Nervous System , Chemokine CXCL10/metabolism , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurogenic Inflammation , Serum Amyloid A Protein/metabolism , Single-Cell Analysis , Transcription Factors/genetics
4.
Immunity ; 54(7): 1417-1432.e7, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34004142

ABSTRACT

The transcriptional repressor ZEB2 regulates development of many cell fates among somatic, neural, and hematopoietic lineages, but the basis for its requirement in these diverse lineages is unclear. Here, we identified a 400-basepair (bp) region located 165 kilobases (kb) upstream of the Zeb2 transcriptional start site (TSS) that binds the E proteins at several E-box motifs and was active in hematopoietic lineages. Germline deletion of this 400-bp region (Zeb2Δ-165mice) specifically prevented Zeb2 expression in hematopoietic stem cell (HSC)-derived lineages. Zeb2Δ-165 mice lacked development of plasmacytoid dendritic cells (pDCs), monocytes, and B cells. All macrophages in Zeb2Δ-165 mice were exclusively of embryonic origin. Using single-cell chromatin profiling, we identified a second Zeb2 enhancer located at +164-kb that was selectively active in embryonically derived lineages, but not HSC-derived ones. Thus, Zeb2 expression in adult, but not embryonic, hematopoiesis is selectively controlled by the -165-kb Zeb2 enhancer.


Subject(s)
Enhancer Elements, Genetic/genetics , Hematopoiesis/genetics , Transcription, Genetic/genetics , Zinc Finger E-box Binding Homeobox 2/genetics , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Chromatin/genetics , Dendritic Cells/physiology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Monocytes/physiology
5.
Nat Immunol ; 17(10): 1176-86, 2016 10.
Article in English | MEDLINE | ID: mdl-27548431

ABSTRACT

Inflammasomes are positioned to rapidly escalate the intensity of inflammation by activating interleukin (IL)-1ß, IL-18 and cell death by pyroptosis. However, negative regulation of inflammasomes remains poorly understood, as is the signaling cascade that dampens inflammasome activity. We found that rapid NLRP3 inflammasome activation was directly inhibited by protein kinase A (PKA), which was induced by prostaglandin E2 (PGE2) signaling via the PGE2 receptor E-prostanoid 4 (EP4). PKA directly phosphorylated the cytoplasmic receptor NLRP3 and attenuated its ATPase function. We found that Ser295 in human NLRP3 was critical for rapid inhibition and PKA phosphorylation. Mutations in NLRP3-encoding residues adjacent to Ser295 have been linked to the inflammatory disease CAPS (cryopyrin-associated periodic syndromes). NLRP3-S295A phenocopied the human CAPS mutants. These data suggest that negative regulation at Ser295 is critical for restraining the NLRP3 inflammasome and identify a molecular basis for CAPS-associated NLRP3 mutations.


Subject(s)
Cryopyrin-Associated Periodic Syndromes/immunology , Cyclic AMP-Dependent Protein Kinases/metabolism , Inflammasomes/metabolism , Monocytes/physiology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Adenosine Triphosphatases/metabolism , Animals , Cell Line , Cryopyrin-Associated Periodic Syndromes/genetics , Dinoprostone/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mutation/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Phenotype , Phosphorylation/genetics , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Serine/genetics , Signal Transduction/genetics
6.
Immunity ; 51(4): 638-654.e9, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31561945

ABSTRACT

Macrophages are strongly adapted to their tissue of residence. Yet, little is known about the cell-cell interactions that imprint the tissue-specific identities of macrophages in their respective niches. Using conditional depletion of liver Kupffer cells, we traced the developmental stages of monocytes differentiating into Kupffer cells and mapped the cellular interactions imprinting the Kupffer cell identity. Kupffer cell loss induced tumor necrosis factor (TNF)- and interleukin-1 (IL-1) receptor-dependent activation of stellate cells and endothelial cells, resulting in the transient production of chemokines and adhesion molecules orchestrating monocyte engraftment. Engrafted circulating monocytes transmigrated into the perisinusoidal space and acquired the liver-associated transcription factors inhibitor of DNA 3 (ID3) and liver X receptor-α (LXR-α). Coordinated interactions with hepatocytes induced ID3 expression, whereas endothelial cells and stellate cells induced LXR-α via a synergistic NOTCH-BMP pathway. This study shows that the Kupffer cell niche is composed of stellate cells, hepatocytes, and endothelial cells that together imprint the liver-specific macrophage identity.


Subject(s)
Endothelial Cells/physiology , Hepatic Stellate Cells/physiology , Hepatocytes/physiology , Kupffer Cells/physiology , Liver/cytology , Macrophages/physiology , Monocytes/physiology , Animals , Cell Communication , Cell Differentiation , Cells, Cultured , Cellular Microenvironment , Female , Gene Expression Regulation , Inhibitor of Differentiation Proteins/genetics , Inhibitor of Differentiation Proteins/metabolism , Liver X Receptors/genetics , Liver X Receptors/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Notch/metabolism
7.
Immunity ; 48(1): 91-106.e6, 2018 01 16.
Article in English | MEDLINE | ID: mdl-29343444

ABSTRACT

CD103+ dendritic cells are critical for cross-presentation of tumor antigens. Here we have shown that during immunotherapy, large numbers of cells expressing CD103 arose in murine tumors via direct differentiation of Ly6c+ monocytic precursors. These Ly6c+CD103+ cells could derive from bone-marrow monocytic progenitors (cMoPs) or from peripheral cells present within the myeloid-derived suppressor cell (MDSC) population. Differentiation was controlled by inflammation-induced activation of the transcription factor p53, which drove upregulation of Batf3 and acquisition of the Ly6c+CD103+ phenotype. Mice with a targeted deletion of p53 in myeloid cells selectively lost the Ly6c+CD103+ population and became unable to respond to multiple forms of immunotherapy and immunogenic chemotherapy. Conversely, increasing p53 expression using a p53-agonist drug caused a sustained increase in Ly6c+CD103+ cells in tumors during immunotherapy, which markedly enhanced the efficacy and duration of response. Thus, p53-driven differentiation of Ly6c+CD103+ monocytic cells represents a potent and previously unrecognized target for immunotherapy.


Subject(s)
Antigen-Presenting Cells/physiology , Monocytes/physiology , Myeloid Cells/metabolism , Neoplasms/immunology , Tumor Suppressor Protein p53/metabolism , Animals , Antigen-Presenting Cells/immunology , Antigens, CD/metabolism , Antigens, Ly/metabolism , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Flow Cytometry , Humans , Immunotherapy/methods , Integrin alpha Chains/metabolism , Mice , Monocytes/immunology , Myeloid Cells/physiology
8.
Immunity ; 47(5): 890-902.e4, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29166589

ABSTRACT

Granulocyte-monocyte progenitors (GMPs) and monocyte-dendritic cell progenitors (MDPs) produce monocytes during homeostasis and in response to increased demand during infection. Both progenitor populations are thought to derive from common myeloid progenitors (CMPs), and a hierarchical relationship (CMP-GMP-MDP-monocyte) is presumed to underlie monocyte differentiation. Here, however, we demonstrate that mouse MDPs arose from CMPs independently of GMPs, and that GMPs and MDPs produced monocytes via similar but distinct monocyte-committed progenitors. GMPs and MDPs yielded classical (Ly6Chi) monocytes with gene expression signatures that were defined by their origins and impacted their function. GMPs produced a subset of "neutrophil-like" monocytes, whereas MDPs gave rise to a subset of monocytes that yielded monocyte-derived dendritic cells. GMPs and MDPs were also independently mobilized to produce specific combinations of myeloid cell types following the injection of microbial components. Thus, the balance of GMP and MDP differentiation shapes the myeloid cell repertoire during homeostasis and following infection.


Subject(s)
Dendritic Cells/physiology , Granulocyte Precursor Cells/physiology , Monocytes/physiology , Myeloid Progenitor Cells/physiology , Animals , Antigens, Ly/analysis , Cell Differentiation , Leukosialin/analysis , Mice , Sequence Analysis, RNA , Transcriptome
9.
Immunity ; 47(1): 12-14, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28723545

ABSTRACT

Sickness in mammals can lead to cognition deficits, although the underlying mechanisms remain elusive. In a recent Nature Medicine article, Garré et al. (2017) report that sickness-induced cortical dendritic spine loss and impaired memory formation is mediated by CX3CR1+ monocyte-derived TNF-α.


Subject(s)
Dendritic Spines/physiology , Mental Disorders/immunology , Monocytes/physiology , Motor Neurons/physiology , Nerve Net , Neuronal Plasticity , Virus Diseases/immunology , Animals , CX3C Chemokine Receptor 1 , Humans , Memory , Mental Disorders/etiology , Mental Disorders/psychology , Mice , Monocytes/virology , Motor Neurons/virology , Poly I-C/immunology , Receptors, Chemokine/metabolism , Tumor Necrosis Factor-alpha/metabolism , Virus Diseases/complications , Virus Diseases/psychology
10.
Physiol Rev ; 98(4): 2523-2569, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30156496

ABSTRACT

Research during the last decade has generated numerous insights on the presence, phenotype, and function of myeloid cells in cardiovascular organs. Newer tools with improved detection sensitivities revealed sizable populations of tissue-resident macrophages in all major healthy tissues. The heart and blood vessels contain robust numbers of these cells; for instance, 8% of noncardiomyocytes in the heart are macrophages. This number and the cell's phenotype change dramatically in disease conditions. While steady-state macrophages are mostly monocyte independent, macrophages residing in the inflamed vascular wall and the diseased heart derive from hematopoietic organs. In this review, we will highlight signals that regulate macrophage supply and function, imaging applications that can detect changes in cell numbers and phenotype, and opportunities to modulate cardiovascular inflammation by targeting macrophage biology. We strive to provide a systems-wide picture, i.e., to focus not only on cardiovascular organs but also on tissues involved in regulating cell supply and phenotype, as well as comorbidities that promote cardiovascular disease. We will summarize current developments at the intersection of immunology, detection technology, and cardiovascular health.


Subject(s)
Cardiovascular System/physiopathology , Macrophages/physiology , Animals , Humans , Inflammation/physiopathology , Monocytes/physiology , Phenotype
11.
Immunity ; 45(3): 626-640, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27637148

ABSTRACT

Interferon regulatory factor-8 (IRF8) has been proposed to be essential for development of monocytes, plasmacytoid dendritic cells (pDCs) and type 1 conventional dendritic cells (cDC1s) and remains highly expressed in differentiated DCs. Transcription factors that are required to maintain the identity of terminally differentiated cells are designated "terminal selectors." Using BM chimeras, conditional Irf8(fl/fl) mice and various promotors to target Cre recombinase to different stages of monocyte and DC development, we have identified IRF8 as a terminal selector of the cDC1 lineage controlling survival. In monocytes, IRF8 was necessary during early but not late development. Complete or late deletion of IRF8 had no effect on pDC development or survival but altered their phenotype and gene-expression profile leading to increased T cell stimulatory function but decreased type 1 interferon production. Thus, IRF8 differentially controls the survival and function of terminally differentiated monocytes, cDC1s, and pDCs.


Subject(s)
Cell Differentiation/physiology , Dendritic Cells/metabolism , Dendritic Cells/physiology , Interferon Regulatory Factors/metabolism , Transcription Factors/metabolism , Animals , Interferon Type I/metabolism , Mice , Mice, Inbred C57BL , Monocytes/metabolism , Monocytes/physiology , Promoter Regions, Genetic/physiology , T-Lymphocytes/metabolism , T-Lymphocytes/physiology
12.
Immunol Rev ; 294(1): 188-204, 2020 03.
Article in English | MEDLINE | ID: mdl-31782165

ABSTRACT

Rheumatoid arthritis (RA) risk has a large genetic component (~60%) that is still not fully understood. This has hampered the design of effective treatments that could promise lifelong remission. RA is a polygenic disease with 106 known genome-wide significant associated loci and thousands of small effect causal variants. Our current understanding of RA risk has suggested cell-type-specific contexts for causal variants, implicating CD4 + effector memory T cells, as well as monocytes, B cells and stromal fibroblasts. While these cellular states and categories are still mechanistically broad, future studies may identify causal cell subpopulations. These efforts are propelled by advances in single cell profiling. Identification of causal cell subpopulations may accelerate therapeutic intervention to achieve lifelong remission.


Subject(s)
Arthritis, Rheumatoid/genetics , B-Lymphocytes/physiology , CD4-Positive T-Lymphocytes/physiology , Fibroblasts/physiology , Monocytes/physiology , Animals , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Immunologic Memory , Risk
13.
Immunol Cell Biol ; 101(10): 947-963, 2023.
Article in English | MEDLINE | ID: mdl-37694300

ABSTRACT

Macrophages have previously been characterized based on phenotypical and functional differences into suggested simplified subtypes of MØ, M1, M2a and M2c. These macrophage subtypes can be generated in a well-established primary monocyte culture model that produces cells expressing accepted subtype surface markers. To determine how these subtypes retain functional similarities and better understand their formation, we generated all four subtypes from the same donors. Comparative whole-cell proteomics confirmed that four distinct macrophage subtypes could be induced from the same donor material, with > 50% of 5435 identified proteins being significantly altered in abundance between subtypes. Functional assessment highlighted that these distinct protein expression profiles are primed to enable specific cell functions, indicating that this shifting proteome is predictive of meaningful changes in cell characteristics. Importantly, the 2552 proteins remained consistent in abundance across all macrophage subtypes examined, demonstrating maintenance of a stable core proteome that likely enables swift polarity changes. We next explored the cross-polarization capabilities of preactivated M1 macrophages treated with dexamethasone. Importantly, these treated cells undergo a partial repolarization toward the M2c surface markers but still retain the M1 functional phenotype. Our investigation of polarized macrophage subtypes therefore provides evidence of a sliding scale of macrophage functionality, with these data sets providing a valuable benchmark resource for further studies of macrophage polarity, with relevance for cell therapy development and drug discovery.


Subject(s)
Proteome , Proteomics , Proteome/metabolism , Cells, Cultured , Macrophages/metabolism , Monocytes/physiology
14.
Small ; 19(11): e2205429, 2023 03.
Article in English | MEDLINE | ID: mdl-36638251

ABSTRACT

Fluorescent nanodiamonds (FNDs) with negative nitrogen-vacancy (NV- ) defect centers are great probes for biosensing applications, with potential to act as biomarkers for cell differentiation. To explore this concept, uptake of FNDs (≈120 nm) by THP-1 monocytes and monocyte-derived M0-macrophages is studied. The time course analysis of FND uptake by monocytes confirms differing FND-cell interactions and a positive time-dependence. No effect on cell viability, proliferation, and differentiation potential into macrophages is observed, while cells saturated with FNDs, unload the FNDs completely by 25 cell divisions and subsequently take up a second dose effectively. FND uptake variations by THP-1 cells at early exposure-times indicate differing phagocytic capability. The cell fraction that exhibits relatively enhanced FND uptake is associated to a macrophage phenotype which derives from spontaneous monocyte differentiation. In accordance, chemical-differentiation of the THP-1 cells into M0-macrophages triggers increased and homogeneous FND uptake, depleting the fraction of cells that were non-responsive to FNDs. These observations imply that FND uptake allows for distinction between the two cell subtypes based on phagocytic capacity. Overall, FNDs demonstrate effective cell labeling of monocytes and macrophages, and are promising candidates for sensing biological processes that involve cell differentiation.


Subject(s)
Biosensing Techniques , Fluorescent Dyes , Macrophages , Monocytes , Nanodiamonds , Phagocytosis , Nanodiamonds/chemistry , Nanodiamonds/toxicity , Nitrogen/chemistry , Fluorescent Dyes/chemistry , Fluorescent Dyes/toxicity , Humans , Cell Line , Monocytes/cytology , Monocytes/drug effects , Monocytes/physiology , Macrophages/cytology , Macrophages/drug effects , Macrophages/physiology , Cell Survival/drug effects , Cell Proliferation/drug effects , Cell Differentiation/drug effects , Phagocytosis/drug effects
15.
Immunity ; 40(1): 3-5, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-24439261

ABSTRACT

A study by Epelman et al. (2014) in this issue of Immunity demonstrates that diverse subpopulations of macrophages reside in the adult heart and can be maintained by multiple mechanisms involving both local proliferation and contributions from monocytes.


Subject(s)
Macrophages/immunology , Monocytes/physiology , Myocarditis/immunology , Myocardium/immunology , Animals
16.
Immunity ; 40(1): 91-104, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-24439267

ABSTRACT

Cardiac macrophages are crucial for tissue repair after cardiac injury but are not well characterized. Here we identify four populations of cardiac macrophages. At steady state, resident macrophages were primarily maintained through local proliferation. However, after macrophage depletion or during cardiac inflammation, Ly6c(hi) monocytes contributed to all four macrophage populations, whereas resident macrophages also expanded numerically through proliferation. Genetic fate mapping revealed that yolk-sac and fetal monocyte progenitors gave rise to the majority of cardiac macrophages, and the heart was among a minority of organs in which substantial numbers of yolk-sac macrophages persisted in adulthood. CCR2 expression and dependence distinguished cardiac macrophages of adult monocyte versus embryonic origin. Transcriptional and functional data revealed that monocyte-derived macrophages coordinate cardiac inflammation, while playing redundant but lesser roles in antigen sampling and efferocytosis. These data highlight the presence of multiple cardiac macrophage subsets, with different functions, origins, and strategies to regulate compartment size.


Subject(s)
Macrophages/immunology , Monocytes/physiology , Myocarditis/immunology , Myocardium/immunology , Animals , Antigen Presentation , Antigens, Ly/metabolism , Cell Death , Cell Differentiation , Cell Lineage , Cells, Cultured , Fetal Development , Heart/embryology , Homeostasis , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocytes, Cardiac/immunology , Phagocytosis , Receptors, CCR2/metabolism , Transcriptome , Yolk Sac/cytology
17.
Immunity ; 41(6): 947-59, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25500368

ABSTRACT

Nonresolving inflammation expands a heterogeneous population of myeloid suppressor cells capable of inhibiting T cell function. This heterogeneity has confounded the functional dissection of individual myeloid subpopulations and presents an obstacle for antitumor immunity and immunotherapy. Using genetic manipulation of cell death pathways, we found the monocytic suppressor-cell subset, but not the granulocytic subset, requires continuous c-FLIP expression to prevent caspase-8-dependent, RIPK3-independent cell death. Development of the granulocyte subset requires MCL-1-mediated control of the intrinsic mitochondrial death pathway. Monocytic suppressors tolerate the absence of MCL-1 provided cytokines increase expression of the MCL-1-related protein A1. Monocytic suppressors mediate T cell suppression, whereas their granulocytic counterparts lack suppressive function. The loss of the granulocytic subset via conditional MCL-1 deletion did not alter tumor incidence implicating the monocytic compartment as the functionally immunosuppressive subset in vivo. Thus, death pathway modulation defines the development, survival, and function of myeloid suppressor cells.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Granulocytes/physiology , Monocytes/physiology , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Myeloid Cells/physiology , Neoplasms, Experimental/immunology , Animals , Apoptosis/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , CD8-Positive T-Lymphocytes/immunology , Carcinogenesis/genetics , Caspase 8/metabolism , Cell Differentiation/genetics , Cell Line, Tumor , Cell Lineage/genetics , Coculture Techniques , Fas-Associated Death Domain Protein/genetics , Fas-Associated Death Domain Protein/metabolism , Immune Tolerance/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Minor Histocompatibility Antigens , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Neoplasm Transplantation , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/genetics , Signal Transduction/genetics
18.
Circ Res ; 128(12): 2017-2036, 2021 06 11.
Article in English | MEDLINE | ID: mdl-34110909

ABSTRACT

The association between inflammation, infection, and venous thrombosis has long been recognized; yet, only in the last decades have we begun to understand the mechanisms through which the immune and coagulation systems interact and reciprocally regulate one another. These interconnected networks mount an effective response to injury and pathogen invasion, but if unregulated can result in pathological thrombosis and organ damage. Neutrophils, monocytes, and platelets interact with each other and the endothelium in host defense and also play critical roles in the formation of venous thromboembolism. This knowledge has advanced our understanding of both human physiology and pathophysiology, as well as identified mechanisms of anticoagulant resistance and novel therapeutic targets for the prevention and treatment of thrombosis. In this review, we discuss the contributions of inflammation and infection to venous thromboembolism.


Subject(s)
Infections/complications , Inflammation/complications , Venous Thromboembolism/etiology , Adaptive Immunity , Blood Coagulation/physiology , Blood Platelets/physiology , Endothelium, Vascular/physiology , Extracellular Traps , Extracellular Vesicles/physiology , Fibrinolysis , Hematopoiesis , Hemostasis/physiology , Humans , Immune System/physiology , Leukocytes/physiology , Monocytes/physiology , Neutrophils/physiology , Venous Thromboembolism/prevention & control , Venous Thromboembolism/therapy
19.
Circ Res ; 128(11): 1766-1779, 2021 05 28.
Article in English | MEDLINE | ID: mdl-34043424

ABSTRACT

Cardiac injury remains a major cause of morbidity and mortality worldwide. Despite significant advances, a full understanding of why the heart fails to fully recover function after acute injury, and why progressive heart failure frequently ensues, remains elusive. No therapeutics, short of heart transplantation, have emerged to reliably halt or reverse the inexorable progression of heart failure in the majority of patients once it has become clinically evident. To date, most pharmacological interventions have focused on modifying hemodynamics (reducing afterload, controlling blood pressure and blood volume) or on modifying cardiac myocyte function. However, important contributions of the immune system to normal cardiac function and the response to injury have recently emerged as exciting areas of investigation. Therapeutic interventions aimed at harnessing the power of immune cells hold promise for new treatment avenues for cardiac disease. Here, we review the immune response to heart injury, its contribution to cardiac fibrosis, and the potential of immune modifying therapies to affect cardiac repair.


Subject(s)
Heart Failure/therapy , Heart Injuries/therapy , Immunotherapy/methods , Adaptive Immunity , B-Lymphocytes/physiology , Bioengineering , Cytokines/metabolism , Disease Progression , Eosinophils/physiology , Fibroblasts/physiology , Fibrosis , Heart Failure/etiology , Heart Failure/immunology , Heart Injuries/immunology , Humans , Immunotherapy, Adoptive , Macrophages/physiology , Mast Cells/physiology , Monocytes/physiology , Myocardium/pathology , Myocytes, Cardiac/physiology , Neutrophils/physiology , Receptors, Chimeric Antigen , T-Lymphocytes/physiology , T-Lymphocytes/transplantation
20.
Arterioscler Thromb Vasc Biol ; 42(5): e131-e144, 2022 05.
Article in English | MEDLINE | ID: mdl-35387476

ABSTRACT

BACKGROUND: The CCL2 (CC-chemokine ligand 2)/CCR2 (CC-chemokine receptor 2) axis governs monocyte recruitment to atherosclerotic lesions. Genetic and epidemiological studies show strong associations of CCL2 levels with atherosclerotic disease. Still, experimental studies testing pharmacological inhibition of CCL2 or CCR2 in atheroprone mice apply widely different approaches and report variable results, thus halting clinical translation. METHODS: We systematically searched the literature for studies employing pharmacological CCL2/CCR2 blockade in atheroprone mice and meta-analyzed their effects on lesion size and morphology. RESULTS: In a meta-analysis of 14 studies testing 11 different agents, CCL2/CCR2 blockade attenuated atherosclerotic lesion size in the aortic root or arch (g=-0.75 [-1.17 to -0.32], P=6×10-4; N=171/171 mice in experimental/control group), the carotid (g=-2.39 [-4.23 to -0.55], P=0.01; N=24/25), and the femoral artery (g=-2.38 [-3.50 to -1.26], P=3×10-5; N=10/10). Furthermore, CCL2/CCR2 inhibition reduced intralesional macrophage accumulation and increased smooth muscle cell content and collagen deposition. The effects of CCL2/CCR2 inhibition on lesion size correlated with reductions in plaque macrophage accumulation, in accord with a prominent role of CCL2/CCR2 signaling in monocyte recruitment. Subgroup analyses showed comparable efficacy of different CCL2- and CCR2-inhibitors in reducing lesion size and intralesional macrophages. The quality assessment revealed high risk of detection bias due to lack of blinding during outcome assessment, as well as evidence of attrition and reporting bias. CONCLUSIONS: Preclinical evidence suggests that pharmacological targeting of CCL2 or CCR2 might lower atherosclerotic lesion burden, but the majority of existing studies suffer major quality issues that highlight the need for additional high-quality research.


Subject(s)
Atherosclerosis , Chemokine CCL2 , Animals , Atherosclerosis/drug therapy , Atherosclerosis/genetics , Atherosclerosis/prevention & control , Chemokine CCL2/genetics , Chemokines , Macrophages/pathology , Mice , Mice, Inbred C57BL , Monocytes/physiology , Receptors, CCR2/genetics
SELECTION OF CITATIONS
SEARCH DETAIL