Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 144
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Rev Mol Cell Biol ; 24(6): 430-447, 2023 06.
Article in English | MEDLINE | ID: mdl-36596869

ABSTRACT

Genes specifying long non-coding RNAs (lncRNAs) occupy a large fraction of the genomes of complex organisms. The term 'lncRNAs' encompasses RNA polymerase I (Pol I), Pol II and Pol III transcribed RNAs, and RNAs from processed introns. The various functions of lncRNAs and their many isoforms and interleaved relationships with other genes make lncRNA classification and annotation difficult. Most lncRNAs evolve more rapidly than protein-coding sequences, are cell type specific and regulate many aspects of cell differentiation and development and other physiological processes. Many lncRNAs associate with chromatin-modifying complexes, are transcribed from enhancers and nucleate phase separation of nuclear condensates and domains, indicating an intimate link between lncRNA expression and the spatial control of gene expression during development. lncRNAs also have important roles in the cytoplasm and beyond, including in the regulation of translation, metabolism and signalling. lncRNAs often have a modular structure and are rich in repeats, which are increasingly being shown to be relevant to their function. In this Consensus Statement, we address the definition and nomenclature of lncRNAs and their conservation, expression, phenotypic visibility, structure and functions. We also discuss research challenges and provide recommendations to advance the understanding of the roles of lncRNAs in development, cell biology and disease.


Subject(s)
RNA, Long Noncoding , RNA, Long Noncoding/genetics , Cell Nucleus/genetics , Chromatin/genetics , Regulatory Sequences, Nucleic Acid , RNA Polymerase II/genetics
2.
Nat Immunol ; 19(6): 526-537, 2018 06.
Article in English | MEDLINE | ID: mdl-29777212

ABSTRACT

After activation, cells of the myeloid lineage undergo robust metabolic transitions, as well as discrete epigenetic changes, that can dictate both ongoing and future inflammatory responses. In atherosclerosis, in which macrophages play central roles in the initiation, growth, and ultimately rupture of arterial plaques, altered metabolism is a key feature that dictates macrophage function and subsequent disease progression. This Review explores how factors central to the plaque microenvironment (for example, altered cholesterol metabolism, oxidative stress, hypoxia, apoptotic and necrotic cells, and hyperglycemia) shape the metabolic rewiring of macrophages in atherosclerosis as well as how these metabolic shifts in turn alter macrophage immune-effector and tissue-reparative functions. Finally, this overview offers insight into the challenges and opportunities of harnessing metabolism to modulate aberrant macrophage responses in disease.


Subject(s)
Atherosclerosis/immunology , Atherosclerosis/metabolism , Macrophages/immunology , Macrophages/metabolism , Animals , Humans
3.
Nat Immunol ; 18(6): 642-653, 2017 06.
Article in English | MEDLINE | ID: mdl-28436955

ABSTRACT

It remains unclear whether activated inflammatory macrophages can adopt features of tissue-resident macrophages, or what mechanisms might mediate such a phenotypic conversion. Here we show that vitamin A is required for the phenotypic conversion of interleukin 4 (IL-4)-activated monocyte-derived F4/80intCD206+PD-L2+MHCII+ macrophages into macrophages with a tissue-resident F4/80hiCD206-PD-L2-MHCII-UCP1+ phenotype in the peritoneal cavity of mice and during the formation of liver granulomas in mice infected with Schistosoma mansoni. The phenotypic conversion of F4/80intCD206+ macrophages into F4/80hiCD206- macrophages was associated with almost complete remodeling of the chromatin landscape, as well as alteration of the transcriptional profiles. Vitamin A-deficient mice infected with S. mansoni had disrupted liver granuloma architecture and increased mortality, which indicates that failure to convert macrophages from the F4/80intCD206+ phenotype to F4/80hiCD206- may lead to dysregulated inflammation during helminth infection.


Subject(s)
Granuloma/immunology , Liver/immunology , Macrophages/immunology , Schistosomiasis mansoni/immunology , Vitamin A Deficiency/immunology , Animals , Antigens, Differentiation/metabolism , Flow Cytometry , Histocompatibility Antigens Class II/metabolism , Interleukin-4/immunology , Lectins, C-Type/metabolism , Liver/pathology , Macrophages/drug effects , Macrophages/metabolism , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Mannose Receptor , Mannose-Binding Lectins/metabolism , Mice , Peritoneal Cavity/cytology , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Real-Time Polymerase Chain Reaction , Receptors, Cell Surface/metabolism , Schistosoma mansoni , Schistosomiasis mansoni/pathology , Tretinoin/pharmacology , Uncoupling Protein 1/metabolism , Vitamins/pharmacology
4.
Nat Immunol ; 17(6): 677-86, 2016 06.
Article in English | MEDLINE | ID: mdl-27089382

ABSTRACT

Mycobacterium tuberculosis (Mtb) survives in macrophages by evading delivery to the lysosome and promoting the accumulation of lipid bodies, which serve as a bacterial source of nutrients. We found that by inducing the microRNA (miRNA) miR-33 and its passenger strand miR-33*, Mtb inhibited integrated pathways involved in autophagy, lysosomal function and fatty acid oxidation to support bacterial replication. Silencing of miR-33 and miR-33* by genetic or pharmacological means promoted autophagy flux through derepression of key autophagy effectors (such as ATG5, ATG12, LC3B and LAMP1) and AMPK-dependent activation of the transcription factors FOXO3 and TFEB, which enhanced lipid catabolism and Mtb xenophagy. These data define a mammalian miRNA circuit used by Mtb to coordinately inhibit autophagy and reprogram host lipid metabolism to enable intracellular survival and persistence in the host.


Subject(s)
Autophagy/genetics , Lipid Metabolism/genetics , Lysosomes/physiology , Macrophages/physiology , MicroRNAs/metabolism , Mycobacterium tuberculosis/physiology , Tuberculosis/genetics , Animals , Cells, Cultured , Host-Pathogen Interactions , Humans , Immune Evasion , Lysosomes/microbiology , Macrophages/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Signal Transduction , Transcription Factors/metabolism
5.
Proc Natl Acad Sci U S A ; 121(15): e2400675121, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38564634

ABSTRACT

Atherosclerosis is fueled by a failure to resolve lipid-driven inflammation within the vasculature that drives plaque formation. Therapeutic approaches to reverse atherosclerotic inflammation are needed to address the rising global burden of cardiovascular disease (CVD). Recently, metabolites have gained attention for their immunomodulatory properties, including itaconate, which is generated from the tricarboxylic acid-intermediate cis-aconitate by the enzyme Immune Responsive Gene 1 (IRG1/ACOD1). Here, we tested the therapeutic potential of the IRG1-itaconate axis for human atherosclerosis. Using single-cell RNA sequencing (scRNA-seq), we found that IRG1 is up-regulated in human coronary atherosclerotic lesions compared to patient-matched healthy vasculature, and in mouse models of atherosclerosis, where it is primarily expressed by plaque monocytes, macrophages, and neutrophils. Global or hematopoietic Irg1-deficiency in mice increases atherosclerosis burden, plaque macrophage and lipid content, and expression of the proatherosclerotic cytokine interleukin (IL)-1ß. Mechanistically, absence of Irg1 increased macrophage lipid accumulation, and accelerated inflammation via increased neutrophil extracellular trap (NET) formation and NET-priming of the NLRP3-inflammasome in macrophages, resulting in increased IL-1ß release. Conversely, supplementation of the Irg1-itaconate axis using 4-octyl itaconate (4-OI) beneficially remodeled advanced plaques and reduced lesional IL-1ß levels in mice. To investigate the effects of 4-OI in humans, we leveraged an ex vivo systems-immunology approach for CVD drug discovery. Using CyTOF and scRNA-seq of peripheral blood mononuclear cells treated with plasma from CVD patients, we showed that 4-OI attenuates proinflammatory phospho-signaling and mediates anti-inflammatory rewiring of macrophage populations. Our data highlight the relevance of pursuing IRG1-itaconate axis supplementation as a therapeutic approach for atherosclerosis in humans.


Subject(s)
Atherosclerosis , Plaque, Atherosclerotic , Animals , Humans , Mice , Atherosclerosis/drug therapy , Atherosclerosis/genetics , Cholesterol , Inflammation/metabolism , Leukocytes, Mononuclear/metabolism , Lipids , Plaque, Atherosclerotic/drug therapy , Succinates/metabolism
6.
Cell ; 145(3): 341-55, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21529710

ABSTRACT

In atherosclerosis, the accumulation of apolipoprotein B-lipoproteins in the matrix beneath the endothelial cell layer of blood vessels leads to the recruitment of monocytes, the cells of the immune system that give rise to macrophages and dendritic cells. Macrophages derived from these recruited monocytes participate in a maladaptive, nonresolving inflammatory response that expands the subendothelial layer due to the accumulation of cells, lipid, and matrix. Some lesions subsequently form a necrotic core, triggering acute thrombotic vascular disease, including myocardial infarction, stroke, and sudden cardiac death. This Review discusses the central roles of macrophages in each of these stages of disease pathogenesis.


Subject(s)
Atherosclerosis/immunology , Macrophages/immunology , Animals , Atherosclerosis/metabolism , Dendritic Cells/immunology , Humans , Macrophages/pathology , Monocytes/pathology , Signal Transduction
7.
Nat Immunol ; 14(6): 543-53, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23644505

ABSTRACT

Phagocytosis is a fundamental cellular process that is pivotal for immunity as it coordinates microbial killing, innate immune activation and antigen presentation. An essential step in this process is phagosome acidification, which regulates many functions of these organelles that allow phagosomes to participate in processes that are essential to both innate and adaptive immunity. Here we report that acidification of phagosomes containing Gram-positive bacteria is regulated by the NLRP3 inflammasome and caspase-1. Active caspase-1 accumulates on phagosomes and acts locally to control the pH by modulating buffering by the NADPH oxidase NOX2. These data provide insight into a mechanism by which innate immune signals can modify cellular defenses and establish a new function for the NLRP3 inflammasome and caspase-1 in host defense.


Subject(s)
Carrier Proteins/immunology , Caspase 1/immunology , Inflammasomes/immunology , Membrane Glycoproteins/immunology , NADPH Oxidases/immunology , Phagosomes/immunology , Animals , Carrier Proteins/metabolism , Caspase 1/metabolism , Cells, Cultured , Enzyme Activation/immunology , Flow Cytometry , HEK293 Cells , Host-Pathogen Interactions/immunology , Humans , Hydrogen-Ion Concentration , Immunoblotting , Inflammasomes/metabolism , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Membrane Glycoproteins/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Microscopy, Electron , NADPH Oxidase 2 , NADPH Oxidases/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Phagocytosis/immunology , Phagosomes/metabolism , Phagosomes/microbiology , Phagosomes/ultrastructure , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Staphylococcus aureus/immunology , Staphylococcus aureus/physiology
8.
Nat Immunol ; 14(8): 812-20, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23812099

ABSTRACT

Particulate ligands, including cholesterol crystals and amyloid fibrils, induce production of interleukin 1ß (IL-1ß) dependent on the cytoplasmic sensor NLRP3 in atherosclerosis, Alzheimer's disease and diabetes. Soluble endogenous ligands, including oxidized low-density lipoprotein (LDL), amyloid-ß and amylin peptides, accumulate in such diseases. Here we identify an endocytic pathway mediated by the pattern-recognition receptor CD36 that coordinated the intracellular conversion of those soluble ligands into crystals or fibrils, which resulted in lysosomal disruption and activation of the NLRP3 inflammasome. Consequently, macrophages that lacked CD36 failed to elicit IL-1ß production in response to those ligands, and targeting CD36 in atherosclerotic mice resulted in lower serum concentrations of IL-1ß and accumulation of cholesterol crystals in plaques. Collectively, our findings highlight the importance of CD36 in the accrual and nucleation of NLRP3 ligands from within the macrophage and position CD36 as a central regulator of inflammasome activation in sterile inflammation.


Subject(s)
Alzheimer Disease/immunology , Atherosclerosis/immunology , CD36 Antigens/immunology , Carrier Proteins/immunology , Diabetes Mellitus, Type 2/immunology , Inflammation/immunology , Animals , CD36 Antigens/genetics , Carrier Proteins/genetics , Inflammasomes/immunology , Interleukin-1beta/immunology , Lipoproteins, LDL/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Microscopy, Fluorescence , NLR Family, Pyrin Domain-Containing 3 Protein , RNA/chemistry , RNA/genetics , Real-Time Polymerase Chain Reaction
9.
Proc Natl Acad Sci U S A ; 119(37): e2210321119, 2022 09 13.
Article in English | MEDLINE | ID: mdl-36001732

ABSTRACT

Long noncoding RNAs (lncRNAs) have emerged as critical regulators of gene expression, yet their contribution to immune regulation in humans remains poorly understood. Here, we report that the primate-specific lncRNA CHROMR is induced by influenza A virus and SARS-CoV-2 infection and coordinates the expression of interferon-stimulated genes (ISGs) that execute antiviral responses. CHROMR depletion in human macrophages reduces histone acetylation at regulatory regions of ISG loci and attenuates ISG expression in response to microbial stimuli. Mechanistically, we show that CHROMR sequesters the interferon regulatory factor (IRF)-2-dependent transcriptional corepressor IRF2BP2, thereby licensing IRF-dependent signaling and transcription of the ISG network. Consequently, CHROMR expression is essential to restrict viral infection of macrophages. Our findings identify CHROMR as a key arbitrator of antiviral innate immune signaling in humans.


Subject(s)
COVID-19 , DNA-Binding Proteins , Immunity, Innate , Influenza A virus , Influenza, Human , RNA, Long Noncoding , SARS-CoV-2 , Transcription Factors , COVID-19/genetics , COVID-19/immunology , DNA-Binding Proteins/metabolism , Humans , Immunity, Innate/genetics , Influenza A virus/immunology , Influenza, Human/genetics , Influenza, Human/immunology , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/physiology , SARS-CoV-2/immunology , Transcription Factors/metabolism
10.
Nat Immunol ; 13(2): 136-43, 2012 Jan 08.
Article in English | MEDLINE | ID: mdl-22231519

ABSTRACT

Atherosclerotic plaque formation is fueled by the persistence of lipid-laden macrophages in the artery wall. The mechanisms by which these cells become trapped, thereby establishing chronic inflammation, remain unknown. Here we found that netrin-1, a neuroimmune guidance cue, was secreted by macrophages in human and mouse atheroma, where it inactivated the migration of macrophages toward chemokines linked to their egress from plaques. Acting via its receptor, UNC5b, netrin-1 inhibited the migration of macrophages directed by the chemokines CCL2 and CCL19, activation of the actin-remodeling GTPase Rac1 and actin polymerization. Targeted deletion of netrin-1 in macrophages resulted in much less atherosclerosis in mice deficient in the receptor for low-density lipoprotein and promoted the emigration of macrophages from plaques. Thus, netrin-1 promoted atherosclerosis by retaining macrophages in the artery wall. Our results establish a causative role for negative regulators of leukocyte migration in chronic inflammation.


Subject(s)
Atherosclerosis/immunology , Cell Movement/immunology , Macrophages/immunology , Nerve Growth Factors/metabolism , Plaque, Atherosclerotic/immunology , Tumor Suppressor Proteins/metabolism , Actins/metabolism , Animals , Cells, Cultured , Chemokine CCL19/metabolism , Chemokine CCL2/metabolism , Chimera/metabolism , Gene Deletion , Humans , Mice , Nerve Growth Factors/genetics , Netrin Receptors , Netrin-1 , Neuropeptides/metabolism , Polymerization , Receptors, Cell Surface/metabolism , Tumor Suppressor Proteins/genetics , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein/metabolism
13.
J Mol Cell Cardiol ; 163: 1-8, 2022 02.
Article in English | MEDLINE | ID: mdl-34582824

ABSTRACT

The field of cardio-oncology has emerged in response to the increased risk of cardiovascular disease (CVD) in patients with cancer. However, recent studies suggest a more complicated CVD-cancer relationship, wherein development of CVD, either prior to or following a cancer diagnosis, can also lead to increased risk of cancer and worse outcomes for patients. In this review, we describe the current evidence base, across epidemiological as well as preclinical studies, which supports the emerging concept of 'reverse-cardio oncology', or CVD-induced acceleration of cancer pathogenesis.


Subject(s)
Cardiovascular Diseases , Neoplasms , Cardiovascular Diseases/complications , Humans , Medical Oncology , Neoplasms/complications
14.
Nat Immunol ; 11(2): 155-61, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20037584

ABSTRACT

In atherosclerosis and Alzheimer's disease, deposition of the altered self components oxidized low-density lipoprotein (LDL) and amyloid-beta triggers a protracted sterile inflammatory response. Although chronic stimulation of the innate immune system is believed to underlie the pathology of these diseases, the molecular mechanisms of activation remain unclear. Here we show that oxidized LDL and amyloid-beta trigger inflammatory signaling through a heterodimer of Toll-like receptors 4 and 6. Assembly of this newly identified heterodimer is regulated by signals from the scavenger receptor CD36, a common receptor for these disparate ligands. Our results identify CD36-TLR4-TLR6 activation as a common molecular mechanism by which atherogenic lipids and amyloid-beta stimulate sterile inflammation and suggest a new model of TLR heterodimerization triggered by coreceptor signaling events.


Subject(s)
CD36 Antigens/immunology , Inflammation/immunology , Signal Transduction/immunology , Toll-Like Receptor 4/immunology , Toll-Like Receptor 6/immunology , Amyloid beta-Peptides/immunology , Animals , Atherosclerosis/immunology , Atherosclerosis/metabolism , Blotting, Western , CD36 Antigens/metabolism , Cell Line , Chemokines/biosynthesis , Chemokines/immunology , Gene Expression , Humans , Immunoprecipitation , Inflammation/metabolism , Lipoproteins, LDL/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/immunology , Microglia/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 6/metabolism
15.
Trends Immunol ; 40(3): 179-181, 2019 03.
Article in English | MEDLINE | ID: mdl-30745266

ABSTRACT

Macrophages in the heart have dual roles in injury and repair after myocardial infarction, and understanding the two sides of this coin using traditional 'bulk cell' technologies has been challenging. By combining genetic fate-mapping and single-cell transcriptomics, a new study (Nat. Immunol. 2019;20:29-39) reveals how distinct macrophage populations expand and diverge across the healthy heart and after infarction.


Subject(s)
Macrophages/immunology , Myocardial Infarction/immunology , Myocardium/pathology , Animals , Cellular Reprogramming Techniques , Humans , Mice , Single-Cell Analysis , Transcriptome , Wound Healing
16.
Immunity ; 38(4): 628-30, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-23601680

ABSTRACT

Why patients with chronic kidney disease have elevated cardiovascular risk remains elusive. In this issue of Immunity, Speer et al. (2013) show that natural modification of high density lipoprotein promotes hypertension through a Toll-like receptor-dependent mechanism.

17.
Circ Res ; 126(11): 1590-1612, 2020 05 22.
Article in English | MEDLINE | ID: mdl-32437300

ABSTRACT

Adipose tissue (AT) plays a central role in both metabolic health and pathophysiology. Its expansion in obesity results in increased mortality and morbidity, with contributions to cardiovascular disease, diabetes mellitus, fatty liver disease, and cancer. Obesity prevalence is at an all-time high and is projected to be 50% in the United States by 2030. AT is home to a large variety of immune cells, which are critical to maintain normal tissue functions. For example, γδ T cells are fundamental for AT innervation and thermogenesis, and macrophages are required for recycling of lipids released by adipocytes. The expansion of visceral white AT promotes dysregulation of its immune cell composition and likely promotes low-grade chronic inflammation, which has been proposed to be the underlying cause for the complications of obesity. Interestingly, weight loss after obesity alters the AT immune compartment, which may account for the decreased risk of developing these complications. Recent technological advancements that allow molecular investigation on a single-cell level have led to the discovery of previously unappreciated heterogeneity in many organs and tissues. In this review, we will explore the heterogeneity of immune cells within the visceral white AT and their contributions to homeostasis and pathology.


Subject(s)
Adipose Tissue, White/immunology , Leukocytes/immunology , Obesity/immunology , Adipose Tissue, White/cytology , Adipose Tissue, White/pathology , Animals , Humans , Obesity/pathology
18.
Circ Res ; 126(11): 1565-1589, 2020 05 22.
Article in English | MEDLINE | ID: mdl-32437306

ABSTRACT

The escalating problem of obesity and its multiple metabolic and cardiovascular complications threatens the health and longevity of humans throughout the world. The cause of obesity and one of its chief complications, insulin resistance, involves the participation of multiple distinct organs and cell types. From the brain to the periphery, cell-intrinsic and intercellular networks converge to stimulate and propagate increases in body mass and adiposity, as well as disturbances of insulin sensitivity. This review focuses on the roles of the cadre of innate immune cells, both those that are resident in metabolic organs and those that are recruited into these organs in response to cues elicited by stressors such as overnutrition and reduced physical activity. Beyond the typical cast of innate immune characters invoked in the mechanisms of metabolic perturbation in these settings, such as neutrophils and monocytes/macrophages, these actors are joined by bone marrow-derived cells, such as eosinophils and mast cells and the intriguing innate lymphoid cells, which are present in the circulation and in metabolic organ depots. Upon high-fat feeding or reduced physical activity, phenotypic modulation of the cast of plastic innate immune cells ensues, leading to the production of mediators that affect inflammation, lipid handling, and metabolic signaling. Furthermore, their consequent interactions with adaptive immune cells, including myriad T-cell and B-cell subsets, compound these complexities. Notably, many of these innate immune cell-elicited signals in overnutrition may be modulated by weight loss, such as that induced by bariatric surgery. Recently, exciting insights into the biology and pathobiology of these cell type-specific niches are being uncovered by state-of-the-art techniques such as single-cell RNA-sequencing. This review considers the evolution of this field of research on innate immunity in obesity and metabolic perturbation, as well as future directions.


Subject(s)
Immunity, Innate , Metabolic Syndrome/immunology , Obesity/immunology , Animals , Humans , Metabolic Syndrome/pathology , Obesity/pathology
19.
Circ Res ; 127(3): 335-353, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32336197

ABSTRACT

RATIONALE: Regression of atherosclerosis is an important clinical goal; however, the pathways that mediate the resolution of atherosclerotic inflammation and reversal of plaques are poorly understood. Regulatory T cells (Tregs) have been shown to be atheroprotective, yet the numbers of these immunosuppressive cells decrease with disease progression, and whether they contribute to atherosclerosis regression is not known. OBJECTIVE: We investigated the roles of Tregs in the resolution of atherosclerotic inflammation, tissue remodeling, and plaque contraction during atherosclerosis regression. METHODS AND RESULTS: Using multiple independent mouse models of atherosclerosis regression, we demonstrate that an increase in plaque Tregs is a common signature of regressing plaques. Single-cell RNA-sequencing of plaque immune cells revealed that unlike Tregs from progressing plaques that expressed markers of natural Tregs derived from the thymus, Tregs in regressing plaques lacked Nrp1 expression, suggesting that they are induced in the periphery during lipid-lowering therapy. To test whether Tregs are required for resolution of atherosclerotic inflammation and plaque regression, Tregs were depleted using CD25 monoclonal antibody in atherosclerotic mice during apolipoprotein B antisense oligonucleotide-mediated lipid lowering. Morphometric analyses revealed that Treg depletion blocked plaque remodeling and contraction, and impaired hallmarks of inflammation resolution, including dampening of the T helper 1 response, alternative activation of macrophages, efferocytosis, and upregulation of specialized proresolving lipid mediators. CONCLUSIONS: Our data establish essential roles for Tregs in resolving atherosclerotic cardiovascular disease and provide mechanistic insight into the pathways governing plaque remodeling and regression of disease.


Subject(s)
Aorta/metabolism , Atherosclerosis/metabolism , Macrophage Activation , Macrophages/metabolism , Plaque, Atherosclerotic , T-Lymphocytes, Regulatory/metabolism , Animals , Antibodies/pharmacology , Aorta/drug effects , Aorta/immunology , Aorta/pathology , Apolipoprotein B-100/genetics , Apolipoprotein B-100/metabolism , Atherosclerosis/drug therapy , Atherosclerosis/immunology , Atherosclerosis/pathology , Cells, Cultured , Diet, High-Fat , Disease Models, Animal , Female , Inflammation Mediators/metabolism , Interleukin-2 Receptor alpha Subunit/antagonists & inhibitors , Interleukin-2 Receptor alpha Subunit/metabolism , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/immunology , Male , Mice, Knockout, ApoE , Neuropilin-1/genetics , Neuropilin-1/metabolism , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/metabolism , Proprotein Convertase 9/genetics , Proprotein Convertase 9/metabolism , Receptors, LDL/genetics , Receptors, LDL/metabolism , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology
SELECTION OF CITATIONS
SEARCH DETAIL