Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Cell Rep ; 43(7): 114447, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38963761

ABSTRACT

Obesity and type 2 diabetes cause a loss in brown adipose tissue (BAT) activity, but the molecular mechanisms that drive BAT cell remodeling remain largely unexplored. Using a multilayered approach, we comprehensively mapped a reorganization in BAT cells. We uncovered a subset of macrophages as lipid-associated macrophages (LAMs), which were massively increased in genetic and dietary model of BAT expansion. LAMs participate in this scenario by capturing extracellular vesicles carrying damaged lipids and mitochondria released from metabolically stressed brown adipocytes. CD36 scavenger receptor drove LAM phenotype, and CD36-deficient LAMs were able to increase brown fat genes in adipocytes. LAMs released transforming growth factor ß1 (TGF-ß1), which promoted the loss of brown adipocyte identity through aldehyde dehydrogenase 1 family member A1 (Aldh1a1) induction. These findings unfold cell dynamic changes in BAT during obesity and identify LAMs as key responders to tissue metabolic stress and drivers of loss of brown adipocyte identity.

2.
Front Immunol ; 15: 1415565, 2024.
Article in English | MEDLINE | ID: mdl-38989285

ABSTRACT

How the microbiome regulates responses of systemic innate immune cells is unclear. In the present study, our purpose was to document a novel mechanism by which the microbiome mediates crosstalk with the systemic innate immune system. We have identified a family of microbiome Bacteroidota-derived lipopeptides-the serine-glycine (S/G) lipids, which are TLR2 ligands, access the systemic circulation, and regulate proinflammatory responses of splenic monocytes. To document the role of these lipids in regulating systemic immunity, we used oral gavage with an antibiotic to decrease the production of these lipids and administered exogenously purified lipids to increase the systemic level of these lipids. We found that decreasing systemic S/G lipids by decreasing microbiome Bacteroidota significantly enhanced splenic monocyte proinflammatory responses. Replenishing systemic levels of S/G lipids via exogenous administration returned splenic monocyte responses to control levels. Transcriptomic analysis demonstrated that S/G lipids regulate monocyte proinflammatory responses at the level of gene expression of a small set of upstream inhibitors of TLR and NF-κB pathways that include Trem2 and Irf4. Consistent with enhancement in proinflammatory cytokine responses, decreasing S/G lipids lowered gene expression of specific pathway inhibitors. Replenishing S/G lipids normalized gene expression of these inhibitors. In conclusion, our results suggest that microbiome-derived S/G lipids normally establish a level of buffered signaling activation necessary for well-regulated innate immune responses in systemic monocytes. By regulating gene expression of inflammatory pathway inhibitors such as Trem2, S/G lipids merit broader investigation into the potential dysfunction of other innate immune cells, such as microglia, in diseases such as Alzheimer's disease.


Subject(s)
Monocytes , Signal Transduction , Monocytes/immunology , Monocytes/metabolism , Monocytes/drug effects , Animals , Mice , Microbiota/immunology , Mice, Inbred C57BL , Immunity, Innate , Toll-Like Receptor 2/metabolism , Gene Expression Regulation/drug effects , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Lipopeptides/pharmacology , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , NF-kappa B/metabolism , Inflammation/immunology , Interferon Regulatory Factors/metabolism , Interferon Regulatory Factors/genetics , Male , Lipids , Spleen/immunology , Spleen/metabolism , Cytokines/metabolism , Female
3.
Front Immunol ; 14: 1148188, 2023.
Article in English | MEDLINE | ID: mdl-36875144

ABSTRACT

Adipose tissue macrophages (ATMs) bolster obesity-induced metabolic dysfunction and represent a targetable population to lessen obesity-associated health risks. However, ATMs also facilitate adipose tissue function through multiple actions, including adipocyte clearance, lipid scavenging and metabolism, extracellular remodeling, and supporting angiogenesis and adipogenesis. Thus, high-resolution methods are needed to capture macrophages' dynamic and multifaceted functions in adipose tissue. Herein, we review current knowledge on regulatory networks critical to macrophage plasticity and their multifaceted response in the complex adipose tissue microenvironment.


Subject(s)
Adipocytes , Adipose Tissue , Humans , Adipogenesis , Macrophages , Obesity
4.
Front Immunol ; 14: 1102403, 2023.
Article in English | MEDLINE | ID: mdl-36817480

ABSTRACT

MicroRNA-150 (miR-150) has been shown to play a general role in the immune system, but very little is known about its role on CD4+ T cell responses. During T cell responses against superantigen Staphylococcal Enterotoxin A, miR-150 expression was down-regulated in antigen-specific CD4+ T cells but up-regulated in CD8+ T cells. CD4+ and CD8+ T cell clonal expansion was greater in miR-150-KO mice than in WT mice, but miR-150 selectively repressed IL-2 production in CD4+ T cells. Transcriptome analysis of CD4+ T cells demonstrated that apoptosis and mTOR pathways were highly enriched in the absence of miR-150. Mechanistic studies confirmed that miR-150 promoted apoptosis specifically in antigen-specific CD4+ T cells, but not in bystander CD4+ nor in CD8+ T cells. Furthermore, inhibition of mTOR-linked mitochondrial superoxidedismutase-2 increased apoptosis in miR-150-/- antigen-specific CD4+ T. Thus, miR-150 impacts CD4+ T cell helper activity by attenuating IL-2 production along with clonal expansion, and suppresses superoxidedismutase to promote apoptosis.


Subject(s)
CD4-Positive T-Lymphocytes , MicroRNAs , Mice , Animals , CD8-Positive T-Lymphocytes , Interleukin-2/metabolism , Down-Regulation , Cell Survival , TOR Serine-Threonine Kinases/metabolism , MicroRNAs/metabolism , Superoxide Dismutase/metabolism
5.
Trends Cell Biol ; 33(6): 517-529, 2023 06.
Article in English | MEDLINE | ID: mdl-36272883

ABSTRACT

Brown adipose tissue (BAT) controls mammalian core body temperature by non-shivering thermogenesis. BAT is extraordinarily rich in mitochondria, which have the peculiarity of generating heat by uncoupled respiration. Since the mitochondrial activity of BAT is subject to cycles of activation and deactivation in response to environmental temperature changes, an integrated mitochondrial quality control (MQC) system is of fundamental importance to ensure BAT physiology. Here, we provide an overview of the conventional and alternative mechanisms through which thermogenic adipocytes selectively remove damaged parts of mitochondria and how macrophages participate in the MQC system by removing extracellular mitochondrial waste to maintain the thermogenic function of BAT.


Subject(s)
Adipose Tissue, Brown , Mitochondria , Humans , Animals , Adipose Tissue, Brown/metabolism , Adipocytes/metabolism , Mammals
6.
Genes (Basel) ; 13(11)2022 11 09.
Article in English | MEDLINE | ID: mdl-36360310

ABSTRACT

Macrophages are central players in systemic inflammation associated with obesity and aging, termed meta-inflammation and inflammaging. Activities of macrophages elicited by the two chronic conditions display shared and distinct patterns mechanistically, resulting in multifaceted actions for their pathogenic roles. Drastically expanded tissue macrophage populations under obesity and aging stress attribute to both enhanced recruitment and local expansion. Importantly, molecular networks governing the multifaceted actions of macrophages are directly altered by environmental cues and subsequently contribute to metabolic reprogramming, resulting in meta-inflammation in obesity or inflammaging in aging. In this review, we will summarize how meta-inflammation and inflammaging affect macrophages and the molecular mechanisms involved in these processes.


Subject(s)
Inflammation , Macrophages , Humans , Inflammation/metabolism , Macrophages/metabolism , Aging/genetics , Obesity/genetics , Obesity/metabolism , Leukocyte Count
7.
Genes (Basel) ; 13(10)2022 10 15.
Article in English | MEDLINE | ID: mdl-36292751

ABSTRACT

Obesity is a growing health problem that affects both children and adults. The increasing prevalence of childhood obesity is associated with comorbidities such as cardiovascular disease, type 2 diabetes and metabolic syndrome due to chronic low-grade inflammation present at early stages of the disease. In pediatric patients suffering from obesity, the role of epigenetics, the gut microbiome and intrauterine environment have emerged as causative factors Interestingly, pediatric obesity is strongly associated with low birth weight. Accelerated weight gain oftentimes occurs in these individuals during the post-natal period, which can lead to increased risk of adiposity and metabolic disease. The pathophysiology of obesity is complex and involves biological and physiological factors compounded by societal factors such as family and community. On a cellular level, adipocytes contained within adipose tissue become dysregulated and further contribute to development of comorbidities similar to those present in adults with obesity. This review provides an overview of the current understanding of adipose tissue immune, inflammatory and metabolic adaptation of the adipose tissue in obesity. Early cellular changes as well as the role of immune cells and inflammation on the progression of disease in pivotal pediatric clinical trials, adult studies and mouse models are emphasized. Understanding the initial molecular and cellular changes that occur during obesity can facilitate new and improved treatments aimed at early intervention and subsequent prevention of adulthood comorbidities.


Subject(s)
Diabetes Mellitus, Type 2 , Pediatric Obesity , Pediatrics , Mice , Animals , Child , Humans , Diabetes Mellitus, Type 2/metabolism , Pediatric Obesity/epidemiology , Pediatric Obesity/genetics , Adipose Tissue/metabolism , Inflammation/metabolism
8.
Immunometabolism (Cobham) ; 4(3): e00005, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35966635

ABSTRACT

Obesity is a prevalent health risk by inducing chronic, low-grade inflammation and insulin resistance, in part from adipose tissue inflammation perpetuated by activated B cells and other resident immune cells. However, regulatory mechanisms controlling B-cell actions in adipose tissue remain poorly understood, limiting therapeutic innovations. MicroRNAs are potent regulators of immune cell dynamics through fine-tuning a network of downstream genes in multiple signaling pathways. In particular, miR-150 is crucial to B-cell development and suppresses obesity-associated inflammation via regulating adipose tissue B-cell function. Herein, we review the effect of microRNAs on B-cell development, activation, and function and highlight miR-150-regulated B-cell actions during obesity which modulate systemic inflammation and insulin resistance. In this way, we hope to promote translational discoveries that mitigate obesity-induced health risks by targeting microRNA-regulated B-cell actions.

9.
J Leukoc Biol ; 112(6): 1535-1542, 2022 12.
Article in English | MEDLINE | ID: mdl-35726704

ABSTRACT

Macrophages are widely distributed immune cells that play central roles in a variety of physiologic and pathologic processes, including obesity and cardiovascular disease (CVD). They are highly plastic cells that execute diverse functions according to a combination of signaling and environmental cues. While macrophages have traditionally been understood to polarize to either proinflammatory M1-like or anti-inflammatory M2-like states, evidence has shown that they exist in a spectrum of states between those 2 phenotypic extremes. In obesity-related disease, M1-like macrophages exacerbate inflammation and promote insulin resistance, while M2-like macrophages reduce inflammation, promoting insulin sensitivity. However, polarization markers are expressed inconsistently in adipose tissue macrophages, and they additionally exhibit phenotypes differing from the M1/M2 paradigm. In atherosclerotic CVD, activated plaque macrophages can also exist in a range of proinflammatory or anti-inflammatory states. Some of these macrophages scavenge lipids, developing into heterogeneous foam cell populations. To better characterize the many actions of macrophages in human disease, we have designed a novel set of computational tools: MacSpectrum and AtheroSpectrum. These tools provide information on the inflammatory polarization status, differentiation, and foaming of macrophages in both human and mouse samples, allowing for better characterization of macrophage subpopulations based on their function. Using these tools, we identified disease-relevant cell states in obesity and CVD, including the novel concept that macrophage-derived foam cell formation can follow homeostatic noninflammatory or pathogenic inflammatory foaming programs.


Subject(s)
Atherosclerosis , Insulin Resistance , Humans , Mice , Animals , Macrophages , Inflammation/pathology , Obesity , Anti-Inflammatory Agents
10.
Cells ; 11(8)2022 04 14.
Article in English | MEDLINE | ID: mdl-35456015

ABSTRACT

Obesity-induced adipose tissue dysfunction is bolstered by chronic, low-grade inflammation and impairs systemic metabolic health. Adipose tissue macrophages (ATMs) perpetuate local inflammation but are crucial to adipose tissue homeostasis, exerting heterogeneous, niche-specific functions. Diversified macrophage actions are shaped through finely regulated factors, including microRNAs, which post-transcriptionally alter macrophage activation. Numerous studies have highlighted microRNAs' importance to immune function and potential as inflammation-modulatory. This review summarizes current knowledge of regulatory networks governed by microRNAs in ATMs in white adipose tissue under obesity stress.


Subject(s)
MicroRNAs , Adipose Tissue/metabolism , Humans , Inflammation/metabolism , Macrophages/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Obesity/metabolism
11.
Cell Metab ; 34(4): 533-548.e12, 2022 04 05.
Article in English | MEDLINE | ID: mdl-35305295

ABSTRACT

Recent findings have demonstrated that mitochondria can be transferred between cells to control metabolic homeostasis. Although the mitochondria of brown adipocytes comprise a large component of the cell volume and undergo reorganization to sustain thermogenesis, it remains unclear whether an intercellular mitochondrial transfer occurs in brown adipose tissue (BAT) and regulates adaptive thermogenesis. Herein, we demonstrated that thermogenically stressed brown adipocytes release extracellular vesicles (EVs) that contain oxidatively damaged mitochondrial parts to avoid failure of the thermogenic program. When re-uptaken by parental brown adipocytes, mitochondria-derived EVs reduced peroxisome proliferator-activated receptor-γ signaling and the levels of mitochondrial proteins, including UCP1. Their removal via the phagocytic activity of BAT-resident macrophages is instrumental in preserving BAT physiology. Depletion of macrophages in vivo causes the abnormal accumulation of extracellular mitochondrial vesicles in BAT, impairing the thermogenic response to cold exposure. These findings reveal a homeostatic role of tissue-resident macrophages in the mitochondrial quality control of BAT.


Subject(s)
Adipose Tissue, Brown , Thermogenesis , Adipocytes, Brown/metabolism , Adipose Tissue, Brown/metabolism , Macrophages/metabolism , Mitochondria/metabolism , Thermogenesis/physiology , Uncoupling Protein 1/metabolism
12.
ExRNA ; 42022 Aug 30.
Article in English | MEDLINE | ID: mdl-36866026

ABSTRACT

Background and Objective: Obesity affects hundreds of millions of people worldwide and is characterized by chronic inflammation and insulin resistance, leading to Type II diabetes and atherosclerotic cardiovascular disease. Extracellular RNAs (exRNAs) are among the components which effect immune actions under obese conditions, and technological advances in recent years have rapidly increased our understanding of their roles and functions. Here we review essential background information on exRNAs and vesicles as well as the impact of immune-derived exRNAs in obesity-related disease. We also offer perspectives on clinical applications of exRNAs and future research directions. Methods: We searched PubMed for articles relevant to immune-derived exRNAs in obesity. Articles written in English and published prior to May 25, 2022 were included. Key Content and Findings: We report findings on the roles of immune-derived exRNAs which are important in obesity-related disease. We also highlight several exRNAs derived from other cell lineages which act on immune cells in metabolic disease. Conclusions: ExRNAs produced by immune cells have profound local and systemic effects under obese conditions and can impact metabolic disease phenotypes. Immune-derived exRNAs represent an important target for future research and therapy.

13.
Circulation ; 145(3): 206-218, 2022 01 18.
Article in English | MEDLINE | ID: mdl-34913723

ABSTRACT

BACKGROUND: Whereas several interventions can effectively lower lipid levels in people at risk for atherosclerotic cardiovascular disease (ASCVD), cardiovascular event risks remain, suggesting an unmet medical need to identify factors contributing to cardiovascular event risk. Monocytes and macrophages play central roles in atherosclerosis, but studies have yet to provide a detailed view of macrophage populations involved in increased ASCVD risk. METHODS: A novel macrophage foaming analytics tool, AtheroSpectrum, was developed using 2 quantitative indices depicting lipid metabolism and the inflammatory status of macrophages. A machine learning algorithm was developed to analyze gene expression patterns in the peripheral monocyte transcriptome of MESA participants (Multi-Ethnic Study of Atherosclerosis; set 1; n=911). A list of 30 genes was generated and integrated with traditional risk factors to create an ASCVD risk prediction model (30-gene cardiovascular disease risk score [CR-30]), which was subsequently validated in the remaining MESA participants (set 2; n=228); performance of CR-30 was also tested in 2 independent human atherosclerotic tissue transcriptome data sets (GTEx [Genotype-Tissue Expression] and GSE43292). RESULTS: Using single-cell transcriptomic profiles (GSE97310, GSE116240, GSE97941, and FR-FCM-Z23S), AtheroSpectrum detected 2 distinct programs in plaque macrophages-homeostatic foaming and inflammatory pathogenic foaming-the latter of which was positively associated with severity of atherosclerosis in multiple studies. A pool of 2209 pathogenic foaming genes was extracted and screened to select a subset of 30 genes correlated with cardiovascular event in MESA set 1. A cardiovascular disease risk score model (CR-30) was then developed by incorporating this gene set with traditional variables sensitive to cardiovascular event in MESA set 1 after cross-validation generalizability analysis. The performance of CR-30 was then tested in MESA set 2 (P=2.60×10-4; area under the receiver operating characteristic curve, 0.742) and 2 independent data sets (GTEx: P=7.32×10-17; area under the receiver operating characteristic curve, 0.664; GSE43292: P=7.04×10-2; area under the receiver operating characteristic curve, 0.633). Model sensitivity tests confirmed the contribution of the 30-gene panel to the prediction model (likelihood ratio test; df=31, P=0.03). CONCLUSIONS: Our novel computational program (AtheroSpectrum) identified a specific gene expression profile associated with inflammatory macrophage foam cells. A subset of 30 genes expressed in circulating monocytes jointly contributed to prediction of symptomatic atherosclerotic vascular disease. Incorporating a pathogenic foaming gene set with known risk factors can significantly strengthen the power to predict ASCVD risk. Our programs may facilitate both mechanistic investigations and development of therapeutic and prognostic strategies for ASCVD risk.


Subject(s)
Atherosclerosis/therapy , Cardiovascular Diseases/therapy , Foam Cells/cytology , Macrophages/cytology , Aged , Aged, 80 and over , Atherosclerosis/etiology , Atherosclerosis/genetics , Cardiovascular Diseases/complications , Coronary Artery Disease/complications , Coronary Artery Disease/genetics , Coronary Artery Disease/therapy , Female , Humans , Male , Middle Aged , Plaque, Atherosclerotic/complications , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/therapy , ROC Curve , Risk , Vascular Calcification/complications , Vascular Calcification/genetics , Vascular Calcification/therapy
14.
J Exp Med ; 219(1)2022 01 03.
Article in English | MEDLINE | ID: mdl-34807232

ABSTRACT

Immune checkpoint inhibitor (ICI) therapy continues to revolutionize melanoma treatment, but only a subset of patients respond. Major efforts are underway to develop minimally invasive predictive assays of ICI response. Using single-cell transcriptomics, we discovered a unique CD8 T cell blood/tumor-shared subpopulation in melanoma patients with high levels of oxidative phosphorylation (OXPHOS), the ectonucleotidases CD38 and CD39, and both exhaustion and cytotoxicity markers. We called this population with high levels of OXPHOS "CD8+ TOXPHOS cells." We validated that higher levels of OXPHOS in tumor- and peripheral blood-derived CD8+ TOXPHOS cells correlated with ICI resistance in melanoma patients. We then developed an ICI therapy response predictive model using a transcriptomic profile of CD8+ TOXPHOS cells. This model is capable of discerning responders from nonresponders using either tumor or peripheral blood CD8 T cells with high accuracy in multiple validation cohorts. In sum, CD8+ TOXPHOS cells represent a critical immune population to assess ICI response with the potential to be a new target to improve outcomes in melanoma patients.


Subject(s)
CD8-Positive T-Lymphocytes/drug effects , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy/methods , Melanoma/therapy , Oxidative Phosphorylation/drug effects , T-Lymphocyte Subsets/drug effects , Adult , Aged , Algorithms , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/immunology , Female , Gene Expression Profiling/methods , Humans , Immune Checkpoint Inhibitors/immunology , Male , Melanoma/genetics , Melanoma/immunology , Middle Aged , Models, Genetic , Outcome Assessment, Health Care/methods , RNA-Seq/methods , Single-Cell Analysis/methods , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
15.
Int Immunopharmacol ; 99: 107980, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34298397

ABSTRACT

IF1 is a mitochondrial protein involved in the regulation of ATP synthase activity. The role of IF1 remains to be established in inflammatory bowel diseases (IBD). In this study, we report that IF1 gene inactivation generated protection against IBD in the dextran sodium sulfate (DSS) model. IF1 gene knockout (IF1-KO) mice developed less severe colitis than the wild type (WT) mice as judged by parameters including disease activity index (DAI), body weight loss, inflammatory cytokines, leukocyte infiltration and bacterial invasion in the colon tissue. The intestinal barrier integrity was protected in the colon tissue of IF1-KO mice through a reduction in apoptosis and inflammasomal activity. The protection was abolished in the KO mice after substitution of the immune cells with the wild type cells following bone marrow transplantation. Depletion of neutrophils with anti-Gr-1 antibody abolished the protection from colitis in IF1-KO mice. Neutrophil number was decreased in the peripheral blood of IF1-KO mice, which was associated with a reduction in LC3A/B proteins in the KO neutrophils in Rapamycin-induced autophagy response. Inhibition of autophagy with the lysosome inhibitor Chloroquine (CQ) decreased the absolute number of neutrophils in WT mice and protected the mice from colitis. Taken together, these findings suggest that IF1 may contribute to the pathogenesis of IBD through acceleration of neutrophil autophagy. The activity is attenuated in the IF1-KO mice through reduction of autophagy in neutrophils leading to resistance to IBD.


Subject(s)
Colitis/genetics , Intestinal Mucosa/cytology , Mitochondrial Proteins/genetics , Neutrophil Infiltration/genetics , Animals , Apoptosis/drug effects , Autophagy/drug effects , Chloroquine/pharmacology , Colitis/chemically induced , Colitis/drug therapy , Colon/microbiology , Cytokines/blood , Dextran Sulfate , Down-Regulation/drug effects , Female , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Lysosomes/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration/drug effects , Neutrophils/drug effects , Weight Loss/drug effects
16.
Nat Immunol ; 22(2): 154-165, 2021 02.
Article in English | MEDLINE | ID: mdl-33398185

ABSTRACT

Inflammatory caspase sensing of cytosolic lipopolysaccharide (LPS) triggers pyroptosis and the concurrent release of damage-associated molecular patterns (DAMPs). Collectively, DAMPs are key determinants that shape the aftermath of inflammatory cell death. However, the identity and function of the individual DAMPs released are poorly defined. Our proteomics study revealed that cytosolic LPS sensing triggered the release of galectin-1, a ß-galactoside-binding lectin. Galectin-1 release is a common feature of inflammatory cell death, including necroptosis. In vivo studies using galectin-1-deficient mice, recombinant galectin-1 and galectin-1-neutralizing antibody showed that galectin-1 promotes inflammation and plays a detrimental role in LPS-induced lethality. Mechanistically, galectin-1 inhibition of CD45 (Ptprc) underlies its unfavorable role in endotoxin shock. Finally, we found increased galectin-1 in sera from human patients with sepsis. Overall, we uncovered galectin-1 as a bona fide DAMP released as a consequence of cytosolic LPS sensing, identifying a new outcome of inflammatory cell death.


Subject(s)
Alarmins/metabolism , Endotoxemia/immunology , Galectin 1/metabolism , Inflammation Mediators/metabolism , Inflammation/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Macrophages/metabolism , Phosphate-Binding Proteins/metabolism , Adult , Aged , Aged, 80 and over , Alarmins/deficiency , Alarmins/genetics , Animals , Case-Control Studies , Disease Models, Animal , Endotoxemia/chemically induced , Endotoxemia/metabolism , Endotoxemia/pathology , Female , Galectin 1/blood , Galectin 1/deficiency , Galectin 1/genetics , HeLa Cells , Humans , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Leukocyte Common Antigens/metabolism , Lipopolysaccharides , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Necroptosis , Phosphate-Binding Proteins/deficiency , Phosphate-Binding Proteins/genetics , RAW 264.7 Cells , Sepsis/blood , Sepsis/diagnosis , Signal Transduction , Up-Regulation
17.
Article in English | MEDLINE | ID: mdl-32973073

ABSTRACT

INTRODUCTION: Diabetic retinopathy (DR) is the leading cause of blindness among the working population in the USA. Current therapies, including anti-vascular endothelial growth factor treatments, cannot completely reverse the visual defects induced by DR. MicroRNA-150 (miR-150) is a regulator that suppresses inflammation and pathological angiogenesis. In patients with diabetes, miR-150 is downregulated. As chronic inflammation is a major contributor to the pathogenesis of DR, whether diabetes-associated decrease of miR-150 is merely associated with the disease progression or decreased miR-150 causes retinal inflammation and pathological angiogenesis is still unknown. RESEARCH DESIGN AND METHODS: We used high-fat diet (HFD)-induced type 2 diabetes (T2D) in wild type (WT) and miR-150 knockout (miR-150-/-) mice for this study and compared retinal function and microvasculature morphology. RESULTS: We found that WT mice fed with an HFD for only 1 month had a significant decrease of miR-150 in the blood and retina, and retinal light sensitivity also decreased. The miR-150-/- mice on the HFD developed diabetes similar to that of the WT. At 7-8 months old, miR-150-/- mice under normal diet had increased degeneration of retinal capillaries compared with WT mice, indicating that miR-150 is important in maintaining the structural integrity of retinal microvasculature. Deletion of miR-150 worsened HFD-induced retinal dysfunction as early as 1 month after the diet regimen, and it exacerbated HFD-induced T2DR by further increasing retinal inflammation and microvascular degeneration. CONCLUSION: These data suggest that decreased miR-150 caused by obesity or diabetic insults is not merely correlated to the disease progression, but it contributes to the retinal dysfunction and inflammation, as well as the development of DR.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , MicroRNAs , Animals , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Inflammation/genetics , Mice , Mice, Obese , MicroRNAs/genetics , Obesity/complications , Obesity/genetics
18.
Proc Natl Acad Sci U S A ; 117(10): 5430-5441, 2020 03 10.
Article in English | MEDLINE | ID: mdl-32094172

ABSTRACT

Multiple sclerosis (MS) is an autoimmune inflammatory demyelinating disease of the central nervous system. Dysregulation of STAT3, a transcription factor pivotal to various cellular processes including Th17 cell differentiation, has been implicated in MS. Here, we report that STAT3 is activated in infiltrating monocytic cells near active MS lesions and that activation of STAT3 in myeloid cells is essential for leukocyte infiltration, neuroinflammation, and demyelination in experimental autoimmune encephalomyelitis (EAE). Genetic disruption of Stat3 in peripheral myeloid lineage cells abrogated EAE, which was associated with decreased antigen-specific T helper cell responses. Myeloid cells from immunized Stat3 mutant mice exhibited impaired antigen-presenting functions and were ineffective in driving encephalitogenic T cell differentiation. Single-cell transcriptome analyses of myeloid lineage cells from preclinical wild-type and mutant mice revealed that loss of myeloid STAT3 signaling disrupted antigen-dependent cross-activation of myeloid cells and T helper cells. This study identifies a previously unrecognized requisite for myeloid cell STAT3 in the activation of myelin-reactive T cells and suggests myeloid STAT3 as a potential therapeutic target for autoimmune demyelinating disease.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Lymphocyte Activation , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Myeloid Cells/immunology , STAT3 Transcription Factor/metabolism , T-Lymphocyte Subsets/immunology , Animals , CD11b Antigen/analysis , Cell Differentiation , Encephalomyelitis, Autoimmune, Experimental/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Multiple Sclerosis/genetics , STAT3 Transcription Factor/genetics , Single-Cell Analysis , Transcriptome
19.
Mol Cell ; 75(3): 644-660.e5, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31398325

ABSTRACT

Cell-cell communication via ligand-receptor signaling is a fundamental feature of complex organs. Despite this, the global landscape of intercellular signaling in mammalian liver has not been elucidated. Here we perform single-cell RNA sequencing on non-parenchymal cells isolated from healthy and NASH mouse livers. Secretome gene analysis revealed a highly connected network of intrahepatic signaling and disruption of vascular signaling in NASH. We uncovered the emergence of NASH-associated macrophages (NAMs), which are marked by high expression of triggering receptors expressed on myeloid cells 2 (Trem2), as a feature of mouse and human NASH that is linked to disease severity and highly responsive to pharmacological and dietary interventions. Finally, hepatic stellate cells (HSCs) serve as a hub of intrahepatic signaling via HSC-derived stellakines and their responsiveness to vasoactive hormones. These results provide unprecedented insights into the landscape of intercellular crosstalk and reprogramming of liver cells in health and disease.


Subject(s)
Cell Communication/genetics , Liver/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Sequence Analysis, RNA , Animals , Cellular Reprogramming/genetics , Disease Models, Animal , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Humans , Ligands , Liver/pathology , Macrophages/metabolism , Macrophages/pathology , Mice , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Signal Transduction/genetics , Single-Cell Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...