Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
J Immunol ; 197(9): 3650-3661, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27683748

RESUMO

Dynamic changes of adipose tissue leukocytes, including adipose tissue macrophage (ATM) and adipose tissue dendritic cells (ATDCs), contribute to obesity-induced inflammation and metabolic disease. However, clear discrimination between ATDC and ATM in adipose tissue has limited progress in the field of immunometabolism. In this study, we use CD64 to distinguish ATM and ATDC, and investigated the temporal and functional changes in these myeloid populations during obesity. Flow cytometry and immunostaining demonstrated that the definition of ATM as F4/80+CD11b+ cells overlaps with other leukocytes and that CD45+CD64+ is specific for ATM. The expression of core dendritic cell genes was enriched in CD11c+CD64- cells (ATDC), whereas core macrophage genes were enriched in CD45+CD64+ cells (ATM). CD11c+CD64- ATDCs expressed MHC class II and costimulatory receptors, and had similar capacity to stimulate CD4+ T cell proliferation as ATMs. ATDCs were predominantly CD11b+ conventional dendritic cells and made up the bulk of CD11c+ cells in adipose tissue with moderate high-fat diet exposure. Mixed chimeric experiments with Ccr2-/- mice demonstrated that high-fat diet-induced ATM accumulation from monocytes was dependent on CCR2, whereas ATDC accumulation was less CCR2 dependent. ATDC accumulation during obesity was attenuated in Ccr7-/- mice and was associated with decreased adipose tissue inflammation and insulin resistance. CD45+CD64+ ATM and CD45+CD64-CD11c+ ATDCs were identified in human obese adipose tissue and ATDCs were increased in s.c. adipose tissue compared with omental adipose tissue. These results support a revised strategy for unambiguous delineation of ATM and ATDC, and suggest that ATDCs are independent contributors to adipose tissue inflammation during obesity.


Assuntos
Tecido Adiposo/imunologia , Células Dendríticas/imunologia , Inflamação/imunologia , Macrófagos/imunologia , Obesidade/imunologia , Animais , Células Cultivadas , Dieta Hiperlipídica , Perfilação da Expressão Gênica , Humanos , Imunofenotipagem , Resistência à Insulina , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores CCR2/genética , Receptores CCR7/genética , Receptores de IgG/metabolismo
2.
J Immunol Res ; 2024: 2506586, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38974097

RESUMO

Elevated infant fecal concentrations of the bacterial-derived lipid 12,13-dihydroxy-9Z-octadecenoic acid (12,13-diHOME) increase the risk for childhood atopy and asthma. However, the mechanisms by which this lipid contributes to disease development are largely unknown. We hypothesized that macrophages, which are key to both antimicrobial and antigen responses, are functionally and epigenetically modified by 12,13-diHOME leading to short- and long-term dysfunction with consequences for both antimicrobial and antigenic responses. Macrophages exposed to 12,13-diHOME are skewed toward inflammatory IL-1ß highCD206low cells, a phenomenon that is further amplified in the presence of common microbial-, aero-, and food-allergens. These IL-1ß highCD206low macrophages also exhibit reduced bacterial phagocytic capacity. In primary immune cell coculture assays involving peanut allergen stimulation, 12,13-diHOME promotes both IL-1ß and IL-6 production, memory B cell expansion, and increased IgE production. Exposure to 12,13-diHOME also induces macrophage chromatin remodeling, specifically diminishing access to interferon-stimulated response elements resulting in reduced interferon-regulated gene expression upon bacterial lipopolysaccharide stimulation. Thus 12,13-diHOME reprograms macrophage effector function, B-cell interactions and promotes epigenetic modifications that exacerbate inflammatory response to allergens and mutes antimicrobial response along the interferon axis. These observations offer plausible mechanisms by which this lipid promotes early-life pathogenic microbiome development and innate immune dysfunction associated with childhood allergic sensitization.


Assuntos
Alérgenos , Epigênese Genética , Macrófagos , Macrófagos/imunologia , Macrófagos/metabolismo , Alérgenos/imunologia , Humanos , Interleucina-1beta/metabolismo , Células Cultivadas , Linfócitos B/imunologia , Linfócitos B/metabolismo , Inflamação/imunologia , Imunoglobulina E/imunologia , Animais
3.
Gut Microbes ; 16(1): 2290661, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38117587

RESUMO

Early life gut microbiome composition has been correlated with childhood obesity, though microbial functional contributions to disease origins remain unclear. Here, using an infant birth cohort (n = 349) we identify a distinct fecal microbiota composition in 1-month-old infants with the lowest rate of exclusive breastfeeding, that relates with higher relative risk for obesity and overweight phenotypes at two years. Higher-risk infant fecal microbiomes exhibited accelerated taxonomic and functional maturation and broad-ranging metabolic reprogramming, including reduced concentrations of neuro-endocrine signals. In vitro, exposure of enterocytes to fecal extracts from higher-risk infants led to upregulation of genes associated with obesity and with expansion of nutrient sensing enteroendocrine progenitor cells. Fecal extracts from higher-risk infants also promoted enterocyte barrier dysfunction. These data implicate dysregulation of infant microbiome functional development, and more specifically promotion of enteroendocrine signaling and epithelial barrier impairment in the early-life developmental origins of childhood obesity.


Assuntos
Microbioma Gastrointestinal , Microbiota , Obesidade Infantil , Lactente , Humanos , Criança , Enterócitos , Microbioma Gastrointestinal/fisiologia , Fezes
4.
JCI Insight ; 6(8)2021 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-33724954

RESUMO

Despite studies implicating adipose tissue T cells (ATT) in the initiation and persistence of adipose tissue inflammation, fundamental gaps in knowledge regarding ATT function impedes progress toward understanding how obesity influences adaptive immunity. We hypothesized that ATT activation and function would have tissue-resident-specific properties and that obesity would potentiate their inflammatory properties. We assessed ATT activation and inflammatory potential within mouse and human stromal vascular fraction (SVF). Surprisingly, murine and human ATTs from obese visceral white adipose tissue exhibited impaired inflammatory characteristics upon stimulation. Both environmental and cell-intrinsic factors are implicated in ATT dysfunction. Soluble factors from obese SVF inhibit ATT activation. Additionally, chronic signaling from macrophage major histocompatibility complex II (MHCII) is necessary for ATT impairment in obese adipose tissue but is independent of increased PD1 expression. To assess intracellular signaling mechanisms responsible for ATT inflammation impairments, single-cell RNA sequencing of ATTs was performed. ATTs in obese adipose tissue exhibit enrichment of genes characteristic of T cell exhaustion and increased expression of coinhibitory receptor Btla. In sum, this work suggests that obesity-induced ATTs have functional characteristics and gene expression resembling T cell exhaustion induced by local soluble factors and cell-to-cell interactions in adipose tissue.


Assuntos
Imunidade Adaptativa/imunologia , Tecido Adiposo/imunologia , Gordura Intra-Abdominal/imunologia , Obesidade/imunologia , Linfócitos T/imunologia , Tecido Adiposo Branco/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Macrófagos/imunologia , Masculino , Camundongos , Pessoa de Meia-Idade , Receptor de Morte Celular Programada 1/imunologia , Receptores Imunológicos/imunologia , Subpopulações de Linfócitos T/imunologia
5.
Mol Cell Endocrinol ; 505: 110740, 2020 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-31987897

RESUMO

Adipose tissue derived chronic inflammation is a critical component of obesity induced type II diabetes. Major histocompatibility complex II (MHCII) mediated T cell activation within adipose tissue is one mechanism that contributes to this phenotype. However, the contribution of dendritic cells as professional antigen presenting cells in adipose issue has not previously been explored. Using ItgaxCre x MHCIIfl/fl (M11cKO) mice we observed adipose tissue specific changes in adipose tissue leukocytes. While there was a complete knockout of MHCII in dendritic cells, MHCII was also absent on the majority of macrophages. This resulted in reduction of TCR expression in CD4+ T cells in obese adipose tissue, and an increase in CD8+ and CD4+ CD8+ double positive T cells with decreased CD4+ T cells independent of diet type. Increased CD8+ cells were not observed in the spleen, suggesting adipose tissue T cell regulation is tissue specific. In vitro studies demonstrated more potent antigen presentation function in adipose tissue dendritic cells compared to macrophages. Obese M11cKO mice had decreased CD11c+ adipose tissue macrophages. Despite the changes of immune cellularity in adipose tissue, M11cKO largely did not change inflammatory gene expression in adipose tissue and did not demonstrate differences in glucose and insulin intolerance. Overall MHCII expression on CD11c+ cells is important for maintaining CD4+ and CD8+ adipose tissue T cells, but these cellular changes fail to alter inflammatory output and systemic metabolism.


Assuntos
Tecido Adiposo/patologia , Células Dendríticas/patologia , Homeostase , Obesidade/imunologia , Transdução de Sinais , Linfócitos T/imunologia , Animais , Apresentação de Antígeno/imunologia , Antígenos CD11/metabolismo , Proliferação de Células , Regulação da Expressão Gênica , Glucose/metabolismo , Antígenos de Histocompatibilidade Classe II/metabolismo , Inflamação/genética , Inflamação/patologia , Resistência à Insulina , Macrófagos/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Baço/patologia
6.
Front Physiol ; 11: 903, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32848853

RESUMO

Adipose tissue pathology in obese patients often features impaired adipogenesis, angiogenesis, and chronic low-grade inflammation, all of which are regulated in large part by adipose tissue stromal vascular cells [SVC; i.e., non-adipocyte cells within adipose tissue including preadipocytes, endothelial cells (ECs), and immune cells]. Exercise is known to increase subcutaneous adipose tissue lipolysis, but the impact of exercise on SVCs in adipose tissue has not been explored. The purpose of this study was to assess the effects of a session of exercise on preadipocyte, EC, macrophage, and T cell content in human subcutaneous adipose tissue. We collected abdominal subcutaneous adipose tissue samples from 10 obese adults (BMI 33 ± 3 kg/m2, body fat 41 ± 7%) 12 h after a 60 min acute session of endurance exercise (80 ± 3%HRpeak) vs. no acute exercise session. SVCs were isolated by collagenase digestion and stained for flow cytometry. We found that acute exercise reduced preadipocyte content (38 ± 7 vs. 30 ± 13%SVC; p = 0.04). The reduction was driven by a decrease in CD34hi preadipocytes (18 ± 5 vs. 13 ± 6%SVC; p = 0.002), a subset of preadipocytes that generates high lipolytic rate adipocytes ex vivo. Acute exercise did not alter EC content. Acute exercise also did not change total immune cell, macrophage, or T cell content, and future work should assess the effects of exercise on subpopulations of these cells. We conclude that exercise may rapidly regulate the subcutaneous adipose tissue preadipocyte pool in ways that may help attenuate the high lipolytic rates that are commonly found in obesity.

7.
Obesity (Silver Spring) ; 28(6): 1086-1097, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32281747

RESUMO

OBJECTIVE: Weight regain after weight loss is common, and there is evidence to suggest negative effects on health because of weight cycling. This study sought to investigate the impact of weight regain in formerly obese mice on adipose tissue architecture and stromal cell function. METHODS: A diet-switch model was employed for obesity induction, weight loss, and weight regain in mice. Flow cytometry quantified adipose tissue leukocytes in adipose tissue. Liver and adipose tissue depots were compared to determine tissue-specific effects of weight cycling. RESULTS: Epididymal white adipose tissue of formerly obese mice failed to expand in response to repeat exposure to high-fat diet and retained elevated numbers of macrophages and T cells. Weight regain was associated with disproportionally elevated liver mass, hepatic triglyceride content, serum insulin concentration, and serum transaminase concentration. These effects occurred despite an extended 6-month weight loss cycle and they demonstrate that formerly obese mice maintain durable alterations in their physiological response to weight regain. Conditioned media from epididymal adipose tissue of formerly obese mice inhibited adipogenesis of 3T3-L1 preadipocytes, suggesting a potential mechanism to explain failed epididymal adipose tissue expansion during weight regain. CONCLUSIONS: Metabolic abnormalities related to defects in adipose tissue expansion and ongoing dysfunction manifest in formerly obese mice during weight regain.


Assuntos
Tecido Adiposo/metabolismo , Fígado Gorduroso/metabolismo , Obesidade/metabolismo , Aumento de Peso/fisiologia , Animais , Dieta Hiperlipídica , Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos
8.
PLoS One ; 11(7): e0159217, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27454769

RESUMO

Non-alcoholic fatty liver disease is a prevalent problem throughout the western world. Liver sinusoidal endothelial cells (LSEC) have been shown to play important roles in liver injury and repair, but their role in the underlying pathogenetic mechanisms of non-alcoholic fatty liver disease remains undefined. Here, we evaluated the effects of steatosis on LSEC gene expression in a murine model of non-alcoholic fatty liver disease and an immortalized LSEC line. Using microarray we identified distinct gene expression profiles following exposure to free fatty acids. Gene pathway analysis showed a number of differentially expressed genes including those involved in lipid metabolism and signaling and inflammation. Interestingly, in contrast to hepatocytes, fatty acids led to decreased expression of pro-inflammatory chemokines including CCL2 (MCP-1), CXCL10 and CXCL16 in both primary and LSEC cell lines. Chemokine downregulation translated into a significant inhibition of monocyte migration and LSECs isolated from steatotic livers demonstrated a similar shift towards an anti-inflammatory phenotype. Overall, these pathways may represent a compensatory mechanism to reverse the liver damage associated with non-alcoholic fatty liver disease.

9.
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA