Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 255
Filtrar
1.
Clin Exp Immunol ; 216(3): 280-292, 2024 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-38334487

RESUMO

Gestational diabetes mellitus (GDM) is a frequent and serious complication of pregnancy, often associated with obesity. Metabolic dysfunction and metainflammation are evident in both obesity and GDM. In this cross-sectional study, we aimed at defining the direct contribution of the immune system in GDM, across the main metabolic tissues, specifically focussing on elucidating the roles of obesity and GDM to the clinical outcome. Using immunoassays and multicolour flow cytometry, cytokine profiles and immune cell frequencies were measured in maternal circulation and central metabolic tissues [placenta and visceral adipose tissue (VAT)] in GDM-diagnosed (n = 28) and normal glucose tolerant (n = 32) women undergoing caesarean section. Participants were sub-grouped as non-obese [body mass index (BMI) < 30 kg/m2] or obese (BMI ≥ 30 kg/m2). Unsupervised data analysis was performed on the flow cytometry data set to identify functional alterations. GDM obese participants had significantly elevated circulating IL-6 and IL-17A levels. GDM non-obese participants had elevated circulating IL-12p70, elevated placental IL-17A, and VAT IFN-γ production. Unsupervised clustering of immune populations across the three biological sites simultaneously, identified different NK- and T-cell phenotypes that were altered in NGT obese and GDM non-obese participants, while a classical tissue monocyte cluster was increased in GDM obese participants. In this study, there was significant evidence of subclinical inflammation, and significant alterations in clusters of NK cells, T cells, and tissue monocyte populations in GDM. While increased adiposity assimilates with increased inflammation in the non-pregnant state, this overt relationship may not be as evident during pregnancy and warrants further examination in future longitudinal studies.


Assuntos
Diabetes Gestacional , Inflamação , Obesidade , Humanos , Feminino , Gravidez , Diabetes Gestacional/imunologia , Diabetes Gestacional/sangue , Adulto , Obesidade/imunologia , Inflamação/imunologia , Estudos Transversais , Gordura Intra-Abdominal/imunologia , Gordura Intra-Abdominal/metabolismo , Placenta/imunologia , Placenta/metabolismo , Células Matadoras Naturais/imunologia , Interleucina-17/sangue , Citocinas/sangue , Citocinas/metabolismo , Interleucina-6/sangue , Índice de Massa Corporal , Linfócitos T/imunologia , Interferon gama/sangue
2.
Geroscience ; 44(3): 1761-1778, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35477832

RESUMO

Adipose tissue dysfunction is strongly linked to the development of chronic inflammation and cardiometabolic disorders in aging. While much attention has been given to the role of resident adipose tissue immune cells in the disruption of homeostasis in obesity, age-specific effects remain understudied. Here, we identified and characterized a population of γδ T cells, which show unique age-dependent accumulation in the visceral adipose tissue (VAT) of both mice and humans. Diet-induced obesity likewise increased γδ T cell numbers; however, the effect was greater in the aged where the increase was independent of fat mass. γδ T cells in VAT express a tissue-resident memory T cell phenotype (CD44hiCD62LlowCD69+) and are predominantly IL-17A-producing cells. Transcriptome analyses of immunomagnetically purified γδ T cells identified significant age-associated differences in expression of genes related to inflammation, immune cell composition, and adipocyte differentiation, suggesting age-dependent qualitative changes in addition to the quantitative increase. Genetic deficiency of γδ T cells in old age improved the metabolic phenotype, characterized by increased respiratory exchange ratio, and lowered levels of IL-6 both systemically and locally in VAT. Decreased IL-6 was predominantly due to reduced production by non-immune stromal cells, primarily preadipocytes, and adipose-derived stem cells. Collectively, these findings suggest that an age-dependent increase of tissue-resident γδ T cells in VAT contributes to local and systemic chronic inflammation and metabolic dysfunction in aging.


Assuntos
Envelhecimento , Inflamação , Gordura Intra-Abdominal , Subpopulações de Linfócitos T , Animais , Inflamação/imunologia , Interleucina-6/metabolismo , Gordura Intra-Abdominal/imunologia , Camundongos , Obesidade/metabolismo , Subpopulações de Linfócitos T/imunologia
3.
J Clin Endocrinol Metab ; 107(1): 53-66, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34519830

RESUMO

CONTEXT: Circulating adiponectin levels are decreased in pregnant women with obesity or gestational diabetes, and this is believed to contribute to the insulin resistance and increased risk of fetal overgrowth associated with these conditions. However, the molecular mechanisms regulating adiponectin secretion from maternal adipose tissues in pregnancy are poorly understood. OBJECTIVE: We tested the hypothesis that obesity in pregnancy is associated with adipose tissue insulin resistance and increased adiponectin ubiquitination and degradation, caused by inflammation and endoplasmic reticulum (ER) stress. METHODS: Visceral adipose tissues were collected from lean and obese pregnant humans and mice. Total and ubiquitinated adiponectin, and markers of inflammation, ER stress, and insulin resistance were examined in adipose tissues. The role of insulin, inflammation, and ER stress in mediating adiponectin ubiquitination and degradation was examined using 3T3L-1 adipocytes. RESULTS: Obesity in pregnancy is associated with adipose tissue inflammation, ER stress, insulin resistance, increased adiponectin ubiquitination, and decreased total abundance of adiponectin. Adiponectin ubiquitination was increased in visceral fat of obese pregnant women as compared to lean pregnant women. We further observed that insulin prevents, whereas ER stress and inflammation promote, adiponectin ubiquitination and degradation in differentiated 3T3-L1 adipocytes. CONCLUSION: We have identified adiponectin ubiquitination as a key mechanism by which obesity diminishes adiponectin secretion in pregnancy. This information will help us better understand the mechanisms controlling maternal insulin resistance and fetal growth in pregnancy and may provide a foundation for the development of strategies aimed at improving adiponectin production in pregnant women with obesity or gestational diabetes.


Assuntos
Adiponectina/metabolismo , Diabetes Gestacional/metabolismo , Insulina/metabolismo , Obesidade Materna/metabolismo , Células 3T3-L1 , Adipócitos/metabolismo , Adiponectina/análise , Adulto , Animais , Estudos de Coortes , Diabetes Gestacional/imunologia , Modelos Animais de Doenças , Feminino , Humanos , Recém-Nascido , Resistência à Insulina/imunologia , Gordura Intra-Abdominal/imunologia , Gordura Intra-Abdominal/patologia , Masculino , Camundongos , Obesidade Materna/imunologia , Obesidade Materna/patologia , Gravidez , Proteólise , Ubiquitinação/imunologia
4.
J Clin Endocrinol Metab ; 107(1): 150-166, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34487152

RESUMO

CONTEXT: Chronic glucocorticoid (GC) overexposure, resulting from endogenous Cushing's syndrome (CS) or exogenous GC therapy, causes several adverse outcomes, including persistent central fat accumulation associated with a low-grade inflammation. However, no previous multiomics studies in visceral adipose tissue (VAT) from patients exposed to high levels of unsuppressed GC during active CS or after remission are available yet. OBJECTIVE: To determine the persistent VAT transcriptomic alterations and epigenetic fingerprints induced by chronic hypercortisolism. METHODS: We employed a translational approach combining high-throughput data on endogenous CS patients and a reversible CS mouse model. We performed RNA sequencing and chromatin immunoprecipitation sequencing on histone modifications (H3K4me3, H3K27ac, and H3K27me3) to identify persistent transcriptional and epigenetic signatures in VAT produced during active CS and maintained after remission. RESULTS: VAT dysfunction was associated with low-grade proinflammatory status, macrophage infiltration, and extracellular matrix remodeling. Most notably, chronic hypercortisolism caused a persistent circadian rhythm disruption in VAT through core clock genes modulation. Importantly, changes in the levels of 2 histone modifications associated to gene transcriptional activation (H3K4me3 and H3K27ac) correlated with the observed differences in gene expression during active CS and after CS remission. CONCLUSION: We identified for the first time the persistent transcriptional and epigenetic signatures induced by hypercortisolism in VAT, providing a novel integrated view of molecular components driving the long-term VAT impairment associated with CS.


Assuntos
Neoplasias das Glândulas Suprarrenais/complicações , Síndrome de Cushing/metabolismo , Glucocorticoides/efeitos adversos , Gordura Intra-Abdominal/imunologia , Obesidade Abdominal/genética , Administração Oral , Neoplasias das Glândulas Suprarrenais/diagnóstico , Neoplasias das Glândulas Suprarrenais/imunologia , Neoplasias das Glândulas Suprarrenais/urina , Adulto , Animais , Biópsia , Sequenciamento de Cromatina por Imunoprecipitação , Corticosterona/administração & dosagem , Corticosterona/efeitos adversos , Estudos Transversais , Síndrome de Cushing/imunologia , Síndrome de Cushing/patologia , Modelos Animais de Doenças , Epigenoma/efeitos dos fármacos , Epigenoma/imunologia , Feminino , Glucocorticoides/administração & dosagem , Glucocorticoides/metabolismo , Humanos , Hidrocortisona/metabolismo , Hidrocortisona/urina , Inflamação/induzido quimicamente , Inflamação/imunologia , Inflamação/metabolismo , Gordura Intra-Abdominal/metabolismo , Gordura Intra-Abdominal/patologia , Masculino , Camundongos , Pessoa de Meia-Idade , Obesidade Abdominal/imunologia , Obesidade Abdominal/patologia , RNA-Seq , Transcriptoma/efeitos dos fármacos , Transcriptoma/imunologia
5.
Biochem Pharmacol ; 192: 114723, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34364887

RESUMO

Both obesity and aging are associated with the development of metabolic diseases such as type 2 diabetes and cardiovascular disease. Chronic low-grade inflammation of adipose tissue is one of the mechanisms implicated in the progression of these diseases. Obesity and aging trigger adipose tissue alterations that ultimately lead to a pro-inflammatory phenotype of the adipose tissue-resident immune cells. Obesity and aging also share other features such as a higher visceral vs. subcutaneous adipose tissue ratio and a decreased lifespan. Here, we review the common characteristics of obesity and aging and the alterations in white adipose tissue and resident immune cells. We focus on the adipose tissue metabolic derangements in obesity and aging such as inflammation and adipose tissue remodeling.


Assuntos
Adipócitos Brancos/imunologia , Tecido Adiposo Branco/imunologia , Envelhecimento/imunologia , Distribuição da Gordura Corporal/métodos , Obesidade/imunologia , Adipócitos Brancos/metabolismo , Adipócitos Brancos/patologia , Tecido Adiposo Branco/metabolismo , Tecido Adiposo Branco/patologia , Envelhecimento/metabolismo , Envelhecimento/patologia , Animais , Humanos , Gordura Intra-Abdominal/imunologia , Gordura Intra-Abdominal/metabolismo , Gordura Intra-Abdominal/patologia , Obesidade/metabolismo , Obesidade/patologia
6.
Front Immunol ; 12: 702506, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34421908

RESUMO

Type 1 diabetes (T1D) is a proinflammatory pathology that leads to the specific destruction of insulin producing ß-cells and hyperglycaemia. Much of the knowledge about type 1 diabetes (T1D) has focused on mechanisms of disease progression such as adaptive immune cells and the cytokines that control their function, whereas mechanisms linked with the initiation of the disease remain unknown. It has been hypothesized that in addition to genetics, environmental factors play a pivotal role in triggering ß-cell autoimmunity. The BioBreeding Diabetes Resistant (BBDR) and LEW1.WR1 rats have been used to decipher the mechanisms that lead to virus-induced T1D. Both animals develop ß-cell inflammation and hyperglycemia upon infection with the parvovirus Kilham Rat Virus (KRV). Our earlier in vitro and in vivo studies indicated that KRV-induced innate immune upregulation early in the disease course plays a causal role in triggering ß-cell inflammation and destruction. Furthermore, we recently found for the first time that infection with KRV induces inflammation in visceral adipose tissue (VAT) detectable as early as day 1 post-infection prior to insulitis and hyperglycemia. The proinflammatory response in VAT is associated with macrophage recruitment, proinflammatory cytokine and chemokine upregulation, endoplasmic reticulum (ER) and oxidative stress responses, apoptosis, and downregulation of adipokines and molecules that mediate insulin signaling. Downregulation of inflammation suppresses VAT inflammation and T1D development. These observations are strikingly reminiscent of data from obesity and type 2 diabetes (T2D) in which VAT inflammation is believed to play a causal role in disease mechanisms. We propose that VAT inflammation and dysfunction may be linked with the mechanism of T1D progression.


Assuntos
Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/virologia , Gordura Intra-Abdominal/imunologia , Gordura Intra-Abdominal/virologia , Infecções por Parvoviridae/imunologia , Animais , Humanos , Parvovirus/imunologia , Ratos
7.
Front Immunol ; 12: 702835, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34421909

RESUMO

Arising incidence of metabolic disorders and related diseases caused by obesity is a global health concern. Elucidating the role of the immune system in this process will help to understand the related mechanisms and develop treatment strategies. Here, we have focused on innate immune cells in visceral adipose tissue (VAT) and summarized the roles of these cells in maintaining the homeostasis of VAT. Furthermore, this review reveals the importance of quantitative and functional changes of innate immune cells when the metabolic microenvironment changes due to obesity or excess lipids, and confirms that these changes eventually lead to the occurrence of chronic inflammation and metabolic diseases of VAT. Two perspectives are reviewed, which include sequential changes in various innate immune cells in the steady state of VAT and its imbalance during obesity. Cross-sectional interactions between various innate immune cells at the same time point are also reviewed. Through delineation of a comprehensive perspective of VAT homeostasis in obesity-induced chronic inflammation, and ultimately metabolic dysfunction and disease, we expect to clarify the complex interactive networks among distinct cell populations and propose that these interactions should be taken into account in the development of biotherapeutic strategies.


Assuntos
Homeostase/imunologia , Imunidade Inata/imunologia , Gordura Intra-Abdominal/imunologia , Obesidade/imunologia , Animais , Humanos
8.
Front Immunol ; 12: 669629, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34305899

RESUMO

Background and aims: Group 2 innate lymphoid cells (ILC2s) have been implicated in the regulation of metabolic homeostasis in mice. Methods: In this study, the role of ILC2s in white adipose tissue (WAT) was investigated using ST2, an IL-33 receptor that is expressed on ILC2 knockout mice. Results: The deficiency of ST2 decreased ILC2s in WAT, whereas ex-ILC2, which acquired group 1 innate lymphoid cell (ILC1)-like traits, was increased. This led to significant metabolic disorders such as visceral fat obesity, decreased browning in WAT, reduction of energy metabolism, and impaired glucose tolerance, compared to wild type (WT) mice. Those metabolic abnormalities of ST2-knockout (ST2KO) mice were not ameliorated by IL-33 administration, but impaired glucose tolerance and visceral fat obesity were significantly improved by transplantation of ILCs from the bone marrow of WT mice. The relative expression of Cd36 in WAT increased due to the deficiency of ST2, and the storage of saturated fatty acids in WAT of ST2KO mice was significantly higher than that of WT mice. Moreover, saturated fatty acids aggravated the chronic inflammation in adipocytes, promoted the differentiation of M1-like macrophages, and inhibited that of M2-like macrophages. Conclusions: Our results indicated that ILC2 regulates diet-induced obesity and chronic inflammation through the regulation of saturated fatty acid absorption in visceral adipose tissue.


Assuntos
Glicemia/metabolismo , Ácidos Graxos/metabolismo , Intolerância à Glucose/metabolismo , Imunidade Inata , Resistência à Insulina , Gordura Intra-Abdominal/metabolismo , Linfócitos/metabolismo , Absorção Fisiológica , Transferência Adotiva , Animais , Glicemia/efeitos dos fármacos , Antígenos CD36/metabolismo , Intolerância à Glucose/tratamento farmacológico , Intolerância à Glucose/genética , Intolerância à Glucose/imunologia , Homeostase , Imunidade Inata/efeitos dos fármacos , Inflamação/imunologia , Inflamação/metabolismo , Mediadores da Inflamação/metabolismo , Proteína 1 Semelhante a Receptor de Interleucina-1/genética , Proteína 1 Semelhante a Receptor de Interleucina-1/metabolismo , Interleucina-33/farmacologia , Gordura Intra-Abdominal/efeitos dos fármacos , Gordura Intra-Abdominal/imunologia , Linfócitos/efeitos dos fármacos , Linfócitos/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/imunologia , Obesidade/metabolismo , Células RAW 264.7
9.
Front Endocrinol (Lausanne) ; 12: 703170, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34322094

RESUMO

3,5-diiodo-thyronine (T2), an endogenous metabolite of thyroid hormones, exerts beneficial metabolic effects. When administered to overweight rats receiving a high fat diet (HFD), it significantly reduces body fat accumulation, which is a risk factor for the development of an inflammatory state and of related metabolic diseases. In the present study, we focused our attention on T2 actions aimed at improving the adverse effects of long-lasting HFD such as the adipocyte inflammatory response. For this purpose, three groups of rats were used throughout: i) receiving a standard diet for 14 weeks; ii) receiving a HFD for 14 weeks, and iii) receiving a HFD for 14 weeks with a simultaneous daily injection of T2 for the last 4 weeks. The results showed that T2 administration ameliorated the expression profiles of pro- and anti-inflammatory cytokines, reduced macrophage infiltration in white adipose tissue, influenced their polarization and reduced lymphocytes recruitment. Moreover, T2 improved the expression of hypoxia markers, all altered in HFD rats, and reduced angiogenesis by decreasing the pro-angiogenic miR126 expression. Additionally, T2 reduced the oxidative damage of DNA, known to be associated to the inflammatory status. This study demonstrates that T2 is able to counteract some adverse effects caused by a long-lasting HFD and to produce beneficial effects on inflammation. Irisin and SIRT1 pathway may represent a mechanism underlying the above described effects.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Di-Iodotironinas/farmacologia , Hipóxia/tratamento farmacológico , Inflamação/tratamento farmacológico , Gordura Intra-Abdominal/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Neovascularização Patológica/tratamento farmacológico , Adipocinas/metabolismo , Animais , Dano ao DNA , Hipóxia/metabolismo , Hipóxia/patologia , Inflamação/etiologia , Inflamação/patologia , Gordura Intra-Abdominal/imunologia , Gordura Intra-Abdominal/metabolismo , Macrófagos/imunologia , Masculino , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Sobrepeso/fisiopatologia , Estresse Oxidativo , Ratos , Ratos Wistar
10.
Front Immunol ; 12: 690069, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34322121

RESUMO

Anti-inflammatory therapies have the potential to become an effective treatment for obesity-related diseases. However, the huge gap of immune system between human and rodent leads to limitations of drug discovery. This work aims at constructing a transgenic pig model with higher risk of metabolic diseases and outlining the immune responses at the early stage of metaflammation by transcriptomic strategy. We used CRISPR/Cas9 techniques to targeted knock-in three humanized disease risk genes, GIPRdn , hIAPP and PNPLA3I148M . Transgenic effect increased the risk of metabolic disorders. Triple-transgenic pigs with short-term diet intervention showed early symptoms of type 2 diabetes, including glucose intolerance, pancreatic lipid infiltration, islet hypertrophy, hepatic lobular inflammation and adipose tissue inflammation. Molecular pathways related to CD8+ T cell function were significantly activated in the liver and visceral adipose samples from triple-transgenic pigs, including antigen processing and presentation, T-cell receptor signaling, co-stimulation, cytotoxicity, and cytokine and chemokine secretion. The similar pro-inflammatory signaling in liver and visceral adipose tissue indicated that there might be a potential immune crosstalk between the two tissues. Moreover, genes that functionally related to liver antioxidant activity, mitochondrial function and extracellular matrix showed distinct expression between the two groups, indicating metabolic stress in transgenic pigs' liver samples. We confirmed that triple-transgenic pigs had high coincidence with human metabolic diseases, especially in the scope of inflammatory signaling at early stage metaflammation. Taken together, this study provides a valuable large animal model for the clinical study of metaflammation and metabolic diseases.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Diabetes Mellitus Tipo 2/imunologia , Gordura Intra-Abdominal/imunologia , Fígado/imunologia , Ativação Linfocitária , Hepatopatia Gordurosa não Alcoólica/imunologia , Precursor de Proteína beta-Amiloide/genética , Animais , Animais Geneticamente Modificados , Glicemia/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Citocinas/genética , Citocinas/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Modelos Animais de Doenças , Mediadores da Inflamação/metabolismo , Gordura Intra-Abdominal/metabolismo , Gordura Intra-Abdominal/patologia , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Lipase/genética , Lipídeos/sangue , Fígado/metabolismo , Fígado/patologia , Masculino , Proteínas de Membrana/genética , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/patologia , Receptores dos Hormônios Gastrointestinais/genética , Suínos/genética , Transcriptoma
11.
Inflammation ; 44(6): 2260-2269, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34165676

RESUMO

The anti-inflammatory adipokine CTRP-3 might affect innate immune reactions such as NOD1. The impact of CTRP-3 on NOD1-mediated inflammation in adipocytes and monocytic cells as well as on NOD1 expression was investigated. Murine 3T3-L1 pre-adipocytes and adipocytes as well as human THP-1 monocyte-like cells were co-stimulated with the synthetic NOD1 agonist Tri-DAP and recombinant CTRP-3. Gonadal adipose tissue and primary adipocytes were obtained from a murine model carrying a knockout (KO) of CTRP-3 in adipocytes but not in stroma-vascular cells. Wildtype mice with lipopolysaccharide (LPS)-induced elevated NOD1 expression were treated with CTRP-3. Secreted inflammatory cytokines in cell supernatants were measured by ELISA and mRNA levels were quantified by RT-PCR. Pro-inflammatory chemokine and cytokine secretion (MCP-1, RANTES, TNFα) was induced by NOD1 activation in adipocytes and monocyte-like cells, and MCP-1 and RANTES release was effectively inhibited by pre-incubation of cells with CTRP-3. CTRP-3 also antagonized LPS-triggered induction of NOD1 gene expression in murine adipose tissue, whereas adipocyte CTRP-3 deficiency upregulated NOD1 expression in adipose tissue. CTRP-3 is an effective antagonist of peptidoglycan-induced, NOD1-mediated inflammation and of LPS-induced NOD1 expression. Since basal NOD1 expression is increased by adipocyte CTRP-3 deficiency, there have to be also inflammation-independent mechanisms of NOD1 expression regulation by CTRP-3.


Assuntos
Adipócitos/metabolismo , Adipocinas/metabolismo , Gordura Intra-Abdominal/metabolismo , Proteína Adaptadora de Sinalização NOD1/metabolismo , Síndrome de Resposta Inflamatória Sistêmica/metabolismo , Células 3T3-L1 , Adipócitos/imunologia , Adipocinas/genética , Animais , Citocinas/metabolismo , Modelos Animais de Doenças , Humanos , Mediadores da Inflamação/metabolismo , Gordura Intra-Abdominal/imunologia , Lipopolissacarídeos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Adaptadora de Sinalização NOD1/genética , Transdução de Sinais , Gordura Subcutânea/imunologia , Gordura Subcutânea/metabolismo , Síndrome de Resposta Inflamatória Sistêmica/induzido quimicamente , Síndrome de Resposta Inflamatória Sistêmica/genética , Síndrome de Resposta Inflamatória Sistêmica/imunologia , Células THP-1
12.
FASEB J ; 35(6): e21650, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33993539

RESUMO

Mesenchymal stem cells from healthy adipose tissue are adipocytes progenitors with immunosuppressive potential that are used for years in cell therapy. Whether adipose stem cells (ASC) may prevent inflammation in early obesity is not known. To address this question, we performed a kinetic study of high-fat (HF) diet induced obesity in mice to follow the immune regulating functions of adipose stem cells (ASC) isolated from the subcutaneous (SAT) and the visceral adipose tissue (VAT). Our results show that, early in obesity and before inflammation was detected, HF diet durably and differently activated ASC from SAT and VAT. Subcutaneous ASC from HF-fed mice strongly inhibited the proliferation of activated T lymphocytes, whereas visceral ASC selectively inhibited TNFα expression by macrophages and simultaneously released higher concentrations of IL6. These depot specific differences may contribute to the low-grade inflammation that develops with obesity in VAT while inflammation in SAT is delayed. The mechanisms involved differ from those already described for naïve cells activation with inflammatory cytokines and probably engaged metabolic activation. These results evidence that adipose stem cells are metabolic sensors acquiring an obesity-primed immunocompetent state in answer to depot-specific intrinsic features with overnutrition, placing these cells ahead of inflammation in the local dialog with immune cells.


Assuntos
Tecido Adiposo/imunologia , Inflamação/imunologia , Gordura Intra-Abdominal/imunologia , Células-Tronco Mesenquimais/imunologia , Obesidade/fisiopatologia , Gordura Subcutânea/imunologia , Linfócitos T/imunologia , Tecido Adiposo/patologia , Animais , Inflamação/patologia , Gordura Intra-Abdominal/patologia , Ativação Linfocitária , Masculino , Células-Tronco Mesenquimais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Gordura Subcutânea/patologia , Linfócitos T/patologia
13.
Sci Rep ; 11(1): 10409, 2021 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-34001955

RESUMO

The calcium sensing receptor (CaSR) is a G-protein coupled receptor that especially plays an important role in the sensing of extracellular calcium to maintain its homeostasis. Several in-vitro studies demonstrated that CaSR plays a role in adipose tissue metabolism and inflammation, resulting in systemic inflammation and contributing to atherosclerosis development. The aim of this study was to investigate whether adipocyte CaSR plays a role in adipose tissue inflammation in-vivo and atherosclerosis development. By using a newly established conditional mature adipocyte specific CaSR deficient mouse on a hyperlipidemic and atherosclerosis prone Apoe-/- background it could be shown that CaSR deficiency in adipocytes does neither contribute to initiation nor to progression of atherosclerotic plaques as judged by the unchanged lesion size or composition. Additionally, CaSR deficiency did not influence gonadal visceral adipose tissue (vAT) inflammation in-vivo, although a small decrease in gonadal visceral adipose cholesterol content could be observed. In conclusion, adipocyte CaSR seems not to be involved in vAT inflammation in-vivo and does not influence atherosclerosis development in hyperlipidemic Apoe-/- mice.


Assuntos
Adipócitos/metabolismo , Hiperlipidemias/complicações , Gordura Intra-Abdominal/patologia , Placa Aterosclerótica/imunologia , Receptores de Detecção de Cálcio/deficiência , Animais , Modelos Animais de Doenças , Humanos , Hiperlipidemias/genética , Hiperlipidemias/imunologia , Inflamação/imunologia , Inflamação/patologia , Gordura Intra-Abdominal/citologia , Gordura Intra-Abdominal/imunologia , Gordura Intra-Abdominal/metabolismo , Camundongos , Camundongos Knockout para ApoE , Placa Aterosclerótica/genética , Placa Aterosclerótica/patologia , Receptores de Detecção de Cálcio/genética
14.
Nat Commun ; 12(1): 2388, 2021 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-33888702

RESUMO

To unravel the pathogenesis of obesity and its complications, we investigate the interplay between circadian clocks and NF-κB pathway in human adipose tissue. The circadian clock function is impaired in omental fat from obese patients. ChIP-seq analyses reveal that the core clock activator, BMAL1 binds to several thousand target genes. NF-κB competes with BMAL1 for transcriptional control of some targets and overall, BMAL1 chromatin binding occurs in close proximity to NF-κB consensus motifs. Obesity relocalizes BMAL1 occupancy genome-wide in human omental fat, thereby altering the transcription of numerous target genes involved in metabolic inflammation and adipose tissue remodeling. Eventually, clock dysfunction appears at early stages of obesity in mice and is corrected, together with impaired metabolism, by NF-κB inhibition. Collectively, our results reveal a relationship between NF-κB and the molecular clock in adipose tissue, which may contribute to obesity-related complications.


Assuntos
Fatores de Transcrição ARNTL/metabolismo , Relógios Circadianos/imunologia , Gordura Intra-Abdominal/patologia , NF-kappa B/metabolismo , Obesidade/complicações , Adipócitos/imunologia , Adipócitos/metabolismo , Adiponectina/genética , Adulto , Animais , Biópsia , Estudos de Casos e Controles , Células Cultivadas , Sequenciamento de Cromatina por Imunoprecipitação , Relógios Circadianos/genética , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Humanos , Inflamação/imunologia , Inflamação/patologia , Gordura Intra-Abdominal/imunologia , Masculino , Células-Tronco Mesenquimais , Camundongos Transgênicos , Pessoa de Meia-Idade , Obesidade/imunologia , Obesidade/metabolismo , Obesidade/patologia , Omento/imunologia , Omento/patologia , Proteínas Circadianas Period/genética , Cultura Primária de Células , Transcrição Gênica
15.
Int J Mol Sci ; 22(8)2021 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-33921758

RESUMO

Inflammatory bowel diseases (IBDs), chronic inflammatory disorders affecting the gastrointestinal tract, include Crohn's disease and ulcerative colitis. There are increasing clinical and experimental data showing that obesity, especially visceral adiposity, plays a substantial role in the pathogenesis of IBD. Obesity seems to be an important risk factor also for IBD disease severity and clinical outcomes. Visceral adipose tissue is an active multifunctional metabolic organ involved in lipid storage and immunological and endocrine activity. Bowel inflammation penetrates the surrounding adipose tissue along the mesentery. Mesenteric fat serves as a barrier to inflammation and controls immune responses to the translocation of gut bacteria. At the same time, mesenteric adipose tissue may be the principal source of cytokines and adipokines responsible for inflammatory processes associated with IBD. This review is particularly focusing on the potential role of adipokines in IBD pathogenesis and their possible use as promising therapeutic targets.


Assuntos
Tecido Adiposo/metabolismo , Doenças Inflamatórias Intestinais/metabolismo , Adipocinas/metabolismo , Tecido Adiposo/imunologia , Animais , Humanos , Doenças Inflamatórias Intestinais/imunologia , Gordura Intra-Abdominal/imunologia , Gordura Intra-Abdominal/metabolismo , Obesidade Abdominal/imunologia , Obesidade Abdominal/metabolismo
16.
J Exp Med ; 218(6)2021 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-33881452

RESUMO

A unique population of Foxp3+ regulatory T cells (TRs) resides in visceral adipose tissue (VAT) that regulates adipose inflammation and helps preserve insulin sensitivity. Inducible T cell co-stimulator (ICOS) is highly expressed on effector (e)TRs that migrate to nonlymphoid tissues, and contributes to their maintenance and function in models of autoimmunity. In this study, we report an unexpected cell-intrinsic role for ICOS expression and downstream phosphoinositide 3-kinase (PI3K) signaling in limiting the abundance, VAT-associated phenotype, and function of TRs specifically in VAT. Icos-/- mice and mice expressing a knock-in form of ICOS that cannot activate PI3K had increased VAT-TR abundance and elevated expression of canonical VAT-TR markers. Loss of ICOS signaling facilitated enhanced accumulation of TRs to VAT associated with elevated CCR3 expression, and resulted in reduced adipose inflammation and heightened insulin sensitivity in the context of a high-fat diet. Thus, we have uncovered a new and surprising molecular pathway that regulates VAT-TR accumulation and function.


Assuntos
Tecido Adiposo/imunologia , Proteína Coestimuladora de Linfócitos T Induzíveis/imunologia , Transdução de Sinais/imunologia , Linfócitos T Reguladores/imunologia , Animais , Autoimunidade/imunologia , Dieta Hiperlipídica/métodos , Feminino , Fatores de Transcrição Forkhead/imunologia , Inflamação/imunologia , Insulina/imunologia , Resistência à Insulina/imunologia , Gordura Intra-Abdominal/imunologia , Masculino , Camundongos , Obesidade/imunologia , Fosfatidilinositol 3-Quinases/imunologia
17.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-33753509

RESUMO

Foxp3+CD4+ regulatory T cells (Tregs) regulate most types of immune response as well as several processes important for tissue homeostasis, for example, metabolism and repair. Dedicated Treg compartments-with distinct transcriptomes, T cell receptor repertoires, and growth/survival factor dependencies-have been identified in several nonlymphoid tissues. These Tregs are specifically adapted to function and operate in their home tissue-When, where, and how do they take on their specialized characteristics? We recently reported that a splenic Treg population expressing low levels of the transcription factor PPARγ (peroxisome proliferator-activated receptor gamma) contains precursors of Tregs residing in visceral adipose tissue. This finding made sense given that PPARγ, the "master regulator" of adipocyte differentiation, is required for the accumulation and function of Tregs in visceral adipose tissue but not in lymphoid tissues. Here we use single-cell RNA sequencing, single-cell Tcra and Tcrb sequencing, and adoptive-transfer experiments to show that, unexpectedly, the splenic PPARγlo Treg population is transcriptionally heterogeneous and engenders Tregs in multiple nonlymphoid tissues beyond visceral adipose tissue, such as skin and liver. The existence of a general pool of splenic precursors for nonlymphoid-tissue Tregs opens possibilities for regulating their emergence experimentally or therapeutically.


Assuntos
Gordura Intra-Abdominal/imunologia , PPAR alfa/metabolismo , Baço/imunologia , Linfócitos T Reguladores/imunologia , Transcriptoma , Transferência Adotiva , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , PPAR alfa/genética , RNA-Seq , Análise de Célula Única
18.
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
19.
Nat Rev Immunol ; 21(9): 597-611, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33772242

RESUMO

The FOXP3+CD4+ regulatory T (Treg) cells located in non-lymphoid tissues differ in phenotype and function from their lymphoid organ counterparts. Tissue Treg cells have distinct transcriptomes, T cell receptor repertoires and growth and survival factor dependencies that arm them to survive and operate in their home tissue. Their functions extend beyond immune surveillance to tissue homeostasis, including regulation of local and systemic metabolism, promotion of tissue repair and regeneration, and control of the proliferation, differentiation and fate of non-lymphoid cell progenitors. Treg cells in diverse tissues share a common FOXP3+CD4+ precursor located within lymphoid organs. This precursor undergoes definitive specialization once in the home tissue, following a multilayered array of common and tissue-distinct transcriptional programmes. Our deepening knowledge of tissue Treg cell biology will inform ongoing attempts to harness Treg cells for precision immunotherapeutics.


Assuntos
Linfócitos T Reguladores/imunologia , Animais , Feminino , Homeostase/imunologia , Humanos , Gordura Intra-Abdominal/citologia , Gordura Intra-Abdominal/imunologia , Masculino , Camundongos , Modelos Imunológicos , Músculo Esquelético/citologia , Músculo Esquelético/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Pele/citologia , Pele/imunologia , Linfócitos T Reguladores/fisiologia , Transcriptoma
20.
Adipocyte ; 10(1): 124-130, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-33595419

RESUMO

Fatty acid desaturase 1 (FADS1) polymorphisms alter fatty acid content in subcutaneous adipose tissue (SAT); however, existing evidence is limited and conflicting regarding the association between FADS1 variants and SAT inflammatory status. To advance this area, we conducted an exploratory study to investigate whether the common rs174537 polymorphism in FADS1 was associated with immune cell profiles in abdominal and femoral SAT in individuals with obesity. FADS1 gene expression and immune cell profiles in SAT depots were assessed by qPCR and flow cytometry, respectively. Although FADS1 gene expression was associated with genotype, no associations were observed with immune cell profiles in either depot. Our study provides additional evidence that rs174537 in FADS1 has minimal impact on inflammatory status in obese SAT.


Assuntos
Tecido Adiposo/imunologia , Ácidos Graxos Dessaturases/genética , Gordura Subcutânea/metabolismo , Tecido Adiposo/metabolismo , Adulto , Dessaturase de Ácido Graxo Delta-5 , Ácidos Graxos Dessaturases/imunologia , Ácidos Graxos Dessaturases/metabolismo , Ácidos Graxos/metabolismo , Feminino , Fêmur/metabolismo , Genótipo , Humanos , Gordura Intra-Abdominal/imunologia , Masculino , Pessoa de Meia-Idade , Obesidade/metabolismo , Projetos Piloto , Polimorfismo de Nucleotídeo Único/genética , Gordura Subcutânea/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...