Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Gut ; 71(12): 2463-2480, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35017197

RESUMO

OBJECTIVES: Gut microbiota is a key component in obesity and type 2 diabetes, yet mechanisms and metabolites central to this interaction remain unclear. We examined the human gut microbiome's functional composition in healthy metabolic state and the most severe states of obesity and type 2 diabetes within the MetaCardis cohort. We focused on the role of B vitamins and B7/B8 biotin for regulation of host metabolic state, as these vitamins influence both microbial function and host metabolism and inflammation. DESIGN: We performed metagenomic analyses in 1545 subjects from the MetaCardis cohorts and different murine experiments, including germ-free and antibiotic treated animals, faecal microbiota transfer, bariatric surgery and supplementation with biotin and prebiotics in mice. RESULTS: Severe obesity is associated with an absolute deficiency in bacterial biotin producers and transporters, whose abundances correlate with host metabolic and inflammatory phenotypes. We found suboptimal circulating biotin levels in severe obesity and altered expression of biotin-associated genes in human adipose tissue. In mice, the absence or depletion of gut microbiota by antibiotics confirmed the microbial contribution to host biotin levels. Bariatric surgery, which improves metabolism and inflammation, associates with increased bacterial biotin producers and improved host systemic biotin in humans and mice. Finally, supplementing high-fat diet-fed mice with fructo-oligosaccharides and biotin improves not only the microbiome diversity, but also the potential of bacterial production of biotin and B vitamins, while limiting weight gain and glycaemic deterioration. CONCLUSION: Strategies combining biotin and prebiotic supplementation could help prevent the deterioration of metabolic states in severe obesity. TRIAL REGISTRATION NUMBER: NCT02059538.


Assuntos
Diabetes Mellitus Tipo 2 , Microbioma Gastrointestinal , Obesidade Mórbida , Complexo Vitamínico B , Humanos , Camundongos , Animais , Prebióticos , Obesidade Mórbida/cirurgia , Biotina/farmacologia , Complexo Vitamínico B/farmacologia , Camundongos Endogâmicos C57BL , Obesidade/metabolismo , Inflamação
2.
Diabetologia ; 64(1): 240-254, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33125520

RESUMO

AIM/HYPOTHESIS: Altered adipose tissue secretory profile contributes to insulin resistance and type 2 diabetes in obesity. Preclinical studies have identified senescent cells as a cellular source of proinflammatory factors in adipose tissue of obese mice. In humans, potential links with obesity comorbidities are poorly defined. Here, we investigated adipose tissue senescent status and relationships with metabolic complications in human obesity. METHODS: The study includes a prospective cohort of 227 individuals with severe obesity. A photometric method was used to quantify senescence-associated ß-galactosidase (SA-ß-gal) activity in paired subcutaneous and omental adipose tissue biopsies obtained during gastric surgery. Gene and secretory profiling was performed in adipose tissue biopsies and in human primary pre-adipocytes in the presence or absence of senolytic drugs targeting senescent cells. Participants were phenotyped for anthropometric and bioclinical variables, metabolic complications and gastric surgery-induced improvement to address relationships with adipose tissue SA-ß-gal. RESULTS: SA-ß-gal activity was sevenfold higher in subcutaneous than in omental adipose tissue and not associated with BMI or chronological age. Several factors, including insulin-like growth factor binding protein 3 (IGFBP3), plasminogen activator inhibitor 1 (PAI1), C-C motif chemokine ligand 2 (CCL2) and IL-6, were upregulated in subcutaneous adipose tissue in relation with SA-ß-gal (p for linear trend across tertiles <0.05) and in pre-adipocytes cultured with inflammatory macrophage conditioned media. Senolytic treatment reduced SA-ß-gal staining and normalised these alterations. In the whole population, subcutaneous adipose tissue SA-ß-gal activity was positively associated with serum leptin, markers of insulin resistance and increased trunk fat mass. Metabolic complications, including type 2 diabetes and dyslipidaemia, were more prevalent in patients with high levels of SA-ß-gal, but improved with bariatric surgery whatever the initial adipose tissue senescent status. CONCLUSIONS/INTERPRETATION: This study highlights a phenotype of senescence in adipose tissue of severely obese individuals, which characterises prominently subcutaneous fat depots. Subcutaneous adipose tissue senescence is significantly linked to altered glucose metabolism and body fat distribution. Elimination of senescent cells through senolytic treatment could alleviate metabolic complications in severely obese people. Graphical abstract.


Assuntos
Glicemia/análise , Composição Corporal/fisiologia , Senescência Celular/fisiologia , Obesidade Mórbida/fisiopatologia , Gordura Subcutânea/enzimologia , beta-Galactosidase/metabolismo , Adipócitos/fisiologia , Cirurgia Bariátrica , Biópsia , Estudos de Coortes , Feminino , Humanos , Resistência à Insulina , Masculino , Obesidade Mórbida/metabolismo , Obesidade Mórbida/cirurgia , Estudos Prospectivos , Gordura Subcutânea/patologia , Resultado do Tratamento
3.
Mol Metab ; 39: 101007, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32360426

RESUMO

OBJECTIVE: Obesity is characterized by systemic and low-grade tissue inflammation. In the intestine, alteration of the intestinal barrier and accumulation of inflammatory cells in the epithelium are important contributors of gut inflammation. Recent studies demonstrated the role of the aryl hydrocarbon receptor (AhR) in the maintenance of immune cells at mucosal barrier sites. A wide range of ligands of external and local origin can activate this receptor. We studied the causal relationship between AhR activation and gut inflammation in obesity. METHODS: Jejunum samples from subjects with normal weight and severe obesity were phenotyped according to T lymphocyte infiltration in the epithelium from lamina propria and assayed for the mRNA level of AhR target genes. The effect of an AhR agonist was studied in mice and Caco-2/TC7 cells. AhR target gene expression, permeability to small molecules and ions, and location of cell-cell junction proteins were recorded under conditions of altered intestinal permeability. RESULTS: We showed that a low AhR tone correlated with a high inflammatory score in the intestinal epithelium in severe human obesity. Moreover, AhR activation protected junctional complexes in the intestinal epithelium in mice challenged by an oral lipid load. AhR ligands prevented chemically induced damage to barrier integrity and cytokine expression in Caco-2/TC7 cells. The PKC and p38MAPK signaling pathways were involved in this AhR action. CONCLUSIONS: The results of these series of human, mouse, and cell culture experiments demonstrate the protective effect of AhR activation in the intestine targeting particularly tight junctions and cytokine expression. We propose that AhR constitutes a valuable target to protect intestinal functions in metabolic diseases, which can be achieved in the future via food or drug ligands.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Mucosa Intestinal/metabolismo , Obesidade/metabolismo , Receptores de Hidrocarboneto Arílico/metabolismo , Adiposidade/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Biomarcadores , Linhagem Celular , Comorbidade , Citocinas/metabolismo , Células Epiteliais/metabolismo , Feminino , Humanos , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Jejuno/metabolismo , Metabolismo dos Lipídeos , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Pessoa de Meia-Idade , Modelos Biológicos , Obesidade/etiologia , Obesidade/patologia , Permeabilidade , Receptores de Hidrocarboneto Arílico/genética , Transdução de Sinais , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/metabolismo
4.
FASEB J ; 30(1): 241-51, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26362817

RESUMO

In mice, nutritional supplementation with the trans-10,cis-12 isomer of linoleic acid (t10,c12-CLA) promotes lipoatrophy, hyperinsulinemia, and macrophage infiltration in white adipose tissue (WAT). We explored the dynamics of these interrelated responses over 2 consecutive 7 d periods of t10,c12-CLA administration and withdrawal. t10,c12-CLA down-regulated lipogenic and lipolytic gene expression and increased collagen deposition, but with no evidence of cross-linking. An abundant CD45(+) cell infiltrate, comprising prominently CD206(+)CD11c(-) macrophages, was found in WAT in association with an anti-inflammatory gene signature. Infiltration of natural killer (NK) and dendritic cells contributed to WAT's innate immune response to t10,c12-CLA. Less abundant adaptive immune cells colonized WAT, including B, NK T, γδ T, and αß T cells. By contrast, T-regulatory cell abundance was not affected. Interruption of treatment allowed recovery of WAT mass and normalization of insulinemia, coincident with regain of WAT homeostasis owing to a coordinated reversion of genic, structural, and immune deregulations. These data revealed a striking resilience of WAT after a short-term metabolic injury induced by t10,c12-CLA, which relies on alternatively activated M2 macrophage engagement. In addition, the temporal links between variations in WAT alterations and insulinemia upon t10,c12-CLA manipulation strengthen the view that WAT dysfunctional status is critically involved in altered glucose homeostasis.


Assuntos
Tecido Adiposo Branco/efeitos dos fármacos , Ácidos Linoleicos Conjugados/farmacologia , Ativação de Macrófagos , Macrófagos/efeitos dos fármacos , Adaptação Fisiológica , Tecido Adiposo Branco/citologia , Animais , Células Cultivadas , Feminino , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T/efeitos dos fármacos
5.
Endocrinology ; 156(11): 4047-58, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26295369

RESUMO

Prior studies demonstrated increased plasma IgE in diabetic patients, but the direct participation of IgE in diabetes or obesity remains unknown. This study found that plasma IgE levels correlated inversely with body weight, body mass index, and body fat mass among a population of randomly selected obese women. IgE receptor FcϵR1-deficient (Fcer1a(-/-)) mice and diet-induced obesity (DIO) mice demonstrated that FcϵR1 deficiency in DIO mice increased food intake, reduced energy expenditure, and increased body weight gain but improved glucose tolerance and glucose-induced insulin secretion. White adipose tissue from Fcer1a(-/-) mice showed an increased expression of phospho-AKT, CCAAT/enhancer binding protein-α, peroxisome proliferator-activated receptor-γ, glucose transporter-4 (Glut4), and B-cell lymphoma 2 (Bcl2) but reduced uncoupling protein 1 (UCP1) and phosphorylated c-Jun N-terminal kinase (JNK) expression, tissue macrophage accumulation, and apoptosis, suggesting that IgE reduces adipogenesis and glucose uptake but induces energy expenditure, adipocyte apoptosis, and white adipose tissue inflammation. In 3T3-L1 cells, IgE inhibited the expression of CCAAT/enhancer binding protein-α and peroxisome proliferator-activated receptor-γ, and preadipocyte adipogenesis and induced adipocyte apoptosis. IgE reduced the 3T3-L1 cell expression of Glut4, phospho-AKT, and glucose uptake, which concurred with improved glucose tolerance in Fcer1a(-/-) mice. This study established two novel pathways of IgE in reducing body weight gain in DIO mice by suppressing adipogenesis and inducing adipocyte apoptosis while worsening glucose tolerance by reducing Glut4 expression, glucose uptake, and insulin secretion.


Assuntos
Metabolismo Energético/genética , Obesidade/genética , Receptores de IgE/genética , Aumento de Peso/genética , Células 3T3-L1 , Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Animais , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Dieta Hiperlipídica/efeitos adversos , Feminino , Expressão Gênica , Teste de Tolerância a Glucose , Humanos , Immunoblotting , Imunoglobulina E/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Obesos , Obesidade/etiologia , Obesidade/metabolismo , Obesidade Mórbida/sangue , PPAR gama/genética , PPAR gama/metabolismo , Interferência de RNA , Receptores de IgE/deficiência , Reação em Cadeia da Polimerase Via Transcriptase Reversa
6.
Nat Med ; 21(6): 610-8, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25939064

RESUMO

Accumulation of visceral adipose tissue correlates with elevated inflammation and increased risk of metabolic diseases. However, little is known about the molecular mechanisms that control its pathological expansion. Transcription factor interferon regulatory factor 5 (IRF5) has been implicated in polarizing macrophages towards an inflammatory phenotype. Here we demonstrate that mice lacking Irf5, when placed on a high-fat diet, show no difference in the growth of their epididymal white adipose tissue (epiWAT) but they show expansion of their subcutaneous white adipose tissue, as compared to wild-type (WT) mice on the same diet. EpiWAT from Irf5-deficient mice is marked by accumulation of alternatively activated macrophages, higher collagen deposition that restricts adipocyte size, and enhanced insulin sensitivity compared to epiWAT from WT mice. In obese individuals, IRF5 expression is negatively associated with insulin sensitivity and collagen deposition in visceral adipose tissue. Genome-wide analysis of gene expression in adipose tissue macrophages highlights the transforming growth factor ß1 (TGFB1) gene itself as a direct target of IRF5-mediated inhibition. This study uncovers a new function for IRF5 in controlling the relative mass of different adipose tissue depots and thus insulin sensitivity in obesity, and it suggests that inhibition of IRF5 may promote a healthy metabolic state during this condition.


Assuntos
Tecido Adiposo Branco/metabolismo , Inflamação/genética , Fatores Reguladores de Interferon/genética , Obesidade/genética , Animais , Dieta Hiperlipídica , Regulação da Expressão Gênica , Humanos , Inflamação/tratamento farmacológico , Inflamação/patologia , Resistência à Insulina/genética , Macrófagos , Camundongos , Obesidade/tratamento farmacológico , Obesidade/patologia , Fator de Crescimento Transformador beta1/biossíntese
7.
Mol Immunol ; 60(1): 54-61, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24769492

RESUMO

Polyreactive antibodies represent a significant fraction of immune repertoires and play an important role in the immune defense and immune homeostasis. Polyreactive B-cell receptors (BCR), however, are frequently expressed by B-cell lymphomas. It was suggested that polyreactive BCR on lymphoma cells might deliver stimulation signals by binding to various endogenous or exogenous antigens, thus promoting the survival of the malignant cells. In addition to natural polyreactive antibodies, immune repertoires contain antibodies that acquire polyreactivity after exposure to different redox-active substances such as reactive oxygen species, iron ions and heme. Here, we demonstrate that an antibody cloned from a patient's splenic marginal zone B-cell lymphoma acquires physiologically relevant binding affinity to various autoantigens following exposure to heme. We elucidated the mechanisms underlying polyreactive antigen binding. The results obtained in this study imply that antigen-binding receptors expressed on some malignant cells acquire polyreactivity after exposure to redox substances that are released at sites of inflammation or as a result of cellular damage. The acquisition of novel BCR specificities under hemolytic or inflammatory conditions may play an important role in the physiopathology of certain B-cell malignancies.


Assuntos
Dipeptídeos/imunologia , Hemina/imunologia , Imunoglobulina G/imunologia , Linfoma de Zona Marginal Tipo Células B/imunologia , Sequência de Aminoácidos , Anticorpos/imunologia , Especificidade de Anticorpos/imunologia , Reações Antígeno-Anticorpo/imunologia , Autoantígenos/imunologia , Sequência de Bases , Células Cultivadas , Feminino , Humanos , Cadeias Pesadas de Imunoglobulinas/genética , Região Variável de Imunoglobulina/genética , Cadeias kappa de Imunoglobulina/genética , Inflamação/imunologia , Pessoa de Meia-Idade , Receptores de Antígenos de Linfócitos B/imunologia , Análise de Sequência de DNA
8.
Diabetes ; 63(6): 1966-77, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24520123

RESUMO

Proinflammatory cytokines are critically involved in the alteration of adipose tissue biology leading to deterioration of glucose homeostasis in obesity. Here we show a pronounced proinflammatory signature of adipose tissue macrophages in type 2 diabetic obese patients, mainly driven by increased NLRP3-dependent interleukin (IL)-1ß production. IL-1ß release increased with glycemic deterioration and decreased after gastric bypass surgery. A specific enrichment of IL-17- and IL-22-producing CD4(+) T cells was found in adipose tissue of type 2 diabetic obese patients. Coculture experiments identified the effect of macrophage-derived IL-1ß to promote IL-22 and IL-17 production by human adipose tissue CD4(+) T cells. Reciprocally, adipose tissue macrophages express IL-17 and IL-22 receptors, making them sensitive to IL-17 and IL-22. IL-22 increased IL-1ß release by inducing pro-IL-1ß transcription through activation of C-Jun pathways in macrophages. In sum, these human data identified IL-1ß and the T-cell cytokine IL-22 as key players of a paracrine inflammatory pathway previously unidentified in adipose tissue, with a pathological relevance to obesity-induced type 2 diabetes. These results provide an additional rationale for targeting IL-1ß in obesity-linked type 2 diabetes and may have important implications for the conception of novel combined anti-IL-1ß and anti-IL-22 immunotherapy in human obesity.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Inflamação/patologia , Resistência à Insulina , Interleucina-1beta/metabolismo , Interleucinas/metabolismo , Obesidade/metabolismo , Tecido Adiposo/patologia , Adulto , Western Blotting , Linhagem Celular , Técnicas de Cocultura , Diabetes Mellitus Tipo 2/genética , Feminino , Amplificação de Genes , Hemoglobinas Glicadas , Humanos , Inflamação/genética , Resistência à Insulina/genética , Masculino , Obesidade/genética , Reação em Cadeia da Polimerase em Tempo Real , Linfócitos T/metabolismo , Interleucina 22
9.
Br J Haematol ; 155(4): 487-97, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21933172

RESUMO

The occurrence of alloantibodies against Factor VIII (FVIII) is the main iatrogenic complication in haemophilia A (HA). Anti-FVIII autoantibodies may also spontaneously appear in non-HA patients, leading to acquired haemophilia A. In both contexts, the antibody response against FVIII is complex and difficult to analyse due to the lack of suitable tools. Our purpose was to comprehensively map, at the amino acid level, discontinuous epitopes of the C2 domain of FVIII targeted by patients' antibodies. We synthesized 33 synthetic peptides, which were predicted by the bioinformatic algorithm PEPOP to mimic C2 domain discontinuous epitopes. Using an inhibition assay based on the x-MAP technology, we evaluated their ability to block the binding to the C2 domain of anti-C2 domain antibodies from pooled plasma samples. Nine peptides were thus selected and tested again in individual plasma samples. Our results support the view that C2 domain epitopes are organized as an epitopic mosaic distributed around the molecule, showed that each patient displayed a specific anti-C2 epitopic profile, and confirmed the complexity and variability of the immune response against the C2 domain of FVIII. This ability to finely map epitopes could be further used to follow the antibody specificity modifications over time.


Assuntos
Epitopos/imunologia , Fator VIII/imunologia , Hemofilia A/imunologia , Peptídeos/imunologia , Sequência de Aminoácidos , Autoanticorpos/imunologia , Biologia Computacional , Mapeamento de Epitopos , Epitopos/análise , Fator VIII/química , Fator VIII/metabolismo , Hemofilia A/sangue , Humanos , Isoanticorpos/imunologia , Modelos Moleculares , Dados de Sequência Molecular , Peptídeos/síntese química , Estabilidade Proteica
10.
Am J Pathol ; 174(5): 1575-87, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19349365

RESUMO

CD4+CD25+ regulatory T cells (Tregs) play a critical role in preventing immune aggression. One way in which Tregs exert immune surveillance activities is by modifying the function of antigen presenting cells (APCs) such as dendritic cells, macrophages, and B cells. Tregs can induce apoptosis of APCs or inhibit their activation and function, thereby regulating subsequent innate and adaptive immune responses. These actions of Tregs are mediated by both soluble factors (interleukin [IL]-10, transforming growth factor-beta, perforins, granzymes) and cell-associated molecules (cytotoxic T lymphocyte antigen 4, lymphocyte activation gene-3, CD18, neuropilin-1, LFA-1/CD11a, CD39), of which cytotoxic T lymphocyte antigen 4 has a key role. However, in autoimmunity, chronically activated APCs under the influence of intracellular signaling pathways, such as phosphatidyl inositol 3 kinase, JAK-STAT, MAPK, and nuclear factor-kappaB pathways, can escape surveillance by Tregs, leading to the activation of T cells that are refractory to suppression by Tregs. Moreover, APCs and APC-derived inflammatory cytokines such as tumor necrosis factor, IL-6, IL-1beta, and IL-23 can render Tregs defective and can also reciprocally enhance the activity of the IL-17-producing pathogenic Th17 T cell subset. Emerging knowledge of the importance of APC-Treg interactions in maintaining immune tolerance and aberrations in this cross talk in autoimmune diseases provides a rationale for therapeutic approaches specifically targeting this axis of the immune system.


Assuntos
Células Apresentadoras de Antígenos/fisiologia , Doenças Autoimunes/imunologia , Autoimunidade/fisiologia , Linfócitos T CD4-Positivos/metabolismo , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Linfócitos T Reguladores/fisiologia , Doenças Autoimunes/patologia , Doenças Autoimunes/terapia , Humanos
11.
Immunogenetics ; 56(3): 204-13, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15146297

RESUMO

Nontemplate (N)-nucleotide addition by the terminal dideoxynucleotidyl transferase (TdT) at the junctions of rearranging V( D) J gene segments greatly contribute to antigen-receptor diversity. TdT has been identified in several vertebrate species, where it is highly conserved. We report here the isolation of two forms of TdT mRNA in an amphibian, the Mexican axolotl. The isoform TdT1 shares all of the conserved structural motifs required for TdT activity and displays an average of 50-58% similarity at the amino acid level with TdT of other species. The second axolotl TdT variant ( TdT2) differs from TdT1 by a 57-amino acid deletion located between amino acids 165-222 of TdT1, including the first helix-hairpin-helix DNA-binding motif. During ontogeny, TdT products are first detected in the head of 6-week-old larvae and further in the head and trunk of 8-month-old larvae. These developmental stages correspond to the first detection of RAG1 and antigen-receptor (TCRbeta and IgHmicro) products in axolotl larvae. Our results suggest that in contrast to mammalian development, N diversity occurs early in axolotl development to diversify the primary repertoire. Phylogenetic analyses reveal that TdT and DNA polymerase mu(Pol mu) genes are closely related, and that both enzymes were already present in the common ancestor of jawed vertebrates.


Assuntos
Ambystoma mexicanum/genética , DNA Nucleotidilexotransferase/genética , Sistema Hematopoético/metabolismo , Ambystoma mexicanum/embriologia , Ambystoma mexicanum/crescimento & desenvolvimento , Sequência de Aminoácidos , Animais , Clonagem Molecular , DNA Nucleotidilexotransferase/química , DNA Nucleotidilexotransferase/metabolismo , DNA Complementar/química , DNA Complementar/isolamento & purificação , Evolução Molecular , Expressão Gênica , Isoenzimas/genética , Isoenzimas/metabolismo , Dados de Sequência Molecular , Filogenia , Alinhamento de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA