Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Biol Chem ; 297(5): 101229, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34599964

RESUMO

Men have a statistically higher risk of metabolic and cardiovascular disease than premenopausal women, but the mechanisms mediating these differences are elusive. Chronic inflammation during obesity contributes to disease risk and is significantly more robust in males. Prior work demonstrated that compared with obese males, obese females have reduced proinflammatory adipose tissue macrophages (ATMs). Given the paucity of data on how sex hormones contribute to macrophage responses in obesity, we sought to understand the role of sex hormones in promoting obesity-induced myeloid inflammation. We used gonadectomy, estrogen receptor-deficient alpha chimeras, and androgen-insensitive mice to model sex hormone deficiency. These models were evaluated in diet-induced obesity conditions (high-fat diet [HFD]) and in vitro myeloid assays. We found that ovariectomy increased weight gain and adiposity. Ovariectomized females had increased ATMs and bone marrow myeloid colonies compared with sham-gonadectomized females. In addition, castrated males exposed to HFD had improved glucose tolerance, insulin sensitivity, and adiposity with fewer Ly6chi monocytes and bone marrow myeloid colonies compared with sham-gonadectomized males, although local adipose inflammation was enhanced. Similar findings were observed in androgen-insensitive mice; however, these mice had fewer CD11c+ ATMs, implying a developmental role for androgens in myelopoiesis and adipose inflammation. We concluded that gonadectomy results in convergence of metabolic and inflammatory responses to HFD between the sexes, and that myeloid estrogen receptor alpha contributes minimally to diet-induced inflammatory responses, whereas loss of androgen-receptor signaling improves metabolic and inflammatory outcomes. These studies demonstrate that sex hormones play a critical role in sex differences in obesity, metabolic dysfunction, and myeloid inflammation.


Assuntos
Tecido Adiposo/metabolismo , Hormônios Esteroides Gonadais/metabolismo , Macrófagos/metabolismo , Obesidade/metabolismo , Caracteres Sexuais , Animais , Dieta Hiperlipídica/efeitos adversos , Feminino , Inflamação/induzido quimicamente , Inflamação/metabolismo , Masculino , Camundongos , Obesidade/induzido quimicamente
2.
Aging (Albany NY) ; 12(2): 1725-1746, 2020 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-31983693

RESUMO

Aging, like obesity, is associated with metabolic and inflammatory alterations within adipose tissue in older individuals. Younger females are protected from adipose inflammation, but older post-menopausal females exhibit exaggerated visceral adiposity correlated with increased disease risk. Obesity accelerates the onset and progression of age-associated diseases, but it is unclear if aging and obesity drive adipose tissue dysfunction in a sexually dimorphic fashion. We investigated adipose tissue metabolism and inflammation in a diet-induced obesity model in young and old mice. We identified age related sex differences in adipose tissue macrophages (ATMs), fibrosis and lipid metabolism in male and female visceral fat depot (GWAT). Although aging normalized body weights between the sexes, females remained protected from proinflammatory ATMs and stimulated lipolysis failed to adversely affect the inflammatory state even with obesity. Older obese males had augmented CD11c+ ATMs and higher insulin levels, while females showed increased visceral adiposity and exaggerated Pparγ, and Pgc1α expression. Obesity in aging demonstrated similar expression of GWAT p53, p16, p21, Timp1 and Tgfß1 in both sexes. Our studies suggest that even with aging, female GWAT shows an attenuated inflammatory response compared to males due to an efficient oxidative metabolism combined with an active tissue remodeling state.


Assuntos
Adaptação Fisiológica , Tecido Adiposo/metabolismo , Envelhecimento/metabolismo , Metabolismo Energético , Obesidade/metabolismo , Tecido Adiposo/patologia , Adiposidade , Fatores Etários , Animais , Biomarcadores/metabolismo , Senescência Celular , Dieta Hiperlipídica , Matriz Extracelular/metabolismo , Feminino , Fibrose , Imuno-Histoquímica , Gordura Intra-Abdominal/metabolismo , Metabolismo dos Lipídeos , Lipólise , Fígado/imunologia , Fígado/metabolismo , Fígado/patologia , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Fatores Sexuais
3.
Biol Sex Differ ; 10(1): 16, 2019 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-30944030

RESUMO

BACKGROUND: Weight loss by surgery or lifestyle changes is strongly recommended for obese individuals to improve metabolic health, but the underlying impairments that persist from a history of obesity remain unclear. Recent investigations demonstrate a persistent inflammatory state with weight loss and bariatric surgery, but the mechanism and impact are not fully understood. Additionally, these studies have not been performed in females although women are the majority of individuals undergoing weight loss interventions. METHODS: The goal of this study was to determine the sex differences in metabolically induced inflammation after dietary weight loss (WL) or bariatric surgery. Following a 60% high-fat diet (HFD) for 12 weeks, C57Bl/6j mice underwent either a dietary switch to normal chow for WL or vertical sleeve gastrectomy (VSG) and were evaluated 8 weeks after intervention. WL effects on myelopoiesis were further evaluated with bone marrow chimeras. RESULTS: Both sexes had a decrease in adiposity and total weight following WL or VSG intervention. With HFD, females had very little inflammation and no further increase with WL, but males had persistent inflammation even after WL despite metabolic improvement. Interestingly, after VSG, myeloid inflammation was increased in the livers of males and to a lesser extent in females. CONCLUSIONS: These studies demonstrate that regardless of sex, it is critical to assess an individuals' history of obesity rather than just rely on current weight status in medical decision-making. There are long-lasting effects on tissue inflammation in both sexes especially with surgical weight loss. Dietary change is overall most effective to improve meta-inflammation in obese males on its own or in combination with surgical weight loss.


Assuntos
Obesidade/dietoterapia , Obesidade/cirurgia , Caracteres Sexuais , Redução de Peso , Adiposidade , Animais , Cirurgia Bariátrica , Glicemia , Dieta Hiperlipídica , Dieta Redutora , Feminino , Inflamação , Insulina/sangue , Leucócitos/imunologia , Fígado/imunologia , Masculino , Camundongos Endogâmicos C57BL , Obesidade/sangue
4.
Endocrinology ; 160(2): 293-312, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30544158

RESUMO

Males are known to have profound adipose tissue macrophage (ATM) accumulation in gonadal white adipose tissue (GWAT) during obesity, whereas females are protected from such an inflammatory response even with increased adiposity. The inflammatory tone in males is linked to insulin resistance and might be the underlying cause for sex differences in metabolic disease. Factors regulating the meta-inflammatory response remain unclear but enhanced lipid storage in females may explain the reduced inflammatory response to high-fat diets. In this study, we evaluated lean and obese females with stimulated lipolysis to understand whether a stress release of free fatty acids (FFAs) could induce female ATMs. We demonstrate that in both lean and obese females, GWAT CD11c- resident ATMs accumulate with ß-3 adrenergic receptor-stimulated lipolysis. Lipolysis elevated serum FFA, triglyceride, and IL-6 levels in females that corresponded to significant phosphorylated hormone-sensitive lipase and adipose triglyceride lipase protein expression in obese female GWAT compared with males. Increased lipolytic response in obese females was associated with crown-like structures and induced Il6, Mcp1, Arg1, and Mgl1 expression in obese female GWAT, suggesting an environment of lipid clearance and adipose remodeling. With this finding we next investigated whether lipid storage and lipolytic mediators differed by sex. Diacylglycerol, ceramides, phospholipids, and certain fatty acid species associated with inflammation were elevated in male GWAT compared with obese female GWAT. Overall, our data demonstrate a role for GWAT lipid storage and lipolytic metabolites to induce inflammation in males and induce remodeling in females that might explain sex differences in overall metabolic health.


Assuntos
Tecido Adiposo/imunologia , Lipólise , Obesidade/imunologia , Caracteres Sexuais , Animais , Dieta Hiperlipídica , Feminino , Lipase/metabolismo , Macrófagos , Masculino , Camundongos Endogâmicos C57BL , Receptores Adrenérgicos beta 3/metabolismo , Esterol Esterase/metabolismo , Ativação Transcricional
5.
J Biol Chem ; 293(23): 8775-8786, 2018 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-29636416

RESUMO

Obesity-induced chronic inflammation is associated with metabolic disease. Results from mouse models utilizing a high-fat diet (HFD) have indicated that an increase in activated macrophages, including CD11c+ adipose tissue macrophages (ATMs), contributes to insulin resistance. Obesity primes myeloid cell production from hematopoietic stem cells (HSCs) and Toll-like receptor 4 (TLR4), and the downstream TIR domain-containing adapter protein-inducing interferon-ß (TRIF)- and MyD88-mediated pathways regulate production of similar myeloid cells after lipopolysaccharide stimulation. However, the role of these pathways in HFD-induced myelopoiesis is unknown. We hypothesized that saturated fatty acids and HFD alter myelopoiesis by activating TLR4 pathways in HSCs, differentially producing pro-inflammatory CD11c+ myeloid cells that contribute to obesity-induced metabolic disease. Results from reciprocal bone marrow transplants (BMTs) with Tlr4-/- and WT mice indicated that TLR4 is required for HFD-induced myelopoiesis and production of CD11c+ ATMs. Experiments with homozygous knockouts of Irakm (encoding a suppressor of MyD88 inactivation) and Trif in competitive BMTs revealed that MyD88 is required for HFD expansion of granulocyte macrophage progenitors and that Trif is required for pregranulocyte macrophage progenitor expansion. A comparison of WT, Tlr4-/-, Myd88-/-, and Trif-/- mice on HFD demonstrated that TLR4 plays a role in the production of CD11c+ ATMs, and both Myd88-/- and Trif-/- mice produced fewer ATMs than WT mice. Moreover, HFD-induced TLR4 activation inhibited macrophage proliferation, leading to greater accumulation of recruited CD11c+ ATMs. Our results indicate that HFD potentiates TLR4 and both its MyD88- and TRIF-mediated downstream pathways within progenitors and adipose tissue and leads to macrophage polarization.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/imunologia , Antígeno CD11c/imunologia , Macrófagos/patologia , Fator 88 de Diferenciação Mieloide/imunologia , Mielopoese , Obesidade/patologia , Receptor 4 Toll-Like/imunologia , Tecido Adiposo/imunologia , Tecido Adiposo/patologia , Animais , Dieta Hiperlipídica/efeitos adversos , Inflamação/etiologia , Inflamação/imunologia , Inflamação/patologia , Macrófagos/imunologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Obesos , Obesidade/complicações , Obesidade/etiologia , Obesidade/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...