Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Diabetes ; 70(3): 720-732, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33323395

RESUMO

CD11c+ macrophages/dendritic cells (MDCs) are increased and display the classically activated M1-like phenotype in obese adipose tissue (AT) and may contribute to AT inflammation and insulin resistance. Stat1 is a key transcription factor for MDC polarization into the M1-like phenotype. Here, we examined the role of Stat1 in obesity-induced AT MDC polarization and inflammation and insulin resistance using mice with specific knockout of Stat1 in MDCs (cKO). Stat1 was upregulated and phosphorylated, indicating activation, early and persistently in AT and AT MDCs of wild-type mice fed a high-fat diet (HFD). Compared with littermate controls, cKO mice fed an HFD (16 weeks) had reductions in MDC (mainly CD11c+ macrophage) M1-like polarization and interferon-γ-expressing T-helper type 1 (Th1) cells but increases in interleukin 5-expressing Th2 cells and eosinophils in perigonadal and inguinal AT, and enhanced inguinal AT browning, with increased energy expenditure. cKO mice compared with controls also had significant reductions in triglyceride content in the liver and skeletal muscle and exhibited improved insulin sensitivity and glucose tolerance. Taken together, our results demonstrate that Stat1 in MDCs plays an important role in obesity-induced MDC M1-like polarization and AT inflammation and contributes to insulin resistance and metabolic dysfunctions in obese mice.


Assuntos
Tecido Adiposo/metabolismo , Antígeno CD11c/metabolismo , Dieta Hiperlipídica/efeitos adversos , Inflamação/metabolismo , Resistência à Insulina/fisiologia , Obesidade/imunologia , Obesidade/metabolismo , Fator de Transcrição STAT1/metabolismo , Adulto , Animais , Western Blotting , Antígeno CD11c/genética , Células Cultivadas , Feminino , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Inflamação/genética , Inflamação/imunologia , Resistência à Insulina/genética , Interleucina-5/metabolismo , Masculino , Camundongos , Obesidade/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT1/genética
2.
J Clin Lipidol ; 14(6): 850-858, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33011137

RESUMO

BACKGROUND: Accumulation of lipoprotein X (LpX) in blood can cause severe hypercholesterolemia and cutaneous xanthomas. Monocytes sensitively sense lipid changes in circulation and contribute to inflammation. However, how monocytes respond to LpX is undefined. OBJECTIVE: We examined the phenotype of monocytes from a patient, who had LpX, severe hypercholesterolemia, and extensive cutaneous xanthomas, and effects of semiselective plasmapheresis therapy (SPPT). METHOD: Fluorescence-activated cell sorting and adhesion assays were used to examine monocyte phenotype and ex vivo oxidized low-density lipoprotein uptake and adhesion in the patient before and after treatment with SPPT. Effects of plasma from the patient on the phenotype and adhesion of monocytes from a healthy participant were determined. RESULTS: SPPT improved hypercholesterolemia and cutaneous xanthomas. Before treatment, the patient had lower frequency of nonclassical monocytes but higher frequency of intermediate monocytes than the control participant. Before treatment, monocytes from the patient with LpX showed more intracellular lipid accumulation, alterations in several cell surface markers and intracellular cytokines, as well as enhanced oxidized low-density lipoprotein uptake and reduced adhesion compared with control. After SPPT, the phenotypes of monocytes from the patient with LpX were similar to control monocytes. Incubation with plasma from the patient before treatment as compared with plasma from the control participant or the patient after treatment increased CD11c expression and adhesion of monocytes from a healthy participant. CONCLUSION: LpX-induced hypercholesterolemia increased lipid accumulation and altered the phenotype of monocytes, which may contribute to cutaneous xanthoma development. Removal of LpX by SPPT reduced lipid accumulation and improved monocyte phenotype, likely contributing to xanthoma resolution.


Assuntos
Lipoproteína-X/sangue , Monócitos/citologia , Fenótipo , Humanos , Hipercolesterolemia/sangue , Hipercolesterolemia/imunologia
3.
Cholesterol ; 2014: 843468, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24672716

RESUMO

Our current understanding of oxysterol metabolism during different disease states such as obesity and dyslipidemia is limited. Therefore, the aim of this study was to determine the effect of diet-induced obesity on the tissue distribution of various oxysterols and the mRNA expression of key enzymes involved in oxysterol metabolism. To induce obesity, male C57BL/6J mice were fed a high fat-cholesterol diet for 24 weeks. Following diet-induced obesity, plasma levels of 4 ß -hydroxycholesterol, 5,6 α -epoxycholesterol, 5,6 ß -epoxycholesterol, 7 α -hydroxycholesterol, 7 ß -hydroxycholesterol, and 27-hydroxycholesterol were significantly (P < 0.05) increased. In the liver and adipose tissue of the obese mice, 4 ß -hydroxycholesterol was significantly (P < 0.05) increased, whereas 27-hydroxycholesterol was increased only in the adipose tissue. No significant changes in either hepatic or adipose tissue mRNA expression were observed for oxysterol synthesizing enzymes 4 ß -hydroxylase, 27-hydroxylase, or 7 α -hydroxylase. Hepatic mRNA expression of SULT2B1b, a key enzyme involved in oxysterol detoxification, was significantly (P < 0.05) elevated in the obese mice. Interestingly, the appearance of the large HDL1 lipoprotein was observed with increased oxysterol synthesis during obesity. In diet-induced obese mice, dietary intake and endogenous enzymatic synthesis of oxysterols could not account for the increased oxysterol levels, suggesting that nonenzymatic cholesterol oxidation pathways may be responsible for the changes in oxysterol metabolism.

4.
Arterioscler Thromb Vasc Biol ; 34(1): 34-43, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24158516

RESUMO

OBJECTIVE: T cells, particularly CD8(+) T cells, are major participants in obesity-linked adipose tissue (AT) inflammation. We examined the mechanisms of CD8(+) T-cell accumulation and activation in AT and the role of CD11a, a ß2 integrin. APPROACH AND RESULTS: CD8(+) T cells in AT of obese mice showed activated phenotypes with increased proliferation and interferon-γ expression. In vitro, CD8(+) T cells from mouse AT displayed increased interferon-γ expression and proliferation to stimulation with interleukin-12 and interleukin-18, which were increased in obese AT. CD11a was upregulated in CD8(+) T cells in obese mice. Ablation of CD11a in obese mice dramatically reduced T-cell accumulation, activation, and proliferation in AT. Adoptive transfer showed that CD8(+) T cells from wild-type mice, but not from CD11a-deficient mice, infiltrated into AT of recipient obese wild-type mice. CD11a deficiency also reduced tumor necrosis factor-α-producing and interleukin-12-producing macrophages in AT and improved insulin resistance. CONCLUSIONS: Combined action of cytokines in obese AT induces proliferative response of CD8(+) T cells locally, which, along with increased infiltration, contributes to CD8(+) T-cell accumulation and activation in AT. CD11a plays a crucial role in AT inflammation by participating in T-cell infiltration and activation.


Assuntos
Tecido Adiposo/imunologia , Antígeno CD11a/metabolismo , Linfócitos T CD8-Positivos/imunologia , Quimiotaxia de Leucócito , Ativação Linfocitária , Obesidade/imunologia , Paniculite/imunologia , Transferência Adotiva , Animais , Antígeno CD11a/genética , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Mediadores da Inflamação/metabolismo , Resistência à Insulina , Interferon gama/metabolismo , Interleucina-12/metabolismo , Interleucina-18/metabolismo , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Obesos , Obesidade/sangue , Obesidade/genética , Paniculite/sangue , Paniculite/genética , Fatores de Tempo , Fator de Necrose Tumoral alfa/metabolismo , Aumento de Peso
5.
Atherosclerosis ; 223(2): 342-9, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22770993

RESUMO

OBJECTIVE: To identify the role of triglyceride-rich lipoproteins (TGRLs) and apoE, a major apolipoprotein in TGRLs, in adipose tissue inflammation with high-fat diet (HFD)-induced obesity. METHODS: Male apoE(-/-) and C57BL/6J wild-type (WT) mice fed HFD for 12 weeks were assessed for metabolic and inflammatory parameters. ApoE(-/-) and WT mice were orally gavaged with [(3)H]palmitic acid to examine the role of apoE in fat delivery to adipose tissue. VLDL from obese apoE(-/-) mice were intravenously injected into lean WT or apoE(-/-) mice to test potential contribution of TGRLs-derived fat delivery to inflammation in adipose tissue and the role of apoE. RESULTS: ApoE(-/-) mice gained less body weight, and had less fat mass and lower triglyceride levels in skeletal muscle than WT. ApoE(-/-) mice on HFD had better insulin sensitivity than WT even when comparing body weight-matched mice. Compared to WT mice, apoE(-/-) mice on HFD had lower levels of inflammatory cytokines/chemokines and CD11c in adipose tissue, and lower levels of inflammatory markers in skeletal muscle. At 6 h after oral gavage with [(3)H]palmitic acid, incorporation of [(3)H]palmitic acid into adipose tissue and skeletal muscle was lower in apoE(-/-) mice. After repeated daily injection for 3 days, VLDL from obese apoE(-/-) mice induced inflammation in adipose tissue of recipient WT but not apoE(-/-) mice. CONCLUSION: In HFD-induced obesity, apoE plays an important role in inflammation in adipose tissue and skeletal muscle, likely by mediating TGRL-derived fat delivery to these tissues.


Assuntos
Tecido Adiposo/metabolismo , Apolipoproteínas E/metabolismo , Lipoproteínas VLDL/metabolismo , Obesidade/metabolismo , Paniculite/metabolismo , Triglicerídeos/metabolismo , Tecido Adiposo/imunologia , Adiposidade , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Glicemia/metabolismo , Citocinas/metabolismo , Dieta Hiperlipídica , Modelos Animais de Doenças , Mediadores da Inflamação/metabolismo , Injeções Intravenosas , Insulina/sangue , Lipoproteínas VLDL/administração & dosagem , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/metabolismo , Obesidade/etiologia , Obesidade/genética , Obesidade/imunologia , Estresse Oxidativo , Ácido Palmítico/administração & dosagem , Ácido Palmítico/metabolismo , Paniculite/etiologia , Paniculite/genética , Paniculite/imunologia , Paniculite/prevenção & controle , Fatores de Tempo , Triglicerídeos/administração & dosagem , Aumento de Peso
6.
Atherosclerosis ; 219(1): 100-8, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21824616

RESUMO

OBJECTIVE: We studied the effects of weight loss induced by either a low-fat normal diet (ND) or restriction of high-fat diet (HFD) on hepatic steatosis, inflammation in the liver and adipose tissue (AT), and blood monocytes of obese mice. METHODS: In mice with HFD-induced obesity, weight loss was achieved by switching from HFD to ND and maintaining on ND ad libitum or by restricting HFD intake to match body weight of mice with ND-induced weight loss. After diet interventions for 4 weeks, hepatic steatosis, hepatic and AT inflammation, and blood CD11c(+) monocytes were examined. RESULTS: At 4 weeks after switching diets, body weight was reduced by 23% from baseline. To achieve the same reduced body weight required restricting calorie intake from HFD. Weight loss with either ND or HFD restriction decreased body fat mass and ameliorated liver steatosis; both effects were greater with ND-induced weight loss than HFD restriction-induced weight loss. Weight loss with ND but not HFD restriction normalized blood CD11c(+) monocytes and attenuated hepatic inflammation assessed by chemokine and CD11c expression. In contrast, weight loss with HFD restriction significantly reduced chemokine levels and CD11c(+) cells in AT compared to obese controls, and tended to reduce AT chemokines and CD11c(+) cells more than ND-induced weight loss. CONCLUSION: In mice with diet-induced obesity, weight loss with ND was superior in alleviating hepatic inflammation and steatosis, whereas weight loss with HFD calorie restriction provided greater amelioration of AT inflammation.


Assuntos
Tecido Adiposo/patologia , Fígado/patologia , Obesidade/metabolismo , Redução de Peso/fisiologia , Tecido Adiposo/efeitos dos fármacos , Animais , Antígeno CD11c/biossíntese , Restrição Calórica , Quimiocina CCL2/sangue , Dieta com Restrição de Gorduras , Dieta Hiperlipídica , Fígado Gorduroso/etiologia , Fígado Gorduroso/terapia , Inflamação/etiologia , Inflamação/terapia , Fígado/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/fisiologia
7.
Arterioscler Thromb Vasc Biol ; 30(2): 186-92, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19910635

RESUMO

OBJECTIVE: To examine CD11c, a beta(2)-integrin, on adipose tissue (AT) leukocytes and blood monocytes and its role in diet-induced obesity. METHODS AND RESULTS: High-fat diet-induced obese C57BL/6 mice, CD11c-deficient mice, and obese humans were studied. CD11c, leukocytes, and chemokines/cytokines were examined in AT and/or blood by flow cytometry, RNase protection assay, quantitative polymerase chain reaction, or enzyme-linked immunosorbent assay. Obese C57BL/6 mice had increased CD11c in AT and blood compared with lean controls. CD11c messenger RNA positively correlated with monocyte chemoattractant protein 1 in human visceral AT. Obese humans with metabolic syndrome had a higher CD11c level on blood monocytes compared with lean humans. Low-fat diet-induced weight loss reduced blood monocyte CD11c in obese mice and humans. Mouse and human monocyte CD11c levels and mouse AT CD11c messenger RNA correlated with insulin resistance. CD11c deficiency in mice did not alter weight gain but decreased inflammation, evidenced by a lower T-cell number and reduced levels of major histocompatibility complex class II, C-C chemokine ligand 2 (CCL5), CCL4, and interferon gamma in AT, and ameliorated insulin resistance and glucose intolerance associated with diet-induced obesity. CONCLUSIONS: Diet-induced obesity increased CD11c in both AT and blood in mice and humans. CD11c plays an important role in T-cell accumulation and activation in AT, and contributes to insulin resistance associated with obesity.


Assuntos
Antígeno CD11c/metabolismo , Inflamação/imunologia , Gordura Intra-Abdominal/imunologia , Leucócitos/imunologia , Monócitos/imunologia , Obesidade/imunologia , Animais , Biomarcadores/metabolismo , Antígeno CD11b/genética , Antígeno CD11c/sangue , Antígeno CD11c/genética , Estudos de Casos e Controles , Quimiocina CCL2/genética , Quimiocinas/metabolismo , Dieta Redutora , Gorduras na Dieta , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Intolerância à Glucose/imunologia , Humanos , Inflamação/fisiopatologia , Resistência à Insulina , Gordura Intra-Abdominal/fisiopatologia , Ativação de Macrófagos , Masculino , Síndrome Metabólica/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/complicações , Obesidade/dietoterapia , Obesidade/fisiopatologia , Reação em Cadeia da Polimerase , RNA Mensageiro/metabolismo , Linfócitos T/imunologia , Resultado do Tratamento , Redução de Peso
8.
Circulation ; 115(8): 1029-38, 2007 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-17296858

RESUMO

BACKGROUND: Obesity is associated with chronic inflammation, which includes increased macrophage accumulation in adipose tissue (AT) and upregulation of chemokines and cytokines. T cells also play important roles in chronic inflammatory diseases such as atherosclerosis but have not been well studied in obesity. METHODS AND RESULTS: Flow cytometric analysis showed higher numbers of T cells and macrophages in AT of diet-induced obese insulin-resistant male mice than in lean mice and obese females (P<0.05). RNase protection assay, ELISA, and flow cytometry indicated gender-dependent upregulation of mRNA and protein levels of regulated on activation, normal T cell expressed and secreted (RANTES) and its receptor CCR5 in AT of obese mice. Adipocytes, stromal/vascular cells from mouse AT, and human and murine adipocytes expressed RANTES. RANTES mRNA levels were negatively correlated with adiponectin in mouse AT. Adiponectin-deficient mice fed high-fat diet showed higher RANTES mRNA levels in AT than wild-type mice. Activated T cells coincubated with preadipocytes in vitro significantly suppressed preadipocyte-to-adipocyte differentiation. Obese humans with metabolic syndrome had higher mRNA levels of RANTES and CCR5 in subcutaneous AT than lean humans. RANTES and CCR5 mRNA levels were significantly higher in visceral than subcutaneous AT of morbidly obese humans. RANTES mRNA levels were positively correlated with CD3 and CD11b in human visceral AT. CONCLUSIONS: Obesity is associated with increased accumulation of T cells and macrophages in AT, which may play important roles in obesity-related disease by influencing preadipocyte/adipocyte functions. RANTES is an adipokine that is upregulated in AT by obesity in both mice and humans.


Assuntos
Adiponectina/fisiologia , Tecido Adiposo/imunologia , Tecido Adiposo/metabolismo , Quimiocina CCL5/fisiologia , Obesidade/metabolismo , Linfócitos T/imunologia , Adipócitos/química , Adiponectina/genética , Animais , Antígeno CD11b/genética , Complexo CD3/genética , Diferenciação Celular , Quimiocina CCL5/análise , Quimiocina CCL5/genética , Fatores Quimiotáticos/análise , Feminino , Humanos , Fígado/química , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/imunologia , RNA Mensageiro/análise , Receptores CCR5/análise , Receptores CCR5/genética , Linfócitos T/fisiologia , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...