Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Diabetes ; 71(11): 2372-2383, 2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-36006465

RESUMO

Obesity with dysfunctional adipose cells is the major cause of the current epidemic of type 2 diabetes (T2D). We examined senescence in human adipose tissue cells from age- and BMI-matched individuals who were lean, obese, and obese with T2D. In obese individuals and, more pronounced, those with T2D, we found mature and fully differentiated adipose cells to exhibit increased senescence similar to what we previously have shown in the progenitor cells. The degree of adipose cell senescence was positively correlated with whole-body insulin resistance and adipose cell size. Adipose cell protein analysis revealed dysfunctional cells in T2D with increased senescence markers reduced PPAR-γ, GLUT4, and pS473AKT. Consistent with a recent study, we found the cell cycle regulator cyclin D1 to be increased in obese cells and further elevated in T2D cells, closely correlating with senescence markers, ambient donor glucose, and, more inconsistently, plasma insulin levels. Furthermore, fully differentiated adipose cells were susceptible to experimentally induced senescence and to conditioned medium increasing cyclin D1 and responsive to senolytic agents. Thus, fully mature human adipose cells from obese individuals, particularly those with T2D become senescent, and SASP secretion by senescent progenitor cells can play an important role in addition to donor hyperinsulinemia.


Assuntos
Diabetes Mellitus Tipo 2 , Resistência à Insulina , Insulinas , Humanos , Diabetes Mellitus Tipo 2/metabolismo , Ciclina D1/metabolismo , Meios de Cultivo Condicionados/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Tecido Adiposo/metabolismo , Obesidade/metabolismo , Resistência à Insulina/fisiologia , Glucose/metabolismo , Biomarcadores/metabolismo , Insulinas/metabolismo
2.
Aging Cell ; 21(3): e13557, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35146866

RESUMO

Senescence of adipose precursor cells (APC) impairs adipogenesis, contributes to the age-related subcutaneous adipose tissue (SAT) dysfunction, and increases risk of type 2 diabetes (T2D). First-degree relatives of T2D individuals (FDR) feature restricted adipogenesis, reflecting the detrimental effects of APC senescence earlier in life and rendering FDR more vulnerable to T2D. Epigenetics may contribute to these abnormalities but the underlying mechanisms remain unclear. In previous methylome comparison in APC from FDR and individuals with no diabetes familiarity (CTRL), ZMAT3 emerged as one of the top-ranked senescence-related genes featuring hypomethylation in FDR and associated with T2D risk. Here, we investigated whether and how DNA methylation changes at ZMAT3 promote early APC senescence. APC from FDR individuals revealed increases in multiple senescence markers compared to CTRL. Senescence in these cells was accompanied by ZMAT3 hypomethylation, which caused ZMAT3 upregulation. Demethylation at this gene in CTRL APC led to increased ZMAT3 expression and premature senescence, which were reverted by ZMAT3 siRNA. Furthermore, ZMAT3 overexpression in APC determined senescence and activation of the p53/p21 pathway, as observed in FDR APC. Adipogenesis was also inhibited in ZMAT3-overexpressing APC. In FDR APC, rescue of ZMAT3 methylation through senolytic exposure simultaneously downregulated ZMAT3 expression and improved adipogenesis. Interestingly, in human SAT, aging and T2D were associated with significantly increased expression of both ZMAT3 and the P53 senescence marker. Thus, DNA hypomethylation causes ZMAT3 upregulation in FDR APC accompanied by acquisition of the senescence phenotype and impaired adipogenesis, which may contribute to FDR predisposition for T2D.


Assuntos
Metilação de DNA , Diabetes Mellitus Tipo 2 , Adipócitos/metabolismo , Adipogenia/genética , Senescência Celular/genética , Metilação de DNA/genética , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Proteína Supressora de Tumor p53/metabolismo
3.
Diabetologia ; 62(10): 1835-1841, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31451866

RESUMO

Ageing and diabetes lead to similar organ dysfunction that is driven by parallel molecular mechanisms, one of which is cellular senescence. The abundance of senescent cells in various tissues increases with age, obesity and diabetes. Senescent cells have been directly implicated in the generation of insulin resistance. Recently, drugs that preferentially target senescent cells, known as senolytics, have been described and recently entered clinical trials. In this review, we explore the biological links between ageing and diabetes, specifically focusing on cellular senescence. We summarise the current data on cellular senescence in key target tissues associated with the development and clinical phenotypes of type 2 diabetes and discuss the therapeutic potential of targeting cellular senescence in diabetes.


Assuntos
Envelhecimento/metabolismo , Senescência Celular/fisiologia , Diabetes Mellitus Tipo 2/metabolismo , Envelhecimento/genética , Animais , Senescência Celular/genética , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patologia , Humanos
4.
Nat Commun ; 10(1): 2757, 2019 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-31227697

RESUMO

Inappropriate expansion of the adipose cells in the subcutaneous adipose tissue (SAT) is a characteristic of hypertrophic obesity and of individuals with genetic predisposition for T2D (first-degree relatives; FDR). It is associated with insulin resistance, a dysfunctional, adipose tissue and reduced adipogenesis. We examined the regulation of adipogenesis in human SAT precursor cells and found ZNF521 to be a critical regulator of early adipogenic commitment and precursor cells leaving the cell cycle. However, neither altered upstream signalling nor lack of SAT progenitor cells could explain the reduced adipogenesis in hypertrophic obesity. Instead, we show that progenitor cells undergoing poor differentiation are characterized by senescence, inability to suppress p53/P16INK4 and secretion of factors reducing adipogenesis in non-senescent cells. We found aging, FDR and established T2D to be associated with increased progenitor cell senescence, reduced adipogenesis and hypertrophic expansion of the SAT adipose cells.


Assuntos
Adipogenia , Senescência Celular/fisiologia , Proteínas de Ligação a DNA/metabolismo , Obesidade/patologia , Gordura Subcutânea/patologia , Adipócitos , Adulto , Idoso , Envelhecimento/fisiologia , Biópsia por Agulha , Células Cultivadas , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Proteínas de Ligação a DNA/genética , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/genética , Feminino , Predisposição Genética para Doença , Humanos , Hipertrofia/patologia , Masculino , Pessoa de Meia-Idade , Obesidade/etiologia , Cultura Primária de Células , Transdução de Sinais/fisiologia , Células-Tronco/fisiologia , Gordura Subcutânea/citologia , Proteína Supressora de Tumor p53/metabolismo , Adulto Jovem
5.
Nat Cell Biol ; 20(4): 443-454, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29593329

RESUMO

Excessive fat accumulation is a major risk factor for the development of type 2 diabetes mellitus and other common conditions, including cardiovascular disease and certain types of cancer. Here, we identify a mechanism that regulates adiposity based on the activator of autophagy TP53INP2. We report that TP53INP2 is a negative regulator of adipogenesis in human and mouse preadipocytes. In keeping with this, TP53INP2 ablation in mice caused enhanced adiposity, which was characterized by greater cellularity of subcutaneous adipose tissue and increased expression of master adipogenic genes. TP53INP2 modulates adipogenesis through autophagy-dependent sequestration of GSK3ß into late endosomes. GSK3ß sequestration was also dependent on ESCRT activity. As a result, TP53INP2 promotes greater ß-catenin levels and induces the transcriptional activity of TCF/LEF transcription factors. These results demonstrate a link between autophagy, sequestration of GSK3ß into late endosomes and inhibition of adipogenesis in vivo.


Assuntos
Adipócitos/enzimologia , Adipogenia , Tecido Adiposo/enzimologia , Adiposidade , Autofagia , Glicogênio Sintase Quinase 3 beta/metabolismo , Proteínas Nucleares/metabolismo , beta Catenina/metabolismo , Células 3T3-L1 , Adipócitos/patologia , Tecido Adiposo/patologia , Adulto , Animais , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Endossomos/enzimologia , Feminino , Glicogênio Sintase Quinase 3 beta/genética , Humanos , Hiperplasia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Proteínas Nucleares/genética , Transporte Proteico , Espanha , Suécia , Fatores de Transcrição TCF/genética , Fatores de Transcrição TCF/metabolismo , Fatores de Tempo , Ativação Transcricional , Via de Sinalização Wnt , beta Catenina/genética
6.
Atherosclerosis ; 241(1): 27-35, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25957567

RESUMO

The subcutaneous adipose tissue (SAT) is the largest and least harmful adipose depot to store excess lipids. However, SAT has a limited ability to expand and recruit new cells. When the SAT adipose cells become expanded (hypertrophic obesity), this leads to a dysregulated and dysfunctional SAT and the accumulation of ectopic fat in many depots. Increased hepatic and visceral fat are well-known ectopic fat depots and reflect the inability of SAT to accommodate excess fat. Ectopic fat also leads to paracrine and endocrine effects and promotes the metabolic profile of the Metabolic Syndrome. In addition, ectopic fat accumulation in and around the heart and vessels are considered to be active and cross talk with the tissues, thereby enhancing several aspects associated with the risk of developing cardiovascular disease and atherosclerosis.


Assuntos
Adipogenia , Doenças Cardiovasculares/etiologia , Obesidade/fisiopatologia , Gordura Subcutânea/fisiopatologia , Adipócitos/metabolismo , Animais , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/fisiopatologia , Metabolismo Energético , Humanos , Síndrome Metabólica/etiologia , Síndrome Metabólica/metabolismo , Síndrome Metabólica/fisiopatologia , Obesidade/complicações , Obesidade/metabolismo , Fatores de Risco , Transdução de Sinais , Gordura Subcutânea/metabolismo
7.
Trends Endocrinol Metab ; 26(4): 193-200, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25703677

RESUMO

The adipose tissue is crucial in regulating insulin sensitivity and risk for diabetes through its lipid storage capacity and thermogenic and endocrine functions. Subcutaneous adipose tissue (SAT) stores excess lipids through expansion of adipocytes (hypertrophic obesity) and/or recruitment of new precursor cells (hyperplastic obesity). Hypertrophic obesity in humans, a characteristic of genetic predisposition for diabetes, is associated with abdominal obesity, ectopic fat accumulation, and the metabolic syndrome (MS), while the ability to recruit new adipocytes prevents this. We review the regulation of adipogenesis, its relation to SAT expandability and the risks of ectopic fat accumulation, and insulin resistance. The actions of GLUT4 in SAT, including a novel family of lipids enhancing insulin sensitivity/secretion, and the function of bone morphogenetic proteins (BMPs) in white and beige/brown adipogenesis in humans are highlighted.


Assuntos
Adipogenia , Regulação para Baixo , Transportador de Glucose Tipo 4/metabolismo , Resistência à Insulina , Síndrome Metabólica/metabolismo , Modelos Biológicos , Gordura Subcutânea Abdominal/metabolismo , Tecido Adiposo Marrom/imunologia , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Marrom/patologia , Adiposidade , Animais , Transportador de Glucose Tipo 4/genética , Humanos , Hipertrofia , Metabolismo dos Lipídeos , Síndrome Metabólica/genética , Síndrome Metabólica/imunologia , Síndrome Metabólica/patologia , Especificidade de Órgãos , Gordura Subcutânea Abdominal/imunologia , Gordura Subcutânea Abdominal/patologia
8.
Diabetes ; 64(5): 1670-81, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25605802

RESUMO

The limited expandability of subcutaneous adipose tissue, due to reduced ability to recruit and differentiate new adipocytes, prevents its buffering effect in obesity and is characterized by expanded adipocytes (hypertrophic obesity). Bone morphogenetic protein-4 (BMP4) plays a key role in regulating adipogenic precursor cell commitment and differentiation. We found BMP4 to be induced and secreted by differentiated (pre)adipocytes, and BMP4 was increased in large adipose cells. However, the precursor cells exhibited a resistance to BMP4 owing to increased secretion of the BMP inhibitor Gremlin-1 (GREM1). GREM1 is secreted by (pre)adipocytes and is an inhibitor of both BMP4 and BMP7. BMP4 alone, and/or silencing GREM1, increased transcriptional activation of peroxisome proliferator-activated receptor γ and promoted the preadipocytes to assume an oxidative beige/brown adipose phenotype including markers of increased mitochondria and PGC1α. Driving white adipose differentiation inhibited the beige/brown markers, suggesting the presence of multipotent adipogenic precursor cells. However, silencing GREM1 and/or adding BMP4 during white adipogenic differentiation reactivated beige/brown markers, suggesting that increased BMP4 preferentially regulates the beige/brown phenotype. Thus, BMP4, secreted by white adipose cells, is an integral feedback regulator of both white and beige adipogenic commitment and differentiation, and resistance to BMP4 by GREM1 characterizes hypertrophic obesity.


Assuntos
Adipogenia/fisiologia , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo/metabolismo , Proteína Morfogenética Óssea 4/metabolismo , Adipogenia/genética , Adulto , Proteína Morfogenética Óssea 4/antagonistas & inibidores , Proteína Morfogenética Óssea 4/genética , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Regulação da Expressão Gênica/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Pessoa de Meia-Idade , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Obesidade/metabolismo , Interferência de RNA , RNA Interferente Pequeno
10.
Proc Natl Acad Sci U S A ; 110(7): 2563-8, 2013 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-23359679

RESUMO

Inability to recruit new adipose cells following weight gain leads to inappropriate enlargement of existing cells (hypertrophic obesity) associated with inflammation and a dysfunctional adipose tissue. We found increased expression of WNT1 inducible signaling pathway protein 2 (WISP2) and other markers of WNT activation in human abdominal s.c. adipose tissue characterized by hypertrophic obesity combined with increased visceral fat accumulation and insulin resistance. WISP2 activation in the s.c. adipose tissue, but not in visceral fat, identified the metabolic syndrome in equally obese individuals. WISP2 is a novel adipokine, highly expressed and secreted by adipose precursor cells. Knocking down WISP2 induced spontaneous differentiation of 3T3-L1 and human preadipocytes and allowed NIH 3T3 fibroblasts to become committed to the adipose lineage by bone morphogenetic protein 4 (BMP4). WISP2 forms a cytosolic complex with the peroxisome proliferator-activated receptor γ (PPARγ) transcriptional activator zinc finger protein 423 (Zfp423), and this complex is dissociated by BMP4 in a SMAD-dependent manner, thereby allowing Zfp423 to enter the nucleus, activate PPARγ, and commit the cells to the adipose lineage. The importance of intracellular Wisp2 protein for BMP4-induced adipogenic commitment and PPARγ activation was verified by expressing a mutant Wisp2 protein lacking the endoplasmic reticulum signal and secretion sequence. Secreted Wnt/Wisp2 also inhibits differentiation and PPARγ activation, albeit not through Zfp423 nuclear translocation. Thus adipogenic commitment and differentiation is regulated by the cross-talk between BMP4 and canonical WNT signaling and where WISP2 plays a key role. Furthermore, they link WISP2 with hypertrophic obesity and the metabolic syndrome.


Assuntos
Tecido Adiposo/metabolismo , Proteína Morfogenética Óssea 4/metabolismo , Proteínas de Sinalização Intercelular CCN/metabolismo , Células-Tronco Mesenquimais/fisiologia , PPAR gama/metabolismo , Proteínas Repressoras/metabolismo , Análise de Variância , Animais , Proteínas de Sinalização Intercelular CCN/genética , Diferenciação Celular/fisiologia , Proteínas de Ligação a DNA/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Immunoblotting , Imunoprecipitação , Camundongos , Microscopia de Fluorescência , Células NIH 3T3 , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Repressoras/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estatísticas não Paramétricas , Fatores de Transcrição/metabolismo
11.
Diabetes ; 61(5): 1217-24, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22447857

RESUMO

Overweight characterized by inappropriate expansion of adipose cells (hypertrophic obesity) is associated with the metabolic syndrome and is caused by an inability to recruit and differentiate new precursor cells. We examined the role of bone morphogenetic protein 4 (BMP4) and WNT activation in the regulation of human adipose cell differentiation. Cluster of differentiation (CD)14(+)/45(+) and CD31(+) cells were first removed before the remaining stromal vascular cells of human subcutaneous biopsy specimens were differentiated with/without different WNT inhibitors and/or BMP4. Inhibition of WNT and induction of Dickkopf 1 (DKK1) were markers of precursor cells undergoing excellent differentiation. The addition of DKK1 inhibited WNT activation and promoted adipogenesis in cells with a low degree of differentiation. The positive effect of DKK1, inhibiting cellular WNT activation by binding to the Kremen/LDL receptor-related protein receptors, was not seen with inhibitors of secreted WNT ligands. BMP4 increased differentiation, and BMP4 in the presence of DKK1 produced an additive effect. There was an apparent cross-talk between differentiation and commitment because BMP4 expression increased in differentiating adipocytes, and the addition of the BMP4 inhibitor, Noggin, reduced precursor cell differentiation. Thus, differentiated human adipose cells can promote adipogenesis via endogenous BMP4 activation, and the impaired adipogenesis in hypertrophic obesity is mainly due to an inability to suppress canonical WNT and to induce DKK1.


Assuntos
Adipogenia/fisiologia , Proteína Morfogenética Óssea 4/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Obesidade/metabolismo , Proteínas Wnt/antagonistas & inibidores , Células 3T3-L1 , Adipócitos/metabolismo , Adipócitos/patologia , Adulto , Idoso , Animais , Biomarcadores , Proteína Morfogenética Óssea 4/genética , Diferenciação Celular , Humanos , Hipertrofia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Ligantes , Camundongos , Pessoa de Meia-Idade , Obesidade/genética , PPAR gama/metabolismo , Proteínas Wnt/metabolismo , Adulto Jovem
12.
J Atheroscler Thromb ; 17(4): 332-41, 2010 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-20124732

RESUMO

Metabolic syndrome is associated with dysfunctional adipose tissue that is most likely a consequence of the enlargement of adipocytes and infiltration of macrophages into adipose tissue. Obesity and ectopic lipid deposition are major risk factors for diseases ranging from insulin resistance to type 2 diabetes and atherosclerosis. Enlargement of adipocytes, due to impaired adipocyte differentiation, leads to a chronic state of inflammation in the adipocytes and adipose tissue with a reduction in the secretion of adiponectin and increase in the secretion of proinflammatory cytokines such as interleukin (IL)-6, IL-8 and monocyte chemoattractant protein (MCP)-1. The secretion of cytokines like tumour necrosis factor (TNF)- alpha, mainly from macrophages, enhances local inflammation. These proinflammatory cytokines might also substantially affect cardiovascular function and morphology. Furthermore, a proinflammatory state in adipose tissue can lead to local insulin resistance with an impaired inhibitory effect of insulin on the release of FFAs and endothelial dysfunction that clearly promotes cardiovascular diseases and type 2 diabetes. The underlying mechanisms of ectopic fat accumulation in various tissues and the impact on metabolic syndrome and its association with insulin resistance are discussed.


Assuntos
Tecido Adiposo/fisiopatologia , Aterosclerose/fisiopatologia , Inflamação/fisiopatologia , Animais , Humanos
13.
J Biol Chem ; 285(18): 14031-41, 2010 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-20179324

RESUMO

Canonical Wnt ligands are secreted by several cell types in the adipose tissue. We examined if mature adipocytes can also be target cells and found that canonical Wnt activation by Wnt3a induced a marked dedifferentiation of both 3T3-L1 and human adipocytes. Typical adipogenic markers were reduced while undifferentiated cell markers like Pref-1/Dlk1, Wnt10b, and Gata2 were increased. The cells also became insulin-resistant with impaired upstream insulin signaling and reduced glucose uptake. Wnt3a stabilized beta-catenin in the absence of the LRP6 receptor and with maintained axin and Dickkopf-1 protein expression. PPARgamma was repressed and PPARgamma ligands could not restore the adipogenic markers or reduce the beta-catenin levels. The dedifferentiated adipocytes expressed the myofibroblast marker alpha-smooth muscle actin and were also susceptible to osteogenic transdifferentiation. These results identify a novel pathway in mature adipose cells that is critical for maintaining the normal adipocyte phenotype and insulin sensitivity.


Assuntos
Adipócitos/metabolismo , Desdiferenciação Celular/fisiologia , Resistência à Insulina/fisiologia , Transdução de Sinais/fisiologia , Proteínas Wnt/metabolismo , Células 3T3-L1 , Adipócitos/citologia , Animais , Antígenos de Diferenciação/biossíntese , Antígenos de Diferenciação/genética , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Células L , Proteínas Relacionadas a Receptor de LDL/genética , Proteínas Relacionadas a Receptor de LDL/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Camundongos , PPAR gama/genética , PPAR gama/metabolismo , Proteínas Wnt/genética , Proteína Wnt3 , Proteína Wnt3A , beta Catenina/genética , beta Catenina/metabolismo
14.
Am J Physiol Endocrinol Metab ; 297(5): E999-E1003, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19622783

RESUMO

Obesity is associated mainly with adipose cell enlargement in adult man (hypertrophic obesity), whereas the formation of new fat cells (hyperplastic obesity) predominates in the prepubertal age. Adipose cell size, independent of body mass index, is negatively correlated with whole body insulin sensitivity. Here, we review recent findings linking hypertrophic obesity with inflammation and a dysregulated adipose tissue, including local cellular insulin resistance with reduced IRS-1 and GLUT4 protein content. In addition, the number of preadipocytes in the abdominal subcutaneous adipose tissue capable of undergoing differentiation to adipose cells is reduced in hypertrophic obesity. This is likely to promote ectopic lipid accumulation, a well-known finding in these individuals and one that promotes insulin resistance and cardiometabolic risk. We also review recent results showing that TNFα, but not MCP-1, resistin, or IL-6, completely prevents normal adipogenesis in preadipocytes, activates Wnt signaling, and induces a macrophage-like phenotype in the preadipocytes. In fact, activated preadipocytes, rather than macrophages, may completely account for the increased release of chemokines and cytokines by the adipose tissue in obesity. Understanding the molecular mechanisms for the impaired preadipocyte differentiation in the subcutaneous adipose tissue in hypertrophic obesity is a priority since it may lead to new ways of treating obesity and its associated metabolic complications.


Assuntos
Adipogenia/fisiologia , Inflamação/fisiopatologia , Obesidade/fisiopatologia , Adipócitos/efeitos dos fármacos , Adipócitos/fisiologia , Tecido Adiposo/fisiologia , Diferenciação Celular/fisiologia , Humanos , Fator de Necrose Tumoral alfa/farmacologia , Proteínas Wnt/fisiologia
15.
Diabetes ; 58(7): 1550-7, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19351711

RESUMO

OBJECTIVE: We examined preadipocyte differentiation in obese and nonobese individuals and the effect of cytokines and wingless-type MMTV (mouse mammary tumor virus) integration site family, member 3A (Wnt3a) protein on preadipocyte differentiation and phenotype. RESEARCH DESIGN AND METHODS: Abdominal subcutaneous adipose tissue biopsies were obtained from a total of 51 donors with varying BMI. After isolation of the adipose and stromalvascular cells, inflammatory cells (CD14- and CD45-positive cells) were removed by immune magnetic separation. CD133-positive cells, containing early progenitor cells, were also isolated and quantified. The CD14- and CD45-negative preadipocytes were cultured with tumor necrosis factor (TNF)-alpha, interleukin (IL)-6, resistin, or Wnt3a with or without a differentiation cocktail. RESULTS: The number of preadipocytes able to differentiate to adipose cells was negatively correlated with both BMI and adipocyte cell size of the donors, whereas the number of CD133-positive cells was positively correlated with BMI, suggesting an impaired differentiation of preadipocytes in obesity. Cultured preadipocytes, like freshly isolated mature adipocytes, from obese individuals had an increased expression of mitogen-activated protein 4 kinase 4 (MAP4K4), which is known to inhibit peroxisome proliferator-activated receptor-gamma induction. TNF-alpha, but not IL-6 or resistin, increased Wnt10b, completely inhibited the normal differentiation of the preadipocytes, and instead induced a proinflammatory and macrophage-like phenotype of the cells. CONCLUSIONS: The apparent number of preadipocytes in the abdominal subcutaneous tissue that can undergo differentiation is reduced in obesity with enlarged fat cells, possibly because of increased MAP4K4 levels. TNF-alpha promoted a macrophage-like phenotype of the preadipocytes, including several macrophage markers. These results document the plasticity of human preadipocytes and the inverse relationship between lipid storage and proinflammatory capacity.


Assuntos
Abdome/patologia , Adipócitos/citologia , Adipócitos/patologia , Inflamação/fisiopatologia , Obesidade/patologia , Fator de Necrose Tumoral alfa/fisiologia , Proteínas Wnt/fisiologia , Antígeno AC133 , Adipócitos/transplante , Adulto , Idoso , Antígenos CD/análise , Cirurgia Bariátrica , Biópsia , Biópsia por Agulha , Índice de Massa Corporal , Diferenciação Celular , Tamanho Celular , Ensaio de Imunoadsorção Enzimática , Glicoproteínas/análise , Humanos , Inflamação/patologia , Pessoa de Meia-Idade , Peptídeos/análise , Reação em Cadeia da Polimerase , Proteína Wnt3 , Proteína Wnt3A
16.
Diabetes Metab Res Rev ; 24(8): 595-603, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18756581

RESUMO

Type 2 diabetes is the most common metabolic disorder today and has reached epidemic proportions in many countries. Insulin resistance and inflammation play a central role in the pathogenesis of type 2 diabetes and are present long before the onset of the disease. During this time, many of the complications associated with type 2 diabetes are initiated. Of major concern is the two- to fourfold increase in cardiovascular morbidity and mortality in this group compared to a nondiabetic population. Obesity, characterized by enlarged fat cells, and insulin resistance are, like type 2 diabetes, associated with impaired adipogenesis and a low-grade chronic inflammation that to a large extent emanates from the adipose tissue. Both these processes contribute to unfavourable alterations of the circulating levels of several bioactive molecules (adipokines) that are secreted from the adipose tissue, many of which have documented inhibitory effects on insulin sensitivity in the liver and peripheral tissues and, in addition, have negative effects on the cardiovascular system.Here we review current knowledge of the adipose tissue as an endocrine organ, the local and systemic effects of a chronic state of low-grade inflammation residing in the adipose tissue, and, in particular, the effects of inflammation and circulating adipokines on the vascular wall.


Assuntos
Adipocinas/fisiologia , Tecido Adiposo/irrigação sanguínea , Tecido Adiposo/fisiopatologia , Inflamação/fisiopatologia , Resistência à Insulina , Doenças Vasculares/fisiopatologia , Células 3T3 , Adipócitos/patologia , Adipócitos/fisiologia , Animais , Diabetes Mellitus Tipo 2/etiologia , Humanos , Macrófagos/fisiologia , Camundongos
17.
Arterioscler Thromb Vasc Biol ; 27(11): 2276-83, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17823366

RESUMO

The metabolic syndrome is associated with a dysregulated adipose tissue; in part a consequence of adipose cell enlargement and the associated infiltration of macrophages. Adipose cell enlargement leads to a proinflammatory state in the cells with reduced secretion of adiponectin and with increased secretion of several cytokines and chemokines including interleukin (IL)-6, IL-8, and MCP-1. MCP-1 has been shown to play an important role for the associated recruitment of macrophages into the adipose tissue. The increased release of cytokines leads to an impaired differentiation of the preadipocytes with reduced lipid accumulation and induction of adiponectin, thus promoting ectopic lipid storage. In particular tumor necrosis factor (TNF) alpha, but also IL-6, has been shown to induce these effects in preadipocytes and this is associated with an increased Wnt signaling maintaining the cells in an undifferentiated and proinflammatory state. The proinflammatory state in the adipose tissue also leads to a local insulin resistance including an impaired inhibitory effect of insulin on FFA release. The insulin resistance further supports the proinflammatory state because insulin, by itself, is both antilipolytic and antiinflammatory by antagonizing cytokine-induced activation of STAT signaling.


Assuntos
Tecido Adiposo/imunologia , Inflamação/fisiopatologia , Síndrome Metabólica/imunologia , Adipogenia/imunologia , Aterosclerose/imunologia , Citocinas/imunologia , Citocinas/metabolismo , Humanos , Insulina/fisiologia , Ativação de Macrófagos/imunologia , Síndrome Metabólica/fisiopatologia
18.
Biochem Biophys Res Commun ; 357(3): 700-6, 2007 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-17442272

RESUMO

Type 2 diabetes and obesity with enlarged fat cells are associated with low-grade systemic inflammation, impaired adipogenesis as well as the recruitment of inflammatory cells into the adipose tissue. Cytokines like TNFalpha and IL-6 are secreted by the inflammatory cells and have been shown to impair normal adipocyte differentiation. An important mechanism whereby these cytokines inhibit adipogenesis is by maintaining an active Wnt-signaling pathway. Also other cytokines like MCP-1 and resistin are involved in the inflammatory process and are secreted by macrophages. If these cytokines also affect Wnt-signaling and adipocyte differentiation is currently unclear. In the present study, we show that while TNFalpha is able to maintain an active Wnt-signaling, induce inflammation and completely block adipose cell differentiation, no effect was found by either MCP-1 or resistin on these processes. Addition of the thiazolidinedione, pioglitazone, was found to antagonize the effect of TNFalpha on the Wnt-signaling process and, consequently, promote adipogenesis.


Assuntos
Adipogenia/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Proteínas Wnt/fisiologia , Células 3T3-L1 , Adipócitos/citologia , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Animais , Western Blotting , Diferenciação Celular/efeitos dos fármacos , Quimiocina CCL2/metabolismo , Quimiocina CCL2/farmacologia , Ciclina D1/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Hipoglicemiantes/farmacologia , Ácidos Indolacéticos/metabolismo , Camundongos , Pioglitazona , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Resistina/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/fisiologia , Tiazolidinedionas/farmacologia , Fatores de Tempo , Fatores de Transcrição/genética , beta Catenina/metabolismo
19.
J Biol Chem ; 281(14): 9507-16, 2006 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-16464856

RESUMO

Obesity with enlarged fat cells is associated with high local concentrations of interleukin-6 (IL-6) and tumor necrosis factor alpha (TNFalpha) in the adipose tissue. We examined the effects of this inflammatory state on 3T3-L1 preadipocyte development and differentiation to mature adipose cells. Both IL-6 and TNFalpha impaired the normal differentiation pattern and lipid accumulation. However, IL-6 allowed a normal early induction of differentiation with inhibition of Wnt10b and Pref-1, whereas expression of CCAAT/enhancer-binding protein alpha, in contrast to peroxisome proliferator-activated receptor gamma, was markedly reduced. TNFalpha also allowed a normal early induction of differentiation, whereas the terminal differentiation to adipose cells was completely prevented. However, both cytokines induced an inflammatory phenotype of the cells but with different profiles. Remarkably, both IL-6 and TNFalpha maintained and augmented the canonical Wnt signaling associated with low axin and high low density lipoprotein receptor-related protein (LRD), Dishevelled, and beta-catenin levels. TNFalpha, but not IL-6, activated Wnt10b expression, whereas IL-6 increased the apparent phosphorylation of Dishevelled. Thus, both IL-6 and TNFalpha prevent the normal development of preadipocytes to fully differentiated adipose cells and, instead, promote an inflammatory phenotype of the adipocytes. These results provide an explanation as to why obesity and diabetes are associated with both local and systemic inflammation, insulin resistance, and ectopic lipid accumulation.


Assuntos
Adipócitos/fisiologia , Diferenciação Celular/fisiologia , Interleucina-6/fisiologia , Metabolismo dos Lipídeos , Obesidade/fisiopatologia , Fator de Necrose Tumoral alfa/fisiologia , Proteínas Wnt/fisiologia , Tecido Adiposo/fisiologia , Técnicas de Cultura de Células , Diabetes Mellitus/fisiopatologia , Humanos , Inflamação , Fenótipo , Transdução de Sinais
20.
Obes Res ; 12(3): 454-60, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15044662

RESUMO

OBJECTIVE: To examine the possibility that interleukin-6 (IL-6) can act as a paracrine regulator in adipose tissue by examining effects on adipogenic genes and measuring interstitial IL-6 concentrations in situ. RESEARCH METHODS AND PROCEDURES: Circulating and interstitial IL-6 concentrations in abdominal and femoral adipose tissue were measured using the calibrated microdialysis technique in 20 healthy male subjects. The effects of adipose cell enlargement on gene expression and IL-6 secretion were examined, as well as the effect of IL-6 in vitro on gene expression of adiponectin and other markers of adipocyte differentiation. RESULTS: The IL-6 concentration in the interstitial fluid was approximately 100-fold higher than that in plasma, suggesting that IL-6 may be a paracrine regulator of adipose tissue. This was further supported by the finding that adding IL-6 in vitro at similar concentrations down-regulated the expression of adiponectin, aP2, and PPARgamma-2 in cultured human adipose tissue. In addition, gene expression and release of IL-6, both in vivo and in vitro, correlated with adipose cell size. DISCUSSION: These data suggest that IL-6 may be a paracrine regulator of adipose tissue. Furthermore, increased adipose tissue production of IL-6 after hypertrophic enlargement of the adipose cells may detrimentally affect systemic insulin action by inducing adipose tissue dysfunction with impaired differentiation of the pre-adipocytes and/or adipocytes and lower adiponectin.


Assuntos
Adipócitos/citologia , Tecido Adiposo/fisiologia , Diferenciação Celular/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intercelular , Interleucina-6/fisiologia , Abdome , Adiponectina , Tecido Adiposo/química , Adulto , Diferenciação Celular/genética , Tamanho Celular , Fêmur , Expressão Gênica/efeitos dos fármacos , Humanos , Interleucina-6/genética , Interleucina-6/farmacologia , Masculino , Microdiálise , Pessoa de Meia-Idade , Proteínas/genética , Receptores Citoplasmáticos e Nucleares/genética , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA