Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Obesity (Silver Spring) ; 28(6): 1086-1097, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32281747

RESUMO

OBJECTIVE: Weight regain after weight loss is common, and there is evidence to suggest negative effects on health because of weight cycling. This study sought to investigate the impact of weight regain in formerly obese mice on adipose tissue architecture and stromal cell function. METHODS: A diet-switch model was employed for obesity induction, weight loss, and weight regain in mice. Flow cytometry quantified adipose tissue leukocytes in adipose tissue. Liver and adipose tissue depots were compared to determine tissue-specific effects of weight cycling. RESULTS: Epididymal white adipose tissue of formerly obese mice failed to expand in response to repeat exposure to high-fat diet and retained elevated numbers of macrophages and T cells. Weight regain was associated with disproportionally elevated liver mass, hepatic triglyceride content, serum insulin concentration, and serum transaminase concentration. These effects occurred despite an extended 6-month weight loss cycle and they demonstrate that formerly obese mice maintain durable alterations in their physiological response to weight regain. Conditioned media from epididymal adipose tissue of formerly obese mice inhibited adipogenesis of 3T3-L1 preadipocytes, suggesting a potential mechanism to explain failed epididymal adipose tissue expansion during weight regain. CONCLUSIONS: Metabolic abnormalities related to defects in adipose tissue expansion and ongoing dysfunction manifest in formerly obese mice during weight regain.


Assuntos
Tecido Adiposo/metabolismo , Fígado Gorduroso/metabolismo , Obesidade/metabolismo , Aumento de Peso/fisiologia , Animais , Dieta Hiperlipídica , Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos
2.
J Leukoc Biol ; 103(4): 615-628, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29493813

RESUMO

Obesity-related changes in adipose tissue leukocytes, in particular adipose tissue macrophages (ATMs) and dendritic cells (ATDCs), are implicated in metabolic inflammation, insulin resistance, and altered regulation of adipocyte function. We evaluated stromal cell and white adipose tissue (WAT) expansion dynamics with high fat diet (HFD) feeding for 3-56 days, quantifying ATMs, ATDCs, endothelial cells (ECs), and preadipocytes (PAs) in visceral epididymal WAT and subcutaneous inguinal WAT. To better understand mechanisms of the early response to obesity, we evaluated ATM proliferation and lipid accumulation. ATMs, ATDCs, and ECs increased with rapid WAT expansion, with ATMs derived primarily from a CCR2-independent resident population. WAT expansion stimulated proliferation in resident ATMs and ECs, but not CD11c+ ATMs or ATDCs. ATM proliferation was unperturbed in Csf2- and Rag1-deficient mice with WAT expansion. Additionally, ATM apoptosis decreased with WAT expansion, and proliferation and apoptosis reverted to baseline with weight loss. Adipocytes reached maximal hypertrophy at 28 days of HFD, coinciding with a plateau in resident ATM accumulation and the appearance of lipid-laden CD11c+ ATMs in visceral epididymal WAT. ATM increases were proportional to tissue expansion and adipocyte hypertrophy, supporting adipocyte-mediated regulation of resident ATMs. The appearance of lipid-laden CD11c+ ATMs at peak adipocyte size supports a role in responding to ectopic lipid accumulation within adipose tissue. In contrast, ATDCs increase independently of proliferation and may be derived from circulating precursors. These changes precede and establish the setting in which large-scale adipose tissue infiltration of CD11c+ ATMs, inflammation, and adipose tissue dysfunction contributes to insulin resistance.


Assuntos
Tecido Adiposo Branco/citologia , Proliferação de Células , Células Dendríticas/citologia , Endotélio Vascular/citologia , Lipídeos/análise , Macrófagos/citologia , Obesidade/fisiopatologia , Tecido Adiposo Branco/metabolismo , Animais , Células Dendríticas/metabolismo , Dieta Hiperlipídica/efeitos adversos , Endotélio Vascular/metabolismo , Feminino , Humanos , Inflamação/fisiopatologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL
3.
J Biol Chem ; 292(22): 9051-9062, 2017 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-28424263

RESUMO

Obesity and its associated complications such as insulin resistance and non-alcoholic fatty liver disease are reaching epidemic proportions. In mice, the TGF-ß superfamily is implicated in the regulation of white and brown adipose tissue differentiation. The kielin/chordin-like protein (KCP) is a secreted regulator of the TGF-ß superfamily pathways that can inhibit both TGF-ß and activin signals while enhancing bone morphogenetic protein (BMP) signaling. However, KCP's effects on metabolism and obesity have not been studied in animal models. Therefore, we examined the effects of KCP loss or gain of function in mice that were maintained on either a regular or a high-fat diet. KCP loss sensitized the mice to obesity and associated complications such as glucose intolerance and adipose tissue inflammation and fibrosis. In contrast, transgenic mice that expressed KCP in the kidney, liver, and adipose tissues were resistant to developing high-fat diet-induced obesity and had significantly reduced white adipose tissue. Moreover, KCP overexpression shifted the pattern of SMAD signaling in vivo, increasing the levels of phospho (P)-SMAD1 and decreasing P-SMAD3. Adipocytes in culture showed a cell-autonomous effect in response to added TGF-ß1 or BMP7. Metabolic profiling indicated increased energy expenditure in KCP-overexpressing mice and reduced expenditure in the KCP mutants with no effect on food intake or activity. These findings demonstrate that shifting the TGF-ß superfamily signaling with a secreted protein can alter the physiology and thermogenic properties of adipose tissue to reduce obesity even when mice are fed a high-fat diet.


Assuntos
Adipócitos/metabolismo , Proteínas de Transporte/metabolismo , Gorduras na Dieta/efeitos adversos , Síndrome Metabólica/metabolismo , Obesidade/metabolismo , Transdução de Sinais , Adipócitos/patologia , Animais , Proteína Morfogenética Óssea 7/genética , Proteína Morfogenética Óssea 7/metabolismo , Proteínas de Transporte/genética , Gorduras na Dieta/farmacologia , Síndrome Metabólica/induzido quimicamente , Síndrome Metabólica/genética , Síndrome Metabólica/patologia , Camundongos , Camundongos Knockout , Obesidade/induzido quimicamente , Obesidade/genética , Obesidade/patologia , Especificidade de Órgãos/genética , Proteína Smad3/genética , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
4.
Diabetes ; 66(2): 392-406, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28108608

RESUMO

Obesity causes dramatic proinflammatory changes in the adipose tissue immune environment, but relatively little is known regarding how this inflammation responds to weight loss (WL). To understand the mechanisms by which meta-inflammation resolves during WL, we examined adipose tissue leukocytes in mice after withdrawal of a high-fat diet. After 8 weeks of WL, mice achieved similar weights and glucose tolerance values as age-matched lean controls but showed abnormal insulin tolerance. Despite fat mass normalization, total and CD11c+ adipose tissue macrophage (ATM) content remained elevated in WL mice for up to 6 months and was associated with persistent fibrosis in adipose tissue. ATMs in formerly obese mice demonstrated a proinflammatory profile, including elevated expression of interferon-γ, tumor necrosis factor-α, and interleukin-1ß. T-cell-deficient Rag1-/- mice showed a degree of ATM persistence similar to that in WT mice, but with reduced inflammatory gene expression. ATM proliferation was identified as the predominant mechanism by which ATMs are retained in adipose tissue with WL. Our study suggests that WL does not completely resolve obesity-induced ATM activation, which may contribute to the persistent adipose tissue damage and reduced insulin sensitivity observed in formerly obese mice.


Assuntos
Tecido Adiposo/imunologia , Proliferação de Células , Macrófagos/imunologia , Obesidade/imunologia , Redução de Peso/imunologia , Tecido Adiposo/citologia , Animais , Peso Corporal , Dieta Hiperlipídica , Citometria de Fluxo , Imunofluorescência , Perfilação da Expressão Gênica , Teste de Tolerância a Glucose , Proteínas de Homeodomínio/genética , Immunoblotting , Imuno-Histoquímica , Inflamação/imunologia , Insulina/metabolismo , Interferon gama/imunologia , Interleucina-1beta/imunologia , Macrófagos/citologia , Masculino , Camundongos , Camundongos Knockout , Camundongos Obesos , Linfócitos T , Fator de Necrose Tumoral alfa/imunologia
5.
Obesity (Silver Spring) ; 24(3): 597-605, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26916240

RESUMO

OBJECTIVE: The relationship between adipose tissue fibrosis, adipocyte hypertrophy, and preadipocyte hyperplasia in the context of obesity and the correlation of these tissue-based phenomena with systemic metabolic disease are poorly defined. The goal of this study was to clarify the relationship between adipose tissue fibrosis, adipocyte hypertrophy, and preadipocyte hyperplasia in human obesity and determine the correlation of these adipose-tissue based phenomena with diabetes. METHODS: Visceral and subcutaneous adipose tissues from humans with obesity collected during bariatric surgery were studied with QRTPCR, immunohistochemistry, and flow cytometry for expression of collagens and fibrosis-related proteins, adipocyte size, and preadipocyte frequency. Results were correlated with clinical characteristics including diabetes status. RESULTS: Fibrosis was decreased, hypertrophy was increased, and preadipocyte frequency and fibrotic gene expression were decreased in adipose tissues from diabetic subjects compared to non-diabetic subjects. These differences were greater in visceral compared to subcutaneous adipose tissue. CONCLUSIONS: These data are consistent with the hypothesis that adipose tissue fibrosis in the context of human obesity limits adipocyte hypertrophy and is associated with a reciprocal increase in adipocyte hyperplasia, with beneficial effects on systemic metabolism. These findings suggest adipose tissue fibrosis as a potential target for manipulation of adipocyte metabolism.


Assuntos
Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Hiperplasia/metabolismo , Obesidade/metabolismo , Cirurgia Bariátrica , Feminino , Fibrose , Humanos , Hipertrofia/metabolismo , Masculino , Pessoa de Meia-Idade , Gordura Subcutânea/metabolismo
6.
Adipocyte ; 4(4): 264-72, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26451282

RESUMO

The extracellular matrix (ECM) plays important roles in maintaining adequate adipose tissue function and in metabolic regulation. Here we have examined the organization of a relatively unexplored adipose tissue ECM component, elastin and its response to diet induced obesity in mice. Additionally, we have explored the regulation and requirement of macrophage metalloelastase, MMP-12, in adipose tissue ECM remodeling in obesity. In visceral fat depots, elastin fibers form a mesh-like net that becomes denser with diet-induced obesity. In contrast, the elastin fibers in subcutaneous adipose depots are more linear in organization, and are tightly associated with adipose tissue macrophages (ATMs). We found that Mmp12 is produced predominantly by ATMs and can be induced with both short- and long-term high fat diet challenge and rapid remodeling induced by lipolysis. This contrasts with Mmp14 and Timp1 which are further induced only after chronic obesity in non-ATM populations. We examined obese transgenic Mmp12 (-/-) mice and found an increase in gene expression of ECM genes with diet-induced obesity, but showed few significant differences in metabolic parameters, elastin matrix density, or in adipose tissue inflammation. Together, these studies reveal the architecture and diet-induced regulation of the elastin matrix and suggest that MMP-12 is not required for elastin matrix remodeling or for the metabolic dysfunction that occurs with obesity.

7.
J Biol Chem ; 290(21): 13250-62, 2015 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-25869128

RESUMO

Women of reproductive age are protected from metabolic disease relative to postmenopausal women and men. Most preclinical rodent studies are skewed toward the use of male mice to study obesity-induced metabolic dysfunction because of a similar protection observed in female mice. How sex differences in obesity-induced inflammatory responses contribute to these observations is unknown. We have compared and contrasted the effects of high fat diet-induced obesity on glucose metabolism and leukocyte activation in multiple depots in male and female C57Bl/6 mice. With both short term and long term high fat diet, male mice demonstrated increased weight gain and CD11c(+) adipose tissue macrophage content compared with female mice despite similar degrees of adipocyte hypertrophy. Competitive bone marrow transplant studies demonstrated that obesity induced a preferential contribution of male hematopoietic cells to circulating leukocytes and adipose tissue macrophages compared with female cells independent of the sex of the recipient. Sex differences in macrophage and hematopoietic cell in vitro activation in response to obesogenic cues were observed to explain these results. In summary, this report demonstrates that male and female leukocytes and hematopoietic stem cells have cell-autonomous differences in their response to obesity that contribute to an amplified response in males compared with females.


Assuntos
Glicemia/metabolismo , Dieta Hiperlipídica/efeitos adversos , Células-Tronco Hematopoéticas/citologia , Inflamação/imunologia , Obesidade/etiologia , Tecido Adiposo/citologia , Tecido Adiposo/imunologia , Tecido Adiposo/metabolismo , Animais , Biomarcadores/análise , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Feminino , Citometria de Fluxo , Teste de Tolerância a Glucose , Células-Tronco Hematopoéticas/metabolismo , Imuno-Histoquímica , Inflamação/complicações , Inflamação/patologia , Lipídeos/análise , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mielopoese/fisiologia , Obesidade/metabolismo , Obesidade/patologia , Fatores Sexuais , Aumento de Peso
8.
Cell Rep ; 9(2): 605-17, 2014 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-25310975

RESUMO

An adaptive immune response triggered by obesity is characterized by the activation of adipose tissue CD4(+) T cells by unclear mechanisms. We have examined whether interactions between adipose tissue macrophages (ATMs) and CD4(+) T cells contribute to adipose tissue metainflammation. Intravital microscopy identifies dynamic antigen-dependent interactions between ATMs and T cells in visceral fat. Mice deficient in major histocompatibility complex class II (MHC II) showed protection from diet-induced obesity. Deletion of MHC II expression in macrophages led to an adipose tissue-specific decrease in the effector/memory CD4(+) T cells, attenuation of CD11c(+) ATM accumulation, and improvement in glucose intolerance by increasing adipose tissue insulin sensitivity. Ablation experiments demonstrated that the maintenance of proliferating conventional T cells is dependent on signals from CD11c(+) ATMs in obese mice. These studies demonstrate the importance of MHCII-restricted signals from ATMs that regulate adipose tissue T cell maturation and metainflammation.


Assuntos
Tecido Adiposo/imunologia , Linfócitos T CD4-Positivos/imunologia , Genes MHC da Classe II , Ativação Linfocitária , Macrófagos/imunologia , Obesidade/imunologia , Tecido Adiposo/citologia , Animais , Antígeno CD11c/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Células Cultivadas , Deleção de Genes , Antígenos HLA-DR/genética , Antígenos HLA-DR/metabolismo , Humanos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL
9.
Mol Metab ; 3(6): 664-75, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25161889

RESUMO

Obesity is associated with an activated macrophage phenotype in multiple tissues that contributes to tissue inflammation and metabolic disease. To evaluate the mechanisms by which obesity potentiates myeloid activation, we evaluated the hypothesis that obesity activates myeloid cell production from bone marrow progenitors to potentiate inflammatory responses in metabolic tissues. High fat diet-induced obesity generated both quantitative increases in myeloid progenitors as well as a potentiation of inflammation in macrophages derived from these progenitors. In vivo, hematopoietic stem cells from obese mice demonstrated the sustained capacity to preferentially generate inflammatory CD11c(+) adipose tissue macrophages after serial bone marrow transplantation. We identified that hematopoietic MyD88 was important for the accumulation of CD11c(+) adipose tissue macrophage accumulation by regulating the generation of myeloid progenitors from HSCs. These findings demonstrate that obesity and metabolic signals potentiate leukocyte production and that dietary priming of hematopoietic progenitors contributes to adipose tissue inflammation.

10.
Annu Rev Nutr ; 34: 57-76, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24850386

RESUMO

The ability of adipose tissue to adapt to a changing nutrient environment is critical to the maintenance of metabolic control. Nutrient excess and deficiency alter the shape of adipose tissue drastically and trigger many events that are collectively known as adipose tissue remodeling. Remodeling of adipose tissue involves more than adipocytes and is controlled by an extensive network of stromal cells and extracellular matrix proteins. Prominent players in this process are adipose tissue macrophages, which are a specialized leukocyte present in lean and obese states that contributes to adipose tissue inflammation. The interest in adipose tissue remodeling has been accelerated by the current epidemic of obesity and the chronic generation of signals that lead to expansion of adipose tissue. It is clear that evidence of dysfunctional remodeling events is a hallmark of obesity associated with metabolic disease. This review summarizes and highlights the recent work in this area and provides a framework in which to consider how adipose tissue macrophages contribute to the remodeling events in lean and obese states. Advancing our understanding of the involvement of macrophages in adipose tissue remodeling will promote one aspect of the new field of "immunometabolism," which connects control systems developed for regulation of immunity with those that control metabolism. It will also provide insight into how physiologic and pathophysiologic remodeling differs in adipose tissue and identify potential nodes for intervention to break the link between obesity and disease.


Assuntos
Adaptação Fisiológica , Tecido Adiposo/imunologia , Adiposidade , Macrófagos/imunologia , Modelos Biológicos , Neovascularização Fisiológica , Tecido Adiposo/irrigação sanguínea , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Animais , Matriz Extracelular/imunologia , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Humanos , Macrófagos/metabolismo , Neovascularização Patológica , Obesidade/imunologia , Obesidade/metabolismo , Obesidade/patologia
11.
Methods Enzymol ; 537: 17-30, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24480339

RESUMO

Adipose tissue is composed of a variety of cell types that include mature adipocytes, endothelial cells, fibroblasts, adipocyte progenitors, and a range of inflammatory leukocytes. These cells work in concert to promote nutrient storage in adipose tissue depots and vary widely based on location. In addition, overnutrition and obesity impart significant changes in the architecture of adipose tissue that are strongly associated with metabolic dysfunction. Recent studies have called attention to the importance of adipose tissue microenvironments in regulating adipocyte function and therefore require techniques that preserve cellular interactions and permit detailed analysis of three-dimensional structures in fat. This chapter summarizes our experience with the use of laser scanning confocal microscopy for imaging adipose tissue in rodents.


Assuntos
Adipócitos/ultraestrutura , Tecido Adiposo Branco/ultraestrutura , Microscopia Confocal/métodos , Obesidade/patologia , Diferenciação Celular/genética , Microambiente Celular , Humanos , Macrófagos/metabolismo , Macrófagos/patologia , Obesidade/genética , Linfócitos T/patologia , Linfócitos T/ultraestrutura
12.
Endocrinology ; 154(12): 4548-59, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24140711

RESUMO

Thrombospondin 1 (THBS1 or TSP-1) is a circulating glycoprotein highly expressed in hypertrophic visceral adipose tissues of humans and mice. High-fat diet (HFD) feeding induces the robust increase of circulating THBS1 in the early stages of HFD challenge. The loss of Thbs1 protects male mice from diet-induced weight gain and adipocyte hypertrophy. Hyperinsulinemic euglycemic clamp study has demonstrated that Thbs1-null mice are protected from HFD-induced insulin resistance. Tissue-specific glucose uptake study has revealed that the insulin-sensitive phenotype of Thbs1-null mice is mostly mediated by skeletal muscles. Further assessments of the muscle phenotype using RNA sequencing, quantitative PCR, and histological studies have demonstrated that Thbs1-null skeletal muscles are protected from the HFD-dependent induction of Col3a1 and Col6a1, coupled with a new collagen deposition. At the same time, the Thbs1-null mice display a better circadian rhythm and higher amplitude of energy expenditure with a browning phenotype in sc adipose tissues. These results suggest that THBS1, which circulates in response to a HFD, may induce insulin resistance and fibrotic tissue damage in skeletal muscles as well as the de-browning of sc adipose tissues in the early stages of a HFD challenge. Our study may shed new light on the pathogenic role played by a circulating extracellular matrix protein in the cross talk between adipose tissues and skeletal muscles during obesity progression.


Assuntos
Gorduras na Dieta/efeitos adversos , Fibrose/induzido quimicamente , Resistência à Insulina/fisiologia , Doenças Musculares/etiologia , Trombospondina 1/metabolismo , Células 3T3-L1 , Tecido Adiposo Branco/metabolismo , Animais , Gorduras na Dieta/administração & dosagem , Relação Dose-Resposta a Droga , Epididimo , Regulação da Expressão Gênica/efeitos dos fármacos , Técnica Clamp de Glucose , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Obesidade , Trombospondina 1/genética , Transcriptoma
13.
Diabetes ; 62(8): 2762-72, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23493569

RESUMO

The proinflammatory activation of leukocytes in adipose tissue contributes to metabolic disease. How crosstalk between immune cells initiates and sustains adipose tissue inflammation remains an unresolved question. We have examined the hypothesis that adipose tissue macrophages (ATMs) interact with and regulate the function of T cells. Dietary obesity was shown to activate the proliferation of effector memory CD4(+) T cells in adipose tissue. Our studies further demonstrate that ATMs are functional antigen-presenting cells that promote the proliferation of interferon-γ-producing CD4(+) T cells in adipose tissue. ATMs from lean and obese visceral fat process and present major histocompatibility complex (MHC) class II-restricted antigens. ATMs were sufficient to promote proliferation and interferon-γ production from antigen-specific CD4(+) T cells in vitro and in vivo. Diet-induced obesity increased the expression of MHC II and T-cell costimulatory molecules on ATMs in visceral fat, which correlated with an induction of T-cell proliferation in that depot. Collectively, these data indicate that ATMs provide a functional link between the innate and adaptive immune systems within visceral fat in mice.


Assuntos
Tecido Adiposo/imunologia , Células Apresentadoras de Antígenos/imunologia , Linfócitos T CD4-Positivos/imunologia , Macrófagos/imunologia , Obesidade/imunologia , Tecido Adiposo/metabolismo , Animais , Células Apresentadoras de Antígenos/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Dieta , Teste de Tolerância a Glucose , Inflamação/imunologia , Insulina/sangue , Resistência à Insulina/imunologia , Ativação Linfocitária/imunologia , Macrófagos/metabolismo , Camundongos , Obesidade/metabolismo , Fagocitose/imunologia
14.
Bone ; 50(2): 477-89, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21872687

RESUMO

Wnt10b is an established regulator of mesenchymal stem cell (MSC) fate that inhibits adipogenesis and stimulates osteoblastogenesis, thereby impacting bone mass in vivo. However, downstream mechanisms through which Wnt10b exerts these effects are poorly understood. Moreover, whether other endogenous Wnt ligands also modulate MSC fate remains to be fully addressed. In this study, we identify Wnt6 and Wnt10a as additional Wnt family members that, like Wnt10b, are downregulated during development of white adipocytes in vivo and in vitro, suggesting that Wnt6 and/or Wnt10a may also inhibit adipogenesis. To assess the relative activities of Wnt6, Wnt10a and Wnt10b to regulate mesenchymal cell fate, we used gain- and loss-of function approaches in bipotential ST2 cells and in 3T3-L1 preadipocytes. Enforced expression of Wnt10a stabilizes ß-catenin, suppresses adipogenesis and stimulates osteoblastogenesis to a similar extent as Wnt10b, whereas stable expression of Wnt6 has a weaker effect on these processes than Wnt10a or Wnt10b. In contrast, knockdown of endogenous Wnt6 is associated with greater preadipocyte differentiation and impaired osteoblastogenesis than knockdown of Wnt10a or Wnt10b, suggesting that, among these Wnt ligands, Wnt6 is the most potent endogenous regulator of MSC fate. Finally, we show that knockdown of ß-catenin completely prevents the inhibition of adipogenesis and stimulation of osteoblast differentiation by Wnt6, Wnt10a or Wnt10b. Potential mechanisms whereby Wnts regulate fate of MSCs downstream of ß-catenin are also investigated. In conclusion, this study identifies Wnt10a and Wnt6 as additional regulators of MSC fate and demonstrates that mechanisms downstream of ß-catenin are required for Wnt6, Wnt10a and Wnt10b to influence differentiation of mesenchymal precursors.


Assuntos
Adipogenia , Proteínas do Tecido Nervoso/metabolismo , Osteoblastos/metabolismo , Osteogênese , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Células 3T3-L1 , Animais , Linhagem da Célula , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Osteoblastos/citologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Transdução de Sinais
15.
J Immunol ; 187(12): 6208-16, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22075699

RESUMO

Age-related adiposity has been linked to chronic inflammatory diseases in late life. To date, the studies on adipose tissue leukocytes and aging have not taken into account the heterogeneity of adipose tissue macrophages (ATMs), nor have they examined how age impacts other leukocytes such as T cells in fat. Therefore, we have performed a detailed examination of ATM subtypes in young and old mice using state of the art techniques. Our results demonstrate qualitative changes in ATMs with aging that generate a decrease in resident type 2 (M2) ATMs. The profile of ATMs in old fat shifts toward a proinflammatory environment with increased numbers of CD206(-)CD11c(-) (double-negative) ATMs. The mechanism of this aging-induced shift in the phenotypic profile of ATMs was found to be related to a decrease in peroxisome proliferator-activated receptor-γ expression in ATMs and alterations in chemokine/chemokine receptor expression profiles. Furthermore, we have revealed a profound and unexpected expansion of adipose tissue T cells in visceral fat with aging that includes a significant induction of regulatory T cells in fat. Our findings demonstrate a unique inflammatory cell signature in the physiologic context of aging adipose tissue that differs from those induced in setting of diet-induced obesity.


Assuntos
Envelhecimento/imunologia , Envelhecimento/patologia , Senescência Celular/imunologia , Gordura Intra-Abdominal/imunologia , Gordura Intra-Abdominal/patologia , Leucócitos Mononucleares/imunologia , Animais , Separação Celular/métodos , Imunofenotipagem , Inflamação/imunologia , Inflamação/patologia , Gordura Intra-Abdominal/citologia , Contagem de Leucócitos , Leucócitos Mononucleares/classificação , Leucócitos Mononucleares/patologia , Contagem de Linfócitos , Macrófagos Peritoneais/classificação , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/citologia , Monócitos/imunologia , Baço/citologia , Baço/imunologia , Subpopulações de Linfócitos T/classificação , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/patologia
16.
RNA Biol ; 6(5): 522-5, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19713761

RESUMO

Polyadenylation is a nearly universal mRNA processing step in eukaryotic gene expression and it takes place in a macromolecular machinery termed the mRNA 3' processing complex. In a recent study, we reported the purification and comprehensive characterization of functional mammalian mRNA 3' processing complexes. Our study defined the protein composition of this machinery, revealed new potential links between mRNA 3' processing and other cellular processes, and characterized basic structural features of the "core" mRNA 3' processing complex. These results provided new insights into the mechanism of mRNA 3'-end formation.


Assuntos
Processamento de Terminações 3' de RNA , Precursores de RNA/genética , Regiões 3' não Traduzidas , Animais , Humanos , Mamíferos , Complexos Multiproteicos , Poliadenilação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...