Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
PLoS Biol ; 19(5): e3000988, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33979328

RESUMO

Although visceral adipocytes located within the body's central core are maintained at approximately 37°C, adipocytes within bone marrow, subcutaneous, and dermal depots are found primarily within the peripheral shell and generally exist at cooler temperatures. Responses of brown and beige/brite adipocytes to cold stress are well studied; however, comparatively little is known about mechanisms by which white adipocytes adapt to temperatures below 37°C. Here, we report that adaptation of cultured adipocytes to 31°C, the temperature at which distal marrow adipose tissues and subcutaneous adipose tissues often reside, increases anabolic and catabolic lipid metabolism, and elevates oxygen consumption. Cool adipocytes rely less on glucose and more on pyruvate, glutamine, and, especially, fatty acids as energy sources. Exposure of cultured adipocytes and gluteal white adipose tissue (WAT) to cool temperatures activates a shared program of gene expression. Cool temperatures induce stearoyl-CoA desaturase-1 (SCD1) expression and monounsaturated lipid levels in cultured adipocytes and distal bone marrow adipose tissues (BMATs), and SCD1 activity is required for acquisition of maximal oxygen consumption at 31°C.


Assuntos
Adipócitos Brancos/metabolismo , Regulação da Temperatura Corporal/fisiologia , Adaptação Fisiológica , Adipócitos/metabolismo , Adipócitos/fisiologia , Adipócitos Marrons/metabolismo , Adipócitos Brancos/fisiologia , Tecido Adiposo/metabolismo , Tecido Adiposo Branco/metabolismo , Animais , Temperatura Baixa , Ácidos Graxos/metabolismo , Feminino , Metabolismo dos Lipídeos/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Consumo de Oxigênio , Ratos , Ratos Sprague-Dawley , Estearoil-CoA Dessaturase/metabolismo
2.
J Biol Chem ; 294(18): 7296-7307, 2019 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-30877201

RESUMO

The E3 ubiquitin ligase parkin is a critical regulator of mitophagy and has been identified as a susceptibility gene for type 2 diabetes (T2D). However, its role in metabolically active tissues that precipitate T2D development is unknown. Specifically, pancreatic ß cells and adipocytes both rely heavily on mitochondrial function in the regulation of optimal glycemic control to prevent T2D, but parkin's role in preserving quality control of ß cell or adipocyte mitochondria is unclear. Although parkin has been reported previously to control mitophagy, here we show that, surprisingly, parkin is dispensable for glucose homeostasis in both ß cells and adipocytes during diet-induced insulin resistance in mice. We observed that insulin secretion, ß cell formation, and islet architecture were preserved in parkin-deficient ß cells and islets, suggesting that parkin is not necessary for control of ß cell function and islet compensation for diet-induced obesity. Although transient parkin deficiency mildly impaired mitochondrial turnover in ß cell lines, parkin deletion in primary ß cells yielded no deficits in mitochondrial clearance. In adipocyte-specific deletion models, lipid uptake and ß-oxidation were increased in cultured cells, whereas adipose tissue morphology, glucose homeostasis, and beige-to-white adipocyte transition were unaffected in vivo In key metabolic tissues where mitochondrial dysfunction has been implicated in T2D development, our experiments unexpectedly revealed that parkin is not an essential regulator of glucose tolerance, whole-body energy metabolism, or mitochondrial quality control. These findings highlight that parkin-independent processes maintain ß cell and adipocyte mitochondrial quality control in diet-induced obesity.


Assuntos
Adipócitos/metabolismo , Homeostase , Células Secretoras de Insulina/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Adipócitos/citologia , Adipócitos/enzimologia , Adiposidade , Animais , Peso Corporal , Diferenciação Celular , Diabetes Mellitus Tipo 2/metabolismo , Metabolismo Energético , Feminino , Teste de Tolerância a Glucose , Resistência à Insulina , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/enzimologia , Masculino , Camundongos , Mitocôndrias/metabolismo , Oxirredução
3.
Blood ; 114(15): 3244-54, 2009 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-19567879

RESUMO

Alternatively activated (M2) macrophages play critical roles in diverse chronic diseases, including parasite infections, cancer, and allergic responses. However, little is known about the acquisition and maintenance of their phenotype. We report that M2-macrophage marker genes are epigenetically regulated by reciprocal changes in histone H3 lysine-4 (H3K4) and histone H3 lysine-27 (H3K27) methylation; and the latter methylation marks are removed by the H3K27 demethylase Jumonji domain containing 3 (Jmjd3). We found that continuous interleukin-4 (IL-4) treatment leads to decreased H3K27 methylation, at the promoter of M2 marker genes, and a concomitant increase in Jmjd3 expression. Furthermore, we demonstrate that IL-4-dependent Jmjd3 expression is mediated by STAT6, a major transcription factor of IL-4-mediated signaling. After IL-4 stimulation, activated STAT6 is increased and binds to consensus sites at the Jmjd3 promoter. Increased Jmjd3 contributes to the decrease of H3K27 dimethylation and trimethylation (H3K27me2/3) marks as well as the transcriptional activation of specific M2 marker genes. The decrease in H3K27me2/3 and increase in Jmjd3 recruitment were confirmed by in vivo studies using a Schistosoma mansoni egg-challenged mouse model, a well-studied system known to support an M2 phenotype. Collectively, these data indicate that chromatin remodeling is mechanistically important in the acquisition of the M2-macrophage phenotype.


Assuntos
Epigênese Genética/imunologia , Ativação de Macrófagos/imunologia , Macrófagos/imunologia , Schistosoma mansoni/imunologia , Esquistossomose mansoni/imunologia , Animais , Montagem e Desmontagem da Cromatina/genética , Montagem e Desmontagem da Cromatina/imunologia , Modelos Animais de Doenças , Feminino , Marcadores Genéticos/genética , Marcadores Genéticos/imunologia , Histonas/genética , Histonas/imunologia , Humanos , Interleucina-4/genética , Interleucina-4/imunologia , Histona Desmetilases com o Domínio Jumonji , Ativação de Macrófagos/genética , Metilação , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Oxirredutases N-Desmetilantes/genética , Oxirredutases N-Desmetilantes/imunologia , Regiões Promotoras Genéticas/genética , Regiões Promotoras Genéticas/imunologia , Fator de Transcrição STAT6/genética , Fator de Transcrição STAT6/imunologia , Esquistossomose mansoni/genética , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Ativação Transcricional/genética , Ativação Transcricional/imunologia
4.
Diabetes ; 70(9): 1970-1984, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34088712

RESUMO

Mechanisms by which autosomal recessive mutations in Lmna cause familial partial lipodystrophy type 2 (FPLD2) are poorly understood. To investigate the function of lamin A/C in adipose tissue, we created mice with an adipocyte-specific loss of Lmna (Lmna ADKO). Although Lmna ADKO mice develop and maintain adipose tissues in early postnatal life, they show a striking and progressive loss of white and brown adipose tissues as they approach sexual maturity. Lmna ADKO mice exhibit surprisingly mild metabolic dysfunction on a chow diet, but on a high-fat diet they share many characteristics of FPLD2 including hyperglycemia, hepatic steatosis, hyperinsulinemia, and almost undetectable circulating adiponectin and leptin. Whereas Lmna ADKO mice have reduced regulated and constitutive bone marrow adipose tissue with a concomitant increase in cortical bone, FPLD2 patients have reduced bone mass and bone mineral density compared with controls. In cell culture models of Lmna deficiency, mesenchymal precursors undergo adipogenesis without impairment, whereas fully differentiated adipocytes have increased lipolytic responses to adrenergic stimuli. Lmna ADKO mice faithfully reproduce many characteristics of FPLD2 and thus provide a unique animal model to investigate mechanisms underlying Lmna-dependent loss of adipose tissues.


Assuntos
Adipócitos/metabolismo , Adipogenia/fisiologia , Lamina Tipo A/genética , Lipodistrofia Parcial Familiar/genética , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/metabolismo , Animais , Densidade Óssea/fisiologia , Modelos Animais de Doenças , Lamina Tipo A/metabolismo , Lipodistrofia Parcial Familiar/metabolismo , Camundongos , Camundongos Knockout
5.
Mol Metab ; 39: 100992, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32325263

RESUMO

OBJECTIVE: Obesity is a key risk factor for many secondary chronic illnesses, including type 2 diabetes and cardiovascular disease. Canonical Wnt/ß-catenin signaling is established as an important endogenous inhibitor of adipogenesis. This pathway is operative in mature adipocytes; however, its roles in this context remain unclear due to complexities of Wnt signaling and differences in experimental models. In this study, we used novel cultured cell and mouse models to investigate functional roles of Wnts secreted from adipocytes. METHODS: We generated adipocyte-specific Wntless (Wls) knockout mice and cultured cell models to investigate molecular and metabolic consequences of disrupting Wnt secretion from mature adipocytes. To characterize Wls-deficient cultured adipocytes, we evaluated the expression of Wnt target and lipogenic genes and the downstream functional effects on carbohydrate and lipid metabolism. We also investigated the impact of adipocyte-specific Wls deletion on adipose tissues and global glucose metabolism in mice fed normal chow or high-fat diets. RESULTS: Many aspects of the Wnt signaling apparatus are expressed and operative in mature adipocytes, including the Wnt chaperone Wntless. Deletion of Wntless in cultured adipocytes results in the inhibition of de novo lipogenesis and lipid monounsaturation, likely through repression of Srebf1 (SREBP1c) and Mlxipl (ChREBP) and impaired cleavage of immature SREBP1c into its active form. Adipocyte-specific Wls knockout mice (Wls-/-) have lipogenic gene expression in adipose tissues and isolated adipocytes similar to that of controls when fed a normal chow diet. However, closer investigation reveals that a subset of Wnts and downstream signaling targets are upregulated within stromal-vascular cells of Wls-/- mice, suggesting that adipose tissues defend loss of Wnt secretion from adipocytes. Interestingly, this compensation is lost with long-term high-fat diet challenges. Thus, after six months of a high-fat diet, Wls-/- mice are characterized by decreased adipocyte lipogenic gene expression, reduced visceral adiposity, and improved glucose homeostasis. CONCLUSIONS: Taken together, these studies demonstrate that adipocyte-derived Wnts regulate de novo lipogenesis and lipid desaturation and coordinate the expression of lipogenic genes in adipose tissues. In addition, we report that Wnt signaling within adipose tissues is defended, such that a loss of Wnt secretion from adipocytes is sensed and compensated for by neighboring stromal-vascular cells. With chronic overnutrition, this compensatory mechanism is lost, revealing that Wls-/- mice are resistant to diet-induced obesity, adipocyte hypertrophy, and metabolic dysfunction.


Assuntos
Adipócitos/metabolismo , Regulação da Expressão Gênica , Lipogênese/genética , Doenças Metabólicas/etiologia , Doenças Metabólicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Biomarcadores , Células Cultivadas , Dieta/efeitos adversos , Modelos Animais de Doenças , Suscetibilidade a Doenças , Glucose/metabolismo , Imuno-Histoquímica , Insulina/metabolismo , Metabolismo dos Lipídeos/genética , Doenças Metabólicas/diagnóstico , Camundongos , Camundongos Knockout , Receptores Acoplados a Proteínas G/genética , Via de Sinalização Wnt
6.
Mol Metab ; 42: 101078, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32919095

RESUMO

OBJECTIVE: Canonical Wnt/ß-catenin signaling is a well-studied endogenous regulator of mesenchymal cell fate determination, promoting osteoblastogenesis and inhibiting adipogenesis. However, emerging genetic evidence in humans links a number of Wnt pathway members to body fat distribution, obesity, and metabolic dysfunction, suggesting that this pathway also functions in adipocytes. Recent studies in mice have uncovered compelling evidence that the Wnt signaling pathway plays important roles in adipocyte metabolism, particularly under obesogenic conditions. However, complexities in Wnt signaling and differences in experimental models and approaches have thus far limited our understanding of its specific roles in this context. METHODS: To investigate roles of the canonical Wnt pathway in the regulation of adipocyte metabolism, we generated adipocyte-specific ß-catenin (ß-cat) knockout mouse and cultured cell models. We used RNA sequencing, ChIP sequencing, and molecular approaches to assess expression of Wnt targets and lipogenic genes. We then used functional assays to evaluate effects of ß-catenin deficiency on adipocyte metabolism, including lipid and carbohydrate handling. In mice maintained on normal chow and high-fat diets, we assessed the cellular and functional consequences of adipocyte-specific ß-catenin deletion on adipose tissues and systemic metabolism. RESULTS: We report that in adipocytes, the canonical Wnt/ß-catenin pathway regulates de novo lipogenesis (DNL) and fatty acid monounsaturation. Further, ß-catenin mediates effects of Wnt signaling on lipid metabolism in part by transcriptional regulation of Mlxipl and Srebf1. Intriguingly, adipocyte-specific loss of ß-catenin is sensed and defended by CD45-/CD31- stromal cells to maintain tissue-wide Wnt signaling homeostasis in chow-fed mice. With long-term high-fat diet, this compensatory mechanism is overridden, revealing that ß-catenin deletion promotes resistance to diet-induced obesity and adipocyte hypertrophy and subsequent protection from metabolic dysfunction. CONCLUSIONS: Taken together, our studies demonstrate that Wnt signaling in adipocytes is required for lipogenic gene expression, de novo lipogenesis, and lipid desaturation. In addition, adipose tissues rigorously defend Wnt signaling homeostasis under standard nutritional conditions, such that stromal-vascular cells sense and compensate for adipocyte-specific loss. These findings underscore the critical importance of this pathway in adipocyte lipid metabolism and adipose tissue function.


Assuntos
Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Via de Sinalização Wnt/fisiologia , Adipócitos/fisiologia , Adipogenia/fisiologia , Tecido Adiposo/fisiologia , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Diferenciação Celular , Células Cultivadas , Expressão Gênica/genética , Regulação da Expressão Gênica/genética , Metabolismo dos Lipídeos , Lipogênese/fisiologia , Camundongos , Camundongos Knockout , Obesidade , Proteína de Ligação a Elemento Regulador de Esterol 1 , Células Estromais/metabolismo , Via de Sinalização Wnt/efeitos dos fármacos , Via de Sinalização Wnt/genética , Proteína Wnt1/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
7.
Cell Metab ; 28(2): 187-189, 2018 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-30089239

RESUMO

Adipocytes are generally thought to be terminally differentiated cells; however, recent evidence suggests a subset may have greater plasticity in certain contexts. In this issue of Cell Metabolism, Wang et al. (2018) demonstrate a novel capacity for mammary adipocytes to dedifferentiate into preadipocyte-like precursors during lactation and redifferentiate upon weaning.


Assuntos
Adipócitos Brancos , Lactação , Diferenciação Celular , Feminino , Humanos , Desmame
8.
Cell Rep ; 15(11): 2510-23, 2016 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-27264173

RESUMO

High levels of collagen deposition in human and mouse breast tumors are associated with poor outcome due to increased local invasion and distant metastases. Using a genetic approach, we show that, in mice, the action of the fibrillar collagen receptor discoidin domain receptor 2 (DDR2) in both tumor and tumor-stromal cells is critical for breast cancer metastasis yet does not affect primary tumor growth. In tumor cells, DDR2 in basal epithelial cells regulates the collective invasion of tumor organoids. In stromal cancer-associated fibroblasts (CAFs), DDR2 is critical for extracellular matrix production and the organization of collagen fibers. The action of DDR2 in CAFs also enhances tumor cell collective invasion through a pathway distinct from the tumor-cell-intrinsic function of DDR2. This work identifies DDR2 as a potential therapeutic target that controls breast cancer metastases through its action in both tumor cells and tumor-stromal cells at the primary tumor site.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Fibroblastos Associados a Câncer/metabolismo , Fibroblastos Associados a Câncer/patologia , Receptor com Domínio Discoidina 2/metabolismo , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/patologia , Alelos , Animais , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Progressão da Doença , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Matriz Extracelular/metabolismo , Feminino , Deleção de Genes , Humanos , Queratina-14/metabolismo , Neoplasias Pulmonares/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Invasividade Neoplásica , Organoides/patologia , Células Estromais/patologia , Microambiente Tumoral
9.
Nat Cell Biol ; 15(6): 677-87, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23644467

RESUMO

Increased stromal collagen deposition in human breast tumours correlates with metastases. We show that activation of the collagen I receptor DDR2 (discoidin domain receptor 2) regulates SNAIL1 stability by stimulating ERK2 activity, in a Src-dependent manner. Activated ERK2 directly phosphorylates SNAIL1, leading to SNAIL1 nuclear accumulation, reduced ubiquitylation and increased protein half-life. DDR2-mediated stabilization of SNAIL1 promotes breast cancer cell invasion and migration in vitro, and metastasis in vivo. DDR2 expression was observed in most human invasive ductal breast carcinomas studied, and was associated with nuclear SNAIL1 and absence of E-cadherin expression. We propose that DDR2 maintains SNAIL1 level and activity in tumour cells that have undergone epithelial-mesenchymal transition (EMT), thereby facilitating continued tumour cell invasion through collagen-I-rich extracellular matrices by sustaining the EMT phenotype. As such, DDR2 could be an RTK (receptor tyrosine kinase) target for the treatment of breast cancer metastasis.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/patologia , Transição Epitelial-Mesenquimal , Receptores Proteína Tirosina Quinases/metabolismo , Receptores Mitogênicos/metabolismo , Fatores de Transcrição/metabolismo , Animais , Caderinas/biossíntese , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Receptores com Domínio Discoidina , Feminino , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Invasividade Neoplásica , Metástase Neoplásica , Fosforilação , Interferência de RNA , RNA Interferente Pequeno , Receptores Proteína Tirosina Quinases/genética , Receptores de Colágeno/metabolismo , Receptores Mitogênicos/genética , Transdução de Sinais , Fatores de Transcrição da Família Snail
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA