Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Metab ; 36(5): 1030-1043.e7, 2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38670107

RESUMO

The mechanisms of hepatic stellate cell (HSC) activation and the development of liver fibrosis are not fully understood. Here, we show that deletion of a nuclear seven transmembrane protein, TM7SF3, accelerates HSC activation in liver organoids, primary human HSCs, and in vivo in metabolic-dysfunction-associated steatohepatitis (MASH) mice, leading to activation of the fibrogenic program and HSC proliferation. Thus, TM7SF3 knockdown promotes alternative splicing of the Hippo pathway transcription factor, TEAD1, by inhibiting the splicing factor heterogeneous nuclear ribonucleoprotein U (hnRNPU). This results in the exclusion of the inhibitory exon 5, generating a more active form of TEAD1 and triggering HSC activation. Furthermore, inhibiting TEAD1 alternative splicing with a specific antisense oligomer (ASO) deactivates HSCs in vitro and reduces MASH diet-induced liver fibrosis. In conclusion, by inhibiting TEAD1 alternative splicing, TM7SF3 plays a pivotal role in mitigating HSC activation and the progression of MASH-related fibrosis.


Assuntos
Proteínas de Ligação a DNA , Cirrose Hepática , Fatores de Transcrição de Domínio TEA , Fatores de Transcrição , Fatores de Transcrição de Domínio TEA/metabolismo , Animais , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Cirrose Hepática/genética , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Humanos , Camundongos , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , Processamento Alternativo , Camundongos Endogâmicos C57BL , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Células Estreladas do Fígado/metabolismo , Masculino , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Fígado Gorduroso/genética , Camundongos Knockout
2.
Nat Metab ; 6(5): 880-898, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38605183

RESUMO

The obesity epidemic continues to worsen worldwide, driving metabolic and chronic inflammatory diseases. Thiazolidinediones, such as rosiglitazone (Rosi), are PPARγ agonists that promote 'M2-like' adipose tissue macrophage (ATM) polarization and cause insulin sensitization. As ATM-derived small extracellular vesicles (ATM-sEVs) from lean mice are known to increase insulin sensitivity, we assessed the metabolic effects of ATM-sEVs from Rosi-treated obese male mice (Rosi-ATM-sEVs). Here we show that Rosi leads to improved glucose and insulin tolerance, transcriptional repolarization of ATMs and increased sEV secretion. Administration of Rosi-ATM-sEVs rescues obesity-induced glucose intolerance and insulin sensitivity in vivo without the known thiazolidinedione-induced adverse effects of weight gain or haemodilution. Rosi-ATM-sEVs directly increase insulin sensitivity in adipocytes, myotubes and primary mouse and human hepatocytes. Additionally, we demonstrate that the miRNAs within Rosi-ATM-sEVs, primarily miR-690, are responsible for these beneficial metabolic effects. Thus, using ATM-sEVs with specific miRNAs may provide a therapeutic path to induce insulin sensitization.


Assuntos
Tecido Adiposo , Vesículas Extracelulares , Resistência à Insulina , Macrófagos , Rosiglitazona , Animais , Rosiglitazona/farmacologia , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/efeitos dos fármacos , Camundongos , Macrófagos/metabolismo , Macrófagos/efeitos dos fármacos , Tecido Adiposo/metabolismo , Tecido Adiposo/efeitos dos fármacos , Masculino , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Obesidade/metabolismo , Insulina/metabolismo , Adipócitos/metabolismo , Adipócitos/efeitos dos fármacos , Camundongos Endogâmicos C57BL
3.
Cell Genom ; 3(7): 100339, 2023 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-37492105

RESUMO

Loss-of-function mutations in hepatocyte nuclear factor 1A (HNF1A) are known to cause rare forms of diabetes and alter hepatic physiology through unclear mechanisms. In the general population, 1:100 individuals carry a rare, protein-coding HNF1A variant, most of unknown functional consequence. To characterize the full allelic series, we performed deep mutational scanning of 11,970 protein-coding HNF1A variants in human hepatocytes and clinical correlation with 553,246 exome-sequenced individuals. Surprisingly, we found that ∼1:5 rare protein-coding HNF1A variants in the general population cause molecular gain of function (GOF), increasing the transcriptional activity of HNF1A by up to 50% and conferring protection from type 2 diabetes (odds ratio [OR] = 0.77, p = 0.007). Increased hepatic expression of HNF1A promoted a pro-atherogenic serum profile mediated in part by enhanced transcription of risk genes including ANGPTL3 and PCSK9. In summary, ∼1:300 individuals carry a GOF variant in HNF1A that protects carriers from diabetes but enhances hepatic secretion of atherogenic lipoproteins.

4.
iScience ; 25(11): 105270, 2022 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-36304109

RESUMO

The seven-transmembrane superfamily member 3 protein (TM7SF3) is a p53-regulated homeostatic factor that attenuates cellular stress and the unfolded protein response. Here we show that TM7SF3 localizes to nuclear speckles; eukaryotic nuclear bodies enriched in splicing factors. This unexpected location for a trans -membranal protein enables formation of stable complexes between TM7SF3 and pre-mRNA splicing factors including DHX15, LARP7, HNRNPU, RBM14, and HNRNPK. Indeed, TM7SF3 regulates alternative splicing of >330 genes, mainly at the 3'end of introns by directly modulating the activity of splicing factors such as HNRNPK. These effects are observed both in cell lines and primary human pancreatic islets. Accordingly, silencing of TM7SF3 results in differential expression of 1465 genes (about 7% of the human genome); with 844 and 621 genes being up- or down-regulated, respectively. Our findings implicate TM7SF3, as a resident protein of nuclear speckles and suggest a role for seven-transmembrane proteins as regulators of alternative splicing.

5.
Nat Cell Biol ; 24(6): 954-967, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35637408

RESUMO

Epidemiological studies demonstrate an association between breast cancer (BC) and systemic dysregulation of glucose metabolism. However, how BC influences glucose homeostasis remains unknown. We show that BC-derived extracellular vesicles (EVs) suppress pancreatic insulin secretion to impair glucose homeostasis. EV-encapsulated miR-122 targets PKM in ß-cells to suppress glycolysis and ATP-dependent insulin exocytosis. Mice receiving high-miR-122 EVs or bearing BC tumours exhibit suppressed insulin secretion, enhanced endogenous glucose production, impaired glucose tolerance and fasting hyperglycaemia. These effects contribute to tumour growth and are abolished by inhibiting EV secretion or miR-122, restoring PKM in ß-cells or supplementing insulin. Compared with non-cancer controls, patients with BC have higher levels of circulating EV-encapsulated miR-122 and fasting glucose concentrations but lower fasting insulin; miR-122 levels are positively associated with glucose and negatively associated with insulin. Therefore, EV-mediated impairment of whole-body glycaemic control may contribute to tumour progression and incidence of type 2 diabetes in some patients with BC.


Assuntos
Neoplasias da Mama , Diabetes Mellitus Tipo 2 , Vesículas Extracelulares , MicroRNAs , Animais , Neoplasias da Mama/patologia , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Vesículas Extracelulares/metabolismo , Feminino , Glucose/metabolismo , Homeostase , Humanos , Insulina/metabolismo , Secreção de Insulina , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo
6.
Diabetes ; 71(7): 1508-1524, 2022 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-35472707

RESUMO

In obesity, increased mitochondrial metabolism with the accumulation of oxidative stress leads to mitochondrial damage and ß-cell dysfunction. In particular, ß-cells express antioxidant enzymes at relatively low levels and are highly vulnerable to oxidative stress. Early in the development of obesity, ß-cells exhibit increased glucose-stimulated insulin secretion in order to compensate for insulin resistance. This increase in ß-cell function under the condition of enhanced metabolic stress suggests that ß-cells possess a defense mechanism against increased oxidative damage, which may become insufficient or decline at the onset of type 2 diabetes. Here, we show that metabolic stress induces ß-cell hypoxia inducible factor 2α (HIF-2α), which stimulates antioxidant gene expression (e.g., Sod2 and Cat) and protects against mitochondrial reactive oxygen species (ROS) and subsequent mitochondrial damage. Knockdown of HIF-2α in Min6 cells exaggerated chronic high glucose-induced mitochondrial damage and ß-cell dysfunction by increasing mitochondrial ROS levels. Moreover, inducible ß-cell HIF-2α knockout mice developed more severe ß-cell dysfunction and glucose intolerance on a high-fat diet, along with increased ROS levels and decreased islet mitochondrial mass. Our results provide a previously unknown mechanism through which ß-cells defend against increased metabolic stress to promote ß-cell compensation in obesity.


Assuntos
Antioxidantes , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Diabetes Mellitus Tipo 2 , Animais , Antioxidantes/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Glucose/farmacologia , Camundongos , Camundongos Knockout , Obesidade , Espécies Reativas de Oxigênio/metabolismo
7.
Nat Metab ; 3(9): 1163-1174, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34489604

RESUMO

In chronic obesity, hepatocytes become insulin resistant and exert important effects on systemic metabolism. Here we show that in early onset obesity (4 weeks high-fat diet), hepatocytes secrete exosomes that enhance insulin sensitivity both in vitro and in vivo. These beneficial effects were due to exosomal microRNA miR-3075, which is enriched in these hepatocyte exosomes. FA2H is a direct target of miR-3075 and small interfering RNA depletion of FA2H in adipocytes, myocytes and primary hepatocytes leads to increased insulin sensitivity. In chronic obesity (16-18 weeks of a high-fat diet), hepatocyte exosomes promote a state of insulin resistance. These chronic obese hepatocyte exosomes do not directly cause impaired insulin signalling in vitro but do promote proinflammatory activation of macrophages. Taken together, these studies show that in early onset obesity, hepatocytes produce exosomes that express high levels of the insulin-sensitizing miR-3075. In chronic obesity, this compensatory effect is lost and hepatocyte-derived exosomes from chronic obese mice promote insulin resistance.


Assuntos
Exossomos/metabolismo , Hepatócitos/metabolismo , Resistência à Insulina/genética , Obesidade/metabolismo , Adipócitos/metabolismo , Animais , Dieta Hiperlipídica , Macrófagos/metabolismo , Camundongos , Células Musculares/metabolismo , RNA Interferente Pequeno/genética
8.
Cell Metab ; 33(9): 1744-1762, 2021 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-34496230

RESUMO

Exosomes are nanoparticles secreted by all cell types and are a large component of the broader class of nanoparticles termed extracellular vesicles (EVs). Once secreted, exosomes gain access to the interstitial space and ultimately the circulation, where they exert local paracrine or distal systemic effects. Because of this, exosomes are important components of an intercellular and intraorgan communication system capable of carrying biologic signals from one cell type or tissue to another. The exosomal cargo consists of proteins, lipids, miRNAs, and other RNA species, and many of the biologic effects of exosomes have been attributed to miRNAs. Exosomal miRNAs have also been used as disease biomarkers. The field of exosome biology and metabolism is rapidly expanding, with new discoveries and reports appearing on a regular basis, and it is possible that potential therapeutic approaches for the use of exosomes or miRNAs in metabolic diseases will be initiated in the near future.


Assuntos
Exossomos , Vesículas Extracelulares , Doenças Metabólicas , MicroRNAs , Comunicação Celular , Exossomos/metabolismo , Vesículas Extracelulares/metabolismo , Humanos , Doenças Metabólicas/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo
9.
Sci Rep ; 10(1): 7375, 2020 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-32355198

RESUMO

Secreted animal lectins of the galectin family are key players in cancer growth and metastasis. Here we show that galectin-8 (gal-8) induces the expression and secretion of cytokines and chemokines such as SDF-1 and MCP-1 in a number of cell types. This involves gal-8 binding to a uPAR/LRP1/integrin complex that activates JNK and the NFkB pathway. Cytokine and chemokine secretion, induced by gal-8, promotes migration of cancer cells toward cells treated with this lectin. Indeed, immune-competent gal-8 knockout (KO) mice express systemic lower levels of cytokines and chemokines while the opposite is true for gal-8 transgenic animals. Accordingly, gal-8 KO mice experience reduced tumor size and smaller and fewer metastatic lesions when injected with cancer cells. These results suggest the existence of a 'vicious cycle' whereby gal-8 secreted by the tumor microenvironment, promotes secretion of chemoattractants at the metastatic niche that promote further recruitment of tumor cells to that site. This study further implicate gal-8 in control of cancer progression and metastasis through its effects on the production of immunoregulatory cytokines.


Assuntos
Movimento Celular , Quimiocina CXCL12/metabolismo , Galectinas/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Experimentais/metabolismo , Animais , Quimiocina CCL2/genética , Quimiocina CXCL12/genética , Galectinas/genética , Camundongos , Camundongos Knockout , Metástase Neoplásica , Proteínas de Neoplasias/genética , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia
10.
Cell Metab ; 31(1): 162-173.e5, 2020 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-31708444

RESUMO

Insulin resistance is a major factor in obesity-linked type 2 diabetes. PPARγ is a master regulator of adipogenesis, and small molecule agonists, termed thiazolidinediones, are potent therapeutic insulin sensitizers. Here, we studied the role of transcriptional co-activator with PDZ-binding motif (TAZ) as a transcriptional co-repressor of PPARγ. We found that adipocyte-specific TAZ knockout (TAZ AKO) mice demonstrate a constitutively active PPARγ state. Obese TAZ AKO mice show improved glucose tolerance and insulin sensitivity compared to littermate controls. PPARγ response genes are upregulated in adipose tissue from TAZ AKO mice and adipose tissue inflammation was also decreased. In vitro and in vivo mechanistic studies revealed that the TAZ-PPARγ interaction is partially dependent on ERK-mediated Ser112 PPARγ phosphorylation. As adipocyte PPARγ Ser112 phosphorylation is increased in obesity, repression of PPARγ activity by TAZ could contribute to insulin resistance. These results identify TAZ as a new factor in the development of obesity-induced insulin resistance.


Assuntos
Adipócitos/metabolismo , Glucose/metabolismo , Resistência à Insulina/genética , PPAR gama/metabolismo , Transativadores/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Adipócitos/enzimologia , Adipogenia/genética , Animais , Linhagem Celular , Dieta Hiperlipídica , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Teste de Tolerância a Glucose , Humanos , Imuno-Histoquímica , Inflamação/genética , Inflamação/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Camundongos Obesos , PPAR gama/genética , Fosforilação , Transativadores/genética
11.
Sci Rep ; 9(1): 14779, 2019 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-31611602

RESUMO

Insulin resistance is a key feature of obesity and type 2 diabetes. PU.1 is a master transcription factor predominantly expressed in macrophages but after HFD feeding PU.1 expression is also significantly increased in adipocytes. We generated adipocyte specific PU.1 knockout mice using adiponectin cre to investigate the role of PU.1 in adipocyte biology, insulin and glucose homeostasis. In HFD-fed obese mice systemic glucose tolerance and insulin sensitivity were improved in PU.1 AKO mice and clamp studies indicated improvements in both adipose and liver insulin sensitivity. At the level of adipose tissue, macrophage infiltration and inflammation was decreased and glucose uptake was increased in PU.1 AKO mice compared with controls. While PU.1 deletion in adipocytes did not affect the gene expression of PPARg itself, we observed increased expression of PPARg target genes in eWAT from HFD fed PU.1 AKO mice compared with controls. Furthermore, we observed decreased phosphorylation at serine 273 in PU.1 AKO mice compared with fl/fl controls, indicating that PPARg is more active when PU.1 expression is reduced in adipocytes. Therefore, in obesity the increased expression of PU.1 in adipocytes modifies the adipocyte PPARg cistrome resulting in impaired glucose tolerance and insulin sensitivity.


Assuntos
Adipócitos/metabolismo , Resistência à Insulina , Insulina/metabolismo , Obesidade/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transativadores/metabolismo , Células 3T3-L1 , Animais , Células Cultivadas , Dieta Hiperlipídica/efeitos adversos , Técnicas de Inativação de Genes , Teste de Tolerância a Glucose , Insulina/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Obesos , Obesidade/sangue , Obesidade/etiologia , Obesidade/genética , Proteínas Proto-Oncogênicas/genética , Transativadores/genética , Regulação para Cima
12.
Cell Metab ; 29(2): 457-474.e5, 2019 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-30595478

RESUMO

The nature of obesity-associated islet inflammation and its impact on ß cell abnormalities remains poorly defined. Here, we explore immune cell components of islet inflammation and define their roles in regulating ß cell function and proliferation. Islet inflammation in obese mice is dominated by macrophages. We identify two islet-resident macrophage populations, characterized by their anatomical distributions, distinct phenotypes, and functional properties. Obesity induces the local expansion of resident intra-islet macrophages, independent of recruitment from circulating monocytes. Functionally, intra-islet macrophages impair ß cell function in a cell-cell contact-dependent manner. Increased engulfment of ß cell insulin secretory granules by intra-islet macrophages in obese mice may contribute to restricting insulin secretion. In contrast, both intra- and peri-islet macrophage populations from obese mice promote ß cell proliferation in a PDGFR signaling-dependent manner. Together, these data define distinct roles and mechanisms for islet macrophages in the regulation of islet ß cells.


Assuntos
Inflamação/imunologia , Células Secretoras de Insulina/metabolismo , Macrófagos/imunologia , Obesidade/metabolismo , Receptores do Fator de Crescimento Derivado de Plaquetas/imunologia , Animais , Linhagem Celular , Proliferação de Células , Secreção de Insulina , Células Secretoras de Insulina/patologia , Macrófagos/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos
14.
Diabetes ; 66(7): 1879-1889, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28424159

RESUMO

Cellular stress and proinflammatory cytokines induce phosphorylation of insulin receptor substrate (IRS) proteins at Ser sites that inhibit insulin and IGF-I signaling. We therefore examined the effects of mutation of five "inhibitory" Ser phosphorylation sites on IRS2 function in transgenic mice that overexpress, selectively in pancreatic ß-cells, either wild-type (WT) or a mutated IRS2 protein (IRS25A). Islets size, number, and mRNA levels of catalase and superoxide dismutase were increased, whereas those of nitric oxide synthase were decreased, in 7- to 10-week-old IRS25A-ß mice compared with IRS2WT-ß mice. However, glucose homeostasis and insulin secretion in IRS25A-ß mice were impaired when compared with IRS2WT-ß mice or to nontransgenic mice. This was associated with reduced mRNA levels of Glut2 and islet ß-cell transcription factors such as Nkx6.1 and MafA Similarly, components mediating the unfolded protein response were decreased in islets of IRS25A-ß mice in accordance with their decreased insulin secretion. The beneficial effects of IRS25A on ß-cell proliferation and ß-cell transcription factors were evident only in 5- to 8-day-old mice. These findings suggest that elimination of inhibitory Ser phosphorylation sites of IRS2 exerts short-term beneficial effects in vivo; however, their sustained elimination leads to impaired ß-cell function.


Assuntos
Retroalimentação Fisiológica , Proteínas Substratos do Receptor de Insulina/genética , Insulina/metabolismo , RNA Mensageiro/metabolismo , Animais , Glicemia/metabolismo , Catalase/genética , Catalase/metabolismo , Proliferação de Células/genética , Transportador de Glucose Tipo 2/genética , Transportador de Glucose Tipo 2/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Células Secretoras de Insulina , Ilhotas Pancreáticas/patologia , Fatores de Transcrição Maf Maior/genética , Fatores de Transcrição Maf Maior/metabolismo , Camundongos , Camundongos Transgênicos , Mutação , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase/metabolismo , Tamanho do Órgão , Fosforilação , Transdução de Sinais , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo
15.
Cell Death Differ ; 24(1): 132-143, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27740623

RESUMO

Earlier reported small interfering RNA (siRNA) high-throughput screens, identified seven-transmembrane superfamily member 3 (TM7SF3) as a novel inhibitor of pancreatic ß-cell death. Here we show that TM7SF3 maintains protein homeostasis and promotes cell survival through attenuation of ER stress. Overexpression of TM7SF3 inhibits caspase 3/7 activation. In contrast, siRNA-mediated silencing of TM7SF3 accelerates ER stress and activation of the unfolded protein response (UPR). This involves inhibitory phosphorylation of eukaryotic translation initiation factor 2α activity and increased expression of activating transcription factor-3 (ATF3), ATF4 and C/EBP homologous protein, followed by induction of apoptosis. This process is observed both in human pancreatic islets and in a number of cell lines. Some of the effects of TM7SF3 silencing are evident both under basal conditions, in otherwise untreated cells, as well as under different stress conditions induced by thapsigargin, tunicamycin or a mixture of pro-inflammatory cytokines (tumor necrosis factor alpha, interleukin-1 beta and interferon gamma). Notably, TM7SF3 is a downstream target of p53: activation of p53 by Nutlin increases TM7SF3 expression in a time-dependent manner, although silencing of p53 abrogates this effect. Furthermore, p53 is found in physical association with the TM7SF3 promoter. Interestingly, silencing of TM7SF3 promotes p53 activity, suggesting the existence of a negative-feedback loop, whereby p53 promotes expression of TM7SF3 that acts to restrict p53 activity. Our findings implicate TM7SF3 as a novel p53-regulated pro-survival homeostatic factor that attenuates the development of cellular stress and the subsequent induction of the UPR.


Assuntos
Glicoproteínas de Membrana/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Fator 3 Ativador da Transcrição/metabolismo , Fator 4 Ativador da Transcrição/metabolismo , Animais , Apoptose/efeitos dos fármacos , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Caspase 3/metabolismo , Caspase 7/metabolismo , Linhagem Celular , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Células HEK293 , Células Hep G2 , Humanos , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/genética , Camundongos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Tapsigargina/toxicidade , Fator de Transcrição CHOP/metabolismo , Tunicamicina/toxicidade , Resposta a Proteínas não Dobradas/efeitos dos fármacos , eIF-2 Quinase/antagonistas & inibidores , eIF-2 Quinase/metabolismo
16.
Biochem Biophys Res Commun ; 465(4): 851-6, 2015 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-26319551

RESUMO

High-throughput siRNA screening was employed to identify novel genes that regulate cytokine-induced death of pancreatic ß-cells. One of the 'hits' was Nedd4 family interacting protein 1 (Ndfip1), an adaptor and activator of Nedd4-family ubiquitin ligases. Silencing of Ndfip1 inhibited cytokine-induced apoptosis of mouse and human pancreatic islets and promoted glucose-stimulated insulin secretion. These effects were associated with an increase in the cellular content of JunB, a potent inhibitor of ER stress and apoptosis. Silencing of Ndfip1 also increased the expression of ATF4, IRE-1α, and the spliced form of XBP that govern the unfolded protein response (UPR) and relieve cytokine-induced ER stress, while overexpression of Ndfip1 exerted opposite effects. These findings implicate Ndfip1 in the degradation of JunB; inhibition of the UPR and insulin secretion; and promotion of cytokine-induced death of pancreatic ß-cells.


Assuntos
Apoptose/fisiologia , Proteínas de Transporte/metabolismo , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Proteínas de Membrana/metabolismo , Animais , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/genética , Linhagem Celular , Células Cultivadas , Citocinas/metabolismo , Estresse do Retículo Endoplasmático , Ensaios de Triagem em Larga Escala , Humanos , Insulina/metabolismo , Secreção de Insulina , Peptídeos e Proteínas de Sinalização Intercelular , Masculino , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Proteólise , RNA Interferente Pequeno/genética , Fatores de Transcrição/metabolismo , Resposta a Proteínas não Dobradas
17.
J Biol Chem ; 288(8): 5682-93, 2013 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-23275337

RESUMO

Selective serotonin reuptake inhibitors (SSRIs) are antidepressants used for the treatment of mood and anxiety disorders. Here, we demonstrate that incubation (2 h) of murine islets or Min6 ß cell line with the SSRIs paroxetine, fluoxetine, or sertraline inhibited insulin-induced Tyr phosphorylation of insulin receptor substrate (IRS)-2 protein and the activation of its downstream targets Akt and the ribosomal protein S6 kinase-1 (S6K1). Inhibition was dose-dependent with half-maximal effects at ∼15-20 µM. It correlated with a rapid dephosphorylation and activation of the IRS kinase GSK3ß. Introduction of GSK3ß siRNAs eliminated the inhibitory effects of the SSRIs. Inhibition of IRS-2 action by 30 µM SSRI was associated with a marked inhibition of glucose-stimulated insulin secretion from murine and human pancreatic islets. Secretion induced by basic secretagogues (KCl and Arg) was not affected by these drugs. Prolonged treatment (16 h) of Min6 cells with sertraline resulted in the induction of inducible nitric oxide synthase; activation of endoplasmic reticulum stress, and the initiation of the unfolded protein response, manifested by enhanced transcription of ATF4 and C/EBP homologous protein. This triggered an apoptotic process, manifested by enhanced caspase 3/7 activity, which resulted in ß cell death. These findings implicate SSRIs as inhibitors of IRS protein function and insulin action through the activation of GSK3ß. They further suggest that SSRIs inhibit insulin secretion; induce the unfolded protein response; activate an apoptotic process, and trigger ß cell death. Given that SSRIs promote insulin resistance while inhibiting insulin secretion, these drugs might accelerate the transition from an insulin-resistant state to overt diabetes.


Assuntos
Células Secretoras de Insulina/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Animais , Apoptose , Morte Celular , Linhagem Celular , Diabetes Mellitus/induzido quimicamente , Diabetes Mellitus/metabolismo , Fluoxetina/farmacologia , Humanos , Insulina/metabolismo , Proteínas Substratos do Receptor de Insulina/metabolismo , Resistência à Insulina , Células Secretoras de Insulina/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Paroxetina/farmacologia , Sertralina/farmacologia , Transdução de Sinais , Resposta a Proteínas não Dobradas
18.
Diabetes ; 59(9): 2188-97, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20547979

RESUMO

OBJECTIVE: Cellular stress and proinflammatory cytokines induce phosphorylation of insulin receptor substrate (IRS) proteins at Ser sites that inhibit insulin and IGF-1 signaling. Here, we examined the role of Ser phosphorylation of IRS-2 in mediating the inhibitory effects of proinflammatory cytokines and cellular stress on beta-cell function. RESEARCH DESIGN AND METHODS: Five potential inhibitory Ser sites located proximally to the P-Tyr binding domain of IRS-2 were mutated to Ala. These IRS-2 mutants, denoted IRS-2(5A), and their wild-type controls (IRS-2(WT)) were introduced into adenoviral constructs that were infected into Min6 cells or into cultured murine islets. RESULTS: When expressed in cultured mouse islets, IRS-2(5A) was better than IRS-2(WT) in protecting beta-cells from apoptosis induced by a combination of IL-1beta, IFN-gamma, TNF-alpha, and Fas ligand. Cytokine-treated islets expressing IRS2(5A) secreted significantly more insulin in response to glucose than did islets expressing IRS-2(WT). This could be attributed to the higher transcription of Pdx1 in cytokine-treated islets that expressed IRS-2(5A). Accordingly, transplantation of 200 islets expressing IRS2(5A) into STZ-induced diabetic mice restored their ability to respond to a glucose load similar to naïve mice. In contrast, mice transplanted with islets expressing IRS2(WT) maintained sustained hyperglycemia 3 days after transplantation. CONCLUSIONS: Elimination of a physiological negative feedback control mechanism along the insulin-signaling pathway that involves Ser/Thr phosphorylation of IRS-2 affords protection against the adverse effects of proinflammatory cytokines and improves beta-cell function under stress. Genetic approaches that promote IRS2(5A) expression in pancreatic beta-cells, therefore, could be considered a rational treatment against beta-cell failure after islet transplantation.


Assuntos
Proteínas Substratos do Receptor de Insulina/fisiologia , Células Secretoras de Insulina/fisiologia , Insulina/fisiologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Glicemia/metabolismo , Células CHO , Caspases/metabolismo , Cricetinae , Cricetulus , Citocinas/farmacologia , Diabetes Mellitus Experimental/cirurgia , Glucose/farmacologia , Teste de Tolerância a Glucose , Proteínas de Homeodomínio/genética , Insulina/metabolismo , Proteínas Substratos do Receptor de Insulina/genética , Secreção de Insulina , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Transplante das Ilhotas Pancreáticas/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Serina/fisiologia , Transdução de Sinais , Transativadores/genética , Transfecção
19.
Mol Cell Neurosci ; 36(3): 305-12, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17728140

RESUMO

Certain selective serotonin reuptake inhibitors (SSRIs) induce the clinical and biochemical manifestations of a metabolic syndrome by as yet unknown mechanism. Here we demonstrate that incubation (1 h) of rat hepatoma Fao cells with the SSRIs paroxetine and sertraline, but not with the atypical antipsychotic drug olanzapine, inhibited the insulin-stimulated Tyr phosphorylation of the insulin receptor substrate-1 (IRS-1) with half-maximal effects at approximately 10 microM. This inhibition correlated with a rapid phosphorylation and activation of a number of Ser/Thr IRS-1 kinases including JNK, S6K1, ERK and p38 MAPK, but not PKB (Akt). JNK appears as a key player activated by SSRIs because specific JNK inhibitors partially eliminated the effects of these drugs. The SSRIs induced the phosphorylation of IRS-1 on S307 and S408, which inhibits IRS-1 function and insulin signaling. These results implicate selected SSRIs as inhibitors of insulin signaling and as potential inducers of cellular insulin resistance.


Assuntos
Resistência à Insulina/fisiologia , Proteínas Quinases JNK Ativadas por Mitógeno/efeitos dos fármacos , Fosfoproteínas/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/efeitos dos fármacos , Inibidores Seletivos de Recaptação de Serotonina/efeitos adversos , Transdução de Sinais/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Diabetes Mellitus/induzido quimicamente , Diabetes Mellitus/metabolismo , Diabetes Mellitus/fisiopatologia , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Insulina/metabolismo , Proteínas Substratos do Receptor de Insulina , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Síndrome Metabólica/induzido quimicamente , Síndrome Metabólica/metabolismo , Síndrome Metabólica/fisiopatologia , Paroxetina/efeitos adversos , Fosfoproteínas/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Ratos , Serina/metabolismo , Sertralina/efeitos adversos , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...