Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Biochim Biophys Acta Gen Subj ; 1861(8): 2039-2047, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28435021

RESUMO

BACKGROUND: Compensation of the pancreatic ß cell functional mass in response to metabolic stress is key to the pathogenesis of Type 2 Diabetes. The mTORC2 pathway governs fuel metabolism and ß cell functional mass. It is unknown whether mTORC2 is required for regulating metabolic stress-induced ß cell compensation. METHODS: We challenged four-week-old ß-cell-specific Rictor (a key component of mTORC2)-knockout mice with a high fat diet (HFD) for 4weeks and measured metabolic and pancreatic morphological parameters. We performed ex vivo experiments to analyse ß cell insulin secretion and electrophysiology characteristics. Adenoviral-mediated overexpression and lentiviral-ShRNA-mediated knocking down proteins were applied in Min6 cells and cultured primary mouse islets. RESULTS: ßRicKO mice showed a significant glucose intolerance and a reduced plasma insulin level and an unchanged level ß cell mass versus the control mice under HFD. A HFD or palmitate treatment enhanced both glucose-induced insulin secretion (GIIS) and the PMA (phorbol 12-myristate 13-acetate)-induced insulin secretion in the control islets but not in the ßRicKO islets. The KO ß cells showed similar glucose-induced Ca2+ influx but lower membrane capacitance increments versus the control cells. The enhanced mTORC2/PKC proteins levels in the control HFD group were ablated by Rictor deletion. Replenishing PKCα by overexpression of PKCα-T638D restored the defective GIIS in ßRicKO islets. CONCLUSIONS: The mTORC2/Rictor pathway modulates ß cell compensatory GIIS under nutrient overload mediated by its phosphorylation of PKCα. GENERAL SIGNIFICANCE: This study suggests that the mTORC2/PKC pathway in ß cells is involved in the pathogenesis of T2D.


Assuntos
Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Complexos Multiproteicos/fisiologia , Proteína Quinase C-alfa/fisiologia , Transdução de Sinais/fisiologia , Estresse Fisiológico/fisiologia , Serina-Treonina Quinases TOR/fisiologia , Animais , Diabetes Mellitus Tipo 2/etiologia , Dieta Hiperlipídica , Secreção de Insulina , Alvo Mecanístico do Complexo 2 de Rapamicina , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Acetato de Tetradecanoilforbol/farmacologia
2.
Biochim Biophys Acta ; 1840(1): 577-85, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24144566

RESUMO

BACKGROUND: It has been recognized that insulin hypersecretion can lead to the development of insulin resistance and type 2 diabetes mellitus. There is substantial evidence demonstrating that thiazolidinediones are able to delay and prevent the progression of pancreatic ß-cell dysfunction. However, the mechanism underlying the protective effect of thiazolidinediones on ß-cell function remains elusive. METHODS: We synchronously detected the effects of troglitazone on insulin secretion and AMP-activated protein kinase (AMPK) activity under various conditions in isolated rat islets and MIN6 cells. RESULTS: Long-term exposure to high glucose stimulated insulin hypersecretion and inhibited AMPK activity in rat islets. Troglitazone-suppressed insulin hypersecretion was closely related to the activation of AMPK. This action was most prominent at the moderate concentration of glucose. Glucose-stimulated insulin secretion was decreased by long-term troglitazone treatment, but significantly increased after the drug withdrawal. Compound C, an AMPK inhibitor, reversed troglitazone-suppressed insulin secretion in MIN6 cells and rat islets. Knockdown of AMPKα2 showed a similar result. In MIN6 cells, troglitazone blocked high glucose-closed ATP-sensitive K(+) (KATP) channel and decreased membrane potential, along with increased voltage-dependent potassium channel currents. Troglitazone suppressed intracellular Ca(2+) response to high glucose, which was abolished by treatment with compound C. CONCLUSION: Our results suggest that troglitazone provides ß-cell "a rest" through activating AMPK and inhibiting insulin hypersecretion, and thus restores its response to glucose. GENERAL SIGNIFICANCE: These data support that AMPK activation may be an important mechanism for thiazolidinediones preserving ß-cell function.


Assuntos
Cálcio/metabolismo , Cromanos/farmacologia , Hipoglicemiantes/farmacologia , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Proteínas Quinases/metabolismo , Tiazolidinedionas/farmacologia , Quinases Proteína-Quinases Ativadas por AMP , Animais , Células Cultivadas , Eletrofisiologia , Glucose/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Masculino , Proteínas Quinases/química , Proteínas Quinases/genética , RNA Interferente Pequeno/genética , Ratos , Ratos Sprague-Dawley , Troglitazona
3.
Blood ; 119(21): 4868-77, 2012 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-22474250

RESUMO

Alterations in gene expression after chemotherapy may potentially help to identify mediators that induce suppression or regeneration in bone marrow. This paper reports our observation that the expression of the chemokine monokine induced by IFN-γ (Mig) and its receptor CXCR3 was significantly activated in mice after treatment with the chemotherapeutic agent 5-fluorouracil (5-FU). The neutralization of antibodies against the activated Mig increased the survival rate and accelerated BM recovery after chemotherapy. In addition, elevation of Mig plasma levels after 5-FU treatment corresponded with increased mortality. The cell cycle-inhibiting effect of the prophylactic administration of Mig protected hematopoietic progenitor cells (HPCs) from 1-ß-d-arabinofuranosylcytosine in spleen colony assays and enhanced the irradiated recipients' survival. In CXCR3(-/-) mice, Mig did not propagate BM suppression, indicating that the suppressive effect of Mig is dependent on CXCR3. On the one hand, Mig stimulated p70 S6K and Erk1/2 pathways in mesenchymal stroma cells, inhibiting mesenchymal stroma cell-dependent HPC expansion. Moreover, Mig suppressed the STAT5 pathway in HPCs, inhibiting leukocyte differentiation. Our results strongly suggest that Mig contributes to the acute lethal toxicity arising from 5-FU administration. Neutralization of Mig may offer new strategies to alleviate BM toxicity with potentially dramatic implications for chemotherapy.


Assuntos
Antineoplásicos/efeitos adversos , Antineoplásicos/farmacologia , Células da Medula Óssea/efeitos dos fármacos , Quimiocina CXCL9/genética , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/mortalidade , Tolerância Imunológica/efeitos dos fármacos , Animais , Anticorpos/farmacologia , Células da Medula Óssea/patologia , Células Cultivadas , Quimiocina CXCL9/antagonistas & inibidores , Quimiocina CXCL9/imunologia , Quimiocina CXCL9/metabolismo , Quimiocinas/genética , Quimiocinas/metabolismo , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/genética , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/prevenção & controle , Fluoruracila/efeitos adversos , Fluoruracila/farmacologia , Expressão Gênica/efeitos dos fármacos , Perfilação da Expressão Gênica , Tolerância Imunológica/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise em Microsséries , Ratos , Ratos Wistar , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
4.
Theranostics ; 14(5): 2058-2074, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38505613

RESUMO

Rationale: NPC1 is a protein localized on the lysosome membrane regulating intracellular cholesterol transportation and maintaining normal lysosome function. GWAS studies have found that NPC1 variants in T2D was a pancreatic islet expression quantitative trait locus, suggesting a potential role of NPC1 in T2D islet pathophysiology. Methods: Two-week-old Npc1-/- mice and wild type littermates were employed to examine pancreatic ß cell morphology and functional changes induced by loss of Npc1. Single cell RNA sequencing was conducted on primary islets. Npc1-/- Min6 cell line was generated using CRISPR/Cas9 gene editing. Seahorse XF24 was used to analyze primary islet and Min6 cell mitochondria respiration. Ultra-high-resolution cell imaging with Lattice SIM2 and electron microscope imaging were used to observe mitochondria and lysosome in primary islet ß and Min6 cells. Mitophagy Dye and mt-Keima were used to measure ß cell mitophagy. Results: In Npc1-/- mice, we found that ß cell survival and pancreatic ß cell mass expansion as well as islet glucose induced insulin secretion in 2-week-old mice were reduced. Npc1 loss retarded postnatal ß cell differentiation and growth as well as impaired mitochondria oxidative phosphorylation (OXPHOS) function to increase mitochondrial superoxide production, which might be attributed to impaired autophagy flux particularly mitochondria autophagy (mitophagy) induced by dysfunctional lysosome in Npc1 null ß cells. Conclusion: Our study revealed that NPC1 played an important role in maintaining normal lysosome function and mitochondria turnover, which ensured establishment of sufficient mitochondria OXPHOS for islet ß cells differentiation and maturation.


Assuntos
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Ilhotas Pancreáticas , Animais , Camundongos , Diferenciação Celular , Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Mitocôndrias/metabolismo , Proteína C1 de Niemann-Pick/metabolismo
5.
Cell Metab ; 34(2): 256-268.e5, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-35108513

RESUMO

In diabetes, glucagon secretion from pancreatic α cells is dysregulated. The underlying mechanisms, and whether dysfunction occurs uniformly among cells, remain unclear. We examined α cells from human donors and mice using electrophysiological, transcriptomic, and computational approaches. Rising glucose suppresses α cell exocytosis by reducing P/Q-type Ca2+ channel activity, and this is disrupted in type 2 diabetes (T2D). Upon high-fat feeding of mice, α cells shift toward a "ß cell-like" electrophysiological profile in concert with indications of impaired identity. In human α cells we identified links between cell membrane properties and cell surface signaling receptors, mitochondrial respiratory chain complex assembly, and cell maturation. Cell-type classification using machine learning of electrophysiology data demonstrated a heterogenous loss of "electrophysiologic identity" in α cells from donors with type 2 diabetes. Indeed, a subset of α cells with impaired exocytosis is defined by an enrichment in progenitor and lineage markers and upregulation of an immature transcriptomic phenotype, suggesting important links between α cell maturation state and dysfunction.


Assuntos
Diabetes Mellitus Tipo 2 , Células Secretoras de Glucagon , Ilhotas Pancreáticas , Animais , Diabetes Mellitus Tipo 2/metabolismo , Exocitose/fisiologia , Glucagon/metabolismo , Células Secretoras de Glucagon/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Camundongos
6.
Biochim Biophys Acta Mol Basis Dis ; 1867(12): 166261, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34455055

RESUMO

Rapamycin insensitive companion of mechanistic target of Rapamycin (Rictor), the key component of mTOR complex 2 (mTORC2), controls both ß-cell proliferation and function. We sought to study whether long chain acyl-CoA synthetase 4 (Acsl4) worked downstream of Rictor/mTORC2 to maintain ß-cell functional mass. We found Acsl4 was positively regulated by Rictor at transcriptional and posttranslational levels in mouse ß-cell. Infecting adenovirus expressing Acsl4 in ß-cell-specific-Rictor-knockout (ßRicKO) islets and Min6 cells knocking down Rictor with lentivirus-expressing siRNA-oligos targeting Rictor(siRic), recovered the ß-cell dysplasia but not dysfunction. Cell bioenergetic experiment performed with Seahorse XF showed that Acsl4 could not rescue the dampened glucose oxidation in Rictor-lacking ß-cell, but further promoted lipid oxidation. Transposase-Accessible Chromatin (ATAC) and H3K27Ac chromatin immunoprecipitation (ChIP) sequencing studies reflected the epigenetic elevated molecular signature for ß-cell dedifferentiation and mitigated oxidative defense/response. These results were confirmed by the observations of elevated acetylation and ubiquitination of FoxO1, increased protein levels of Gpx1 and Hif1an, excessive reactive oxygen species (ROS) production and diminished MafA in Acsl4 overexpressed Rictor-lacking ß-cells. In these cells, antioxidant treatment significantly recovered MafA level and insulin content. Inducing lipid oxidation alone could not mimic the effect of Acsl4 in Rictor lacking ß-cell. Our study suggested that Acsl4 function in ß-cell was context dependent and might facilitate ß-cell dedifferentiation with attenuated Rictor/mTORC2 activity or insulin signaling via posttranslational inhibiting FoxO1 and epigenetically enhancing ROS induced MafA degradation.


Assuntos
Desdiferenciação Celular/genética , Coenzima A Ligases/genética , Proteína Forkhead Box O1/genética , Células Secretoras de Insulina/metabolismo , Proteína Companheira de mTOR Insensível à Rapamicina/genética , Animais , Proliferação de Células/genética , Epigenômica , Regulação da Expressão Gênica/genética , Glutationa Peroxidase/genética , Humanos , Insulina/genética , Insulina/metabolismo , Células Secretoras de Insulina/patologia , Metabolismo dos Lipídeos/genética , Alvo Mecanístico do Complexo 2 de Rapamicina/genética , Camundongos , Oxigenases de Função Mista/genética , Espécies Reativas de Oxigênio/metabolismo , Glutationa Peroxidase GPX1
7.
Nat Commun ; 11(1): 2538, 2020 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-32439909

RESUMO

Compromised ß-cell identity is emerging as an important contributor to ß-cell failure in diabetes; however, the precise mechanism independent of hyperglycemia is under investigation. We have previously reported that mTORC1/Raptor regulates functional maturation in ß-cells. In the present study, we find that diabetic ß-cell specific Raptor-deficient mice (ßRapKOGFP) show reduced ß-cell mass, loss of ß-cell identity and acquisition of α-cell features; which are not reversible upon glucose normalization. Deletion of Raptor directly impairs ß-cell identity, mitochondrial metabolic coupling and protein synthetic activity, leading to ß-cell failure. Moreover, loss of Raptor activates α-cell transcription factor MafB (via modulating C/EBPß isoform ratio) and several α-cell enriched genes i.e. Etv1 and Tspan12, thus initiates ß- to α-cell reprograming. The present findings highlight mTORC1 as a metabolic rheostat for stabilizing ß-cell identity and repressing α-cell program at normoglycemic level, which might present therapeutic opportunities for treatment of diabetes.


Assuntos
Diferenciação Celular , Plasticidade Celular , Diabetes Mellitus/patologia , Células Secretoras de Insulina/patologia , Proteína Regulatória Associada a mTOR/metabolismo , Animais , Glicemia/metabolismo , Diferenciação Celular/genética , Plasticidade Celular/genética , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Regulação da Expressão Gênica , Células Secretoras de Glucagon/metabolismo , Células Secretoras de Glucagon/patologia , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Fator de Transcrição MafB/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos , Camundongos Knockout , Proteína Regulatória Associada a mTOR/genética , Transdução de Sinais
8.
Diabetes ; 69(1): 48-59, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31649162

RESUMO

Statins are cholesterol-lowering agents that increase the incidence of diabetes and impair glucose tolerance via their detrimental effects on nonhepatic tissues, such as pancreatic islets, but the underlying mechanism has not been determined. In atorvastatin (ator)-treated high-fat diet-fed mice, we found reduced pancreatic ß-cell size and ß-cell mass, fewer mature insulin granules, and reduced insulin secretion and glucose tolerance. Transcriptome profiling of primary pancreatic islets showed that ator inhibited the expression of pancreatic transcription factor, mechanistic target of rapamycin (mTOR) signaling, and small G protein (sGP) genes. Supplementation of the mevalonate pathway intermediate geranylgeranyl pyrophosphate (GGPP), which is produced by 3-hydroxy-3-methylglutaryl-CoA (HMG-CoA) reductase, significantly restored the attenuated mTOR activity, v-maf musculoaponeurotic fibrosarcoma oncogene homolog A (MafA) expression, and ß-cell function after ator, lovastatin, rosuvastatin, and fluvastatin treatment; this effect was potentially mediated by sGP prenylation. Rab5a, the sGP in pancreatic islets most affected by ator treatment, was found to positively regulate mTOR signaling and ß-cell function. Rab5a knockdown mimicked the effect of ator treatment on ß-cells. Thus, ator impairs ß-cell function by regulating sGPs, for example, Rab5a, which subsequently attenuates islet mTOR signaling and reduces functional ß-cell mass. GGPP supplementation could constitute a new approach for preventing statin-induced hyperglycemia.


Assuntos
Atorvastatina/farmacologia , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Ácido Mevalônico/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Contagem de Células , Células Cultivadas , Feminino , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/fisiologia , Ilhotas Pancreáticas/crescimento & desenvolvimento , Masculino , Redes e Vias Metabólicas/genética , Camundongos , Camundongos Endogâmicos C57BL , Tamanho do Órgão/genética , Fosfatos de Poli-Isoprenil/farmacologia , Transdução de Sinais/genética
9.
Neurochem Res ; 34(12): 2233-42, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19557515

RESUMO

The effects of sodium metabisulfite (SMB), a general food preservative, on potassium currents in rat dorsal root ganglion (DRG) neurons were investigated using the whole-cell patch-clamp technique. SMB increased the amplitudes of both transient outward potassium currents and delayed rectifier potassium current in concentration- and voltage-dependent manner. The transient outward potassium currents (TOCs) include a fast inactivating (A-current or IA) current and a slow inactivating (D-current or ID) current. SMB majorly increased IA, and ID was little affected. SMB did not affect the activation process of transient outward currents (TOCs), but the inactivation curve of TOCs was shifted to more positive potentials. The inactivation time constants of TOCs were also increased by SMB. For delayed rectifier potassium current (IK), SMB shifted the activation curve to hyperpolarizing direction. SMB differently affected TOCs and IK, its effects major on A-type K+ channels, which play a role in adjusting pain sensitivity in response to peripheral redox conditions. SMB did not increase TOCs and IK when adding DTT in pipette solution. These results suggested that SMB might oxidize potassium channels, which relate to adjusting pain sensitivity in pain-sensing DRG neurons.


Assuntos
Gânglios Espinais/fisiologia , Neurônios/fisiologia , Canais de Potássio/efeitos dos fármacos , Sulfitos/farmacologia , Animais , Potenciais da Membrana/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Dor , Potássio/metabolismo , Canais de Potássio/metabolismo , Ratos
10.
Diabetes ; 68(10): 1950-1964, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31345937

RESUMO

Immature pancreatic ß-cells are highly proliferative, and the expansion of ß-cells during the early neonatal period largely determines functional ß-cell mass; however, the mechanisms are poorly characterized. We generated Ngn3RapKO mice (ablation of Raptor, an essential component of mechanistic target of rapamycin [mTORC1] in Ngn3+ endocrine progenitor cells) and found that mTORC1 was dispensable for endocrine cell lineage formation but specifically regulated both proliferation and identity maintenance of neonatal ß-cells. Ablation of Raptor in neonatal ß-cells led to autonomous loss of cell identity, decelerated cell cycle progression, compromised proliferation, and caused neonatal diabetes as a result of inadequate establishment of functional ß-cell mass at postnatal day 14. Completely different from mature ß-cells, Raptor regulated G1/S and G2/M phase cell cycle transition, thus permitting a high proliferation rate in neonatal ß-cells. Moreover, Ezh2 was identified as a critical downstream target of mTORC1 in neonatal ß-cells, which was responsible for G2/M phase transition and proliferation. Our discovery of the dual effect of mTORC1 in immature ß-cells has revealed a potential target for replenishing functional ß-cell pools by promoting both expansion and functional maturation of newly formed immature ß-cells.


Assuntos
Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Diabetes Mellitus/metabolismo , Células Secretoras de Insulina/metabolismo , Proteína Regulatória Associada a mTOR/metabolismo , Animais , Animais Recém-Nascidos , Ciclo Celular/fisiologia , Diabetes Mellitus/genética , Células Secretoras de Insulina/citologia , Camundongos , Camundongos Knockout , Fosforilação , Proteína Regulatória Associada a mTOR/genética , Transdução de Sinais/fisiologia
11.
Biochem Biophys Res Commun ; 371(4): 756-61, 2008 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-18466765

RESUMO

Reproduction is accurately regulated by metabolic states in mammals. Adiponectin regulates luteinizing hormone (LH) secretion in the pituitary and energy homeostasis in the hypothalamus. We further investigated the gonadotropin-releasing hormone (GnRH) secretion regulation by adiponectin and its related molecular and electrophysiological mechanisms. The results showed that adiponectin receptors (AdipR1 and 2) were expressed in GT1-7 cells derived from hypothalamus neurons. GnRH secretion was inhibited via activation of AMP-activated protein kinase (AMPK). Moreover, we revealed that hyperpolarization of plasma membrane potentials and reduction of calcium influx was also caused by adiponectin.


Assuntos
Adiponectina/fisiologia , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Potenciais da Membrana , Quinases Proteína-Quinases Ativadas por AMP , Adiponectina/farmacologia , Animais , Cálcio/metabolismo , Linhagem Celular , Ativação Enzimática , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Hipotálamo/citologia , Hipotálamo/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Proteínas Quinases/metabolismo , Receptores de Adiponectina/metabolismo
12.
J Endocrinol ; 238(2): 137-149, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29929986

RESUMO

An increasing amount of evidence suggests that the delayed effect of antibiotics (abx) on gut microbiota after its cessation is not as favorable as its immediate effect on host metabolism. However, it is not known how the diverse abx-dependent metabolic effects influence diabetic subjects and how gut microbiota is involved. Here, we treated db/db mice with abx cocktail for 12 days and discontinued for 24 days. We found that db/db mice showed decreased body weight and blood glucose after abx treatment, which rapidly caught up after abx cessation. Twenty-four days after abx withdrawal, db/db mice exhibit increased plasma, hepatic total cholesterol (TC) levels and liver weight. The gut microbiota composition at that time showed decreased relative abundances (RAs) of Desulfovibrionaceae and Rikenellaceae, increased RA of Erysipelotrichaceae and Mogibacteriaceae, which were correlating with the reduced short-chain fatty acids (SCFAs) in gut content, such as propionic acid and valeric acid and with the elevated fecal taurine-conjugated bile acids (BAs) levels. The molecular biology studies showed inhibited hepatic BA synthesis from cholesterol, impeded intracellular transportation and biliary excretion of cholesterol that all conferred to liver TC accumulation. The associations among alterations of gut microbiota composition, microbial metabolite profiles and host phenotypes suggested the existence of gut microbiota-linked mechanisms that mediate the unfavorable delayed effects of abx on db/db mice cholesterol metabolism. Thus, we call upon the caution of applying abx in diabetic animal models for studying microbiota-host interaction and in type 2 diabetes subjects for preventing chronic cardiovascular consequences.


Assuntos
Antibacterianos/farmacologia , Glicemia/efeitos dos fármacos , Composição Corporal/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Diabetes Mellitus Tipo 2 , Microbioma Gastrointestinal/efeitos dos fármacos , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Animais , Glicemia/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/microbiologia , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/microbiologia , Diabetes Mellitus Tipo 2/patologia , Teste de Tolerância a Glucose , Insulina/metabolismo , Resistência à Insulina/fisiologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fatores de Tempo
13.
Nat Commun ; 8: 15755, 2017 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-28598424

RESUMO

Diabetes is associated with beta cell mass loss and islet dysfunctions. mTORC1 regulates beta cell survival, proliferation and function in physiological and pathological conditions, such as pregnancy and pancreatectomy. Here we show that deletion of Raptor, which is an essential component of mTORC1, in insulin-expressing cells promotes hypoinsulinemia and glucose intolerance. Raptor-deficient beta cells display reduced glucose responsiveness and exhibit a glucose metabolic profile resembling fetal beta cells. Knockout islets have decreased expression of key factors of functional maturation and upregulation of neonatal markers and beta cell disallowed genes, resulting in loss of functional maturity. Mechanistically, Raptor-deficient beta cells show reduced expression of DNA-methyltransferase 3a and altered patterns of DNA methylation at loci that are involved in the repression of disallowed genes. The present findings highlight a novel role of mTORC1 as a core mechanism governing postnatal beta cell maturation and physiologic beta cell mass during adulthood.


Assuntos
Diferenciação Celular , Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Proteína Regulatória Associada a mTOR/metabolismo , Animais , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , DNA Metiltransferase 3A , Diabetes Mellitus Tipo 2/genética , Feminino , Glucose/metabolismo , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/citologia , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Regulatória Associada a mTOR/genética
14.
Biochim Biophys Acta ; 1718(1-2): 67-73, 2005 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-16298331

RESUMO

The effects of sulfur dioxide (SO(2)) derivatives (bisulfite and sulfite, 1:3 M/M) on voltage-dependent sodium channel in isolated rat ventricular myocyte were studied using the whole cell patch-clamp technique. SO(2) derivatives increased sodium current (I(Na)) in a concentration-dependent manner. SO(2) derivatives at 10 microM significantly shifted steady-state inactivation curve of I(Na) to more positive potentials, but did not affect the activation curve. SO(2) derivatives markedly shifted the curve of time-dependent recovery of I(Na) from inactivation to the left, and accelerated the recovery of I(Na). SO(2) derivatives also significantly shortened the activation and inactivation time constants of I(Na). These results indicated that SO(2) derivatives produced concentration-dependent stimulation of cardiac sodium channels, which due mainly to the interaction of the drug with sodium channels in the inactivated state.


Assuntos
Miócitos Cardíacos/efeitos dos fármacos , Canais de Sódio/efeitos dos fármacos , Sulfitos/farmacologia , Dióxido de Enxofre/química , Animais , Ventrículos do Coração/citologia , Transporte de Íons/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Técnicas de Patch-Clamp , Ratos , Ratos Wistar , Sódio/metabolismo
15.
Food Chem Toxicol ; 44(3): 355-63, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16182427

RESUMO

The effects of sulfur dioxide (SO(2)) derivatives (bisulfite and sulfite, 1:3M/M) on voltage-dependent L-type calcium current (I(Ca,L)) in isolated rat ventricular myocytes were studied using the whole cell patch-clamp technique. SO(2) derivatives increased I(Ca,L) in a concentration-dependent manner. SO(2) derivatives shifted both the steady-state activation and the inactivation curves of I(Ca,L) to more positive potentials, the effect on the latter being more pronounced. SO(2) derivatives markedly accelerated the recovery of I(Ca,L) from inactivation. SO(2) derivatives also significantly shortened the fast and slow time constants of inactivation. These results suggested that SO(2) inhalation might cause cardiac myocyte injury through increasing intracellular calcium via voltage-gated calcium channels.


Assuntos
Poluentes Atmosféricos/farmacologia , Canais de Cálcio Tipo L/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Dióxido de Enxofre/farmacologia , Poluentes Atmosféricos/toxicidade , Animais , Canais de Cálcio Tipo L/metabolismo , Células Cultivadas , Relação Dose-Resposta a Droga , Potenciais da Membrana/efeitos dos fármacos , Miócitos Cardíacos/citologia , Técnicas de Patch-Clamp , Ratos , Ratos Wistar , Dióxido de Enxofre/toxicidade
16.
J Mol Endocrinol ; 56(3): 249-59, 2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-26744456

RESUMO

The Sidt2 global knockout mouse (Sidt2(-/-)) has impaired insulin secretion. The aim of this study was to assess the role of SIDT2 protein in glucose-induced insulin secretion in primary cultured mouse ß-cells. The major metabolic and electrophysiological steps of glucose-induced insulin secretion of primary cultured ß-cells from Sidt2(-/-) mice were investigated. The ß-cells from Sidt2(-/-) mice had normal NAD(P)H responses and KATP and KV currents. However, they exhibited a lower [Ca(2+)]i peak height when stimulated with 20mM glucose compared with those from WT mice. Furthermore, it took a longer time for the [Ca(2+)]i of ß-cell from Sidt2(-/-) mice to reach the peak. Pretreatment with ryanodine or 2-aminoethoxydiphenyl borate (2-APB) did not change [Ca(2+)]i the response pattern to glucose in Sidt2(-/-) cells. Extraordinarily, pretreatment with bafilomycin A1(Baf-A1) led to a comparable [Ca(2+)]i increase pattern between these two groups, suggesting that calcium traffic from the intracellular acidic compartment is defective in Sidt2(-/-) ß-cells. Bath-mediated application of 50nM nicotinic acid adenine dinucleotide phosphate (NAADP) normalized the [Ca(2+)]i response of Sidt2(-/-) ß-cells. Finally, glucose-induced CD38 expression increased to a comparable level between Sidt2(-/-) and WT islets, suggesting that Sidt2(-/-) islets generated NAADP normally. We conclude that Sidt2 is involved in NAADP-mediated release of calcium from insulin secretory granules and thus regulates insulin secretion.


Assuntos
Cálcio/metabolismo , Insulina/metabolismo , Proteínas de Membrana/metabolismo , NADP/análogos & derivados , Vesículas Secretórias/metabolismo , ADP-Ribosil Ciclase 1/genética , ADP-Ribosil Ciclase 1/metabolismo , Animais , Sinalização do Cálcio , Expressão Gênica , Perfilação da Expressão Gênica , Glucose/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Lisossomos/metabolismo , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , NADP/metabolismo , Proteínas de Transporte de Nucleotídeos , Transporte Proteico
17.
Endocrine ; 53(1): 117-28, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26818915

RESUMO

Beta cell replication is the major component for maintenance of beta cell mass in adult rodents; however, little is known about what is the earliest signals that initiate rodent beta cell proliferation. The mTORC1 pathway integrates signals from growth factors and nutrients and regulates cell growth and survival. Here, we used normoglycemic 60 % partial-pancreatectomy (60 % Px) mouse model to determine whether mTORC1 pathway was required for compensatory beta cell proliferation. C57BL/6 J male mice were subjected to 60 % Px or sham operation, and subsequently treated with either rapamycin or vehicle for 7 days. Metabolic profile, pancreatic beta cell mass, and proliferation were examined, and expression levels of cell cycle regulators were determined. Beta cell proliferation was increased by 2.5-fold, and mTORC1 signaling was activated in islets post-Px. Rapamycin treatment impaired glucose tolerance and glucose stimulating insulin secretion in 60 % Px mice, but did not affect their insulin sensitivity in peripheral tissue. Rapamycin inhibited mTORC1 activity in beta cells, suppressed compensatory beta cell proliferation and growth, and reduced beta cell mass and insulin content in 60 % Px mice. Px caused an increase of the cyclin D2 at protein level and promoted cyclin D2 nuclear localization in an mTOR-dependent manner. Disrupting mTORC1 signaling suppressed cell proliferation and simultaneously diminished cyclin D2 protein abundance in RINm5F cells. Our data demonstrated that mTORC1 plays an essential role in beta cell adaption to significant beta cell mass loss in 60 % Px model and in early compensatory beta cell proliferation via cyclin D2 pathway.


Assuntos
Proliferação de Células/fisiologia , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Complexos Multiproteicos/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Glicemia/metabolismo , Ciclina D2/metabolismo , Insulina/sangue , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Endogâmicos C57BL , Pancreatectomia
18.
Food Chem Toxicol ; 43(2): 225-32, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15621334

RESUMO

The effects of sodium metabisulfite (SMB), a food preservative mostly used in food and drug industries, on voltage-dependent potassium currents in acutely isolated hippocampal CA1 pyramidal neurons of rat were studied using the whole-cell patch-clamp techniques. SMB increased transient outward potassium current (IA) and delayed rectifier potassium current (IK) in a concentration-dependent manner. 10 microM SMB shifted the steady-state activation curve of IK to more negative potentials, and the steady-state inactivation curves of IA and IK to more positive potentials. Time to peak of IA was not affected, but the decay of IA was delayed by SMB. However, the activation and inactivation time constants of IK were both decreased by SMB. These results suggested that SMB differently affected IA and IK, and it would decrease the excitability of hippocampal neuron by increasing potassium currents.


Assuntos
Conservantes de Alimentos/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Potássio/farmacocinética , Células Piramidais/fisiologia , Sulfitos/farmacologia , Animais , Relação Dose-Resposta a Droga , Eletrofisiologia , Hipocampo/citologia , Técnicas In Vitro , Potenciais da Membrana/fisiologia , Técnicas de Patch-Clamp , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/fisiologia , Células Piramidais/efeitos dos fármacos , Ratos , Ratos Wistar
19.
Environ Toxicol Pharmacol ; 20(1): 35-41, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21783565

RESUMO

The effect of sodium metabisulfite (SMB) on voltage-gated sodium channel currents (I(Na)) was examined in freshly isolated rat hippocampal CA1 neurons using whole-cell patch-clamp technique under voltage-clamp conditions. SMB irreversibly enhanced I(Na) in a concentration-dependent manner, shifted the inactivation curve to more positive potential, without affecting the current activation curve. In addition, SMB increased the time to peak and the inactivation time constant of I(Na). Superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPx) could all partly inhibit the effect of SMB on the sodium current. These results suggested that SMB have neuronal toxicity by increasing the excitability of neurons and its mechanism might involve the oxidative damage on ion channels.

20.
Hypertension ; 65(3): 622-8, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25624344

RESUMO

Recent studies have shown that somatic mutations in the KCNJ5, ATP1A1, ATP2B3, and CACNA1D genes are associated with the pathogenesis of aldosterone-producing adenoma. Clinical profile and biochemical characteristics of the mutations in Chinese patients with aldosterone-producing adenoma remain unclear. In this study, we performed DNA sequencing in 168 Chinese patients with aldosterone-producing adenoma and found 129 somatic mutations in KCNJ5, 4 in ATP1A1, 1 in ATP2B3, and 1 in CACNA1D. KCNJ5 mutations were more prevalent in female patients and were associated with larger adenomas, higher aldosterone excretion, and lower minimal serum K(+) concentration. More interestingly, we identified a novel somatic KCNJ5 mutation (c.445-446insGAA, p.T148-T149insR) that could enhance CYP11B2 mRNA upregulation and aldosterone release. This mutation could also cause membrane depolarization and intercellular Ca(2+) increase. In conclusion, somatic KCNJ5 mutations are conspicuously more popular than mutations of other genes in aldosterone-producing adenomas of Chinese patients. The T148-T149insR mutation in KCNJ5 may influence K(+) channel selectivity and autonomous aldosterone production.


Assuntos
Neoplasias do Córtex Suprarrenal/genética , Neoplasias do Córtex Suprarrenal/metabolismo , Adenoma Adrenocortical/genética , Adenoma Adrenocortical/metabolismo , Aldosterona/metabolismo , Povo Asiático/genética , Mutação/genética , Fenótipo , Adulto , Sequência de Bases , Canais de Cálcio Tipo L/genética , Feminino , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/genética , Humanos , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , ATPases Transportadoras de Cálcio da Membrana Plasmática/genética , Potássio/sangue , Prevalência , Estudos Retrospectivos , Fatores Sexuais , ATPase Trocadora de Sódio-Potássio/genética , Carga Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA