Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Arch Biochem Biophys ; 749: 109789, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37852426

RESUMO

Clathrin-dependent endocytosis is a key process for secretory cells, in which molecules on the plasma membrane are both degraded and recycled in a stimulus-dependent manner. There are many reports showing that disruption of endocytosis is involved in the onset of various diseases. Recently, it has been reported that such disruption in pancreatic ß-cells causes impaired insulin secretion and might be associated with the pathology of diabetes mellitus. Compared with exocytosis, there are few reports on the molecular mechanism of endocytosis in pancreatic ß-cells. We previously reported that GDP-bound Rab27a regulates endocytosis through its GDP-dependent effectors after insulin secretion. In this study, we identified heat shock protein family A member 8 (HSPA8) as a novel interacting protein for GDP-bound Rab27a. HSPA8 directly bound GDP-bound Rab27a via the ß2 region of its substrate binding domain (SBD). The ß2 fragment was capable of inhibiting the interaction between HSPA8 and GDP-bound Rab27a, and suppressed glucose-induced clathrin-dependent endocytosis in pancreatic ß-cells. The region also affected clathrin dynamics on purified clathrin-coated vesicles (CCVs). These results suggest that the interaction between GDP-bound Rab27a and HSPA8 regulates clathrin disassembly from CCVs and subsequent vesicle transport. The regulatory stages in endocytosis by HSPA8 differ from those for other GDP-bound Rab27a effectors. This study shows that GDP-bound Rab27a dominantly regulates each stage in glucose-induced endocytosis through its specific effectors in pancreatic ß-cells.


Assuntos
Clatrina , Proteínas rab de Ligação ao GTP , Secreção de Insulina , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas rab27 de Ligação ao GTP/metabolismo , Clatrina/metabolismo , Endocitose/fisiologia , Glucose/metabolismo , Insulina/metabolismo
2.
Biol Pharm Bull ; 46(4): 630-635, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37005308

RESUMO

The improvement of type 2 diabetes mellitus induced by naturally occurring polyphenols, known as flavonoids, has received considerable attention. However, there is a dearth of information regarding the effect of the trihydroxyflavone apigenin on pancreatic ß-cell function. In the present study, the anti-diabetic effect of apigenin on pancreatic ß-cell insulin secretion, apoptosis, and the mechanism underlying its anti-diabetic effects, were investigated in the INS-ID ß-cell line. The results showed that apigenin concentration-dependently facilitated 11.1-mM glucose-induced insulin secretion, which peaked at 30 µM. Apigenin also concentration-dependently inhibited the expression of endoplasmic reticulum (ER) stress signaling proteins, CCAAT/enhancer binding protein (C/EBP) homologous protein (CHOP) and cleaved caspase-3, which was elevated by thapsigargin in INS-1D cells, with peak suppression at 30 µM. This was strongly correlated with the results of flow cytometric analysis of annexin V/propidium iodide (PI) staining and DNA fragmentation analysis. Moreover, the increased expression of thioredoxin-interacting protein (TXNIP) induced by thapsigargin was remarkably reduced by apigenin in a concentration-dependent manner. These results suggest that apigenin is an attractive candidate with remarkable and potent anti-diabetic effects on ß-cells, which are mediated by facilitating glucose-stimulated insulin secretion and preventing ER stress-mediated ß-cell apoptosis, the latter of which may be possibly mediated by reduced expression of CHOP and TXNIP, thereby promoting ß-cell survival and function.


Assuntos
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Humanos , Diabetes Mellitus Tipo 2/metabolismo , Apigenina/farmacologia , Tapsigargina/metabolismo , Tapsigargina/farmacologia , Apoptose , Estresse do Retículo Endoplasmático , Glucose/metabolismo , Fator de Transcrição CHOP/metabolismo
3.
Biol Pharm Bull ; 46(5): 640-646, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37121690

RESUMO

Although diabetes is associated with an increased risk of various diseases, including cancer and infectious diseases, no definitive cure has yet been found. Long-term treatment for blood glucose control significantly reduces the QOL. Pancreatic ß-cells are the only cells that can lower blood glucose levels by secreting insulin. Therefore, maintaining insulin-secreting ß-cells is crucial in preventing the progression of diabetes and improving the QOL. We have investigated the mechanisms for the regulation of insulin secretion, the prevention of ß-cell apoptosis, and the increase in ß-cell mass. In particular, we have elucidated the involvement of type I diacylglycerol kinase (DGK) in the regulation of insulin secretion and the effects of nitric oxide (NO) signaling and natural products in suppressing ß-cell death. In addition, we have elucidated the function of DGKδ as a suppressor of ß-cell proliferation. This review introduces the findings of our study leading to development of novel anti-diabetic therapeutics that targets pancreatic ß-cells.


Assuntos
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Humanos , Diabetes Mellitus Tipo 2/tratamento farmacológico , Qualidade de Vida , Insulina/metabolismo , Secreção de Insulina , Transdução de Sinais , Apoptose
4.
Front Nutr ; 10: 1336133, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38249612

RESUMO

Introduction: Nobiletin is a polymethoxyflavonoid abundant in citrus peels and has been reported to have various bioactive effects. We have previously reported that nobiletin inhibits endoplasmic reticulum stress-induced apoptosis in the pancreatic ß-cell line INS-1 and that continuous subcutaneous administration of nobiletin suppresses the progression of diabetes by protecting ß-cells in type 2 diabetic db/db mice. In the present study, we investigated effects of oral ingestion of Shiikuwasha extract rich in nobiletin on the pathogenesis of type 2 diabetes in db/db mice. Materials and methods: A Shiikuwasha extract was dissolved in MediDrop sucralose. Twenty-four mice were equally divided in three groups and fed with vehicle or low or high dose of Shiikuwasha extract for 4 weeks. Blood glucose levels, pancreatic ß-cell mass, serum insulin levels, pancreatic insulin content, and other biomarkers were measured and compared between the groups. Results: The group that freely ingested the Shiikuwasha extract containing higher concentration of nobiletin (Shiikuwasha H) showed lower blood glucose levels. At the end of the experiment, the Shiikuwasha H group exhibited improved glucose tolerance, lower serum glycoalbumin levels, and an increase in ß-cell area per pancreas compared with the control group. Body weight, food intake, and serum biomarkers related to liver function and lipid metabolism of the Shiikuwasha H group were not different from those of the control group, although water intake of the former was significantly decreased than that of the latter. Conclusion: Our results suggest that the oral ingestion of Shiikuwasha extract preserves pancreatic ß-cell mass in diabetic mice, which might be attributed to ameliorating the progression of diabetes.

5.
Biochem Biophys Res Commun ; 637: 108-116, 2022 12 31.
Artigo em Inglês | MEDLINE | ID: mdl-36395692

RESUMO

Low concentrations of nitric oxide (NO) produced by constitutive NO synthase (cNOS) has been shown to suppress apoptosis in pancreatic ß-cells. In the present study, the influence of asymmetric dimethylarginine (ADMA), the major endogenous inhibitor of NOS, on the apoptosis-suppressive effect of NO was investigated. The expression of dimethylarginine dimethylaminohydrolase 2 (DDAH2), an ADMA-metabolizing enzyme, in INS-1 ß-cells and in mouse pancreatic islets was drastically reduced by in vitro exposure to high-concentration glucose (20 mM) and by in vivo treatment of mice with the insulin receptor blocker S661, which resulted in hyperglycemia, respectively. In line with this, a higher ADMA level was observed in INS-1 cells exposed to 20 mM glucose. The treatment of INS-1 cells with ADMA, similarly to with the NOS inhibitor NG-nitro-L-arginine methyl ester, significantly facilitated 20 mM glucose-induced increase in cleaved caspase-3 protein expression. Furthermore, increased protein expression of cleaved caspase-3 and CHOP was observed in INS-1 cells with knockdown of DDAH2. These results suggest that ADMA accumulation through a decrease in DDAH2 expression in ß-cells, which is induced under hyperglycemic conditions, facilitates ß-cell apoptosis through suppression of cNOS-mediated NO production.


Assuntos
Hiperglicemia , Óxido Nítrico , Animais , Camundongos , Caspase 3 , Apoptose , Glucose
6.
Yakugaku Zasshi ; 142(5): 457-463, 2022.
Artigo em Japonês | MEDLINE | ID: mdl-35491149

RESUMO

Depression of lipid metabolism in ß-cells has been indicated to be one of the causes of impaired insulin secretion in type 2 diabetes. Diacylglycerol (DAG) is an important lipid mediator and is known to regulate insulin secretion in pancreatic ß-cells. Intracellular DAG accumulation is involved in ß-cell dysfunction in the pathogenesis of type 2 diabetes; thus, the regulation of intracellular DAG levels is likely important for maintaining the ß-cell function. We focused on diacylglycerol kinases (DGKs), which strictly regulate intracellular DAG levels, and analyzed the function of type I DGKs (DGKα, γ), which are activated by intracellular Ca2+ and expressed in the cytoplasm, in ß-cells. The suppression of the DGKα and γ expression decreased the insulin secretory response, and the decreased expression of DGKα and γ was observed in islets of diabetic model mice. In the pancreatic ß-cell line MIN6, 1 µM R59949 (a type I DGK inhibitor) and 10 µM DiC8 (a cell permeable DAG analog) enhanced glucose-induced [Ca2+]i oscillation in a PKC-dependent manner, while 10 µM R59949 and 100 µM DiC8 suppressed [Ca2+]i oscillation and voltage-dependent Ca2+ channel activity in a PKC-independent manner. These results suggest that the intracellular accumulation of DAG by the loss of the DGKα and γ functions regulates insulin secretion in a dual manner depending on the degree of DAG accumulation. The regulation of the insulin secretory response through DAG metabolism by type I DGKs may change depending on the degree of progression of type 2 diabetes.


Assuntos
Diabetes Mellitus Tipo 2 , Insulinas , Animais , Diabetes Mellitus Tipo 2/metabolismo , Diacilglicerol Quinase/fisiologia , Diglicerídeos/metabolismo , Secreção de Insulina , Insulinas/metabolismo , Camundongos
7.
Biochem Biophys Res Commun ; 600: 136-141, 2022 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-35219102

RESUMO

Liver fibrosis is a major consequence of chronic liver disease, where excess extracellular matrix is deposited, due caused by the activation of hepatic stellate cells (HSCs). The suppression of collagen production in HSCs is therefore regarded as a therapeutic target of liver fibrosis. The present study investigated effects of harmine, which is a ß-carboline alkaloid and known as an inhibitor of dual-specificity tyrosine-regulated kinases (DYRKs), on the production of collagen in HSCs. LX-2 cells, a human HSC cell line, were treated with harmine (0-10 µM) for 48 h in the presence or absence of TGF-ß1 (5 ng/ml). The expression of collagen type I α1 (COL1A1) and DYRK isoforms was investigated by Western blotting, quantitative RT-PCR, or immunofluorescence. The influence of knockdown of each DYRK isoform on the COL1A1 expression was further investigated. The expression of COL1A1 was markedly increased by treating with TGF-ß1 for 48 h in LX-2 cells. Harmine (10 µM) significantly inhibited the increased expression of COL1A1. LX-2 cells expressed mRNAs of DYRK1A, DYRK1B, DYRK2, and DYRK4, although the expression of DYRK4 was much lower than the others. Knockdown of DYRK1B, but not DYRK1A or DYRK2, with siRNA significantly suppressed TGF-ß1-induced increase in COL1A1 expression. These results suggest that harmine suppresses COL1A1 expression via inhibiting DYRK1B in HSCs and therefore might be effective for the treatment of liver fibrosis.


Assuntos
Cadeia alfa 1 do Colágeno Tipo I , Harmina , Células Estreladas do Fígado , Proteínas Serina-Treonina Quinases , Proteínas Tirosina Quinases , Fator de Crescimento Transformador beta1 , Cadeia alfa 1 do Colágeno Tipo I/antagonistas & inibidores , Cadeia alfa 1 do Colágeno Tipo I/biossíntese , Harmina/farmacologia , Células Estreladas do Fígado/efeitos dos fármacos , Células Estreladas do Fígado/metabolismo , Humanos , Cirrose Hepática/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Quinases Dyrk
8.
FASEB J ; 35(5): e21420, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33774855

RESUMO

Although an aberrant reduction in pancreatic ß-cell mass contributes to the pathogenesis of diabetes, the mechanism underlying the regulation of ß-cell mass is poorly understood. Here, we show that diacylglycerol kinase δ (DGKδ) is a key enzyme in the regulation of ß-cell mass. DGKδ expression was detected in the nucleus of ß-cells. We developed ß-cell-specific DGKδ knockout (ßDGKδ KO) mice, which showed lower blood glucose, higher plasma insulin levels, and better glucose tolerance compared to control mice. Moreover, an increased number of small islets and Ki-67-positive islet cells, as well as elevated cyclin B1 expression in the islets, were detected in the pancreas of ßDGKδ KO mice. DGKδ knockdown in the ß-cell line MIN6 induced significant increases in bromodeoxyuridine (BrdU) incorporation and cyclin B1 expression. Finally, we confirmed that streptozotocin-induced hyperglycemia and ß-cell loss were alleviated in ßDGKδ KO mice. Thus, suppressing the expression or enzymatic activity of DGKδ that functions as a suppressor of ß-cell proliferation could be a novel therapeutic approach to increase ß-cell mass for the treatment of diabetes.


Assuntos
Encéfalo/enzimologia , Proliferação de Células , Diabetes Mellitus Experimental/complicações , Diacilglicerol Quinase/fisiologia , Hiperglicemia/prevenção & controle , Células Secretoras de Insulina/metabolismo , Animais , Hiperglicemia/etiologia , Hiperglicemia/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais
9.
Nihon Yakurigaku Zasshi ; 155(4): 209-213, 2020.
Artigo em Japonês | MEDLINE | ID: mdl-32612030

RESUMO

The number of patients with type 2 diabetes mellitus (T2DM) has rapidly increased, especially in East and Southeast Asia. In these areas, in general, people have especially vulnerable ß-cells and insulin secretion deficiency and reduced ß-cell mass are the primary cause of T2DM. Therefore, the alleviation of such ß-cell dysfunction would provide therapeutic approaches to prevent the development of T2DM. Nobiletin, a polymethoxylated flavonoid found in citrus fruits, has been shown to improve obesity and insulin resistance in T2DM model mice. We focused on ß-cells and investigated the effects of nobiletin on insulin secretion and ß-cell apoptosis. In ß-cell line INS-1, nobiletin increased glucose-induced insulin secretion (GSIS) in a concentration-dependent manner, which was inhibited by an Epac inhibitor. In addition, nobiletin at 10 µM inhibited thapsigargin-induced apoptosis, which was inhibited by a PKA inhibitor. Nobiletin also suppressed thapsigargin-induced increases in cleaved caspase-3 and phosphorylated JNK. Thus, nobiletin is suggested to promote GSIS and prevent ER stress-induced ß-cell apoptosis, which are mediated via Epac and PKA-dependent pathways, respectively. In summary, nobiletin is suggested to exhibit insulinotropic and anti-apoptotic effects on ß-cells, which are one of the causes of its anti-diabetic effect. Moreover, nobiletin seems to be able to alleviate the development of T2DM by protecting ß-cells from apoptosis.


Assuntos
Citrus , Diabetes Mellitus Tipo 2 , Flavonas , Células Secretoras de Insulina , Animais , Apoptose , Diabetes Mellitus Tipo 2/tratamento farmacológico , Flavonas/farmacologia , Flavonas/uso terapêutico , Flavonoides/farmacologia , Flavonoides/uso terapêutico , Humanos , Camundongos
10.
Sci Rep ; 10(1): 5198, 2020 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-32251344

RESUMO

Sialidase cleaves sialic acid residues from a sialoglycoconjugate: oligosaccharides, glycolipids and glycoproteins that contain sialic acid. Histochemical imaging of the mouse pancreas using a benzothiazolylphenol-based sialic acid derivative (BTP3-Neu5Ac), a highly sensitive histochemical imaging probe used to assess sialidase activity, showed that pancreatic islets have intense sialidase activity. The sialidase inhibitor 2,3-dehydro-2-deoxy-N-acetylneuraminic acid (DANA) remarkably enhances glutamate release from hippocampal neurons. Since there are many similar processes between synaptic vesicle exocytosis and secretory granule exocytosis, we investigated the effect of DANA on insulin release from ß-cells. Insulin release was induced in INS-1D cells by treatment with 8.3 mM glucose, and the release was enhanced by treatment with DANA. In a mouse intraperitoneal glucose tolerance test, the increase in serum insulin levels was enhanced by intravenous injection with DANA. However, under fasting conditions, insulin release was not enhanced by treatment with DANA. Calcium oscillations induced by 8.3 mM glucose treatment of INS-1D cells were not affected by DANA. Blood insulin levels in sialidase isozyme Neu3-deficient mice were significantly higher than those in WT mice under ad libitum feeding conditions, but the levels were not different under fasting conditions. These results indicate that DANA is a glucose-dependent potentiator of insulin secretion. The sialidase inhibitor may be useful for anti-diabetic treatment with a low risk of hypoglycemia.


Assuntos
Glucose/fisiologia , Secreção de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/efeitos dos fármacos , Ácido N-Acetilneuramínico/análogos & derivados , Neuraminidase/antagonistas & inibidores , Animais , Benzotiazóis/química , Sinalização do Cálcio/efeitos dos fármacos , Corantes/análise , Avaliação Pré-Clínica de Medicamentos , Jejum/sangue , Teste de Tolerância a Glucose , Hipoglicemiantes/farmacologia , Injeções Intravenosas , Insulina/sangue , Secreção de Insulina/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ácido N-Acetilneuramínico/farmacologia , Neuraminidase/fisiologia , Ácidos Siálicos/química
11.
J Pharmacol Sci ; 140(2): 178-186, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31279581

RESUMO

The role of type I diacylglycerol kinases (DGKs) in the regulation of insulin secretion was investigated in MIN6 ß-cells. In intracellular Ca2+ concentration ([Ca2+]i) measurement experiments, 1 µM R59949, a type I DGK inhibitor, and 10 µM DiC8, a diacylglycerol (DAG) analog, amplified 22.2 mM glucose-induced [Ca2+]i oscillations in a protein kinase C (PKC)-dependent manner, whereas 10 µM R59949 and 100 µM DiC8 decreased [Ca2+]i independent of PKC. High concentrations of R59949 and DiC8 attenuated voltage-dependent Ca2+ channel currents. According to these results, 22.2 mM glucose-stimulated insulin secretion (GSIS) was potentiated by 1 µM R59949 but suppressed by 10 µM of the same. The DGKα inhibitor R59022 showed a similar dual effect. Conversely, DiC8 at 10 and 100 µM potentiated GSIS, although 100 µM DiC8 decreased [Ca2+]i. These results suggest that DAG accumulated through declined type I DGK activity shows a dual effect on insulin secretion depending on the degree of accumulation; a mild DAG accumulation induces a PKC-dependent stimulatory effect on insulin secretion, whereas an excessive DAG accumulation suppresses it in a PKC-independent manner, possibly via attenuation of VDCC activity.


Assuntos
Diacilglicerol Quinase/fisiologia , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Animais , Cálcio/metabolismo , Canais de Cálcio/metabolismo , Células Cultivadas , Diacilglicerol Quinase/antagonistas & inibidores , Diglicerídeos/metabolismo , Relação Dose-Resposta a Droga , Glucose/farmacologia , Secreção de Insulina/efeitos dos fármacos , Camundongos , Piperidinas/farmacologia , Proteína Quinase C/fisiologia , Quinazolinonas/farmacologia
12.
Am J Physiol Cell Physiol ; 316(3): C434-C443, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30649920

RESUMO

ß-Cell swelling induces membrane depolarization, which has been suggested to be caused at least partly by the activation of cation channels. Here, we show the identification of the cation channels. In isolated mouse pancreatic ß-cells, the exposure to 30% hypotonic solution elicited an increase in cytosolic Ca2+ concentration ([Ca2+]c). The [Ca2+]c elevation was partially inhibited by ruthenium red, a blocker of several Ca2+-permeable channels including transient receptor potential vanilloid receptors [transient receptor potential cation channel subfamily V (TRPV)], and by nicardipine, but not by the depletion of intracellular Ca2+ stores with thapsigargin and caffeine. The hypotonic stimulation also increased insulin secretion from isolated mouse islets, which was significantly suppressed by ruthenium red. Expression of TRPV2 and TRPV4 was confirmed in mouse pancreatic islets and the MIN6 ß-cell line by RT-PCR, Western blot, and immunohistochemical analyses. However, neither 4α-phorbol 12,13-didecanoate nor GSK1016790A, TRPV4 activators, showed any apparent effect on [Ca2+]c in isolated mouse ß-cells or in MIN6 cells. In contrast, probenecid, a TRPV2 activator, induced an increase in [Ca2+]c in MIN6 cells, which was attenuated by ruthenium red. Moreover, the [Ca2+]c elevation induced by 30% hypotonic stimulation was significantly reduced by knockdown of TRPV2 with siRNA and by tranilast, a TRPV2 inhibitor. The knockdown of TRPV2 also decreased insulin secretion induced by the hypotonic stimulation. In addition, glucose-stimulated insulin secretion was also significantly reduced in the TRPV2-knockdown MIN6 cells. These results suggest that osmotic cell swelling activates TRPV2 in mouse ß-cells, thereby causing membrane depolarization and subsequent activation of voltage-dependent Ca2+ channels and insulin secretion.


Assuntos
Canais de Cálcio/metabolismo , Secreção de Insulina/fisiologia , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Canais de Cátion TRPV/metabolismo , Animais , Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Membrana Celular/metabolismo , Células Cultivadas , Citosol/metabolismo , Glucose/metabolismo , Masculino , Camundongos , RNA Interferente Pequeno/metabolismo
13.
Yakugaku Zasshi ; 136(12): 1623-1629, 2016.
Artigo em Japonês | MEDLINE | ID: mdl-27904096

RESUMO

Pancreatic ß-cell dysfunction is a major feature of type 2 diabetes. Therefore maintenance of ß-cell function is essential to preventing the onset and progression of type 2 diabetes. To elucidate the mechanisms underlying the regulation of insulin secretion and ß-cell survival, we particularly focused on the roles of gasotransmitters in pancreatic ß-cells. Nitric oxide (NO) and hydrogen sulfide (H2S) are recognized as toxic gases. However, they are also vital physiological and pathophysiological mediators in various cell types. NO, generated from L-arginine by reactions catalyzed by NO synthases, is a well-known neurotransmitter and smooth muscle relaxation factor. In pancreatic ß-cells, induction of nitric oxide synthase 2 (NOS2) by inflammatory cytokines generates a large amount of NO, which contributes to the impairment of ß-cell function and induction of ß-cell apoptosis, which are, in turn, involved in the development of type 1 diabetes. In contrast, a physiological level of NO, generated by constitutive NOS (cNOS), acts as a positive or negative regulator of insulin secretion and ß-cell survival, depending on concentration. H2S generated from L-cysteine has been shown to play a role of neuromodulator, and this gas possesses cytoprotective properties. In pancreatic ß-cells, H2S functions as a potent suppressor of insulin secretion. Furthermore, chronic exposure to high glucose induces H2S production by increasing the expression of a H2S-producing enzyme, cystathionine γ-lyase (CSE). H2S generated by CSE prevents ß-cell apoptosis via an antioxidant mechanism. Here, we describe the current understanding of the function of gasotransmitters in regulating insulin secretion and pancreatic ß-cell survival.


Assuntos
Diabetes Mellitus Tipo 2/fisiopatologia , Diabetes Mellitus Tipo 2/terapia , Sulfeto de Hidrogênio , Células Secretoras de Insulina/fisiologia , Terapia de Alvo Molecular , Óxido Nítrico/fisiologia , Antioxidantes , Apoptose/efeitos dos fármacos , Arginina/metabolismo , Catálise , Cistationina gama-Liase/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Relação Dose-Resposta a Droga , Humanos , Sulfeto de Hidrogênio/metabolismo , Sulfeto de Hidrogênio/farmacologia , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Óxido Nítrico/metabolismo , Óxido Nítrico/farmacologia , Óxido Nítrico Sintase/fisiologia
14.
Yakugaku Zasshi ; 136(3): 461-5, 2016.
Artigo em Japonês | MEDLINE | ID: mdl-26935087

RESUMO

The appropriate secretion of insulin from pancreatic ß-cells is essential for regulating blood glucose levels. Glucose-stimulated insulin secretion (GSIS) involves the following steps: Glucose uptake by pancreatic ß-cells is metabolized to produce ATP. Increased ATP levels result in the closure of ATP-sensitive K(+) (KATP) channels, resulting in membrane depolarization that activates voltage-dependent Ca(2+) channels to subsequently trigger insulin secretion. In addition to this primary mechanism through KATP channels, insulin secretion is regulated by cyclic AMP and diacylglycerol (DAG), which mediate the effects of receptor agonists such as GLP-1 and acetylcholine. Glucose by itself can also increase the levels of these second messengers. Recently, we have shown an obligatory role of diacylglycerol kinase (DGK), an enzyme catalyzing the conversion of DAG to phosphatidic acid, in GSIS. Of the 10 known DGK isoforms, we focused on type-I DGK isoforms (i.e., DGKα, DGKß, and DGKγ), which are activated by Ca(2+). The protein expression of DGKα and DGKγ was detected in mouse pancreatic islets and the pancreatic ß-cell line MIN6. Depletion of these DGKs by a specific inhibitor or siRNA decreased both [Ca(2+)]i and insulin secretion in MIN6 cells. Similar [Ca(2+)]i responses were induced by DiC8, a membrane-permeable DAG analog. These results suggest that DGKα and DGKγ play crucial roles in insulin secretion, and that their depletion impairs insulin secretion through DAG accumulation. In this article, we review the current understanding of the roles of DAG- and DGK-signaling in pancreatic ß-cells, and discuss their pathophysiological roles in the progression of type-2 diabetes.


Assuntos
Diabetes Mellitus Tipo 2/etiologia , Diabetes Mellitus Tipo 2/metabolismo , Diacilglicerol Quinase/fisiologia , Descoberta de Drogas , Células Secretoras de Insulina/metabolismo , Metabolismo dos Lipídeos , Terapia de Alvo Molecular , Trifosfato de Adenosina/metabolismo , Animais , Canais de Cálcio/metabolismo , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diglicerídeos/metabolismo , Glucose/metabolismo , Humanos , Insulina/metabolismo , Secreção de Insulina , Canais KATP/metabolismo , Camundongos
15.
Biol Pharm Bull ; 38(10): 1599-605, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26424020

RESUMO

Our previous study using apoptosis analysis suggested that Ca(2+) release through inositol 1,4,5-trisphosphate (IP3) receptors and the subsequent Ca(2+) influx through store-operated channels (SOCs) constitute a triggering signal for H2O2-induced ß-cell apoptosis. In the present study, we further examined the obligatory role of early Ca(2+) responses in ß-cell apoptosis induction. H2O2 induced elevation of the cytosolic Ca(2+) concentration ([Ca(2+)]c) consisting of two phases: an initial transient [Ca(2+)]c elevation within 30 min and a slowly developing one thereafter. The first phase was almost abolished by 2-aminoethoxydiphenylborate (2-APB), which blocks IP3 receptors and cation channels including SOCs, while the second phase was only partially inhibited by 2-APB. The inhibition by 2-APB of the second phase was not observed when 2-APB was added 30 min after the treatment with H2O2. 2-APB also largely inhibited elevation of the mitochondrial Ca(2+) concentration ([Ca(2+)]m) induced by H2O2 when 2-APB was applied simultaneously with H2O2, but not when applied 30 min after H2O2 application. In addition, 2-APB inhibited the release of mitochondrial cytochrome c to the cytosol induced by H2O2 when 2-APB was applied simultaneously with H2O2 but not 30 min post-treatment. H2O2-induced [Ca(2+)]m elevation and cell death were not inhibited by Ru360, an inhibitor of the mitochondrial calcium uniporter (MCU). These results suggest that the H2O2-induced initial [Ca(2+)]c elevation, occurring within 30 min and mediated by Ca(2+) release through IP3 receptors and subsequent Ca(2+) influx through SOCs, leads to [Ca(2+)]m elevation, possibly through a mechanism independent of MCU, thereby inducing cytochrome c release and consequent apoptosis.


Assuntos
Apoptose/fisiologia , Cálcio/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Células Secretoras de Insulina/metabolismo , Animais , Apoptose/efeitos dos fármacos , Compostos de Boro/farmacologia , Linhagem Celular , Citocromos c/metabolismo , Peróxido de Hidrogênio , Receptores de Inositol 1,4,5-Trifosfato/antagonistas & inibidores , Células Secretoras de Insulina/efeitos dos fármacos , Mitocôndrias/metabolismo , Ratos
16.
Biol Pharm Bull ; 38(5): 669-73, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25947912

RESUMO

Diacylglycerol (DAG) is a lipid signal messenger and plays a physiological role in ß-cells. Since defective glucose homeostasis increases de novo DAG synthesis, DAG may also contribute to ß-cell dysfunction in type 2 diabetes. Although the primary function of DAG is to activate protein kinase C (PKC), the role of PKC in insulin secretion is controversial: PKC has been reported to act as both a positive and negative regulator of insulin secretion. In addition to the PKC pathway, DAG has also been shown to mediate other pathways such as the Munc-13-dependent pathway in ß-cells. The intracellular levels of DAG are strictly regulated by diacylglycerol kinase (DGK); however, the role of DGK in ß-cells and their involvement in ß-cell failure in type 2 diabetes remain to be fully elucidated. We have recently reported the roles of type I DGK, DGKα and γ, in insulin secretion from ß-cells. DGKα and γ were activated by glucose or high K(+) stimulation in ß-cells, and the inhibition of the DGKs by a type I DGK inhibitor or by knockdown with small interfering RNA (siRNA) decreased insulin secretion. Thus, DGKα and γ are suggested to be activated in response to elevated [Ca(2+)]i in ß-cells and to act as positive regulators of insulin secretion. In this article, we review the current understanding of the roles of DAG and DGK in ß-cell function and their involvement in the development of ß-cell dysfunction in type 2 diabetes.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Diacilglicerol Quinase/metabolismo , Diglicerídeos/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Proteína Quinase C/metabolismo , Animais , Cálcio/metabolismo , Glucose/metabolismo , Humanos , Secreção de Insulina
17.
Biol Pharm Bull ; 38(3): 476-81, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25757931

RESUMO

The effects of green tea catechins on glucose-stimulated insulin secretion (GSIS) were investigated in the ß-cell line INS-1D. Epigallocatechin gallate (EGCG) at 10 µM or gallocatechin gallate (GCG) at 30 µM caused significant inhibitory effects on GSIS, and each of these at 100 µM almost abolished it. In contrast, epicatechin (EC) or catechin (CA) had no effect on GSIS at concentrations up to 100 µM. We thus investigated the structure-activity relationship by using epigallocatechin (EGC) and gallocatechin (GC) containing a trihydroxyl group in the B-ring, and epicatechin gallate (ECG) and catechin gallate (CG) containing the gallate moiety. EGC, GC, and ECG caused an inhibition of GSIS, although significant effects were obtained only at 100 µM. At this concentration, EGC almost abolished GSIS, whereas GC and ECG partially inhibited it. In contrast, CG did not affect GSIS at concentrations up to 100 µM. EGCG also abolished the insulin secretion induced by tolbutamide, an ATP-sensitive K(+) channel blocker, and partially inhibited that induced by 30 mM K(+). Moreover, EGCG, but not EC, inhibited the oscillation of intracellular Ca(2+) concentration induced by 11.1 mM glucose. These results suggest that some catechins at supraphysiological concentrations have inhibitory effects on GSIS, the potency of which depends on their structure; the order of potency was EGCG>GCG>EGC>GC≈ECG. The inhibitory effects seem to be mediated by the inhibition of voltage-dependent Ca(2+) channels, which is caused, at least in part, by membrane hyperpolarization resulting from the activation of K(+) channels.


Assuntos
Catequina/farmacologia , Glucose/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Insulina/metabolismo , Chá/química , Cálcio/metabolismo , Canais de Cálcio/metabolismo , Catequina/análogos & derivados , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Relação Estrutura-Atividade
18.
J Pharmacol Sci ; 123(4): 295-300, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24285083

RESUMO

An involvement of inducible nitric oxide (NO) synthase (NOS) in pancreatic ß-cell degeneration during the process of type 1 diabetes has been well discussed. Recently, there is growing evidence for pivotal roles of constitutive NOS (cNOS) in ß-cells; the presence of NOS1 and NOS3 in pancreatic ß-cells and the effects of low-concentration NO, which is assumed to be derived from cNOS, on ß-cell functions have been reported. However, the roles of cNOS-derived NO in ß-cells are still under debate. One of the reasons seems to be that NO has multiple biological activities, which are dependent on its concentration. In ß-cells, NO has been shown to exert positive and negative regulation of insulin secretion and anti- and pro-apoptotic activities, which is likely to be dependent on concentrations. In this review article, we will describe the current understanding of the roles of NO in pancreatic ß-cells, especially focusing on cNOS-derived NO and its differential roles depending on concentrations.


Assuntos
Diabetes Mellitus Tipo 1/patologia , Células Secretoras de Insulina/patologia , Células Secretoras de Insulina/fisiologia , Óxido Nítrico Sintase/fisiologia , Óxido Nítrico/metabolismo , Óxido Nítrico/fisiologia , Apoptose , Linhagem Celular , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Humanos , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/enzimologia , Células Secretoras de Insulina/metabolismo
19.
Endocrinology ; 154(11): 4089-98, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24035999

RESUMO

Diacylglycerol kinase (DGK) catalyzes the conversion of diacylglycerol (DAG) to phosphatidic acid. This study investigated the expression and function of DGK in pancreatic ß-cells. mRNA expression of type I DGK isoforms (α, ß, γ) was detected in mouse pancreatic islets and the ß-cell line MIN6. Protein expression of DGKα and DGKγ was also detected in mouse ß-cells and MIN6 cells. The type I DGK inhibitor R59949 inhibited high K(+)- and glucose-induced insulin secretion in MIN6 cells. Moreover, single knockdown of DGKα or DGKγ by small interfering RNA slightly but significantly decreased glucose- and high K(+)-induced insulin secretions, and the double knockdown further decreased them to the levels comparable with those induced by R59949. R59949 and DiC8, a membrane permeable DAG analog, decreased intracellular Ca(2+) concentration elevated by glucose and high K(+) in MIN6 cells. Real-time imaging in MIN6 cells expressing green fluorescent protein-tagged DGKα or DGKγ showed that the DGK activator phorbol 12-myristate 13-acetate rapidly induced translocation of DGKγ to the plasma membrane, whereas high K(+) slowly translocated DGKα and DGKγ to the plasma membrane. R59949 increased the DAG content in MIN6 cells when stimulated with high KCl, whereas it did not increase the DAG content but decreased the phosphatidic acid content when stimulated with high glucose. Finally, R59949 was confirmed to inhibit high K(+)-induced insulin secretion from mouse islets and glucose-induced insulin secretion from rat islets. These results suggest that DGKα and DGKγ are present in ß-cells and that the depression of these DGKs causes a decrease in intracellular Ca(2+) concentration, thereby reducing insulin secretion.


Assuntos
Diacilglicerol Quinase/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Animais , Cálcio/metabolismo , Linhagem Celular Tumoral , Diacilglicerol Quinase/genética , Secreção de Insulina , Masculino , Camundongos , Piperidinas/farmacologia , Isoformas de Proteínas , Quinazolinonas/farmacologia , Ratos , Ratos Wistar
20.
Biol Pharm Bull ; 34(7): 954-8, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21719997

RESUMO

Reactive oxygen species, including hydrogen peroxide (H(2)O(2)), are known to induce ß-cell apoptosis. The present study investigated the role of Ca(2+) in H(2)O(2)-induced apoptosis of the ß-cell line INS-1. Annexin V assay with flow cytometry and DNA ladder assay demonstrated that treatment of INS-1 cells with 100 µM H(2)O(2) for 18 h significantly increased apoptotic cells. A comparable level of apoptosis was also observed after 18 h when the cells were treated with 100 µM H(2)O(2) only for initial 30 min. The H(2)O(2)-induced apoptosis was abolished by 1,2-bis(o-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid tetra(acetoxymethyl)ester (BAPTA/AM), a chelator of intracellular Ca(2+), by 2-aminoethoxydiphenylborate (2-APB), a blocker of inositol 1,4,5-trisphosphate (IP(3)) receptors and cation channels, and by xestospongin D, a blocker of IP(3) receptors, and was partially blocked by SKF-96365, a non-selective cation channel blocker. However, nicardipine, an L-type voltage-dependent Ca(2+) channel blocker, or N-(p-amylcinnamoyl)anthranilic acid (ACA), a TRPM2 blocker, had little effect on the apoptosis. The inhibitory effect of BAPTA/AM or 2-APB on the H(2)O(2)-induced apoptosis was largely attenuated when the drug was added 30 min or 1 h after start of the treatment with H(2)O(2). These results suggest that the initial intracellular Ca(2+) elevation induced by H(2)O(2), which is mediated via IP(3) receptors and store-operated cation channels, plays an obligatory role in the induction of ß-cell apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Cálcio/metabolismo , Peróxido de Hidrogênio/farmacologia , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Ilhotas Pancreáticas/metabolismo , Transdução de Sinais , Linhagem Celular , Humanos , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...