Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Biogerontology ; 25(5): 819-836, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38748336

RESUMEN

An over-activation of the mechanistic target of rapamycin (mTOR) pathway promotes senescence and age-related diseases like type 2 diabetes. Besides, the regenerative potential of pancreatic islets deteriorates with aging. Nevertheless, the role of mTOR on senescence promoted by metabolic stress in islet cells as well as its relevance for electrophysiological aspects is not yet known. Here, we investigated whether parameters suggested to be indicative for senescence are induced in vitro in mouse islet cells by glucotoxicity and if mTOR inhibition plays a protective role against this. Islet cells exhibit a significant increase (~ 76%) in senescence-associated beta-galactosidase (SA-beta-gal) activity after exposure to glucotoxicity for 72 h. Glucotoxicity does not markedly influence p16INK4a protein within 72 h, but p16INK4a levels increase significantly after a 7-days incubation period. mTOR inhibition with a low rapamycin concentration (1 nM) entirely prevents the glucotoxicity-mediated increase of SA-beta-gal and p16INK4a. At the functional level, reactive oxygen species, calcium homeostasis, and electrical activity are disturbed by glucotoxicity, and rapamycin fails to prevent this. In contrast, rapamycin significantly attenuates the insulin hypersecretion promoted by glucotoxicity by modifying the mRNA levels of Vamp2 and Snap25 genes, related to insulin exocytosis. Our data indicate an influence of glucotoxicity on pancreatic islet-cell senescence and a reduction of the senescence markers by mTOR inhibition, which is relevant to preserve the regenerative potential of the islets. Decreasing the influence of mTOR on islet cells exposed to glucotoxicity attenuates insulin hypersecretion, but is not sufficient to prevent electrophysiological disturbances, indicating the involvement of mTOR-independent mechanisms.


Asunto(s)
Senescencia Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Islotes Pancreáticos , Serina-Treonina Quinasas TOR , Animales , Serina-Treonina Quinasas TOR/metabolismo , Ratones , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Senescencia Celular/efectos de los fármacos , Insulina/metabolismo , Sirolimus/farmacología , beta-Galactosidasa/metabolismo , Secreción de Insulina/efectos de los fármacos , Glucosa/metabolismo , Masculino , Ratones Endogámicos C57BL , Células Cultivadas , Especies Reactivas de Oxígeno/metabolismo
2.
Gen Comp Endocrinol ; 326: 114068, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-35671834

RESUMEN

BACKGROUND: Reports in recent years have shown that pancreatic ß-cell pyroptosis represents a critical mechanism involved with the progressive failure of pancreatic function. Previous research from our laboratory has indicated that artemether can increase the number of cells in pancreatic islets of db/db mice. In this study, we further examined whether artesunate (ART) protects pancreatic ß-cells from the damage of streptozotocin (STZ) by inhibiting pyroptosis. MATERIALS AND METHODS: In vitro, MIN6 cells exposed to 1 mM STZ were treated with ART (0.8 or 1.6 µM). The effects of ART on STZ-treated cells were evaluated through CCK-8 assay, flow cytometry and western blot, and further compared the effects of ART with the NLRP3 inhibitor, Mcc950 upon pyroptosis pathway proteins using western blot. In vivo, Male C57 mice were administered with a single intraperitoneal injection of STZ, and those with confirmed diabetes mellitus were given ART (0.5 or 1.0 mg/ml in drinking water) for 18 days. The effects of ART on STZ-induced diabetes were assessed by the observation of the general situation, glucose tolerance test, hematoxylin-eosin (HE) staining and immunohistochemistry. RESULTS: In MIN6 cells treated with STZ, we found that ART increased cell viability, decreased the number of late apoptotic cells (including pyroptosis cells) and inhibited the expression of proteins associated with the pyroptosis pathway. In STZ-induced animal model, the administration of ART reduced blood glucose levels, improved the consumption status within this diabetic mouse model and inhibited the expression of proteins include in the pyroptosis pathway in mice pancreats. CONCLUSIONS: Inhibition of pyroptosis may be a critical mechanism through which artesunate exerts protective effects upon pancreatic ß cells.


Asunto(s)
Artesunato , Diabetes Mellitus Experimental , Células Secretoras de Insulina , Animales , Artesunato/efectos adversos , Artesunato/farmacología , Caspasa 1/metabolismo , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Masculino , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteínas de Unión a Fosfato/metabolismo , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Estreptozocina
3.
J Endocrinol Invest ; 44(9): 1897-1904, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33486704

RESUMEN

BACKGROUND: High-density lipoprotein (HDL) is considered a complex plasma-circulating particle with subfractions that vary in function, size, and chemical composition. We sought to test the effects of HDL, and HDL subfractions on insulin secretion and cholesterol efflux in the ß-cell line MIN-6. METHODS: We used total HDL and HDL subfractions 2a, 2b, 3a, 3b, and 3c, isolated from human plasma, to test insulin secretion under different glucose concentrations as well as insulin content and cholesterol efflux in the insulinoma MIN-6 cell line. RESULTS: Incubation of MIN-6 cells with low glucose and total HDL increased insulin release two-fold. Meanwhile, when high glucose and HDL were used, insulin release increased more than five times. HDL subfractions 2a, 2b, 3a, 3b, and 3c elicited higher insulin secretion and cholesterol efflux than their respective controls, at both low and high glucose concentrations. The insulin content of the MIN-6 cells incubated with low glucose and any of the five HDL subclasses had a modest reduction compared with their controls. However, there were no statistically significant differences between each HDL subfraction on their capacity of eliciting insulin secretion, insulin content, or cholesterol efflux. CONCLUSIONS: HDL can trigger insulin secretion under low, normal, and high glucose conditions. We found that all HDL subfractions exhibit very similar capacity to increase insulin secretion and cholesterol efflux. This is the first report demonstrating that HDL subfractions act both as insulin secretagogues (under low glucose) and insulin secretion enhancers (under high glucose) in the MIN-6 cell line.


Asunto(s)
Colesterol/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Lipoproteínas HDL/sangre , Adulto , Animales , Línea Celular Tumoral , Femenino , Glucosa/farmacología , Humanos , Masculino , Ratones , Persona de Mediana Edad
4.
Acta Biochim Biophys Sin (Shanghai) ; 53(1): 46-53, 2021 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-33242093

RESUMEN

Endoplasmic reticulum (ER) stress plays a critical role in pancreatic ß cell destruction which leads to the pathogenesis of type 1 diabetes mellitus (T1DM). Vitamin D (VD) has been reported to reduce the risk of T1DM; however, it remains unknown whether VD affects ER stress in pancreatic ß cells. In this study, we investigated the role of the active form of VD, 1,25-dihydroxyvitamin D3 [1,25-(OH)2D3], in ER stress-induced ß cell apoptosis and explored its potential mechanism in mouse insulinoma cell line mouse insulinoma 6 (MIN6). The results of cell counting kit-8 (CCK8) and flow cytometric analyses showed that 1,25-(OH)2D3 caused a significant increase in the viability of MIN6 cells injured by H2O2. The protein kinase like ER kinase (PERK) signal pathway, one of the most conserved branches of ER stress, was found to be involved in this process. H2O2 activated the phosphorylation of PERK, upregulated the activating transcription factor 4 (ATF4) and C/EBP homologous protein (CHOP) expression, and subsequently initiated cell apoptosis, which were significantly reversed by 1,25-(OH)2D3 pretreatment. In addition, GSK2606414, a specific inhibitor of PERK, suppressed PERK phosphorylation and reduced the expressions of ATF4 and CHOP, leading to a significant decrease in ß cell apoptosis induced by H2O2. Taken together, the present findings firstly demonstrated that 1,25-(OH)2D3 could prevent MIN6 cells against ER stress-associated apoptosis by inhibiting the PERK-ATF4-CHOP pathway. Therefore, our results suggested that 1,25-(OH)2D3 might serve as a potential therapeutic target for preventing pancreatic ß cell destruction in T1DM.


Asunto(s)
Factor de Transcripción Activador 4/antagonistas & inhibidores , Calcitriol/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Sustancias Protectoras/farmacología , Transducción de Señal/efectos de los fármacos , Factor de Transcripción CHOP/antagonistas & inhibidores , eIF-2 Quinasa/antagonistas & inhibidores , Adenina/análogos & derivados , Adenina/farmacología , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Peróxido de Hidrógeno/toxicidad , Indoles/farmacología , Células Secretoras de Insulina/citología , Ratones
5.
J Cell Biochem ; 121(1): 840-855, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31452250

RESUMEN

Pancreatic stellate cells (PSCs) secrete various factors, which can influence the ß-cell function. The identification of stellate cell infiltration into the islets in pancreatic diseases suggests possible existence of cross-talk between these cells. To elucidate the influence of PSCs on ß-cell function, mouse PSCs were cocultured with Min6 cells using the Transwell inserts. Glucose-stimulated insulin secretion from Min6 cells in response to PSCs was quantified by enzyme-linked immunosorbent assay and insulin gene expression was measured by quantitative polymerase chain reaction. Upon cytometric identification of IL6 in PSC culture supernatants, Min6 cells were cultured with IL6 to assess its influence on the insulin secretion and gene expression. PLC-IP3 pathway inhibitors were added in the cocultures, to determine the influence of PSC-secreted IL6 on Glucose-stimulated insulin secretion from Min6 cells. Increased insulin secretion with a concomitant decrease in total insulin content was noticed in PSC-cocultured Min6 cells. Although increased GSIS was noted from IL6-treated Min6 cells, no change in the total insulin content was noted. Coculture of Min6 cells with PSCs or their exposure to IL6 did not alter either the expression of ß-cell-specific genes or that of miRNA-375. PSC-cocultured Min6 cells, in the presence of PLC-IP3 pathway inhibitors (U73122, Neomycin, and Xestospongin C), did not revoke the observed increase in GSIS. In conclusion, the obtained results indicate that augmented insulin secretion from Min6 cells in response to PSC secretions is independent of IL6-mediated PLC-IP3 pathway.


Asunto(s)
Glucosa/farmacología , Secreción de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Insulinoma/metabolismo , Interleucina-6/metabolismo , Células Estrelladas Pancreáticas/metabolismo , Animales , Células Cultivadas , Técnicas de Cocultivo , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/efectos de los fármacos , Insulinoma/patología , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Células Estrelladas Pancreáticas/citología , Células Estrelladas Pancreáticas/efectos de los fármacos , Edulcorantes/farmacología
6.
J Cell Physiol ; 234(11): 19852-19865, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30963563

RESUMEN

Wnt proteins act mainly as paracrine signals regulating cell proliferation and differentiation. The canonical Wnt pathway has recently been associated with pancreas development and the onset of type 2 diabetes in rodent and human but the underlying mechanisms are still unclear. The aim of this work was threefold: (a) to screen for Wnt expressed by murine pancreas/islet cells, (b) to investigate whether the Wnt gene expression profile can be changed in hyperplastic islets from type 2 prediabetic mice (fed a high-fat diet), and (c) to verify whether soluble factors (namely Wnts) released by pancreatic islets affect insulin secretion and proliferation of a beta-cell line in vitro condition. The majority of the Wnt subtypes are expressed by islet cells, such as Wnts 2, 2b, 3, 3a, 4, 5a, 5b, 6, 7a, 7b, 8a, 8b, 9a, 9b, and 11, while in the whole pancreas homogenates were found the same subtypes, except Wnts 3, 6, 7a, and 7b. Among all the Wnts, the Wnts 3a and 5b showed a significantly increased gene expression in hyperplastic islets from prediabetic mice compared with those from control mice. Furthermore, we observed that coculture with hyperplastic or nonhyperplastic islets did not change the secretory function of the mouse insulinoma clone 6 (MIN6) beta cells but induced a significant increase in cell proliferation in this lineage, which was partially blocked by the IWR-1 and IWP-2 Wnt inhibitors. In conclusion, we demonstrated that murine pancreas/islet cells can secrete Wnts, and that islet-released Wnts may participate in the regulation of beta-cell mass under normal and prediabetic conditions.


Asunto(s)
Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Proteínas Wnt/metabolismo , Tejido Adiposo/metabolismo , Animales , Línea Celular , Proliferación Celular , Dieta Alta en Grasa , Embrión de Mamíferos/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Secreción de Insulina , Masculino , Ratones Endogámicos C57BL , Estado Prediabético/genética , Estado Prediabético/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Wnt/genética , Vía de Señalización Wnt
7.
Int J Mol Sci ; 20(21)2019 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-31690008

RESUMEN

The zinc transporter ZnT8 (SLC30A8) localises to insulin secretory granules of ß-cells where it facilitates zinc uptake for insulin crystallisation. ZnT8 abundance has been linked to ß-cell survival and functional phenotype. However, the consequences of ZnT8 haploinsufficiency for ß-cell zinc trafficking and function remain unclear. Since investigations in human populations have shown SLC30A8 truncating polymorphisms to decrease the risk of developing Type 2 Diabetes, we hypothesised that ZnT8 haploinsufficiency would improve ß-cell function and maintain the endocrine phenotype. We used CRISPR/Cas9 technology to generate ZnT8 haploinsufficient mouse MIN6 ß-cells and showed that ZnT8 haploinsufficiency is associated with downregulation of mRNAs for Slc39a8 and Slc39a14, which encode for the zinc importers, Znt- and Irt-related proteins 8 (ZIP8) and 14 (ZIP14), and with lowered total cellular zinc content. ZnT8 haploinsufficiency disrupts expression of a distinct array of important ß-cell markers, decreases cellular proliferation via mitogen-activated protein (MAP) kinase cascades and downregulates insulin gene expression. Thus, ZnT8 cooperates with zinc importers of the ZIP family to maintain ß-cell zinc homeostasis. In contrast to the hypothesis, lowered ZnT8 expression reduces MIN6 cell survival by affecting zinc-dependent transcription factors that control the ß-cell phenotype.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Transportador 8 de Zinc/genética , Zinc/metabolismo , Animales , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Línea Celular , Regulación hacia Abajo , Haploinsuficiencia , Sistema de Señalización de MAP Quinasas , Ratones , Fenotipo
8.
Cell Physiol Biochem ; 51(5): 2185-2197, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30537728

RESUMEN

BACKGROUND/AIMS: VCP-interacting membrane selenoprotein (VIMP), an ER resident selenoprotein, is highly expressed in ß-cells, however, the role of VIMP in ß-cells has not been characterized. In this study, we studied the relationship between VIMP deficiency and ß-cell survival in MIN6 insulinoma cells. METHODS: To determine the role of VIMP in ß-cells, lentiviral VIMP shRNAs were used to knock down (KD) expression of VIMP in MIN6 cells. Cell death was quantified by propidium iodide (PI) staining followed by flow cytometric analyses using a FACS Caliber and FlowJo software. Cell apoptosis and proliferation were determined by TUNEL assay and Ki67 staining, respectively. Cell cycle was analyzed after PI staining. RESULTS: The results show that 1) VIMP suppression induces ß-cell apoptosis, which is associated with a decrease in Bcl-xL, and the ß-cell apoptosis induced by VIMP suppression can be inhibited by overexpression of Bcl-xL; 2) VIMP knockdown (KD) decreases cell proliferation and G1 cell cycle arrest by accumulating p27 and decreasing E2F1; 3) VIMP KD suppresses unfolded protein response (UPR) activation by regulating the IRE1α and PERK pathways; 4) VIMP KD increases insulin secretion. CONCLUSION: These results suggest that VIMP may function as a novel regulator to modulate ß-cell survival, proliferation, cell cycle, UPR and insulin secretion in MIN6 cells.


Asunto(s)
Regulación hacia Abajo , Puntos de Control de la Fase G1 del Ciclo Celular , Regulación Neoplásica de la Expresión Génica , Células Secretoras de Insulina/patología , Insulinoma/genética , Proteínas de la Membrana/genética , Neoplasias Pancreáticas/genética , Selenoproteínas/genética , Animales , Apoptosis , Muerte Celular , Línea Celular Tumoral , Técnicas de Silenciamiento del Gen , Células Secretoras de Insulina/metabolismo , Insulinoma/patología , Ratones , Neoplasias Pancreáticas/patología , Respuesta de Proteína Desplegada
9.
Histochem Cell Biol ; 150(4): 395-401, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30145684

RESUMEN

Valproate (VPA), an FDA approved anti-epileptic drug with a half-life of 12-18 h in humans, has been shown to perturb the vacuolar proton pump (vH+-ATPase) function in yeasts by inhibiting myo-inositol phosphate synthase, the first and rate-limiting enzyme in inositol biosynthesis, thereby resulting in inositol depletion. vH+-ATPase transfers protons (H+) across cell membranes, which help maintain pH gradients within cells necessary for various cellular functions including secretion. This proton pump has a membrane (V0) and a soluble cytosolic (V1) domain, with C-subunit associated with V1. In secretory cells such as neurons and insulin-secreting beta cells, vH+-ATPase acidifies vesicles essential for secretion. In this study, we demonstrate that exposure of insulin-secreting Min6 cells to a clinical dose of VPA results in inositol depletion and loss of co-localization of subunit C of vH+-ATPase with insulin-secreting granules. Consequently, a reduction of glucose-stimulated insulin secretion is observed following VPA exposure. These results merit caution and the reassessment of the clinical use of VPA.


Asunto(s)
Glucosa/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Ácido Valproico/farmacología , Animales , Secreción de Insulina , Ratones , Células Tumorales Cultivadas , Ácido Valproico/química
10.
Traffic ; 16(5): 493-509, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25615411

RESUMEN

The pre-exocytotic behavior of insulin granules was studied against the background of the entirety of submembrane granules in MIN6 cells, and the characteristics were compared with the macroscopic secretion pattern and the cytosolic Ca(2+) concentration of MIN6 pseudo-islets at 22°C, 32°C and 37°C. The mobility of granules labeled by insulin-EGFP and the fusion events were assessed by TIRF microscopy utilizing an observer-independent algorithm. In the z-dimension, 40 mm K(+) or 30 mm glucose increased the granule turnover. The effect of high K(+) was quickly reversible. The increase by glucose was more sustained and modified the efficacy of a subsequent K(+) stimulus. The effect size of glucose increased with physiological temperature whereas that of high K(+) did not. The mobility in the x/y-dimension and the fusion rates were little affected by the stimuli, in contrast to secretion. Fusion and secretion, however, had the same temperature dependence. Granules that appeared and fused within one image sequence had significantly larger caging diameters than pre-existent granules that underwent fusion. These in turn had a different mobility than residence-matched non-fusing granules. In conclusion, delivery to the membrane, tethering and fusion of granules are differently affected by insulinotropic stimuli. Fusion rates and secretion do not appear to be tightly coupled.


Asunto(s)
Membrana Celular/metabolismo , Exocitosis , Células Secretoras de Insulina/efectos de los fármacos , Insulina/metabolismo , Fusión de Membrana/efectos de los fármacos , Vesículas Secretoras/metabolismo , Animales , Calcio/metabolismo , Línea Celular , Membrana Celular/efectos de los fármacos , Citosol/metabolismo , Exocitosis/efectos de los fármacos , Glucosa/farmacología , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Ratones , Microscopía Fluorescente , Cloruro de Potasio/farmacología , Vesículas Secretoras/efectos de los fármacos
11.
Amino Acids ; 49(8): 1401-1414, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28624907

RESUMEN

Oxidized tyrosine products are commonly found in food with high protein content and have been demonstrated to cause damage of liver and kidney in our previous studies. Dityrosine (Dityr) is a typical oxidized tyrosine product. Due to its structural homology with thyroid hormones T3, we assumed that one of the endocrine systems most likely considered in connection with its disruption by Dityr may be the T3 action. T3 plays important roles in insulin synthesis, and thyroid hormone resistance (RTH) is associated with the impairment of glucose metabolism. Therefore, this study determined whether Dityr exposure impaired T3 function in pancreas leading to glucose metabolism disruption. After 10-week gavage with Dityr, mice exhibited impaired glucose tolerance and disturbed energy metabolism. The elevated free THs content in plasma, the up-regulation of THs synthesis-specific genes expressions in thyroid glands, and the increased thyroid follicles histology shapes and areas indicated that Dityr enhanced the THs synthesis in thyroid glands. In addition, Dityr-induced RTH, which reflected as elevated plasma free THs in the presence of unsuppressed thyroid stimulating hormone. The mRNA downregulation of membrane transporter of T3 (MCT8) and co-activator factors (RXRα, Src-1), together with the decreased protein level of thyroid hormone receptor ß1 (TRß1) in pancreas illustrated that the activation ability of T3 to downstream gene involved in insulin synthesis was suppressed by Dityr. In MIN-6 cell experiment, T3 improved glucose-stimulated insulin secretion by upregulating mRNA levels of insulin synthesis-related genes (Ins2, MafA, Pdx1) and T3 action-related genes, as well as increasing protein level of TRß1. These data suggest that Dityr suppress T3-regulated insulin synthesis stimulated by glucose via an indirect way of decreasing sensibility to T3 in pancreas. All these findings indicate that Dityr can disrupt THs function in pancreas leading to glucose metabolism disorder.


Asunto(s)
Células Secretoras de Insulina/efectos de los fármacos , Insulina/metabolismo , Triyodotironina/metabolismo , Tirosina/análogos & derivados , Animales , Células Cultivadas , Metabolismo Energético/efectos de los fármacos , Secreción de Insulina , Masculino , Ratones , Ratones Endogámicos C57BL , Tirosina/administración & dosificación
12.
Clin Exp Pharmacol Physiol ; 44(12): 1254-1262, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28699234

RESUMEN

Morin is a flavonoid contained in guava that is known to reduce hyperglycemia in diabetes. Insulin secretion has been demonstrated to increase following the administration of morin. The present study is designed to investigate the potential mechanism(s) of morin-induced insulin secretion in the MIN6 cell line. First, we identified that morin induced a dose-dependent increase in insulin secretion and intracellular calcium content in MIN6 cells. Morin potentiated glucose-stimulated insulin secretion (GSIS). Additionally, we used siRNA for the ablation of imidazoline receptor protein (NISCH) expression in MIN6 cells. Interestingly, the effects of increased insulin secretion by morin and canavanine were markedly reduced in Si-NISCH cells. Moreover, we used KU14R to block imidazoline I3 receptor (I-3R) that is known to enhance insulin release from the pancreatic ß-cells. Without influence on the basal insulin secretion, KU14R dose-dependently inhibited the increased insulin secretion induced by morin or efaroxan in MIN6 cells. Additionally, effects of increased insulin secretion by morin or efaroxan were reduced by diazoxide at the dose sufficient to open KATP channels and attenuated by nifedipine at the dose used to inhibit L-type calcium channels. Otherwise, phospholipase C (PLC) is introduced to couple with imidazoline receptor (I-R). The PLC inhibitor dose-dependently inhibited the effects of morin in MIN6 cells. Similar blockade was also observed in protein kinase C (PKC) inhibitor-treated cells. Taken together, we found that morin increases insulin secretion via the activation of I-R in pancreatic cells. Therefore, morin would be useful to develop in the research and treatment of diabetic disorders.


Asunto(s)
Flavonoides/farmacología , Receptores de Imidazolina/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Insulina/metabolismo , Animales , Técnicas de Cultivo de Célula , Línea Celular , Relación Dosis-Respuesta a Droga , Silenciador del Gen , Glucosa/farmacología , Receptores de Imidazolina/genética , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Ratones
13.
Biochim Biophys Acta ; 1841(9): 1217-26, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24814049

RESUMEN

BACKGROUNDS: High-density lipoprotein (HDL) has been proposed to enhance ß-cell functions. Clinical studies have suggested that apolipoprotein M (apoM), which rides mainly on HDL, is involved in diabetes; however, the underlying mechanism has not yet been elucidated. Recently, apoM was shown to be a carrier for sphingosine 1-phosphate (S1P), a bioactive lipid mediator. In the present study, we investigated the modulation of insulin secretion by apoM through the action of S1P. METHODS AND RESULTS: We overexpressed apoM in the livers of C57BL6 mice using adenovirus gene transfer and found that the blood glucose levels under ad libitum feeding conditions were lower in the apoM-overexpressing mice. While an insulin tolerance test revealed that insulin sensitivity was not significantly affected, a glucose tolerance test revealed that apoM-overexpressing mice had a better glucose tolerance because of enhanced insulin secretion, a phenomenon that was reversed by treatment with VPC 23019, an antagonist against S1P1 and S1P3 receptor. In vitro experiments with MIN6 cells also revealed that apoM-containing lipoproteins enhanced insulin secretion, which was again inhibited by VPC 23019. ApoM retarded the degradation of S1P, and an increase in Pdx1 expression, the attenuation of endoreticulum stress, and the phosphorylation of Akt, AmpK, and Erk were observed as possible underlying mechanisms for the effect of S1P, maintained at a high concentration by apoM, on the increase in insulin secretion. CONCLUSIONS: ApoM augmented insulin secretion by maintaining the S1P concentration under both in vivo and in vitro conditions.


Asunto(s)
Apolipoproteínas/genética , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Lisofosfolípidos/metabolismo , Esfingosina/análogos & derivados , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Adenoviridae/genética , Animales , Apolipoproteínas/metabolismo , Apolipoproteínas M , Transporte Biológico , Glucemia/metabolismo , Línea Celular , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación de la Expresión Génica , Vectores Genéticos , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Insulina/biosíntesis , Secreción de Insulina , Células Secretoras de Insulina/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosforilación , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Lisoesfingolípidos/genética , Receptores de Lisoesfingolípidos/metabolismo , Transducción de Señal , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato , Transactivadores/genética , Transactivadores/metabolismo
14.
Growth Factors ; 33(1): 1-7, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25370781

RESUMEN

We have reported earlier that murine-regenerating gene mReg2 protects MIN6 mouse insulinoma cells from ER stress and caspase-mediated apoptosis. In apoptotic cells, DNA damage is induced by the nuclear translocation of mitochondrial apoptosis-inducing factor (AIF). Here we tested the hypothesis that mReg2 may regulate Scythe and/or hsp70 which influence the nuclear import of AIF. Treatment with thapsigargin (Tg) or doxorubicin induced an increase in nuclear AIF in MIN6 cells carrying the empty transfection vector (MIN6-VC) but not in cells overexpressing mReg2 (MIN6-mReg2). On one hand, nuclear Scythe was higher in the nucleus of MIN6-mReg2 compared with that in MIN6-VC cells. mReg2 did not alter the expression of AIF or Scythe. On the other hand, mReg2 induced the expression of hsp70 which is known to promote cytosolic retention of AIF. We conclude that mReg2 inhibits AIF-mediated apoptosis by promoting the nuclear presence of Scythe and inducing hsp70.


Asunto(s)
Factor Inductor de la Apoptosis/metabolismo , Núcleo Celular/metabolismo , Células Secretoras de Insulina/metabolismo , Proteínas/metabolismo , Transporte Activo de Núcleo Celular , Animales , Línea Celular Tumoral , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Ratones , Chaperonas Moleculares/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Asociadas a Pancreatitis , Proteínas/genética
15.
Biochem Biophys Res Commun ; 468(4): 752-7, 2015 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-26585489

RESUMEN

Regulation of ATP-sensitive inwardly rectifying potassium (KATP) channel plays a critical role in metabolism-secretion coupling of pancreatic ß-cells. Released insulin from ß-cells inhibits insulin and glucagon secretion with autocrine and paracrine modes. However, molecular mechanism by which insulin inhibits hormone secretion remains elusive. Here, we investigated the effect of autocrine insulin on surface abundance of KATP channel in mouse clonal ß-cell line, MIN6. High glucose increased plasmalemmal sulfonylurea receptor 1 (SUR1), a component of KATP channel as well as exogenous insulin treatment. SUR1 trafficking by high glucose or insulin was blocked by inhibition of phosphoinositide 3-kinase (PI3K) with wortmannin. Pretreatment with brefeldin A or silencing of vesicle-associated membrane protein 2 (VAMP2) abolished insulin-mediated upregulation of surface SUR1. Functionally, glucose-stimulated cytosolic Ca(2+) ([Ca(2+)]i) increase was blunted by insulin or diazoxide, a KATP channel opener. Insulin-induced suppression of [Ca(2+)]i oscillation was prevented by an insulin receptor blocker. These results provide a novel molecular mechanism for autocrine negative feedback regulation of insulin secretion.


Asunto(s)
Comunicación Autocrina/fisiología , Calcio/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Proteína 2 de Membrana Asociada a Vesículas/metabolismo , Animales , Línea Celular , Membrana Celular/metabolismo , Secreción de Insulina , Canales KATP , Ratones , Potasio/metabolismo
16.
Phytother Res ; 29(10): 1672-5, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26178652

RESUMEN

Ethno-botanical inspired isolation from plant Scoparia dulcis Linn. (Sweet Broomweed) yielded six compounds, coixol (1), glutinol (2), glutinone (3), friedelin (4), betulinic acid (5), and tetratriacontan-1-ol (6). There structures were identified using mass and 1D- and 2D-NMR spectroscopy techniques. Compounds 1-6 were evaluated for their insulin secretory activity on isolated mice islets and MIN-6 pancreatic ß-cell line, and compounds 1 and 2 were found to be potent and mildly active, respectively. Compound 1 was further evaluated for insulin secretory activity on MIN-6 cells. Compound 1 was subjected to in vitro cytotoxicity assay against MIN-6, 3T3 cell lines, and islet cells, and in vivo acute toxicity test in mice that was found to be non-toxic. The insulin secretory activity of compounds 1 and 2 supported the ethno-botanic uses of S. dulcis as an anti-diabetic agent.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Hipoglucemiantes/farmacología , Extractos Vegetales/uso terapéutico , Scoparia , Células 3T3 , Animales , Insulina , Islotes Pancreáticos , Masculino , Ratones , Nepal , Ratas , Ratas Wistar
17.
Mol Nutr Food Res ; 68(1): e2200842, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37990402

RESUMEN

SCOPE: Consuming goat milk is known to benefit high-fat diet-fed and streptozocin (STZ)-induced diabetic rats, but the underlying mechanisms are unknown. This study is conducted to investigate the metabolic effects of a goat milk diet (a form of goat milk powder) on glucose homeostasis and pancreatic conditions in a mouse model of Type 2 diabetes mellitus (T2DM) induced by STZ. METHODS AND RESULTS: T2DM mice are fed with a goat-milk-based diet containing 10.3% w/w goat milk powder for 10 weeks for investigating the in vivo effects; a ß-cell line MIN6 cells are used to test the in vitro effects of digested goat milk (DGM). Goat milk diet improves the deleterious effects of STZ on fasting glucose levels and glucose tolerance, accelerates pancreatic structure recovery, and alters blood metabolites in mice. Based on the significant differences observed in metabolites, the key pathways, metabolite regulatory enzymes, metabolite molecular modules, and biochemical reactions are identified as critical integrated pathways. DGM promotes the cell activity, glucose transportation, and AKT activation in cultured STZ-treated MIN6 cells in vitro. CONCLUSIONS: Goat milk diet improves glucose homeostasis and pancreatic conditions of T2DM mice, in association with improved blood metabolite profiles and activation of pancreatic AKT pathway.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Ratones , Ratas , Animales , Diabetes Mellitus Tipo 2/metabolismo , Leche/química , Diabetes Mellitus Experimental/metabolismo , Proteínas Proto-Oncogénicas c-akt , Polvos , Glucosa/metabolismo , Dieta Alta en Grasa/efectos adversos , Cabras/metabolismo , Glucemia/metabolismo , Estreptozocina , Insulina
18.
Cells ; 13(12)2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38920672

RESUMEN

Recently, we successfully utilized noninvasive magnetic resonance and bioluminescence imaging to track MIN6 cells subcutaneously transplanted in immunocompromised nude mice for up to 64 days. In this study, we further used bioluminescence imaging to investigate the immune rejection of MIN6 cells in immunocompetent C3H mice. A total of 5 × 106 luciferase-transfected MIN6 cells were implanted into the subcutaneous space of each nude or C3H mouse. After transplantation, hypoglycemia and persistent bioluminescence signals were observed in eight of eight (100%) nude mice and five of nine (56%) C3H mice (p < 0.05). We then presensitized a group of C3H mice with C57BL/6 spleen cells just prior to transplantation (n = 14). Interestingly, none of them had hypoglycemia or persistent bioluminescence signals (p < 0.01 vs. C3H mice without presensitization). Histological examination of the grafts revealed a lack or minimal presence of insulin-positive cells in recipients without hypoglycemia and persistent bioluminescence signals. In contrast, recipients with hypoglycemia and persistent bioluminescence signals showed a significant presence of insulin-positive cells in their grafts. Our results indicate that rejection of MIN6 cells occurred in C3H mice and could be enhanced by presensitization with C57BL/6 spleen cells and that bioluminescence imaging is a useful noninvasive tool for detecting rejection of subcutaneously transplanted MIN6 cells.


Asunto(s)
Rechazo de Injerto , Mediciones Luminiscentes , Ratones Endogámicos C3H , Animales , Ratones , Rechazo de Injerto/inmunología , Mediciones Luminiscentes/métodos , Ratones Endogámicos C57BL , Ratones Desnudos , Línea Celular Tumoral , Bazo
19.
Ann Biomed Eng ; 52(9): 2610-2626, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38829457

RESUMEN

Interactions between cells are of fundamental importance in affecting cell function. In vivo, endothelial cells and islet cells are close to each other, which makes endothelial cells essential for islet cell development and maintenance of islet cell function. We used endothelial cells to construct 3D pseudo-islets, which demonstrated better glucose regulation and greater insulin secretion compared to conventional pseudo-islets in both in vivo and in vitro trials. However, the underlying mechanism of how endothelial cells promote beta cell function localized within islets is still unknown. We performed transcriptomic sequencing, differential gene analysis, and enrichment analysis on two types of pseudo-islets to show that endothelial cells can promote the function of internal beta cells in pseudo-islets through the BTC-EGFR-JAK/STAT signaling pathway. Min6 cells secreted additional BTC after co-culture of endothelial cells with MIN6 cells outside the body. After BTC knockout in vitro, we found that beta cells functioned differently: insulin secretion levels decreased significantly, while the expression of key proteins in the EGFR-mediated JAK/STAT signaling pathway simultaneously decreased, further confirming our results. Through our experiments, we elucidate the molecular mechanisms by which endothelial cells maintain islet function in vitro, which provides a theoretical basis for the construction of pseudo-islets and islet cell transplants for the treatment of diabetes mellitus.


Asunto(s)
Células Endoteliales , Transducción de Señal , Animales , Ratones , Línea Celular , Células Endoteliales/metabolismo , Receptores ErbB/metabolismo , Receptores ErbB/genética , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Quinasas Janus/metabolismo , Factores de Transcripción STAT/metabolismo
20.
Cells ; 13(11)2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38891081

RESUMEN

This study unveils verapamil's compelling cytoprotective and proliferative effects on pancreatic ß-cells amidst diabetic stressors, spotlighting its unforeseen role in augmenting cholecystokinin (CCK) expression. Through rigorous investigations employing MIN6 ß-cells and zebrafish models under type 1 and type 2 diabetic conditions, we demonstrate verapamil's capacity to significantly boost ß-cell proliferation, enhance glucose-stimulated insulin secretion, and fortify cellular resilience. A pivotal revelation of our research is verapamil's induction of CCK, a peptide hormone known for its role in nutrient digestion and insulin secretion, which signifies a novel pathway through which verapamil exerts its therapeutic effects. Furthermore, our mechanistic insights reveal that verapamil orchestrates a broad spectrum of gene and protein expressions pivotal for ß-cell survival and adaptation to immune-metabolic challenges. In vivo validation in a zebrafish larvae model confirms verapamil's efficacy in fostering ß-cell recovery post-metronidazole infliction. Collectively, our findings advocate for verapamil's reevaluation as a multifaceted agent in diabetes therapy, highlighting its novel function in CCK upregulation alongside enhancing ß-cell proliferation, glucose sensing, and oxidative respiration. This research enriches the therapeutic landscape, proposing verapamil not only as a cytoprotector but also as a promoter of ß-cell regeneration, thereby offering fresh avenues for diabetes management strategies aimed at preserving and augmenting ß-cell functionality.


Asunto(s)
Colecistoquinina , Células Secretoras de Insulina , Verapamilo , Pez Cebra , Animales , Ratones , Línea Celular , Proliferación Celular/efectos de los fármacos , Colecistoquinina/metabolismo , Colecistoquinina/farmacología , Modelos Animales de Enfermedad , Glucosa/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Regeneración/efectos de los fármacos , Verapamilo/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA