Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 564
Filtrar
Más filtros

Medicinas Complementárias
Tipo del documento
Intervalo de año de publicación
1.
Phytomedicine ; 128: 155396, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38547617

RESUMEN

BACKGROUND: Abnormalities in glucose metabolism may be the underlying cause of ß-cell dysfunction and identity impairment resulting from high glucose exposure. In China, Coptis deltoidea C. Y. Cheng et Hsiao (YL) has demonstrated remarkable hypoglycemic effects. HYPOTHESIS/PURPOSE: To investigate the hypoglycemic effect of YL and determine the mechanism of YL in treating diabetes. METHODS: A type 2 diabetes mouse model was used to investigate the pharmacodynamics of YL. YL was administrated once daily for 8 weeks. The hypoglycemic effect of YL was assessed by fasting blood glucose, an oral glucose tolerance test, insulin levels, and other indexes. The underlying mechanism of YL was examined by targeting glucose metabolomics, western blotting, and qRT-PCR. Subsequently, the binding capacity between predicted AMP-activated protein kinase (AMPK) and important components of YL (Cop, Ber, and Epi) were validated by molecular docking and surface plasmon resonance. Then, in AMPK knockdown MIN6 cells, the mechanisms of Cop, Ber, and Epi were inversely confirmed through evaluations encompassing glucose-stimulated insulin secretion, markers indicative of ß-cell identity, and the examination of glycolytic genes and products. RESULTS: YL (0.9 g/kg) treatment exerted notable hypoglycemic effects and protected the structural integrity and identity of pancreatic ß-cells. Metabolomic analysis revealed that YL inhibited the hyperactivated glycolysis pathway in diabetic mice, thereby regulating the products of the tricarboxylic acid cycle. KEGG enrichment revealed the intimate relationship of this process with the AMPK signaling pathway. Cop, Ber, and Epi in YL displayed high binding affinities for AMPK protein. These compounds played a pivotal role in preserving the identity of pancreatic ß-cells and amplifying insulin secretion. The mechanism underlying this process involved inhibition of glucose uptake, lowering intracellular lactate levels, and elevating acetyl coenzyme A and ATP levels through AMPK signaling. The use of a glycolytic inhibitor corroborated that attenuation of glycolysis restored ß-cell identity and function. CONCLUSION: YL demonstrates significant hypoglycemic efficacy. We elucidated the potential mechanisms underlying the protective effects of YL and its active constituents on ß-cell function and identity by observing glucose metabolism processes in pancreatic tissue and cells. In this intricate process, AMPK plays a pivotal regulatory role.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Coptis , Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Hipoglucemiantes , Células Secretoras de Insulina , Transducción de Señal , Animales , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Hipoglucemiantes/farmacología , Transducción de Señal/efectos de los fármacos , Ratones , Diabetes Mellitus Experimental/tratamiento farmacológico , Masculino , Coptis/química , Glucemia/efectos de los fármacos , Insulina/metabolismo , Ratones Endogámicos C57BL , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/química , Simulación del Acoplamiento Molecular , Prueba de Tolerancia a la Glucosa , Extractos Vegetales/farmacología
2.
J Endocrinol ; 261(1)2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38305305

RESUMEN

Metabolic syndrome (MetS) is an increasing global health threat and strong risk factor for type 2 diabetes (T2D). MetS causes both hyperinsulinemia and islet size overexpansion, and pancreatic ß-cell failure impacts insulin and proinsulin secretion, mitochondrial density, and cellular identity loss. The low-density lipoprotein receptor knockout (LDLr-/-) model combined with high-fat diet (HFD) has been used to study alterations in multiple organs, but little is known about the changes to ß-cell identity resulting from MetS. Osteocalcin (OC), an insulin-sensitizing protein secreted by bone, shows promising impact on ß-cell identity and function. LDLr-/- mice at 12 months were fed chow or HFD for 3 months ± 4.5 ng/h OC. Islets were examined by immunofluorescence for alterations in nuclear Nkx6.1 and PDX1 presence, insulin-glucagon colocalization, islet size and %ß-cell and islet area by insulin and synaptophysin, and mitochondria fluorescence intensity by Tomm20. Bone mineral density (BMD) and %fat changes were examined by Piximus Dexa scanning. HFD-fed mice showed fasting hyperglycemia by 15 months, increased weight gain, %fat, and fasting serum insulin and proinsulin; concurrent OC treatment mitigated weight increase and showed lower proinsulin-to-insulin ratio, and higher BMD. HFD increased %ß and %islet area, while simultaneous OC-treatment with HFD was comparable to chow-fed mice. Significant reductions in nuclear PDX1 and Nkx6.1 expression, increased insulin-glucagon colocalization, and reduction in ß-cell mitochondria fluorescence intensity were noted with HFD, but largely prevented with OC administration. OC supplementation here suggests a benefit to ß-cell identity in LDLr-/- mice and offers intriguing clinical implications for countering metabolic syndrome.


Asunto(s)
Diabetes Mellitus Tipo 2 , Hiperinsulinismo , Células Secretoras de Insulina , Islotes Pancreáticos , Síndrome Metabólico , Animales , Ratones , Diabetes Mellitus Tipo 2/metabolismo , Dieta Alta en Grasa/efectos adversos , Glucagón/metabolismo , Hiperinsulinismo/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Lipoproteínas LDL , Síndrome Metabólico/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Osteocalcina/metabolismo , Proinsulina/metabolismo , Aumento de Peso
3.
In Vitro Cell Dev Biol Anim ; 60(1): 23-35, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38117455

RESUMEN

It has been well established that the circulating taurine affects the insulin synthesis in pancreatic islet ß-cells, whereas miR-7a and LIM-homeodomain transcription factor Isl-1 are important intracellular factors regulating insulin transcription and synthesis. However, it still remains unknown whether taurine regulates insulin synthesis by affecting miR-7a and/or Isl-1 expressions in mouse pancreatic islet ß-cells. The present study was thus proposed to identify the effects of taurine on the expressions of miR-7a and/or Isl-1 and their relations to insulin synthesis in mouse pancreatic islet ß-cells by using miR-7a2 knockout (KO) and taurine transporter (TauT) KO mouse models and the related in vitro experiments. The results demonstrated that taurine supplement significantly decreased the pancreas miR-7a expression, but sharply upregulated the pancreas Isl-1 and insulin expressions, and serum insulin levels. However, the enhanced effects of taurine on Isl-1 expression and insulin synthesis were mitigated in the TauT KO and miR-7a2 KO mice. In addition, our results confirmed that taurine markedly increased pancreas RAF1 and ERK1/2 expressions. Collectively, the present study firstly demonstrates that taurine regulates insulin synthesis through TauT/miR-7a/RAF1/ERK1/2/Isl-1 signaling pathway, which are crucial for our understanding the mechanisms of taurine affecting insulin synthesis, and also potential for establishing the therapeutic strategies for diabetes and the diseases related to metabolism.


Asunto(s)
Células Secretoras de Insulina , MicroARNs , Animales , Ratones , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Sistema de Señalización de MAP Quinasas , Ratones Noqueados , MicroARNs/genética , MicroARNs/metabolismo , Taurina/farmacología , Taurina/metabolismo
4.
J Ethnopharmacol ; 315: 116564, 2023 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-37244407

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Fufang-Zhenzhu-Tiaozhi capsule (FTZ), a Traditional Chinese Medicine (TCM) patent prescription commonly used in clinical practice, has a significant curative effect on hyperglycemia and hyperlipidemia. Previous studies have shown that FTZ can treat diabetes, but the effect of FTZ on ß-cell regeneration needs to be further explored in T1DM mice. AIM OF THE STUDY: The aim is to investigate the role of FTZ in promoting ß-cell regeneration in T1DM mice, and to further explore its mechanism. MATERIALS AND METHODS: C57BL/6 mice were used as control. NOD/LtJ mice were divided into the Model group and FTZ group. Oral glucose tolerance, fasting blood glucose, and fasting insulin level were measured. Immunofluorescence staining was used to detect the level of ß-cell regeneration and the composition of α-cells and ß-cells in islets. Hematoxylin and eosin staining was used to detect the infiltration degree of inflammatory cells. The apoptosis of islet cells was detected by terminal deoxynucleotidyl transferase deoxyuridine triphosphate nick end labeling. Western blotting was used to detect the expression levels of Pancreas/duodenum homeobox protein 1 (PDX-1), V-maf musculoaponeurotic fibrosarcoma oncogene homolog A (MAFA), and Neurogenin-3 (NGN3). RESULTS: FTZ could increase insulin levels and reduce the glucose level of T1DM mice and promote ß-cell regeneration. FTZ also inhibited the invasion of inflammatory cells and the islet cell apoptosis, and maintained the normal composition of islet cells, thus preserving the quantity and quality of ß-cells. Furthermore, FTZ promoting ß-cell regeneration was accompanied by increasing the expression of PDX-1, MAFA, and NGN3. CONCLUSION: FTZ can restore the insulin-secreting function of the impaired pancreatic islet, improve blood glucose level, possibly via the enhancing ß cell regeneration via upregulation of PDX-1, MAFA, and NGN3 in T1DM mice, and may be a potential therapeutic drug for T1DM.


Asunto(s)
Diabetes Mellitus Tipo 1 , Células Secretoras de Insulina , Islotes Pancreáticos , Ratones , Animales , Diabetes Mellitus Tipo 1/metabolismo , Glucemia/metabolismo , Ratones Endogámicos NOD , Ratones Endogámicos C57BL , Islotes Pancreáticos/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina , Regeneración , Proliferación Celular
5.
J Trace Elem Med Biol ; 79: 127217, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37224745

RESUMEN

OBJECTIVES: Zinc, which is found in high concentrations in the ß-cells of the pancreas, is also a critical component for the endocrine functions of the pancreas. SLC30A8/ZnT8 is the carrier protein responsible for the transport of zinc from the cytoplasm to the insulin granules. The aim of this study was to investigate how dietary zinc status affects pancreatic beta cell activation and ZnT8 levels in infant male rats born to zinc-deficient mothers. METHODS: The study was performed on male pups born to mothers fed a zinc-deficient diet. A total of 40 male rats were divided into 4 equal groups. Group 1: In addition to maternal zinc deficiency, this group was fed a zinc-deficient diet. Group 2: In addition to maternal zinc deficiency, this group was fed a standard diet. Group 3: In addition to maternal zinc deficiency, this group was fed a standard diet and received additional zinc supplementation. Group 4: Control group. Pancreas ZnT8 levels were determined by ELISA method and insulin-positive cell ratios in ß-cells by immunohistochemistry. RESULTS: The highest pancreatic ZnT8 levels and anti-insulin positive cell ratios in the current study were obtained in Group 3 and Group 4. In our study, the lowest pancreatic ZnT8 levels were obtained in Group 1 and Group 2, and the lowest pancreatic anti-insulin positive cell ratios were obtained in Group 1. CONCLUSION: The results of the present study; in rats fed a zinc-deficient diet after maternal zinc deficiency has been established shows that ZnT8 levels and anti-insulin positive cell ratios in pancreatic tissue, which is significantly suppressed, reach control values with intraperitoneal zinc supplementation.


Asunto(s)
Proteínas de Transporte de Catión , Células Secretoras de Insulina , Islotes Pancreáticos , Ratas , Masculino , Animales , Células Secretoras de Insulina/metabolismo , Zinc/metabolismo , Proteínas de Transporte de Catión/metabolismo , Islotes Pancreáticos/metabolismo , Transportador 8 de Zinc/metabolismo , Insulina/metabolismo
6.
J Med Food ; 26(6): 357-367, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37083465

RESUMEN

We examined the effects of HM-chromanone (HMC) on alleviating hyperglycemia and protecting pancreatic ß-cells from streptozotocin (STZ)-induced damage in C57BL/6J mice. HMC was administered to STZ-induced diabetic mice at 10 or 30 mg/kg, for 14 days. Thereafter, changes in fasting blood glucose levels, insulin-secretion, histopathological examination of pancreas islet cell and apoptotic protein levels, and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay were determined. The results revealed that HMC dose-dependently improved blood glucose concentrations and alleviated pancreatic islet cells damage. In diabetic mice, degeneration of the islet cells was observed wherein they appeared shrunken, with hyaline deterioration, nuclear dissolution, and condensation. However, morphology of the islet cell was restored, and nuclei were visibly rounded in the HMC (30 mg/kg)-administered diabetic mice. In addition, ß-cell numbers were markedly increased in HMC mice compared to STZ-induced diabetic mice, and the number of cells stained with glucagon was decreased. HMC markedly decreased the expression of proapoptotic proteins and increased antiapoptotic proteins, and the number of apoptotic cells detected by TUNEL was elevated. HMC decreased expression of interleukin (IL)-1ß, IL-6, and tumor necrosis factor-α in diabetic mice. Moreover, HMC increased antioxidant-enzymes activity, and decreased reactive oxygen species generation. In conclusion, the results demonstrate the potential of HMC to alleviate hyperglycemia by protecting the pancreatic ß-cells in diabetic mice.


Asunto(s)
Diabetes Mellitus Experimental , Hiperglucemia , Células Secretoras de Insulina , Islotes Pancreáticos , Ratones , Animales , Estreptozocina/efectos adversos , Insulina , Glucemia/metabolismo , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Ratones Endogámicos C57BL , Islotes Pancreáticos/metabolismo , Hiperglucemia/tratamiento farmacológico , Hiperglucemia/metabolismo , Células Secretoras de Insulina/metabolismo , Antioxidantes/farmacología , Antioxidantes/metabolismo
7.
J Clin Invest ; 133(8)2023 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-37066881

RESUMEN

Insulin secretion by pancreatic ß cells is a dynamic and highly regulated process due to the central importance of insulin in enabling efficient utilization and storage of glucose. Multiple regulatory layers enable ß cells to adapt to acute changes in nutrient availability as well as chronic changes in metabolic demand. While epigenetic factors have been well established as regulators of chronic ß cell adaptations to insulin resistance, their role in acute adaptations in response to nutrient stimulation has been relatively unexplored. In this issue of the JCI, Wortham et al. report that short-term dynamic changes in histone modifications regulated insulin secretion and acute ß cell adaptations in response to fasting and feeding cycles. These findings highlight the importance of investigating whether other epigenetic mechanisms may contribute to acute physiologic adaptations in ß cells.


Asunto(s)
Resistencia a la Insulina , Células Secretoras de Insulina , Secreción de Insulina , Cebollas/metabolismo , Insulina/metabolismo , Glucosa/metabolismo , Resistencia a la Insulina/fisiología , Células Secretoras de Insulina/metabolismo
8.
J Integr Med ; 21(1): 99-105, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36481247

RESUMEN

OBJECTIVE: To investigate the effect of ferulic acid, a natural compound, on pancreatic beta cell viability, Ca2+ channels, and insulin secretion. METHODS: We studied the effects of ferulic acid on rat insulinoma cell line viability using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide viability assay. The whole-cell patch-clamp technique and enzyme-linked immunosorbent assay were also used to examine the action of ferulic acid on Ca2+ channels and insulin secretion, respectively. RESULTS: Ferulic acid did not affect cell viability during exposures up to 72 h. The electrophysiological study demonstrated that ferulic acid rapidly and concentration-dependently increased L-type Ca2+ channel current, shifting its activation curve in the hyperpolarizing direction with a decreased slope factor, while the voltage dependence of inactivation was not affected. On the other hand, ferulic acid have no effect on T-type Ca2+ channels. Furthermore, ferulic acid significantly increased insulin secretion, an effect inhibited by nifedipine and Ca2+-free extracellular fluid, confirming that ferulic acid-induced insulin secretion in these cells was mediated by augmenting Ca2+ influx through L-type Ca2+ channel. Our data also suggest that this may be a direct, nongenomic action. CONCLUSION: This is the first electrophysiological demonstration that acute ferulic acid treatment could increase L-type Ca2+ channel current in pancreatic ß cells by enhancing its voltage dependence of activation, leading to insulin secretion.


Asunto(s)
Células Secretoras de Insulina , Insulina , Ratas , Animales , Secreción de Insulina , Insulina/farmacología , Células Secretoras de Insulina/metabolismo , Ácidos Cumáricos/farmacología , Ácidos Cumáricos/metabolismo , Calcio/metabolismo
9.
Zhongguo Zhong Yao Za Zhi ; 47(16): 4403-4410, 2022 Aug.
Artículo en Chino | MEDLINE | ID: mdl-36046869

RESUMEN

The present study investigated the anti-oxidative and anti-apoptotic effects and molecular mechanisms of catalpol on the H_2O_2-induced pancreatic ß-cells(INS-1 cells).The oxidative damage model of INS-1 cells was induced and optimized by the stimulation of H_2O_2 of different concentrations for different time.CCK-8 assay was used to detect cell viability after catalpol intervention(1, 5, 10, 20, 40, 80, and 160 µmol·L~(-1)) for 24 h.Intracellular reactive oxygen species(ROS), superoxide dismutase(SOD), and lipid peroxide malondialdehyde(MDA) were measured by DCFH-DA fluorescent probe, WST-1, and TBA respectively.Moreover, the apo-ptotic effect was detected by AO-EB and Annexin V-FITC/PI staining.In addition, the protein expression levels were detected by Wes-tern blot, and intracellular insulin concentration was measured by ELISA.The results showed that the oxidative damage model of INS-1 cells was stably induced by 50 µmol·L~(-1) H_2O_2 treatment for 2 h, and catalpol at 1-80 µmol·L~(-1) did not affect cell viability of INS-1 cells.Compared with the conditions in the model group, 1, 5, and 10 µmol·L~(-1) catalpol intervention for 2 h could protect INS-1 cells from oxidative damage(P<0.001), reduce ROS and MDA, increase SOD, and inhibit excessive cell apoptosis.Moreover, 1, 5, and 10 µmol·L~(-1) catalpol could also up-regulate the phosphorylation of nuclear transcription factor NF-E2 related factors, negatively regulate Kelch-like ECH-associated protein 1(Keap1), phosphorylation of extracellular signal-regulated kinase(ERK), and heme oxyge-nase 1(HO-1), and promote the protein expression of pancreatic-duodenal homeobox factor-1(PDX-1) and glucose transporter 2(GLUT2).In addition, 1, 5, and 10 µmol·L~(-1) catalpol increased insulin secretion of INS-1 cells under oxidative damage in the high-glucose culture medium, indicating function recovery of pancreatic ß cells.PDX-1 is a key nuclear transcription factor of pancreatic ß cell function that directly regulates GLUT2 and insulin synthesis, and affects glucose homeostasis.In conclusion, catalpol can reduce the oxidative damage and apoptosis of INS-1 cells, activate antioxidant pathway, protect the function of pancreatic ß cells, and improve insulin synthesis and secretion.


Asunto(s)
Células Secretoras de Insulina , Apoptosis , Glucosa/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Glucósidos Iridoides , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/metabolismo
10.
Nutrients ; 14(15)2022 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-35893876

RESUMEN

Background: Pancreatic beta cells regulate bioenergetics efficiency and secret insulin in response to glucose and nutrient availability. The mechanistic Target of Rapamycin (mTOR) network orchestrates pancreatic progenitor cell growth and metabolism by nucleating two complexes, mTORC1 and mTORC2. Objective: To determine the impact of mTORC1/mTORC2 inhibition on amino acid metabolism in mouse pancreatic beta cells (Beta-TC-6 cells, ATCC-CRL-11506) using high-resolution metabolomics (HRM) and live-mitochondrial functions. Methods: Pancreatic beta TC-6 cells were incubated for 24 h with either: RapaLink-1 (RL); Torin-2 (T); rapamycin (R); metformin (M); a combination of RapaLink-1 and metformin (RLM); Torin-2 and metformin (TM); compared to the control. We applied high-resolution mass spectrometry (HRMS) LC-MS/MS untargeted metabolomics to compare the twenty natural amino acid profiles to the control. In addition, we quantified the bioenergetics dynamics and cellular metabolism by live-cell imaging and the MitoStress Test XF24 (Agilent, Seahorse). The real-time, live-cell approach simultaneously measures the oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) to determine cellular respiration and metabolism. Statistical significance was assessed using ANOVA on Ranks and post-hoc Welch t-Tests. Results: RapaLink-1, Torin-2, and rapamycin decreased L-aspartate levels compared to the control (p = 0.006). Metformin alone did not affect L-aspartate levels. However, L-asparagine levels decreased with all treatment groups compared to the control (p = 0.03). On the contrary, L-glutamate and glycine levels were reduced only by mTORC1/mTORC2 inhibitors RapaLink-1 and Torin-2, but not by rapamycin or metformin. The metabolic activity network model predicted that L-aspartate and AMP interact within the same activity network. Live-cell bioenergetics revealed that ATP production was significantly reduced in RapaLink-1 (122.23 ± 33.19), Torin-2 (72.37 ± 17.33) treated cells, compared to rapamycin (250.45 ± 9.41) and the vehicle control (274.23 ± 38.17), p < 0.01. However, non-mitochondrial oxygen consumption was not statistically different between RapaLink-1 (67.17 ± 3.52), Torin-2 (55.93 ± 8.76), or rapamycin (80.01 ± 4.36, p = 0.006). Conclusions: Dual mTORC1/mTORC2 inhibition by RapaLink-1 and Torin-2 differentially altered the amino acid profile and decreased mitochondrial respiration compared to rapamycin treatment which only blocks the FRB domain on mTOR. Third-generation mTOR inhibitors may alter the mitochondrial dynamics and reveal a bioenergetics profile that could be targeted to reduce mitochondrial stress.


Asunto(s)
Células Secretoras de Insulina , Metformina , Aminoácidos/metabolismo , Animales , Ácido Aspártico/metabolismo , Cromatografía Liquida , Metabolismo Energético , Células Secretoras de Insulina/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Metformina/farmacología , Ratones , Oxígeno/metabolismo , Transducción de Señal , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Espectrometría de Masas en Tándem
11.
Phytomedicine ; 102: 154190, 2022 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-35636173

RESUMEN

BACKGROUND: Islet transplantation is an effective treatment for the type 1 and severe type 2 diabetes, but it is restricted by the severe lack of pancreas donors. In vitro differentiation of pancreatic progenitors into insulin-secreting cells is one of the hopeful strategies in the cell transplantation therapy of diabetes. Isoastragaloside I is one of the saponin molecules found in Astragalus membranaceus, which has been demonstrated to alleviate insulin resistance and glucose intolerance in obese mice. STUDY DESIGN: We established mouse pancreatic ductal organoids (mPDOs) with progenitor characteristics and an insulin promoter-driven EGFP reporter system to screen astragalus saponin components for monomers that can promote insulin-producing cell differentiation. METHODS: mPDOs treated with or without astragalus saponin monomers were investigated by the insulin promoter-driven EGFP reporter, quantitative PCR, immunofluorescence and flow cytometry to evaluate the expression of endocrine progenitor and ß-cell markers. RESULTS: Isoastragaloside I significantly promoted the expression of ß-cell differentiation genes, which was demonstrated by the activation of the insulin promoter-driven EGFP reporter, as well as the significant increase of mRNA levels of the endocrine progenitor marker Ngn3 and the ß-cell markers insulin1 and insulin2. Immunostaining studies indicated that the ß-cell-specific C-peptide was upregulated in isoastragaloside I-treated mPDOs. FACS analysis revealed that the ratio of C-peptide-secreting cells in isoastragaloside I-treated mPDOs was over 40%. Glucose tolerance tests demonstrated that the differentiated mPDOs could secrete C-peptide in response to glucose stimulation. CONCLUSIONS: We discover a novel strategy of inducing pancreatic ductal progenitors to differentiate into insulin-producing cells using isoastragaloside I. This approach can be potentially applied to ß-cell transplantation in diabetes therapies.


Asunto(s)
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Saponinas , Animales , Péptido C/metabolismo , Diferenciación Celular/fisiología , Diabetes Mellitus Tipo 2/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Ratones , Organoides/metabolismo , Saponinas/metabolismo , Saponinas/farmacología
12.
Arch Toxicol ; 96(7): 1915-1920, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35416515

RESUMEN

The toxic potential of H2O2 is limited, even if intracellular concentrations of H2O2 under conditions of oxidative stress increase to the micromolar concentration range. Its toxicity is mostly restricted to the oxidation of highly reactive thiol groups, some of which are functionally very important. Subsequently, the HO· radical is generated spontaneously from H2O2 in the Fenton reaction. The HO· radical is extremely toxic and destroys any biological structure. Due to the high reactivity, its action is limited to a locally restricted site of its generation. On the other hand, H2O2 with its stability and long half-life can reach virtually any site and distribute its toxic effect all over the cell. Thereby HO·, in spite of its ultra-short half-life (10-9 s), can execute its extraordinary toxic action at any target of the cell. In this oxidative stress scenario, H2O2 is the pro-radical, that spreads the toxic action of the HO· radical. It is the longevity of the H2O2 molecule allowing it to distribute its toxic action from the site of origin all over the cell and may even mediate intercellular communication. Thus, H2O2 acts as a spreader by transporting it to sites where the extremely short-lived toxic HO· radical can arise in the presence of "free iron". H2O2 and HO· act in concert due to their different complementary chemical properties. They are dependent upon each other while executing the toxic effects in oxidative stress under diabetic metabolic conditions in particular in the highly vulnerable pancreatic beta cell, which in contrast to many other cell types is so badly protected against oxidative stress due to its extremely low H2O2 inactivating enzyme capacity.


Asunto(s)
Radical Hidroxilo , Células Secretoras de Insulina , Peróxido de Hidrógeno/metabolismo , Peróxido de Hidrógeno/toxicidad , Radical Hidroxilo/química , Radical Hidroxilo/metabolismo , Células Secretoras de Insulina/metabolismo , Hierro/metabolismo , Oxidación-Reducción
13.
Free Radic Biol Med ; 183: 89-103, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35318102

RESUMEN

Selenoprotein P (SELENOP) is a major selenium (Se)-containing protein (selenoprotein) in human plasma that is mainly synthesized in the liver. SELENOP transports Se to the cells, while SELENOP synthesized in peripheral tissues is incorporated in a paracrine/autocrine manner to maintain the levels of cellular selenoproteins, called the SELENOP cycle. Pancreatic ß cells, responsible for the synthesis and secretion of insulin, are known to express SELENOP. Here, using MIN6 cells as a mouse model for pancreatic ß cells and Selenop small interfering (si)RNA, we found that Selenop gene knockdown (KD) resulted in decreased cell viability, cellular pro/insulin levels, insulin secretion, and levels of several cellular selenoproteins, including glutathione peroxidase 4 (Gpx4) and selenoprotein K (Selenok). These dysfunctions induced by Selenop siRNA were recovered by the addition of Se. Ferroptosis-like cell death, regulated by Gpx4, was involved in the decrease of cell viability by Selenop KD, while stress-induced nascent granule degradation (SINGD), regulated by Selenok, was responsible for the decrease in proinsulin. SINGD was also observed in the pancreatic ß cells of Selenop knockout mice. These findings indicate a significant role of SELENOP expression for the function of pancreatic ß cells by maintaining the levels of cellular selenoproteins such as GPX4 and SELENOK.


Asunto(s)
Ferroptosis , Células Secretoras de Insulina , Selenio , Selenoproteína P , Animales , Glutatión Peroxidasa/genética , Glutatión Peroxidasa/metabolismo , Células Secretoras de Insulina/metabolismo , Ratones , Selenio/metabolismo , Selenoproteína P/genética , Selenoproteína P/metabolismo
14.
Tissue Eng Regen Med ; 19(3): 603-616, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35212973

RESUMEN

BACKGROUND: Endogenous pancreatic ß-cell regeneration is a promising therapeutic approach for enhancing ß-cell function and neogenesis in diabetes. Various findings have reported that regeneration might occur via stimulating ß-cell proliferation, neogenesis, or conversion from other pancreatic cells to ß-like cells. Although the current scenario illustrates numerous therapeutic strategies and approaches that concern endogenous ß-cell regeneration, all of them have not been successful to a greater extent because of cost effectiveness, availability of suitable donors and rejection in case of transplantation, or lack of scientific evidence for many phytochemicals derived from plants that have been employed in traditional medicine. Therefore, the present study aims to investigate the effect of gymnemic acid (GA) on ß-cell regeneration in streptozotocin-induced type 1 diabetic rats and high glucose exposed RIN5-F cells. METHODS: The study involves histopathological and immunohistochemical analysis to examine the islet's architecture. Quantitative polymerase chain reaction (qPCR) and/or immunoblot were employed to quantify the ß-cell regeneration markers and cell cycle proliferative markers. RESULTS: The immunoexpression of E-cadherin, ß-catenin, and phosphoinositide 3-kinases/protein kinase B were significantly increased in GA-treated diabetic rats. On the other hand, treatment with GA upregulated the pancreatic regenerative transcription factor viz. pancreatic duodenal homeobox 1, Neurogenin 3, MafA, NeuroD1, and ß-cells proliferative markers such as CDK4, and Cyclin D1, with a simultaneous downregulation of the forkhead box O, glycogen synthase kinase-3, and p21cip1 in diabetic treated rats. Adding to this, we noticed increased nuclear localization of Pdx1 in GA treated high glucose exposed RIN5-F cells. CONCLUSION: Our results suggested that GA acts as a potential therapeutic candidate for endogenous ß-cell regeneration in treating type 1 diabetes.


Asunto(s)
Diabetes Mellitus Experimental , Células Secretoras de Insulina , Animales , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Glucosa/metabolismo , Células Secretoras de Insulina/metabolismo , Ratas , Regeneración , Saponinas , Triterpenos
15.
J Biochem Mol Toxicol ; 36(4): e23004, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35191145

RESUMEN

Saponin gypenoside A (GP) has shown its potential to handle diabetes mellitus. MicroRNA-150-3p (miR-150-3p) is closely related to the dysfunction of pancreatic ß cells by targeting PDX1. Given the function of GP is related to its regulation on different miRs, the current study assessed the role of miR-150-3p as a therapeutic target for the hypoglycemic effects of GP. Pancreatic ß cell dysfunction was induced in mice using the high-fatty diet (HFD) method and then handled with GP. Changes in insulin release and resistance and the activity of the miR-150-3p/PDX1 axis were detected. The expression of miR-150-3p was induced to confirm its central in the effects of GP. The results of in vivo tests were then validated with in vitro assays. HFD administration suppressed glucose tolerance, delayed insulin release, and induced insulin resistance and pancreas apoptosis in mice, which was indicative of the dysfunction of ß pancreatic cells. Changes in pancreatic ß function were associated with the increased expression of miR-150-3p and suppressed expression of PDX1. After the administration of GP, the impairments of the pancreas were alleviated and the expression of miR-150-3p was inhibited, contributing to the restored level of PDX1. The injection of miR-150-3p agomir counteracted the protective effects of GP. In in vitro assays, the pretransfection of miR-150-3p mimetics also counteracted the protective effects of GP on pancreatic ß cells against palmitic acid. Collectively, miR-150-3p played a key role in the protective effects of GP against pancreatic ß cell dysfunction by inhibiting PDX1 expression.


Asunto(s)
Células Secretoras de Insulina , MicroARNs , Animales , Gynostemma , Proteínas de Homeodominio/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Extractos Vegetales , Transducción de Señal , Transactivadores/metabolismo
16.
Cell Metab ; 34(2): 285-298.e7, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35108515

RESUMEN

The central nervous system has long been thought to regulate insulin secretion, an essential process in the maintenance of blood glucose levels. However, the anatomical and functional connections between the brain and insulin-producing pancreatic ß cells remain undefined. Here, we describe a functional transneuronal circuit connecting the hypothalamus to ß cells in mice. This circuit originates from a subpopulation of oxytocin neurons in the paraventricular hypothalamic nucleus (PVNOXT), and it reaches the islets of the endocrine pancreas via the sympathetic autonomic branch to innervate ß cells. Stimulation of PVNOXT neurons rapidly suppresses insulin secretion and causes hyperglycemia. Conversely, silencing of these neurons elevates insulin levels by dysregulating neuronal signaling and secretory pathways in ß cells and induces hypoglycemia. PVNOXT neuronal activity is triggered by glucoprivation. Our findings reveal that a subset of PVNOXT neurons form functional multisynaptic circuits with ß cells in mice to regulate insulin secretion, and their function is necessary for the ß cell response to hypoglycemia.


Asunto(s)
Células Secretoras de Insulina , Animales , Hipotálamo/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Ratones , Oxitocina/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo
17.
Diabetes ; 71(5): 961-977, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35113157

RESUMEN

To date, identification of nonislet-specific transcriptional factors in the regulation of insulin gene expression has been little studied. Here, we report that the expression level of the transcription factor YY1 is increased dramatically in both human and mouse pancreatic ß-cells after birth. Nevertheless, the physiological role of YY1 during ß-cell development and its regulatory mechanism in ß-cell function remain largely unknown. After ß-cell ablation of Yy1, we observed rapid onset of hyperglycemia, impaired glucose tolerance, and reduced ß-cell mass in neonatal and adult mice. These mice also had hypoinsulinemia with normal insulin sensitivity compared with their wild-type littermates, manifesting as a type 1 diabetic phenotype. Mechanistically, genome-wide RNA sequencing has defined dysregulated insulin signaling and defective glucose responsiveness in ß-cells devoid of YY1. Integrative analyses coupled with chromatin immunoprecipitation assays targeting YY1, and histone modifications, including H3K4me1, H3K27ac, and H3K27me3, have further identified Ins1 and Ins2 as direct gene targets of YY1. Luciferase reporter assays and loss- and gain-of-function experiments also demonstrated that YY1 binds to the enhancer regions in exon 2 of Ins1 and Ins2, activating insulin transcription and, therefore, proinsulin and insulin production in pancreatic ß-cells. YY1 also directly interacts with RNA polymerase II, potentially stabilizing the enhancer-promoter interaction in the multiprotein-DNA complex during transcription initiation. Taken together, our findings suggest a role for YY1 as a transcriptional activator of insulin gene expression, assisting ß-cell maturation and function after birth. These analyses may advance our understanding of ß-cell biology and provide clinically relevant insights targeting the pathophysiological origins of diabetes.


Asunto(s)
Diabetes Mellitus , Células Secretoras de Insulina , Animales , Diabetes Mellitus/metabolismo , Glucosa/metabolismo , Homeostasis , Insulina/metabolismo , Insulina Regular Humana , Células Secretoras de Insulina/metabolismo , Ratones , Factor de Transcripción YY1/genética , Factor de Transcripción YY1/metabolismo
18.
Expert Opin Ther Targets ; 26(1): 57-67, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35138971

RESUMEN

INTRODUCTION: Current treatment for type 1 diabetes (T1D) is centered around insulin supplementation to manage the effects of pancreatic ß cell loss. GDF15 is a potential preventative therapy against T1D progression that could work to curb increasing disease incidence. AREAS COVERED: This paper discusses the known actions of GDF15, a pleiotropic protein with metabolic, feeding, and immunomodulatory effects, connecting them to highlight the open opportunities for future research. The role of GDF15 in the prevention of insulitis and protection of pancreatic ß cells against pro-inflammatory cytokine-mediated cellular stress are examined and the pharmacological promise of GDF15 and critical areas of future research are discussed. EXPERT OPINION: GDF15 shows promise as a potential intervention but requires further development. Preclinical studies have shown poor efficacy, but this result may be confounded by the measurement of gross GDF15 instead of the active form. Additionally, the effect of GDF15 in the induction of anorexia and nausea-like behavior and short-half-life present significant challenges to its deployment, but a systems pharmacology approach paired with chronotherapy may provide a possible solution to therapy for this currently unpreventable disease.


Asunto(s)
Diabetes Mellitus Tipo 1 , Células Secretoras de Insulina , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/metabolismo , Factor 15 de Diferenciación de Crecimiento/metabolismo , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo
19.
Mol Med Rep ; 25(3)2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35039874

RESUMEN

Type­2 diabetes mellitus (T2DM) causes several complications that affect the quality of life and life span of patients. Hyperbaric oxygen therapy (HBOT) has been used to successfully treat several diseases, including carbon monoxide poisoning, ischemia, infections and diabetic foot ulcer, and increases insulin sensitivity in T2DM. The present study aimed to determine the effect of HBOT on ß­cell function and hepatic gluconeogenesis in streptozotocin (STZ)­induced type­2 diabetic mice. To establish a T2DM model, 7­week­old male C57BL/6J mice were fed a high­fat diet (HFD) and injected once daily with low­dose STZ for 3 days after 1­week HFD feeding. At the 14th week, HFD+HBOT and T2DM+HBOT groups received 1­h HBOT (2 ATA; 100% pure O2) daily from 5:00 to 6:00 p.m. for 7 days. The HFD and T2DM groups were maintained under normobaric oxygen conditions and used as controls. During HBOT, the 12­h nocturnal food intake and body weight were measured daily. Moreover, blood glucose was measured by using a tail vein prick and a glucometer. After the final HBO treatment, all mice were sacrificed to conduct molecular biology experiments. Fasting insulin levels of blood samples of sacrificed mice were measured by an ultrasensitive ELISA kit. Pancreas and liver tissues were stained with hematoxylin and eosin, while immunohistochemistry was performed to determine the effects of HBOT on insulin resistance. TUNEL was used to determine the effects of HBOT on ß­cell apoptosis, and immunoblotting was conducted to determine the ß­cell apoptosis pathway. HBOT notably reduced fasting blood glucose and improved insulin sensitivity in T2DM mice. After HBOT, ß­cell area and ß­cell mass in T2DM mice were significantly increased. HBOT significantly decreased the ß­cell apoptotic rate in T2DM mice via the pancreatic Bcl­2/caspase­3/poly(ADP­ribose) polymerase (PARP) apoptosis pathway. Moreover, HBOT improved the morphology of the liver tissue and increased hepatic glycogen storage in T2DM mice. These findings suggested that HBOT ameliorated the insulin sensitivity of T2DM mice by decreasing the ß­cell apoptotic rate via the pancreatic Bcl­2/caspase­3/PARP apoptosis pathway.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Gluconeogénesis/fisiología , Oxigenoterapia Hiperbárica/métodos , Células Secretoras de Insulina/metabolismo , Hígado/metabolismo , Animales , Apoptosis/fisiología , Glucemia/metabolismo , Western Blotting , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/etiología , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/etiología , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Ayuno/sangre , Prueba de Tolerancia a la Glucosa/métodos , Humanos , Insulina/sangre , Células Secretoras de Insulina/citología , Masculino , Ratones Endogámicos C57BL
20.
J Clin Endocrinol Metab ; 107(1): 230-240, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34473295

RESUMEN

CONTEXT: Vitamin D regulates glucose homeostasis pathways, but effects of vitamin D supplementation on ß-cell function remain unclear. OBJECTIVE: To investigate the effects of vitamin D3 supplementation on insulin sensitivity and ß-cell function. METHODS: This is a prespecified secondary analysis of the Vitamin D and Type 2 Diabetes study. Overweight/obese adults at high risk for type 2 diabetes (prediabetes) were randomly treated with vitamin D3 4000 IU or matching placebo daily for 24 months. MAIN OUTCOME: Disposition index (DI), as an estimate of ß-cell function, was calculated as the product of Homeostasis Model Assessment 2 indices derived from C-peptide values (HOMA2%Scpep) and C-peptide response during the first 30 minutes of a 75-g oral glucose tolerance test (OGTT). RESULTS: Mean age was 60.5 ± 9.8 years and body mass index was 31.9 ± 4.4 kg/m2. Mean serum 25(OH)D level increased from 27.9 ± 10.3 ng/mL at baseline to 54.9 ng/mL at 2 years in the vitamin D group and was unchanged (28.5 ± 10.0 ng/mL) in the placebo group. The baseline DI predicted incident diabetes independent of the intervention. In the entire cohort, there were no significant differences in changes in DI, HOMA2%Scpep, or C-peptide response between the 2 groups. Among participants with baseline 25(OH)D level <12 ng/mL, the mean percent differences for DI between the vitamin D and placebo groups was 8.5 (95% CI, 0.2-16.8). CONCLUSIONS: Supplementation with vitamin D3 for 24 months did not improve an OGTT-derived index of ß-cell function in people with prediabetes not selected based on baseline vitamin D status; however, there was benefit among those with very low baseline vitamin D status.


Asunto(s)
Colecalciferol/administración & dosificación , Diabetes Mellitus Tipo 2/epidemiología , Células Secretoras de Insulina/metabolismo , Estado Prediabético/dietoterapia , Deficiencia de Vitamina D/dietoterapia , Anciano , Glucemia/metabolismo , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/prevención & control , Suplementos Dietéticos , Femenino , Estudios de Seguimiento , Prueba de Tolerancia a la Glucosa , Humanos , Incidencia , Insulina/metabolismo , Resistencia a la Insulina , Masculino , Persona de Mediana Edad , Sobrepeso/sangre , Sobrepeso/complicaciones , Sobrepeso/dietoterapia , Sobrepeso/metabolismo , Estado Prediabético/sangre , Estado Prediabético/diagnóstico , Estado Prediabético/metabolismo , Resultado del Tratamiento , Vitamina D/análogos & derivados , Vitamina D/sangre , Deficiencia de Vitamina D/sangre , Deficiencia de Vitamina D/diagnóstico , Deficiencia de Vitamina D/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA