Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
FASEB J ; 38(4): e23490, 2024 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-38363581

RESUMO

Appropriate Ca2+ concentration in the endoplasmic reticulum (ER), modulating cytosolic Ca2+ signal, serves significant roles in physiological function of pancreatic ß cells. To maintaining ER homeostasis, Ca2+ movement across the ER membrane is always accompanied by a simultaneous K+ flux in the opposite direction. KCNH6 was proven to modulate insulin secretion by controlling plasma membrane action potential duration and intracellular Ca2+ influx. Meanwhile, the specific function of KCNH6 in pancreatic ß-cells remains unclear. In this study, we found that KCNH6 exhibited mainly ER localization and Kcnh6 ß-cell-specific knockout (ßKO) mice suffered from abnormal glucose tolerance and impaired insulin secretion in adulthood. ER Ca2+ store was overloaded in islets of ßKO mice, which contributed to ER stress and ER stress-induced apoptosis in ß cells. Next, we verified that ethanol treatment induced increases in ER Ca2+ store and apoptosis in pancreatic ß cells, whereas adenovirus-mediated KCNH6 overexpression in islets attenuated ethanol-induced ER stress and apoptosis. In addition, tail-vein injections of KCNH6 lentivirus rescued KCNH6 expression in ßKO mice, restored ER Ca2+ overload and attenuated ER stress in ß cells, which further confirms that KCNH6 protects islets from ER stress and apoptosis. These data suggest that KCNH6 on the ER membrane may help to stabilize intracellular ER Ca2+ stores and protect ß cells from ER stress and apoptosis. In conclusion, our study reveals the protective potential of KCNH6-targeting drugs in ER stress-induced diabetes.


Assuntos
Diabetes Mellitus , Células Secretoras de Insulina , Camundongos , Animais , Secreção de Insulina , Diabetes Mellitus/metabolismo , Células Secretoras de Insulina/metabolismo , Retículo Endoplasmático/metabolismo , Estresse do Retículo Endoplasmático/fisiologia , Cálcio/metabolismo , Etanol , Insulina/metabolismo
2.
Biochem Biophys Res Commun ; 589: 165-172, 2022 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-34922198

RESUMO

Low serum bicarbonate is closely related to type 2 diabetes mellitus. However, the precise role of bicarbonate on glucose homeostasis and insulin secretion remains unknown. In this study, we investigated the effects of bicarbonate concentration on pancreatic ß-cells. It was observed that the high bicarbonate concentration of the cell culture medium significantly increased the glucose-induced insulin secretion (GSIS) levels in mouse islets, MIN6, and the INS-1E ß cells. MIN6 cells presented an impaired GSIS; the cells produced a lower bicarbonate concentration when co-cultured with Capan-1 than when with CFPAC-1. NBCe1, a major bicarbonate transporter was observed to block the increasing insulin secretions, which were promoted by a high concentration of bicarbonate. In addition, higher extracellular bicarbonate concentration significantly increased the intracellular cAMP level, pHi, and calcium concentration with a 16.7 mM of glucose stimulation. Further study demonstrated that a low concentration of extracellular bicarbonate significantly impaired the functioning of pancreatic ß cells by reducing coupling Ca2+ influx, whose process may be modulated by NBCe1. Taken together, our results conclude that bicarbonate may serve as a novel target in diabetes prevention-related research.


Assuntos
Bicarbonatos/farmacologia , Glucose/farmacologia , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Animais , Cálcio/metabolismo , Linhagem Celular , AMP Cíclico/metabolismo , Concentração de Íons de Hidrogênio , Secreção de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/efeitos dos fármacos , Camundongos , Simportadores de Sódio-Bicarbonato/metabolismo
3.
FASEB J ; 34(11): 15015-15028, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32918525

RESUMO

Adult patients with dysfunction in human ether-a-go-go 2 (hERG2) protein, encoded by KCNH6, present with hypoinsulinemia and hyperglycemia. However, the mechanism of KCNH6 action in glucose disorders has not been clearly defined. Previous studies identified that sustained endoplasmic reticulum (ER) stress-mediated apoptosis of pancreatic ß-cells and directly contributed to diabetes. In the present study, we showed that Kcnh6 knockout (KO) mice had impaired glucose tolerance mediated by high ER stress levels, and showed increased apoptosis and elevated intracellular calcium levels in pancreatic ß-cells. In contrast, KCNH6 overexpression in islets isolated from C57BL/6J mice attenuated ER stress induced by thapsigargin or palmitic acid. This effect contributed to better preservation of ß-cells, as reflected in increased ß cell survival and enhanced glucose-stimulated insulin secretion. These results were further corroborated by studies evaluating KCNH6 overexpression in KO islets. Similarly, induction of Kcnh6 in KO mice by lentivirus injection improved glucose tolerance by reducing pancreatic ER stress and apoptosis. Our data provide new insights into how Kcnh6 deficiency causes ER calcium depletion and ß cell dysfunction.


Assuntos
Apoptose , Estresse do Retículo Endoplasmático , Canais de Potássio Éter-A-Go-Go/fisiologia , Células Secretoras de Insulina/citologia , Substâncias Protetoras/farmacologia , Tapsigargina/farmacologia , Animais , Cálcio/metabolismo , Inibidores Enzimáticos/farmacologia , Feminino , Glucose/farmacologia , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ácido Palmítico/farmacologia
4.
Horm Metab Res ; 52(9): 669-675, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32750722

RESUMO

Adult patients with a dysfunctional ether-a-go-go 2 (hERG2) protein, which is encoded by the KCNH6 gene, present with hyperinsulinemia and hyperglycemia. However, the mechanism of KCNH6 in glucose metabolism disorders has not been clearly defined. It has been proposed that sustained endoplasmic reticulum (ER) stress is closely concerned with hepatic insulin resistance and inflammation. Here, we demonstrate that Kcnh6 knockout (KO) mice had impaired glucose tolerance and increased levels of hepatic apoptosis, in addition to displaying an increased insulin resistance that was mediated by high ER stress levels. By contrast, overexpression of KCNH6 in primary hepatocytes led to a decrease in ER stress and apoptosis induced by thapsigargin. Similarly, induction of Kcnh6 by tail vein injection into KO mice improved glucose tolerance by reducing ER stress and apoptosis. Furthermore, we show that KCNH6 alleviated hepatic ER stress, apoptosis, and inflammation via the NFκB-IκB kinase (IKK) pathway both in vitro and in vivo. In summary, our study provides new insights into the causes of ER stress and subsequent induction of primary hepatocytes apoptosis.


Assuntos
Estresse do Retículo Endoplasmático , Canais de Potássio Éter-A-Go-Go/fisiologia , Intolerância à Glucose/patologia , Glucose/metabolismo , Resistência à Insulina , Hepatopatias/patologia , Animais , Apoptose , Intolerância à Glucose/etiologia , Intolerância à Glucose/metabolismo , Hepatopatias/etiologia , Hepatopatias/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
5.
Mol Metab ; 80: 101885, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38246588

RESUMO

OBJECTIVE: Genome-scale CRISPR-Cas9 knockout coupled with single-cell RNA sequencing (scRNA-seq) has been used to identify function-related genes. However, this method may knock out too many genes, leading to low efficiency in finding genes of interest. Insulin secretion is controlled by several electrophysiological events, including fluxes of KATP depolarization and K+ repolarization. It is well known that glucose stimulates insulin secretion from pancreatic ß-cells, mainly via the KATP depolarization channel, but whether other nutrients directly regulate the repolarization K+ channel to promote insulin secretion is unknown. METHODS: We used a system involving CRISPR-Cas9-mediated knockout of all 83 K+ channels and scRNA-seq in a pancreatic ß cell line to identify genes associated with insulin secretion. RESULTS: The expression levels of insulin genes were significantly increased after all-K+ channel knockout. Furthermore, Kcnb1 and Kcnh6 were the two most important repolarization K+ channels for the increase in high-glucose-dependent insulin secretion that occurred upon application of specific inhibitors of the channels. Kcnh6 currents, but not Kcnb1 currents, were reduced by one of the amino acids, lysine, in both transfected cells, primary cells and mice with ß-cell-specific deletion of Kcnh6. CONCLUSIONS: Our function-related CRISPR screen with scRNA-seq identifies Kcnh6 as a lysine-specific channel.


Assuntos
Insulina , Lisina , Camundongos , Animais , Secreção de Insulina , Lisina/metabolismo , Insulina/metabolismo , Glucose/farmacologia , Trifosfato de Adenosina/metabolismo
6.
Oxid Med Cell Longev ; 2022: 3739556, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36217412

RESUMO

KCNH6 has been proven to affect glucose metabolism and insulin secretion both in humans and mice. Further study revealed that Kcnh6 knockout (KO) mice showed impaired glucose tolerance. However, the precise function of KCNH6 in the liver remains unknown. Mitochondria have been suggested to maintain intracellular Ca2+ homeostasis; ROS generation and defective mitochondria can cause glucose metabolism disorders, including type 2 diabetes (T2D). Here, we found that Kcnh6 attenuated glucose metabolism disorders by decreasing PEPCK and G6pase abundance and induced Glut2 and IRS2 expression. Overexpression of Kcnh6 increased hepatic glucose uptake and glycogen synthesis. Kcnh6 attenuated intracellular and mitochondrial calcium levels in primary hepatocytes and reduced intracellular ROS and mitochondrial superoxide production. Kcnh6 suppressed oxidative stress by inhibiting mitochondrial pathway activation and NADPH oxidase expression. Experiments demonstrated that Kcnh6 expression improved hepatic glucose metabolism disorder through the c-Jun N-terminal kinase and p38MAPK signaling pathways. These results were confirmed by experiments evaluating the extent to which forced Kcnh6 expression rescued metabolic disorder in KO mice. In conclusion, KCNH6 enhanced hepatic glucose metabolism by regulating mitochondrial Ca2+ levels and inhibiting oxidative stress. As liver glucose metabolism is key to T2D, understanding KCNH6 functions may provide new insights into the causes of diabetes.


Assuntos
Diabetes Mellitus Tipo 2 , Canais de Potássio Éter-A-Go-Go , Glucose , Mitocôndrias , Estresse Oxidativo , Animais , Cálcio/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Canais de Potássio Éter-A-Go-Go/metabolismo , Glucose/metabolismo , Glicogênio/metabolismo , Humanos , Insulina/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Fígado/metabolismo , Camundongos , Camundongos Knockout , Mitocôndrias/metabolismo , NADPH Oxidases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Superóxidos/metabolismo
7.
Int J Endocrinol ; 2021: 4350418, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34938332

RESUMO

Impaired fasting glucose (IFG) is a reversible intermediate hyperglycemia stage with an increasing risk of diabetes and related complications. Our study was designed to identify the relationship between the serum anion gap and the risk of progressing to impaired fasting glucose and diabetes. Here, we performed a prospective, population-based study among 1191 Chinese individuals aged 22-87 years who took health examinations annually between 2006 and 2012 including clinical features and plasma metabolites. All of the participants had no history of diabetes or related chronic complications. Logistic regression analysis was designed to examine the associations between clinical and metabolomic factors and the risk of developing IFG or diabetes. Among them, 58 subjects whose fasting glucose were between 6.1 and 7 mmol/L were diagnosed as IFG or diabetes. After adjusting for age, sex, body mass index (BMI), high-density lipoprotein (HDL), low-density lipoprotein (LDL), alanine aminotransferase (ALT), aspartate aminotransferase (AST), systolic blood pressure (SBP), diastolic blood pressure (DBP), potassium, and albumin at baseline, the participants in the upper tertiles of serum anion gap (SAG) had higher odds of progressing to IFG or diabetes than those in the lower tertiles. A receiver operating characteristic (ROC) curve was analyzed, and the optimal cutoff level for the anion gap to predict incident IFG or diabetes was 13.76 mmol/L, and the area under the ROC curve (AUC) was 0.623. Our data demonstrate that a higher serum anion gap is associated with the risk of developing IFG or diabetes.

8.
Cell Cycle ; 20(19): 2053-2065, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34494921

RESUMO

Diabetes mellitus is caused by either insulin resistance or insulin deficiency. The pancreatic ß cells are the primary producers of insulin. Large-scale CRISPR screens combined with single-cell RNA sequencing (scRNA-seq) on ß cells has identified novel insulin regulators and revealed the presence of a highly complex inner network. Here, we performed pooled CRISPR delivery with single-cell transcriptome analysis on the MIN6 cell line, a pancreatic ß-cell line. We have presented the scRNA-seq readout and demonstrated that the MIN6 cell line might develop genetic heterogeneity with increasing passage number based on GO and KEGG pathway analysis. Both computational and biological analyses revealed that the function of MIN6 cell lines could be divided into five clusters, including endocrine cells, basal cells, epithelial cells, and neuroendocrine cells. The fifth cluster was different from the other four clusters due to the differentially expressed insulin transcription and was called the lncRNA-enriched cluster. The experiments also confirmed that uncharacterized lncRNAs GM26917 and Cenpw were associated with insulin transcription. This study provides information that can be used to systematically characterize insulin regulator genes and other genes that control protein folding and vesicle trafficking.


Assuntos
Células Secretoras de Insulina , Insulina , Animais , Linhagem Celular , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Camundongos , Análise de Célula Única
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa