Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Cell Rep ; 43(4): 113992, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38536815

ABSTRACT

Insulin is packaged into secretory granules that depart the Golgi and undergo a maturation process that involves changes in the protein and lipid composition of the granules. Here, we show that insulin secretory granules form physical contacts with the endoplasmic reticulum and that the lipid exchange protein oxysterol-binding protein (OSBP) is recruited to these sites in a Ca2+-dependent manner. OSBP binding to insulin granules is positively regulated by phosphatidylinositol-4 (PI4)-kinases and negatively regulated by the PI4 phosphate (PI(4)P) phosphatase Sac2. Loss of Sac2 results in excess accumulation of cholesterol on insulin granules that is normalized when OSBP expression is reduced, and both acute inhibition and small interfering RNA (siRNA)-mediated knockdown of OSBP suppress glucose-stimulated insulin secretion without affecting insulin production or intracellular Ca2+ signaling. In conclusion, we show that lipid exchange at endoplasmic reticulum (ER)-granule contact sites is involved in the exocytic process and propose that these contacts act as reaction centers with multimodal functions during insulin granule maturation.


Subject(s)
Cholesterol , Endoplasmic Reticulum , Insulin Secretion , Insulin , Minor Histocompatibility Antigens , Receptors, Steroid , Secretory Vesicles , Endoplasmic Reticulum/metabolism , Secretory Vesicles/metabolism , Animals , Cholesterol/metabolism , Insulin/metabolism , Receptors, Steroid/metabolism , Phosphatidylinositol Phosphates/metabolism , Mice , Humans , Calcium/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Glucose/metabolism
2.
Nat Commun ; 14(1): 8040, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-38086799

ABSTRACT

Epigenetic dysregulation may influence disease progression. Here we explore whether epigenetic alterations in human pancreatic islets impact insulin secretion and type 2 diabetes (T2D). In islets, 5,584 DNA methylation sites exhibit alterations in T2D cases versus controls and are associated with HbA1c in individuals not diagnosed with T2D. T2D-associated methylation changes are found in enhancers and regions bound by ß-cell-specific transcription factors and associated with reduced expression of e.g. CABLES1, FOXP1, GABRA2, GLR1A, RHOT1, and TBC1D4. We find RHOT1 (MIRO1) to be a key regulator of insulin secretion in human islets. Rhot1-deficiency in ß-cells leads to reduced insulin secretion, ATP/ADP ratio, mitochondrial mass, Ca2+, and respiration. Regulators of mitochondrial dynamics and metabolites, including L-proline, glycine, GABA, and carnitines, are altered in Rhot1-deficient ß-cells. Islets from diabetic GK rats present Rhot1-deficiency. Finally, RHOT1methylation in blood is associated with future T2D. Together, individuals with T2D exhibit epigenetic alterations linked to mitochondrial dysfunction in pancreatic islets.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , Islets of Langerhans , Humans , Rats , Animals , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Insulin Secretion , Insulin/metabolism , DNA Methylation , Islets of Langerhans/metabolism , Insulin-Secreting Cells/metabolism , Transcription Factors/metabolism , Epigenesis, Genetic , Mitochondria/genetics , Mitochondria/metabolism , Repressor Proteins/metabolism , Forkhead Transcription Factors/metabolism
3.
J Endocrinol ; 258(2)2023 08 01.
Article in English | MEDLINE | ID: mdl-37159865

ABSTRACT

Glucagon is the principal glucose-elevating hormone that forms the first-line defence against hypoglycaemia. Along with insulin, glucagon also plays a key role in maintaining systemic glucose homeostasis. The cells that secrete glucagon, pancreatic α-cells, are electrically excitable cells and use electrical activity to couple its hormone secretion to changes in ambient glucose levels. Exactly how glucose regulates α-cells has been a topic of debate for decades but it is clear that electrical signals generated by the cells play an important role in glucagon secretory response. Decades of studies have already revealed the key players involved in the generation of these electrical signals and possible mechanisms controlling them to tune glucagon release. This has offered the opportunity to fully understand the enigmatic α-cell physiology. In this review, we describe the current knowledge on cellular electrophysiology and factors regulating excitability, glucose sensing, and glucagon secretion. We also discuss α-cell pathophysiology and the perspective of addressing glucagon secretory defects in diabetes for developing better diabetes treatment, which bears the hope of eliminating hypoglycaemia as a clinical problem in diabetes care.


Subject(s)
Diabetes Mellitus , Glucagon-Secreting Cells , Hypoglycemia , Humans , Glucagon , Insulin , Glucose , Cell Physiological Phenomena , Electrophysiology
4.
Biochem Biophys Res Commun ; 649: 87-92, 2023 03 15.
Article in English | MEDLINE | ID: mdl-36758483

ABSTRACT

Type 2 diabetes (T2D) is associated with low-grade inflammation. Here we investigate if the anti-inflammatory cytokine interleukin-4 (IL-4) affects glucose-stimulated insulin secretion (GSIS) in human islets from non-diabetic (ND) and type-2 diabetic (T2D) donors. We first confirmed that GSIS is reduced in islets from T2D donors. Treatment with IL-4 for 48 h had no further effect on GSIS in these islets but significantly reduced secretion in ND islets. Acute treatment with IL-4 for 1 h had no effect on GSIS in ND islets which led us to suspect that IL-4 affects a slow cellular mechanism such as gene transcription. IL-4 has been reported to regulate miR-378a-3p and, indeed, we found that this microRNA was increased with IL-4 treatment. However, overexpression of miR-378a-3p in the human beta cell line EndoC-ßH1 did not affect GSIS. MiR-378a-3p is transcribed from the same gene as peroxisome proliferator-activated receptor gamma co-activator 1 beta (PCG-1ß) and we found that IL-4 treatment showed a clear tendency to increased gene expression of PCG-1ß. PCG-1ß is a co-activator of peroxisome proliferator-activated receptor gamma (PPARγ) and, the gene expression of PPARγ was also increased with IL-4 treatment. Our data suggests that the protective role of IL-4 on beta cell survival comes at the cost of lowered insulin secretion, presumably involving the PPARγ-pathway.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , Islets of Langerhans , MicroRNAs , Humans , Insulin Secretion , Diabetes Mellitus, Type 2/metabolism , Interleukin-4/pharmacology , Interleukin-4/metabolism , PPAR gamma/metabolism , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Glucose/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Islets of Langerhans/metabolism
5.
Acta Physiol (Oxf) ; 236(1): e13857, 2022 09.
Article in English | MEDLINE | ID: mdl-35753051

ABSTRACT

AIM: SYT11 and SYT13, two calcium-insensitive synaptotagmins, are downregulated in islets from type 2 diabetic donors, but their function in insulin secretion is unknown. To address this, we investigated the physiological role of these two synaptotagmins in insulin-secreting cells. METHODS: Correlations between gene expression levels were performed using previously described RNA-seq data on islets from 188 human donors. SiRNA knockdown was performed in EndoC-ßH1 and INS-1 832/13 cells. Insulin secretion was measured with ELISA. Patch-clamp was used for single-cell electrophysiology. Confocal microscopy was used to determine intracellular localization. RESULTS: Human islet expression of the transcription factor PDX1 was positively correlated with SYT11 (p = 2.4e-10 ) and SYT13 (p < 2.2e-16 ). Syt11 and Syt13 both co-localized with insulin, indicating their localization in insulin granules. Downregulation of Syt11 in INS-1 832/13 cells (siSYT11) resulted in increased basal and glucose-induced insulin secretion. Downregulation of Syt13 (siSYT13) decreased insulin secretion induced by glucose and K+ . Interestingly, the cAMP-raising agent forskolin was unable to enhance insulin secretion in siSYT13 cells. There was no difference in insulin content, exocytosis, or voltage-gated Ca2+ currents in the two models. Double knockdown of Syt11 and Syt13 (DKD) resembled the results in siSYT13 cells. CONCLUSION: SYT11 and SYT13 have similar localization and transcriptional regulation, but they regulate insulin secretion differentially. While downregulation of SYT11 might be a compensatory mechanism in type-2 diabetes, downregulation of SYT13 reduces the insulin secretory response and overrules the compensatory regulation of SYT11 in a way that could aggravate the disease.


Subject(s)
Calcium , Insulin-Secreting Cells , Calcium/metabolism , Glucose/metabolism , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Synaptotagmins/genetics , Synaptotagmins/metabolism
6.
Curr Opin Pharmacol ; 63: 102199, 2022 04.
Article in English | MEDLINE | ID: mdl-35245797

ABSTRACT

Diabetes is the result of dysregulation of both insulin and glucagon. Still, insulin has attracted much more attention than glucagon. Glucagon is released from alpha cells in the islets of Langerhans in response to low glucose and certain amino acids. Drugs with the primary aim of targeting glucagon signalling are scarce. However, glucagon is often administered to counteract severe hypoglycaemia, and commonly used diabetes medications such as GLP-1 analogues, sulfonylureas and SGLT2-inhibitors also affect alpha cells. Indeed, there are physiological and developmental similarities between the alpha cell and the insulin-secreting beta cell and new data confirm that alpha cells can be converted into insulin-secreting cells. These aspects and attributes, the need to find novel therapies targeting the alpha cell and more are considered in this review.


Subject(s)
Glucagon-Secreting Cells , Insulin-Secreting Cells , Islets of Langerhans , Glucagon/metabolism , Glucagon-Like Peptide 1/metabolism , Glucagon-Secreting Cells/metabolism , Glucose/metabolism , Homeostasis , Humans , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism
7.
iScience ; 25(4): 103995, 2022 Apr 15.
Article in English | MEDLINE | ID: mdl-35310942

ABSTRACT

MicroRNAs (miRNAs) are short non-coding RNAs that regulate gene expression via mRNA targeting, playing important roles in the pancreatic islets. We aimed to identify molecular pathways and genomic regulatory regions associated with altered miRNA expression due to glycemic status, which could contribute to the development of type 2 diabetes (T2D). To this end, miRNAs were identified by a combination of differential miRNA expression and correlation analysis in human islet samples from donors with normal and elevated blood glucose levels. Analysis and clustering of highly correlated, experimentally validated gene targets of these miRNAs revealed two islet-specific clusters, which were associated with key aspects of islet functions and included a high number of T2D-related genes. Finally, cis-eQTLs and public GWAS data integration uncovered suggestive genomic signals of association with insulin secretion and T2D. The miRNA-driven network-based approach presented in this study contributes to a better understanding of impaired insulin secretion in T2D pathogenesis.

8.
Diabetes ; 71(2): 275-284, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34753799

ABSTRACT

MicroRNAs (miRNAs) are part of deregulated insulin secretion in type 2 diabetes (T2D) development. Rodent models have suggested miR-200c to be involved, but the role and potential as therapeutic target of this miRNA in human islets are not clear. Here we report increased expression of miR-200c in islets from T2D as compared with nondiabetic (ND) donors and display results showing reduced glucose-stimulated insulin secretion in EndoC-ßH1 cells overexpressing miR-200c. We identify transcription factor ETV5 as the top rank target of miR-200c in human islets using TargetScan in combination with Pearson correlation analysis of miR-200c and mRNA expression data from the same human donors. Among other targets were JAZF1, as earlier shown in miR-200 knockout mice. Accordingly, linear model analysis of ETV5 and JAZF1 gene expression showed reduced expression of both genes in islets from human T2D donors. Western blot analysis confirmed the reduced expression of ETV5 on the protein level in EndoC-ßH1 cells overexpressing miR-200c, and luciferase assay validated ETV5 as a direct target of miR-200c. Finally, LNA knockdown of miR-200c increased glucose-stimulated insulin secretion in islets from T2D donors approximately threefold. Our data reveal a vital role of the miR-200c-ETV5 axis in ß-cell dysfunction and pathophysiology of T2D.


Subject(s)
DNA-Binding Proteins/genetics , Diabetes Mellitus, Type 2 , Insulin Secretion/genetics , Islets of Langerhans/metabolism , MicroRNAs/genetics , Transcription Factors/genetics , Animals , Cells, Cultured , DNA-Binding Proteins/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Down-Regulation/genetics , Gene Expression Regulation , Glucose/pharmacology , Humans , Insulin/metabolism , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Islets of Langerhans/pathology , Mice , MicroRNAs/metabolism , Transcription Factors/metabolism
9.
Clin Med Insights Endocrinol Diabetes ; 14: 11795514211031204, 2021.
Article in English | MEDLINE | ID: mdl-34345195

ABSTRACT

Cystic fibrosis-related diabetes mellitus (CFRD) is the most common non-pulmonary co-morbidity in cystic fibrosis (CF). CF is caused by mutations in the cystic fibrosis transmembrane conductance regulator gene (CFTR), which leads to aberrant luminal fluid secretions in organs such as the lungs and pancreas. How dysfunctional CFTR leads to CFRD is still under debate. Both intrinsic effects of dysfunctional CFTR in hormone secreting cells of the islets and effects of exocrine damage have been proposed. In the current review, we discuss these non-mutually exclusive hypotheses with a special focus on how dysfunctional CFTR in endocrine cells may contribute to an altered glucose homeostasis. We outline the proposed role of CFTR in the molecular pathways of ß-cell insulin secretion and α-cell glucagon secretion, and touch upon the importance of the exocrine pancreas and intra-pancreatic crosstalk for proper islet function.

10.
Methods Mol Biol ; 2128: 25-54, 2020.
Article in English | MEDLINE | ID: mdl-32180184

ABSTRACT

The polygenic background of selectively bred diabetes models mimics the etiology of type 2 diabetes. So far, three different rodent models (Goto-Kakizaki rats, Nagoya-Shibata-Yasuda mice, and Oikawa-Nagao mice) have been established in the diabetes research field by continuous selective breeding for glucose tolerance from outbred rodent stocks. The origin of hyperglycemia in these rodents is mainly insulin secretion deficiency from the pancreatic ß-cells and mild insulin resistance in insulin target organs. In this chapter, we summarize backgrounds and phenotypes of these rodent models to highlight their importance in diabetes research. Then, we introduce experimental methodologies to evaluate ß-cell exocytosis as a putative common defect observed in these rodent models.


Subject(s)
Diabetes Mellitus, Experimental/genetics , Selective Breeding/genetics , Animals , Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Exocytosis , Gene Expression Profiling/methods , Glucose Intolerance , Insulin Resistance/physiology , Insulin Secretion/physiology , Insulin-Secreting Cells/chemistry , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/physiology , Mice , Mice, Inbred C3H , Patch-Clamp Techniques/methods , Phenotype , Rats , Rats, Wistar
11.
Diabetes ; 69(6): 1193-1205, 2020 06.
Article in English | MEDLINE | ID: mdl-32198214

ABSTRACT

Obesity is a risk factor for type 2 diabetes (T2D); however, not all obese individuals develop the disease. In this study, we aimed to investigate the cause of differential insulin secretion capacity of pancreatic islets from donors with T2D and non-T2D (ND), especially obese donors (BMI ≥30 kg/m2). Islets from obese donors with T2D had reduced insulin secretion, decreased ß-cell exocytosis, and higher expression of fatty acid translocase CD36. We tested the hypothesis that CD36 is a key molecule in the reduced insulin secretion capacity. Indeed, CD36 overexpression led to decreased insulin secretion, impaired exocytosis, and reduced granule docking. This was accompanied by reduced expression of the exocytotic proteins SNAP25, STXBP1, and VAMP2, likely because CD36 induced downregulation of the insulin receptor substrate (IRS) proteins, suppressed the insulin-signaling phosphatidylinositol 3-kinase/AKT pathway, and increased nuclear localization of the transcription factor FoxO1. CD36 antibody treatment of the human ß-cell line EndoC-ßH1 increased IRS1 and exocytotic protein levels, improved granule docking, and enhanced insulin secretion. Our results demonstrate that ß-cells from obese donors with T2D have dysfunctional exocytosis likely due to an abnormal lipid handling represented by differential CD36 expression. Hence, CD36 could be a key molecule to limit ß-cell function in T2D associated with obesity.


Subject(s)
CD36 Antigens/metabolism , Diabetes Mellitus, Type 2/etiology , Exocytosis/physiology , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Obesity/complications , Antibodies/pharmacology , CD36 Antigens/genetics , Cell Line , Gene Expression Regulation/drug effects , Humans , Islets of Langerhans/cytology
12.
PLoS One ; 15(2): e0228497, 2020.
Article in English | MEDLINE | ID: mdl-32017788

ABSTRACT

There are growing demands to ensure animal health and, from a broader perspective, animal welfare, especially for farmed animals. In addition to the newly developed welfare assessment protocols, which provide a harmonised method to measure animal health during farm visits, the question has been raised whether data from existing data collections can be used for an assessment without a prior farm visit. Here, we explore the possibilities of developing animal health scores for fattening pig herds using a) official meat inspection results, b) data on antibiotic usage and c) data from the QS (QS Qualität und Sicherheit GmbH) Salmonella monitoring programme in Germany. The objective is to aggregate and combine these register-like data into animal health scores that allow the comparison and benchmark of participating pig farms according to their health status. As the data combined in the scores have different units of measure and are collected in different abattoirs with possibly varying recording practices, we chose a relative scoring approach using z-transformations of different entrance variables. The final results are aggregated scores in which indicators are combined and weighted based on expert opinion according to their biological significance for animal health. Six scores have been developed to describe different focus areas, such as "Respiratory Health", "External Injuries/ Alterations", "Animal Management", "Antibiotic Usage", "Salmonella Status" and "Mortality". These "focus" area scores are finally combined into an "Overall Score". To test the scoring method, existing routine data from 1,747 pig farm units in Germany are used; these farm units are members of the QS Qualität und Sicherheit GmbH (QS) quality system. In addition, the scores are directly validated for 38 farm units. For these farm units, the farmers and their veterinarians provided their perceptions concerning the actual health status and existing health problems. This process allowed a comparison of the scoring results with actual health information using kappa coefficients as a measure of similarity. The score testing of the focus area scores using real information resulted in normalised data. The results of the validation showed satisfactory agreement between the calculated scores for the project farm units and the actual health information provided by the related farmers and veterinarians. In conclusion, the developed scoring method could become a viable benchmark and risk assessment instrument for animal health on a larger scale under the conditions of the German system.


Subject(s)
Animal Husbandry/methods , Anti-Bacterial Agents/therapeutic use , Drug Utilization/statistics & numerical data , Pork Meat/analysis , Salmonella Infections, Animal/prevention & control , Animal Welfare , Animals , Germany , Private Sector , Public Sector , Registries , Swine , Swine Diseases/microbiology , Swine Diseases/prevention & control
13.
Semin Cell Dev Biol ; 103: 41-50, 2020 07.
Article in English | MEDLINE | ID: mdl-31983511

ABSTRACT

The pancreatic islets of Langerhans consist of several hormone-secreting cell types important for blood glucose control. The insulin secreting ß-cells are the best studied of these cell types, but less is known about the glucagon secreting α-cells. The α-cells secrete glucagon as a response to low blood glucose. The major function of glucagon is to release glucose from the glycogen stores in the liver. In both type 1 and type 2 diabetes, glucagon secretion is dysregulated further exaggerating the hyperglycaemia, and in type 1 diabetes α-cells fail to counter regulate hypoglycaemia. Although glucagon has been recognized for almost 100 years, the understanding of how glucagon secretion is regulated and how glucagon act within the islet is far from complete. However, α-cell research has taken off lately which is promising for future knowledge. In this review we aim to highlight α-cell regulation and glucagon secretion with a special focus on recent discoveries from human islets. We will present some novel aspects of glucagon function and effects of selected glucose lowering agents on glucagon secretion.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Glucagon-Secreting Cells/metabolism , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Humans
14.
J Endocrinol ; 2019 Feb 01.
Article in English | MEDLINE | ID: mdl-30721137

ABSTRACT

Cystic fibrosis-related diabetes (CFRD) is a common complication for patients with cystic fibrosis (CF), a disease caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR). The cause of CFRD is unclear, but a commonly observed reduction in first-phase insulin secretion suggests defects at the beta cell level. Here we aimed to examine beta- and alpha-cell function in the Cftrtm1EUR/F508del mouse model (C57BL/6J), which carries the most common human mutation in CFTR, the F508del mutation. CFTR expression, beta cell mass, insulin granule distribution, hormone secretion and single cell capacitance changes were evaluated using islets (or beta cells) from F508del mice and age-matched wild-type mice aged 7-10 weeks. Granular pH was measured with DND-189 fluorescence. Serum glucose, insulin and glucagon levels were measured in vivo, and glucose tolerance was assessed using IPGTT. We show increased secretion of proinsulin and concomitant reduced secretion of C-peptide in islets from F508del mice compared to WT mice. Exocytosis and number of docked granules was reduced. We confirmed reduced granular pH by CFTR stimulation. We detected decreased pancreatic beta cell area, but unchanged beta cell number. Moreover, the F508del mutation caused failure to suppress glucagon secretion leading to hyperglucagonemia. In conclusion, F508del mice have beta cell defects resulting in 1) reduced number of docked insulin granules and reduced exocytosis, and 2) potential defective proinsulin cleavage and secretion of immature insulin. These observations provide insight into the functional role of CFTR in pancreatic islets and contribute to increased understanding of the pathogenesis of CFRD.

15.
Sci Rep ; 8(1): 16858, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30442920

ABSTRACT

Transient hepatic steatosis upon liver resection supposes functional relationships between lipid metabolism and liver regeneration. Repin1 has been suggested as candidate gene for obesity and dyslipidemia by regulating key genes of lipid metabolism and lipid storage. Herein, we characterized the regenerative potential of mice with a hepatic deletion of Repin1 (LRep1-/-) after partial hepatectomy (PH) in order to determine the functional significance of Repin1 in liver regeneration. Lipid dynamics and the regenerative response were analyzed at various time points after PH. Hepatic Repin1 deficiency causes a significantly decreased transient hepatic lipid accumulation. Defects in lipid uptake, as analyzed by decreased expression of the fatty acid transporter Cd36 and Fatp5, may contribute to attenuated and shifted lipid accumulation, accompanied by altered extent and chronological sequence of liver cell proliferation in LRep1-/- mice. In vitro steatosis experiments with primary hepatocytes also revealed attenuated lipid accumulation and occurrence of smaller lipid droplets in Repin1-deficient cells, while no direct effect on proliferation in HepG2 cells was observed. Based on these results, we propose that hepatocellular Repin1 might be of functional significance for early accumulation of lipids in hepatocytes after PH, facilitating efficient progression of liver regeneration.


Subject(s)
DNA-Binding Proteins/deficiency , Fatty Liver/metabolism , Liver Regeneration , Liver/metabolism , Organ Specificity , Animals , Cell Proliferation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fatty Acids/metabolism , Glycogen/metabolism , Hep G2 Cells , Hepatocytes/metabolism , Hepatocytes/ultrastructure , Humans , Lipid Metabolism , Liver/pathology , Liver/physiopathology , Liver/surgery , Liver Function Tests , Male , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins
16.
Diabetes Obes Metab ; 20 Suppl 2: 11-19, 2018 09.
Article in English | MEDLINE | ID: mdl-30230181

ABSTRACT

Pancreatic islet hormone secretion is central in the maintenance of blood glucose homeostasis. During development of hyperglycaemia, the ß-cell is under pressure to release more insulin to compensate for increased insulin resistance. Failure of the ß-cells to secrete enough insulin results in type 2 diabetes (T2D). MicroRNAs (miRNAs) are short non-coding RNA molecules suitable for rapid regulation of the changes in target gene expression needed in ß-cell adaptations. Moreover, miRNAs are involved in the maintenance of α-cell and ß-cell phenotypic identities via cell-specific, or cell-enriched expression. Although many of the abundant miRNAs are highly expressed in both cell types, recent research has focused on the role of miRNAs in ß-cells. It has been shown that highly abundant miRNAs, such as miR-375, are involved in several cellular functions indispensable in maintaining ß-cell phenotypic identity, almost acting as "housekeeping genes" in the context of hormone secretion. Despite the abundance and importance of miR-375, it has not been shown to be differentially expressed in T2D islets. On the contrary, the less abundant miRNAs such as miR-212/miR-132, miR-335, miR-130a/b and miR-152 are deregulated in T2D islets, wherein the latter three miRNAs were shown to play key roles in regulating ß-cell metabolism. In this review, we focus on ß-cell function and describe miRNAs involved in insulin biosynthesis and processing, glucose uptake and metabolism, electrical activity and Ca2+ -influx and exocytosis of the insulin granules. We present current status on miRNA regulation in α-cells, and finally we discuss the involvement of miRNAs in ß-cell dysfunction underlying T2D pathogenesis.


Subject(s)
Insulin-Secreting Cells/physiology , MicroRNAs/physiology , Animals , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Glucagon-Secreting Cells/metabolism , Glucagon-Secreting Cells/physiology , Humans , Insulin/biosynthesis , Insulin Secretion/physiology , Insulin-Secreting Cells/metabolism , Mice , Rats
17.
Curr Opin Pharmacol ; 43: 46-52, 2018 12.
Article in English | MEDLINE | ID: mdl-30144686

ABSTRACT

Failure of the ß-cell to secrete enough insulin is a major contributing factor in the pathogenesis of type-2 diabetes (T2D). MicroRNAs provide an extra layer in the regulation of protein expression, and are thus involved in ß-cell compensation during development of the disease. In this review, we discuss how microRNAs can regulate their target protein expression and phenotypic output, present the status of nutritional regulation of microRNA expression, and summarize work on microRNA expression in human islets. In conclusion, current data lend support to microRNAs being essential regulators of insulin secretion. Future work will describe microRNAs in α-cell function, details of the microRNA-mRNA network, and possibilities to use microRNAs as biomarkers and in therapeutic treatment of T2D and complications.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 2/metabolism , Insulin/blood , Islets of Langerhans/metabolism , MicroRNAs/metabolism , Animals , Biomarkers/blood , Blood Glucose/genetics , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/therapy , Diffusion of Innovation , Gene Expression Regulation , Genetic Therapy/methods , Humans , MicroRNAs/genetics , MicroRNAs/therapeutic use , Signal Transduction
18.
Environ Geochem Health ; 40(2): 865-885, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29027593

ABSTRACT

To understand how extraction of different energy sources impacts water resources requires assessment of how water chemistry has changed in comparison with the background values of pristine streams. With such understanding, we can develop better water quality standards and ecological interpretations. However, determination of pristine background chemistry is difficult in areas with heavy human impact. To learn to do this, we compiled a master dataset of sulfate and barium concentrations ([SO4], [Ba]) in Pennsylvania (PA, USA) streams from publically available sources. These elements were chosen because they can represent contamination related to oil/gas and coal, respectively. We applied changepoint analysis (i.e., likelihood ratio test) to identify pristine streams, which we defined as streams with a low variability in concentrations as measured over years. From these pristine streams, we estimated the baseline concentrations for major bedrock types in PA. Overall, we found that 48,471 data values are available for [SO4] from 1904 to 2014 and 3243 data for [Ba] from 1963 to 2014. Statewide [SO4] baseline was estimated to be 15.8 ± 9.6 mg/L, but values range from 12.4 to 26.7 mg/L for different bedrock types. The statewide [Ba] baseline is 27.7 ± 10.6 µg/L and values range from 25.8 to 38.7 µg/L. Results show that most increases in [SO4] from the baseline occurred in areas with intensive coal mining activities, confirming previous studies. Sulfate inputs from acid rain were also documented. Slight increases in [Ba] since 2007 and higher [Ba] in areas with higher densities of gas wells when compared to other areas could document impacts from shale gas development, the prevalence of basin brines, or decreases in acid rain and its coupled effects on [Ba] related to barite solubility. The largest impacts on PA stream [Ba] and [SO4] are related to releases from coal mining or burning rather than oil and gas development.


Subject(s)
Acid Rain , Barium/analysis , Coal Mining , Hydraulic Fracking , Natural Gas , Rivers , Sulfates/analysis , Water Pollutants, Chemical/analysis , Appalachian Region , Datasets as Topic , Geology , Human Activities , Humans , Pennsylvania , Time Factors
19.
Physiol Rep ; 5(21)2017 Nov.
Article in English | MEDLINE | ID: mdl-29122960

ABSTRACT

MicroRNAs contribute to the maintenance of optimal cellular functions by fine-tuning protein expression levels. In the pancreatic ß-cells, imbalances in the exocytotic machinery components lead to impaired insulin secretion and type 2 diabetes (T2D). We hypothesize that dysregulated miRNA expression exacerbates ß-cell dysfunction, and have earlier shown that islets from the diabetic GK-rat model have increased expression of miRNAs, including miR-335-5p (miR-335). Here, we aim to determine the specific role of miR-335 during development of T2D, and the influence of this miRNA on glucose-stimulated insulin secretion and Ca2+-dependent exocytosis. We found that the expression of miR-335 negatively correlated with secretion index in human islets of individuals with prediabetes. Overexpression of miR-335 in human EndoC-ßH1 and in rat INS-1 832/13 cells (OE335) resulted in decreased glucose-stimulated insulin secretion, and OE335 cells showed concomitant reduction in three exocytotic proteins: SNAP25, Syntaxin-binding protein 1 (STXBP1), and synaptotagmin 11 (SYT11). Single-cell capacitance measurements, complemented with TIRF microscopy of the granule marker NPY-mEGFP demonstrated a significant reduction in exocytosis in OE335 cells. The reduction was not associated with defective docking or decreased Ca2+ current. More likely, it is a direct consequence of impaired priming of already docked granules. Earlier reports have proposed reduced granular priming as the cause of reduced first-phase insulin secretion during prediabetes. Here, we show a specific role of miR-335 in regulating insulin secretion during this transition period. Moreover, we can conclude that miR-335 has the capacity to modulate insulin secretion and Ca2+-dependent exocytosis through effects on granular priming.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Insulin/metabolism , MicroRNAs/metabolism , Animals , Cell Line , Disease Models, Animal , Exocytosis , Humans , Insulin Secretion , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Munc18 Proteins/metabolism , Rats , Synaptosomal-Associated Protein 25/metabolism , Synaptotagmins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL