Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 9.998
Filter
1.
Biomolecules ; 14(6)2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38927074

ABSTRACT

Diabetes imposes a huge burden worldwide. Islet transplantation is an alternative therapy for diabetes. However, tacrolimus, a kind of immunosuppressant after organ transplantation, is closely related to post-transplant diabetes mellitus. Mesenchymal stem cells (MSCs) have attracted interest for their potential to alleviate diabetes. In vivo experiments revealed that human menstrual blood-derived stem cells (MenSCs) treatment improved tacrolimus-induced blood glucose, body weight, and glucose tolerance disorders in mice. RNA sequencing was used to analyze the potential therapeutic targets of MenSCs. In this study, we illustrated that cystathionine ß-synthase (CBS) contributed to tacrolimus -induced islet dysfunction. Using ß-cell lines (MIN6, ß-TC-6), we demonstrated that MenSCs ameliorated tacrolimus-induced islet dysfunction in vitro. Moreover, MenSC reduced the tacrolimus-induced elevation of CBS levels and significantly enhanced the viability, anti-apoptotic ability, glucose-stimulated insulin secretion (GSIS), and glycolytic flux of ß-cells. We further revealed that MenSCs exerted their therapeutic effects by inhibiting CBS expression to activate the IL6/JAK2/STAT3 pathway. In conclusion, we showed that MenSCs may be a potential strategy to improve tacrolimus-induced islet dysfunction.


Subject(s)
Cystathionine beta-Synthase , Interleukin-6 , STAT3 Transcription Factor , Tacrolimus , Humans , STAT3 Transcription Factor/metabolism , Tacrolimus/pharmacology , Interleukin-6/metabolism , Animals , Mice , Female , Cystathionine beta-Synthase/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Janus Kinase 2/metabolism , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Menstruation/blood , Menstruation/drug effects , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/cytology , Signal Transduction/drug effects , Insulin Secretion/drug effects , Cell Line
2.
Biochem Biophys Res Commun ; 725: 150254, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-38901223

ABSTRACT

Decreased pancreatic ß-cell volume is a serious problem in patients with type 2 diabetes mellitus, and there is a need to establish appropriate treatments. Increasingly, sodium/glucose cotransporter 2 (SGLT2) inhibitors, which have a protective effect on pancreatic ß-cells, are being prescribed to treat diabetes; however, the underlying mechanism is not well understood. We previously administered SGLT2 inhibitor dapagliflozin to a mouse model of type 2 diabetes and found significant changes in gene expression in the early-treated group, which led us to hypothesize that epigenetic regulation was a possible mechanism of these changes. Therefore, we performed comprehensive DNA methylation analysis by methylated DNA immunoprecipitation using isolated pancreatic islets after dapagliflozin administration to diabetic model mice. As a result, we identified 31 genes with changes in expression due to DNA methylation changes. Upon immunostaining, cystic fibrosis transmembrane conductance regulator and cadherin 24 were found to be upregulated in islets in the dapagliflozin-treated group. These molecules may contribute to the maintenance of islet morphology and insulin secretory capacity, suggesting that SGLT2 inhibitors' protective effect on pancreatic ß-cells is accompanied by DNA methylation changes, and that the effect is long-term and not temporary. In future diabetes care, SGLT2 inhibitors may be expected to have positive therapeutic effects, including pancreatic ß-cell protection.


Subject(s)
Benzhydryl Compounds , DNA Methylation , Diabetes Mellitus, Type 2 , Glucosides , Islets of Langerhans , Sodium-Glucose Transporter 2 Inhibitors , Animals , Benzhydryl Compounds/pharmacology , Benzhydryl Compounds/therapeutic use , DNA Methylation/drug effects , Glucosides/pharmacology , Glucosides/therapeutic use , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/pathology , Mice , Islets of Langerhans/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/pathology , Male , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Mice, Inbred C57BL , Disease Models, Animal , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/pathology , Epigenesis, Genetic/drug effects , Gene Expression Regulation/drug effects , Cadherins/metabolism , Cadherins/genetics
3.
Sci Adv ; 10(23): eadk3081, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38848367

ABSTRACT

Clinical outcomes for total-pancreatectomy followed by intraportal islet autotransplantation (TP-IAT) to treat chronic pancreatitis (CP) are suboptimal due to pancreas inflammation, oxidative stress during islet isolation, and harsh engraftment conditions in the liver's vasculature. We describe a thermoresponsive, antioxidant macromolecule poly(polyethylene glycol citrate-co-N-isopropylacrylamide) (PPCN) to protect islet redox status and function and to enable extrahepatic omentum islet engraftment. PPCN solution transitions from a liquid to a hydrogel at body temperature. Islets entrapped in PPCN and exposed to oxidative stress remain functional and support long-term euglycemia, in contrast to islets entrapped in a plasma-thrombin biologic scaffold. In the nonhuman primate (NHP) omentum, PPCN is well-tolerated and mostly resorbed without fibrosis at 3 months after implantation. In NHPs, autologous omentum islet transplantation using PPCN restores normoglycemia with minimal exogenous insulin requirements for >100 days. This preclinical study supports TP-IAT with PPCN in patients with CP and highlights antioxidant properties as a mechanism for islet function preservation.


Subject(s)
Islets of Langerhans Transplantation , Islets of Langerhans , Omentum , Oxidative Stress , Islets of Langerhans Transplantation/methods , Omentum/metabolism , Animals , Islets of Langerhans/metabolism , Islets of Langerhans/drug effects , Oxidative Stress/drug effects , Citric Acid/pharmacology , Humans , Antioxidants/pharmacology , Pancreatitis, Chronic/metabolism , Pancreatitis, Chronic/surgery , Pancreatitis, Chronic/pathology , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Male , Phase Transition
4.
Int J Mol Sci ; 25(11)2024 May 31.
Article in English | MEDLINE | ID: mdl-38892240

ABSTRACT

A detailed study of palmitate metabolism in pancreatic islets subject to different experimental conditions, like varying concentrations of glucose, as well as fed or starved conditions, has allowed us to explore the interaction between the two main plasma nutrients and its consequences on hormone secretion. Palmitate potentiates glucose-induced insulin secretion in a concentration-dependent manner, in a physiological range of both palmitate (0-2 mM) and glucose (6-20 mM) concentrations; at glucose concentrations lower than 6 mM, no metabolic interaction with palmitate was apparent. Starvation (48 h) increased islet palmitate oxidation two-fold, and the effect was resistant to its inhibition by glucose (6-20 mM). Consequently, labelled palmitate and glucose incorporation into complex lipids were strongly suppressed, as well as glucose-induced insulin secretion and its potentiation by palmitate. 2-bromostearate, a palmitate oxidation inhibitor, fully recovered the synthesis of complex lipids and insulin secretion. We concluded that palmitate potentiation of the insulin response to glucose is not attributable to its catabolic mitochondrial oxidation but to its anabolism to complex lipids: islet lipid biosynthesis is dependent on the uptake of plasma fatty acids and the supply of α-glycerol phosphate from glycolysis. Islet secretion of glucagon and somatostatin showed a similar dependence on palmitate anabolism as insulin. The possible mechanisms implicated in the metabolic coupling between glucose and palmitate were commented on. Moreover, possible mechanisms responsible for islet gluco- or lipotoxicity after a long-term stimulation of insulin secretion were also discussed. Our own data on the simultaneous stimulation of insulin, glucagon, and somatostatin by glucose, as well as their modification by 2-bromostearate in perifused rat islets, give support to the conclusion that increased FFA anabolism, rather than its mitochondrial oxidation, results in a potentiation of their stimulated release. Starvation, besides suppressing glucose stimulation of insulin secretion, also blocks the inhibitory effect of glucose on glucagon secretion: this suggests that glucagon inhibition might be an indirect or direct effect of insulin, but not of glucose. In summary, there seems to exist three mechanisms of glucagon secretion stimulation: 1. glucagon stimulation through the same secretion coupling mechanism as insulin, but in a different range of glucose concentrations (0 to 5 mM). 2. Direct or indirect inhibition by secreted insulin in response to glucose (5-20 mM). 3. Stimulation by increased FFA anabolism in glucose intolerance or diabetes in the context of hyperlipidemia, hyperglycemia, and hypo-insulinemia. These conclusions were discussed and compared with previous published data in the literature. Specially, we discussed the mechanism for inhibition of glucagon release by glucose, which was apparently contradictory with the secretion coupling mechanism of its stimulation.


Subject(s)
Glucagon , Glucose , Insulin Secretion , Insulin , Islets of Langerhans , Glucose/metabolism , Animals , Insulin/metabolism , Glucagon/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans/drug effects , Insulin Secretion/drug effects , Fatty Acids/metabolism , Rats , Palmitates/metabolism , Palmitates/pharmacology , Oxidation-Reduction/drug effects
5.
Nutrients ; 16(12)2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38931174

ABSTRACT

Zinc deficiency has been associated with the worsening of diabetes while zinc supplementation has been proposed to ameliorate diabetes. This study examined the effects of marginal zinc deficiency (MZD) and zinc supplementation (ZS) on obesity, glycemic control, pancreatic islets, hepatic steatosis and renal function of Zucker diabetic fatty (ZDF) rats. Male ZDF rats were fed an MZD, zinc control (ZC) or ZS diet (4, 30 and 300 mg Zn/kg diet, respectively), and lean Zucker rats were fed a ZC diet for 8 weeks. MZD and ZS did not alter body weight or whole-body composition in ZDF rats. MZD ZDF rats had reduced zinc concentrations in the femur and pancreas, a greater number of enlarged pancreatic islets and a diminished response to an oral glucose load based on a 1.8-fold greater incremental area-under-the-curve (AUC) for glucose compared to ZC ZDF. ZS ZDF rats had elevated serum, femur and pancreatic zinc concentrations, unchanged pancreatic parameters and a 50% reduction in the AUC for insulin compared to ZC ZDF rats, suggesting greater insulin sensitivity. Dietary zinc intake did not alter hepatic steatosis, creatinine clearance, or levels of proteins that contribute to insulin signaling, inflammation or zinc transport in epididymal fat. Potential adverse effects of ZS were suggested by reduced hepatic copper concentrations and elevated serum urea compared to ZC ZDF rats. In summary, ZS improved the pancreatic insulin response but not the glucose handling. In contrast, reduced zinc status in ZDF rats led to impaired glucose tolerance and a compensatory increase in the number and size of pancreatic islets which could lead to ß-cell exhaustion.


Subject(s)
Dietary Supplements , Insulin , Islets of Langerhans , Rats, Zucker , Zinc , Animals , Zinc/deficiency , Male , Insulin/blood , Islets of Langerhans/metabolism , Islets of Langerhans/drug effects , Rats , Blood Glucose/metabolism , Obesity/metabolism , Insulin Resistance , Pancreas/metabolism , Pancreas/drug effects , Liver/metabolism , Liver/drug effects , Diabetes Mellitus, Experimental/drug therapy
6.
Am J Physiol Endocrinol Metab ; 327(1): E27-E41, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38690938

ABSTRACT

Pancreatic endocrine cells employ a sophisticated system of paracrine and autocrine signals to synchronize their activities, including glutamate, which controls hormone release and ß-cell viability by acting on glutamate receptors expressed by endocrine cells. We here investigate whether alteration of the excitatory amino acid transporter 2 (EAAT2), the major glutamate clearance system in the islet, may occur in type 2 diabetes mellitus and contribute to ß-cell dysfunction. Increased EAAT2 intracellular localization was evident in islets of Langerhans from T2DM subjects as compared with healthy control subjects, despite similar expression levels. Chronic treatment of islets from healthy donors with high-glucose concentrations led to the transporter internalization in vesicular compartments and reduced [H3]-d-glutamate uptake (65 ± 5% inhibition), phenocopying the findings in T2DM pancreatic sections. The transporter relocalization was associated with decreased Akt phosphorylation protein levels, suggesting an involvement of the phosphoinositide 3-kinase (PI3K)/Akt pathway in the process. In line with this, PI3K inhibition by a 100-µM LY294002 treatment in human and clonal ß-cells caused the transporter relocalization in intracellular compartments and significantly reduced the glutamate uptake compared to control conditions, suggesting that hyperglycemia changes the trafficking of the transporter to the plasma membrane. Upregulation of the glutamate transporter upon treatment with the antibiotic ceftriaxone rescued hyperglycemia-induced ß-cells dysfunction and death. Our data underscore the significance of EAAT2 in regulating islet physiology and provide a rationale for potential therapeutic targeting of this transporter to preserve ß-cell survival and function in diabetes.NEW & NOTEWORTHY The glutamate transporter SLC1A2/excitatory amino acid transporter 2 (EAAT2) is expressed on the plasma membrane of pancreatic ß-cells and controls islet glutamate clearance and ß-cells survival. We found that the EAAT2 membrane expression is lost in the islets of Langerhans from type 2 diabetes mellitus (T2DM) patients due to hyperglycemia-induced downregulation of the phosphoinositide 3-kinase/Akt pathway and modification of its intracellular trafficking. Pharmacological rescue of EAAT2 expression prevents ß-cell dysfunction and death, suggesting EAAT2 as a new potential target of intervention in T2DM.


Subject(s)
Diabetes Mellitus, Type 2 , Excitatory Amino Acid Transporter 2 , Glutamic Acid , Hyperglycemia , Islets of Langerhans , Excitatory Amino Acid Transporter 2/metabolism , Humans , Diabetes Mellitus, Type 2/metabolism , Glutamic Acid/metabolism , Hyperglycemia/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans/drug effects , Male , Middle Aged , Female , Protein Transport , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Aged , Adult , Animals , Phosphatidylinositol 3-Kinases/metabolism
7.
Am J Physiol Endocrinol Metab ; 327(1): E103-E110, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38775725

ABSTRACT

The incretin axis is an essential component of postprandial insulin secretion and glucose homeostasis. There are two incretin hormones, glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP), which exert multiple actions throughout the body. A key cellular target for the incretins are pancreatic ß-cells, where they potentiate nutrient-stimulated insulin secretion. This feature of incretins has made this system an attractive target for therapeutic interventions aimed at controlling glycemia. Here, we discuss the role of GIP in both ß-cells and α-cells within the islet, to stimulate insulin and glucagon secretion, respectively. Moreover, we discuss how glucagon secretion from α-cells has important insulinotropic actions in ß-cells through an axis termed α- to ß-cell communication. These recent advances have elevated the potential of GIP and glucagon as a therapeutic targets, coinciding with emerging compounds that pharmacologically leverage the actions of these two peptides in the context of diabetes and obesity.


Subject(s)
Gastric Inhibitory Polypeptide , Glucagon , Insulin Secretion , Insulin , Islets of Langerhans , Gastric Inhibitory Polypeptide/metabolism , Humans , Glucagon/metabolism , Insulin Secretion/physiology , Animals , Insulin/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans/drug effects , Insulin-Secreting Cells/metabolism , Glucagon-Secreting Cells/metabolism , Incretins/metabolism , Glucagon-Like Peptide 1/metabolism
8.
Peptides ; 178: 171254, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38815655

ABSTRACT

The incretin hormones, glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP), are rapidly degraded by dipeptidyl peptidase-4 (DPP-4) to their major circulating metabolites GLP-1(9-36) and GIP(3-42). This study investigates the possible effects of these metabolites, and the equivalent exendin molecule Ex(9-39), on pancreatic islet morphology and constituent alpha and beta cells in high-fat diet (HFD) fed mice. Male Swiss TO-mice (6-8 weeks-old) were maintained on a HFD or normal diet (ND) for 4 months and then received twice-daily subcutaneous injections of GLP-1(9-36), GIP(3-42), Ex(9-39) (25 nmol/kg bw) or saline vehicle (0.9% (w/v) NaCl) over a 60-day period. Metabolic parameters were monitored and excised pancreatic tissues were used for immunohistochemical analysis. Body weight and assessed metabolic indices were not changed by peptide administration. GLP-1(9-36) significantly (p<0.001) increased islet density per mm2 tissue, that was decreased (p<0.05) by HFD. Islet, beta and alpha cell areas were increased (p<0.01) following HFD and subsequently reduced (p<0.01-p<0.001) by GIP(3-42) and Ex(9-39) treatment. While GLP-1(9-36) did not affect islet and beta cell areas in HFD mice, it significantly (p<0.01) decreased alpha cell area. Compared to ND and HFD mice, GIP(3-42) treatment significantly (p<0.05) increased beta cell proliferation. Whilst HFD increased (p<0.001) beta cell apoptosis, this was reduced (p<0.01-p<0.001) by both GLP-1(9-36) and GIP(3-42). These data indicate that the major circulating forms of GLP-1 and GIP, namely GLP-1(9-36) and GIP(3-42) previously considered largely inactive, may directly impact pancreatic morphology, with an important protective effect on beta cell health under conditions of beta cell stress.


Subject(s)
Diet, High-Fat , Gastric Inhibitory Polypeptide , Glucagon-Like Peptide 1 , Incretins , Insulin-Secreting Cells , Animals , Gastric Inhibitory Polypeptide/pharmacology , Gastric Inhibitory Polypeptide/metabolism , Male , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide 1/pharmacology , Mice , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Diet, High-Fat/adverse effects , Incretins/pharmacology , Incretins/metabolism , Peptide Fragments/pharmacology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Apoptosis/drug effects , Insulin/metabolism
9.
Int J Biol Macromol ; 271(Pt 2): 132376, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38750865

ABSTRACT

Diabetes is a complex metabolic disease and islet transplantation is a promising approach for the treatment of diabetes. Unfortunately, the transplanted islets at the subcutaneous site are also affected by various adverse factors such as poor vascularization and hypoxia. In this study, we utilize biocompatible copolymers l-lactide and D,l-lactide to manufacture a biomaterial scaffold with a mesh-like structure via 3D printing technology, providing a material foundation for encapsulating pancreatic islet cells. The scaffold maintains the sustained release of vascular endothelial growth factor (VEGF) and a slow release of oxygen from calcium peroxide (CPO), thereby regulating the microenvironment for islet survival. This helps to improve insufficient subcutaneous vascularization and reduce islet death due to hypoxia post-transplantation. By pre-implanting VEGF-CPO scaffolds subcutaneously into diabetic rats, a sufficiently vascularized site is formed, thereby ensuring early survival of transplanted islets. In a word, the VEGF-CPO scaffold shows good biocompatibility both in vitro and in vivo, avoids the adverse effects on the implanted islets, and displays promising clinical transformation prospects.


Subject(s)
Biocompatible Materials , Diabetes Mellitus, Experimental , Islets of Langerhans Transplantation , Islets of Langerhans , Printing, Three-Dimensional , Tissue Scaffolds , Vascular Endothelial Growth Factor A , Animals , Tissue Scaffolds/chemistry , Rats , Islets of Langerhans Transplantation/methods , Vascular Endothelial Growth Factor A/metabolism , Diabetes Mellitus, Experimental/therapy , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Islets of Langerhans/drug effects , Islets of Langerhans/blood supply , Islets of Langerhans/metabolism , Male , Neovascularization, Physiologic/drug effects , Rats, Sprague-Dawley , Peroxides
10.
Endocrinology ; 165(6)2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38626085

ABSTRACT

Reducing ghrelin by ghrelin gene knockout (GKO), ghrelin-cell ablation, or high-fat diet feeding increases islet size and ß-cell mass in male mice. Here we determined if reducing ghrelin also enlarges islets in females and if pregnancy-associated changes in islet size are related to reduced ghrelin. Islet size and ß-cell mass were larger (P = .057 for ß-cell mass) in female GKO mice. Pregnancy was associated with reduced ghrelin and increased liver-expressed antimicrobial peptide-2 (LEAP2; a ghrelin receptor antagonist) in wild-type mice. Ghrelin deletion and pregnancy each increased islet size (by ∼19.9-30.2% and ∼34.9-46.4%, respectively), percentage of large islets (>25 µm2×103, by ∼21.8-42% and ∼21.2-41.2%, respectively), and ß-cell mass (by ∼15.7-23.8% and ∼65.2-76.8%, respectively). Neither islet cross-sectional area, ß-cell cross-sectional area, nor ß-cell mass correlated with plasma ghrelin, although all positively correlated with LEAP2 (P = .081 for islet cross-sectional area). In ad lib-fed mice, there was an effect of pregnancy, but not ghrelin deletion, to change (raise) plasma insulin without impacting blood glucose. Similarly, there was an effect of pregnancy, but not ghrelin deletion, to change (lower) blood glucose area under the curve during a glucose tolerance test. Thus, genetic deletion of ghrelin increases islet size and ß-cell cross-sectional area in female mice, similar to males. Yet, despite pregnancy-associated reductions in ghrelin, other factors appear to govern islet enlargement and changes to insulin sensitivity and glucose tolerance in the setting of pregnancy. In the case of islet size and ß-cell mass, one of those factors may be the pregnancy-associated increase in LEAP2.


Subject(s)
Ghrelin , Islets of Langerhans , Animals , Female , Male , Mice , Pregnancy , Antimicrobial Cationic Peptides , Blood Glucose/metabolism , Ghrelin/metabolism , Insulin/metabolism , Insulin/blood , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Mice, Inbred C57BL , Mice, Knockout , Organ Size/drug effects
11.
J Pharm Pharmacol ; 76(7): 861-872, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38652540

ABSTRACT

OBJECTIVES: Dopamine and related receptors are evidenced in pancreatic endocrine tissue, but the impact on islet ß-cell stimulus-secretion as well as (patho)physiological role are unclear. METHODS: The present study has evaluated islet cell signalling pathways and biological effects of dopamine, as well as alterations of islet dopamine in rodent models of diabetes of different aetiology. KEY FINDINGS: The dopamine precursor l-DOPA partially impaired glucose tolerance in mice and attenuated glucose-, exendin-4, and alanine-induced insulin secretion. The latter effect was echoed by the attenuation of glucose-induced [Ca2+]i dynamics and elevation of ATP levels in individual mouse islet cells. l-DOPA significantly decreased ß-cell proliferation rates, acting predominantly via the D2 receptor, which was most abundant at the mRNA level. The administration of streptozotocin (STZ) or high-fat diet (HFD) in mice significantly elevated numbers of dopamine-positive islet cells, with HFD also increasing colocalization of dopamine with insulin. At the same time, colocalization of dopamine with glucagon was increased in STZ-treated and pregnant mice, but unaffected by HFD. CONCLUSION: These findings highlight a role for dopamine receptor signalling in islet cell biology adaptations to various forms of metabolic stress.


Subject(s)
Diet, High-Fat , Dopamine , Insulin , Signal Transduction , Animals , Dopamine/metabolism , Mice , Signal Transduction/drug effects , Female , Male , Insulin/metabolism , Stress, Physiological , Islets of Langerhans/metabolism , Islets of Langerhans/drug effects , Levodopa/pharmacology , Diabetes Mellitus, Experimental/metabolism , Mice, Inbred C57BL , Receptors, Dopamine D2/metabolism , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/drug effects , Pregnancy , Cell Proliferation/drug effects , Adaptation, Physiological , Streptozocin , Exenatide/pharmacology , Glucagon/metabolism , Glucose/metabolism , Insulin Secretion/drug effects
12.
Cell Transplant ; 33: 9636897241246577, 2024.
Article in English | MEDLINE | ID: mdl-38646716

ABSTRACT

Calcineurin inhibitors (CNIs) are critical in preventing rejection posttransplantation but pose an increased risk of post-transplant diabetes (PTD). Recent studies show that late conversion from CNIs to belatacept, a costimulation blocker, improves HbA1c in kidney transplant recipients with PTD or de novo diabetes. This study investigates whether the observed effects on PTD stem solely from CNI withdrawal or if belatacept influences PTD independently. The study assessed the impact of tacrolimus and belatacept on insulin secretion in MIN6 cells (a beta cell line) and rat islets. Tacrolimus and belatacept were administered to the cells and islets, followed by assessments of cell viability and insulin secretion. Tacrolimus impaired insulin secretion without affecting cell viability, while belatacept showed no detrimental effects on either parameter. These findings support clinical observations of improved HbA1c upon switching from tacrolimus to belatacept. Belatacept holds promise in islet or pancreas transplantation, particularly in patients with unstable diabetes. Successful cases of islet transplantation treated with belatacept without severe hypoglycemia highlight its potential in managing PTD. Further research is needed to fully understand the metabolic changes accompanying the transition from CNIs to belatacept. Preserving insulin secretion emerges as a promising avenue for investigation in this context.


Subject(s)
Abatacept , Immunosuppressive Agents , Insulin , Tacrolimus , Tacrolimus/therapeutic use , Tacrolimus/pharmacology , Abatacept/therapeutic use , Abatacept/pharmacology , Animals , Rats , Insulin/metabolism , Immunosuppressive Agents/therapeutic use , Immunosuppressive Agents/pharmacology , Humans , Male , Insulin Secretion/drug effects , Mice , Islets of Langerhans Transplantation/methods , Cell Line , Cell Survival/drug effects , Diabetes Mellitus/drug therapy , Diabetes Mellitus/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism
13.
Cells ; 13(8)2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38667300

ABSTRACT

Interleukin-6 (IL6) is a pleiotropic cytokine implicated in metabolic disorders and inflammation, yet its precise influence on insulin secretion and glucose metabolism remains uncertain. This study examined IL6 expression in pancreatic islets from individuals with/without diabetes, alongside a series of functional experiments, including siRNA silencing; IL6 treatment; and assessments of glucose uptake, cell viability, apoptosis, and expression of key ß-cell genes, which were conducted in both INS-1 cells and human islets to elucidate the effect of IL6 on insulin secretion. Serum levels of IL6 from Emirati patients with type 2 diabetes (T2D) were measured, and the effect of antidiabetic drugs on IL6 levels was studied. The results revealed that IL6 mRNA expression was higher in islets from diabetic and older donors compared to healthy or young donors. IL6 expression correlated negatively with PDX1, MAFB, and NEUROD1 and positively with SOX4, HES1, and FOXA1. Silencing IL6 in INS-1 cells reduced insulin secretion and glucose uptake independently of apoptosis or oxidative stress. Reduced expression of IL6 was associated with the downregulation of Ins, Pdx1, Neurod1, and Glut2 in INS-1 cells. In contrast, IL6 treatment enhanced insulin secretion in INS-1 cells and human islets and upregulated insulin expression. Serum IL6 levels were elevated in patients with T2D and associated with higher glucose, HbA1c, and triglycerides, regardless of glucose-lowering medications. This study provides a new understanding of the role of IL6 in ß-cell function and the pathophysiology of T2D. Our data highlight differences in the response to IL6 between INS-1 cells and human islets, suggesting the presence of species-specific variations across different experimental models. Further research is warranted to unravel the precise mechanisms underlying the observed effects of IL-6 on insulin secretion.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Secretion , Interleukin-6 , Islets of Langerhans , Humans , Interleukin-6/metabolism , Interleukin-6/blood , Insulin Secretion/drug effects , Islets of Langerhans/metabolism , Islets of Langerhans/drug effects , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/blood , Male , Middle Aged , Female , Adult , Glucose/metabolism , Insulin/metabolism , Insulin/blood , Rats , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/drug effects , Animals , Cell Line , Aged , Apoptosis/drug effects
14.
Am J Physiol Endocrinol Metab ; 326(5): E673-E680, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38446636

ABSTRACT

Residual beta cells are present in most patients with longstanding type 1 diabetes but it is unknown whether these beta cells react normally to different stimuli. Moreover a defect in proinsulin conversion and abnormal alpha cell response are also part of the islet dysfunction. A three-phase [euglycemia, hyperglycemia, and hyperglycemia + glucagon-like peptide 1 (GLP-1)] clamp was performed in patients with longstanding type 1 diabetes. Intravenous arginine boluses were administered at the end of each phase. On another day, a mixed meal stimulation test with a subsequent intravenous arginine bolus was performed. C-peptide was detectable in a subgroup of subjects at baseline (2/15) or only after stimulation (3/15). When detectable, C-peptide increased 2.9-fold [95% CI: 1.2-7.1] during the hyperglycemia phase and 14.1-fold [95% CI: 3.1-65.2] during the hyperglycemia + GLP-1 phase, and 22.3-fold [95% CI: 5.6-89.1] during hyperglycemia + GLP-1 + arginine phase when compared with baseline. The same subset of patients with a C-peptide response were identified during the mixed meal stimulation test as during the clamp. There was an inhibition of glucagon secretion (0.72-fold, [95% CI: 0.63-0.84]) during the glucose clamp irrespective of the presence of detectable beta cell function. Proinsulin was only present in a subset of subjects with detectable C-peptide (3/15) and proinsulin mimicked the C-peptide response to the different stimuli when detectable. Residual beta cells in longstanding type 1 diabetes respond adequately to different stimuli and could be of clinical benefit.NEW & NOTEWORTHY If beta cell function is detectable, the beta cells react relatively normal to the different stimuli except for the first phase response to intravenous glucose. An oral mixed meal followed by an intravenous arginine bolus can identify residual beta cell function/mass as well as the more commonly used glucose potentiated arginine-induced insulin secretion during a hyperglycemic clamp.


Subject(s)
Arginine , C-Peptide , Diabetes Mellitus, Type 1 , Food, Formulated , Glucagon-Like Peptide 1 , Glucose , Islets of Langerhans , Adult , Female , Humans , Male , Middle Aged , Arginine/administration & dosage , Arginine/pharmacology , Blood Glucose/metabolism , C-Peptide/blood , C-Peptide/metabolism , Diabetes Mellitus, Type 1/metabolism , Glucagon/metabolism , Glucagon-Like Peptide 1/administration & dosage , Glucagon-Like Peptide 1/metabolism , Glucose/administration & dosage , Glucose/metabolism , Glucose Clamp Technique , Hyperglycemia/metabolism , Insulin/metabolism , Insulin/administration & dosage , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/physiology , Islets of Langerhans/metabolism , Islets of Langerhans/drug effects
15.
Mol Cell Endocrinol ; 588: 112202, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38552943

ABSTRACT

Developmental exposure to endocrine disruptors like bisphenol A (BPA) are implicated in later-life metabolic dysfunction. Leveraging a unique sheep model of developmental programming, we conducted an exploratory analysis of the programming effects of BPA on the endocrine pancreas. Pregnant ewes were administered environmentally relevant doses of BPA during gestational days (GD) 30-90, and pancreata from female fetuses and adult offspring were analyzed. Prenatal BPA exposure induced a trend toward decreased islet insulin staining and ß-cell count, increased glucagon staining and α-cell count, and increased α-cell/ß-cell ratio. Findings were most consistent in fetal pancreata assessed at GD90 and in adult offspring exposed to the lowest BPA dose. While not assessed in fetuses, adult islet fibrosis was increased. Collectively, these data provide further evidence that early-life BPA exposure is a likely threat to human metabolic health. Future studies should corroborate these findings and decipher the molecular mechanisms of BPA's developmental endocrine toxicity.


Subject(s)
Benzhydryl Compounds , Islets of Langerhans , Phenols , Prenatal Exposure Delayed Effects , Animals , Benzhydryl Compounds/toxicity , Female , Phenols/toxicity , Pregnancy , Sheep , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/pathology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Endocrine Disruptors/toxicity , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Maternal Exposure/adverse effects , Insulin/metabolism , Fetus/drug effects , Glucagon-Secreting Cells/drug effects , Glucagon-Secreting Cells/metabolism , Glucagon-Secreting Cells/pathology
16.
Cells ; 13(5)2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38474380

ABSTRACT

Islet transplantation is a therapeutic option to replace ß-cell mass lost during type 1 or type 3c diabetes. Innate immune responses, particularly the instant blood-mediated inflammatory reaction and activation of monocytes, play a major role in the loss of transplanted islet tissue. In this study, we aimed to investigate the inhibition of toll-like receptor 4 (TLR4) on innate inflammatory responses. We first demonstrate a significant loss of graft function shortly after transplant through the assessment of miR-375 and miR-200c in plasma as biomarkers. Using in vitro models, we investigate how targeting TLR4 mitigates islet damage and immune cell activation during the peritransplant period. The results of this study support the application of TAK-242 as a therapeutic agent to reduce inflammatory and innate immune responses to islets immediately following transplantation into the hepatic portal vein. Therefore, TLR4 may serve as a target to improve islet transplant outcomes in the future.


Subject(s)
Immunity, Innate , Islets of Langerhans Transplantation , Islets of Langerhans , MicroRNAs , Sulfonamides , Toll-Like Receptor 4 , Immunity, Innate/drug effects , Islets of Langerhans Transplantation/methods , Sulfonamides/pharmacology , Toll-Like Receptor 4/antagonists & inhibitors , Islets of Langerhans/drug effects , Islets of Langerhans/immunology , Humans
17.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167136, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38531483

ABSTRACT

Farnesoid X receptor (FXR) improves the function of islets, especially in the setting of Roux-en-Y gastric bypass (RYGB). Here we investigated how FXR activation regulates ß-cell proliferation and explored the potential link between FXR signaling and the menin pathway in controlling E2F3 expression, a key transcription factor for controlling adult ß-cell proliferation. Stimulation with the FXR agonist GW4064 or chenodeoxycholic acid (CDCA) increased E2F3 expression and ß-cell proliferation. Consistently, E2F3 knockdown abolished GW4064-induced proliferation. Treatment with GW4064 increased E2F3 expression in ß-cells via enhancing Steroid receptor coactivator-1 (SRC1) recruitment, increasing the pro-transcriptional acetylation of histone H3 at the E2f3 promoter. GW4064 treatment also decreased the association between FXR and menin, leading to the induction of FXR-mediated SRC1 recruitment. Mimicking the impact of FXR agonists, RYGB also increased E2F3 expression and ß-cell proliferation in GK rats and SD rats. These findings unravel the crucial role of the FXR/menin signaling in epigenetically controlling E2F3 expression and ß-cell proliferation, a mechanism possibly underlying RYGB-induced ß-cell proliferation.


Subject(s)
Cell Proliferation , E2F3 Transcription Factor , Epigenesis, Genetic , Insulin-Secreting Cells , Receptors, Cytoplasmic and Nuclear , Animals , Rats , Cell Proliferation/drug effects , Epigenesis, Genetic/drug effects , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/genetics , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/drug effects , Male , E2F3 Transcription Factor/metabolism , E2F3 Transcription Factor/genetics , Rats, Wistar , Histones/metabolism , Isoxazoles/pharmacology , Signal Transduction/drug effects , Islets of Langerhans/metabolism , Islets of Langerhans/drug effects , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology
18.
J Ultrasound Med ; 43(6): 1131-1141, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38414281

ABSTRACT

OBJECTIVES: Diabetes mellitus is a complex heterogenous metabolic disease that significantly affects the world population. Although many treatments exist, including medications such as metformin, sulfonylureas, and glucagon-like peptide-1 (GLP) receptor agonist, there is growing interest in finding alternative methods to noninvasively treat this disease. It has been previously shown that low-intensity ultrasound stimulation of pancreatic ß-cells in mice can elicit insulin secretion as a potential treatment for this disease. This is desirable as therapeutic ultrasound has the ability to induce bioeffects while selectively focusing deep within tissues, allowing for modulation of hormone secretion in the pancreas to mitigate insufficient levels of insulin. METHODS: Exactly 800 kHz ultrasound with intensity 0.5 W/cm2 was administered 5 minutes continuously, that is, 100% duty cycle, to donor pancreatic human islets, followed by 1 hour incubation and RT-qPCR to assess the effect of ultrasound stimulation on gene expression. The genes were insulin (INS), glucagon (Glu), amylin (Amy), and binding immunoglobulin protein (BiP). Nine donor pancreatic human islets were used to assess insulin and glucagon secretion, while eight samples were used for amylin and BiP. Fold change (FC) was calculated to analyze the effect of ultrasound stimulation on the gene expression of the donor islet cells. High-glucose and thapsigargin-treated islets were utilized as positive controls. Cell viability testing was done using a Trypan Blue Exclusion Test. RESULTS: Ultrasound stimulation did not cause a statistically significant upregulation in any of the tested genes (INS FC = 1.15, P-value = .5692; Glu FC = 1.60, P-value = .2231; Amy FC, P-value = .2863; BiP FC = 2.68, P-value = .3907). CONCLUSIONS: The results of this study show that the proposed ultrasound treatment parameters do not appear to significantly affect gene expression of any gene tested.


Subject(s)
Insulin , Islets of Langerhans , Ultrasonic Therapy , Humans , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Ultrasonic Therapy/methods , Glucagon , Gene Expression/drug effects , Islet Amyloid Polypeptide/pharmacology
19.
Histochem Cell Biol ; 161(5): 391-408, 2024 May.
Article in English | MEDLINE | ID: mdl-38368592

ABSTRACT

Apelin-13 is a peptide hormone that regulates pancreatic endocrine functions, and its benefits on the endocrine pancreas are of interest. This study aims to investigate the potential protective effects of apelin-13 in cisplatin-induced endocrine pancreatic damage. Twenty-four rats were divided into four groups: control, apelin-13, cisplatin, and cisplatin + apelin-13. Caspase-3, TUNEL, and Ki-67 immunohistochemical staining were used as markers of apoptosis and mitosis. NF-κB/p65 and TNFα were used to show inflammation. ß-cells and α-cells were also evaluated with insulin and glucagon staining in the microscopic examination. Pancreatic tissue was subjected to biochemical analyses of glutathione (GSH) and malondialdehyde (MDA). Apelin-13 ameliorated cisplatin-induced damage in the islets of Langerhans. The immunopositivity of apelin-13 on ß-cells and α-cells was found to be increased compared to the cisplatin group (p = 0.001, p = 0.001). Mitosis and apoptosis were significantly higher in the cisplatin group (p = 0.001). Apelin-13 reduced TNFα, NF-κB/p65 positivity, and apoptosis caused by cisplatin (p = 0.001, p = 0.001, p = 0.001). While cisplatin caused a significant increase in MDA levels (p = 0.001), apelin caused a significant decrease in MDA levels (p = 0.001). The results demonstrated a significant decrease in pancreatic tissue GSH levels following cisplatin treatment (p = 0.001). Nevertheless, apelin-13 significantly enhanced cisplatin-induced GSH reduction (p = 0.001). On the other hand, the serum glucose level, which was measured as 18.7 ± 2.5 mmol/L in the cisplatin group, decreased to 13.8 ± 0.7 mmol/L in the cisplatin + apelin-13 group (p = 0.001). The study shows that apelin-13 ameliorated cisplatin-induced endocrine pancreas damage by reducing oxidative stress and preventing apoptosis.


Subject(s)
Cisplatin , Intercellular Signaling Peptides and Proteins , Animals , Cisplatin/pharmacology , Rats , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Male , Apoptosis/drug effects , Islets of Langerhans/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/pathology , Rats, Wistar
20.
J Clin Endocrinol Metab ; 109(7): 1745-1753, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38252888

ABSTRACT

CONTEXT: In previous SURPASS studies tirzepatide reduced hemoglobin glycated A1c (HbA1c) and body weight and improved markers of insulin sensitivity and ß-cell function to a greater extent than comparators. OBJECTIVE: Explore changes in biomarkers of ß-cell function and insulin sensitivity and in efficacy profiles in baseline biomarker quartile analyses with tirzepatide compared to semaglutide. DESIGN: Post hoc analysis of SURPASS-2 phase 3 trial (participants randomly assigned to receive weekly subcutaneous tirzepatide or semaglutide for 40 weeks). SETTING: Post hoc analysis of 128 sites in 8 countries. PARTICIPANTS: A total of 1879 participants with type 2 diabetes. INTERVENTIONS: Once-weekly tirzepatide (5, 10, 15 mg) or semaglutide 1 mg. MAIN OUTCOMES MEASURES: Change in homeostatic model assessment indices for pancreatic ß-cell function (HOMA2-B) and for insulin resistance (HOMA2-IR), fasting glucagon, fasting C-peptide, and fasting insulin. RESULTS: At week 40, a greater increase in HOMA2-B was seen with tirzepatide (5, 10, 15 mg) doses (96.9-120.4%) than with semaglutide 1 mg (84.0%) (P < .05). There was a greater reduction in HOMA2-IR with all doses of tirzepatide (15.5%-24.0%) than with semaglutide 1 mg (5.1%) (P < .05). Tirzepatide 10 and 15 mg resulted in a significant reduction in both fasting C-peptide (5.2%-6.0%) and fasting glucagon (53.0%-55.3%) compared with an increase of C-peptide (3.3%) and a reduction of glucagon (47.7%) with semaglutide 1 mg (P < .05). HbA1c and body weight reductions were greater with all tirzepatide doses than semaglutide within each HOMA2-B and HOMA2-IR baseline quartile. CONCLUSION: In this post hoc analysis, improvements in HbA1c and weight loss were consistent and significantly higher with tirzepatide, regardless of baseline ß-cell function and insulin resistance, compared with semaglutide.


Subject(s)
Biomarkers , Diabetes Mellitus, Type 2 , Glucagon-Like Peptides , Hypoglycemic Agents , Insulin Resistance , Humans , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Male , Female , Middle Aged , Glucagon-Like Peptides/administration & dosage , Glucagon-Like Peptides/pharmacology , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Biomarkers/blood , Glycated Hemoglobin/analysis , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Blood Glucose/drug effects , Blood Glucose/metabolism , Aged , Adult , Double-Blind Method , Insulin/blood , Insulin/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Treatment Outcome , Glucagon-Like Peptide-2 Receptor , Gastric Inhibitory Polypeptide
SELECTION OF CITATIONS
SEARCH DETAIL
...