Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 685
Filter
1.
Clin Diabetes Endocrinol ; 10(1): 23, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39123252

ABSTRACT

This review explores the immunomodulatory potential of Teplizumab and its impact on pancreatic ß-cell function in T1D. Characterized by the autoimmune destruction of insulin-producing beta cells, T1D's management involves maintaining glycemic control through exogenous insulin. Teplizumab, a humanized monoclonal antibody targeting the CD3 antigen, has shown promise in delaying T1D onset and preserving residual ß-cell function. The review employs a narrative approach, synthesizing evidence from diverse clinical trials and studies gathered through a meticulous literature search. It scrutinizes Teplizumab's mechanisms of action, including its influence on autoreactive CD8 + T cells and regulatory T cells, offering insights into its immunological pathways. The synthesis of findings from various trials demonstrates Teplizumab's efficacy in preserving C-peptide levels and reducing exogenous insulin requirements, particularly in recent-onset T1D. Considering Teplizumab's real-world implications, the paper addresses potential obstacles, including side effects, patient selection criteria, and logistical challenges. It also emphasizes exploring combination therapies and personalized treatment strategies to maximize Teplizumab's benefits. The review contributes a nuanced perspective on Teplizumab's clinical implications and future directions in T1D management, bridging theoretical understanding with practical considerations.

2.
Nutrients ; 16(15)2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39125265

ABSTRACT

Uncarboxylated osteocalcin (ucOC) is a hormone secreted by osteoblasts that strengthens bone during mineralization and is a biomarker for ongoing bone formation. It also regulates glucose homeostasis by stimulating insulin secretion from pancreatic ß-cells. However, its effect on ß-cells under hyperglycemic diabetic conditions is unclear. The objective of this study was to investigate ucOC's effect on insulin secretion in ß-cells maintained under high glucose conditions. We hypothesized that hyperglycemia potentiates insulin secretion in response to ucOC stimulation. Using INS-1 cells, we performed insulin secretion experiments, intracellular calcium recordings, and RT-qPCR to determine ucOC's effect on glucose-stimulated insulin secretion (GSIS)-related genes. The results reveal that ucOC significantly increased insulin secretion under hyperglycemic conditions compared to lower glucose levels. High glucose conditions also potentiated the effect of ucOC on calcium signals, which enhanced insulin secretion. The increase in intracellular calcium was due to an influx from the extracellular space via voltage-dependent calcium channels (VDCCs). Interestingly, the treatment of cells with NPS-2143, a GPRC6A blocker, failed to abolish the calcium signals. Uncarboxylated osteocalcin upregulated the expression of GSIS-related genes under high glucose conditions (450 mg/dL) compared to cells under standard culture conditions (200 mg/dL). In conclusion, hyperglycemia potentiates ucOC-induced insulin secretion in ß-cells by opening VDCCs and upregulating GSIS genes. These findings provide a better understanding of ucOC's mechanism in the diabetic state and could lead to alternative treatments to stimulate insulin secretion.


Subject(s)
Hyperglycemia , Insulin Secretion , Insulin-Secreting Cells , Osteocalcin , Animals , Osteocalcin/metabolism , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/drug effects , Hyperglycemia/metabolism , Rats , Insulin Secretion/drug effects , Insulin/metabolism , Glucose/metabolism , Calcium/metabolism , Cell Line , Calcium Signaling/drug effects
3.
Medicina (Kaunas) ; 60(8)2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39202492

ABSTRACT

Abelmoschus manihot (L.) Medic flower (AMf) exhibits both nutritional value and bioactivities such as antioxidative, anti-inflammatory, neuroprotective, cardioprotective, and hepatoprotective effects. The aim of this investigation was to examine the potential impact of three different solvent extracts of AMf: supercritical CO2 extraction extract, water extract, and ethanol extract (AME), on management of diabetes. All three extracts demonstrated significant inhibitory effects on α-glucosidase (IC50 = 157-261 µg/mL) and lipase (IC50 = 401-577 µg/mL) activities while enhancing the α-amylase activity (32.4-41.8 folds at 200 µg/mL). Moreover, all three extracts exhibited notable inhibition of the formation of advanced glycation end-products, including the Amadori products (inhibition rates = 15.7-36.6%) and the dicarbonyl compounds (inhibition rates = 18.6-28.3%). Among the three extracts, AME exhibited the most pronounced inhibitory effect. AME displayed substantial in vitro and intracellular antioxidative activity, and effectively reduced ROS production (135% at 500 µg/mL) in ß-cells under hyperglycemic (HG) conditions. AME also enhanced the activity and gene expression of antioxidant enzymes, which were markedly decreased in the HG-induced ß-cells. Furthermore, AME protected ß-cell viability and maintained normal insulin secretion under HG conditions, likely due to its ability to reduce oxidative stress within ß-cells. This study demonstrated the potential of AME in preventing and managing diabetes and its associated complications. Further in vivo research is necessary to thoroughly elucidate the preventive effects and their underlying mechanisms.


Subject(s)
Abelmoschus , Flowers , Hypoglycemic Agents , Plant Extracts , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Flowers/chemistry , Abelmoschus/chemistry , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Antioxidants/pharmacology , Antioxidants/therapeutic use , Animals , Rats
4.
Biochem Biophys Res Commun ; 733: 150596, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39197196

ABSTRACT

PURPOSE: The high morbidity and mortality associated with type 2 diabetes mellitus (T2DM) pose a significant global health challenge, necessitating the development of more efficient anti-diabetic drugs with fewer side effects. This study investigated the intervention of vitamin D3 combined with glibenclamide in rats with T2DM to elucidate its effects on pancreatic ß-cells through the NF-κB pathway. METHODS: Twenty-four healthy male Sprague-Dawley (SD) rats were randomly assigned to four groups: the control group (CG), the model group (MG), the glibenclamide group (GG), and the glibenclamide + vitamin D3 group (GDG). After inducing the T2DM model using high-fat and high-sugar diet and intraperitoneal injection of streptozotocin, the rats in the GG group were administered glibenclamide orally (0.6 mg/kg/day), while those in the GDG group received both glibenclamide (0.6 mg/kg/day) and vitamin D3 (500 IU/kg/day) in corn oil for a duration of 8 weeks. Biochemical indices were measured, and histopathological changes in pancreatic tissue and islet ß cells were observed using hematoxylin and eosin staining. The expression of pancreatic nuclear factor κB (NF-κB), islet ß-cells, and inflammatory cytokines were assessed using the TUNEL method and PCR. RESULTS: According to the data from this current study, the GDG group showed significant positive differences in plasma biochemical indices, as well as in the expression of ß cells, NF-κB p65, TNF-α, IL-1ß, INF-γ, and Fas, compared to the GG and CG groups (P < 0.05). CONCLUSION: The results suggest that vitamin D has beneficial effects on T2DM by improving the functions of islet ß cells through inhibition of the NF-κB signaling pathway. Therefore, it is suggested that vitamin D supplementation, when used alongside antidiabetic drugs, may more effectively prevent and treat T2DM.

5.
Sci Rep ; 14(1): 19863, 2024 08 27.
Article in English | MEDLINE | ID: mdl-39191834

ABSTRACT

The significant advances in the differentiation of human pluripotent stem (hPS) cells into pancreatic endocrine cells, including functional ß-cells, have been based on a detailed understanding of the underlying developmental mechanisms. However, the final differentiation steps, leading from endocrine progenitors to mono-hormonal and mature pancreatic endocrine cells, remain to be fully understood and this is reflected in the remaining shortcomings of the hPS cell-derived islet cells (SC-islet cells), which include a lack of ß-cell maturation and variability among different cell lines. Additional signals and modifications of the final differentiation steps will have to be assessed in a combinatorial manner to address the remaining issues and appropriate reporter lines would be useful in this undertaking. Here we report the generation and functional validation of hPS cell reporter lines that can monitor the generation of INS+ and GCG+ cells and their resolution into mono-hormonal cells (INSeGFP, INSeGFP/GCGmCHERRY) as well as ß-cell maturation (INSeGFP/MAFAmCHERRY) and function (INSGCaMP6). The reporter hPS cell lines maintained strong and widespread expression of pluripotency markers and differentiated efficiently into definitive endoderm and pancreatic progenitor (PP) cells. PP cells from all lines differentiated efficiently into islet cell clusters that robustly expressed the corresponding reporters and contained glucose-responsive, insulin-producing cells. To demonstrate the applicability of these hPS cell reporter lines in a high-content live imaging approach for the identification of optimal differentiation conditions, we adapted our differentiation procedure to generate SC-islet clusters in microwells. This allowed the live confocal imaging of multiple SC-islets for a single condition and, using this approach, we found that the use of the N21 supplement in the last stage of the differentiation increased the number of monohormonal ß-cells without affecting the number of α-cells in the SC-islets. The hPS cell reporter lines and the high-content live imaging approach described here will enable the efficient assessment of multiple conditions for the optimal differentiation and maturation of SC-islets.


Subject(s)
Cell Differentiation , Genes, Reporter , Insulin-Secreting Cells , Islets of Langerhans , Pluripotent Stem Cells , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Cell Line , Insulin/metabolism , Green Fluorescent Proteins/metabolism , Green Fluorescent Proteins/genetics
6.
Viruses ; 16(8)2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39205219

ABSTRACT

The COVID-19 pandemic has revealed a bidirectional relationship between SARS-CoV-2 infection and diabetes mellitus. Existing evidence strongly suggests hyperglycemia as an independent risk factor for severe COVID-19, resulting in increased morbidity and mortality. Conversely, recent studies have reported new-onset diabetes following SARS-CoV-2 infection, hinting at a potential direct viral attack on pancreatic beta cells. In this review, we explore how hyperglycemia, a hallmark of diabetes, might influence SARS-CoV-2 entry and accessory proteins in pancreatic ß-cells. We examine how the virus may enter and manipulate such cells, focusing on the role of the spike protein and its interaction with host receptors. Additionally, we analyze potential effects on endosomal processing and accessory proteins involved in viral infection. Our analysis suggests a complex interplay between hyperglycemia and SARS-CoV-2 in pancreatic ß-cells. Understanding these mechanisms may help unlock urgent therapeutic strategies to mitigate the detrimental effects of COVID-19 in diabetic patients and unveil if the virus itself can trigger diabetes onset.


Subject(s)
COVID-19 , Hyperglycemia , Insulin-Secreting Cells , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Virus Internalization , Insulin-Secreting Cells/virology , Insulin-Secreting Cells/metabolism , Humans , Hyperglycemia/virology , Hyperglycemia/metabolism , Hyperglycemia/complications , SARS-CoV-2/physiology , COVID-19/virology , COVID-19/complications , COVID-19/metabolism , Spike Glycoprotein, Coronavirus/metabolism , Diabetes Mellitus/virology , Diabetes Mellitus/metabolism
7.
Front Endocrinol (Lausanne) ; 15: 1412411, 2024.
Article in English | MEDLINE | ID: mdl-39015185

ABSTRACT

Early in the development of Type 2 diabetes (T2D), metabolic stress brought on by insulin resistance and nutrient overload causes ß-cell hyperstimulation. Herein we summarize recent studies that have explored the premise that an increase in the intracellular Ca2+ concentration ([Ca2+]i), brought on by persistent metabolic stimulation of ß-cells, causes ß-cell dysfunction and failure by adversely affecting ß-cell function, structure, and identity. This mini-review builds on several recent reviews that also describe how excess [Ca2+]i impairs ß-cell function.


Subject(s)
Calcium Signaling , Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , Stress, Physiological , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Humans , Calcium Signaling/physiology , Animals , Stress, Physiological/physiology , Diabetes Mellitus, Type 2/metabolism , Calcium/metabolism , Insulin Resistance/physiology
8.
Cells ; 13(13)2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38994961

ABSTRACT

Cytokine-induced ß-cell apoptosis is a major pathogenic mechanism in type 1 diabetes (T1D). Despite significant advances in understanding its underlying mechanisms, few drugs have been translated to protect ß-cells in T1D. Epigenetic modulators such as bromodomain-containing BET (bromo- and extra-terminal) proteins are important regulators of immune responses. Pre-clinical studies have demonstrated a protective effect of BET inhibitors in an NOD (non-obese diabetes) mouse model of T1D. However, the effect of BET protein inhibition on ß-cell function in response to cytokines is unknown. Here, we demonstrate that I-BET, a BET protein inhibitor, protected ß-cells from cytokine-induced dysfunction and death. In vivo administration of I-BET to mice exposed to low-dose STZ (streptozotocin), a model of T1D, significantly reduced ß-cell apoptosis, suggesting a cytoprotective function. Mechanistically, I-BET treatment inhibited cytokine-induced NF-kB signaling and enhanced FOXO1-mediated anti-oxidant response in ß-cells. RNA-Seq analysis revealed that I-BET treatment also suppressed pathways involved in apoptosis while maintaining the expression of genes critical for ß-cell function, such as Pdx1 and Ins1. Taken together, this study demonstrates that I-BET is effective in protecting ß-cells from cytokine-induced dysfunction and apoptosis, and targeting BET proteins could have potential therapeutic value in preserving ß-cell functional mass in T1D.


Subject(s)
Apoptosis , Cytokines , Insulin-Secreting Cells , NF-kappa B , Signal Transduction , Animals , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , NF-kappa B/metabolism , Mice , Cytokines/metabolism , Signal Transduction/drug effects , Apoptosis/drug effects , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Forkhead Box Protein O1/metabolism , Mice, Inbred NOD , Male , Mice, Inbred C57BL
9.
Int J Mol Sci ; 25(13)2024 Jun 27.
Article in English | MEDLINE | ID: mdl-39000139

ABSTRACT

Epinephrine influences the function of pancreatic ß-cells, primarily through the α2A-adrenergic receptor (α2A-AR) on their plasma membrane. Previous studies indicate that epinephrine transiently suppresses insulin secretion, whereas prolonged exposure induces its compensatory secretion. Nonetheless, the impact of epinephrine-induced α2A-AR signaling on the survival and function of pancreatic ß-cells, particularly the impact of reprogramming after their removal from sustained epinephrine stimulation, remains elusive. In the present study, we applied MIN6, a murine insulinoma cell line, with 3 days of high concentration epinephrine incubation and 2 days of standard incubation, explored cell function and activity, and analyzed relevant regulatory pathways. The results showed that chronic epinephrine incubation led to the desensitization of α2A-AR and enhanced insulin secretion. An increased number of docked insulin granules and impaired Syntaxin-2 was found after chronic epinephrine exposure. Growth curve and cell cycle analyses showed the inhibition of cell proliferation. Transcriptome analysis showed the occurrence of endoplasmic reticulum stress (ER stress) and oxidative stress, such as the presence of BiP, CHOP, IRE1, ATF4, and XBP, affecting cellular endoplasmic reticulum function and survival, along with UCP2, OPA1, PINK, and PRKN, associated with mitochondrial dysfunction. Consequently, we conclude that chronic exposure to epinephrine induces α2A-AR desensitization and leads to ER and oxidative stress, impairing protein processing and mitochondrial function, leading to modified pancreatic ß-cell secretory function and cell fate.


Subject(s)
Endoplasmic Reticulum Stress , Epinephrine , Insulin-Secreting Cells , Insulin , Oxidative Stress , Animals , Epinephrine/pharmacology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/drug effects , Oxidative Stress/drug effects , Mice , Endoplasmic Reticulum Stress/drug effects , Insulin/metabolism , Insulin Secretion/drug effects , Receptors, Adrenergic, alpha-2/metabolism , Receptors, Adrenergic, alpha-2/genetics , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Signal Transduction/drug effects , Mitochondria/metabolism , Mitochondria/drug effects
10.
Curr Issues Mol Biol ; 46(7): 7621-7667, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39057094

ABSTRACT

The ß-cells within the pancreas play a pivotal role in insulin production and secretion, responding to fluctuations in blood glucose levels. However, factors like obesity, dietary habits, and prolonged insulin resistance can compromise ß-cell function, contributing to the development of Type 2 Diabetes (T2D). A critical aspect of this dysfunction involves ß-cell dedifferentiation and transdifferentiation, wherein these cells lose their specialized characteristics and adopt different identities, notably transitioning towards progenitor or other pancreatic cell types like α-cells. This process significantly contributes to ß-cell malfunction and the progression of T2D, often surpassing the impact of outright ß-cell loss. Alterations in the expressions of specific genes and transcription factors unique to ß-cells, along with epigenetic modifications and environmental factors such as inflammation, oxidative stress, and mitochondrial dysfunction, underpin the occurrence of ß-cell dedifferentiation and the onset of T2D. Recent research underscores the potential therapeutic value for targeting ß-cell dedifferentiation to manage T2D effectively. In this review, we aim to dissect the intricate mechanisms governing ß-cell dedifferentiation and explore the therapeutic avenues stemming from these insights.

11.
Sci Rep ; 14(1): 16809, 2024 07 22.
Article in English | MEDLINE | ID: mdl-39039202

ABSTRACT

Long-term exposure to hyperglycemic conditions leads to ß-cell dysfunction, particularly mitochondrial dysfunction, and inflammatory and oxidative stress responses, which are considered the primary causes of ß-cell death and the hallmarks of diabetes. Plant-active ingredients may play a key role in glycemic control. Epigallocatechin gallate (EGCG) is a characteristic catechin derived from tea that possesses anti-diabetic properties. Nonetheless, its underlying mechanisms remain elusive. Herein, the protective role of EGCG on high glucose (33 mM)-induced pancreatic beta cell dysfunction and its possible molecular mechanisms were investigated. Briefly, MIN6 cells were treated with glucose and EGCG (10 µM, 20 µM, and 40 µM) for 48 h. Our results revealed that EGCG dose-dependently restored mitochondrial membrane potential and concomitantly alleviated cell apoptosis. Mechanistically, the expression level of apoptotic protein BAX and Dynamic related protein 1 (DRP1) was significantly downregulated following EGCG treatment, whereas that of the anti-apoptotic protein BCL-2 was significantly upregulated. Taken together, EGCG alleviated high glucose-induced pancreatic beta cell dysfunction by targeting the DRP1-related mitochondrial apoptosis pathway and thus can serve as a nutritional intervention for the preservation of beta cell dysfunction in patients with type 2 diabetes mellitus.


Subject(s)
Apoptosis , Catechin , Dynamins , Glucose , Insulin-Secreting Cells , Mitochondria , Catechin/analogs & derivatives , Catechin/pharmacology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Apoptosis/drug effects , Mitochondria/metabolism , Mitochondria/drug effects , Glucose/metabolism , Dynamins/metabolism , Dynamins/genetics , Animals , Mice , Cell Line , Membrane Potential, Mitochondrial/drug effects , bcl-2-Associated X Protein/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism
12.
Sci Rep ; 14(1): 17691, 2024 07 31.
Article in English | MEDLINE | ID: mdl-39085384

ABSTRACT

Glucocorticoids (GCs) are known to stimulate pancreatic beta (ß)-cell apoptosis via several mechanisms, including oxidative stress. Our previous study suggested an increase in dexamethasone-induced pancreatic ß-cell apoptosis via a reduction of glutathione S-transferase P1 (GSTP1), which is an antioxidant enzyme. Imatinib, which is a tyrosine kinase inhibitor, also exerts antioxidant effect. This study aims to test our hypothesis that imatinib would prevent pancreatic ß-cell apoptosis induced by dexamethasone via increased GSTP1 expression and reduced oxidative stress. Our results revealed that dexamethasone significantly increased apoptosis in INS-1 cells when compared to the control, and that imatinib significantly decreased INS-1 cell apoptosis induced by dexamethasone. Moreover, dexamethasone significantly increased superoxide production in INS-1 cells when compared to the control; however, imatinib, when combined with dexamethasone, significantly reduced superoxide production in INS-1 cells. Dexamethasone significantly decreased GSTP1, p-ERK1/2, and BCL2 protein expression, but significantly increased p-JNK, p-p38, and BAX protein expression in INS-1 cells-all compared to control. Importantly, imatinib significantly ameliorated the effect of dexamethasone on the expression of GSTP1, p-ERK1/2, p-JNK, p-p38 MAPK, BAX, and BCL2. Furthermore-6-(7-nitro-2,1,3-benzoxadiazol-4-ylthio) hexanol (NBDHEX), which is a GSTP1 inhibitor, neutralized the protective effect of imatinib against pancreatic ß-cell apoptosis induced by dexamethasone. In conclusion, imatinib decreases pancreatic ß-cell apoptosis induced by dexamethasone via increased GSTP1 expression and reduced oxidative stress.


Subject(s)
Apoptosis , Dexamethasone , Glutathione S-Transferase pi , Imatinib Mesylate , Insulin-Secreting Cells , Oxidative Stress , Imatinib Mesylate/pharmacology , Dexamethasone/pharmacology , Oxidative Stress/drug effects , Apoptosis/drug effects , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Glutathione S-Transferase pi/metabolism , Animals , Rats , Cell Line , Superoxides/metabolism
13.
Redox Biol ; 75: 103283, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39067330

ABSTRACT

We asked whether acute redox signaling from mitochondria exists concomitantly to fatty acid- (FA-) stimulated insulin secretion (FASIS) at low glucose by pancreatic ß-cells. We show that FA ß-oxidation produces superoxide/H2O2, providing: i) mitochondria-to-plasma-membrane redox signaling, closing KATP-channels synergically with elevated ATP (substituting NADPH-oxidase-4-mediated H2O2-signaling upon glucose-stimulated insulin secretion); ii) activation of redox-sensitive phospholipase iPLA2γ/PNPLA8, cleaving mitochondrial FAs, enabling metabotropic GPR40 receptors to amplify insulin secretion (IS). At fasting glucose, palmitic acid stimulated IS in wt mice; palmitic, stearic, lauric, oleic, linoleic, and hexanoic acids also in perifused pancreatic islets (PIs), with suppressed 1st phases in iPLA2γ/PNPLA8-knockout mice/PIs. Extracellular/cytosolic H2O2-monitoring indicated knockout-independent redox signals, blocked by mitochondrial antioxidant SkQ1, etomoxir, CPT1 silencing, and catalase overexpression, all inhibiting FASIS, keeping ATP-sensitive K+-channels open, and diminishing cytosolic [Ca2+]-oscillations. FASIS in mice was a postprandially delayed physiological event. Redox signals of FA ß-oxidation are thus documented, reaching the plasma membrane, essentially co-stimulating IS.


Subject(s)
Cell Membrane , Fatty Acids , Insulin Secretion , Insulin-Secreting Cells , Mitochondria , Oxidation-Reduction , Signal Transduction , Animals , Mice , Mitochondria/metabolism , Fatty Acids/metabolism , Insulin-Secreting Cells/metabolism , Cell Membrane/metabolism , Mice, Knockout , Insulin/metabolism , Hydrogen Peroxide/metabolism , Group VI Phospholipases A2/metabolism , Group VI Phospholipases A2/genetics , Glucose/metabolism , Receptors, G-Protein-Coupled
14.
Front Endocrinol (Lausanne) ; 15: 1386471, 2024.
Article in English | MEDLINE | ID: mdl-38966213

ABSTRACT

Diabetes mellitus (DM), is a chronic disorder characterized by impaired glucose homeostasis that results from the loss or dysfunction of pancreatic ß-cells leading to type 1 diabetes (T1DM) and type 2 diabetes (T2DM), respectively. Pancreatic ß-cells rely to a great degree on their endoplasmic reticulum (ER) to overcome the increased secretary need for insulin biosynthesis and secretion in response to nutrient demand to maintain glucose homeostasis in the body. As a result, ß-cells are potentially under ER stress following nutrient levels rise in the circulation for a proper pro-insulin folding mediated by the unfolded protein response (UPR), underscoring the importance of this process to maintain ER homeostasis for normal ß-cell function. However, excessive or prolonged increased influx of nascent proinsulin into the ER lumen can exceed the ER capacity leading to pancreatic ß-cells ER stress and subsequently to ß-cell dysfunction. In mammalian cells, such as ß-cells, the ER stress response is primarily regulated by three canonical ER-resident transmembrane proteins: ATF6, IRE1, and PERK/PEK. Each of these proteins generates a transcription factor (ATF4, XBP1s, and ATF6, respectively), which in turn activates the transcription of ER stress-inducible genes. An increasing number of evidence suggests that unresolved or dysregulated ER stress signaling pathways play a pivotal role in ß-cell failure leading to insulin secretion defect and diabetes. In this article we first highlight and summarize recent insights on the role of ER stress and its associated signaling mechanisms on ß-cell function and diabetes and second how the ER stress pathways could be targeted in vitro during direct differentiation protocols for generation of hPSC-derived pancreatic ß-cells to faithfully phenocopy all features of bona fide human ß-cells for diabetes therapy or drug screening.


Subject(s)
Endoplasmic Reticulum Stress , Insulin-Secreting Cells , Unfolded Protein Response , Insulin-Secreting Cells/metabolism , Endoplasmic Reticulum Stress/physiology , Humans , Animals , Unfolded Protein Response/physiology , Diabetes Mellitus/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology
15.
Biochem Pharmacol ; 227: 116444, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39038551

ABSTRACT

Regenerating family protein 2 (Reg2) is a trophic factor which stimulates ß-cell replication and resists islet destruction. However, Reg2 also serves as an islet autoantigen, which makes it complicated to judge the effectiveness in treating diabetes. How Reg2 treatment behaves in non-obese diabetic (NOD) mice is to be investigated. NOD mice were treated with recombinant Reg2 protein, Complete Freund's adjuvant (CFA) + PBS and CFA+Reg2 vaccinations, CFA+PBS- and CFA+Reg2-immunized antisera, and single chain variable fragment (scFv)-Reg2 and mIgG2a-Reg2 antibodies. Glycemic level, bodyweight, serum Reg2 antibody titer, glucose tolerance, and insulin secretion were determined. Islet morphological characteristics, insulitis, cell apoptosis, islet cell components, and T cell infiltration were analyzed by histological examinations. The autoantigenicity of constructed Reg2C and Reg2X fragments was determined in healthy BALB/c mice, and the bioactivity in stimulating cell proliferation and survival was assessed in insulinoma MIN6 cells. Reg2 administration alleviated diabetes in NOD mice with improved glucose tolerance and insulin secretion but elevated serum Reg2 autoantibodies. Histomorphometry showed reduced inflammatory area, TUNEL signal and CD8 + T cell infiltration, and increased ß-cell proportion in support of the islet-protective effect of Reg2 treatment. CFA+PBS and CFA+Reg2 immunizations prevented diabetic onset and alleviated insulitis while injections of the antisera offered mild protections. Antibody treatments accelerated diabetic onset without increasing the overall incidence. Reg2C fragment depletes antigenicity, but reserves protective activity in streptozotocin (STZ)-treated MIN6 cells. In conclusion, Reg2 treatment alleviates type 1 diabetes (T1D) by preserving islet ß-cells, but induces Reg2 autoantibody production which poses a potential risk of accelerating diabetic progression.


Subject(s)
Autoantibodies , Islets of Langerhans , Mice, Inbred BALB C , Mice, Inbred NOD , Pancreatitis-Associated Proteins , Animals , Autoantibodies/immunology , Autoantibodies/blood , Mice , Islets of Langerhans/immunology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Female , Pancreatitis-Associated Proteins/immunology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/prevention & control , Diabetes Mellitus, Type 1/drug therapy , Lithostathine/immunology
16.
Stem Cell Rev Rep ; 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38916841

ABSTRACT

The pathogenesis of diabetes involves complex changes in the expression profiles of mRNA and non-coding RNAs within pancreatic islet cells. Recent progress in induced pluripotent stem cell (iPSC) technology have allowed the modeling of diabetes-associated genes. Our recent study using FOXA2-deficient human iPSC models has highlighted an essential role for FOXA2 in the development of human pancreas. Here, we aimed to provide further insights on the role of microRNAs (miRNAs) by studying the miRNA-mRNA regulatory networks in iPSC-derived islets lacking the FOXA2 gene. Consistent with our previous findings, the absence of FOXA2 significantly downregulated the expression of islet hormones, INS, and GCG, alongside other key developmental genes in pancreatic islets. Concordantly, RNA-Seq analysis showed significant downregulation of genes related to pancreatic development and upregulation of genes associated with nervous system development and lipid metabolic pathways. Furthermore, the absence of FOXA2 in iPSC-derived pancreatic islets resulted in significant alterations in miRNA expression, with 61 miRNAs upregulated and 99 downregulated. The upregulated miRNAs targeted crucial genes involved in diabetes and pancreatic islet cell development. In contrary, the absence of FOXA2 in islets showed a network of downregulated miRNAs targeting genes related to nervous system development and lipid metabolism. These findings highlight the impact of FOXA2 absence on pancreatic islet development and suggesting intricate miRNA-mRNA regulatory networks affecting pancreatic islet cell development.

17.
Cells ; 13(11)2024 May 30.
Article in English | MEDLINE | ID: mdl-38891081

ABSTRACT

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


Subject(s)
Cholecystokinin , Insulin-Secreting Cells , Verapamil , Zebrafish , Animals , Mice , Cell Line , Cell Proliferation/drug effects , Cholecystokinin/metabolism , Cholecystokinin/pharmacology , Disease Models, Animal , Glucose/metabolism , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/drug effects , Regeneration/drug effects , Verapamil/pharmacology
18.
Zhongguo Zhen Jiu ; 44(6): 669-75, 2024 Jun 12.
Article in Chinese | MEDLINE | ID: mdl-38867629

ABSTRACT

OBJECTIVE: To observe the effects of electroacupuncture (EA) on the expression of serum interleukin-1ß (IL-1ß), tumor necrosis factor-α (TNF-α), and the pancreatic nuclear factor-κB (NF-κB) pathway in type 2 diabetes mellitus (T2DM) rats, and to explore the possible mechanism by which EA improving the dedifferentiation of pancreatic ß-cells in the treatment of T2DM. METHODS: Among 18 SPF-grade male Wistar rats, 6 rats were randomly selected as the control group, and the remaining 12 rats were fed with high-sugar and high-fat diet combined with intraperitoneal injection of 2% streptozotocin solution (35 mg/kg) to establish T2DM model. After successful modeling, the 12 rats were randomly divided into a model group and an EA group, with 6 rats in each group. The EA group received EA at bilateral "Zusanli" (ST 36), "Sanyinjiao" (SP 6), "Weiwanxiashu" (EX-B 3), and "Pishu" (BL 20), with continuous wave, frequency of 15 Hz, current intensity of 2 mA, for 20 min each time, once a day, 6 times a week, for a total of 6 weeks. Fasting blood glucose (FBG) levels were measured before modeling and before and after intervention. After intervention, ELISA was used to detect the serum fasting insulin (FINS), IL-1ß and TNF-α levels, and the ß-cell function index (HOMA-ß) and insulin resistance index (HOMA-IR) were calculated; HE staining was used to observe the morphology of the pancreatic islets; Western blot was used to detect the protein expression of pancreatic forkhead box protein O1 (FoxO1), pancreatic and duodenal homeobox 1 (PDX-1), neurogenin 3 (NGN3), and NF-κB p65. RESULTS: After intervention, the FBG in the model group was higher than that in the control group (P<0.01), and the FBG in the EA group was lower than that in the model group (P<0.01). Compared with the control group, the model group had increased levels of serum FINS, IL-1ß, TNF-α, and HOMA-IR (P<0.01), and decreased HOMA-ß (P<0.01), reduced protein expression of pancreatic FoxO1 and PDX-1 (P<0.01), and increased protein expression of pancreatic NGN3 and NF-κB p65 (P<0.01, P<0.05). Compared with the model group, the EA group had lower serum FINS, IL-1ß, TNF-α levels, and HOMA-IR (P<0.01), higher HOMA-ß (P<0.05), increased protein expression of pancreatic FoxO1 and PDX-1 (P<0.01, P<0.05), and decreased protein expression of pancreatic NGN3 and NF-κB p65 (P<0.01, P<0.05). The control group's pancreatic islets showed no obvious abnormalities; the model group's pancreatic islets were irregular in shape and had unclear boundaries with the surrounding area, with immune cell infiltration, reduced ß-cell nuclei, disordered arrangement of islet cells, and increased intercellular spaces; the EA group showed improvements in islet morphology, immune cell infiltration, ß-cell nuclei count, and the arrangement and spacing of islet cells approaching normal. CONCLUSION: EA could lower the blood glucose levels in T2DM rats, alleviate chronic inflammatory responses in the islets, and improve the dedifferentiation of pancreatic ß-cells, which may be related to the inhibition of pancreatic NF-κB pathway expression.


Subject(s)
Diabetes Mellitus, Type 2 , Electroacupuncture , Insulin-Secreting Cells , Interleukin-1beta , NF-kappa B , Rats, Wistar , Animals , Male , Rats , Diabetes Mellitus, Type 2/therapy , Diabetes Mellitus, Type 2/metabolism , Insulin-Secreting Cells/metabolism , NF-kappa B/metabolism , Humans , Interleukin-1beta/metabolism , Tumor Necrosis Factor-alpha/metabolism , Signal Transduction , Cell Dedifferentiation , Blood Glucose/metabolism , Acupuncture Points , Insulin/metabolism
19.
Islets ; 16(1): 2361996, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38833523

ABSTRACT

Epidemiological studies consistently link environmental toxicant exposure with increased Type 2 diabetes risk. Our study investigated the diabetogenic effects of a widely used flame retardant, Dechlorane Plus (DP), on pancreatic ß-cells using rodent and human model systems. We first examined pancreas tissues from male mice exposed daily to oral gavage of either vehicle (corn oil) or DP (10, 100, or 1000 µg/kg per day) and fed chow or high fat diet for 28-days in vivo. DP exposure did not affect islet size or endocrine cell composition in either diet group. Next, we assessed the effect of 48-hour exposure to vehicle (DMSO) or DP (1, 10, or 100 nM) in vitro using immortalized rat ß-cells (INS-1 832/3), primary mouse and human islets, and human stem-cell derived islet-like cells (SC-islets). In INS-1 832/3 cells, DP did not impact glucose-stimulated insulin secretion (GSIS) but significantly decreased intracellular insulin content. DP had no effect on GSIS in mouse islets or SC-islets but had variable effects on GSIS in human islets depending on the donor. DP alone did not affect insulin content in mouse islets, human islets, or SC-islets, but mouse islets co-exposed to DP and glucolipotoxic (GLT) stress conditions (28.7 mM glucose + 0.5 mM palmitate) had reduced insulin content compared to control conditions. Co-exposure of mouse islets to DP + GLT amplified the upregulation of Slc30a8 compared to GLT alone. Our study highlights the importance and challenges of using different in vitro models for studying chemical toxicity.


Subject(s)
Hydrocarbons, Chlorinated , Insulin-Secreting Cells , Polycyclic Compounds , Animals , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Humans , Mice , Male , Polycyclic Compounds/pharmacology , Hydrocarbons, Chlorinated/toxicity , Rats , Insulin/metabolism , Flame Retardants/toxicity , Insulin Secretion/drug effects , Mice, Inbred C57BL , Cells, Cultured
20.
Clin Epigenetics ; 16(1): 78, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38862980

ABSTRACT

Diabetes mellitus is a chronic disease that impairs metabolism, and its prevalence has reached an epidemic proportion globally. Most people affected are with type 2 diabetes mellitus (T2DM), which is caused by a decline in the numbers or functioning of pancreatic endocrine islet cells, specifically the ß-cells that release insulin in sufficient quantity to overcome any insulin resistance of the metabolic tissues. Genetic and epigenetic factors have been implicated as the main contributors to the T2DM. Epigenetic modifiers, histone deacetylases (HDACs), are enzymes that remove acetyl groups from histones and play an important role in a variety of molecular processes, including pancreatic cell destiny, insulin release, insulin production, insulin signalling, and glucose metabolism. HDACs also govern other regulatory processes related to diabetes, such as oxidative stress, inflammation, apoptosis, and fibrosis, revealed by network and functional analysis. This review explains the current understanding of the function of HDACs in diabetic pathophysiology, the inhibitory role of various HDAC inhibitors (HDACi), and their functional importance as biomarkers and possible therapeutic targets for T2DM. While their role in T2DM is still emerging, a better understanding of the role of HDACi may be relevant in improving insulin sensitivity, protecting ß-cells and reducing T2DM-associated complications, among others.


Subject(s)
Diabetes Mellitus, Type 2 , Epigenesis, Genetic , Histone Deacetylase Inhibitors , Histone Deacetylases , Humans , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/physiopathology , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Epigenesis, Genetic/drug effects , Insulin Resistance , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Animals , Oxidative Stress/drug effects , Insulin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL