Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 11.166
Filter
1.
FASEB J ; 38(16): e23885, 2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39139039

ABSTRACT

Liver kinase B1 (LKB1/STK11) is an important regulator of pancreatic ß-cell identity and function. Elimination of Lkb1 from the ß-cell results in improved glucose-stimulated insulin secretion and is accompanied by profound changes in gene expression, including the upregulation of several neuronal genes. The mechanisms through which LKB1 controls gene expression are, at present, poorly understood. Here, we explore the impact of ß cell-selective deletion of Lkb1 on chromatin accessibility in mouse pancreatic islets. To characterize the role of LKB1 in the regulation of gene expression at the transcriptional level, we combine these data with a map of islet active transcription start sites and histone marks. We demonstrate that LKB1 elimination from ß-cells results in widespread changes in chromatin accessibility, correlating with changes in transcript levels. Changes occurred in hundreds of promoter and enhancer regions, many of which were close to neuronal genes. We reveal that dysregulated enhancers are enriched in binding motifs for transcription factors (TFs) important for ß-cell identity, such as FOXA, MAFA or RFX6, and we identify microRNAs (miRNAs) that are regulated by LKB1 at the transcriptional level. Overall, our study provides important new insights into the epigenetic mechanisms by which LKB1 regulates ß-cell identity and function.


Subject(s)
Epigenesis, Genetic , Insulin-Secreting Cells , Protein Serine-Threonine Kinases , Animals , Insulin-Secreting Cells/metabolism , Mice , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Mice, Knockout , AMP-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinases/genetics , Promoter Regions, Genetic , Mice, Inbred C57BL , Male
2.
Sci Rep ; 14(1): 18962, 2024 08 16.
Article in English | MEDLINE | ID: mdl-39152192

ABSTRACT

Cadmium, a common metal pollutant, has been demonstrated to induce type 2 diabetes by disrupting pancreatic ß cells function. In this study, transcriptome microarray was utilized to identify differential gene expression in oxidative damage to pancreatic ß cells following cadmium exposure. The results indicated that a series of mRNAs, LncRNAs, and miRNAs were altered. Of the differentially expressed miRNAs, miR-29a-3p exhibited the most pronounced alteration, with an 11.62-fold increase relative to the control group. Following this, the target gene of miR-29a-3p was identified as Col3a1 through three databases (miRDB, miRTarbase and Tarbase), which demonstrated a decrease across the transcriptome microarray. The upstream target gene of miR-29a-3p was identified as NONMMUT036805, with decreased expression observed in the microarray. Finally, the expression trend of NONMMUT036805/miR-29a-3p/Col3a1 was reversed following NAC pretreatment. This was accompanied by a reduction in oxidative damage indicators, MDA/ROS/GSH-Px appeared to be negatively affected to varying degrees. In conclusion, this study has demonstrated that multiple RNAs are altered during cadmium exposure-induced oxidative damage in pancreatic ß cells. The NONMMUT036805/miR-29a-3p/Col3a1 axis has been shown to be involved in this process, which provides a foundation for the identification of potential targets for cadmium toxicity intervention.


Subject(s)
Cadmium , Insulin-Secreting Cells , MicroRNAs , Oxidative Stress , RNA, Competitive Endogenous , Animals , Mice , Cadmium/toxicity , Cell Line , Gene Expression Profiling , Gene Expression Regulation/drug effects , Gene Regulatory Networks/drug effects , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/drug effects , MicroRNAs/genetics , MicroRNAs/metabolism , Oxidative Stress/drug effects , Oxidative Stress/genetics , RNA, Competitive Endogenous/genetics , RNA, Competitive Endogenous/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcriptome
3.
Int J Mol Sci ; 25(15)2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39125590

ABSTRACT

Ras-related Rap1A GTPase is implicated in pancreas ß-cell insulin secretion and is stimulated by the cAMP sensor Epac2, a guanine exchange factor and activator of Rap1 GTPase. In this study, we examined the differential proteomic profiles of pancreata from C57BL/6 Rap1A-deficient (Null) and control wild-type (WT) mice with nanoLC-ESI-MS/MS to assess targets of Rap1A potentially involved in insulin regulation. We identified 77 overlapping identifier proteins in both groups, with 8 distinct identifier proteins in Null versus 56 distinct identifier proteins in WT mice pancreata. Functional enrichment analysis showed four of the eight Null unique proteins, ERO1-like protein ß (Ero1lß), triosephosphate isomerase (TP1), 14-3-3 protein γ, and kallikrein-1, were exclusively involved in insulin biogenesis, with roles in insulin metabolism. Specifically, the mRNA expression of Ero1lß and TP1 was significantly (p < 0.05) increased in Null versus WT pancreata. Rap1A deficiency significantly affected glucose tolerance during the first 15-30 min of glucose challenge but showed no impact on insulin sensitivity. Ex vivo glucose-stimulated insulin secretion (GSIS) studies on isolated Null islets showed significantly impaired GSIS. Furthermore, in GSIS-impaired islets, the cAMP-Epac2-Rap1A pathway was significantly compromised compared to the WT. Altogether, these studies underscore an essential role of Rap1A GTPase in pancreas physiological function.


Subject(s)
Insulin , Mice, Inbred C57BL , Pancreas , Proteomics , Signal Transduction , rap1 GTP-Binding Proteins , Animals , rap1 GTP-Binding Proteins/metabolism , rap1 GTP-Binding Proteins/genetics , Mice , Proteomics/methods , Insulin/metabolism , Pancreas/metabolism , Insulin-Secreting Cells/metabolism , Mice, Knockout , Guanine Nucleotide Exchange Factors/metabolism , Guanine Nucleotide Exchange Factors/genetics , Insulin Secretion , Male , Glucose/metabolism
4.
Sci Rep ; 14(1): 17844, 2024 08 01.
Article in English | MEDLINE | ID: mdl-39090166

ABSTRACT

This study was to determine whether extracellular vesicles (EVs) derived from insulin-producing cells (IPCs) can modulate naïve mesenchymal stromal cells (MSCs) to become insulin-secreting. MSCs were isolated from human adipose tissue. The cells were then differentiated to generate IPCs by achemical-based induction protocol. EVs were retrieved from the conditioned media of undifferentiated (naïve) MSCs (uneducated EVs) and from that of MSC-derived IPCs (educated EVs) by sequential ultracentrifugation. The obtained EVs were co-cultured with naïve MSCs.The cocultured cells were evaluated by immunofluorescence, flow cytometry, C-peptide nanogold silver-enhanced immunostaining, relative gene expression and their response to a glucose challenge.Immunostaining for naïve MSCs cocultured with educated EVs was positive for insulin, C-peptide, and GAD65. By flow cytometry, the median percentages of insulin-andC-peptide-positive cells were 16.1% and 14.2% respectively. C-peptide nanogoldimmunostaining providedevidence for the intrinsic synthesis of C-peptide. These cells released increasing amounts of insulin and C-peptide in response to increasing glucose concentrations. Gene expression of relevant pancreatic endocrine genes, except for insulin, was modest. In contrast, the results of naïve MSCs co-cultured with uneducated exosomes were negative for insulin, C-peptide, and GAD65. These findings suggest that this approach may overcome the limitations of cell therapy.


Subject(s)
Cell Differentiation , Coculture Techniques , Extracellular Vesicles , Insulin-Secreting Cells , Insulin , Mesenchymal Stem Cells , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Humans , Extracellular Vesicles/metabolism , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/cytology , C-Peptide/metabolism , Cells, Cultured , Glucose/metabolism , Adipose Tissue/cytology , Adipose Tissue/metabolism
5.
Front Endocrinol (Lausanne) ; 15: 1417437, 2024.
Article in English | MEDLINE | ID: mdl-39114287

ABSTRACT

Introduction: Using a non-human primate (NHP) model of maternal Western-style diet (mWSD) feeding during pregnancy and lactation, we previously reported altered offspring beta:alpha cell ratio in vivo and insulin hyper-secretion ex vivo. Mitochondria are known to maintain beta-cell function by producing ATP for insulin secretion. In response to nutrient stress, the mitochondrial network within beta cells undergoes morphological changes to maintain respiration and metabolic adaptability. Given that mitochondrial dynamics have also been associated with cellular fate transitions, we assessed whether mWSD exposure was associated with changes in markers of beta-cell maturity and/or mitochondrial morphology that might explain the offspring islet phenotype. Methods: We evaluated the expression of beta-cell identity/maturity markers (NKX6.1, MAFB, UCN3) via florescence microscopy in islets of Japanese macaque pre-adolescent (1 year old) and peri-adolescent (3-year-old) offspring born to dams fed either a control diet or WSD during pregnancy and lactation and weaned onto WSD. Mitochondrial morphology in NHP offspring beta cells was analyzed in 2D by transmission electron microscopy and in 3D using super resolution microscopy to deconvolve the beta-cell mitochondrial network. Results: There was no difference in the percent of beta cells expressing key maturity markers in NHP offspring from WSD-fed dams at 1 or 3 years of age; however, beta cells of WSD-exposed 3 year old offspring showed increased levels of NKX6.1 per beta cell at 3 years of age. Regardless of maternal diet, the beta-cell mitochondrial network was found to be primarily short and fragmented at both ages in NHP; overall mitochondrial volume increased with age. In utero and lactational exposure to maternal WSD consumption may increase mitochondrial fragmentation. Discussion: Despite mWSD consumption having clear developmental effects on offspring beta:alpha cell ratio and insulin secretory response to glucose, this does not appear to be mediated by changes to beta-cell maturity or the beta-cell mitochondrial network. In general, the more fragmented mitochondrial network in NHP beta cells suggests greater ability for metabolic flexibility.


Subject(s)
Diet, Western , Insulin-Secreting Cells , Maternal Nutritional Physiological Phenomena , Mitochondria , Prenatal Exposure Delayed Effects , Animals , Female , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/ultrastructure , Pregnancy , Mitochondria/metabolism , Mitochondria/ultrastructure , Diet, Western/adverse effects , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/pathology , Male , Lactation
6.
Front Immunol ; 15: 1393248, 2024.
Article in English | MEDLINE | ID: mdl-39114661

ABSTRACT

Objective: Beta cell destruction in type 1 diabetes (T1D) results from the combined effect of inflammation and recurrent autoimmunity. In recent years, the role played by beta cells in the development of T1D has evolved from passive victims of the immune system to active contributors in their own destruction. We and others have demonstrated that perturbations in the islet microenvironment promote endoplasmic reticulum (ER) stress in beta cells, leading to enhanced immunogenicity. Among the underlying mechanisms, secretion of extracellular vesicles (EVs) by beta cells has been suggested to mediate the crosstalk with the immune cell compartment. Methods: To study the role of cellular stress in the early events of T1D development, we generated a novel cellular model for constitutive ER stress by modulating the expression of HSPA5, which encodes BiP/GRP78, in EndoC-ßH1 cells. To investigate the role of EVs in the interaction between beta cells and the immune system, we characterized the EV miRNA cargo and evaluated their effect on innate immune cells. Results: Analysis of the transcriptome showed that HSPA5 knockdown resulted in the upregulation of signaling pathways involved in the unfolded protein response (UPR) and changes the miRNA content of EVs, including reduced levels of miRNAs involved in IL-1ß signaling. Treatment of primary human monocytes with EVs from stressed beta cells resulted in increased surface expression of CD11b, HLA-DR, CD40 and CD86 and upregulation of IL-1ß and IL-6. Conclusion: These findings indicate that the content of EVs derived from stressed beta cells can be a mediator of islet inflammation.


Subject(s)
Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , Extracellular Vesicles , Insulin-Secreting Cells , MicroRNAs , Monocytes , Extracellular Vesicles/metabolism , Extracellular Vesicles/immunology , Monocytes/immunology , Monocytes/metabolism , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/immunology , Humans , Endoplasmic Reticulum Stress/immunology , MicroRNAs/genetics , Inflammation/immunology , Inflammation/metabolism , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Animals , Cell Line , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , Signal Transduction , Unfolded Protein Response/immunology
7.
Cell Mol Life Sci ; 81(1): 345, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39133305

ABSTRACT

BACKGROUND AND AIMS: Hepatitis B virus (HBV)-associated liver cirrhosis (LC), a common condition with high incidence and mortality rates, is often associated with diabetes mellitus (DM). However, the molecular mechanisms underlying impaired glucose regulation during HBV-associated LC remain unclear. METHODS: Data from 63 patients with LC and 62 patients with LC-associated DM were analysed. Co-culture of NK cells and islet ß cell lines were used to study the glucose regulation mechanism. A mouse model of LC was used to verify the effect of S100A8/A9 on the glucose regulation. RESULTS: Higher levels of interferon (IFN)-γ derived from natural killer (NK) cells and lower levels of insulin emerged in the peripheral blood of patients with both LC and DM compared with those from patients with LC only. IFN-γ derived from NK cells facilitated ß cell necroptosis and impaired insulin production. Furthermore, S100A8/A9 elevation in patients with both LC and DM was found to upregulate IFN-γ production in NK cells. Consistently, in the mouse model for LC, mice treated with carbon tetrachloride (CCL4) and S100A8/A9 exhibited increased blood glucose, impaired insulin production, increased IFN-γ, and increased ß cells necroptosis compared with those treated with CCL4. Mechanistically, S100A8/A9 activated the p38 MAPK pathway to increase IFN-γ production in NK cells. These effects were diminished after blocking RAGE. CONCLUSION: Together, the data indicate that IFN-γ produced by NK cells induces ß cell necroptosis via the S100A8/A9-RAGE-p38 MAPK axis in patients with LC and DM. Reduced levels of S100A8/A9, NK cells, and IFN-γ could be valuable for the treatment of LC with DM. Accumulation of S100A8/A9 in patients with LC may indicate the emergence of DM.


Subject(s)
Calgranulin A , Calgranulin B , Hepatitis B virus , Insulin-Secreting Cells , Interferon-gamma , Killer Cells, Natural , Liver Cirrhosis , Necroptosis , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Humans , Animals , Interferon-gamma/metabolism , Calgranulin B/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/virology , Liver Cirrhosis/immunology , Mice , Male , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Insulin-Secreting Cells/virology , Calgranulin A/metabolism , Mice, Inbred C57BL , Female , Middle Aged , Hepatitis B/complications , Hepatitis B/pathology , Hepatitis B/metabolism , Disease Models, Animal , Carbon Tetrachloride
8.
Sci Rep ; 14(1): 18590, 2024 08 10.
Article in English | MEDLINE | ID: mdl-39127728

ABSTRACT

The relationship of adipose tissue insulin resistance (AT-IR, a product of fasting insulin and free fatty acids) and homeostasis-model assessment-insulin resistance (HOMA-IR) to ß-cell function was studied cross-sectionally in the setting of subtle glucose dysregulation. Associations of AT-IR and HOMA-IR with fasting and post-glucose glycemia and ß-cell function inferred from serum insulin kinetics during a 75 g oral glucose tolerance test were studied in 168 young female Japanese students. ß-cell function was evaluated by disposition index calculated as a product of the insulinogenic index (IGI) and Matsuda index. AT-IR, not HOMA-IR, showed positive associations with post-glucose glycemia and area under the glucose response curve although both indices were associated with fasting glycemia. HOMA-IR, not AT-IR, was associated positively with log IGI whereas both indices were inversely associated with Matsuda index. AT-IR, not HOMA-IR, showed inverse associations with log disposition index. Associations of adipose tissue insulin resistance with ß-cell function (inverse) and glucose excursion in young Japanese women may suggest that lipotoxicity to pancreatic ß-cells for decades may be associated with ß cell dysfunction found in Japanese patients with type 2 diabetes. Positive association of HOMA-IR with insulinogenic index may be associated with compensatory increased insulin secretion.


Subject(s)
Adipose Tissue , Blood Glucose , Glucose Tolerance Test , Insulin Resistance , Insulin-Secreting Cells , Insulin , Humans , Female , Insulin-Secreting Cells/metabolism , Adipose Tissue/metabolism , Blood Glucose/metabolism , Young Adult , Adult , Japan , Insulin/blood , Insulin/metabolism , Cross-Sectional Studies , Fasting/blood , East Asian People
9.
Cells ; 13(15)2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39120275

ABSTRACT

Preserving the function and survival of pancreatic beta-cells, in order to achieve long-term glycemic control and prevent complications, is an essential feature for an innovative drug to have clinical value in the treatment of diabetes. Innovative research is developing therapeutic strategies to prevent pathogenic mechanisms and protect beta-cells from the deleterious effects of inflammation and/or chronic hyperglycemia over time. A better understanding of receptors and signaling pathways, and of how they interact with each other in beta-cells, remains crucial and is a prerequisite for any strategy to develop therapeutic tools aimed at modulating beta-cell function and/or mass. Here, we present a comprehensive review of our knowledge on membrane and intracellular receptors and signaling pathways as targets of interest to protect beta-cells from dysfunction and apoptotic death, which opens or could open the way to the development of innovative therapies for diabetes.


Subject(s)
Diabetes Mellitus , Insulin-Secreting Cells , Signal Transduction , Humans , Signal Transduction/drug effects , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/pathology , Animals , Diabetes Mellitus/metabolism , Diabetes Mellitus/drug therapy , Diabetes Mellitus/pathology , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Cell Survival/drug effects
10.
Nat Commun ; 15(1): 6627, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39103322

ABSTRACT

Functional genetics has identified drug targets for metabolic disorders. Opioid use impacts metabolic homeostasis, although mechanisms remain elusive. Here, we explore the OPRD1 gene (encoding delta opioid receptor, DOP) to understand its impact on type 2 diabetes. Large-scale sequencing of OPRD1 and in vitro analysis reveal that loss-of-function variants are associated with higher adiposity and lower hyperglycemia risk, whereas gain-of-function variants are associated with lower adiposity and higher type 2 diabetes risk. These findings align with studies of opium addicts. OPRD1 is expressed in human islets and beta cells, with decreased expression under type 2 diabetes conditions. DOP inhibition by an antagonist enhances insulin secretion from human beta cells and islets. RNA-sequencing identifies pathways regulated by DOP antagonism, including nerve growth factor, circadian clock, and nuclear receptor pathways. Our study highlights DOP as a key player between opioids and metabolic homeostasis, suggesting its potential as a therapeutic target for type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , Receptors, Opioid, delta , Receptors, Opioid, delta/metabolism , Receptors, Opioid, delta/genetics , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Humans , Insulin-Secreting Cells/metabolism , Male , Female , Middle Aged , Insulin/metabolism , Insulin Secretion/drug effects , Insulin Secretion/genetics , Adult
11.
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
12.
Int J Mol Sci ; 25(15)2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39125908

ABSTRACT

Multicenter international clinical trials demonstrated the clinical safety and efficacy by using stem cell educator therapy to treat type 1 diabetes (T1D) and other autoimmune diseases. Previous studies characterized the peripheral blood insulin-producing cells (PB-IPC) from healthy donors with high potential to give rise to insulin-producing cells. PB-IPC displayed the molecular marker glucose transporter 2 (GLUT2), contributing to the glucose transport and sensing. To improve the clinical efficacy of stem cell educator therapy in the restoration of islet ß-cell function, we explored the GLUT2 expression on PB-IPC in recent onset and longstanding T1D patients. In the Food and Drug Administration (FDA)-approved phase 2 clinical studies, patients received one treatment with the stem cell educator therapy. Peripheral blood mononuclear cells (PBMC) were isolated for flow cytometry analysis of PB-IPC and other immune markers before and after the treatment with stem cell educator therapy. Flow cytometry revealed that both recent onset and longstanding T1D patients displayed very low levels of GLUT2 on PB-IPC. After the treatment with stem cell educator therapy, the percentages of GLUT2+CD45RO+ PB-IPC were markedly increased in these T1D subjects. Notably, we found that T1D patients shared common clinical features with patients with other autoimmune and inflammation-associated diseases, such as displaying low or no expression of GLUT2 on PB-IPC at baseline and exhibiting a high profile of the inflammatory cytokine interleukin (IL)-1ß. Flow cytometry demonstrated that their GLUT2 expressions on PB-IPC were also markedly upregulated, and the levels of IL-1ß-positive cells were significantly downregulated after the treatment with stem cell educator therapy. Stem cell educator therapy could upregulate the GLUT2 expression on PB-IPC and restore their function in T1D patients, leading to the improvement of clinical outcomes. The clinical data advances current understanding about the molecular mechanisms underlying the stem cell educator therapy, which can be expanded to treat patients with other autoimmune and inflammation-associated diseases.


Subject(s)
Diabetes Mellitus, Type 1 , Glucose Transporter Type 2 , Insulin-Secreting Cells , Insulin , Humans , Diabetes Mellitus, Type 1/therapy , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/blood , Glucose Transporter Type 2/metabolism , Glucose Transporter Type 2/genetics , Insulin-Secreting Cells/metabolism , Male , Female , Insulin/metabolism , Adult , Leukocytes, Mononuclear/metabolism , Middle Aged , Stem Cell Transplantation
13.
Regen Med ; 19(5): 239-246, 2024 May 03.
Article in English | MEDLINE | ID: mdl-39118533

ABSTRACT

Aim: Type II diabetes (T2D) stems from insulin resistance, with ß-cell dysfunction as a hallmark in its progression. Studies reveal that ß cells undergo apoptosis or dedifferentiation during T2D development. The transcription factor PAX4 is vital for ß differentiation and survival, thus may be a potential enhancer of ß-cell function in T2D islets. Materials & methods: Human PAX4 cDNA was delivered into T2D human islets with an adenoviral vector, and its effects on ß cells were examined. Results: PAX4 gene delivery significantly improved ß-cell survival, and increased ß-cell composition in the T2D human islets. Basal insulin and glucose-stimulated insulin secretion in PAX4-expressing islets were substantially higher than untreated or control-treated T2D human islets. Conclusion: Introduced PAX4 expression in T2D human islets improves ß-cell function, thus could provide therapeutic benefits for T2D treatment.


Type II diabetes (T2D) results from insulin resistance, with ß-cell dysfunction playing a pivotal role in its progression. Deficits in ß-cell mass and function have been attributed primarily to ß-cell death through apoptosis; however, recent studies suggest ß-cell failure can also arise from ß-cell dedifferentiation ­ that is, ß cells undergo a loss of mature identity, adopting either progenitor-like or glucagon-producing α cell states during T2D development. Therefore, a strategy preventing ß-cell dedifferentiation while promoting its survival is beneficial for T2D treatment. In this study, we explored whether PAX4, a critical transcription factor for ß differentiation and survival, could alleviate ß-cell dysfunction in human islets derived from T2D patients. To accomplish that, human PAX4 cDNA was delivered into human islets isolated from T2D donors by an adenoviral vector-based vector, Ad5.Pax4 and its effects on ß-cell function were evaluated. The results showed PAX4 expression significantly improved ß-cell survival and increased ß-cell composition in the T2D islets. Notably, PAX4-treated T2D islets exhibited significantly higher basal insulin secretion and glucose-stimulated insulin secretion than control-treated islets. The data demonstrate that PAX4 gene delivery into T2D human islets enhances ß-cell mass and function, and thus may offer therapeutic benefits in the treatment of T2D.


Subject(s)
Diabetes Mellitus, Type 2 , Homeodomain Proteins , Insulin-Secreting Cells , Insulin , Paired Box Transcription Factors , Humans , Diabetes Mellitus, Type 2/therapy , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Paired Box Transcription Factors/metabolism , Paired Box Transcription Factors/genetics , Insulin-Secreting Cells/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Insulin/metabolism , Insulin Secretion , Gene Transfer Techniques , Cell Survival , Islets of Langerhans/metabolism , Genetic Therapy/methods
14.
FASEB J ; 38(15): e23853, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39120544

ABSTRACT

Sodium butyrate (NaB) improves ß-cell function in preclinical models of diabetes; however, the mechanisms underlying these beneficial effects have not been fully elucidated. In this study, we investigated the impact of NaB on ß-cell function and calcium (Ca2+) signaling using ex vivo and in vitro models of diabetes. Our results show that NaB significantly improved glucose-stimulated insulin secretion in islets from human organ donors with type 2 diabetes and in cytokine-treated INS-1 ß cells. Consistently, NaB improved glucose-stimulated Ca2+ oscillations in mouse islets treated with proinflammatory cytokines. Because the oscillatory phenotype of Ca2+ in the ß cell is governed by changes in endoplasmic reticulum (ER) Ca2+ levels, we explored the relationship between NaB and store-operated calcium entry (SOCE), a rescue mechanism that acts to refill ER Ca2+ levels through STIM1-mediated gating of plasmalemmal Orai channels. We found that NaB treatment preserved basal ER Ca2+ levels and restored SOCE in IL-1ß-treated INS-1 cells. Furthermore, we linked these changes with the restoration of STIM1 levels in cytokine-treated INS-1 cells and mouse islets, and we found that NaB treatment was sufficient to prevent ß-cell death in response to IL-1ß treatment. Mechanistic experiments revealed that NaB mediated these beneficial effects in the ß-cell through histone deacetylase (HDAC) inhibition, iNOS suppression, and modulation of AKT-GSK-3 signaling. Taken together, these data support a model whereby NaB treatment promotes ß-cell function and Ca2+ homeostasis under proinflammatory conditions through pleiotropic effects that are linked with maintenance of SOCE. These results also suggest a relationship between ß-cell SOCE and gut microbiome-derived butyrate that may be relevant in the treatment and prevention of diabetes.


Subject(s)
Butyric Acid , Calcium , Insulin-Secreting Cells , Stromal Interaction Molecule 1 , Animals , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/drug effects , Stromal Interaction Molecule 1/metabolism , Mice , Humans , Butyric Acid/pharmacology , Calcium/metabolism , Cytokines/metabolism , Calcium Signaling/drug effects , Male , Mice, Inbred C57BL , Endoplasmic Reticulum/metabolism , Diabetes Mellitus, Type 2/metabolism
15.
Sci Rep ; 14(1): 19556, 2024 08 22.
Article in English | MEDLINE | ID: mdl-39174611

ABSTRACT

Islet amyloid polypeptide (IAPP) is co-secreted with insulin from pancreatic ß-cells. Its oligomerisation is regarded as disease driving force in type 2 diabetes (T2D) pathology. Up to now, IAPP oligomers have been detected in affected tissues. IAPP oligomer concentrations in blood have not been analysed so far. Using the IAPP single-oligomer-sensitive and monomer-insensitive surface-based fluorescence intensity distribution analysis (sFIDA) technology, levels of IAPP oligomers in blood plasma from healthy controls and people with T2D in different disease stages where determined. Subsequently, the level of IAPP oligomerisation was introduced as the ratio between the IAPP oligomers determined with sFIDA and the total IAPP concentration determined with ELISA. Highest oligomerisation levels were detected in plasma of people with T2D without late complication and without insulin therapy. Their levels stand out significantly from the control group. Healthy controls presented with the lowest oligomerisation levels in plasma. In people with T2D without complications, IAPP oligomerisation levels correlated with disease duration. The results clearly demonstrate that IAPP oligomerisation in insulin-naïve patients correlates with duration of T2D. Although a correlation per se does not identify, which is cause and what is consequence, this result supports the hypothesis that IAPP aggregation is the driving factor of T2D development and progression. The alternative and conventional hypothesis explains development of T2D with increasing insulin resistance causing exhaustion of pancreatic ß-cells due to over-secretion of insulin, and thus IAPP, too, resulting in subsequent IAPP aggregation and fibril deposition in the pancreas. Further experiments and comparative analyses with primary tissues are warranted.


Subject(s)
Diabetes Mellitus, Type 2 , Islet Amyloid Polypeptide , Humans , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Islet Amyloid Polypeptide/blood , Islet Amyloid Polypeptide/metabolism , Male , Middle Aged , Female , Aged , Protein Multimerization , Adult , Case-Control Studies , Insulin/blood , Insulin/metabolism , Insulin-Secreting Cells/metabolism
16.
Sci Rep ; 14(1): 17450, 2024 08 12.
Article in English | MEDLINE | ID: mdl-39134590

ABSTRACT

Because of the advent of genome-editing technology, gene knockout (KO) hamsters have become attractive research models for diverse diseases in humans. This study established a new KO model of diabetes by disrupting the insulin receptor substrate-2 (Irs2) gene in the golden (Syrian) hamster. Homozygous KO animals were born alive but with delayed postnatal growth until adulthood. They showed hyperglycemia, high HbA1c, and impaired glucose tolerance. However, they normally responded to insulin stimulation, unlike Irs2 KO mice, an obese type 2 diabetes (T2D) model. Consistent with this, Irs2 KO hamsters did not increase serum insulin levels upon glucose administration and showed ß-cell hypoplasia in their pancreas. Thus, our Irs2 KO hamster provide a unique T2D animal model that is distinct from the obese T2D models. This model may contribute to a better understanding of the pathophysiology of human non-obese T2D with ß-cell dysfunction, the most common type of T2D in East Asian countries, including Japan.


Subject(s)
Diabetes Mellitus, Type 2 , Disease Models, Animal , Insulin Receptor Substrate Proteins , Mesocricetus , Animals , Cricetinae , Humans , Male , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/genetics , Gene Knockout Techniques , Insulin/metabolism , Insulin Receptor Substrate Proteins/metabolism , Insulin Receptor Substrate Proteins/genetics , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology
17.
Sci Rep ; 14(1): 19234, 2024 08 20.
Article in English | MEDLINE | ID: mdl-39164380

ABSTRACT

The present study investigated the associations of serum gamma-glutamyl transferase (GGT), a marker of fatty liver and oxidative stress, and ALT/AST, a marker of fatty liver, with percentage trunk fat and postload glucose, insulin resistance, and ß-cell function in middle-aged Japanese individuals, whose BMI averaged < 23.0 kg/m2. Pancreatic ß-cell function was assessed using the disposition index calculated by a product of the insulinogenic index (IGI) and Matsuda insulin sensitivity index, a biomarker of early-phase glucose-stimulated insulin secretion and whole-body insulin sensitivity, respectively. Multivariate linear regression analyses revealed that the disposition index was associated inversely with GGT independently of percentage trunk fat, homeostasis model assessment insulin resistance (HOMA-IR), a marker of insulin resistance, and Matsuda index. When IGI was included instead of the disposition index, IGI (inversely) and HOMA-IR were associated with GGT independently of percentage trunk fat and Matsuda index. When the area under the glucose concentration curve (AUCg) during an oral glucose tolerance test was included instead of the disposition index, AUCg and HOMA-IR emerged as independent determinants of GGT. ALT/AST was associated with HOMA-IR alone. Results suggest a different pathophysiologic basis between GGT and ALT/AST in predicting diabetic risk in non-obese Japanese.


Subject(s)
Alanine Transaminase , Insulin Resistance , Insulin Secretion , Insulin-Secreting Cells , gamma-Glutamyltransferase , Humans , gamma-Glutamyltransferase/blood , gamma-Glutamyltransferase/metabolism , Insulin-Secreting Cells/metabolism , Male , Alanine Transaminase/blood , Alanine Transaminase/metabolism , Female , Middle Aged , Japan , Insulin/blood , Insulin/metabolism , Adult , Biomarkers/blood , Blood Glucose/metabolism , Blood Glucose/analysis , Glucose Tolerance Test , East Asian People
18.
Islets ; 16(1): 2392343, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-39154325

ABSTRACT

Replacement of beta cells through transplantation is a potential therapeutic approach for individuals with pancreas removal or poorly controllable type 1 diabetes. However, stress and death of beta cells pose significant challenges. Circulating miRNA has emerged as potential biomarkers reflecting early beta cell stress and death, allowing for timely intervention. The aim of this study was to identify miRNAs as potential biomarkers for beta cell health. Literature review combined with small RNA sequencing was employed to select islet-enriched miRNA. The release of those miRNA was assessed by RT-qPCR in vivo, using a streptozotocin induced diabetes mouse model and in vitro, through mouse and human islets exposed to varying degrees of hypoxic and cytokine stressors. Utilizing the streptozotocin induced model, we identified 18 miRNAs out of 39 candidate islet-enriched miRNA to be released upon islet stress in vivo. In vitro analysis of culture supernatants from cytokine and/or hypoxia stressed islets identified the release of 45 miRNAs from mouse and 8 miRNAs from human islets. Investigation into the biological pathways targeted by the cytokine- and/or hypoxia-induced miRNA suggested the involvement of MAPK and PI3K-Akt signaling pathways in both mouse and human islets. We have identified miRNAs associated with beta cell health and stress. The findings allowed us to propose a panel of 47 islet-related human miRNA that is potentially valuable for application in clinical contexts of beta cell transplantation and presymptomatic early-stage type 1 diabetes.


Subject(s)
Diabetes Mellitus, Experimental , Islets of Langerhans , MicroRNAs , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Humans , Mice , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Islets of Langerhans/metabolism , Insulin-Secreting Cells/metabolism , Real-Time Polymerase Chain Reaction/methods , Stress, Physiological/genetics , Male , RNA-Seq/methods , Mice, Inbred C57BL , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism
19.
Life Sci Alliance ; 7(11)2024 Nov.
Article in English | MEDLINE | ID: mdl-39159974

ABSTRACT

Regeneration of insulin-producing ß-cells is an alternative avenue to manage diabetes, and it is crucial to unravel this process in vivo during physiological responses to the lack of ß-cells. Here, we aimed to characterize how hepatocytes can contribute to ß-cell regeneration, either directly or indirectly via secreted proteins or metabolites, in a zebrafish model of ß-cell loss. Using lineage tracing, we show that hepatocytes do not directly convert into ß-cells even under extreme ß-cell ablation conditions. A transcriptomic analysis of isolated hepatocytes after ß-cell ablation displayed altered lipid- and glucose-related processes. Based on the transcriptomics, we performed a genetic screen that uncovers a potential role of the molybdenum cofactor (Moco) biosynthetic pathway in ß-cell regeneration and glucose metabolism in zebrafish. Consistently, molybdenum cofactor synthesis 2 (Mocs2) haploinsufficiency in mice indicated dysregulated glucose metabolism and liver function. Together, our study sheds light on the liver-pancreas crosstalk and suggests that the molybdenum cofactor biosynthesis pathway should be further studied in relation to glucose metabolism and diabetes.


Subject(s)
Coenzymes , Glucose , Hepatocytes , Insulin-Secreting Cells , Liver , Metalloproteins , Molybdenum Cofactors , Pteridines , Zebrafish , Animals , Insulin-Secreting Cells/metabolism , Pteridines/metabolism , Coenzymes/metabolism , Mice , Liver/metabolism , Liver/cytology , Metalloproteins/metabolism , Metalloproteins/genetics , Hepatocytes/metabolism , Glucose/metabolism , Regeneration/genetics , Pancreas/metabolism , Pancreas/cytology , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
20.
Immunol Rev ; 325(1): 23-45, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39166298

ABSTRACT

Type 1 diabetes (T1D) results from a complex interplay of genetic predisposition, immunological dysregulation, and environmental triggers, that culminate in the destruction of insulin-secreting pancreatic ß cells. This review provides a comprehensive examination of the multiple factors underpinning T1D pathogenesis, to elucidate key mechanisms and potential therapeutic targets. Beginning with an exploration of genetic risk factors, we dissect the roles of human leukocyte antigen (HLA) haplotypes and non-HLA gene variants associated with T1D susceptibility. Mechanistic insights gleaned from the NOD mouse model provide valuable parallels to the human disease, particularly immunological intricacies underlying ß cell-directed autoimmunity. Immunological drivers of T1D pathogenesis are examined, highlighting the pivotal contributions of both effector and regulatory T cells and the multiple functions of B cells and autoantibodies in ß-cell destruction. Furthermore, the impact of environmental risk factors, notably modulation of host immune development by the intestinal microbiome, is examined. Lastly, the review probes human longitudinal studies, unveiling the dynamic interplay between mucosal immunity, systemic antimicrobial antibody responses, and the trajectories of T1D development. Insights garnered from these interconnected factors pave the way for targeted interventions and the identification of biomarkers to enhance T1D management and prevention strategies.


Subject(s)
Autoimmunity , Diabetes Mellitus, Type 1 , Gastrointestinal Microbiome , Genetic Predisposition to Disease , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/genetics , Humans , Animals , Gastrointestinal Microbiome/immunology , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/metabolism , Gene-Environment Interaction , Autoantibodies/immunology , HLA Antigens/genetics , HLA Antigens/immunology , Mice , Disease Models, Animal , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL