Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 17.682
Filtrer
1.
Sci Rep ; 14(1): 17844, 2024 08 01.
Article de Anglais | MEDLINE | ID: mdl-39090166

RÉSUMÉ

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.


Sujet(s)
Différenciation cellulaire , Techniques de coculture , Vésicules extracellulaires , Cellules à insuline , Insuline , Cellules souches mésenchymateuses , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , Humains , Vésicules extracellulaires/métabolisme , Insuline/métabolisme , Cellules à insuline/métabolisme , Cellules à insuline/cytologie , Peptide C/métabolisme , Cellules cultivées , Glucose/métabolisme , Tissu adipeux/cytologie , Tissu adipeux/métabolisme
2.
Nat Commun ; 15(1): 6627, 2024 Aug 05.
Article de Anglais | MEDLINE | ID: mdl-39103322

RÉSUMÉ

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.


Sujet(s)
Diabète de type 2 , Cellules à insuline , Récepteur delta , Récepteur delta/métabolisme , Récepteur delta/génétique , Diabète de type 2/génétique , Diabète de type 2/métabolisme , Humains , Cellules à insuline/métabolisme , Mâle , Femelle , Adulte d'âge moyen , Insuline/métabolisme , Sécrétion d'insuline/effets des médicaments et des substances chimiques , Sécrétion d'insuline/génétique , Adulte
3.
Cell Mol Life Sci ; 81(1): 345, 2024 Aug 12.
Article de Anglais | MEDLINE | ID: mdl-39133305

RÉSUMÉ

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.


Sujet(s)
Calgranuline A , Calgranuline B , Virus de l'hépatite B , Cellules à insuline , Interféron gamma , Cellules tueuses naturelles , Cirrhose du foie , Nécroptose , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Humains , Animaux , Interféron gamma/métabolisme , Calgranuline B/métabolisme , Cirrhose du foie/anatomopathologie , Cirrhose du foie/métabolisme , Cirrhose du foie/virologie , Cirrhose du foie/immunologie , Souris , Mâle , Cellules à insuline/métabolisme , Cellules à insuline/anatomopathologie , Cellules à insuline/virologie , Calgranuline A/métabolisme , Souris de lignée C57BL , Femelle , Adulte d'âge moyen , Hépatite B/complications , Hépatite B/anatomopathologie , Hépatite B/métabolisme , Modèles animaux de maladie humaine , Tétrachloro-méthane
4.
Regen Med ; 19(5): 239-246, 2024 May 03.
Article de Anglais | MEDLINE | ID: mdl-39118533

RÉSUMÉ

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.


Sujet(s)
Diabète de type 2 , Protéines à homéodomaine , Cellules à insuline , Insuline , Facteurs de transcription PAX , Humains , Diabète de type 2/thérapie , Diabète de type 2/génétique , Diabète de type 2/métabolisme , Facteurs de transcription PAX/métabolisme , Facteurs de transcription PAX/génétique , Cellules à insuline/métabolisme , Protéines à homéodomaine/génétique , Protéines à homéodomaine/métabolisme , Insuline/métabolisme , Sécrétion d'insuline , Techniques de transfert de gènes , Survie cellulaire , Ilots pancréatiques/métabolisme , Thérapie génétique/méthodes
5.
FASEB J ; 38(15): e23853, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-39120544

RÉSUMÉ

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.


Sujet(s)
Acide butyrique , Calcium , Cellules à insuline , Molécule-1 d'interaction stromale , Animaux , Cellules à insuline/métabolisme , Cellules à insuline/effets des médicaments et des substances chimiques , Molécule-1 d'interaction stromale/métabolisme , Souris , Humains , Acide butyrique/pharmacologie , Calcium/métabolisme , Cytokines/métabolisme , Signalisation calcique/effets des médicaments et des substances chimiques , Mâle , Souris de lignée C57BL , Réticulum endoplasmique/métabolisme , Diabète de type 2/métabolisme
6.
Sci Rep ; 14(1): 17450, 2024 08 12.
Article de Anglais | MEDLINE | ID: mdl-39134590

RÉSUMÉ

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.


Sujet(s)
Diabète de type 2 , Modèles animaux de maladie humaine , Substrats du récepteur à l'insuline , Cellules à insuline , Mesocricetus , Animaux , Cellules à insuline/métabolisme , Cellules à insuline/anatomopathologie , Diabète de type 2/métabolisme , Diabète de type 2/génétique , Substrats du récepteur à l'insuline/métabolisme , Substrats du récepteur à l'insuline/génétique , Cricetinae , Insuline/métabolisme , Techniques de knock-out de gènes , Mâle , Humains , Glycémie/métabolisme
7.
Cells ; 13(15)2024 Jul 24.
Article de Anglais | MEDLINE | ID: mdl-39120275

RÉSUMÉ

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.


Sujet(s)
Diabète , Cellules à insuline , Transduction du signal , Humains , Transduction du signal/effets des médicaments et des substances chimiques , Cellules à insuline/métabolisme , Cellules à insuline/effets des médicaments et des substances chimiques , Cellules à insuline/anatomopathologie , Animaux , Diabète/métabolisme , Diabète/traitement médicamenteux , Diabète/anatomopathologie , Hypoglycémiants/pharmacologie , Hypoglycémiants/usage thérapeutique , Survie cellulaire/effets des médicaments et des substances chimiques
8.
Nutrients ; 16(15)2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39125265

RÉSUMÉ

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.


Sujet(s)
Hyperglycémie , Sécrétion d'insuline , Cellules à insuline , Ostéocalcine , Animaux , Ostéocalcine/métabolisme , Cellules à insuline/métabolisme , Cellules à insuline/effets des médicaments et des substances chimiques , Hyperglycémie/métabolisme , Rats , Sécrétion d'insuline/effets des médicaments et des substances chimiques , Insuline/métabolisme , Glucose/métabolisme , Calcium/métabolisme , Lignée cellulaire , Signalisation calcique/effets des médicaments et des substances chimiques
9.
Int J Mol Sci ; 25(15)2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39125590

RÉSUMÉ

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.


Sujet(s)
Insuline , Souris de lignée C57BL , Pancréas , Protéomique , Transduction du signal , Protéines G rap1 , Animaux , Protéines G rap1/métabolisme , Protéines G rap1/génétique , Souris , Protéomique/méthodes , Insuline/métabolisme , Pancréas/métabolisme , Cellules à insuline/métabolisme , Souris knockout , Facteurs d'échange de nucléotides guanyliques/métabolisme , Facteurs d'échange de nucléotides guanyliques/génétique , Sécrétion d'insuline , Mâle , Glucose/métabolisme
10.
Int J Mol Sci ; 25(15)2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-39125908

RÉSUMÉ

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.


Sujet(s)
Diabète de type 1 , Transporteur de glucose de type 2 , Cellules à insuline , Insuline , Humains , Diabète de type 1/thérapie , Diabète de type 1/métabolisme , Diabète de type 1/sang , Transporteur de glucose de type 2/métabolisme , Transporteur de glucose de type 2/génétique , Cellules à insuline/métabolisme , Mâle , Femelle , Insuline/métabolisme , Adulte , Agranulocytes/métabolisme , Adulte d'âge moyen , Transplantation de cellules souches
11.
Sci Rep ; 14(1): 18590, 2024 08 10.
Article de Anglais | MEDLINE | ID: mdl-39127728

RÉSUMÉ

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.


Sujet(s)
Tissu adipeux , Glycémie , Hyperglycémie provoquée , Insulinorésistance , Cellules à insuline , Insuline , Humains , Femelle , Cellules à insuline/métabolisme , Tissu adipeux/métabolisme , Glycémie/métabolisme , Jeune adulte , Adulte , Japon , Insuline/sang , Insuline/métabolisme , Études transversales , Jeûne/sang , Peuples d'Asie de l'Est
12.
AIDS ; 38(11): 1725-1727, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-39088830

RÉSUMÉ

We explored the impact of immune dysregulation on pancreatic beta cell injury in HIV patients. Analyzing 105 participant samples, we observed lower IL-21 levels and elevated immune checkpoint levels (e.g. PD-1, CD27+, CD40+) in untreated HIV patients. Notably, soluble TIM-3 correlated positively with improved beta cell function and inversely with beta cell stress, suggesting its potential role in beta cell protection in untreated HIV.


Sujet(s)
Infections à VIH , Cellules à insuline , Humains , Infections à VIH/complications , Infections à VIH/immunologie , Cellules à insuline/immunologie , Mâle , Femelle , Adulte d'âge moyen , Adulte , Récepteur cellulaire-2 du virus de l'hépatite A , Interleukines/sang , Protéines de points de contrôle immunitaires/métabolisme
13.
FASEB J ; 38(16): e23885, 2024 Aug 31.
Article de Anglais | MEDLINE | ID: mdl-39139039

RÉSUMÉ

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.


Sujet(s)
Épigenèse génétique , Cellules à insuline , Protein-Serine-Threonine Kinases , Animaux , Cellules à insuline/métabolisme , Souris , Protein-Serine-Threonine Kinases/génétique , Protein-Serine-Threonine Kinases/métabolisme , microARN/génétique , microARN/métabolisme , Souris knockout , AMP-Activated Protein Kinases/métabolisme , AMP-Activated Protein Kinases/génétique , Régions promotrices (génétique) , Souris de lignée C57BL , Mâle
14.
Front Endocrinol (Lausanne) ; 15: 1417437, 2024.
Article de Anglais | MEDLINE | ID: mdl-39114287

RÉSUMÉ

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.


Sujet(s)
Régime occidental , Cellules à insuline , Phénomènes physiologiques nutritionnels maternels , Mitochondries , Effets différés de l'exposition prénatale à des facteurs de risque , Animaux , Femelle , Cellules à insuline/métabolisme , Cellules à insuline/ultrastructure , Grossesse , Mitochondries/métabolisme , Mitochondries/ultrastructure , Régime occidental/effets indésirables , Effets différés de l'exposition prénatale à des facteurs de risque/métabolisme , Effets différés de l'exposition prénatale à des facteurs de risque/anatomopathologie , Mâle , Lactation
15.
Front Immunol ; 15: 1393248, 2024.
Article de Anglais | MEDLINE | ID: mdl-39114661

RÉSUMÉ

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.


Sujet(s)
Chaperonne BiP du réticulum endoplasmique , Stress du réticulum endoplasmique , Vésicules extracellulaires , Cellules à insuline , microARN , Monocytes , Vésicules extracellulaires/métabolisme , Vésicules extracellulaires/immunologie , Monocytes/immunologie , Monocytes/métabolisme , Cellules à insuline/métabolisme , Cellules à insuline/immunologie , Humains , Stress du réticulum endoplasmique/immunologie , microARN/génétique , Inflammation/immunologie , Inflammation/métabolisme , Diabète de type 1/immunologie , Diabète de type 1/métabolisme , Animaux , Lignée cellulaire , Ilots pancréatiques/immunologie , Ilots pancréatiques/métabolisme , Transduction du signal , Réponse aux protéines mal repliées/immunologie
16.
Nat Commun ; 15(1): 6344, 2024 Jul 27.
Article de Anglais | MEDLINE | ID: mdl-39068220

RÉSUMÉ

Dysfunction of pancreatic δ cells contributes to the etiology of diabetes. Despite their important role, human δ cells are scarce, limiting physiological studies and drug discovery targeting δ cells. To date, no directed δ-cell differentiation method has been established. Here, we demonstrate that fibroblast growth factor (FGF) 7 promotes pancreatic endoderm/progenitor differentiation, whereas FGF2 biases cells towards the pancreatic δ-cell lineage via FGF receptor 1. We develop a differentiation method to generate δ cells from human stem cells by combining FGF2 with FGF7, which synergistically directs pancreatic lineage differentiation and modulates the expression of transcription factors and SST activators during endoderm/endocrine precursor induction. These δ cells display mature RNA profiles and fine secretory granules, secrete somatostatin in response to various stimuli, and suppress insulin secretion from in vitro co-cultured ß cells and mouse ß cells upon transplantation. The generation of human pancreatic δ cells from stem cells in vitro would provide an unprecedented cell source for drug discovery and cell transplantation studies in diabetes.


Sujet(s)
Différenciation cellulaire , Cellules souches pluripotentes , Humains , Animaux , Souris , Cellules souches pluripotentes/métabolisme , Cellules souches pluripotentes/cytologie , Facteur de croissance fibroblastique de type 2/métabolisme , Facteur de croissance fibroblastique de type 2/pharmacologie , Cellules à insuline/métabolisme , Cellules à insuline/cytologie , Cellules à somatostatine/métabolisme , Cellules à somatostatine/cytologie , Endoderme/cytologie , Endoderme/métabolisme , Récepteur FGFR1/métabolisme , Récepteur FGFR1/génétique , Pancréas/cytologie , Pancréas/métabolisme , Somatostatine/métabolisme , Lignage cellulaire , Insuline/métabolisme , Sécrétion d'insuline
17.
Front Immunol ; 15: 1403752, 2024.
Article de Anglais | MEDLINE | ID: mdl-38975343

RÉSUMÉ

Type 1 diabetes (T1D) arises from autoimmune-mediated destruction of insulin-producing pancreatic beta cells. Recent advancements in the technology of generating pancreatic beta cells from human pluripotent stem cells (SC-beta cells) have facilitated the exploration of cell replacement therapies for treating T1D. However, the persistent threat of autoimmunity poses a significant challenge to the survival of transplanted SC-beta cells. Genetic engineering is a promising approach to enhance immune resistance of beta cells as we previously showed by inactivating the Renalase (Rnls) gene. Here, we demonstrate that Rnls loss of function in beta cells shapes autoimmunity by mediating a regulatory natural killer (NK) cell phenotype important for the induction of tolerogenic antigen-presenting cells. Rnls-deficient beta cells mediate cell-cell contact-independent induction of hallmark anti-inflammatory cytokine Tgfß1 in NK cells. In addition, surface expression of regulatory NK immune checkpoints CD47 and Ceacam1 is markedly elevated on beta cells deficient for Rnls. Altered glucose metabolism in Rnls mutant beta cells is involved in the upregulation of CD47 surface expression. These findings are crucial to better understand how genetically engineered beta cells shape autoimmunity, giving valuable insights for future therapeutic advancements to treat and cure T1D.


Sujet(s)
Auto-immunité , Diabète de type 1 , Cellules à insuline , Cellules tueuses naturelles , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Animaux , Cellules à insuline/immunologie , Cellules à insuline/métabolisme , Souris , Diabète de type 1/immunologie , Humains , Antigènes CD47/métabolisme , Antigènes CD47/génétique , Antigènes CD47/immunologie , Facteur de croissance transformant bêta-1/métabolisme , Souris de lignée NOD , Monoamine oxidase
18.
Endocrinology ; 165(8)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38954536

RÉSUMÉ

BACKGROUND: Nephrin is a transmembrane protein with well-established signaling roles in kidney podocytes, and a smaller set of secretory functions in pancreatic ß cells are implicated in diabetes. Nephrin signaling is mediated in part through its 3 cytoplasmic YDxV motifs, which can be tyrosine phosphorylated by high glucose and ß cell injuries. Although in vitro studies demonstrate these phosphorylated motifs can regulate ß cell vesicle trafficking and insulin release, in vivo evidence of their role in this cell type remains to be determined. METHODS: To further explore the role of nephrin YDxV phosphorylation in ß cells, we used a mouse line with tyrosine to phenylalanine substitutions at each YDxV motif (nephrin-Y3F) to inhibit phosphorylation. We assessed islet function via primary islet glucose-stimulated insulin secretion assays and oral glucose tolerance tests. RESULTS: Nephrin-Y3F mice successfully developed pancreatic endocrine and exocrine tissues with minimal structural differences. Unexpectedly, male and female nephrin-Y3F mice showed elevated insulin secretion, with a stronger increase observed in male mice. At 8 months of age, no differences in glucose tolerance were observed between wild-type (WT) and nephrin-Y3F mice. However, aged nephrin-Y3F mice (16 months of age) demonstrated more rapid glucose clearance compared to WT controls. CONCLUSION: Taken together, loss of nephrin YDxV phosphorylation does not alter baseline islet function. Instead, our data suggest a mechanism linking impaired nephrin YDxV phosphorylation to improved islet secretory ability with age. Targeting nephrin phosphorylation could provide novel therapeutic opportunities to improve ß cell function.


Sujet(s)
Hyperglycémie provoquée , Sécrétion d'insuline , Cellules à insuline , Insuline , Protéines membranaires , Animaux , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Phosphorylation , Souris , Mâle , Sécrétion d'insuline/physiologie , Cellules à insuline/métabolisme , Femelle , Insuline/métabolisme , Tyrosine/métabolisme , Vieillissement/métabolisme , Intolérance au glucose/métabolisme , Souris de lignée C57BL , Glucose/métabolisme
19.
Front Endocrinol (Lausanne) ; 15: 1377918, 2024.
Article de Anglais | MEDLINE | ID: mdl-38962677

RÉSUMÉ

With changes in lifestyle behaviors, including dietary structure and habits, the prevalence of Youth-onset Type 2 Diabetes Mellitus (YODM) has increased 2 to 3 times compared to 30 years ago. YODM patients experience complications earlier, progress faster, and exhibit more severe symptoms. However, limited and inconclusive direct evidence, coupled with poor patient compliance, poses challenges in the clinical management of YODM. Apart from the continuous decline in pancreatic ß-cell function and quantity, tissue-specific insulin resistance (IR) is also a typical characteristic of YODM. The main mechanisms of IR in YODM involve different aspects such as obesity, dietary imbalance, abnormal substance metabolism, chronic inflammation, oxidative stress, and hormonal fluctuations during adolescence. For the comprehensive management of YODM, besides achieving good control of blood glucose levels, it may be necessary to apply the most appropriate methods considering the uniqueness of the patient population and the specifics of the disease. Early identification and detection of the disease are crucial. Precise screening of patients with well-functioning pancreatic insulin ß-cells, primarily characterized by IR and obesity, represents the population most likely to achieve diabetes remission or reversal through lifestyle modifications, medications, or even surgical interventions. Additionally, considering potential emotional disorders or the impact of adolescent hormones in these patients, health education for patients and caregivers is essential to make them aware of the long-term benefits of well-controlled blood glucose. In conclusion, adopting comprehensive management measures to achieve diabetes remission or reversal is the ideal goal. Controlling high blood glucose, obesity, and other risk factors related to diabetes complications is the next priority to delay the occurrence and progression of complications. A comprehensive perspective on IR provides insights and references for identifying YODM and its management strategies.


Sujet(s)
Diabète de type 2 , Insulinorésistance , Humains , Diabète de type 2/thérapie , Diabète de type 2/épidémiologie , Diabète de type 2/métabolisme , Adolescent , Prise en charge de la maladie , Mode de vie , Obésité/thérapie , Obésité/épidémiologie , Cellules à insuline/métabolisme
20.
Diabetes ; 73(8): 1255-1265, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38985991

RÉSUMÉ

Inducible pluripotent stem cell-derived human ß-like cells (BLCs) hold promise for both therapy and disease modeling, but their generation remains challenging and their functional analyses beyond transcriptomic and morphological assessments remain limited. Here, we validate an approach using multicellular and single-cell electrophysiological tools to evaluate function of BLCs from pioneer protocols that can be easily adapted to more differentiated BLCs. The multi-electrode arrays (MEAs) measuring the extracellular electrical activity revealed that BLCs, like primary ß-cells, are electrically coupled and produce slow potential (SP) signals that are closely linked to insulin secretion. We also used high-resolution single-cell patch clamp measurements to capture the exocytotic properties, and characterize voltage-gated sodium and calcium currents, and found that they were comparable with those in primary ß- and EndoC-ßH1 cells. The KATP channel conductance is greater than in human primary ß-cells, which may account for the limited glucose responsiveness observed with MEA. We used MEAs to study the impact of the type 2 diabetes-protective SLC30A8 allele (p.Lys34Serfs50*) and found that BLCs with this allele have stronger electrical coupling activity. Our data suggest that BLCs can be used to evaluate the functional impact of genetic variants on ß-cell function and coupling.


Sujet(s)
Cellules souches pluripotentes induites , Cellules à insuline , Transporteur de zinc ZnT-8 , Humains , Cellules souches pluripotentes induites/métabolisme , Cellules à insuline/métabolisme , Cellules à insuline/physiologie , Transporteur de zinc ZnT-8/génétique , Transporteur de zinc ZnT-8/métabolisme , Différenciation cellulaire , Diabète de type 2/métabolisme , Diabète de type 2/génétique , Techniques de patch-clamp , Phénomènes électrophysiologiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE