Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 5.138
Filtrer
1.
Islets ; 16(1): 2379650, 2024 Dec 31.
Article de Anglais | MEDLINE | ID: mdl-39028826

RÉSUMÉ

Islet amyloid polypeptide (IAPP) is a factor that regulates food intake and is secreted from both pancreatic islets and insulinoma cells. Here, we aimed to evaluate IAPP immunohistochemically in islets or insulinoma cells in association with clinical characteristics. We recruited six insulinoma patients and six body mass index-matched control patients with pancreatic diseases other than insulinoma whose glucose tolerance was confirmed to be normal preoperatively. IAPP and IAPP-insulin double staining were performed on pancreatic surgical specimens. We observed that the IAPP staining level and percentage of IAPP-positive beta cells tended to be lower (p = 0.1699) in the islets of insulinoma patients than in those of control patients, which might represent a novel IAPP expression pattern under persistent hyperinsulinemia and hypoglycemia.


Sujet(s)
Insulinome , Polypeptide amyloïde des ilots , Ilots pancréatiques , Tumeurs du pancréas , Insulinome/métabolisme , Insulinome/anatomopathologie , Humains , Mâle , Femelle , Polypeptide amyloïde des ilots/métabolisme , Adulte d'âge moyen , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/anatomopathologie , Ilots pancréatiques/métabolisme , Ilots pancréatiques/anatomopathologie , Adulte , Sujet âgé , Immunohistochimie , Insuline/métabolisme
2.
Sci Rep ; 14(1): 14669, 2024 06 25.
Article de Anglais | MEDLINE | ID: mdl-38918575

RÉSUMÉ

Non-obese diabetes (NOD) mice are an established, spontaneous model of type 1 diabetes in which diabetes develops through insulitis. Using next-generation sequencing, coupled with pathway analysis, the molecular fingerprint of early insulitis was mapped in a cohort of mice ranging from 4 to 12 weeks of age. The resulting dynamic timeline revealed an initial decrease in proliferative capacity followed by the emergence of an inflammatory signature between 6 and 8 weeks that increased to a regulatory plateau between 10 and 12 weeks. The inflammatory signature is identified by the activation of central immunogenic factors such as Infg, Il1b, and Tnfa, and activation of canonical inflammatory signaling. Analysis of the regulatory landscape revealed the transcription factor Atf3 as a potential novel modulator of inflammatory signaling in the NOD islets. Furthermore, the Hedgehog signaling pathway correlated with Atf3 regulation, suggesting that the two play a role in regulating islet inflammation; however, further studies are needed to establish the nature of this connection.


Sujet(s)
Facteur de transcription ATF-3 , Diabète de type 1 , Ilots pancréatiques , Souris de lignée NOD , Transduction du signal , Animaux , Ilots pancréatiques/métabolisme , Ilots pancréatiques/anatomopathologie , Facteur de transcription ATF-3/métabolisme , Facteur de transcription ATF-3/génétique , Souris , Diabète de type 1/génétique , Diabète de type 1/métabolisme , Diabète de type 1/anatomopathologie , Femelle , Inflammation/génétique , Inflammation/anatomopathologie , Inflammation/métabolisme , Protéines Hedgehog/métabolisme , Protéines Hedgehog/génétique , Analyse de profil d'expression de gènes , Modèles animaux de maladie humaine
3.
Cell Rep ; 43(6): 114346, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38850534

RÉSUMÉ

Histopathological heterogeneity in the human pancreas is well documented; however, functional evidence at the tissue level is scarce. Herein, we investigate in situ glucose-stimulated islet and carbachol-stimulated acinar cell secretion across the pancreas head (PH), body (PB), and tail (PT) regions in donors without diabetes (ND; n = 15), positive for one islet autoantibody (1AAb+; n = 7), and with type 1 diabetes (T1D; <14 months duration, n = 5). Insulin, glucagon, pancreatic amylase, lipase, and trypsinogen secretion along with 3D tissue morphometrical features are comparable across regions in ND. In T1D, insulin secretion and beta-cell volume are significantly reduced within all regions, while glucagon and enzymes are unaltered. Beta-cell volume is lower despite normal insulin secretion in 1AAb+, resulting in increased volume-adjusted insulin secretion versus ND. Islet and acinar cell secretion in 1AAb+ are consistent across the PH, PB, and PT. This study supports low inter-regional variation in pancreas slice function and, potentially, increased metabolic demand in 1AAb+.


Sujet(s)
Diabète de type 1 , Insuline , Ilots pancréatiques , Humains , Diabète de type 1/anatomopathologie , Diabète de type 1/métabolisme , Ilots pancréatiques/métabolisme , Ilots pancréatiques/anatomopathologie , Mâle , Insuline/métabolisme , Femelle , Sécrétion d'insuline/effets des médicaments et des substances chimiques , Adulte , Adulte d'âge moyen , Cellules à insuline/métabolisme , Cellules à insuline/anatomopathologie , Cellules acineuses/métabolisme , Cellules acineuses/anatomopathologie , Glucagon/métabolisme , Glucose/métabolisme , Autoanticorps/immunologie , Amylases/métabolisme
4.
Commun Biol ; 7(1): 778, 2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-38937540

RÉSUMÉ

The prevalent RNA alternative splicing (AS) contributes to molecular diversity, which has been demonstrated in cellular function regulation and disease pathogenesis. However, the contribution of AS in pancreatic islets during diabetes progression remains unclear. Here, we reanalyze the full-length single-cell RNA sequencing data from the deposited database to investigate AS regulation across human pancreatic endocrine cell types in non-diabetic (ND) and type 2 diabetic (T2D) individuals. Our analysis demonstrates the significant association between transcriptomic AS profiles and cell-type-specificity, which could be applied to distinguish the clustering of major endocrine cell types. Moreover, AS profiles are enabled to clearly define the mature subset of ß-cells in healthy controls, which is completely lost in T2D. Further analysis reveals that RNA-binding proteins (RBPs), heterogeneous nuclear ribonucleoproteins (hnRNPs) and FXR1 family proteins are predicted to induce the functional impairment of ß-cells through regulating AS profiles. Finally, trajectory analysis of endocrine cells suggests the ß-cell identity shift through dedifferentiation and transdifferentiation of ß-cells during the progression of T2D. Together, our study provides a mechanism for regulating ß-cell functions and suggests the significant contribution of AS program during diabetes pathogenesis.


Sujet(s)
Épissage alternatif , Diabète de type 2 , Cellules à insuline , Analyse de séquence d'ARN , Analyse sur cellule unique , Diabète de type 2/génétique , Diabète de type 2/métabolisme , Humains , Cellules à insuline/métabolisme , Cellules à insuline/anatomopathologie , Transcriptome , Protéines de liaison à l'ARN/génétique , Protéines de liaison à l'ARN/métabolisme , Ilots pancréatiques/métabolisme , Ilots pancréatiques/anatomopathologie
5.
Life Sci ; 351: 122854, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-38901688

RÉSUMÉ

AIMS: To evaluate the cell proliferation and death, and structural morphology of the pancreatic islet cells of the rats with hyperglycemia in the first month of life and compare to those of the control rats. MAIN METHODS: Female Sprague-Dawley newborn rats received Streptozotocin (a beta-cytotoxic drug) at birth for diabetes induction. Control and hyperglycemic animals were euthanized on different days of life: 5, 10, 15, and 30. The pancreas was collected and processed for immunohistochemical analysis of cleaved Caspase-3 (cell death), Ki-67 (cell proliferation), PDX-1 (transcription factor responsible for insulin synthesis), and endocrine hormones (insulin, glucagon, and somatostatin). KEY FINDINGS: Control females showed a higher percentage (%) of Ki-67-positive(+) cells on D10 and D15, a higher % of insulin+ and somatostatin+ cells on D15 and D30, a lower % of PDX-1+ cells on D10, and a higher % of glucagon+ cells on D10 and D30. Hyperglycemic females showed a lower % of Ki-67+ cells on D15, a higher % of cleaved Caspase-3+ cells on D15, and insulin+ cells on D15 and D30. In the comparison among the experimental groups, the hyperglycemic females showed an increased % of cleaved Caspase-3+ and Ki-67+ cells and a lower % of PDX-1+ cells. SIGNIFICANCE: This study enabled a better understanding of the abnormal pancreas development regarding cellular proliferation, apoptosis, and hormonal synthesis in the neonatal period. Thus, the pancreatic islets of hyperglycemic rats do not reestablish the normal endocrine cell population, and cellular apoptosis overcame the proliferative activity of these cells.


Sujet(s)
Animaux nouveau-nés , Prolifération cellulaire , Hyperglycémie , Ilots pancréatiques , Rat Sprague-Dawley , Animaux , Femelle , Hyperglycémie/métabolisme , Hyperglycémie/anatomopathologie , Rats , Ilots pancréatiques/métabolisme , Ilots pancréatiques/anatomopathologie , Diabète expérimental/anatomopathologie , Diabète expérimental/métabolisme , Mort cellulaire , Glucagon/métabolisme , Insuline/métabolisme , Antigène KI-67/métabolisme , Caspase-3/métabolisme , Somatostatine/métabolisme , Apoptose , Transactivateurs , Protéines à homéodomaine
6.
Biochem Biophys Res Commun ; 725: 150254, 2024 Sep 17.
Article de Anglais | MEDLINE | ID: mdl-38901223

RÉSUMÉ

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


Sujet(s)
Composés benzhydryliques , Méthylation de l'ADN , Diabète de type 2 , Glucosides , Ilots pancréatiques , Inhibiteurs du cotransporteur sodium-glucose de type 2 , Animaux , Composés benzhydryliques/pharmacologie , Composés benzhydryliques/usage thérapeutique , Méthylation de l'ADN/effets des médicaments et des substances chimiques , Glucosides/pharmacologie , Glucosides/usage thérapeutique , Diabète de type 2/métabolisme , Diabète de type 2/génétique , Diabète de type 2/traitement médicamenteux , Diabète de type 2/anatomopathologie , Souris , Ilots pancréatiques/métabolisme , Ilots pancréatiques/effets des médicaments et des substances chimiques , Ilots pancréatiques/anatomopathologie , Mâle , Inhibiteurs du cotransporteur sodium-glucose de type 2/pharmacologie , Souris de lignée C57BL , Modèles animaux de maladie humaine , Diabète expérimental/génétique , Diabète expérimental/métabolisme , Diabète expérimental/traitement médicamenteux , Diabète expérimental/anatomopathologie , Épigenèse génétique/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Cadhérines/métabolisme , Cadhérines/génétique
7.
J Pathol ; 263(4-5): 429-441, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38837231

RÉSUMÉ

The Ppy gene encodes pancreatic polypeptide (PP) secreted by PP- or γ-cells, which are a subtype of endocrine cells localised mainly in the islet periphery. For a detailed characterisation of PP cells, we aimed to establish PP cell lines. To this end, we generated a mouse model harbouring the SV40 large T antigen (TAg) in the Rosa26 locus, which is expressed upon Ppy-promoter-mediated Cre-loxP recombination. Whereas Insulin1-CreERT-mediated TAg expression in beta cells resulted in insulinoma, surprisingly, Ppy-Cre-mediated TAg expression resulted in the malignant transformation of Ppy-lineage cells. These mice showed distorted islet structural integrity at 5 days of age compared with normal islets. CK19+ duct-like lesions contiguous with the islets were observed at 2 weeks of age, and mice developed aggressive pancreatic ductal adenocarcinoma (PDAC) at 4 weeks of age, suggesting that PDAC can originate from the islet/endocrine pancreas. This was unexpected as PDAC is believed to originate from the exocrine pancreas. RNA-sequencing analysis of Ppy-lineage islet cells from 7-day-old TAg+ mice showed a downregulation and an upregulation of endocrine and exocrine genes, respectively, in addition to the upregulation of genes and pathways associated with PDAC. These results suggest that the expression of an oncogene in Ppy-lineage cells induces a switch from endocrine cell fate to PDAC. Our findings demonstrate that Ppy-lineage cells may be an origin of PDAC and may provide novel insights into the pathogenesis of pancreatic cancer, as well as possible therapeutic strategies. © 2024 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Sujet(s)
Carcinome du canal pancréatique , Lignage cellulaire , Tumeurs du pancréas , Animaux , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/métabolisme , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/génétique , Tumeurs du pancréas/métabolisme , Souris , Souris transgéniques , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/anatomopathologie , Transformation cellulaire néoplasique/métabolisme , Ilots pancréatiques/anatomopathologie , Ilots pancréatiques/métabolisme , Antigènes transformants de polyomavirus/génétique , Antigènes transformants de polyomavirus/métabolisme , Régulation de l'expression des gènes tumoraux , Humains
8.
J Clin Invest ; 134(14)2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38885342

RÉSUMÉ

While inflammation is beneficial for insulin secretion during homeostasis, its transformation adversely affects ß cells and contributes to diabetes. However, the regulation of islet inflammation for maintaining glucose homeostasis remains largely unknown. Here, we identified pericytes as pivotal regulators of islet immune and ß cell function in health. Islets and pancreatic pericytes express various cytokines in healthy humans and mice. To interfere with the pericytic inflammatory response, we selectively inhibited the TLR/MyD88 pathway in these cells in transgenic mice. The loss of MyD88 impaired pericytic cytokine production. Furthermore, MyD88-deficient mice exhibited skewed islet inflammation with fewer cells, an impaired macrophage phenotype, and reduced IL-1ß production. This aberrant pericyte-orchestrated islet inflammation was associated with ß cell dedifferentiation and impaired glucose response. Additionally, we found that Cxcl1, a pericytic MyD88-dependent cytokine, promoted immune IL-1ß production. Treatment with either Cxcl1 or IL-1ß restored the mature ß cell phenotype and glucose response in transgenic mice, suggesting a potential mechanism through which pericytes and immune cells regulate glucose homeostasis. Our study revealed pericyte-orchestrated islet inflammation as a crucial element in glucose regulation, implicating this process as a potential therapeutic target for diabetes.


Sujet(s)
Inflammation , Interleukine-1 bêta , Facteur de différenciation myéloïde-88 , Péricytes , Transduction du signal , Animaux , Facteur de différenciation myéloïde-88/génétique , Facteur de différenciation myéloïde-88/métabolisme , Souris , Péricytes/métabolisme , Péricytes/anatomopathologie , Péricytes/immunologie , Humains , Inflammation/anatomopathologie , Inflammation/métabolisme , Inflammation/génétique , Inflammation/immunologie , Interleukine-1 bêta/métabolisme , Interleukine-1 bêta/génétique , Interleukine-1 bêta/immunologie , Souris transgéniques , Récepteurs de type Toll/métabolisme , Récepteurs de type Toll/génétique , Chimiokine CXCL1/métabolisme , Chimiokine CXCL1/génétique , Ilots pancréatiques/immunologie , Ilots pancréatiques/métabolisme , Ilots pancréatiques/anatomopathologie , Souris knockout , Cellules à insuline/métabolisme , Cellules à insuline/anatomopathologie , Cellules à insuline/immunologie , Mâle , Glucose/métabolisme
9.
Peptides ; 178: 171254, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38815655

RÉSUMÉ

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


Sujet(s)
Alimentation riche en graisse , Peptide gastrointestinal , Glucagon-like peptide 1 , Incrétines , Cellules à insuline , Animaux , Peptide gastrointestinal/pharmacologie , Peptide gastrointestinal/métabolisme , Mâle , Glucagon-like peptide 1/métabolisme , Glucagon-like peptide 1/pharmacologie , Souris , Cellules à insuline/effets des médicaments et des substances chimiques , Cellules à insuline/métabolisme , Alimentation riche en graisse/effets indésirables , Incrétines/pharmacologie , Incrétines/métabolisme , Fragments peptidiques/pharmacologie , Ilots pancréatiques/effets des médicaments et des substances chimiques , Ilots pancréatiques/métabolisme , Ilots pancréatiques/anatomopathologie , Apoptose/effets des médicaments et des substances chimiques , Insuline/métabolisme
11.
Am J Surg Pathol ; 48(7): 834-838, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38722694

RÉSUMÉ

The presence of epithelial cells within lymph node parenchyma is typically indicative of a metastatic malignancy. However, there are rare instances in which non-neoplastic epithelial or epithelioid cells may be found within lymph nodes, either due to aberrant embryologic migration, mechanical displacement, or physiological trafficking. These can potentially lead to serious potential diagnostic pitfalls, as when such situations are encountered by surgical pathologists, there is substantial risk of overdiagnosing these as metastatic malignancy. Herein, we describe 2 cases of benign pancreatic islet cells within peripancreatic lymph nodes, and underscore the potential for misdiagnosis of this phenomenon as foci of metastatic well-differentiated neuroendocrine tumor. The benign nature of these intranodal islet cells was supported by: (1) the absence of a well-differentiated neuroendocrine tumor in the entirely submitted concomitant pancreatic resection specimen and (2) the presence of an admixture of insulin and glucagon expressing cells by immunohistochemistry in a distribution characteristic of non-neoplastic pancreatic islets. Both cases were incidental microscopic findings in pancreatic resections for intraductal papillary mucinous neoplasms that were previously biopsied and showed associated microscopic areas of fibrosis and chronic pancreatitis and thus this phenomenon may be related to mechanical displacement from prior injury and/or biopsy.


Sujet(s)
Ilots pancréatiques , Noeuds lymphatiques , Tumeurs du pancréas , Humains , Noeuds lymphatiques/anatomopathologie , Ilots pancréatiques/anatomopathologie , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/chirurgie , Tumeurs du pancréas/composition chimique , Mâle , Adulte d'âge moyen , Femelle , Sujet âgé , Métastase lymphatique , Immunohistochimie , Diagnostic différentiel , Résultats fortuits , Erreurs de diagnostic , Marqueurs biologiques tumoraux/analyse , Valeur prédictive des tests , Tumeurs neuroendocrines/anatomopathologie , Tumeurs neuroendocrines/chirurgie
12.
BMC Nephrol ; 25(1): 156, 2024 May 09.
Article de Anglais | MEDLINE | ID: mdl-38724923

RÉSUMÉ

BACKGROUND: Islet transplantation is an effective treatment for diabetes or even its complications. Aim of this study is to investigate efficacy of biomaterial treated islet transplantation on treating diabetic nephropathy. METHODS: Male rats were randomly divided into 6 groups; Control, diabetic control, diabetic transplanted with untreated islets, with platelet rich plasma treated islets, with pancreatic islets homogenate treated islets, or with these biomaterials combination treated islets. Islets cultured with biomaterials and transplanted to diabetic rats. After 60 days, biochemical, oxidative stress, and stereological parameters were assessed. RESULTS: Serum albumin and BUN concentration, decreased and increased respectively, Oxidative stress of kidney impaired, kidney weight, volume of kidney, cortex, medulla, glomerulus, proximal and distal tubules, collecting ducts, vessels, inflammatory, necrotic and fibrotic tissue in diabetic group increased compared to control group (p < 0.001). In treated groups, especially pancreatic islets homogenate treated islets transplanting animals, there was significant changes in kidney weight, and volume of kidney, proximal and distal tubules, Henle's loop and collecting ducts compared with diabetic group (p = 0.013 to p < 0.001). Combination treated islets animals showed significant increase in vessel volume compared to diabetic group (p < 0.001). Necrotic and fibrotic tissue significantly decreased in islets treated than untreated islet animals, it was higher in pancreatic islets homogenate, and combination treated islets groups (p = 0.001). CONCLUSIONS: Biomaterials treated islets transplanting could improve diabetic nephropathy. Improvement of oxidative stress followed by controlling glucose level, and effects of growth factors presenting in biomaterials can be considered as capable underlying mechanism of ameliorating inflammatory, necrotic and fibrotic tissue volume.


Sujet(s)
Matériaux biocompatibles , Diabète expérimental , Néphropathies diabétiques , Transplantation d'ilots de Langerhans , Animaux , Mâle , Rats , Néphropathies diabétiques/anatomopathologie , Transplantation d'ilots de Langerhans/méthodes , Matériaux biocompatibles/usage thérapeutique , Ilots pancréatiques/anatomopathologie , Stress oxydatif , Rat Sprague-Dawley , Résultat thérapeutique
13.
Transplantation ; 108(8): e156-e169, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38578708

RÉSUMÉ

BACKGROUND: Islet transplantation is a promising therapy for patients with type 1 diabetes. However, ischemic injury to the donor islets during cold preservation leads to reduced islet quality and compromises transplant outcome. Several studies imply that liraglutide, a glucagon-like peptide-1 receptor agonist, has a positive effect on promoting islet survival, but its impact on islet cold-ischemic injury remains unexplored. Therefore, the aim of this study was to investigate whether liraglutide can improve islet transplantation efficacy by inhibiting cold-ischemic injury and to explore the underlying mechanisms. METHODS: Liraglutide was applied in a mouse pancreas preservation model and a human islets cold-preservation model, and islet viability, function, oxidative stress levels were evaluated. Furthermore, islet transplantation was performed in a syngeneic mouse model and a human-to-nude mouse islet xenotransplantation model. RESULTS: The supplementation of liraglutide in preservation solution improved islet viability, function, and reduced cell apoptosis. Liraglutide inhibited the oxidative stress of cold-preserved pancreas or islets through upregulating the antioxidant enzyme glutathione levels, inhibiting reactive oxygen species accumulation, and maintaining the mitochondrial membrane integrity, which is associated with the activation of Akt signaling. Furthermore, the addition of liraglutide during cold preservation of donor pancreas or donor islets significantly improved the subsequent transplant outcomes in both syngeneic mouse islet transplantation model and human-to-nude mouse islet xenotransplantation model. CONCLUSIONS: Liraglutide protects islets from cold ischemia-related oxidative stress during preservation and hence improved islet transplantation outcomes, and this protective effect of liraglutide in islets is associated with the activation of Akt signaling.


Sujet(s)
Transplantation d'ilots de Langerhans , Ilots pancréatiques , Liraglutide , Stress oxydatif , Protéines proto-oncogènes c-akt , Transduction du signal , Liraglutide/pharmacologie , Animaux , Stress oxydatif/effets des médicaments et des substances chimiques , Transplantation d'ilots de Langerhans/méthodes , Humains , Protéines proto-oncogènes c-akt/métabolisme , Ilots pancréatiques/effets des médicaments et des substances chimiques , Ilots pancréatiques/métabolisme , Ilots pancréatiques/anatomopathologie , Transduction du signal/effets des médicaments et des substances chimiques , Ischémie froide/effets indésirables , Mâle , Souris , Souris nude , Survie du greffon/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Apoptose/effets des médicaments et des substances chimiques , Transplantation hétérologue , Cryoconservation , Conservation d'organe/méthodes , Lésion d'ischémie-reperfusion/prévention et contrôle , Lésion d'ischémie-reperfusion/anatomopathologie , Lésion d'ischémie-reperfusion/métabolisme
14.
Signal Transduct Target Ther ; 9(1): 104, 2024 Apr 23.
Article de Anglais | MEDLINE | ID: mdl-38654010

RÉSUMÉ

The angiotensin-converting enzyme 2 (ACE2) is a primary cell surface viral binding receptor for SARS-CoV-2, so finding new regulatory molecules to modulate ACE2 expression levels is a promising strategy against COVID-19. In the current study, we utilized islet organoids derived from human embryonic stem cells (hESCs), animal models and COVID-19 patients to discover that fibroblast growth factor 7 (FGF7) enhances ACE2 expression within the islets, facilitating SARS-CoV-2 infection and resulting in impaired insulin secretion. Using hESC-derived islet organoids, we demonstrated that FGF7 interacts with FGF receptor 2 (FGFR2) and FGFR1 to upregulate ACE2 expression predominantly in ß cells. This upregulation increases both insulin secretion and susceptibility of ß cells to SARS-CoV-2 infection. Inhibiting FGFR counteracts the FGF7-induced ACE2 upregulation, subsequently reducing viral infection and replication in the islets. Furthermore, retrospective clinical data revealed that diabetic patients with severe COVID-19 symptoms exhibited elevated serum FGF7 levels compared to those with mild symptoms. Finally, animal experiments indicated that SARS-CoV-2 infection increased pancreatic FGF7 levels, resulting in a reduction of insulin concentrations in situ. Taken together, our research offers a potential regulatory strategy for ACE2 by controlling FGF7, thereby protecting islets from SARS-CoV-2 infection and preventing the progression of diabetes in the context of COVID-19.


Sujet(s)
Angiotensin-converting enzyme 2 , COVID-19 , Facteur de croissance fibroblastique de type 7 , Ilots pancréatiques , Organoïdes , Animaux , Humains , Mâle , Souris , Angiotensin-converting enzyme 2/génétique , Angiotensin-converting enzyme 2/métabolisme , COVID-19/génétique , COVID-19/métabolisme , COVID-19/virologie , COVID-19/anatomopathologie , Facteur de croissance fibroblastique de type 7/génétique , Facteur de croissance fibroblastique de type 7/métabolisme , Cellules souches embryonnaires humaines/métabolisme , Sécrétion d'insuline/génétique , Ilots pancréatiques/métabolisme , Ilots pancréatiques/virologie , Ilots pancréatiques/anatomopathologie , Organoïdes/virologie , Organoïdes/métabolisme , Organoïdes/anatomopathologie , SARS-CoV-2/génétique
15.
Sci Rep ; 14(1): 9100, 2024 04 20.
Article de Anglais | MEDLINE | ID: mdl-38643275

RÉSUMÉ

Diabetes constitutes a major public health problem, with dramatic consequences for patients. Both genetic and environmental factors were shown to contribute to the different forms of the disease. The monogenic forms, found both in humans and in animal models, specially help to decipher the role of key genes in the physiopathology of the disease. Here, we describe the phenotype of early diabetes in a colony of NOD mice, with spontaneous invalidation of Akt2, that we called HYP. The HYP mice were characterised by a strong and chronic hyperglycaemia, beginning around the age of one month, especially in male mice. The phenotype was not the consequence of the acceleration of the autoimmune response, inherent to the NOD background. Interestingly, in HYP mice, we observed hyperinsulinemia before hyperglycaemia occurred. We did not find any difference in the pancreas' architecture of the NOD and HYP mice (islets' size and staining for insulin and glucagon) but we detected a lower insulin content in the pancreas of HYP mice compared to NOD mice. These results give new insights about the role played by Akt2 in glucose homeostasis and argue for the ß cell failure being the primary event in the course of diabetes.


Sujet(s)
Diabète de type 1 , Hyperglycémie , Ilots pancréatiques , Animaux , Humains , Mâle , Souris , Diabète de type 1/génétique , Hyperglycémie/génétique , Insuline , Ilots pancréatiques/anatomopathologie , Souris de lignée NOD , Pancréas/anatomopathologie , Protéines proto-oncogènes c-akt/génétique
16.
Acta Diabetol ; 61(7): 897-907, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38530415

RÉSUMÉ

OBJECTIVE: To examine the association between islet autoantibodies (IAbs) and the retinal neurovascular changes in type 1 diabetes mellitus (T1DM) with no diabetic retinopathy (NDR). METHODS: This cross-sectional study measured the neural retinal structure and microvascular density of 118 NDR eyes using spectral-domain optical coherence tomography angiography. Retinal structure parameters included retinal thickness (RT), inner retinal thickness (iRT), retina never fibral layer thickness (RNFL thickness), ganglion cell complex thickness (GCC thickness), and loss volume of GCC. Microvascular parameters included vessel density of superficial capillary plexus (sVD), vessel density of deep capillary plexus, and vessel density of choroid capillary plexus. Comparison and correlation analyses of these OCTA parameters were made with various IAbs, including glutamic acid decarboxylase antibody (GADA), tyrosine phosphatase-related islet antigen 2 antibody (IA2A), and zinc transporter 8 antibody (ZnT8A). A general linear model was used to understand the association of IAbs with the retina parameters. RESULTS: The IAb positive (IAbs +) group, which included 85 patients, had thinner RT (235.20 ± 18.10 mm vs. 244.40 ± 19.90 mm at fovea, P = 0.021) and thinner iRT (120.10 ± 9.00 mm vs. 124.70 ± 6.90 mm at parafovea, P = 0.015), compared with the IAb negative (IAbs-) group comprising 33 patients. Furthermore, a more severe reduction of RT was demonstrated in the presence of multiple IAbs. Among the three IAbs, GADA was the most significant independent risk factor of all-round RT decrease (ß = -0.20 vs. -0.27 at fovea and parafovea, respectively, P < 0.05), while titers of IA2A negatively affect sVD in the parafovea (ß = -0.316, P = 0.003). CONCLUSIONS: IAbs are associated with neural retinal thinning and microcirculation reduction in T1DM patients before the clinical onset of diabetic retinopathy.


Sujet(s)
Autoanticorps , Diabète de type 1 , Rétinopathie diabétique , Microcirculation , Rétine , Humains , Autoanticorps/sang , Autoanticorps/immunologie , Diabète de type 1/immunologie , Diabète de type 1/anatomopathologie , Diabète de type 1/imagerie diagnostique , Mâle , Femelle , Études transversales , Adulte , Rétinopathie diabétique/immunologie , Rétinopathie diabétique/anatomopathologie , Rétinopathie diabétique/imagerie diagnostique , Rétine/imagerie diagnostique , Rétine/immunologie , Rétine/anatomopathologie , Adulte d'âge moyen , Tomographie par cohérence optique , Ilots pancréatiques/immunologie , Ilots pancréatiques/imagerie diagnostique , Ilots pancréatiques/anatomopathologie , Ilots pancréatiques/vascularisation , Vaisseaux rétiniens/imagerie diagnostique , Vaisseaux rétiniens/anatomopathologie , Jeune adulte
17.
Mol Cell Endocrinol ; 588: 112202, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38552943

RÉSUMÉ

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


Sujet(s)
Composés benzhydryliques , Ilots pancréatiques , Phénols , Effets différés de l'exposition prénatale à des facteurs de risque , Animaux , Composés benzhydryliques/toxicité , Femelle , Phénols/toxicité , Grossesse , Ovis , Effets différés de l'exposition prénatale à des facteurs de risque/induit chimiquement , Effets différés de l'exposition prénatale à des facteurs de risque/anatomopathologie , Ilots pancréatiques/effets des médicaments et des substances chimiques , Ilots pancréatiques/métabolisme , Ilots pancréatiques/anatomopathologie , Perturbateurs endocriniens/toxicité , Cellules à insuline/effets des médicaments et des substances chimiques , Cellules à insuline/métabolisme , Cellules à insuline/anatomopathologie , Exposition maternelle/effets indésirables , Insuline/métabolisme , Foetus/effets des médicaments et des substances chimiques , Cellules à glucagon/effets des médicaments et des substances chimiques , Cellules à glucagon/métabolisme , Cellules à glucagon/anatomopathologie
18.
Histochem Cell Biol ; 161(5): 391-408, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38368592

RÉSUMÉ

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


Sujet(s)
Cisplatine , Protéines et peptides de signalisation intercellulaire , Animaux , Cisplatine/pharmacologie , Rats , Protéines et peptides de signalisation intercellulaire/métabolisme , Protéines et peptides de signalisation intercellulaire/pharmacologie , Mâle , Apoptose/effets des médicaments et des substances chimiques , Ilots pancréatiques/métabolisme , Ilots pancréatiques/effets des médicaments et des substances chimiques , Ilots pancréatiques/anatomopathologie , Rat Wistar
19.
Reprod Biomed Online ; 48(4): 103727, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38402677

RÉSUMÉ

RESEARCH QUESTION: Does type 1 diabetes mellitus (T1DM) affect reproductive health of female patients? What is the potential mechanism of reproductive dysfunction in female patients caused by T1DM? DESIGN: Preliminary assessment of serum levels of female hormones in women with or without T1DM. Then histological and immunological examinations were carried out on the pancreas, ovaries and uteri at different stages in non-obese diabetic (NOD) and Institute of Cancer Research (ICR) mice, as well as assessment of their fertility. A protein array was carried out to detect the changes in serum inflammatory cytokines. Furthermore, RNA-sequencing was used to identify the key abnormal genes/pathways in ovarian and uterine tissues of female NOD mice, which were further verified at the protein level. RESULTS: Testosterone levels were significantly increased (P = 0.0036) in female mice with T1DM. Increasing age in female NOD mice was accompanied by obvious lymphocyte infiltration in the pancreatic islets. Moreover, the levels of serum inflammatory factors in NOD mice were sharply increased with increasing age. The fertility of female NOD mice declined markedly, and most were capable of conceiving only once. Furthermore, ovarian and uterine morphology and function were severely impaired in NOD female mice. Additionally, ovarian and uterine tissues revealed that the differentially expressed genes were primarily enriched in metabolism, cytokine-receptor interactions and chemokine signalling pathways. CONCLUSION: T1DM exerts a substantial impairment on female reproductive health, leading to diminished fertility, potentially associated with immune disorders and alterations in energy metabolism.


Sujet(s)
Diabète de type 1 , Ilots pancréatiques , Humains , Femelle , Animaux , Souris , Diabète de type 1/génétique , Diabète de type 1/métabolisme , Diabète de type 1/anatomopathologie , Souris de lignée NOD , Pancréas/métabolisme , Pancréas/anatomopathologie , Ilots pancréatiques/métabolisme , Ilots pancréatiques/anatomopathologie , Cytokines/métabolisme , Inflammation/métabolisme
20.
Protoplasma ; 261(2): 281-291, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-37798610

RÉSUMÉ

Insulinoma is an excessive insulin-released beta cell tumor. Pancreas cancer is one of the deadliest malignant neoplasms. Exosomes are secreted cell membrane vesicles containing a large number of proteins, lipids, and nucleic acids. The aim of this study is to investigate the effects of exosomes on two cell lines of benign and malignant character. For the first time, exosomes were isolated from pancreatic island-derived progenitor cells (PID-PCs) and applied to INS-1 and MiaPaCa-2 cells. In addition, exosomes isolated from PID-PC, MiaPaca-2, and INS-1 cells were characterized in order to compare their sizes with other previously isolated exosomes. Alix, TSG101, CD9, and CD81 were analyzed. The size and concentration of exosomes and the cell viability were detected. The cells were marked with HSP90, HSF-1, Kaspaz-8, Active-Kaspaz-3, Beclin, and p-Bcl-2. The cell cytotoxicity and insulin levels kit were measured. Alix in all exosomes, and PID-PC, MiaPaca-2 cell lysates; TSG101 in PID-PC and MiaPaca-2 cell lysates; CD9 in INS-1 exosomes were detected. The dimensions of isolated exosomes were 103.6 ± 28.6 nm, 100.7 ± 10 nm, and 147.2 ± 12.3 nm for PID-PCs, MiaPaca-2, and INS-1 cells. The cell viability decreased and HSP90 increased in the MiaPaca-2 cells. The HSF-1 was higher in the control MiaPaca-2 cell compared to the control INS-1 cell, and the exosome-treated MiaPaca-2 cell compared to the exosome-treated INS-1 cell. Beclin and p-Bcl-2 were decreased in the exosome-treated MiaPaca-2 cells. The insulin level in the cell lysates increased compared to cell secretion in INS-1 cells. In conclusion, exosomes isolated from the PID-PC caused cell death in the MiaPaca-2 cells in a time- and dose-dependent manner. The IC50 value determined for MiaPaca-2 cells has no effect on cell viability in INS-1 cells, which best mimics pancreatic beta cells and can be used instead of healthy pancreatic beta cells. Isolated exosomes can kill cancer cells without damaging healthy cells.


Sujet(s)
Exosomes , Insulines , Ilots pancréatiques , Tumeurs du pancréas , Humains , Exosomes/métabolisme , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/anatomopathologie , Cellules souches/métabolisme , Cellules souches/anatomopathologie , Protéines proto-oncogènes c-bcl-2/métabolisme , Ilots pancréatiques/métabolisme , Ilots pancréatiques/anatomopathologie , Insulines/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE