Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 8.974
1.
PLoS One ; 19(5): e0287877, 2024.
Article En | MEDLINE | ID: mdl-38787820

Type 1 diabetes (T1D) is characterized by HLA class I-mediated presentation of autoantigens on the surface of pancreatic ß-cells. Recognition of these autoantigens by CD8+ T cells results in the destruction of pancreatic ß-cells and, consequently, insulin deficiency. Most epitopes presented at the surface of ß-cells derive from the insulin precursor molecule proinsulin. The intracellular processing pathway(s) involved in the generation of these peptides are poorly defined. In this study, we show that a proinsulin B-chain antigen (PPIB5-14) originates from proinsulin molecules that are processed by ER-associated protein degradation (ERAD) and thus originate from ER-resident proteins. Furthermore, screening genes encoding for E2 ubiquitin conjugating enzymes, we identified UBE2G2 to be involved in proinsulin degradation and subsequent presentation of the PPIB10-18 autoantigen. These insights into the pathway involved in the generation of insulin-derived peptides emphasize the importance of proinsulin processing in the ER to T1D pathogenesis and identify novel targets for future T1D therapies.


Autoantigens , Endoplasmic Reticulum-Associated Degradation , Proinsulin , Proteolysis , Ubiquitin-Conjugating Enzymes , Proinsulin/metabolism , Proinsulin/immunology , Proinsulin/genetics , Autoantigens/metabolism , Autoantigens/immunology , Humans , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Conjugating Enzymes/genetics , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Antigen Presentation/immunology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/immunology
2.
Int J Mol Sci ; 25(10)2024 May 11.
Article En | MEDLINE | ID: mdl-38791299

Type 1 diabetes (T1D) affects gastrointestinal (GI) motility, favoring gastroparesis, constipation, and fecal incontinence, which are more prevalent in women. The mechanisms are unknown. Given the G-protein-coupled estrogen receptor's (GPER) role in GI motility, we investigated sex-related diabetes-induced epigenetic changes in GPER. We assessed GPER mRNA and protein expression levels using qPCR and Western blot analyses, and quantified the changes in nuclear DNA methyltransferases and histone modifications (H3K4me3, H3Ac, and H3K27Ac) by ELISA kits. Targeted bisulfite and chromatin immunoprecipitation assays were used to evaluate DNA methylation and histone modifications around the GPER promoter by chromatin immunoprecipitation assays in gastric and colonic smooth muscle tissues of male and female control (CTR) and non-obese diabetic (NOD) mice. GPER expression was downregulated in NOD, with sex-dependent variations. In the gastric smooth muscle, not in colonic smooth muscle, downregulation coincided with differences in methylation ratios between regions 1 and 2 of the GPER promoter of NOD. DNA methylation was higher in NOD male colonic smooth muscle than in NOD females. H3K4me3 and H3ac enrichment decreased in NOD gastric smooth muscle. H3K4me3 levels diminished in the colonic smooth muscle of NOD. H3K27ac levels were unaffected, but enrichment decreased in NOD male gastric smooth muscle; however, it increased in the NOD male colonic smooth muscle and decreased in the female NOD colonic smooth muscle. Male NOD colonic smooth muscle exhibited decreased H3K27ac levels, not female, whereas female NOD colonic smooth muscle demonstrated diminished enrichment of H3ac at the GPER promoter, contrary to male NOD. Sex-specific epigenetic mechanisms contribute to T1D-mediated suppression of GPER expression in the GI tract. These insights advance our understanding of T1D complications and suggest promising avenues for targeted therapeutic interventions.


Colon , DNA Methylation , Epigenesis, Genetic , Histones , Mice, Inbred NOD , Muscle, Smooth , Promoter Regions, Genetic , Receptors, G-Protein-Coupled , Animals , Female , Male , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Muscle, Smooth/metabolism , Mice , Histones/metabolism , Colon/metabolism , Colon/pathology , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/genetics , Receptors, Estrogen/metabolism , Receptors, Estrogen/genetics , Stomach/pathology
3.
Nat Commun ; 15(1): 3744, 2024 May 03.
Article En | MEDLINE | ID: mdl-38702321

Cellular composition and anatomical organization influence normal and aberrant organ functions. Emerging spatial single-cell proteomic assays such as Image Mass Cytometry (IMC) and Co-Detection by Indexing (CODEX) have facilitated the study of cellular composition and organization by enabling high-throughput measurement of cells and their localization directly in intact tissues. However, annotation of cell types and quantification of their relative localization in tissues remain challenging. To address these unmet needs for atlas-scale datasets like Human Pancreas Analysis Program (HPAP), we develop AnnoSpat (Annotator and Spatial Pattern Finder) that uses neural network and point process algorithms to automatically identify cell types and quantify cell-cell proximity relationships. Our study of data from IMC and CODEX shows the higher performance of AnnoSpat in rapid and accurate annotation of cell types compared to alternative approaches. Moreover, the application of AnnoSpat to type 1 diabetic, non-diabetic autoantibody-positive, and non-diabetic organ donor cohorts recapitulates known islet pathobiology and shows differential dynamics of pancreatic polypeptide (PP) cell abundance and CD8+ T cells infiltration in islets during type 1 diabetes progression.


Algorithms , Diabetes Mellitus, Type 1 , Pancreas , Proteomics , Humans , Proteomics/methods , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/metabolism , Pancreas/cytology , Pancreas/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans/cytology , Single-Cell Analysis/methods , Neural Networks, Computer , CD8-Positive T-Lymphocytes/metabolism , Image Cytometry/methods
4.
BMC Endocr Disord ; 24(1): 63, 2024 May 09.
Article En | MEDLINE | ID: mdl-38724988

BACKGROUND: A chronic autoimmune disease with an increasing incidence rate, type 1 diabetes mellitus (T1DM) is typified by the degeneration of the pancreatic beta cells. Diabetes management is significantly impacted by nutrition. Although it has been demonstrated that following the Mediterranean diet (MD) improves metabolic control with type 2 diabetes in children and adults, its effects on children with T1DM have not received much attention. OBJECTIVE: Therefore, the purpose of this study was to assess whether adherence to Mediterranean diet is associated with better metabolic control and body composition in youths with Type 1 Diabetes Mellitus. The study recruited T1DM patients aged 6-18 years at Istanbul University Cerrahpasa Medical Faculty Hospital's Pediatric Endocrinology and Diabetes Outpatient Clinic for follow-up. METHODS: In addition to demographic variables, some anthropometric measurements, body composition and biochemical parameters such as: Trygliceride(TG), Total cholesterol (TC), High density lipoprotein cholesterol (HDL-C), Low density lipoprotein cholesterol (LDL-C), (Aspartate aminotransferase) AST, Alanine transaminase (ALT) and glycated hemoglobin (HbA1c) was analyzed. The time in range (TIR) is a value obtained from continuous glucose monitoring. KIDMED was used to assess the participants' adherence with the MD. RESULTS: Good adherence to the MD resulted in much larger height SDS than poor adherence. Poor adherence to MD resulted in higher body fat than moderate and good adherence. There is positivite correlation between TIR and KIDMED score. Adherence to MD is negatively associated with HbA1c. The regression anaylsis showed that a one-point rise in the KIDMED score would result in a 0.314-unit reduction in the HbA1c value (p < 0.01). CONCLUSIONS: In conclusion, this study found that adhering to MD led to improved anthropometric measurements, biochemistry, and diabetes outcomes. Awareness among children, adolescents with T1DM, and their parents about the benefits of MD compliance for glycemic and metabolic control should be raised.


Body Composition , Diabetes Mellitus, Type 1 , Diet, Mediterranean , Humans , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/diet therapy , Adolescent , Male , Female , Child , Follow-Up Studies , Blood Glucose/metabolism , Blood Glucose/analysis , Glycated Hemoglobin/analysis , Glycated Hemoglobin/metabolism , Patient Compliance
5.
Sci Rep ; 14(1): 11911, 2024 05 24.
Article En | MEDLINE | ID: mdl-38789719

Lack of understanding of the pathophysiology of gastrointestinal (GI) complications in type 1 diabetes (T1D), including altered intestinal transcriptomes and protein expression represents a major gap in the management of these patients. Human enteroids have emerged as a physiologically relevant model of the intestinal epithelium but establishing enteroids from individuals with long-standing T1D has proven difficult. We successfully established duodenal enteroids using endoscopic biopsies from pediatric T1D patients and compared them with aged-matched enteroids from healthy subjects (HS) using bulk RNA sequencing (RNA-seq), and functional analyses of ion transport processes. RNA-seq analysis showed significant differences in genes and pathways associated with cell differentiation and proliferation, cell fate commitment, and brush border membrane. Further validation of these results showed higher expression of enteroendocrine cells, and the proliferating cell marker Ki-67, significantly lower expression of NHE3, lower epithelial barrier integrity, and higher fluid secretion in response to cAMP and elevated calcium in T1D enteroids. Enteroids established from pediatric T1D duodenum identify characteristics of an abnormal intestinal epithelium and are distinct from HS. Our data supports the use of pediatric enteroids as an ex-vivo model to advance studies of GI complications and drug discovery in T1D patients.


Diabetes Mellitus, Type 1 , Duodenum , Intestinal Mucosa , Humans , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/genetics , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Child , Duodenum/metabolism , Duodenum/pathology , Female , Male , Cell Proliferation , Adolescent , Enteroendocrine Cells/metabolism , Enteroendocrine Cells/pathology , Sodium-Hydrogen Exchanger 3/metabolism , Sodium-Hydrogen Exchanger 3/genetics , Cell Differentiation , Organoids/metabolism , Organoids/pathology , Ki-67 Antigen/metabolism
6.
Sci Adv ; 10(20): eadn2136, 2024 May 17.
Article En | MEDLINE | ID: mdl-38758799

Monocytes are immune regulators implicated in the pathogenesis of type 1 diabetes (T1D), an autoimmune disease that targets insulin-producing pancreatic ß cells. We determined that monocytes of recent onset (RO) T1D patients and their healthy siblings express proinflammatory/cytolytic transcriptomes and hypersecrete cytokines in response to lipopolysaccharide exposure compared to unrelated healthy controls (uHCs). Flow cytometry measured elevated circulating abundances of intermediate monocytes and >2-fold more CD14+CD16+HLADR+KLRD1+PRF1+ NK-like monocytes among patients with ROT1D compared to uHC. The intermediate to nonclassical monocyte ratio among ROT1D patients correlated with the decline in functional ß cell mass during the first 24 months after onset. Among sibling nonprogressors, temporal decreases were measured in the intermediate to nonclassical monocyte ratio and NK-like monocyte abundances; these changes coincided with increases in activated regulatory T cells. In contrast, these monocyte populations exhibited stability among T1D progressors. This study associates heightened monocyte proinflammatory/cytolytic activity with T1D susceptibility and progression and offers insight to the age-dependent decline in T1D susceptibility.


Diabetes Mellitus, Type 1 , Disease Progression , Monocytes , Humans , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/genetics , Monocytes/metabolism , Monocytes/immunology , Male , Female , Adolescent , Child , Adult , Cytokines/metabolism , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Young Adult , Case-Control Studies
7.
Cell Commun Signal ; 22(1): 252, 2024 May 02.
Article En | MEDLINE | ID: mdl-38698453

BACKGROUND: Ischemic postconditioning (IPostC) has been reported as a promising method for protecting against myocardial ischemia-reperfusion (MI/R) injury. Our previous study found that the infarct-limiting effect of IPostC is abolished in the heart of diabetes whose cardiac expression of DJ-1 (also called PARK7, Parkinsonism associated deglycase) is reduced. However, the role and in particular the underlying mechanism of DJ-1 in the loss of sensitivity to IPostC-induced cardioprotection in diabetic hearts remains unclear. METHODS: Streptozotocin-induced type 1 diabetic rats were subjected to MI/R injury by occluding the left anterior descending artery (LAD) and followed by reperfusion. IPostC was induced by three cycles of 10s of reperfusion and ischemia at the onset of reperfusion. AAV9-CMV-DJ-1, AAV9-CMV-C106S-DJ-1 or AAV9-DJ-1 siRNA were injected via tail vein to either over-express or knock-down DJ-1 three weeks before inducing MI/R. RESULTS: Diabetic rats subjected to MI/R exhibited larger infarct area, more severe oxidative injury concomitant with significantly reduced cardiac DJ-1 expression and increased PTEN expression as compared to non-diabetic rats. AAV9-mediated cardiac DJ-1 overexpression, but not the cardiac overexpression of DJ-1 mutant C106S, restored IPostC-induced cardioprotection and this effect was accompanied by increased cytoplasmic DJ-1 translocation toward nuclear and mitochondrial, reduced PTEN expression, and increased Nrf-2/HO-1 transcription. Our further study showed that AAV9-mediated targeted DJ-1 gene knockdown aggravated MI/R injury in diabetic hearts, and this exacerbation of MI/R injury was partially reversed by IPostC in the presence of PTEN inhibition or Nrf-2 activation. CONCLUSIONS: These findings suggest that DJ-1 preserves the cardioprotective effect of IPostC against MI/R injury in diabetic rats through nuclear and mitochondrial DJ-1 translocation and that inhibition of cardiac PTEN and activation of Nrf-2/HO-1 may represent the major downstream mechanisms whereby DJ-1 preserves the cardioprotective effect of IPostC in diabetes.


Diabetes Mellitus, Experimental , Ischemic Postconditioning , Myocardial Reperfusion Injury , PTEN Phosphohydrolase , Protein Deglycase DJ-1 , Rats, Sprague-Dawley , Animals , Protein Deglycase DJ-1/metabolism , Protein Deglycase DJ-1/genetics , PTEN Phosphohydrolase/metabolism , PTEN Phosphohydrolase/genetics , Diabetes Mellitus, Experimental/metabolism , Male , Rats , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/genetics , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/complications , Protein Transport , Streptozocin , Myocardial Infarction/metabolism , Myocardial Infarction/pathology
8.
J Diabetes Res ; 2024: 5574968, 2024.
Article En | MEDLINE | ID: mdl-38800586

Islet transplantation (ITx) is an established and safe alternative to pancreas transplantation for type 1 diabetes mellitus (T1DM) patients. However, most ITx recipients lose insulin independence by 3 years after ITx due to early graft loss, such that multiple donors are required to achieve insulin independence. In the present study, we investigated whether skeletal myoblast cells could be beneficial for promoting angiogenesis and maintaining the differentiated phenotypes of islets. In vitro experiments showed that the myoblast cells secreted angiogenesis-related cytokines (vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF), and stromal-derived factor-1α (SDF-1α)), contributed to maintenance of differentiated islet phenotypes, and enhanced islet cell insulin secretion capacity. To verify these findings in vivo, we transplanted islets alone or with myoblast cells under the kidney capsule of streptozotocin-induced diabetic mice. Compared with islets alone, the group bearing islets with myoblast cells had a significantly lower average blood glucose level. Histological examination revealed that transplants with islets plus myoblast cells were associated with a significantly larger insulin-positive area and significantly higher number of CD31-positive microvessels compared to islets alone. Furthermore, islets cotransplanted with myoblast cells showed JAK-STAT signaling activation. Our results suggest two possible mechanisms underlying enhancement of islet graft function with myoblast cells cotransplantation: "indirect effects" mediated by angiogenesis and "direct effects" of myoblast cells on islets via the JAK-STAT cascade. Overall, these findings suggest that skeletal myoblast cells enhance the function of transplanted islets, implying clinical potential for a novel ITx procedure involving myoblast cells for patients with diabetes.


Diabetes Mellitus, Experimental , Insulin , Islets of Langerhans Transplantation , Myoblasts, Skeletal , Neovascularization, Physiologic , Animals , Islets of Langerhans Transplantation/methods , Diabetes Mellitus, Experimental/metabolism , Myoblasts, Skeletal/transplantation , Myoblasts, Skeletal/metabolism , Mice , Male , Insulin/metabolism , Hepatocyte Growth Factor/metabolism , Mice, Inbred C57BL , Vascular Endothelial Growth Factor A/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans/blood supply , Chemokine CXCL12/metabolism , Blood Glucose/metabolism , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/physiopathology , Diabetes Mellitus, Type 1/surgery , Signal Transduction , Insulin Secretion , Cell Differentiation
9.
Sci Rep ; 14(1): 12402, 2024 05 30.
Article En | MEDLINE | ID: mdl-38811610

Evaluating the quality of isolated human islets before transplantation is crucial for predicting the success in treating Type 1 diabetes. The current gold standard involves time-intensive in vivo transplantation into diabetic immunodeficient mice. Given the susceptibility of isolated islets to hypoxia, we hypothesized that hypoxia present in islets before transplantation could indicate compromised islet quality, potentially leading to unfavorable outcomes. To test this hypothesis, we analyzed expression of 39 hypoxia-related genes in human islets from 85 deceased donors. We correlated gene expression profiles with transplantation outcomes in 327 diabetic mice, each receiving 1200 islet equivalents grafted into the kidney capsule. Transplantation outcome was post-transplant glycemic control based on area under the curve of blood glucose over 4 weeks. In linear regression analysis, DDIT4 (R = 0.4971, P < 0.0001), SLC2A8 (R = 0.3531, P = 0.0009) and HK1 (R = 0.3444, P = 0.0012) had the highest correlation with transplantation outcome. A multiple regression model of 11 genes increased the correlation (R = 0.6117, P < 0.0001). We conclude that assessing pre-transplant hypoxia in human islets via gene expression analysis is a rapid, viable alternative to conventional in vivo assessments. This approach also underscores the importance of mitigating pre-transplant hypoxia in isolated islets to improve the success rate of islet transplantation.


Diabetes Mellitus, Experimental , Islets of Langerhans Transplantation , Islets of Langerhans , Humans , Animals , Islets of Langerhans Transplantation/methods , Mice , Islets of Langerhans/metabolism , Diabetes Mellitus, Experimental/therapy , Male , Diabetes Mellitus, Type 1/metabolism , Hypoxia/metabolism , Female , Cell Hypoxia , Middle Aged , Blood Glucose/metabolism
10.
Biomed Pharmacother ; 175: 116731, 2024 Jun.
Article En | MEDLINE | ID: mdl-38761421

Nutraceuticals have gained increasing interest, prompting the need to investigate plant extracts for their beneficial properties and potential side effects. This study aimed to assess the nutraceutical effects of environmentally clean extracts from Rosmarinus officinalis and Gongolaria abies-marina (formerly Cystoseira abies-marina (Phaeophyceae)) on the metabolic profile of streptozotocin-induced diabetic rats. We conducted untargeted LC-QTOF-MS metabolic profiling on six groups of rats: three diabetic groups receiving either a placebo, R. officinalis, or G. abies-marina extracts, and three corresponding control groups. The metabolic analysis revealed significant alterations in the levels of various glycerophospholipids, sterol lipids, and fatty acyls. Both extracts influenced the metabolic profile, partially mitigating diabetes-induced changes. Notably, G. abies-marina extract had a more pronounced impact on the animals' metabolic profiles compared to R. officinalis. In conclusion, our findings suggest that environmentally clean extracts from R. officinalis and G. abies-marina possess nutraceutical potential, as they were able to modulate the metabolic profile in streptozotocin-induced diabetic rats. G. abies-marina extract exhibited a more substantial effect on metabolic alterations induced by diabetes compared to R. officinalis. These results warrant further exploration of these plant extracts for their potential in managing diabetes-related metabolic disturbances.


Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Dietary Supplements , Plant Extracts , Rosmarinus , Animals , Plant Extracts/pharmacology , Rosmarinus/chemistry , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Male , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/metabolism , Rats , Rats, Wistar , Metabolomics , Metabolome/drug effects , Blood Glucose/drug effects , Blood Glucose/metabolism , Streptozocin , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/isolation & purification
12.
Front Endocrinol (Lausanne) ; 15: 1399256, 2024.
Article En | MEDLINE | ID: mdl-38818504

Background: It is well known that metabolic disorders, including type 1 diabetes (T1D), are often associated with reduced male fertility, mainly increasing oxidative stress and impairing the hypothalamus-pituitary-testis (HPT) axis, with consequently altered spermatogenesis and reduced sperm parameters. Herein, using a rat model of T1D obtained by treatment with streptozotocin (STZ), we analyzed several parameters of testicular activity. Methods: A total of 10 adult male Wistar rats were divided into two groups of five: control and T1D, obtained with a single intraperitoneal injection of STZ. After 3 months, the rats were anesthetized and sacrificed; one testis was stored at -80°C for biochemical analysis, and the other was fixed for histological and immunofluorescence analysis. Results: The data confirmed that T1D induced oxidative stress and, consequently, alterations in both testicular somatic and germ cells. This aspect was highlighted by enhanced apoptosis, altered steroidogenesis and Leydig cell maturity, and impaired spermatogenesis. In addition, the blood-testis barrier integrity was compromised, as shown by the reduced levels of structural proteins (N-cadherin, ZO-1, occludin, connexin 43, and VANGL2) and the phosphorylation status of regulative kinases (Src and FAK). Mechanistically, the dysregulation of the SIRT1/NRF2/MAPKs signaling pathways was proven, particularly the reduced nuclear translocation of NRF2, affecting its ability to induce the transcription of genes encoding for antioxidant enzymes. Finally, the stimulation of testicular inflammation and pyroptosis was also confirmed, as highlighted by the increased levels of some markers, such as NF-κB and NLRP3. Conclusion: The combined data allowed us to confirm that T1D has detrimental effects on rat testicular activity. Moreover, a better comprehension of the molecular mechanisms underlying the association between metabolic disorders and male fertility could help to identify novel targets to prevent and treat fertility disorders related to T1D.


Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , NF-E2-Related Factor 2 , NLR Family, Pyrin Domain-Containing 3 Protein , Oxidative Stress , Rats, Wistar , Testis , Animals , Male , Rats , NF-E2-Related Factor 2/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Testis/metabolism , Testis/pathology , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Spermatogenesis , Signal Transduction , Germ Cells/metabolism , Spermatozoa/metabolism
13.
Mol Biol Rep ; 51(1): 677, 2024 May 25.
Article En | MEDLINE | ID: mdl-38796641

BACKGROUND: One of the main causes of diabetic nephropathy is oxidative stress induced by hyperglycemia. Apelin inhibits insulin secretion. Besides, renal expression of TGF-ß is increased in diabetes mellitus (DM). The preventive effect of quercetin (Q) against renal functional disorders and tissue damage developed by DM in rats was assessed. METHODS: Forty male Wistar rats were grouped into normal control (NC), normal + quercetin (NQ: quercetin, 50 mg/kg/day by gavage), diabetic control (DC: streptozotocin, 65 mg/kg, i.p.), diabetic + quercetin pretreatment (D + Qpre), and diabetic + quercetin post-treatment (D + Qpost). All samples (24-hour urine, plasma, pancreatic, and renal tissues) were obtained at the terminal of the experiment. RESULTS: Compared to NC and NQ groups, DM ended in elevated plasma and glucose levels, decreased plasma insulin level, kidney dysfunction, augmented levels of malondialdehyde, decreased level of reduced glutathione, reduced enzymatic activities of superoxide dismutase and catalase, elevated gene expression levels of apelin and TGF-ß, also renal and pancreatic histological damages. Quercetin administration diminished entire the changes. However, the measure of improvement in the D + Qpre group was higher than that of the D + Qpost group. CONCLUSION: Quercetin prevents renal dysfunction induced by DM, which might be related to the diminution of lipid peroxidation, strengthening of antioxidant systems, and prevention of the apelin/ TGF-ß signaling pathway.


Apelin , Diabetes Mellitus, Experimental , Diabetic Nephropathies , Kidney , Oxidative Stress , Quercetin , Rats, Wistar , Transforming Growth Factor beta , Animals , Quercetin/pharmacology , Rats , Male , Transforming Growth Factor beta/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/drug therapy , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/prevention & control , Diabetic Nephropathies/drug therapy , Apelin/metabolism , Oxidative Stress/drug effects , Blood Glucose/metabolism , Blood Glucose/drug effects , Antioxidants/pharmacology , Antioxidants/metabolism , Insulin/metabolism , Insulin/blood , Diabetes Mellitus, Type 1/metabolism , Gene Expression Regulation/drug effects
14.
Front Immunol ; 15: 1386528, 2024.
Article En | MEDLINE | ID: mdl-38590527

Introduction: Inflammation of the placenta is harmful to both the fetus and the mother. Inflammation is strongly associated with diabetes, a common complication of pregnancy. Hofbauer cells (HBCs), unique immune system cells of fetal origin in the placenta, play complex roles, including growth of placental villi and their branching, stromal remodelling, and angiogenesis. Methods: Our study investigated the expression of IL-1ß, IL-10, CYP2C8, CYP2C9, CYP2J2 and sEH in HBCs from patients with type 1 diabetes mellitus (T1DM) and gestational diabetes mellitus (GDM) compared to healthy controls using immunohistochemistry. We also assessed the structure of the villus stroma using Masson´s trichrome. Results: In T1DM, HBCs showed inflammatory activation characterised by increased IL-1ß and decreased CYP epoxygenase expression compared to normal placentas. Conversely, significant inflammation in HBCs appeared less likely in GDM, as levels of IL-1ß and CYP epoxygenases remained stable compared to normal placentas. However, GDM showed a significant increase in sEH expression. Both types of diabetes showed delayed placental villous maturation and hypovascularisation, with GDM showing a more pronounced effect. Conclusion: The expression profiles of IL-1ß, CYP epoxygenases and sEH significantlly differ between controls and diabetic placentas and between T1DM and GDM. These facts suggest an association of the CYP epoxygenase-EETs-sEH axis with IL-1ß expression as well as villous stromal hypovascularisation. Given the stable high expression of IL-10 in both controls and both types of diabetes, it appears that immune tolerance is maintained in HBCs.


Diabetes Mellitus, Type 1 , Diabetes, Gestational , Pregnancy , Humans , Female , Placenta/metabolism , Interleukin-10/metabolism , Diabetes Mellitus, Type 1/metabolism , Inflammation/metabolism
15.
Cell Physiol Biochem ; 58(2): 144-155, 2024 Apr 17.
Article En | MEDLINE | ID: mdl-38639210

Diabetes mellitus type 1 (T1D) and type 2 (T2D) develop due to dysfunction of the Langerhans islet ß-cells in the pancreas, and this dysfunction is mediated by oxidative, endoplasmic reticulum (ER), and mitochondrial stresses. Although the two types of diabetes are significantly different, ß-cell failure and death play a key role in the pathogenesis of both diseases, resulting in hyperglycemia due to a reduced ability to produce insulin. In T1D, ß-cell apoptosis is the main event leading to hyperglycemia, while in T2D, insulin resistance results in an inability to meet insulin requirements. It has been suggested that autophagy promotes ß-cell survival by delaying apoptosis and providing adaptive responses to mitigate the detrimental effects of ER stress and DNA damage, which is directly related to oxidative stress. As people with diabetes are now living longer, they are more susceptible to a different set of complications. There has been a diversification in causes of death, whereby a larger proportion of deaths among individuals with diabetes is attributable to nonvascular conditions; on the other hand, the proportion of cancer-related deaths has remained stable or even increased in some countries. Due to the increasing cases of both T1D and T2D, these diseases become even more socially significant. Hence, we believe that search for any opportunities for control of this disease is an overwhelmingly important target for the modern science. We focus on two differences that are characteristic of the development of diabetes's last periods. One of them shows that all-cause death rates have declined in several diabetes populations, driven in part by large declines in vascular disease mortality but large increases in oncological diseases. Another hypothesis is that some T2D medications could be repurposed to control glycemia in patients with T1D.


Diabetes Mellitus, Type 1 , Diabetes Mellitus, Type 2 , Hyperglycemia , Insulin-Secreting Cells , Humans , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 1/metabolism , Insulin-Secreting Cells/metabolism , Cell Death , Insulin/metabolism , Hyperglycemia/metabolism , Oxidative Stress
16.
Nutr Diabetes ; 14(1): 23, 2024 Apr 23.
Article En | MEDLINE | ID: mdl-38653987

BACKGROUND: The number of patients with type 1 diabetes rises rapidly around the world in recent years. Maternal diabetes has a detrimental effect on reproductive outcomes due to decreased oocyte quality. However, the strategies to improve the oocyte quality and artificial reproductive technology (ART) efficiency of infertile females suffering from diabetes have not been fully studied. In this study, we aimed to examine the effects of nicotinamide mononucleotide (NMN) on oocyte maturation of mouse with type 1 diabetes mouse and explore the underlying mechanisms of NMN's effect. METHODS: Streptozotocin (STZ) was used to establish the mouse models with type 1 diabetes. The successful establishment of the models was confirmed by the results of body weight test, fasting blood glucose test and haematoxylin and eosin (H&E) staining. The in vitro maturation (IVM) rate of oocytes from diabetic mice was examined. Immunofluorescence staining (IF) was performed to examine the reactive oxygen species (ROS) level, spindle/chromosome structure, mitochondrial function, actin dynamics, DNA damage and histone modification of oocytes, which are potential factors affecting the oocyte quality. The quantitative reverse transcription PCR (RT-qPCR) was used to detect the mRNA levels of Sod1, Opa1, Mfn2, Drp1, Sirt1 and Sirt3 in oocytes. RESULTS: The NMN supplementation increased the oocyte maturation rate of the mice with diabetes. Furthermore, NMN supplementation improved the oocyte quality by rescuing the actin dynamics, reversing meiotic defects, improving the mitochondrial function, reducing ROS level, suppressing DNA damage and restoring changes in histone modifications of oocytes collected from the mice with diabetes. CONCLUSION: NMN could improve the maturation rate and quality of oocytes in STZ-induced diabetic mice, which provides a significant clue for the treatment of infertility of the patients with diabetes.


Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Dynamins , Nicotinamide Mononucleotide , Oocytes , Reactive Oxygen Species , Animals , Mice , Female , Oocytes/drug effects , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Experimental/drug therapy , Reactive Oxygen Species/metabolism , Nicotinamide Mononucleotide/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Sirtuin 1/metabolism , Sirtuin 3/metabolism , In Vitro Oocyte Maturation Techniques/methods , Superoxide Dismutase-1 , DNA Damage/drug effects , Streptozocin , Oogenesis/drug effects
17.
Chin J Nat Med ; 22(4): 293-306, 2024 Apr.
Article En | MEDLINE | ID: mdl-38658093

Icariin, a flavonoid glycoside, is extracted from Epimedium. This study aimed to investigate the vascular protective effects of icariin in type 1 diabetic rats by inhibiting high-mobility group box 1 (HMGB1)-related inflammation and exploring its potential mechanisms. The impact of icariin on vascular dysfunction was assessed in streptozotocin (STZ)-induced diabetic rats through vascular reactivity studies. Western blotting and immunofluorescence assays were performed to measure the expressions of target proteins. The release of HMGB1 and pro-inflammation cytokines were measured by enzyme-linked immunosorbent assay (ELISA). The results revealed that icariin administration enhanced acetylcholine-induced vasodilation in the aortas of diabetic rats. It also notably reduced the release of pro-inflammatory cytokines, including interleukin-8 (IL-8), IL-6, IL-1ß, and tumor necrosis factor-alpha (TNF-α) in diabetic rats and high glucose (HG)-induced human umbilical vein endothelial cells (HUVECs). The results also unveiled that the pro-inflammatory cytokines in the culture medium of HUVECs could be increased by rHMGB1. The increased release of HMGB1 and upregulated expressions of HMGB1-related inflammatory factors, including advanced glycation end products (RAGE), Toll-like receptor 4 (TLR4), and phosphorylated p65 (p-p65) in diabetic rats and HG-induced HUVECs, were remarkably suppressed by icariin. Notably, HMGB1 translocation from the nucleus to the cytoplasm in HUVECs under HG was inhibited by icariin. Meanwhile, icariin could activate G protein-coupled estrogen receptor (GPER) and sirt1. To explore the role of GPER and Sirt1 in the inhibitory effect of icariin on HMGB1 release and HMGB-induced inflammation, GPER inhibitor and Sirt1 inhibitor were used in this study. These inhibitors diminished the effects of icariin on HMGB1 release and HMGB1-induced inflammation. Specifically, the GPER inhibitor also negated the activation of Sirt1 by icariin. These findings suggest that icariin activates GPER and increases the expression of Sirt1, which in turn reduces HMGB1 translocation and release, thereby improving vascular endothelial function in type 1 diabetic rats by inhibiting inflammation.


Diabetes Mellitus, Experimental , Flavonoids , HMGB1 Protein , Rats, Sprague-Dawley , Receptors, Cannabinoid , Receptors, G-Protein-Coupled , Signal Transduction , Sirtuin 1 , Animals , HMGB1 Protein/metabolism , HMGB1 Protein/genetics , Sirtuin 1/metabolism , Sirtuin 1/genetics , Flavonoids/pharmacology , Signal Transduction/drug effects , Rats , Male , Humans , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Receptors, G-Protein-Coupled/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Inflammation/drug therapy , Inflammation/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/metabolism , Cytokines/metabolism , Epimedium/chemistry
18.
J Endocrinol ; 262(1)2024 Jul 01.
Article En | MEDLINE | ID: mdl-38642584

Obesity and diabetes represent two increasing and invalidating public health issues that often coexist. It is acknowledged that fat mass excess predisposes to insulin resistance and type 2 diabetes mellitus (T2D), with the increasing incidence of the two diseases significantly associated. Moreover, emerging evidence suggests that obesity might also accelerate the appearance of type 1 diabetes (T1D), which is now a relatively frequent comorbidity in patients with obesity. It is a common clinical finding that not all patients with obesity will develop diabetes at the same level of adiposity, with gender, genetic, and ethnic factors playing an important role in defining the timing of diabetes appearance. The adipose tissue (AT) expandability hypothesis explains this paradigm, indicating that the individual capacity to appropriately store energy surplus in the form of fat within the AT determines and prevents the toxic deposition of lipids in other organs, such as the pancreas. Thus, we posit that when the maximal storing capacity of AT is exceeded, individuals will develop T2D. In this review, we provide insight into mechanisms by which the AT controls pancreas lipid content and homeostasis in case of obesity to offer an adipocentric perspective of pancreatic lipotoxicity in the pathogenesis of diabetes. Moreover, we suggest that improving AT function is a valid therapeutic approach to fighting obesity-associated complications including diabetes.


Adipose Tissue , Diabetes Mellitus, Type 2 , Obesity , Pancreas , Humans , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/etiology , Obesity/metabolism , Adipose Tissue/metabolism , Adipose Tissue/pathology , Pancreas/metabolism , Pancreas/pathology , Lipid Metabolism , Diabetes Mellitus, Type 1/metabolism , Insulin Resistance/physiology , Animals
19.
Int J Mol Sci ; 25(8)2024 Apr 09.
Article En | MEDLINE | ID: mdl-38673735

Experimental animal models of diabetes can be useful for identifying novel targets related to disease, for understanding its physiopathology, and for evaluating emerging antidiabetic treatments. This study aimed to characterize two rat diabetes models: HFD + STZ, a high-fat diet (60% fat) combined with streptozotocin administration (STZ, 35 mg/kg BW), and a model with a single STZ dose (65 mg/kg BW) in comparison with healthy rats. HFD + STZ- induced animals demonstrated a stable hyperglycemia range (350-450 mg/dL), whereas in the STZ-induced rats, we found glucose concentration values with a greater dispersion, ranging from 270 to 510 mg/dL. Moreover, in the HFD + STZ group, the AUC value of the insulin tolerance test (ITT) was found to be remarkably augmented by 6.2-fold higher than in healthy animals (33,687.0 ± 1705.7 mg/dL/min vs. 5469.0 ± 267.6, respectively), indicating insulin resistance (IR). In contrast, a more moderate AUC value was observed in the STZ group (19,059.0 ± 3037.4 mg/dL/min) resulting in a value 2.5-fold higher than the average exhibited by the control group. After microarray experiments on liver tissue from all animals, we analyzed genes exhibiting a fold change value in gene expression <-2 or >2 (p-value <0.05). We found 27,686 differentially expressed genes (DEG), identified the top 10 DEGs and detected 849 coding genes that exhibited opposite expression patterns between both diabetes models (491 upregulated genes in the STZ model and 358 upregulated genes in HFD + STZ animals). Finally, we performed an enrichment analysis of the 849 selected genes. Whereas in the STZ model we found cellular pathways related to lipid biosynthesis and metabolism, in the HFD + STZ model we identified pathways related to immunometabolism. Some phenotypic differences observed in the models could be explained by transcriptomic results; however, further studies are needed to corroborate these findings. Our data confirm that the STZ and the HFD + STZ models are reliable experimental models for human T1D and T2D, respectively. These results also provide insight into alterations in the expression of specific liver genes and could be utilized in future studies focusing on diabetes complications associated with impaired liver function.


Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Liver , Animals , Liver/metabolism , Rats , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Male , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Diet, High-Fat/adverse effects , Transcriptome , Insulin Resistance/genetics , Gene Expression Profiling , Streptozocin , Disease Models, Animal , Blood Glucose/metabolism
20.
Front Endocrinol (Lausanne) ; 15: 1360525, 2024.
Article En | MEDLINE | ID: mdl-38650715

Diabetes is a common chronic metabolic disease with complex causes and pathogenesis. As an immunomodulator, vitamin D has recently become a research hotspot in the occurrence and development of diabetes and its complications. Many studies have shown that vitamin D can reduce the occurrence of diabetes and delay the progression of diabetes complications, and vitamin D can reduce oxidative stress, inhibit iron apoptosis, promote Ca2+ influx, promote insulin secretion, and reduce insulin resistance. Therefore, the prevention and correction of vitamin D deficiency is very necessary for diabetic patients, but further research is needed to confirm what serum levels of vitamin D3 are maintained in the body. This article provides a brief review of the relationship between vitamin D and diabetes, including its acute and chronic complications.


Diabetes Mellitus, Type 1 , Diabetes Mellitus, Type 2 , Disease Progression , Vitamin D Deficiency , Vitamin D , Humans , Diabetes Mellitus, Type 1/metabolism , Vitamin D/blood , Vitamin D/therapeutic use , Diabetes Mellitus, Type 2/metabolism , Vitamin D Deficiency/complications , Child , Adult , Oxidative Stress/drug effects
...