Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.011
Filter
1.
Endocrinology ; 165(8)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38954536

ABSTRACT

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.


Subject(s)
Glucose Tolerance Test , Insulin Secretion , Insulin-Secreting Cells , Insulin , Membrane Proteins , Animals , Membrane Proteins/metabolism , Membrane Proteins/genetics , Phosphorylation , Mice , Male , Insulin Secretion/physiology , Insulin-Secreting Cells/metabolism , Female , Insulin/metabolism , Tyrosine/metabolism , Aging/metabolism , Glucose Intolerance/metabolism , Mice, Inbred C57BL , Glucose/metabolism
2.
Int J Mol Sci ; 25(13)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-39000372

ABSTRACT

Circadian disruption increases the development of cardiovascular disease and diabetes. We found that circadian disruption causes glucose intolerance, cardiac fibrosis and adipocyte tissue dysfunction in male sand rats, Psammomys obesus. Whether these effects occur in female P. obesus is unknown. Male and female P. obesus were fed a high energy diet and exposed to a neutral (12 light:12 dark, control) or short (5 light:19 dark, circadian disruption) photoperiod for 20 weeks. Circadian disruption impaired glucose tolerance in males but not females. It also increased cardiac perivascular fibrosis and cardiac expression of inflammatory marker Ccl2 in males, with no effect in females. Females had reduced proapoptotic Bax mRNA and cardiac Myh7:Myh6 hypertrophy ratio. Cardiac protection in females occurred despite reductions in the clock gene Per2. Circadian disruption increased adipocyte hypertrophy in both males and females. This was concomitant with a reduction in adipocyte differentiation markers Pparg and Cebpa in males and females, respectively. Circadian disruption increased visceral adipose expression of inflammatory mediators Ccl2, Tgfb1 and Cd68 and reduced browning marker Ucp1 in males. However, these changes were not observed in females. Collectively, our study show that sex differentially influences the effects of circadian disruption on glucose tolerance, cardiac function and adipose tissue dysfunction.


Subject(s)
Adipocytes , Fibrosis , Gerbillinae , Glucose Intolerance , Animals , Female , Adipocytes/metabolism , Adipocytes/pathology , Male , Glucose Intolerance/metabolism , Myocardium/metabolism , Myocardium/pathology , Circadian Rhythm
3.
Front Immunol ; 15: 1416543, 2024.
Article in English | MEDLINE | ID: mdl-39050851

ABSTRACT

Introduction: Obesity is associated with a plethora of health complications, including increased susceptibility to infections or decreased vaccine efficacy, partly due to dysregulated immune responses. Monocytes play a crucial role in innate immunity, yet their functional alterations in obesity remain poorly understood. Methods: Here, we employed proteomic and metabolomic analyses to investigate monocyte characteristics in individuals with overweight, obesity, impaired glucose tolerance (IGT), and type 2 diabetes (T2D), compared to lean donors. Results and discussion: Our results revealed distinct molecular signatures in monocytes from individuals with obesity, with significant alterations in pathways related to metabolism, cellular migration, and phagocytosis. Moreover, LPS-induced activation of monocytes unveiled heightened metabolic reprogramming towards glycolysis in subjects with obesity accompanied by dysregulated cytokine responses and elevated oxidative stress. Additionally, monocytes from donors with obesity exhibited increased lipid droplet accumulation. These findings shed light on the immunometabolic dysregulation underlying obesity-associated immune dysfunction, highlighting potential targets for therapeutic intervention.


Subject(s)
Cytokines , Glycolysis , Monocytes , Obesity , Oxidative Stress , Humans , Obesity/immunology , Obesity/metabolism , Monocytes/immunology , Monocytes/metabolism , Cytokines/metabolism , Male , Female , Adult , Middle Aged , Diabetes Mellitus, Type 2/immunology , Diabetes Mellitus, Type 2/metabolism , Proteomics/methods , Glucose Intolerance/immunology , Glucose Intolerance/metabolism
4.
Int J Mol Sci ; 25(14)2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39062999

ABSTRACT

Circadian disruption causes glucose intolerance, cardiac fibrosis, and adipocyte dysfunction in sand rats (Psammomys obesus). Exercise intervention can improve glucose metabolism, insulin sensitivity, adipose tissue function and protect against inflammation. We investigated the influence of exercise on male P. obesus exposed to a short photoperiod (5 h light:19 h dark) and high-energy diet. Exercise reduced glucose intolerance. Exercise reduced cardiac expression of inflammatory marker Ccl2 and Bax:Bcl2 apoptosis ratio. Exercise increased heart:body weight ratio and hypertrophy marker Myh7:Myh6, yet reduced Gata4 expression. No phenotypic changes were observed in perivascular fibrosis and myocyte area. Exercise reduced visceral adipose expression of inflammatory transcription factor Rela, adipogenesis marker Ppard and browning marker Ppargc1a, but visceral adipocyte size was unaffected. Conversely, exercise reduced subcutaneous adipocyte size but did not affect any molecular mediators. Exercise increased ZT7 Bmal1 and Per2 in the suprachiasmatic nucleus and subcutaneous Per2. Our study provides new molecular insights and histological assessments on the effect of exercise on cardiac inflammation, adipose tissue dysfunction and circadian gene expression in P. obesus exposed to short photoperiod and high-energy diet. These findings have implications for the protective benefits of exercise for shift workers in order to reduce the risk of diabetes and cardiovascular disease.


Subject(s)
Adipose Tissue , Gerbillinae , Glucose Intolerance , Photoperiod , Physical Conditioning, Animal , Animals , Male , Glucose Intolerance/metabolism , Adipose Tissue/metabolism , Inflammation/metabolism , Inflammation/pathology , Diet, High-Fat/adverse effects , Myocardium/metabolism , Myocardium/pathology
5.
PLoS One ; 19(6): e0304761, 2024.
Article in English | MEDLINE | ID: mdl-38843265

ABSTRACT

Type 2 diabetes predisposes patients to heart disease, which is the primary cause of death across the globe. Type 2 diabetes often accompanies obesity and is defined by insulin resistance and abnormal glucose handling. Insulin resistance impairs glucose uptake and results in hyperglycemia, which damages tissues such as kidneys, liver, and heart. 2-oxoglutarate (2-OG)- and iron-dependent oxygenases (2-OGDOs), a family of enzymes regulating various aspects of cellular physiology, have been studied for their role in obesity and diet-induced insulin resistance. However, nothing is known of the 2-OGDO family member 2-oxoglutarate and iron-dependent prolyl hydroxylase domain containing protein 1 (OGFOD1) in this setting. OGFOD1 deletion leads to protection in cardiac ischemia-reperfusion injury and cardiac hypertrophy, which are two cardiac events that can lead to heart failure. Considering the remarkable correlation between heart disease and diabetes, the cardioprotection observed in OGFOD1-knockout mice led us to challenge these knockouts with high-fat diet. Wildtype mice fed a high-fat diet developed diet-induced obesity, insulin resistance, and glucose intolerance, but OGFOD1 knockout mice fed this same diet were resistant to diet-induced obesity and insulin resistance. These results support OGFOD1 down-regulation as a strategy for preventing obesity and insulin handling defects.


Subject(s)
Diet, High-Fat , Insulin Resistance , Mice, Knockout , Obesity , Animals , Obesity/metabolism , Obesity/genetics , Mice , Diet, High-Fat/adverse effects , Male , Prolyl Hydroxylases/metabolism , Prolyl Hydroxylases/genetics , Glucose Intolerance/metabolism , Glucose Intolerance/genetics , Mice, Inbred C57BL , Gene Deletion , Cardiomegaly/metabolism , Cardiomegaly/prevention & control , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/genetics
6.
FASEB J ; 38(11): e23715, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38837260

ABSTRACT

Impaired intestinal permeability induces systemic inflammation and metabolic disturbance. The effect of a leaky gut on metabolism in skeletal muscle, a major nutrient consumer, remains unclear. In this study, we aimed to investigate the glucose metabolic function of the whole body and skeletal muscles in a mouse model of diet-induced intestinal barrier dysfunction. At Week 2, we observed higher intestinal permeability in mice fed a titanium dioxide (TiO2)-containing diet than that of mice fed a normal control diet. Subsequently, systemic glucose and insulin tolerance were found to be impaired. In the skeletal muscle, glucose uptake and phosphorylation levels in insulin signaling were lower in the TiO2 group than those in the control group. Additionally, the levels of pro-inflammatory factors were higher in TiO2-fed mice than those in the control group. We observed higher carboxymethyl-lysin (CML) levels in the plasma and intestines of TiO2-fed mice and lower insulin-dependent glucose uptake in CML-treated cultured myotubes than those in the controls. Finally, soluble dietary fiber supplementation improved glucose and insulin intolerance, suppressed plasma CML, and improved intestinal barrier function. These results suggest that an impaired intestinal barrier leads to systemic glucose intolerance, which is associated with glucose metabolism dysfunction in the skeletal muscles due to circulating CML derived from the intestine. This study highlights that the intestinal condition regulates muscle and systemic metabolic health.


Subject(s)
Lysine , Muscle, Skeletal , Titanium , Animals , Mice , Muscle, Skeletal/metabolism , Muscle, Skeletal/drug effects , Male , Lysine/analogs & derivatives , Lysine/metabolism , Mice, Inbred C57BL , Food Additives/pharmacology , Insulin/blood , Insulin/metabolism , Glucose/metabolism , Glucose Intolerance/metabolism , Intestinal Mucosa/metabolism
7.
Endocrinol Diabetes Metab ; 7(4): e487, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38867382

ABSTRACT

INTRODUCTION: High-fat diet (HFD) consumption and being exposed to daily psychological stress, common environmental factors in modern lifestyle, play an important role on metabolic disorders such as glucose homeostasis impairment. The aim of this study was to investigate the effects of high-fat diet (HFD) and psychological stress combination on metabolic response to chronic psychological stress in male rats. METHOD: Male Wistar rats were divided into HFD, and normal diet (ND) groups and then into stress and nonstress subgroups. The diets were applied for 5 weeks, and psychological stress was induced for 7 consecutive days. Then, blood samples were taken to measure glucose, insulin, free fatty acids (FFA), and leptin and corticosterone concentrations. Subsequently, glucose-stimulated insulin release from pancreatic isolated islets was assessed. RESULTS: HFD did not significantly change fasting plasma glucose, insulin and corticosterone levels, whereas increased plasma leptin (7.05 ± 0.33) and FFA (p < 0.01) levels and impaired glucose tolerance. Additionally, HFD and stress combination induced more profound glucose intolerance associated with increased plasma corticosterone (p < 0.01) and leptin (8.63 ± 0.38) levels. However, insulin secretion from isolated islets did not change in the presence of high-fat diet and/or stress. CONCLUSION: HFD should be considered as an intensified factor of metabolic impairments caused by chronic psychological stress.


Subject(s)
Blood Glucose , Corticosterone , Diet, High-Fat , Insulin , Leptin , Rats, Wistar , Stress, Psychological , Animals , Male , Stress, Psychological/metabolism , Diet, High-Fat/adverse effects , Rats , Corticosterone/blood , Insulin/blood , Leptin/blood , Blood Glucose/metabolism , Fatty Acids, Nonesterified/blood , Islets of Langerhans/metabolism , Glucose Intolerance/etiology , Glucose Intolerance/metabolism
8.
Nat Commun ; 15(1): 5413, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38926424

ABSTRACT

Diet composition impacts metabolic health and is now recognized to shape the immune system, especially in the intestinal tract. Nutritional imbalance and increased caloric intake are induced by high-fat diet (HFD) in which lipids are enriched at the expense of dietary fibers. Such nutritional challenge alters glucose homeostasis as well as intestinal immunity. Here, we observed that short-term HFD induced dysbiosis, glucose intolerance and decreased intestinal RORγt+ CD4 T cells, including peripherally-induced Tregs and IL17-producing (Th17) T cells. However, supplementation of HFD-fed male mice with the fermentable dietary fiber fructooligosaccharides (FOS) was sufficient to maintain RORγt+ CD4 T cell subsets and microbial species known to induce them, alongside having a beneficial impact on glucose tolerance. FOS-mediated normalization of Th17 cells and amelioration of glucose handling required the cDC2 dendritic cell subset in HFD-fed animals, while IL-17 neutralization limited FOS impact on glucose tolerance. Overall, we uncover a pivotal role of cDC2 in the control of the immune and metabolic effects of FOS in the context of HFD feeding.


Subject(s)
Dendritic Cells , Diet, High-Fat , Homeostasis , Mice, Inbred C57BL , Oligosaccharides , Animals , Oligosaccharides/pharmacology , Diet, High-Fat/adverse effects , Dendritic Cells/immunology , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Male , Mice , Th17 Cells/immunology , Th17 Cells/metabolism , Th17 Cells/drug effects , Glucose/metabolism , Interleukin-17/metabolism , Dietary Fiber/pharmacology , Glucose Intolerance/immunology , Glucose Intolerance/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Dysbiosis/immunology , Gastrointestinal Microbiome/drug effects
9.
Nat Commun ; 15(1): 5217, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38890307

ABSTRACT

Dietary protein is a critical regulator of metabolic health and aging. Low protein diets are associated with healthy aging in humans, and dietary protein restriction extends the lifespan and healthspan of mice. In this study, we examined the effect of protein restriction (PR) on metabolic health and the development and progression of Alzheimer's disease (AD) in the 3xTg mouse model of AD. Here, we show that PR promotes leanness and glycemic control in 3xTg mice, specifically rescuing the glucose intolerance of 3xTg females. PR induces sex-specific alterations in circulating and brain metabolites, downregulating sphingolipid subclasses in 3xTg females. PR also reduces AD pathology and mTORC1 activity, increases autophagy, and improves the cognition of 3xTg mice. Finally, PR improves the survival of 3xTg mice. Our results suggest that PR or pharmaceutical interventions that mimic the effects of this diet may hold promise as a treatment for AD.


Subject(s)
Alzheimer Disease , Brain , Diet, Protein-Restricted , Disease Models, Animal , Disease Progression , Mice, Transgenic , Animals , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Female , Male , Mice , Brain/metabolism , Brain/pathology , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Autophagy , Glucose Intolerance/metabolism , Sphingolipids/metabolism , Cognition , Mice, Inbred C57BL
10.
Nitric Oxide ; 149: 75-84, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38879114

ABSTRACT

Obesity is commonly linked with white adipose tissue (WAT) dysfunction, setting off inflammation and oxidative stress, both key contributors to the cardiometabolic complications associated with obesity. To improve metabolic and cardiovascular health, countering these inflammatory and oxidative signaling processes is crucial. Offering potential in this context, the activation of nuclear factor erythroid 2-related factor 2 (Nrf2) by nitro-fatty acids (NO2-FA) promote diverse anti-inflammatory signaling and counteract oxidative stress. Additionally, we previously highlighted that nitro-oleic acid (NO2-OA) preferentially accumulates in WAT and provides protection against already established high fat diet (HFD)-mediated impaired glucose tolerance. The precise mechanism accounting for these protective effects remained largely unexplored until now. Herein, we reveal that protective effects of improved glucose tolerance by NO2-OA is absent when Nrf2 is specifically ablated in adipocytes (ANKO mice). NO2-OA treatment did not alter body weight between ANKO and littermate controls (Nrf2fl/fl) mice on both the HFD and low-fat diet (LFD). As expected, at day 76 (before NO2-OA treatment) and notably at day 125 (daily treatment of 15 mg/kg NO2-OA for 48 days), both HFD-fed Nrf2fl/fl and ANKO mice exhibited increased fat mass and reduced lean mass compared to LFD controls. However, throughout the NO2-OA treatment, no distinction was observed between Nrf2fl/fl and ANKO in the HFD-fed mice as well as in the Nrf2fl/fl mice fed a LFD. Glucose tolerance tests revealed impaired glucose tolerance in HFD-fed Nrf2fl/fl and ANKO compared to LFD-fed Nrf2fl/fl mice. Notably, NO2-OA treatment improved glucose tolerance in HFD-fed Nrf2fl/fl but did not yield the same improvement in ANKO mice at days 15, 30, and 55 of treatment. Unraveling the pathways linked to NO2-OA's protective effects in obesity-mediated impairment in glucose tolerance is pivotal within the realm of precision medicine, crucially propelling future applications and refining novel drug-based strategies.


Subject(s)
Adipocytes , Diet, High-Fat , NF-E2-Related Factor 2 , Obesity , Animals , NF-E2-Related Factor 2/metabolism , Obesity/metabolism , Obesity/drug therapy , Diet, High-Fat/adverse effects , Mice , Adipocytes/metabolism , Adipocytes/drug effects , Male , Mice, Inbred C57BL , Glucose Intolerance/metabolism , Oleic Acids/pharmacology , Mice, Knockout
11.
Am J Physiol Endocrinol Metab ; 327(2): E194-E202, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38836778

ABSTRACT

Our previous study revealed that over 50% of recipients with pretransplant impaired glucose tolerance (IGT) improved to normal glucose tolerance after kidney transplantation. However, the mechanism is unclear. We aimed to investigate whether the changes in glucose tolerance are associated with ß-cell function and insulin resistance in Japanese kidney transplant recipients with pretransplant IGT. Of the 265 recipients who received kidney transplantation, 54 with pretransplant IGT were included. We divided the recipients into improvement and nonimprovement groups according to the change in the area under the curve for glucose obtained from the oral glucose tolerance test (OGTT). ß-Cell function was estimated by the insulin secretion sensitivity index-2 (ISSI-2) and the disposition index (DI). Insulin resistance was estimated by the Matsuda index (MI) and the homeostasis model assessment of insulin resistance (HOMA-IR). ISSI-2 and DI increased significantly after transplantation in the improved group (P < 0.01, P < 0.05, respectively), but not in the nonimproved group. ΔISSI-2 and ΔDI were significantly and positively associated with pretransplant 60-min OGTT plasma glucose levels (both P < 0.01). There were no differences in MI or HOMA-IR between these two groups after transplantation. In recipients not on pretransplant dialysis, a significant negative association was found between Δblood urea nitrogen (BUN) and ΔDI (correlation coefficient = -0.48, P < 0.05). In pretransplant IGT recipients, improvements in glucose tolerance after kidney transplantation were linked to improvements in ß-cell function. The higher the 60-min OGTT plasma glucose level, the greater the improvement in posttransplant ß-cell function. Improvements in BUN after transplantation were associated with improvements in ß-cell function.NEW & NOTEWORTHY In recipients with pretransplant impaired glucose tolerance, improvements in glucose tolerance after kidney transplantation were associated with improvements in ß-cell function. The higher the pretransplant 60-min OGTT plasma glucose level, the greater the improvement in posttransplant ß-cell function. Although glucose tolerance is known to be impaired after transplantation, the present study focused on the reason for the improvement in glucose tolerance rather than the development of posttransplantation diabetes mellitus.


Subject(s)
Blood Glucose , Glucose Intolerance , Glucose Tolerance Test , Insulin Resistance , Insulin-Secreting Cells , Kidney Transplantation , Humans , Insulin-Secreting Cells/metabolism , Male , Glucose Intolerance/metabolism , Female , Middle Aged , Insulin Resistance/physiology , Adult , Blood Glucose/metabolism , Aged
12.
BMC Res Notes ; 17(1): 128, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38711110

ABSTRACT

The elemental composition of chemical elements can vary between healthy and diseased tissues, providing essential insights into metabolic processes in physiological and diseased states. This study aimed to evaluate the calcium (Ca) and phosphorus (P) levels in the bones of rats with/without streptozotocin-induced diabetes and/or exposure to infrasound. X-ray fluorescence spectroscopy was used to determine the concentrations of Ca and P in Wistar rat tibiae samples.The results showed a significant decrease in bone P concentration in streptozotocin-induced diabetic rats compared to untreated animals. Similarly, the Ca/P ratio was higher in the streptozotocin-induced diabetic group. No significant differences were observed in bone Ca concentration between the studied groups or between animals exposed and not exposed to infrasound.Moreover, streptozotocin-induced diabetic rats had lower bone P concentration but unaltered bone Ca concentration compared to untreated rats. Infrasound exposure did not impact bone Ca or P levels. The reduced bone P concentration may be associated with an increased risk of bone fractures in diabetes.


Subject(s)
Calcium , Diabetes Mellitus, Experimental , Phosphorus , Rats, Wistar , Streptozocin , Animals , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/chemically induced , Phosphorus/metabolism , Calcium/metabolism , Rats , Male , Spectrometry, X-Ray Emission , Tibia/metabolism , Sound/adverse effects , Bone and Bones/metabolism , Glucose Intolerance/metabolism
13.
Respir Med ; 228: 107664, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38759874

ABSTRACT

BACKGROUND: Impaired glycemic control and the subsequent development of Cystic fibrosis Related Diabetes (CFRD) are prevalent complications, affecting up to 50 % of adults with cystic fibrosis (CF). CFTR modulator (CFTRm) therapies improve pulmonary functions, reduce exacerbation rates, increase survival in people with CF (pwCF) and appear to have a positive effect on extrapulmonary manifestations, such as nutritional state, improvements in upper respiratory symptoms, and quality of life. Initial findings indicate that CFTRm may have a positive impact on short-term glycemic control; however, long-term effects remain uncertain at present. METHODS: In this retrospective study, data were collected and analyzed on 15 pwCF, ages 13-37 years, started on CFTRm therapy. Oral Glucose Tolerance Test (OGTT) results were compared pre- and post-CFTRm therapy. RESULTS: The 120-min OGTT value decreased from 159.7 mg/dL to 130.4 mg/dL post-CFTRm (p = 0.047). The average time elapsed between the two OGTTs was 49.87 months (ranging 9-157 months, median 38 months). Glycemic status improved in six pwCF (two CFRD to normal (NGT)/indeterminate (INDET) glucose tolerance; two impaired glucose tolerance (IGT) to INDET; two INDET to NGT) and worsened in one (IGT to CFRD). Six pwCF and NGT remained stable with no changes in glycemic status throughout the follow-up period. CONCLUSIONS: CFTRm therapy may decelerate the glycemic control deterioration in pwCF over an extended period. These findings indicate the need for periodic OGTTs following the initiation of CFTRm therapy to appropriately adjust insulin requirements and prevent hypoglycemia. Further larger cohorts are required to authenticate and substantiate these findings.


Subject(s)
Blood Glucose , Cystic Fibrosis Transmembrane Conductance Regulator , Cystic Fibrosis , Glucose Tolerance Test , Humans , Cystic Fibrosis/drug therapy , Cystic Fibrosis/physiopathology , Cystic Fibrosis/complications , Adolescent , Adult , Retrospective Studies , Male , Female , Young Adult , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Blood Glucose/metabolism , Blood Glucose/drug effects , Aminophenols/therapeutic use , Quinolones/therapeutic use , Aminopyridines/therapeutic use , Benzodioxoles/therapeutic use , Diabetes Mellitus/drug therapy , Diabetes Mellitus/metabolism , Glycemic Control/methods , Time Factors , Glucose/metabolism , Glucose Intolerance/drug therapy , Glucose Intolerance/metabolism
14.
Nat Commun ; 15(1): 4527, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38811550

ABSTRACT

The IL-22RA1 receptor is highly expressed in the pancreas, and exogenous IL-22 has been shown to reduce endoplasmic reticulum and oxidative stress in human pancreatic islets and promote secretion of high-quality insulin from beta-cells. However, the endogenous role of IL-22RA1 signaling on these cells remains unclear. Here, we show that antibody neutralisation of IL-22RA1 in cultured human islets leads to impaired insulin quality and increased cellular stress. Through the generation of mice lacking IL-22ra1 specifically on pancreatic alpha- or beta-cells, we demonstrate that ablation of murine beta-cell IL-22ra1 leads to similar decreases in insulin secretion, quality and islet regeneration, whilst increasing islet cellular stress, inflammation and MHC II expression. These changes in insulin secretion led to impaired glucose tolerance, a finding more pronounced in female animals compared to males. Our findings attribute a regulatory role for endogenous pancreatic beta-cell IL-22ra1 in insulin secretion, islet regeneration, inflammation/cellular stress and appropriate systemic metabolic regulation.


Subject(s)
Glucose , Homeostasis , Insulin-Secreting Cells , Insulin , Mice, Knockout , Receptors, Interleukin , Animals , Insulin-Secreting Cells/metabolism , Receptors, Interleukin/metabolism , Receptors, Interleukin/genetics , Female , Humans , Male , Insulin/metabolism , Mice , Glucose/metabolism , Insulin Secretion , Mice, Inbred C57BL , Interleukin-22 , Glucose Intolerance/metabolism , Interleukins/metabolism , Interleukins/genetics , Aging/metabolism
15.
FASEB J ; 38(10): e23690, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38795327

ABSTRACT

Alterations to gene transcription and DNA methylation are a feature of many liver diseases including fatty liver disease and liver cancer. However, it is unclear whether the DNA methylation changes are a cause or a consequence of the transcriptional changes. It is even possible that the methylation changes are not required for the transcriptional changes. If DNA methylation is just a minor player in, or a consequence of liver transcriptional change, then future studies in this area should focus on other systems such as histone tail modifications. To interrogate the importance of de novo DNA methylation, we generated mice that are homozygous mutants for both Dnmt3a and Dnmt3b in post-natal liver. These mice are viable and fertile with normal sized livers. Males, but not females, showed increased adipose depots, yet paradoxically, improved glucose tolerance on both control diet and high-fat diets (HFD). Comparison of the transcriptome and methylome with RNA sequencing and whole-genome bisulfite sequencing in adult hepatocytes revealed that widespread loss of methylation in CpG-rich regions in the mutant did not induce loss of homeostatic transcriptional regulation. Similarly, extensive transcriptional changes induced by HFD did not require de novo DNA methylation. The improved metabolic phenotype of the Dnmt3a/3b mutant mice may be mediated through the dysregulation of a subset of glucose and fat metabolism genes which increase both glucose uptake and lipid export by the liver. However, further work is needed to confirm this.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases , DNA Methylation , DNA Methyltransferase 3A , DNA Methyltransferase 3B , Diet, High-Fat , Glucose Intolerance , Liver , Animals , Male , Diet, High-Fat/adverse effects , Liver/metabolism , Mice , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A/metabolism , Glucose Intolerance/metabolism , Glucose Intolerance/genetics , Female , Mice, Inbred C57BL
16.
Front Endocrinol (Lausanne) ; 15: 1376530, 2024.
Article in English | MEDLINE | ID: mdl-38681771

ABSTRACT

Background/Objectives: Glucagon is important in the maintenance of glucose homeostasis, with also effects on lipids. In this study, we aimed to apply a recently developed model of glucagon kinetics to determine the sensitivity of glucagon variations (especially, glucagon inhibition) to insulin levels ("alpha-cell insulin sensitivity"), during oral glucose administration. Subjects/Methods: We studied 50 participants (spanning from normal glucose tolerance to type 2 diabetes) undergoing frequently sampled 5-hr oral glucose tolerance test (OGTT). The alpha-cell insulin sensitivity and the glucagon kinetics were assessed by a mathematical model that we developed previously. Results: The alpha-cell insulin sensitivity parameter (named SGLUCA; "GLUCA": "glucagon") was remarkably variable among participants (CV=221%). SGLUCA was found inversely correlated with the mean glycemic values, as well as with 2-hr glycemia of the OGTT. When stratifying participants into two groups (normal glucose tolerance, NGT, N=28, and impaired glucose regulation/type 2 diabetes, IGR_T2D, N=22), we found that SGLUCA was lower in the latter (1.50 ± 0.50·10-2 vs. 0.26 ± 0.14·10-2 ng·L-1 GLUCA/pmol·L-1 INS, in NGT and IGR_T2D, respectively, p=0.009; "INS": "insulin"). Conclusions: The alpha-cell insulin sensitivity is highly variable among subjects, and it is different in groups at different glucose tolerance. This may be relevant for defining personalized treatment schemes, in terms of dietary prescriptions but also for treatments with glucagon-related agents.


Subject(s)
Blood Glucose , Diabetes Mellitus, Type 2 , Glucagon , Glucose , Adult , Aged , Female , Humans , Male , Middle Aged , Administration, Oral , Blood Glucose/metabolism , Blood Glucose/analysis , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Glucagon/blood , Glucagon-Secreting Cells/metabolism , Glucagon-Secreting Cells/drug effects , Glucose/metabolism , Glucose/administration & dosage , Glucose Intolerance/blood , Glucose Intolerance/metabolism , Glucose Tolerance Test , Insulin/blood , Insulin/administration & dosage , Insulin Resistance , Kinetics , Models, Theoretical
17.
Peptides ; 177: 171214, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38615716

ABSTRACT

In this systematic review, we assessed the safety and possible safety events of native glucose-dependent insulinotropic polypeptide (GIP)(1-42) in human studies with administration of synthetic human GIP. We searched the PubMed database for all trials investigating synthetic human GIP(1-42) administration. A total of 67 studies were included. Study duration ranged from 30 min to 6 days. In addition to healthy individuals, the studies included individuals with impaired glucose tolerance, type 2 diabetes, type 1 diabetes, chronic pancreatitis and secondary diabetes, latent autoimmune diabetes in adults, diabetes caused by a mutation in the hepatocyte nuclear factor 1-alpha gene, end-stage renal disease, chronic renal insufficiency, critical illness, hypoparathyroidism, or cystic fibrosis-related diabetes. Of the included studies, 78% did not mention safety events, 10% of the studies reported that no safety events were observed in relation to GIP administration, and 15% of the studies reported safety events in relation to GIP administration with most frequently reported event being a moderate and transient increased heart rate. Gastrointestinal safety events, and changes in blood pressure were also reported. Plasma concentration of active GIP(1-42) increased linearly with dose independent of participant phenotype. There was no significant correlation between achieved maximal concentration of GIP(1-42) and reported safety events. Clearance rates of GIP(1-42) were similar between participant groups. In conclusion, the available data indicate that GIP(1-42) in short-term (up to 6 days) infusion studies is generally well-tolerated. The long-term safety of continuous GIP(1-42) administration is unknown.


Subject(s)
Gastric Inhibitory Polypeptide , Humans , Peptide Fragments , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Glucose Intolerance/metabolism , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/metabolism
18.
Can J Physiol Pharmacol ; 102(7): 422-428, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38669698

ABSTRACT

We aimed to determine if cheese could reduce glucose intolerance in aged rats with overt type 2 diabetes (T2D). Male Sprague-Dawley rats treated with high-fat diet (HFD) and streptozotocin (STZ) to elicit T2D were hyperglycemic. One week after STZ injection, low-fat (LOW) or regular-fat (REG) cheese was provided for 5 weeks and compared with T2D and low-fat diet reference (REF) groups. Food intake and weight gain were similar in all groups. Oral glucose tolerance tests revealed glucose intolerance in T2D rats that was partially ameliorated by LOW but not REG. Insulin secretion during the oral glucose tolerance test was impaired in T2D and REG at 10 min (p < 0.05) but the iAUC was highly variable in all groups and statistical differences were not detected (p > 0.05). ß-cell mass and pancreatic insulin content in T2D and REG were 50% lower than REF (p < 0.05), whereas LOW was not significantly different. Although isolated islets from all groups responded to glucose, the absolute amount of insulin secreted by T2D and REG was markedly reduced compared with REF, while LOW islets had relatively normal secretion. In conclusion, LOW but not REG cheese enhanced ß-cell recovery from HFD/STZ treatment that led to amelioration of glucose tolerance within 5 weeks.


Subject(s)
Cheese , Diabetes Mellitus, Type 2 , Glucose Intolerance , Insulin Secretion , Insulin-Secreting Cells , Insulin , Rats, Sprague-Dawley , Animals , Male , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/drug effects , Glucose Intolerance/metabolism , Diabetes Mellitus, Type 2/metabolism , Insulin Secretion/drug effects , Rats , Insulin/metabolism , Insulin/blood , Glucose Tolerance Test , Diet, High-Fat/adverse effects , Diet, Fat-Restricted , Disease Models, Animal , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/metabolism
19.
J Biol Chem ; 300(6): 107328, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38679332

ABSTRACT

Management of chronic obesity-associated metabolic disorders is a key challenge for biomedical researchers. During chronic obesity, visceral adipose tissue (VAT) undergoes substantial transformation characterized by a unique lipid-rich hypoxic AT microenvironment which plays a crucial role in VAT dysfunction, leading to insulin resistance (IR) and type 2 diabetes. Here, we demonstrate that obese AT microenvironment triggers the release of miR-210-3p microRNA-loaded extracellular vesicles from adipose tissue macrophages, which disseminate miR-210-3p to neighboring adipocytes, skeletal muscle cells, and hepatocytes through paracrine and endocrine actions, thereby influencing insulin sensitivity. Moreover, EVs collected from Dicer-silenced miR-210-3p-overexpressed bone marrow-derived macrophages induce glucose intolerance and IR in lean mice. Mechanistically, miR-210-3p interacts with the 3'-UTR of GLUT4 mRNA and silences its expression, compromising cellular glucose uptake and insulin sensitivity. Therapeutic inhibition of miR-210-3p in VAT notably rescues high-fat diet-fed mice from obesity-induced systemic glucose intolerance. Thus, targeting adipose tissue macrophage-specific miR-210-3p during obesity could be a promising strategy for managing IR and type 2 diabetes.


Subject(s)
Glucose Transporter Type 4 , Insulin Resistance , Macrophages , MicroRNAs , Obesity , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , Obesity/metabolism , Obesity/genetics , Obesity/pathology , Macrophages/metabolism , Mice , Glucose Transporter Type 4/metabolism , Glucose Transporter Type 4/genetics , Male , Mice, Inbred C57BL , Adipose Tissue/metabolism , Adipose Tissue/pathology , Humans , Diet, High-Fat/adverse effects , Glucose Intolerance/metabolism , Glucose Intolerance/genetics , Glucose Intolerance/pathology , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics , Intra-Abdominal Fat/metabolism , Intra-Abdominal Fat/pathology
20.
Sci Rep ; 14(1): 7670, 2024 04 01.
Article in English | MEDLINE | ID: mdl-38561446

ABSTRACT

Dietary factors such as food texture affect feeding behavior and energy metabolism, potentially causing obesity and type 2 diabetes. We previously found that rats fed soft pellets (SPs) were neither hyperphagic nor overweight but demonstrated glucose intolerance, insulin resistance, and hyperplasia of pancreatic ß-cells. In the present study, we investigated the mechanism of muscle atrophy in rats that had been fed SPs on a 3-h time-restricted feeding schedule for 24 weeks. As expected, the SP rats were normal weight; however, they developed insulin resistance, glucose intolerance, and fat accumulation. In addition, skeletal muscles of SP rats were histologically atrophic and demonstrated disrupted insulin signaling. Furthermore, we learned that the muscle atrophy of the SP rats developed via the IL-6-STAT3-SOCS3 and ubiquitin-proteasome pathways. Our data show that the dietary habit of consuming soft foods can lead to not only glucose intolerance or insulin resistance but also muscle atrophy.


Subject(s)
Diabetes Mellitus, Type 2 , Glucose Intolerance , Insulin Resistance , Rats , Animals , Insulin Resistance/physiology , Glucose Intolerance/metabolism , Diabetes Mellitus, Type 2/metabolism , Insulin/metabolism , Muscular Atrophy/etiology , Muscular Atrophy/metabolism , Muscle, Skeletal/metabolism , Diet , Diet, High-Fat
SELECTION OF CITATIONS
SEARCH DETAIL