Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 25.428
1.
JCI Insight ; 9(11)2024 Jun 10.
Article En | MEDLINE | ID: mdl-38855868

Lactate elevation is a well-characterized biomarker of mitochondrial dysfunction, but its role in diabetic kidney disease (DKD) is not well defined. Urine lactate was measured in patients with type 2 diabetes (T2D) in 3 cohorts (HUNT3, SMART2D, CRIC). Urine and plasma lactate were measured during euglycemic and hyperglycemic clamps in participants with type 1 diabetes (T1D). Patients in the HUNT3 cohort with DKD had elevated urine lactate levels compared with age- and sex-matched controls. In patients in the SMART2D and CRIC cohorts, the third tertile of urine lactate/creatinine was associated with more rapid estimated glomerular filtration rate decline, relative to first tertile. Patients with T1D demonstrated a strong association between glucose and lactate in both plasma and urine. Glucose-stimulated lactate likely derives in part from proximal tubular cells, since lactate production was attenuated with sodium-glucose cotransporter-2 (SGLT2) inhibition in kidney sections and in SGLT2-deficient mice. Several glycolytic genes were elevated in human diabetic proximal tubules. Lactate levels above 2.5 mM potently inhibited mitochondrial oxidative phosphorylation in human proximal tubule (HK2) cells. We conclude that increased lactate production under diabetic conditions can contribute to mitochondrial dysfunction and become a feed-forward component to DKD pathogenesis.


Diabetes Mellitus, Type 1 , Diabetes Mellitus, Type 2 , Diabetic Nephropathies , Glycolysis , Lactic Acid , Humans , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Animals , Mice , Lactic Acid/metabolism , Lactic Acid/blood , Female , Male , Middle Aged , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/complications , Mitochondria/metabolism , Adult , Glomerular Filtration Rate , Aged , Kidney Tubules, Proximal/metabolism , Glucose/metabolism , Oxidative Phosphorylation , Biomarkers/metabolism , Sodium-Glucose Transporter 2/metabolism , Sodium-Glucose Transporter 2/genetics , Sodium-Glucose Transporter 2 Inhibitors/pharmacology
2.
Nat Commun ; 15(1): 4827, 2024 Jun 06.
Article En | MEDLINE | ID: mdl-38844451

Adipose progenitor cells (APCs) are heterogeneous stromal cells and help to maintain metabolic homeostasis. However, the influence of obesity on human APC heterogeneity and the role of APC subpopulations on regulating glucose homeostasis remain unknown. Here, we find that APCs in human visceral adipose tissue contain four subsets. The composition and functionality of APCs are altered in patients with type 2 diabetes (T2D). CD9+CD55low APCs are the subset which is significantly increased in T2D patients. Transplantation of these cells from T2D patients into adipose tissue causes glycemic disturbance. Mechanistically, CD9+CD55low APCs promote T2D development through producing bioactive proteins to form a detrimental niche, leading to upregulation of adipocyte lipolysis. Depletion of pathogenic APCs by inducing intracellular diphtheria toxin A expression or using a hunter-killer peptide improves obesity-related glycemic disturbance. Collectively, our data provide deeper insights in human APC functionality and highlights APCs as a potential therapeutic target to combat T2D. All mice utilized in this study are male.


Diabetes Mellitus, Type 2 , Glucose , Homeostasis , Obesity , Single-Cell Analysis , Stem Cells , Humans , Animals , Single-Cell Analysis/methods , Diabetes Mellitus, Type 2/metabolism , Male , Mice , Stem Cells/metabolism , Glucose/metabolism , Obesity/metabolism , Obesity/pathology , Adipocytes/metabolism , Intra-Abdominal Fat/metabolism , Intra-Abdominal Fat/cytology , Adipose Tissue/metabolism , Adipose Tissue/cytology , Mice, Inbred C57BL , Lipolysis , Female , Middle Aged
3.
Sci Rep ; 14(1): 12910, 2024 06 05.
Article En | MEDLINE | ID: mdl-38839813

Impaired insulin secretory capacity is associated with high glycemic variability in patients with type 2 diabetes (T2DM). However, there are no existing reports on the association between insulin secretory capacity and time in range (TIR). This retrospective study involved 330 T2DM admitted for diabetes education who underwent intermittently scanned continuous glucose monitoring (isCGM) and had their fasting serum C-peptide immunoreactivity (S-CPR) measured within 5 days of admission. The baseline characteristics were as follows: age, 60.2 years; glycated hemoglobin (HbA1c), 9.2%; S-CPR, 2.2 ng/mL; S-CPR index (S-CPR [ng/mL]/fasting plasma glucose [mg/dL] × 100), 1.6; and TIR, 60.3%. TIR correlated significantly with the S-CPR index, which was confirmed by multivariate analysis that included various factors such as HbA1c. Receiver operating characteristic (ROC) analysis showed that 1.88 was the optimal S-CPR index level to predict TIR ≥ 70%. In addition to HbA1c and biguanide use, the S-CPR index was a significant factor associated with TIR > 70%. S-CPR index values of ≥ 1.88 also correlated significantly with TIR > 70%. In conclusion, insulin secretory capacity is associated with TIR in Japanese T2DM, suggesting that the S-CPR index might be a potentially useful biomarker insulin secretory capacity, in association with TIR.Trial registration UMIN0000254333.


Blood Glucose , Diabetes Mellitus, Type 2 , Insulin Secretion , Insulin , Humans , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Male , Female , Middle Aged , Retrospective Studies , Insulin/blood , Aged , Blood Glucose/analysis , Blood Glucose/metabolism , Japan , Glycated Hemoglobin/analysis , Glycated Hemoglobin/metabolism , C-Peptide/blood , East Asian People
4.
PLoS One ; 19(6): e0304870, 2024.
Article En | MEDLINE | ID: mdl-38900754

The underlying causes of breast cancer are diverse, however, there is a striking association between type 2 diabetes and poor patient outcomes. Platelet activation is a common feature of both type 2 diabetes and breast cancer and has been implicated in tumourigenesis through a multitude of pathways. Here transcriptomic analysis of type 2 diabetes patient-derived platelet microvesicles revealed an altered miRNA signature compared with normoglycaemic control patients. Interestingly, interrogation of these data identifies a shift towards an oncogenic signature in type 2 diabetes-derived platelet microvesicles, with increased levels of miRNAs implicated in breast cancer progression and poor prognosis. Functional studies demonstrate that platelet microvesicles isolated from type 2 diabetes patient blood are internalised by triple-negative breast cancer cells in vitro, and that co-incubation with type 2 diabetes patient-derived platelet microvesicles led to significantly increased expression of epithelial to mesenchymal transition markers and triple-negative breast cancer cell invasion compared with platelet microvesicles from healthy volunteers. Together, these data suggest that circulating PMVs in type 2 diabetes patients may contribute to the progression of triple-negative breast cancer.


Blood Platelets , Cell-Derived Microparticles , Diabetes Mellitus, Type 2 , MicroRNAs , Neoplasm Invasiveness , Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Female , Blood Platelets/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Cell-Derived Microparticles/metabolism , Cell Line, Tumor , Middle Aged , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic
5.
Front Endocrinol (Lausanne) ; 15: 1344729, 2024.
Article En | MEDLINE | ID: mdl-38904034

Cuproptosis, a recently discovered form of cell death, stems from an overabundance of copper ions infiltrating mitochondria. These ions directly engage lipoylated proteins, prompting their oligomerization and subsequent loss of iron-sulfur clusters. This sequence induces proteotoxic stress, ultimately culminating in cell death. Type 2 diabetes, a chronic metabolic disorder resulting from a complex interplay of genetic and environmental factors, has not yet been fully understood in terms of its etiology and pathogenesis. Intricately, it is linked to various modalities of cell death, including mitochondrial autophagy, apoptosis, pyroptosis, and ferroptosis. Studies have discovered impaired copper metabolism in individuals with Type 2 diabetes, hinting at a unique role for copper homeostasis in the progression of the disease. To this end, the present research aims to delineate the potential correlation between cuproptosis and Type 2 diabetes by exhaustively reviewing the existing literature. By synthesizing relevant research on cuproptosis, the paper intends to lay the groundwork for a thorough exploration of the pathogenesis of Type 2 diabetes and the development of targeted therapeutic interventions. The ultimate objective is to facilitate a deeper understanding of Type 2 diabetes and to identify novel therapeutic strategies associated with cuproptosis.


Copper , Diabetes Mellitus, Type 2 , Humans , Diabetes Mellitus, Type 2/metabolism , Copper/metabolism , Animals , Mitochondria/metabolism , Cell Death
6.
Cardiovasc Diabetol ; 23(1): 211, 2024 Jun 20.
Article En | MEDLINE | ID: mdl-38902687

BACKGROUND: Recently deorphanized G protein-coupled receptor 146 (GPR146) was shown to respond to signal from a newly identified hormone-cholesin-and to play a role in hepatic lipid metabolism. However, the importance of its biological activity in human organism remains elusive, mainly due to the lack of studies on human tissues up to this point. This study aimed to identify the cholesin receptor-associated genes and clinical factors linked with their expression in cardiovascular system and associated adipose tissues. METHODS: Right cardiac auricle, aortic wall, saphenous vein, and adipose tissue (periaortic-PAT, epicardial-EAT, thymic-TAT) samples were collected during coronary artery bypass grafting. Clinical records of the study participants were assessed for the presence of diabetes, medications taken and serum cholesterol levels. GPR146 mRNA expression in all gathered tissues was assessed with qPCR, and RNA seqencing was performed in selected tissues of 20 individuals to identify pathways associated with GPR146 expression. RESULTS: We included 46 participants [37 male, 23 with type 2 diabetes, median age 68.50 (Q1-Q3: 63.00-72.00) years, BMI 28.39 (26.06-31.49) kg/m2]. GPR146 expression in adipose tissues significantly correlated with BMI, c-peptide, total cholesterol, and LDL concentrations. Selected metabolic pathways were significantly and positively enriched in GPR146-dependent manner. GPR146-coexpressed genes contained key regulators of lipid metabolism involved in such pathways as fatty acid metabolism, tricarboxilic acid cycle and peroxisomal metabolism. Those genes correlated positively with serum concentrations of LDL, HDL, and total cholesterol. SGLT2i treatment was associated with inversion of GPR146-related signature in EAT, suggesting potential impact on cholesin-GPR146 network. CONCLUSIONS: GPR146 expression is associated with serum lipids and metabolically-relevant transcriptomic changes in EAT similar to SGLT2i-associated ones.


Adipose Tissue , Diabetes Mellitus, Type 2 , Receptors, G-Protein-Coupled , Signal Transduction , Sodium-Glucose Transporter 2 Inhibitors , Humans , Male , Middle Aged , Female , Aged , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/genetics , Adipose Tissue/metabolism , Treatment Outcome , Biomarkers/blood , Biomarkers/metabolism
7.
Clin Nutr ; 43(7): 1725-1735, 2024 Jul.
Article En | MEDLINE | ID: mdl-38843581

BACKGROUND: Aging-related type 2 diabetes (T2DM) is characterized by hyperinsulinemia, insulin resistance, and ß-cell dysfunction. However, the underlying molecular mechanisms remain to be unclear. METHODS: We conducted non-targeted metabolomics to compare human serum samples from young adults (YA), elderly adults (EA), and elderly adults with diabetes (EA + DM) of Chinese population. Adult mice and aged mice were intragastrically administered with varespladib every day for two weeks and metabolic characteristics were monitored. Serum levels of arachidonic acid, insulin, and C-peptide, as well as serum activity of secretory phospholipase A2 (sPLA2) were detected in mice. Mouse islet perfusion assays were used to assess insulin secretion ability. Phosphorylated AKT levels were measured to evaluate insulin sensitivities of peripheral tissues in mice. RESULTS: Non-targeted metabolomics analysis of human serum samples revealed differential metabolic signatures among the YA, EA, and EA + DM groups. Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis revealed significant enhancement of arachidonic acid metabolism and glycerophospholipid metabolism in the EA group compared with the YA group. Further analysis identified two metabolic fluxes that favored the accumulation of arachidonic acid in the elderly. Increased levels of arachidonic acid were also confirmed in aged mice with hyperinsulinemia and insulin resistance, together with subsequent glucose intolerance. Conversely, inhibiting the generation of arachidonic acid with varespladib, an inhibitor of sPLA2, reduced aging-associated diabetes by improving hyperinsulinemia and hepatic insulin resistance in aged mice but not in adult mice. Islet perfusion assays also showed that varespladib treatment suppressed the enhanced insulin secretion observed in aged islets. CONCLUSIONS: Collectively, our findings uncover that arachidonic acid serves as a metabolic hub in Chinese elderly population. Our results also suggest that arachidonic acid plays a fundamental role in regulating ß-cell function during aging and point to a novel therapy for aging-associated diabetes.


Aging , Arachidonic Acid , Hyperinsulinism , Insulin Resistance , Animals , Arachidonic Acid/metabolism , Arachidonic Acid/blood , Humans , Mice , Male , Aged , Diabetes Mellitus, Type 2/metabolism , Adult , Mice, Inbred C57BL , Female , Young Adult , Metabolomics , Insulin/blood , Insulin/metabolism , Middle Aged
8.
Sci Rep ; 14(1): 14361, 2024 06 21.
Article En | MEDLINE | ID: mdl-38906977

Diabetic peripheral neuropathy (DPN) is a common complication of type 2 diabetes mellitus (T2DM) that causes peripheral and autonomic nervous system dysfunction. Dysregulation of miRNAs plays a crucial role in DPN development. However, the role of miR-503-5p in DPN remains unknown. Herein, T2DM mice (db/db) were used as a DPN model in vivo, and astrocytes isolated from db/db mice were induced with high glucose levels as a DPN model in vitro. MiR-503-5p expression was analyzed using qRT-PCR. GFAP, MCP-1, and SEPT9 protein levels were analyzed using western blotting and immunofluorescence. Luciferase assays were performed to investigate the interaction between miR-503-5p and SEPT9. We found that miR-503-5p expression decreased in the spinal cord of DPN model mice and astrocytes treated with high glucose (HG). The db/db mice displayed higher body weight and blood glucose, lower mechanical withdrawal threshold and thermal withdrawal latency, and higher GFAP and MCP-1 protein levels than db/m mice. However, tail vein injection of agomiR-503-5p remarkably reversed these parameters, whereas antigomiR-503-5p enhanced them. HG markedly facilitated GFAP and MCP-1 protein expression in astrocytes, whereas miR-503-5p mimic or inhibitor transfection markedly blocked or elevated GFAP and MCP-1 protein expression, respectively, in astrocytes with HG. SEPT9 was a target of miR-503-5p. In addition, SEPT9 protein levels were found to be elevated in db/db mice and astrocytes treated with HG. Treatment with agomiR-503-5p and miR-503-5p mimic was able to reduce SEPT9 protein levels, whereas treatment with antigomiR-503-5p and miR-503-5p inhibitor led to inhibition of the protein. Furthermore, SEPT9 overexpression suppressed the depressing effect of miR-503-5p overexpression in astrocytes subjected to HG doses. In conclusion, miR-503-5p was found to alleviate peripheral neuropathy-induced neuropathic pain in T2DM mice by regulating SEPT9 expression.


Astrocytes , Diabetes Mellitus, Type 2 , Diabetic Neuropathies , MicroRNAs , Septins , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Astrocytes/metabolism , Mice , Septins/genetics , Septins/metabolism , Diabetic Neuropathies/metabolism , Diabetic Neuropathies/genetics , Diabetic Neuropathies/etiology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/genetics , Male , Neuralgia/metabolism , Neuralgia/genetics , Neuralgia/etiology , Disease Models, Animal
9.
Pharmacol Res ; 205: 107257, 2024 Jul.
Article En | MEDLINE | ID: mdl-38866264

Global aging is a tendency of the world, as is the increasing prevalence of diabetes, and the two are closely linked. In our early research, Enteromorpha prolifera oligosaccharide (EPO) possesses the excellent ability of anti-oxidative, anti-inflammatory, and anti-diabetic. We aim to further explore the deeper mechanism of how EPO delays aging and regulates glycometabolism. EPO effectively impacts crotonylation procession to enhance glucose metabolism and reduce cell senescence in aging diabetic rats. Crotonylation modification of XPO1 influences the expression of critical genes, including p53, CDK1, and CCNB1, which affect cell cycle regulation and aging. Additionally, EPO improves glucose metabolism by inhibiting the crotonylation modification of HSPA8-K126 and activating the AKT pathway. EPO promotes crotonylation of histones in intestinal cells, influencing the aging process by increasing the butyric acid-producing bacteria Ruminococcaceae. The observed enhancement in pyrimidine metabolism underscores EPO's potential role in regulating intestinal health, presenting a promising avenue for delaying aging. In summary, our findings affirm EPO as a naturally bioactive ingredient with significant potential for anti-aging and antidiabetic interventions.


Diabetes Mellitus, Type 2 , Hypoglycemic Agents , Oligosaccharides , Animals , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Oligosaccharides/pharmacology , Oligosaccharides/metabolism , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Male , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/drug therapy , Aging/metabolism , Aging/drug effects , Cellular Senescence/drug effects , Rats, Sprague-Dawley , Rats , Humans , Gastrointestinal Microbiome/drug effects
10.
ACS Nano ; 18(24): 15452-15467, 2024 Jun 18.
Article En | MEDLINE | ID: mdl-38830624

Type 2 diabetes (T2D), a prevalent metabolic disorder lacking effective treatments, is associated with lysosomal acidification dysfunction, as well as autophagic and mitochondrial impairments. Here, we report a series of biodegradable poly(butylene tetrafluorosuccinate-co-succinate) polyesters, comprising a 1,4-butanediol linker and varying ratios of tetrafluorosuccinic acid (TFSA) and succinic acid as components, to engineer lysosome-acidifying nanoparticles (NPs). The synthesized NPs are spherical with diameters of ≈100 nm and have low polydispersity and good stability. Notably, TFSA NPs, which are composed entirely of TFSA, exhibit the strongest degradation capability and superior acidifying properties. We further reveal significant downregulation of lysosomal vacuolar (H+)-ATPase subunits, which are responsible for maintaining lysosomal acidification, in human T2D pancreatic islets, INS-1 ß-cells under chronic lipotoxic conditions, and pancreatic tissues of high-fat-diet (HFD) mice. Treatment with TFSA NPs restores lysosomal acidification, autophagic function, and mitochondrial activity, thereby improving the pancreatic function in INS-1 cells and HFD mice with lipid overload. Importantly, the administration of TFSA NPs to HFD mice reduces insulin resistance and improves glucose clearance. These findings highlight the therapeutic potential of lysosome-acidifying TFSA NPs for T2D.


Insulin-Secreting Cells , Lysosomes , Nanoparticles , Lysosomes/metabolism , Lysosomes/drug effects , Animals , Nanoparticles/chemistry , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Mice , Humans , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/pathology , Male , Diet, High-Fat , Mice, Inbred C57BL , Hydrogen-Ion Concentration
11.
Diabetes ; 73(7): 1027-1031, 2024 Jul 01.
Article En | MEDLINE | ID: mdl-38900951

The glucagon-like peptide-1 receptor (GLP-1R) is a class B G protein-coupled receptor involved in the regulation of blood glucose levels and food intake. Stabilized agonists targeting GLP-1R are used in the treatment of type 2 diabetes and have recently become a breakthrough obesity therapy. Here, we revisit a classic article in Diabetes by Thorens et al. that described the cloning, sequencing, and functional expression of the human GLP-1R. The article also demonstrated that exendin4(1-39) was a full agonist of the human GLP-1R whereas exendin4(9-39) was a full antagonist. We discuss how the knowledge imparted by these studies has gone on to inform multiple strands of GLP-1R biology over the past three decades, including pharmacology, signaling, human genetics, structural biology, and chemical biology.


Diabetes Mellitus, Type 2 , Glucagon-Like Peptide-1 Receptor , Humans , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Glucagon-Like Peptide-1 Receptor/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/genetics , Exenatide/therapeutic use , Exenatide/pharmacology , Hypoglycemic Agents/therapeutic use , Hypoglycemic Agents/pharmacology , Animals , Peptides/therapeutic use
12.
Pharmazie ; 79(6): 101-108, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38877681

In this study, we hypothesized that lixisenatide (LIX) and ticagrelor (TIC) could have a protective effect against type 2 diabetes mellitus (T2DM)-induced vascular damage. Furthermore, we explored the possible additional protective effect of co-administering LIX and TIC in the treatment regimen. Methods: 50 male rats were divided into five groups, each comprising 10 rats: C (control), D (T2DM rats), D + LIX (T2DM rats treated with LIX for 4 weeks), D + TIC (T2DM rats treated with TIC for 4 weeks), and D + LIX + TIC (T2DM rats treated with LIX + TIC for 4 weeks). Results: The D group showed an increase in body weight, blood glucose, hemostatic model assessment for insulin resistance (HOMA-IR), aorta reactive oxygen species (ROS), and nuclear factor kappa B (NF-κ B), along with a reduction in serum insulin, aorta superoxide dismutase (SOD), glutathione reduced (GSH), nuclear factor erythroid-2 (NrF2), hemeoxygenase-1 (HO-1), and endothelial nitric oxide synthase (eNOS). Deterioration in the aorta histopathological condition, coupled with a noticeable impairment in vascular reactivity compared to the C group, was observed. A single administration of LIX showed a reduction in body weight, blood glucose, HOMA-IR, aorta ROS, and NF-κ B, accompanied by an increase in serum insulin, aorta SOD, GSH, NrF2, HO-1, and eNOS. Amelioration in the aorta histopathological condition and improved vascular reactivity compared to the D group were reported. Similarly, a single administration of TIC showed a reduction in aorta ROS and NF-κ B, along with an increase in aorta SOD, GSH, NrF2, HO-1, and eNOS. A slight amelioration was detected in the aorta histopathological condition, with improved vascular reactivity compared to the D group. The combined administration of LIX and TIC showed a reduction in aorta ROS and NF-κ B, along with an increase in aorta GSH, SOD, HO-1, and eNOS. This was combined with evident amelioration in the aorta histopathological condition and noticeable improvement in vascular reactivity compared to the single treatment with either LIX or TIC group. Conclusion: The present study introduces clear evidence that the administration of LIX and TIC can improve metabolic and vascular complications of T2DM through modulating eNOS and NrF2 /HO-1 signaling. The combined administration of LIX and TIC produced more significant effects than a single treatment.


Diabetes Mellitus, Experimental , NF-E2-Related Factor 2 , Nitric Oxide Synthase Type III , Peptides , Reactive Oxygen Species , Signal Transduction , Ticagrelor , Animals , Male , Nitric Oxide Synthase Type III/metabolism , Rats , Signal Transduction/drug effects , Ticagrelor/pharmacology , Ticagrelor/administration & dosage , Peptides/pharmacology , Peptides/administration & dosage , NF-E2-Related Factor 2/metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/complications , Reactive Oxygen Species/metabolism , Blood Glucose/drug effects , Insulin Resistance , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Rats, Sprague-Dawley , Heme Oxygenase (Decyclizing)/metabolism , NF-kappa B/metabolism , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/administration & dosage , Heme Oxygenase-1/metabolism , Insulin , Oxidative Stress/drug effects , Superoxide Dismutase/metabolism , Drug Synergism , Glucagon-Like Peptide-2 Receptor
13.
Biomed Res Int ; 2024: 7747599, 2024.
Article En | MEDLINE | ID: mdl-38884019

Introduction: PPIs, or proton pump inhibitors, are the most widely prescribed drugs. There is a debate regarding the relationship between long-term PPI use and the risk of type 2 diabetes mellitus (T2DM). A potential connection between T2DM and PPIs could be an elevated gastrin concentration. This study is aimed at investigating the long-term effects of PPI omeprazole (OZ) on glucose homeostasis and pancreatic gene expression profile in mice. Methods: Healthy adult male BALB/c mice were randomly divided into three equal groups (n = 10 in each one): (1) experimental mice that received OZ 20 mg/kg; (2) control mice that received 30 µl saline per os; (3) intact mice without any interventions. Mice were treated for 30 weeks. Glucose homeostasis was investigated by fasting blood glucose level, oral glucose tolerance test (GTT), insulin tolerance test (ITT), and basal insulin resistance (HOMA-IR). Serum gastrin and insulin concentration were determined by ELISA. Expressions of Sirt1, Pparg, Nfκb1 (p105), Nfe2l2, Cxcl5, Smad3, H2a.z, and H3f3b were measured by RT-PCR. Result: The ROC analysis revealed an increase in fasting blood glucose levels in OZ-treated mice in comparison with control and intact groups during the 30-week experiment. A slight but statistically significant increase in glucose tolerance and insulin sensitivity was observed in OZ-treated mice within 30 weeks of the experiment. The mice treated with OZ exhibited significant increases in serum insulin and gastrin levels, accompanied by a rise in the HOMA-IR level. These animals had a statistically significant increase in Sirt1, Pparg, and Cxcl5 mRNA expression. There were no differences in ß-cell numbers between groups. Conclusion: Long-term OZ treatment induced hypergastrin- and hyperinsulinemia and increased expression of Sirt1, Pparg, and Cxcl5 in mouse pancreatic tissues accompanied by specific changes in glucose metabolism. The mechanism of omeprazole-induced Cxcl5 mRNA expression and its association with pancreatic cancer risk should be investigated.


Blood Glucose , Gastrins , Homeostasis , Insulin Resistance , Mice, Inbred BALB C , Omeprazole , Animals , Omeprazole/pharmacology , Omeprazole/adverse effects , Gastrins/blood , Gastrins/metabolism , Male , Mice , Homeostasis/drug effects , Blood Glucose/metabolism , Insulin/metabolism , Insulin/blood , Gene Expression Regulation/drug effects , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/chemically induced , Glucose Tolerance Test , Proton Pump Inhibitors/pharmacology , Proton Pump Inhibitors/adverse effects , Glucose/metabolism
14.
Front Endocrinol (Lausanne) ; 15: 1320605, 2024.
Article En | MEDLINE | ID: mdl-38872971

Due to the Earth's rotation, the natural environment exhibits a light-dark diurnal cycle close to 24 hours. To adapt to this energy intake pattern, organisms have developed a 24-hour rhythmic diurnal cycle over long periods, known as the circadian rhythm, or biological clock. With the gradual advancement of research on the biological clock, it has become increasingly evident that disruptions in the circadian rhythm are closely associated with the occurrence of type 2 diabetes (T2D). To further understand the progress of research on T2D and the biological clock, this paper reviews the correlation between the biological clock and glucose metabolism and analyzes its potential mechanisms. Based on this, we discuss the potential factors contributing to circadian rhythm disruption and their impact on the risk of developing T2D, aiming to explore new possible intervention measures for the prevention and treatment of T2D in the future. Under the light-dark circadian rhythm, in order to adapt to this change, the human body forms an internal biological clock involving a variety of genes, proteins and other molecules. The main mechanism is the transcription-translation feedback loop centered on the CLOCK/BMAL1 heterodimer. The expression of important circadian clock genes that constitute this loop can regulate T2DM-related blood glucose traits such as glucose uptake, fat metabolism, insulin secretion/glucagon secretion and sensitivity in various peripheral tissues and organs. In addition, sleep, light, and dietary factors under circadian rhythms also affect the occurrence of T2DM.


Circadian Rhythm , Diabetes Mellitus, Type 2 , Diabetes Mellitus, Type 2/metabolism , Humans , Circadian Rhythm/physiology , Animals , Biological Clocks , Circadian Clocks/physiology , Blood Glucose/metabolism
16.
PLoS One ; 19(6): e0304676, 2024.
Article En | MEDLINE | ID: mdl-38875234

Diabetes often results in chronic ulcers that fail to heal. Effective treatment for diabetic wounds has not been achieved, although stem-cell-treatment has shown promise. Hair-follicle-associated-pluripotent (HAP)-stem-cells from bulge area of mouse hair follicle have been shown to differentiate into keratinocytes, vascular endothelial cells, smooth muscle cells, and some other types of cells. In the present study, we developed HAP-cell-sheets to determine their effects on wound healing in type-2 diabetes mellitus (db/db) C57BL/6 mouse model. Flow cytometry analysis showed cytokeratin 15 expression in 64% of cells and macrophage expression in 3.6% of cells in HAP-cell-sheets. A scratch cell migration assay in vitro showed the ability of fibroblasts to migrate and proliferate was enhanced when co-cultured with HAP-cell-sheets. To investigate in vivo effects of the HAP-cell-sheets, they were implanted into 10 mm circular full-thickness resection wounds made on the back of db/db mice. Wound closure was facilitated in the implanted group until day 16. The thickness of epithelium and granulation tissue volume at day 7 were significantly increased by the implantation. CD68 positive area and TGF-ß1 positive area were significantly increased; meanwhile, iNOS positive area was reduced at day 7 in the HAP-cell-sheets implanted group. After 21 days, CD68 positive areas in the implanted group were reduced to under the control group level, and TGF-ß1 positive area had no difference between the two groups. These observations strongly suggest that the HAP-cell-sheets implantation is efficient to facilitate early macrophage activity and to suppress inflammation level. Using immuno-double-staining against CD34 and α-SMA, we found more vigorous angiogenesis in the implanted wound tissue. The present results suggest autologous HAP-cell-sheets can be used to heal refractory diabetic ulcers and have clinical promise.


Cell Movement , Hair Follicle , Mice, Inbred C57BL , Pluripotent Stem Cells , Wound Healing , Animals , Mice , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Diabetes Mellitus, Type 2/metabolism , Male , Cell Proliferation , Transforming Growth Factor beta1/metabolism , Fibroblasts/metabolism , Granulation Tissue/pathology , Macrophages/metabolism , Diabetes Mellitus, Experimental/therapy
17.
Sci Rep ; 14(1): 12743, 2024 06 03.
Article En | MEDLINE | ID: mdl-38830911

Type 2 diabetes (T2D) is implicated as a risk factor for Alzheimer's disease (AD), the most common form of dementia. In this work, we investigated neuroinflammatory responses of primary neurons to potentially circulating, blood-brain barrier (BBB) permeable metabolites associated with AD, T2D, or both. We identified nine metabolites associated with protective or detrimental properties of AD and T2D in literature (lauric acid, asparagine, fructose, arachidonic acid, aminoadipic acid, sorbitol, retinol, tryptophan, niacinamide) and stimulated primary mouse neuron cultures with each metabolite before quantifying cytokine secretion via Luminex. We employed unsupervised clustering, inferential statistics, and partial least squares discriminant analysis to identify relationships between cytokine concentration and disease-associations of metabolites. We identified MCP-1, a cytokine associated with monocyte recruitment, as differentially abundant between neurons stimulated by metabolites associated with protective and detrimental properties of AD and T2D. We also identified IL-9, a cytokine that promotes mast cell growth, to be differentially associated with T2D. Indeed, cytokines, such as MCP-1 and IL-9, released from neurons in response to BBB-permeable metabolites associated with T2D may contribute to AD development by downstream effects of neuroinflammation.


Alzheimer Disease , Chemokine CCL2 , Diabetes Mellitus, Type 2 , Interleukin-9 , Neurons , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Diabetes Mellitus, Type 2/metabolism , Mice , Neurons/metabolism , Chemokine CCL2/metabolism , Interleukin-9/metabolism , Blood-Brain Barrier/metabolism , Cells, Cultured
18.
Gut Microbes ; 16(1): 2361491, 2024.
Article En | MEDLINE | ID: mdl-38868903

Metformin is widely used for treating type 2 diabetes mellitus (T2D). However, the efficacy of metformin monotherapy is highly variable within the human population. Understanding the potential indirect or synergistic effects of metformin on gut microbiota composition and encoded functions could potentially offer new insights into predicting treatment efficacy and designing more personalized treatments in the future. We combined targeted metabolomics and metagenomic profiling of gut microbiomes in newly diagnosed T2D patients before and after metformin therapy to identify potential pre-treatment biomarkers and functional signatures for metformin efficacy and induced changes in metformin therapy responders. Our sequencing data were largely corroborated by our metabolic profiling and identified that pre-treatment enrichment of gut microbial functions encoding purine degradation and glutamate biosynthesis was associated with good therapy response. Furthermore, we identified changes in glutamine-associated amino acid (arginine, ornithine, putrescine) metabolism that characterize differences in metformin efficacy before and after the therapy. Moreover, metformin Responders' microbiota displayed a shifted balance between bacterial lipidA synthesis and degradation as well as alterations in glutamate-dependent metabolism of N-acetyl-galactosamine and its derivatives (e.g. CMP-pseudaminate) which suggest potential modulation of bacterial cell walls and human gut barrier, thus mediating changes in microbiome composition. Together, our data suggest that glutamine and associated amino acid metabolism as well as purine degradation products may potentially condition metformin activity via its multiple effects on microbiome functional composition and therefore serve as important biomarkers for predicting metformin efficacy.


Amino Acids , Bacteria , Biomarkers , Diabetes Mellitus, Type 2 , Gastrointestinal Microbiome , Hypoglycemic Agents , Metformin , Purines , Humans , Metformin/pharmacology , Metformin/therapeutic use , Gastrointestinal Microbiome/drug effects , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/microbiology , Diabetes Mellitus, Type 2/metabolism , Amino Acids/metabolism , Male , Middle Aged , Female , Purines/metabolism , Bacteria/classification , Bacteria/metabolism , Bacteria/genetics , Bacteria/drug effects , Bacteria/isolation & purification , Biomarkers/metabolism , Hypoglycemic Agents/therapeutic use , Hypoglycemic Agents/pharmacology , Aged , Adult , Treatment Outcome , Metabolomics
19.
Front Endocrinol (Lausanne) ; 15: 1332237, 2024.
Article En | MEDLINE | ID: mdl-38872972

Type 2 diabetes mellitus has a worldwide prevalence of 10.5% in the adult population (20-79 years), and by 2045, the prevalence is expected to keep rising to one in eight adults living with diabetes. Mild cognitive impairment has a global prevalence of 19.7% in adults aged 50 years. Both conditions have shown a concerning increase in prevalence rates over the past 10 years, highlighting a growing public health challenge. Future forecasts indicate that the prevalence of dementia (no estimations done for individuals with mild cognitive impairment) is expected to nearly triple by 2050. Type 2 diabetes mellitus is a risk factor for the development of cognitive impairment, and such impairment increase the likelihood of poor glycemic/metabolic control. High phytate intake has been shown to be a protective factor against the development of cognitive impairment in observational studies. Diary phytate intake might reduce the micro- and macrovascular complications of patients with type 2 diabetes mellitus through different mechanisms. We describe the protocol of the first trial (the PHYND trial) that evaluate the effect of daily phytate supplementation over 56 weeks with a two-arm double-blind placebo-controlled study on the progression of mild cognitive impairment, cerebral iron deposition, and retinal involvement in patients with type 2 diabetes mellitus. Our hypothesis proposes that phytate, by inhibiting advanced glycation end product formation and chelating transition metals, will improve cognitive function and attenuate the progression from Mild Cognitive Impairment to dementia in individuals with type 2 diabetes mellitus and mild cognitive impairment. Additionally, we predict that phytate will reduce iron accumulation in the central nervous system, mitigate neurodegenerative changes in both the central nervous system and retina, and induce alterations in biochemical markers associated with neurodegeneration.


Brain , Cognitive Dysfunction , Diabetes Mellitus, Type 2 , Diabetic Retinopathy , Dietary Supplements , Disease Progression , Iron , Phytic Acid , Humans , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/complications , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/etiology , Cognitive Dysfunction/prevention & control , Double-Blind Method , Phytic Acid/administration & dosage , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/drug therapy , Brain/metabolism , Brain/drug effects , Iron/metabolism , Iron/administration & dosage , Female , Male , Middle Aged , Aged , Adult , Administration, Oral
20.
Cell Metab ; 36(6): 1169-1171, 2024 Jun 04.
Article En | MEDLINE | ID: mdl-38838638

In this issue of Cell Metabolism, Fang et al.1 report a novel pH-sensitive cellular signaling mechanism involving the transcription factor SMAD5 that regulates the vesicular secretion of insulin from pancreatic ß cells in response to dietary challenges. Dysregulation of this pathway may contribute to metabolic disorders such as type 2 diabetes mellitus.


Insulin Secretion , Insulin-Secreting Cells , Insulin , Signal Transduction , Smad5 Protein , Insulin/metabolism , Animals , Insulin-Secreting Cells/metabolism , Smad5 Protein/metabolism , Humans , Diabetes Mellitus, Type 2/metabolism , Mice , Hydrogen-Ion Concentration
...