Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 186
Filter
1.
Sci Rep ; 14(1): 10311, 2024 05 05.
Article in English | MEDLINE | ID: mdl-38705920

ABSTRACT

Diabetic individuals with diabetic cardiomyopathy (DbCM) present with abnormal myocardial structure and function. DbCM cannot be accurately diagnosed due to the lack of suitable diagnostic biomarkers. In this study, 171 eligible participants were divided into a healthy control (HC), type 2 diabetes mellitus (T2DM) patients without DbCM (T2DM), or DbCM group. Serum fibrinogen-like protein 1 (FGL-1) and other biochemical parameters were determined for all participants. Serum FGL-1 levels were significantly higher in patients with DbCM compared with those in the T2DM group and HCs. Serum FGL-1 levels were negatively correlated with left ventricular fractional shortening and left ventricular ejection fraction (LVEF) and positively correlated with left ventricular mass index in patients with DbCM after adjusting for age, sex and body mass index. Interaction of serum FGL-1 and triglyceride levels on LVEF was noted in patients with DbCM. A composite marker including serum FGL-1 and triglycerides could differentiate patients with DbCM from those with T2DM and HCs with an area under the curve of 0.773 and 0.789, respectively. Composite marker levels were negatively correlated with N-terminal B-type natriuretic peptide levels in patients with DbCM. Circulating FGL-1 may therefore be a valuable index reflecting cardiac functions in DbCM and to diagnose DbCM.


Subject(s)
Biomarkers , Diabetes Mellitus, Type 2 , Diabetic Cardiomyopathies , Fibrinogen , Humans , Male , Female , Fibrinogen/metabolism , Fibrinogen/analysis , Diabetic Cardiomyopathies/blood , Diabetic Cardiomyopathies/diagnosis , Biomarkers/blood , Middle Aged , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Aged , Ventricular Function, Left , Case-Control Studies , Stroke Volume , Triglycerides/blood
2.
Med Arch ; 76(2): 135-139, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35774047

ABSTRACT

Background: Angiogenesis in diabetic patients is often caused by hyperglycemia induced by hypoxia. Objective: The aim of this study was to analyze the serum level of Hypoxia Inducible Factor -1α (HIF-1α) and Vascular Endothelial Growth Factor (VEGF) between March until Desember 2020. Methods: This is a cross-sectional analytic methods, 135 patients with Type 2 Diabetes 48 samples with Microvascular complication and 87 samples with non-microvascular complication were recruited from the various primary health care centers in Medan city and surrounding areas in North Sumatera. VEGF levels and HIF-1α tested were done with ELISA methods in the laboratory of Medical Faculty, Universitas Sumatera Utara. Statistical analysis was performed using the IBM SPSS Statistics version 24. The significance level was set up to 0.005. Results: The median HIF-1 levels in patients with microvascular complications were lower than those without microvascular complications, with a range of HIF-1α values in non-complicated samples (0.02-13.96) ng/ml and a range of HIF-1α values in vascular complications (0.52- 8.87) mg/dL. There was a significant difference in HIF-1α levels in patients with Type-2 DM with complications compared to those without complications (p<0.05). Median VEGF levels were higher in complicated Type-2 DM. There was no difference in VEGF levels in patients with Type-2 DM with complications compared to those without complications (p > 0.005). Conclusion: HIF-1α and VEGF levels showed the development in vascularity. With the higher level of HIF-1α, an increase in VEGF levels were found, indicating the angiogenesis is occurring. Although complications have not yet occurred, it is predicted that high VEGF values will cause vascular complications in the future.


Subject(s)
Diabetes Mellitus, Type 2 , Diabetic Cardiomyopathies , Hypoxia-Inducible Factor 1, alpha Subunit , Vascular Endothelial Growth Factor A , Cross-Sectional Studies , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetic Cardiomyopathies/blood , Diabetic Cardiomyopathies/pathology , Humans , Hypoxia/blood , Hypoxia/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/blood , Neovascularization, Pathologic/blood , Neovascularization, Pathologic/pathology , Vascular Endothelial Growth Factor A/blood
3.
JAMA Netw Open ; 5(1): e2146124, 2022 01 04.
Article in English | MEDLINE | ID: mdl-35099545

ABSTRACT

Importance: The associations of serum folate and vitamin B12 levels with cardiovascular outcomes among patients with type 2 diabetes (T2D) remain unclear. Objective: To investigate the associations of serum folate and vitamin B12 levels with risk of cardiovascular disease (CVD) mortality among individuals with T2D. Design, Setting, and Participants: This prospective cohort study included 8067 patients with T2D who participated in the National Health and Nutrition Examination Survey (NHANES) from 1999 through 2014 and NHANES III (1988-1994). American Diabetes Association criteria were used to define T2D. Data were analyzed between October 1, 2020, and April 1, 2021. Exposures: Serum folate and vitamin B12 levels. Main Outcomes and Measures: Multivariable Cox proportional hazards regression models were used to compute hazard ratios and 95% CIs for the associations of serum folate and vitamin B12 levels with risks of CVD and all-cause mortality. Two multivariable models were constructed. Restricted cubic spline analyses were used to examine the nonlinear association of serum folate levels and vitamin B12 levels with CVD mortality, and nonlinearity was assessed using the likelihood ratio test. Results: This cohort study included data from 7700 participants in the folate analysis (mean [SE] age, 57.8 [0.3] years; 3882 men [weighted, 50.5%]; median serum folate level, 12.1 ng/mL [IQR, 7.1-19.5 ng/mL]) and 4860 participants for the vitamin B12 analysis (mean [SE] age, 57.8 [0.3] years; 2390 men [weighted, 50.7%]; median serum vitamin B12 level, 506.1 pg/mL [IQR, 369.1-703.5 pg/mL]). During 72 031 person-years of follow-up, 799 CVD deaths were documented for the folate analysis, and during 43 855 person-years of follow-up, 467 CVD deaths were reported for the vitamin B12 analysis. Nonlinear associations were observed for serum levels of folate (P = .04 for nonlinearity) and vitamin B12 (P = .04 for nonlinearity) with risk of CVD mortality among patients with T2D. Compared with participants in the second quartile of serum folate levels (7.1-12.1 ng/mL), the hazard ratios for CVD mortality were 1.43 (95% CI, 1.04-1.98) for participants in the lowest serum folate level quartile (<7.1 ng/mL) and 1.03 (95% CI, 0.74-1.44) for participants in the highest quartile (≥19.5 ng/mL). In addition, compared with participants in the second quartile of serum vitamin B12 levels (369.1-506.0 pg/mL), the hazard ratios for CVD mortality were 1.74 (95% CI, 1.20-2.52) for participants in the lowest quartile (<369.1 pg/mL) and 2.32 (95% CI, 1.60-3.35) for participants in the highest quartile (≥703.5 pg/mL). Similar patterns of association were observed for all-cause mortality (nonlinearity: P = .01 for folate and P = .02 for vitamin B12). Conclusions and Relevance: This cohort study found that both low and high serum levels of vitamin B12 as well as low serum levels of folate were significantly associated with higher risk of CVD mortality among individuals with T2D.


Subject(s)
Cardiovascular Diseases/mortality , Diabetes Mellitus, Type 2/blood , Diabetic Cardiomyopathies/mortality , Folic Acid/blood , Vitamin B 12/blood , Cardiovascular Diseases/blood , Cardiovascular Diseases/etiology , Diabetes Mellitus, Type 2/complications , Diabetic Cardiomyopathies/blood , Diabetic Cardiomyopathies/etiology , Female , Humans , Male , Middle Aged , Nutrition Surveys , Nutritional Status , Proportional Hazards Models , Prospective Studies
4.
Cardiovasc Diabetol ; 21(1): 5, 2022 01 06.
Article in English | MEDLINE | ID: mdl-34991588

ABSTRACT

BACKGROUND: Systemic inflammatory processes plausibly contribute to the development of cardiovascular complications, causing increased morbidity and mortality in type 2 diabetes. Circulating inflammatory markers, i.e., interleukin (IL)-6 and tumour necrosis factor-α, are associated with neurocardiac measures. We examined a broad panel of various inflammatory and inflammation-related serum markers to obtain more detailed insight into the possible neuro-immune interaction between cardiovascular regulation and systemic level of inflammation. METHODS: Serum samples from 100 participants with type 2 diabetes were analysed. Heart rate variability, cardiovascular autonomic reflex tests, and cardiac vagal tone tests were performed based on electrocardiographic readings. Data regarding covariates (demographic-, diabetes-, and cardiovascular risk factors) were registered. RESULTS: Increased serum levels of IL-12/IL-23p40 (p < 0.01) and intercellular adhesion molecule (ICAM)-1 (p < 0.007) were associated with diminished heart rate variability measures. After all adjustments, the associations between IL-12/23p40, SDANN and VLF persisted (p = 0.001). Additionally, serum levels of vascular endothelial growth factor (VEGF)-C were associated with response to standing (p = 0.005). DISCUSSION: The few but robust associations between neurocardiac regulation and serum markers found in this study suggest systemic changes in proinflammatory, endothelial, and lymphatic function, which collectively impacts the systemic cardiovascular function. Our results warrant further exploration of IL-12/IL-23p40, ICAM-1, and VEGF-C as possible cardiovascular biomarkers in T2D that may support future decisions regarding treatment strategies for improved patient care.


Subject(s)
Diabetes Mellitus, Type 2/blood , Diabetic Cardiomyopathies/blood , Heart Rate , Inflammation Mediators/blood , Interleukin-12 Subunit p40/blood , Adult , Aged , Aged, 80 and over , Biomarkers/blood , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/diagnosis , Diabetic Cardiomyopathies/diagnosis , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/physiopathology , Electrocardiography , Female , Humans , Intercellular Adhesion Molecule-1/blood , Male , Middle Aged , Predictive Value of Tests , Prognosis , Up-Regulation , Vascular Endothelial Growth Factor C/blood
5.
Metabolism ; 126: 154918, 2022 01.
Article in English | MEDLINE | ID: mdl-34699838

ABSTRACT

Type 2 diabetes (T2D) is one of the most common causes of chronic kidney disease (CKD) and cardiovascular (CV) disease. Until recently, glycemic and BP control were the cornerstones for preventing progression of CKD and CV disease associated with T2D. However, there has been a paradigm shift in treatment since the publication of the first clinical trial demonstrating benefits of sodium glucose cotransporter 2 (SGLT2) inhibitors in 2015. SGLT2 inhibitors have been shown to reduce the risk of major adverse CV events and progression of kidney disease in the setting of T2D. However, the elucidation of mechanisms of underlying these clinical benefits is the subject of ongoing investigation. Experimental studies have shown that SGLT2 inhibitors have diverse pleiotropic effects such as modulation of neurohormones such as the renin-angiotensin-aldosterone system, increasing hematocrit, altering energy substrate use, and attenuating systemic inflammation and oxidative stress, all of which have been implicated in the CV and kidney protective effects of SGLT2 inhibitors. In this review, we highlight biomarkers linked with diabetic kidney disease and heart failure and discuss how SGLT2 inhibitor-associated changes potentially mediate the cardiorenal protection observed with these therapies.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Diabetic Cardiomyopathies/prevention & control , Diabetic Nephropathies/prevention & control , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Biomarkers/blood , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Diabetic Cardiomyopathies/blood , Diabetic Nephropathies/blood , Humans , Oxidative Stress/physiology , Reactive Oxygen Species/blood
6.
Nutrients ; 13(12)2021 Nov 28.
Article in English | MEDLINE | ID: mdl-34959854

ABSTRACT

Vitamin D (VD) insufficiency is common among patients with diabetes in French Guiana. The study aimed to evaluate the prevalence of VD deficiency in the different type of diabetes encountered and to analyze the relationship between VD deficiency and diabetes complications. METHODS: An observational study was conducted between May 2019 and May 2020 in French Guiana, based on data from the CODIAM study (Diabetes Cohort in French Amazonia), describing the characteristics of patients with diabetes mellitus. Among 600 patients enrolled with diabetes, 361 had an available VD assay. RESULTS: The mean 25(OH)VD (hydroxycalciferol) level was 27.9 ng/mL. The level of VD was inversely proportional to the HbA1c (glycated hemoglobin) level. Patients with angina pectoris had a greater proportion of deficiencies VD < 20 ng/mL than those without angina. By contrast, patients with retinopathy had higher vitamin D concentrations than those without retinopathy. There was no association between vitamin D and arteriopathy, stroke, nephropathy and polyneuropathy. VD deficiency was more frequent in women, and in patients with a high school education. CONCLUSION: The prevalence of VD deficiency was high in patients with diabetes in French Guiana, emphasizing the importance of VD supplementation.


Subject(s)
Diabetes Complications/blood , Diabetes Mellitus/blood , Vitamin D Deficiency/epidemiology , Adult , Angina Pectoris/blood , Angina Pectoris/epidemiology , Angina Pectoris/etiology , Cohort Studies , Diabetes Complications/complications , Diabetic Cardiomyopathies/blood , Diabetic Cardiomyopathies/epidemiology , Diabetic Cardiomyopathies/etiology , Diabetic Retinopathy/blood , Diabetic Retinopathy/epidemiology , Diabetic Retinopathy/etiology , Female , French Guiana/epidemiology , Glycated Hemoglobin/analysis , Humans , Male , Middle Aged , Prevalence , Vitamin D/analogs & derivatives , Vitamin D/blood , Vitamin D Deficiency/etiology
7.
Molecules ; 26(23)2021 Nov 30.
Article in English | MEDLINE | ID: mdl-34885867

ABSTRACT

Metabolic disorders often lead to cardiac complications. Metabolic deregulations during diabetic conditions are linked to mitochondrial dysfunctions, which are the key contributing factors in cardiac hypertrophy. However, the underlying mechanisms involved in diabetes-induced cardiac hypertrophy are poorly understood. In the current study, we initially established a diabetic rat model by alloxan-administration, which was validated by peripheral glucose measurement. Diabetic rats displayed myocardial stiffness and fibrosis, changes in heart weight/body weight, heart weight/tibia length ratios, and enhanced size of myocytes, which altogether demonstrated the establishment of diabetic cardiac hypertrophy (DCH). Furthermore, we examined the expression of genes associated with mitochondrial signaling impairment. Our data show that the expression of PGC-1α, cytochrome c, MFN-2, and Drp-1 was deregulated. Mitochondrial-signaling impairment was further validated by redox-system dysregulation, which showed a significant increase in ROS and thiobarbituric acid reactive substances, both in serum and heart tissue, whereas the superoxide dismutase, catalase, and glutathione levels were decreased. Additionally, the expression levels of pro-apoptotic gene PUMA and stress marker GATA-4 genes were elevated, whereas ARC, PPARα, and Bcl-2 expression levels were decreased in the heart tissues of diabetic rats. Importantly, these alloxan-induced impairments were rescued by N-acetyl cysteine, ascorbic acid, and selenium treatment. This was demonstrated by the amelioration of myocardial stiffness, fibrosis, mitochondrial gene expression, lipid profile, restoration of myocyte size, reduced oxidative stress, and the activation of enzymes associated with antioxidant activities. Altogether, these data indicate that the improvement of mitochondrial dysfunction by protective agents such as N-acetyl cysteine, selenium, and ascorbic acid could rescue diabetes-associated cardiac complications, including DCH.


Subject(s)
Acetylcysteine/therapeutic use , Ascorbic Acid/therapeutic use , Cardiomegaly/drug therapy , Diabetic Cardiomyopathies/drug therapy , Mitochondria, Heart/metabolism , Selenium/therapeutic use , Acetylcysteine/pharmacology , Animals , Antioxidants/pharmacology , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Biomarkers/blood , Blood Glucose/metabolism , Body Weight/drug effects , Calcium/blood , Cardiomegaly/blood , Cardiomegaly/complications , Cardiomegaly/pathology , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use , Cytochromes c/metabolism , Diabetic Cardiomyopathies/blood , Diabetic Cardiomyopathies/complications , Diabetic Cardiomyopathies/pathology , Disease Models, Animal , Down-Regulation , GATA4 Transcription Factor/metabolism , Lipid Peroxidation/drug effects , Lipids/blood , Mitochondria, Heart/drug effects , Myocardium/pathology , Oxidation-Reduction , Oxidative Stress , PPAR alpha/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Selenium/pharmacology
8.
Cardiovasc Diabetol ; 20(1): 163, 2021 08 09.
Article in English | MEDLINE | ID: mdl-34372849

ABSTRACT

BACKGROUND: Patients with diabetes mellitus (DM) are at increased risk of developing heart failure (HF). The "Heart OMics in AGEing" (HOMAGE) trial suggested that spironolactone had beneficial effect on fibrosis and cardiac remodelling in an at risk population, potentially slowing the progression towards HF. We compared the proteomic profile of patients with and without diabetes among patients at risk for HF in the HOMAGE trial. METHODS: Protein biomarkers (n = 276) from the Olink®Proseek-Multiplex cardiovascular and inflammation panels were measured in plasma collected at baseline and 9 months (or last visit) from HOMAGE trial participants including 217 patients with, and 310 without, diabetes. RESULTS: Twenty-one biomarkers were increased and five decreased in patients with diabetes compared to non-diabetics at baseline. The markers clustered mainly within inflammatory and proteolytic pathways, with granulin as the key-hub, as revealed by knowledge-induced network and subsequent gene enrichment analysis. Treatment with spironolactone in diabetic patients did not lead to large changes in biomarkers. The effects of spironolactone on NTproBNP, fibrosis biomarkers and echocardiographic measures of diastolic function were similar in patients with and without diabetes (all interaction analyses p > 0.05). CONCLUSIONS: Amongst patients at risk for HF, those with diabetes have higher plasma concentrations of proteins involved in inflammation and proteolysis. Diabetes does not influence the effects of spironolactone on the proteomic profile, and spironolactone produced anti-fibrotic, anti-remodelling, blood pressure and natriuretic peptide lowering effects regardless of diabetes status.  Trial registration NCT02556450.


Subject(s)
Blood Proteins/analysis , Diabetes Mellitus/blood , Diabetic Cardiomyopathies/blood , Heart Failure/blood , Proteome , Proteomics , Aged , Biomarkers/blood , Diabetes Mellitus/diagnosis , Diabetes Mellitus/drug therapy , Diabetic Cardiomyopathies/diagnosis , Diabetic Cardiomyopathies/drug therapy , Female , Heart Failure/diagnosis , Heart Failure/drug therapy , Humans , Male , Mineralocorticoid Receptor Antagonists/therapeutic use , Predictive Value of Tests , Prospective Studies , Risk Assessment , Risk Factors , Spironolactone/therapeutic use , Time Factors , Treatment Outcome
9.
Cardiovasc Diabetol ; 20(1): 161, 2021 08 03.
Article in English | MEDLINE | ID: mdl-34344360

ABSTRACT

BACKGROUND: Whether differences in circulating long chain acylcarnitines (LCAC) are seen in heart failure (HF) patients with and without diabetes mellitus (DM), and whether these biomarkers report on exercise capacity and clinical outcomes, remains unknown. The objective of the current study was to use metabolomic profiling to identify biomarkers that report on exercise capacity, clinical outcomes, and differential response to exercise in HF patients with and without DM. METHODS: Targeted mass spectrometry was used to quantify metabolites in plasma from participants in the heart failure: a controlled trial investigating outcomes of exercise training (HF-ACTION) trial. Principal components analysis was used to identify 12 uncorrelated factors. The association between metabolite factors, diabetes status, exercise capacity, and time to the primary clinical outcome of all-cause mortality or all-cause hospitalization was assessed. RESULTS: A total of 664 participants were included: 359 (54%) with DM. LCAC factor levels were associated with baseline exercise capacity as measured by peak oxygen consumption (beta 0.86, p = 2 × 10-7, and were differentially associated in participants with and without DM (beta 1.58, p = 8 × 10-8 vs. 0.67, p = 9 × 10-4, respectively; p value for interaction = 0.012). LCAC levels changed to a lesser extent in participants with DM after exercise (mean ∆ 0.09, p = 0.24) than in those without DM (mean ∆ 0.16, p = 0.08). In univariate and multivariate modeling, LCAC factor levels were associated with time to the primary outcome (multivariate HR 0.80, p = 2.74 × 10-8), and were more strongly linked to outcomes in diabetic participants (HR 0.64, p = 3.21 × 10-9 v. HR 0.90, p = 0.104, p value for interaction = 0.001). When analysis was performed at the level of individual metabolites, C16, C16:1, C18, and C18:1 had the greatest associations with both exercise capacity and outcomes, with higher levels associated with worse outcomes. Similar associations with time to the primary clinical outcome were not found in a control group of patients without HF from the CATHeterization GENetics (CATHGEN) study. CONCLUSIONS: LCAC biomarkers are associated with exercise status and clinical outcomes differentially in HF patients with and without DM. Impaired fatty acid substrate utilization and mitochondrial dysfunction both at the level of the skeletal muscle and the myocardium may explain the decreased exercise capacity, attenuated response to exercise training, and poor clinical outcomes seen in patients with HF and DM. Trial Registration clinicaltrials.gov Identifier: NCT00047437.


Subject(s)
Carnitine/analogs & derivatives , Diabetic Cardiomyopathies/blood , Exercise Tolerance , Heart Failure/blood , Aged , Biomarkers/blood , Carnitine/blood , Clinical Trials as Topic , Diabetic Cardiomyopathies/diagnosis , Diabetic Cardiomyopathies/mortality , Diabetic Cardiomyopathies/physiopathology , Female , Health Status , Heart Failure/diagnosis , Heart Failure/mortality , Heart Failure/physiopathology , Hospitalization , Humans , Male , Metabolome , Metabolomics , Middle Aged , Prognosis , Risk Assessment , Risk Factors , Tandem Mass Spectrometry , Time Factors
10.
J Mol Cell Cardiol ; 160: 27-41, 2021 11.
Article in English | MEDLINE | ID: mdl-34224725

ABSTRACT

Irisin, the cleaved form of the fibronectin type III domain containing 5 (FNDC5) protein, is involved in metabolism and inflammation. Recent findings indicated that irisin participated in cardiovascular physiology and pathology. In this study, we investigated the effects of FNDC5/irisin on diabetic cardiomyopathy (DCM) in type 2 diabetic db/db mice. Downregulation of myocardial FNDC5/irisin protein expression and plasma irisin levels was observed in db/db mice compared to db/+ controls. Moreover, echocardiography revealed that db/db mice exhibited normal cardiac systolic function and impaired diastolic function. Adverse structural remodeling, including cardiomyocyte apoptosis, myocardial fibrosis, and cardiac hypertrophy were observed in the hearts of db/db mice. Sixteen-week-old db/db mice were intramyocardially injected with adenovirus encoding FNDC5 or treated with recombinant human irisin via a peritoneal implant osmotic pump for 4 weeks. Both overexpression of myocardial FNDC5 and exogenous irisin administration attenuated diastolic dysfunction and cardiac structural remodeling in db/db mice. Results from in vitro studies revealed that FNDC5/irisin protein expression was decreased in high glucose (HG)/high fat (HF)-treated cardiomyocytes. Increased levels of inducible nitric oxide synthase (iNOS), NADPH oxidase 2 (NOX2), 3-nitrotyrosine (3-NT), reactive oxygen species (ROS), and peroxynitrite (ONOO-) in HG/HF-treated H9C2 cells provided evidence of oxidative/nitrosative stress, which was alleviated by treatment with FNDC5/irisin. Moreover, the mitochondria membrane potential (ΔΨm) was decreased and cytochrome C was released from mitochondria with increased levels of cleaved caspase-3 in HG/HF-treated H9C2 cells, indicating the presence of mitochondria-dependent apoptosis, which was partially reversed by FNDC5/irisin treatment. Mechanistic studies showed that activation of integrin αVß5-AKT signaling and attenuation of oxidative/nitrosative stress were responsible for the cardioprotective effects of FNDC5/irisin. Therefore, FNDC5/irisin mediates cardioprotection in DCM by inhibiting myocardial apoptosis, myocardial fibrosis, and cardiac hypertrophy. These findings implicate that FNDC5/irisin as a potential therapeutic intervention for DCM, especially in type 2 diabetes mellitus (T2DM).


Subject(s)
Cardiotonic Agents/administration & dosage , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/drug therapy , Diabetic Cardiomyopathies/blood , Diabetic Cardiomyopathies/drug therapy , Fibronectins/administration & dosage , Nitrosative Stress/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Vitronectin/metabolism , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Cardiomegaly/prevention & control , Cardiotonic Agents/blood , Disease Models, Animal , Fibronectins/blood , Fibronectins/genetics , Male , Mice , Mitochondria/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Recombinant Proteins/administration & dosage , Treatment Outcome , Ventricular Remodeling/drug effects
11.
Clin Nutr ; 40(6): 3708-3717, 2021 06.
Article in English | MEDLINE | ID: mdl-34130016

ABSTRACT

BACKGROUND: Hyperglycemia can induce the heart to enter an oxygen-restricted environment, which results in diabetic cardiomyopathy (DCM). Microbiota-derived short-chain fatty acids (SCFAs) affect O2 consumption and play crucial roles in modulating metabolic and cardiovascular health. The epigenetic regulation of the hypoxia-inducible factor 3A (HIF3A) gene is implicated in oxidative metabolism in the pathogenesis of diabetes. Identifying the associations between plasma SCFA levels and intronic DNA methylation of HIF3A may reveal useful predictors or provide insights into the disease processes of DCM. METHODS: In this cross-sectional study, we analyzed plasma SCFA levels, HIF3A expression, and CpG methylation of HIF3A intron 1 in peripheral blood from patients with type 2 diabetes presenting with (n = 92) and without (n = 105) cardiomyopathy. RESULTS: Plasma butyric acid levels and HIF3A mRNA expression in peripheral blood were decreased in DCM patients, whereas 3 CpGs in HIF3A intron 1 (CpG 6, CpG 7 and CpG 11) were highly methylated in DCM patients. Interestingly, butyric acid levels positively correlated with HIF3A levels, while a negative association was identified between butyric acid levels and the methylation rates of HIF3A intron 1 at CpG 6. Butyric acid levels also correlated with several clinical/echocardiographic factors in DCM patients. Additionally, the combination of plasma butyric acid levels and HIF3A intron 1 methylation at CpG 6 discriminated DCM patients from type2 diabetes mellitus (T2DM) patients. CONCLUSIONS: The novel associations between plasma butyric acid levels and HIF3A intron 1 methylation at CpG 6 may highlight an underlying mechanism by which the "microbial-myocardial" axis and host-microbe interactions may participate in the pathogenesis of DCM.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Butyric Acid/metabolism , CpG Islands , DNA Methylation , Diabetic Cardiomyopathies/metabolism , Fatty Acids, Volatile/metabolism , Introns , Repressor Proteins/genetics , Adult , Aged , Biomarkers , Butyric Acid/blood , Cross-Sectional Studies , Diabetic Cardiomyopathies/blood , Fatty Acids, Volatile/blood , Female , Humans , Male , Microbiota , Middle Aged
12.
PLoS One ; 16(4): e0250773, 2021.
Article in English | MEDLINE | ID: mdl-33909697

ABSTRACT

Diabetic Cardiomyopathy (DCM) is characterized by myocardial dysfunction caused by diabetes mellitus. After-effects of diabetic cardiomyopathy are far more lethal than non-diabetic cardiomyopathy. More than 300 million people suffer from diabetes and cardiovascular disorder which is expected to be elevated to an alarming figure of 450 million by 2030. Recent studies suggested that miRNA plays important role in the onset of diabetic cardiomyopathy. This study was designed to identify the miRNA that is responsible for the onset of diabetic cardiomyopathy using in silico and in vitro approaches. In this study, to identify the miRNA responsible for the onset of diabetic cardiomyopathy, in silico analysis was done to predict the role of these circulating miRNAs in type 2 diabetic cardiomyopathy. Shared miRNAs that are present in both diseases were selected for further analysis. Total RNA and miRNA were extracted from blood samples taken from type 2 diabetic patients as well as healthy controls to analyze the expression of important genes like AKT, VEGF, IGF, FGF1, ANGPT2 using Real-time PCR. The expression of ANGPT2 was up-regulated and AKT, VEGF, IGF, FGF1 were down-regulated in DCM patients as compared to healthy controls. The miRNA expression of miR-17 was up-regulated and miR-24, miR-150, miR-199a, miR-214, and miR-320a were down-regulated in the DCM patients as compared to healthy controls. This shows that dysregulation of target genes and miRNA may contribute towards the pathogenesis of DCM and more studies should be conducted to elucidate the role of circulating miRNAs to use them as therapeutic and diagnostic options.


Subject(s)
Circulating MicroRNA/genetics , Diabetes Mellitus, Type 2/genetics , Diabetic Cardiomyopathies/genetics , Gene Regulatory Networks , Adult , Aged , Case-Control Studies , Computer Simulation , Diabetes Mellitus, Type 2/blood , Diabetic Cardiomyopathies/blood , Female , Gene Expression Profiling , Gene Expression Regulation , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Pakistan
13.
Int J Mol Sci ; 22(6)2021 Mar 12.
Article in English | MEDLINE | ID: mdl-33809279

ABSTRACT

Skeletal muscle damage is a common clinical manifestation of systemic sclerosis (SSc). C-X-C chemokine ligand 10 (CXCL10) is involved in myopathy and cardiomyopathy development and is associated with a more severe SSc prognosis. Interestingly, the phosphodiesterase type 5 inhibitor (PDE5i) sildenafil reduces CXCL10 sera levels of patients with diabetic cardiomyopathy and in cardiomyocytes. Here, we analyzed the levels of CXCL10 in the sera of 116 SSc vs. 35 healthy subjects and explored differences in 17 SSc patients on stable treatment with sildenafil. CXCL10 sera levels were three-fold higher in SSc vs. healthy controls, independent of subset and antibody positivity. Sildenafil treatment was associated with lower CXCL10 sera levels. Serum CXCL10 strongly correlated with the clinical severity of muscle involvement and with creatine kinase (CK) serum concentration, suggesting a potential involvement in muscle damage in SSc. In vitro, sildenafil dose-dependently reduced CXCL10 release by activated myocytes and impaired cytokine-induced Signal transducer and activator of transcription 1 (STAT1), Nuclear factor-κB (NFκB) and c-Jun N-terminal kinase (JNK) phosphorylation. This was also seen in cardiomyocytes. Sildenafil-induced CXCL10 inhibition at the systemic and human muscle cell level supports the hypothesis that PDE5i could be a potential therapeutic therapy to prevent and treat muscle damage in SSc.


Subject(s)
Chemokine CXCL10/genetics , Cyclic Nucleotide Phosphodiesterases, Type 5/genetics , Diabetic Cardiomyopathies/drug therapy , Scleroderma, Systemic/drug therapy , Sildenafil Citrate/pharmacology , Diabetic Cardiomyopathies/blood , Diabetic Cardiomyopathies/pathology , Female , Humans , JNK Mitogen-Activated Protein Kinases/genetics , Male , Middle Aged , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Myocytes, Cardiac/drug effects , NF-kappa B , Phosphodiesterase 5 Inhibitors/pharmacology , STAT1 Transcription Factor/genetics , Scleroderma, Systemic/blood , Scleroderma, Systemic/pathology
14.
Genome Med ; 13(1): 29, 2021 02 19.
Article in English | MEDLINE | ID: mdl-33608049

ABSTRACT

BACKGROUND: The clinical utility of personal genomic information in identifying individuals at increased risks for dyslipidemia and cardiovascular diseases remains unclear. METHODS: We used data from Biobank Japan (n = 70,657-128,305) and developed novel East Asian-specific genome-wide polygenic risk scores (PRSs) for four lipid traits. We validated (n = 4271) and subsequently tested associations of these scores with 3-year lipid changes in adolescents (n = 620), carotid intima-media thickness (cIMT) in adult women (n = 781), dyslipidemia (n = 7723), and coronary heart disease (CHD) (n = 2374 cases and 6246 controls) in type 2 diabetes (T2D) patients. RESULTS: Our PRSs aggregating 84-549 genetic variants (0.251 < correlation coefficients (r) < 0.272) had comparably stronger association with lipid variations than the typical PRSs derived based on the genome-wide significant variants (0.089 < r < 0.240). Our PRSs were robustly associated with their corresponding lipid levels (7.5 × 10- 103 < P < 1.3 × 10- 75) and 3-year lipid changes (1.4 × 10- 6 < P < 0.0130) which started to emerge in childhood and adolescence. With the adjustments for principal components (PCs), sex, age, and body mass index, there was an elevation of 5.3% in TC (ß ± SE = 0.052 ± 0.002), 11.7% in TG (ß ± SE = 0.111 ± 0.006), 5.8% in HDL-C (ß ± SE = 0.057 ± 0.003), and 8.4% in LDL-C (ß ± SE = 0.081 ± 0.004) per one standard deviation increase in the corresponding PRS. However, their predictive power was attenuated in T2D patients (0.183 < r < 0.231). When we included each PRS (for TC, TG, and LDL-C) in addition to the clinical factors and PCs, the AUC for dyslipidemia was significantly increased by 0.032-0.057 in the general population (7.5 × 10- 3 < P < 0.0400) and 0.029-0.069 in T2D patients (2.1 × 10- 10 < P < 0.0428). Moreover, the quintile of TC-related PRS was moderately associated with cIMT in adult women (ß ± SE = 0.011 ± 0.005, Ptrend = 0.0182). Independent of conventional risk factors, the quintile of PRSs for TC [OR (95% CI) = 1.07 (1.03-1.11)], TG [OR (95% CI) = 1.05 (1.01-1.09)], and LDL-C [OR (95% CI) = 1.05 (1.01-1.09)] were significantly associated with increased risk of CHD in T2D patients (4.8 × 10- 4 < P < 0.0197). Further adjustment for baseline lipid drug use notably attenuated the CHD association. CONCLUSIONS: The PRSs derived and validated here highlight the potential for early genomic screening and personalized risk assessment for cardiovascular disease.


Subject(s)
Asian People/genetics , Atherosclerosis/genetics , Diabetic Cardiomyopathies/genetics , Dyslipidemias/genetics , Genetic Predisposition to Disease , Genome-Wide Association Study , Lipids/blood , Multifactorial Inheritance/genetics , Adolescent , Adult , Atherosclerosis/blood , Carotid Intima-Media Thickness , Coronary Disease/genetics , Diabetes Mellitus, Type 2/genetics , Diabetic Cardiomyopathies/blood , Dyslipidemias/blood , Female , Humans , Risk Factors
15.
Am J Physiol Heart Circ Physiol ; 320(4): H1290-H1302, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33513084

ABSTRACT

Bone marrow-derived mesenchymal stem cells (BM-MSCs) have demonstrated potential in treating diabetic cardiomyopathy. However, patients with diabetes are on multiple drugs and there is a lack of understanding of how transplanted stem cells would respond in presence of such drugs. Metformin is an AMP kinase (AMPK) activator, the widest used antidiabetic drug. In this study, we investigated the effect of metformin on the efficacy of stem cell therapy in a diabetic cardiomyopathy animal model using streptozotocin (STZ) in male Wistar rats. To comprehend the effect of metformin on the efficacy of BM-MSCs, we transplanted BM-MSCs (1 million cells/rat) with or without metformin. Our data demonstrate that transplantation of BM-MSCs prevented cardiac fibrosis and promoted angiogenesis in diabetic hearts. However, metformin supplementation downregulated BM-MSC-mediated cardioprotection. Interestingly, both BM-MSCs and metformin treatment individually improved cardiac function with no synergistic effect of metformin supplementation along with BM-MSCs. Investigating the mechanisms of loss of efficacy of BM-MSCs in the presence of metformin, we found that metformin treatment impairs homing of implanted BM-MSCs in the heart and leads to poor survival of transplanted cells. Furthermore, our data demonstrate that metformin-mediated activation of AMPK is responsible for poor homing and survival of BM-MSCs in the diabetic heart. Hence, the current study confirms that a conflict arises between metformin and BM-MSCs for treating diabetic cardiomyopathy. Approximately 10% of the world population is diabetic to which metformin is prescribed very commonly. Hence, future cell replacement therapies in combination with AMPK inhibitors may be more effective for patients with diabetes.NEW & NOTEWORTHY Metformin treatment reduces the efficacy of mesenchymal stem cell therapy for cardiac repair during diabetic cardiomyopathy. Stem cell therapy in diabetics may be more effective in combination with AMPK inhibitors.


Subject(s)
Cell Movement/drug effects , Diabetes Mellitus, Experimental/drug therapy , Diabetic Cardiomyopathies/surgery , Hypoglycemic Agents/toxicity , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/drug effects , Metformin/toxicity , Myocardium/pathology , AMP-Activated Protein Kinases/metabolism , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Cell Survival/drug effects , Cells, Cultured , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Diabetic Cardiomyopathies/blood , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/pathology , Disease Models, Animal , Fibrosis , Glycated Hemoglobin/metabolism , Insulin/blood , Male , Mesenchymal Stem Cells/metabolism , Myocardium/metabolism , Neovascularization, Physiologic/drug effects , Rats, Wistar , Recovery of Function , Streptozocin
17.
Exp Clin Endocrinol Diabetes ; 129(8): 574-580, 2021 Aug.
Article in English | MEDLINE | ID: mdl-31426113

ABSTRACT

OBJECTIVE: The aim of this study was to evaluate the importance of growth-differentiation factor-15 level and tissue Doppler imaging in the detection of cardiomyopathy in children who have type 1 diabetes mellitus. MATERIALS AND METHODS: Thirty-eight patients (11 males and 27 females) with type 1 diabetes mellitus were included in this study. The control group consisted of 40 age- and gender-matched healthy volunteers. All children underwent a detailed echocardiography, which contained an m-mode, pulse Doppler and tissue Doppler imaging; and growth-differentiation factor-15 level was measured. RESULTS: In this study, there were significant differences between diastolic function parameters of the heart. The mitral isovolumic contraction time, contraction time, and isovolumic relaxation time values were different in the patients than in the controls (p<0.01, p<0.01, p<0.01, respectively). Also, the tricuspid isovolumic contraction time, contraction time, and isovolumic relaxation time values were different in the patients than in the controls (p<0.01, p=0.01, p<0.01, respectively). No statistically significant difference was found between the other M-mode parameters. Mean plasma growth-differentiation factor-15 level was significantly higher in patients than in healthy controls (p<0.01). CONCLUSION: The follow-up of children with type 1 diabetes mellitus in terms of cardiomyopathy and the use of tissue Doppler imaging and growth differentiation factor-15 levels may be useful.


Subject(s)
Diabetes Mellitus, Type 1/complications , Diabetic Cardiomyopathies/diagnosis , Echocardiography, Doppler , Growth Differentiation Factor 15/blood , Adolescent , Child , Child, Preschool , Cross-Sectional Studies , Diabetic Cardiomyopathies/blood , Diabetic Cardiomyopathies/etiology , Female , Humans , Male
18.
Exp Clin Endocrinol Diabetes ; 129(10): 750-756, 2021 Oct.
Article in English | MEDLINE | ID: mdl-32131113

ABSTRACT

BACKGROUND: Type 2 diabetes mellitus (T2DM) promotes the development of atherosclerosis and is a major risk factor for cardiovascular disease. High-sensitivity cardiac troponin I (hs-cTnI) assays fundamentally improved the diagnosis of myocardial injury and even enable the prediction of future cardiovascular events in the general population. However, data about the association of hs-cTnI with cardiovascular risk factors and carotid intima media thickness (cIMT) as a marker of atherosclerosis are limited, especially in patients with T2DM. METHODS: In this cross-sectional study we analyzed clinical and laboratory parameters of 234 patients (43% women) with T2DM and a median age of 65 years (interquartile range: 57-71). The median duration of diabetes mellitus was 10 years (6-17). Anthropometric data, blood pressure, glycemic parameters and lipid profiles were determined. Hs-cTnI plasma concentrations were measured on an ADVIA Centaur XPT immunoassay analyzer and cIMT was evaluated by high-resolution ultrasound. RESULTS: Hs-cTnI plasma concentrations were below the gender-specific 99th percentile in 93% of T2DM patients with a median concentration of 4.0 ng/l (interquartile range: 2.0-10.0). Hs-cTnI was significantly associated with gender, renal function and C-reactive protein in the entire study cohort. Gender-specific analyses revealed cIMT and renal function to be significantly associated with hs-cTnI in men. Contrary, only age was significantly associated with hs-cTnI in women. CONCLUSION: In a real-world clinical setting in patients with T2DM, cIMT is a predictor of subclinical myocardial damage in men, but not in women.


Subject(s)
Carotid Intima-Media Thickness , Diabetes Mellitus, Type 2/complications , Diabetic Cardiomyopathies/diagnosis , Troponin I/blood , Adult , Aged , Cross-Sectional Studies , Diabetic Cardiomyopathies/blood , Diabetic Cardiomyopathies/etiology , Female , Humans , Male , Middle Aged , Prognosis , Sex Factors
19.
Acta Pharmacol Sin ; 42(3): 393-403, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32647341

ABSTRACT

Endoplasmic reticulum stress (ER stress) plays a key role in the development of cardiac hypertrophy and diabetic cardiomyopathy (DCM). Zonisamide (ZNS) was originally developed as an antiepileptic drug. Studies have shown that ZNS suppresses ER stress-induced neuronal cell damage in the experimental models of Parkinson's disease. Herein, we investigated whether ZNS improved DCM by attenuating ER stress-induced apoptosis. C57BL/6J mice were fed with high-fat diet (HFD) and intraperitoneally injected with low-dose streptozotocin (STZ) to induce type 2 diabetes mellitus (T2DM), and then treated with ZNS (40 mg·kg-1·d-1, i.g.) for 16 weeks. We showed that ZNS administration slightly ameliorated the blood glucose levels, but significantly alleviated diabetes-induced cardiac dysfunction and hypertrophy. Furthermore, ZNS administration significantly inhibited the Bax and caspase-3 activity, upregulated Bcl-2 activity, and decreased the proportion of TUNEL-positive cells in heart tissues. We analyzed the hallmarks of ER stress in heart tissues, and revealed that ZNS administration significantly decreased the protein levels of GRP78, XBP-1s, ATF6, PERK, ATF4, and CHOP, and elevated Hrd1 protein. In high glucose (HG)-treated primary cardiomyocytes, application of ZNS (3 µM) significantly alleviated HG-induced cardiomyocyte hypertrophy and apoptosis. ZNS application also suppressed activated ER stress in HG-treated cardiomyocytes. Moreover, preapplication of the specific ER stress inducer tunicamycin (10 ng/mL) eliminated the protective effects of ZNS against HG-induced cardiac hypertrophy and ER stress-mediated apoptosis. Our findings suggest that ZNS improves the cardiac diastolic function in diabetic mice and prevents T2DM-induced cardiac hypertrophy by attenuating ER stress-mediated apoptosis.


Subject(s)
Anticonvulsants/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Diabetic Cardiomyopathies/drug therapy , Endoplasmic Reticulum Stress/drug effects , Zonisamide/therapeutic use , Animals , Apoptosis/drug effects , Blood Glucose/metabolism , Body Weight/drug effects , Cardiomegaly/blood , Cardiomegaly/etiology , Cardiomegaly/prevention & control , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Diabetic Cardiomyopathies/blood , Diabetic Cardiomyopathies/etiology , Diet, High-Fat , Endoplasmic Reticulum Chaperone BiP , Heart/drug effects , Male , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects
20.
Arch Cardiovasc Dis ; 113(11): 736-748, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33189592

ABSTRACT

Besides coronary artery disease, which remains the main cause of heart failure in patients with diabetes, factors independent of coronary artery disease are involved in the development of heart failure in the onset of what is called diabetic cardiomyopathy. Among them, hyperglycaemia - a hallmark of type 2 diabetes - has both acute and chronic deleterious effects on myocardial function, and clearly participates in the establishment of diabetic cardiomyopathy. In the present review, we summarize the cellular and tissular events that occur in a heart exposed to hyperglycaemia, and depict the complex molecular mechanisms proposed to be involved in glucotoxicity. Finally, from a more translational perspective, different therapeutic strategies targeting hyperglycaemia-mediated molecular mechanisms will be detailed.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus/blood , Diabetic Cardiomyopathies/blood , Heart Failure/blood , Hyperglycemia/blood , Myocardium/metabolism , Animals , Blood Glucose/drug effects , Diabetes Mellitus/drug therapy , Diabetes Mellitus/epidemiology , Diabetic Cardiomyopathies/epidemiology , Diabetic Cardiomyopathies/physiopathology , Diabetic Cardiomyopathies/prevention & control , Heart Failure/epidemiology , Heart Failure/physiopathology , Heart Failure/prevention & control , Humans , Hyperglycemia/drug therapy , Hyperglycemia/epidemiology , Hypoglycemic Agents/therapeutic use , Myocardium/pathology , Risk Factors , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...