Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 655
1.
Cardiovasc Diabetol ; 23(1): 164, 2024 May 09.
Article En | MEDLINE | ID: mdl-38724987

Dynamin-related protein 1 (Drp1) is a crucial regulator of mitochondrial dynamics, the overactivation of which can lead to cardiovascular disease. Multiple distinct posttranscriptional modifications of Drp1 have been reported, among which S-nitrosylation was recently introduced. However, the detailed regulatory mechanism of S-nitrosylation of Drp1 (SNO-Drp1) in cardiac microvascular dysfunction in diabetes remains elusive. The present study revealed that mitogen-activated protein kinase kinase kinase kinase 4 (MAP4K4) was consistently upregulated in diabetic cardiomyopathy (DCM) and promoted SNO-Drp1 in cardiac microvascular endothelial cells (CMECs), which in turn led to mitochondrial dysfunction and cardiac microvascular disorder. Further studies confirmed that MAP4K4 promoted SNO-Drp1 at human C644 (mouse C650) by inhibiting glutathione peroxidase 4 (GPX4) expression, through which MAP4K4 stimulated endothelial ferroptosis in diabetes. In contrast, inhibition of MAP4K4 via DMX-5804 significantly reduced endothelial ferroptosis, alleviated cardiac microvascular dysfunction and improved cardiac dysfunction in db/db mice by reducing SNO-Drp1. In parallel, the C650A mutation in mice abolished SNO-Drp1 and the role of Drp1 in promoting cardiac microvascular disorder and cardiac dysfunction. In conclusion, our findings demonstrate that MAP4K4 plays an important role in endothelial dysfunction in DCM and reveal that SNO-Drp1 and ferroptosis activation may act as downstream targets, representing potential therapeutic targets for DCM.


Diabetic Cardiomyopathies , Dynamins , Endothelial Cells , Mice, Inbred C57BL , Signal Transduction , Animals , Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/physiopathology , Diabetic Cardiomyopathies/pathology , Diabetic Cardiomyopathies/enzymology , Diabetic Cardiomyopathies/etiology , Humans , Dynamins/metabolism , Dynamins/genetics , Male , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelial Cells/enzymology , Endothelial Cells/drug effects , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Ferroptosis/drug effects , Disease Models, Animal , Cells, Cultured , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Mitochondria, Heart/enzymology , Mice , Protein Processing, Post-Translational , Coronary Circulation , Intracellular Signaling Peptides and Proteins
2.
Cardiovasc Diabetol ; 23(1): 160, 2024 May 07.
Article En | MEDLINE | ID: mdl-38715043

BACKGROUND: Diabetic cardiomyopathy (DCM) is a crucial complication of long-term chronic diabetes that can lead to myocardial hypertrophy, myocardial fibrosis, and heart failure. There is increasing evidence that DCM is associated with pyroptosis, a form of inflammation-related programmed cell death. Growth differentiation factor 11 (GDF11) is a member of the transforming growth factor ß superfamily, which regulates oxidative stress, inflammation, and cell survival to mitigate myocardial hypertrophy, myocardial infarction, and vascular injury. However, the role of GDF11 in regulating pyroptosis in DCM remains to be elucidated. This research aims to investigate the role of GDF11 in regulating pyroptosis in DCM and the related mechanism. METHODS AND RESULTS: Mice were injected with streptozotocin (STZ) to induce a diabetes model. H9c2 cardiomyocytes were cultured in high glucose (50 mM) to establish an in vitro model of diabetes. C57BL/6J mice were preinjected with adeno-associated virus 9 (AAV9) intravenously via the tail vein to specifically overexpress myocardial GDF11. GDF11 attenuated pyroptosis in H9c2 cardiomyocytes after high-glucose treatment. In diabetic mice, GDF11 alleviated cardiomyocyte pyroptosis, reduced myocardial fibrosis, and improved cardiac function. Mechanistically, GDF11 inhibited pyroptosis by preventing inflammasome activation. GDF11 achieved this by specifically binding to apoptosis-associated speck-like protein containing a CARD (ASC) and preventing the assembly and activation of the inflammasome. Additionally, the expression of GDF11 during pyroptosis was regulated by peroxisome proliferator-activated receptor α (PPARα). CONCLUSION: These findings demonstrate that GDF11 can treat diabetic cardiomyopathy by alleviating pyroptosis and reveal the role of the PPARα-GDF11-ASC pathway in DCM, providing ideas for new strategies for cardioprotection.


Diabetes Mellitus, Experimental , Diabetic Cardiomyopathies , Fibrosis , Growth Differentiation Factors , Inflammasomes , Mice, Inbred C57BL , Myocytes, Cardiac , Pyroptosis , Signal Transduction , Animals , Pyroptosis/drug effects , Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/pathology , Diabetic Cardiomyopathies/prevention & control , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/physiopathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myocytes, Cardiac/drug effects , Diabetes Mellitus, Experimental/metabolism , Cell Line , Inflammasomes/metabolism , Male , Growth Differentiation Factors/metabolism , Rats , Blood Glucose/metabolism , Mice , Glucose/metabolism , Glucose/toxicity , Bone Morphogenetic Proteins , PPAR alpha
3.
Cardiovasc Diabetol ; 23(1): 139, 2024 Apr 25.
Article En | MEDLINE | ID: mdl-38664790

BACKGROUND: Diabetic cardiomyopathy (DCM) poses a growing health threat, elevating heart failure risk in diabetic individuals. Understanding DCM is crucial, with fibroblasts and endothelial cells playing pivotal roles in driving myocardial fibrosis and contributing to cardiac dysfunction. Advances in Multimodal single-cell profiling, such as scRNA-seq and scATAC-seq, provide deeper insights into DCM's unique cell states and molecular landscape for targeted therapeutic interventions. METHODS: Single-cell RNA and ATAC data from 10x Multiome libraries were processed using Cell Ranger ARC v2.0.1. Gene expression and ATAC data underwent Seurat and Signac filtration. Differential gene expression and accessible chromatin regions were identified. Transcription factor activity was estimated with chromVAR, and Cis-coaccessibility networks were calculated using Cicero. Coaccessibility connections were compared to the GeneHancer database. Gene Ontology analysis, biological process scoring, cell-cell communication analysis, and gene-motif correlation was performed to reveal intricate molecular changes. Immunofluorescent staining utilized various antibodies on paraffin-embedded tissues to verify the findings. RESULTS: This study integrated scRNA-seq and scATAC-seq data obtained from hearts of WT and DCM mice, elucidating molecular changes at the single-cell level throughout the diabetic cardiomyopathy progression. Robust and accurate clustering analysis of the integrated data revealed altered cell proportions, showcasing decreased endothelial cells and macrophages, coupled with increased fibroblasts and myocardial cells in the DCM group, indicating enhanced fibrosis and endothelial damage. Chromatin accessibility analysis unveiled unique patterns in cell types, with heightened transcriptional activity in myocardial cells. Subpopulation analysis highlighted distinct changes in cardiomyocytes and fibroblasts, emphasizing pathways related to fatty acid metabolism and cardiac contraction. Fibroblast-centered communication analysis identified interactions with endothelial cells, implicating VEGF receptors. Endothelial cell subpopulations exhibited altered gene expressions, emphasizing contraction and growth-related pathways. Candidate regulators, including Tcf21, Arnt, Stat5a, and Stat5b, were identified, suggesting their pivotal roles in DCM development. Immunofluorescence staining validated marker genes of cell subpopulations, confirming PDK4, PPARγ and Tpm1 as markers for metabolic pattern-altered cardiomyocytes, activated fibroblasts and endothelial cells with compromised proliferation. CONCLUSION: Our integrated scRNA-seq and scATAC-seq analysis unveils intricate cell states and molecular alterations in diabetic cardiomyopathy. Identified cell type-specific changes, transcription factors, and marker genes offer valuable insights. The study sheds light on potential therapeutic targets for DCM.


Diabetic Cardiomyopathies , Single-Cell Analysis , Transcriptome , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/pathology , Diabetic Cardiomyopathies/physiopathology , Animals , Gene Expression Profiling , Chromatin/metabolism , Chromatin/genetics , Mice, Inbred C57BL , Gene Regulatory Networks , Chromatin Assembly and Disassembly , Disease Models, Animal , Male , RNA-Seq , Gene Expression Regulation , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis , Mice , Endothelial Cells/metabolism , Endothelial Cells/pathology
4.
Front Endocrinol (Lausanne) ; 13: 801260, 2022.
Article En | MEDLINE | ID: mdl-35242109

Type 2 diabetes (T2D) patients with SARS-CoV-2 infection hospitalized develop an acute cardiovascular syndrome. It is urgent to elucidate underlying mechanisms associated with the acute cardiac injury in T2D hearts. We performed bioinformatic analysis on the expression profiles of public datasets to identify the pathogenic and prognostic genes in T2D hearts. Cardiac RNA-sequencing datasets from db/db or BKS mice (GSE161931) were updated to NCBI-Gene Expression Omnibus (NCBI-GEO), and used for the transcriptomics analyses with public datasets from NCBI-GEO of autopsy heart specimens with COVID-19 (5/6 with T2D, GSE150316), or dead healthy persons (GSE133054). Differentially expressed genes (DEGs) and overlapping homologous DEGs among the three datasets were identified using DESeq2. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes analyses were conducted for event enrichment through clusterProfile. The protein-protein interaction (PPI) network of DEGs was established and visualized by Cytoscape. The transcriptions and functions of crucial genes were further validated in db/db hearts. In total, 542 up-regulated and 485 down-regulated DEGs in mice, and 811 up-regulated and 1399 down-regulated DEGs in human were identified, respectively. There were 74 overlapping homologous DEGs among all datasets. Mitochondria inner membrane and serine-type endopeptidase activity were further identified as the top-10 GO events for overlapping DEGs. Cardiac CAPNS1 (calpain small subunit 1) was the unique crucial gene shared by both enriched events. Its transcriptional level significantly increased in T2D mice, but surprisingly decreased in T2D patients with SARS-CoV-2 infection. PPI network was constructed with 30 interactions in overlapping DEGs, including CAPNS1. The substrates Junctophilin2 (Jp2), Tnni3, and Mybpc3 in cardiac calpain/CAPNS1 pathway showed less transcriptional change, although Capns1 increased in transcription in db/db mice. Instead, cytoplasmic JP2 significantly reduced and its hydrolyzed product JP2NT exhibited nuclear translocation in myocardium. This study suggests CAPNS1 is a crucial gene in T2D hearts. Its transcriptional upregulation leads to calpain/CAPNS1-associated JP2 hydrolysis and JP2NT nuclear translocation. Therefore, attenuated cardiac CAPNS1 transcription in T2D patients with SARS-CoV-2 infection highlights a novel target in adverse prognostics and comprehensive therapy. CAPNS1 can also be explored for the molecular signaling involving the onset, progression and prognostic in T2D patients with SARS-CoV-2 infection.


COVID-19/epidemiology , Computational Biology , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/genetics , Diabetic Cardiomyopathies/epidemiology , SARS-CoV-2 , Adult , Aged , Aged, 80 and over , Animals , Calpain/genetics , Calpain/physiology , Comorbidity , Diabetes Mellitus, Type 2/physiopathology , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/physiopathology , Humans , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Middle Aged , Mitochondria, Heart/ultrastructure , Muscle Proteins/metabolism , Myocardium/chemistry , Myocardium/metabolism , Myocardium/ultrastructure , Prognosis , Sequence Analysis, RNA , Transcriptome
5.
BMC Cardiovasc Disord ; 22(1): 59, 2022 02 17.
Article En | MEDLINE | ID: mdl-35172745

BACKGROUND: A noninvasive left ventricular (LV) pressure-strain loop (PSL) provides a new method to quantify myocardial work (MW) by combining global longitudinal strain (GLS) and LV pressure, which exerts potential advantages over traditional GLS. We studied the LV PSL and MW in patients with type 2 diabetes mellitus (T2DM). METHODS: This cross-sectional study included 201 subjects (54 healthy controls and 147 T2DM patients) who underwent complete two-dimensional echocardiography (2DE), including 2D speckle-tracking echocardiography (STE), as well as brachial artery pulse pressure measurement. The PSL was used to determine the global myocardial work index (GWI), global constructive work (GCW), global wasted work (GWW), and global work efficiency (GWE) of all study participants. The association between T2DM and LV function was evaluated according to these MW indices. RESULTS: The GLS was significantly lower in the T2DM group than in the control group (P < 0.001), indicating that the LV myocardium had been damaged, although the LV ejection fraction (LVEF) was still normal. The GWI and GWE were decreased (P = 0.022) and the GWW was increased (P < 0.001) in diabetic patients compared with controls, but the GCW was comparable in the two groups (P = 0.160). In all diabetic patients, age, body mass index, systolic blood pressure, smoking history, and LVEF were correlated with GWI, GWW and GWE. CONCLUSIONS: The use of LV PSL is a novel noninvasive technique that could help to depict the relationship between LV myocardial damage and MW in patients with T2DM.


Diabetes Mellitus, Type 2/complications , Diabetic Cardiomyopathies/diagnostic imaging , Echocardiography, Doppler , Ventricular Dysfunction, Left/diagnostic imaging , Ventricular Function, Left , Ventricular Pressure , Adult , Case-Control Studies , Cross-Sectional Studies , Diabetes Mellitus, Type 2/diagnosis , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/physiopathology , Female , Humans , Male , Middle Aged , Observer Variation , Predictive Value of Tests , Reproducibility of Results , Ventricular Dysfunction, Left/etiology , Ventricular Dysfunction, Left/physiopathology
6.
Rev. clín. esp. (Ed. impr.) ; 222(2): 100-111, feb. 2022. ilus
Article Es | IBECS | ID: ibc-204626

La relación entre la diabetes y la insuficiencia cardiaca es compleja y bidireccional. No obstante, la existencia de una miocardiopatía como entidad propia y atribuible exclusivamente a la diabetes ha sido y es motivo de controversia hoy día. Esto es debido, entre otros motivos, a la ausencia de una definición de consenso. Tampoco existe unanimidad en cuanto a los hallazgos fisiopatogénicos presentes en la miocardiopatía diabética ni en su clasificación. Esto añadido a la ausencia de métodos diagnósticos propios o de tratamientos específicos en la enfermedad, limita el conocimiento general de la patología. Sin embargo, los estudios realizados en miocardiopatía diabética sugieren una fisiopatogenia propia diferenciada de la de otras entidades. De la misma manera, nuevos tratamientos han demostrado tener un papel potencial en esta enfermedad. En la siguiente revisión realizamos una actualización de la miocardiopatía diabética (AU)


The relationship between diabetes and heart failure is complex and bidirectional. Nevertheless, the existence of a cardiomyopathy attributable exclusively to diabetes has been and is still the subject of controversy, due, among other reasons, to a lack of a consensus definition. There is also no unanimous agreement in terms of the physiopathogenic findings that need to be present in the definition of diabetic cardiomyopathy or on its classification, which, added to the lack of diagnostic methods and treatments specific for this disease, limits its general understanding. Studies conducted on diabetic cardiomyopathy, however, suggest a unique physiopathogenesis different from that of other diseases. Similarly, new treatments have been shown to play a potential role in this disease. The following review provides an update on diabetic cardiomyopathy (AU)


Humans , Diabetic Cardiomyopathies , Diabetic Cardiomyopathies/diagnosis , Diabetic Cardiomyopathies/physiopathology , Diabetic Cardiomyopathies/therapy
7.
Cells ; 11(2)2022 01 11.
Article En | MEDLINE | ID: mdl-35053356

The incidence and prevalence of diabetes mellitus (DM) are increasing worldwide, and the resulting cardiac complications are the leading cause of death. Among these complications is diabetes-induced cardiomyopathy (DCM), which is the consequence of a pro-inflammatory condition, oxidative stress and fibrosis caused by hyperglycemia. Cardiac remodeling will lead to an imbalance in cell survival and death, which can promote cardiac dysfunction. Since the conventional treatment of DM generally does not address the prevention of cardiac remodeling, it is important to develop new alternatives for the treatment of cardiovascular complications induced by DM. Thus, therapy with mesenchymal stem cells has been shown to be a promising approach for the prevention of DCM because of their anti-apoptotic, anti-fibrotic and anti-inflammatory effects, which could improve cardiac function in patients with DM.


Diabetic Cardiomyopathies/therapy , Mesenchymal Stem Cell Transplantation , Animals , Clinical Trials as Topic , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/physiopathology , Epigenesis, Genetic , Humans , Models, Biological , Vascular Remodeling
8.
Am J Physiol Heart Circ Physiol ; 322(3): H394-H405, 2022 03 01.
Article En | MEDLINE | ID: mdl-35089809

As there is cross talk in functions of the heart and kidney, acute or chronic injury in one of the two organs provokes adaptive and/or maladaptive responses in both organs, leading to cardiorenal syndrome (CRS). Acute kidney injury (AKI) induced by acute heart failure is referred to as type 1 CRS, and a frequent cause of this type of CRS is acute myocardial infarction (AMI). Diabetes mellitus increases the risk of AMI and also the risk of AKI of various causes. However, there have been only a few studies in which animal models of diabetes were used to examine how diabetes modulates AMI-induced AKI. In this review, we summarize findings regarding the mechanisms of type 1 CRS and the impact of diabetes on both AMI and renal susceptibility to AKI and we discuss mechanisms by which diabetes modulates AMI-induced AKI. Hemodynamic alterations induced by AMI could be augmented by diabetes via its detrimental effect on infarct size and contractile function of the noninfarcted region in the heart. Diabetes increases susceptibility of renal cells to hypoxia and oxidative stress by modulation of signaling pathways that regulate cell survival and autophagy. Recent studies have shown that diabetes mellitus even at early stage of cardiomyopathy/nephropathy predisposes the kidney to AMI-induced AKI, in which activation of Toll-like receptors and reactive oxygen species derived from NADPH oxidases are involved. Further analysis of cross talk between diabetic cardiomyopathy and diabetic kidney disease is necessary for obtaining a more comprehensive understanding of modulation of the AMI-AKI axis by diabetes.


Acute Kidney Injury/physiopathology , Cardio-Renal Syndrome/physiopathology , Diabetic Cardiomyopathies/physiopathology , Diabetic Nephropathies/physiopathology , Myocardial Infarction/physiopathology , Acute Kidney Injury/metabolism , Animals , Cardio-Renal Syndrome/metabolism , Diabetic Cardiomyopathies/metabolism , Diabetic Nephropathies/metabolism , Humans , Myocardial Infarction/metabolism
9.
Cardiovasc Diabetol ; 21(1): 5, 2022 01 06.
Article En | MEDLINE | ID: mdl-34991588

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.


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
10.
Cardiovasc Toxicol ; 22(1): 88-96, 2022 01.
Article En | MEDLINE | ID: mdl-34674150

Ciliary neurotrophic factor (CNTF), which is a neural peptide, has been reported to confer cardioprotective effects. However, whether CNTF-based gene delivery could prevent cardiac remodeling in diabetes mellitus remains unknown. In this study, we used adeno-associated viral vector serotype 9 (AAV9)-based cardiac gene delivery to test the effects of CNTF overexpression on adverse ventricular remodeling in streptozotocin-induced type 1 diabetic mice models. Postnatal (P3-P10) mice were peritoneally injected with AAV9 recombinant virus carrying the CNTF gene or EGFP gene. Then, type 1 diabetic models were established by peritoneal injection of streptozotocin (200 mg/kg) in 7-week-old female mice injected with AAV9. 4 weeks later after the establishment of type 1 diabetes mellitus, mouse hearts were removed to assess the degree of cardiac remodeling. We found that CNTF overexpression in mouse cardiomyocytes exacerbated cell apoptosis and cardiac fibrosis coupled with an increased inflammatory response in the heart tissue of diabetic female mice. Taken together, our results suggested that cardiac CNTF gene delivery may not be beneficial in alleviating adverse cardiac remodeling in type 1 diabetes female mice.


Ciliary Neurotrophic Factor/metabolism , Dependovirus/genetics , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 1/complications , Diabetic Cardiomyopathies/metabolism , Gene Transfer Techniques , Genetic Vectors , Myocytes, Cardiac/metabolism , Ventricular Remodeling , Animals , Apoptosis , Ciliary Neurotrophic Factor/genetics , Cytokines/genetics , Cytokines/metabolism , Dependovirus/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Type 1/chemically induced , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/physiopathology , Female , Fibrosis , Inflammation Mediators , Mice, Inbred C57BL , Myocytes, Cardiac/pathology , Streptozocin , Up-Regulation
11.
J Diabetes Investig ; 13(1): 102-111, 2022 Jan.
Article En | MEDLINE | ID: mdl-34228899

AIMS/INTRODUCTION: To investigate the association between cardiovascular autonomic neuropathy (CAN) assessed by the coefficient of variation of the R-R interval and the reduction in the estimated glomerular filtration rate (eGFR) in patients with type 2 diabetes. MATERIALS AND METHODS: This retrospective observational cohort study enrolled type 2 diabetes patients who had their coefficient of variation of the R-R interval measured on an electrocardiogram from January 2005 to December 2018. CAN was defined using the reference coefficient of variation of the R-R interval value based on age and sex. The primary outcome was set as a 40% eGFR decline from baseline. Regression analyses using the Cox proportional hazards model were carried out to evaluate the association. RESULTS: Of the 831 patients, 118 (14.2%) were diagnosed with CAN. In the analysis of the primary outcome, the median follow-up period was 5.3 years, and 25 (21.2%) patients with CAN and 78 (10.9%) patients without CAN developed a 40% eGFR decline. In the univariate regression analysis, CAN was significantly associated with a 40% eGFR decline (hazard ratio 2.42, 95% confidence interval 1.54-3.80). In the multivariate analysis, CAN remained almost significant after adjusting for the prognostic risk factors for CAN and the decline in the renal function, and an interaction with proteinuria was found. In analyses for the interaction effect between CAN and proteinuria, the presence of CAN synergistically increased the risk of an eGFR decline in patients with macroproteinuria. CONCLUSIONS: CAN strongly increased the risk of a 40% eGFR decline from baseline, especially in type 2 diabetes patients with macroproteinuria.


Diabetes Mellitus, Type 2/physiopathology , Diabetic Cardiomyopathies/physiopathology , Diabetic Neuropathies/physiopathology , Proteinuria/physiopathology , Autonomic Nervous System/physiopathology , Diabetes Mellitus, Type 2/complications , Diabetic Cardiomyopathies/etiology , Diabetic Neuropathies/etiology , Electrocardiography , Female , Glomerular Filtration Rate , Humans , Kidney/physiopathology , Male , Middle Aged , Proportional Hazards Models , Proteinuria/etiology , Regression Analysis , Retrospective Studies , Risk Factors
12.
Cardiovasc Res ; 118(1): 212-225, 2022 01 07.
Article En | MEDLINE | ID: mdl-33576380

AIMS: The glucose-driven enzymatic modification of myocardial proteins by the sugar moiety, ß-N-acetylglucosamine (O-GlcNAc), is increased in pre-clinical models of diabetes, implicating protein O-GlcNAc modification in diabetes-induced heart failure. Our aim was to specifically examine cardiac manipulation of the two regulatory enzymes of this process on the cardiac phenotype, in the presence and absence of diabetes, utilising cardiac-targeted recombinant-adeno-associated viral-vector-6 (rAAV6)-mediated gene delivery. METHODS AND RESULTS: In human myocardium, total protein O-GlcNAc modification was elevated in diabetic relative to non-diabetic patients, and correlated with left ventricular (LV) dysfunction. The impact of rAAV6-delivered O-GlcNAc transferase (rAAV6-OGT, facilitating protein O-GlcNAcylation), O-GlcNAcase (rAAV6-OGA, facilitating de-O-GlcNAcylation), and empty vector (null) were determined in non-diabetic and diabetic mice. In non-diabetic mice, rAAV6-OGT was sufficient to impair LV diastolic function and induce maladaptive cardiac remodelling, including cardiac fibrosis and increased Myh-7 and Nppa pro-hypertrophic gene expression, recapitulating characteristics of diabetic cardiomyopathy. In contrast, rAAV6-OGA (but not rAAV6-OGT) rescued LV diastolic function and adverse cardiac remodelling in diabetic mice. Molecular insights implicated impaired cardiac PI3K(p110α)-Akt signalling as a potential contributing mechanism to the detrimental consequences of rAAV6-OGT in vivo. In contrast, rAAV6-OGA preserved PI3K(p110α)-Akt signalling in diabetic mouse myocardium in vivo and prevented high glucose-induced impairments in mitochondrial respiration in human cardiomyocytes in vitro. CONCLUSION: Maladaptive protein O-GlcNAc modification is evident in human diabetic myocardium, and is a critical regulator of the diabetic heart phenotype. Selective targeting of cardiac protein O-GlcNAcylation to restore physiological O-GlcNAc balance may represent a novel therapeutic approach for diabetes-induced heart failure.


Antigens, Neoplasm/metabolism , Diabetic Cardiomyopathies/enzymology , Histone Acetyltransferases/metabolism , Hyaluronoglucosaminidase/metabolism , Myocytes, Cardiac/enzymology , N-Acetylglucosaminyltransferases/metabolism , Protein Processing, Post-Translational , Ventricular Dysfunction, Left/enzymology , Ventricular Function, Left , Ventricular Remodeling , Aged , Animals , Antigens, Neoplasm/genetics , Cell Line , Class I Phosphatidylinositol 3-Kinases/metabolism , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/pathology , Diabetic Cardiomyopathies/physiopathology , Disease Models, Animal , Female , Fibrosis , Gene Expression Regulation , Glycosylation , Histone Acetyltransferases/genetics , Humans , Hyaluronoglucosaminidase/genetics , Male , Mice , Middle Aged , Myocytes, Cardiac/pathology , N-Acetylglucosaminyltransferases/genetics , Phenotype , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/pathology , Ventricular Dysfunction, Left/physiopathology
13.
Cardiovasc Res ; 118(1): 196-211, 2022 01 07.
Article En | MEDLINE | ID: mdl-33483741

AIMS: The aim of this study was to identify the molecular mechanism for hyperglycaemia-induced metabolic memory in endothelial cells (ECs), and to show its critical importance to development of cardiovascular dysfunction in diabetes. METHODS AND RESULTS: Hyperglycaemia induces increased nuclear factor-κB (NF-κB) signalling, up-regulation of miR-27a-3p, down-regulation of nuclear factor erythroid-2 related factor 2 (NRF2) expression, increased transforming growth factor-ß (TGF-ß) signalling, down-regulation of miR-29, and induction of endothelial-to-mesenchymal transition (EndMT), all of which are memorized by ECs and not erased when switched to a low glucose condition, thereby causing perivascular fibrosis and cardiac dysfunction. Similar metabolic memory effects are found for production of nitric oxide (NO), generation of reactive oxygen species (ROS), and the mitochondrial oxygen consumption rate in two different types of ECs. The observed metabolic memory effects in ECs are blocked by NRF2 activator tert-butylhydroquinone and a miR-27a-3p inhibitor. In vivo, the NRF2 activator and miR-27a-3p inhibitor block cardiac perivascular fibrosis and restore cardiovascular function by decreasing NF-κB signalling, down-regulating miR-27a-3p, up-regulating NRF2 expression, reducing TGF-ß signalling, and inhibiting EndMT during insulin treatment of diabetes in streptozotocin-induced diabetic mice, whereas insulin alone does not improve cardiac function. CONCLUSIONS: Our data indicate that disruption of hyperglycaemia-induced EC metabolic memory is required for restoring cardiac function during treatment of diabetes, and identify a novel molecular signalling pathway of NF-κB/miR-27a-3p/NRF2/ROS/TGF-ß/EndMT involved in metabolic memory.


Blood Glucose/metabolism , Diabetic Cardiomyopathies/metabolism , Endothelial Cells/metabolism , Energy Metabolism , Epithelial-Mesenchymal Transition , Animals , Cells, Cultured , Diabetic Cardiomyopathies/drug therapy , Diabetic Cardiomyopathies/pathology , Diabetic Cardiomyopathies/physiopathology , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/pathology , Energy Metabolism/drug effects , Epithelial-Mesenchymal Transition/drug effects , Fibrosis , Humans , Hydroquinones/pharmacology , Male , Mice, Inbred BALB C , MicroRNAs/genetics , MicroRNAs/metabolism , NF-E2-Related Factor 2/agonists , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism
14.
Diabetes ; 71(2): 298-314, 2022 02 01.
Article En | MEDLINE | ID: mdl-34844991

Cardiometabolic diseases, including diabetes and its cardiovascular complications, are the global leading causes of death, highlighting a major unmet medical need. Over the past decade, mitsugumin 53 (MG53), also called TRIM72, has emerged as a powerful agent for myocardial membrane repair and cardioprotection, but its therapeutic value is complicated by its E3 ligase activity, which mediates metabolic disorders. Here, we show that an E3 ligase-dead mutant, MG53-C14A, retains its cardioprotective function without causing metabolic adverse effects. When administered in normal animals, both the recombinant human wild-type MG53 protein (rhMG53-WT) and its E3 ligase-dead mutant (rhMG53-C14A) protected the heart equally from myocardial infarction and ischemia/reperfusion (I/R) injury. However, in diabetic db/db mice, rhMG53-WT treatment markedly aggravated hyperglycemia, cardiac I/R injury, and mortality, whereas acute and chronic treatment with rhMG53-C14A still effectively ameliorated I/R-induced myocardial injury and mortality or diabetic cardiomyopathy, respectively, without metabolic adverse effects. Furthermore, knock-in of MG53-C14A protected the mice from high-fat diet-induced metabolic disorders and cardiac damage. Thus, the E3 ligase-dead mutant MG53-C14A not only protects the heart from acute myocardial injury but also counteracts metabolic stress, providing a potentially important therapy for the treatment of acute myocardial injury in metabolic disorders, including diabetes and obesity.


Membrane Proteins/genetics , Metabolic Syndrome/genetics , Myocardial Reperfusion Injury/prevention & control , Animals , Cells, Cultured , Cytoprotection/genetics , Diabetic Cardiomyopathies/complications , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/physiopathology , Diet, High-Fat , Female , Heart/physiopathology , Humans , Male , Metabolic Syndrome/metabolism , Metabolic Syndrome/pathology , Metabolic Syndrome/physiopathology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Myocardial Reperfusion Injury/etiology , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Signal Transduction/genetics
15.
Metabolism ; 127: 154936, 2022 02.
Article En | MEDLINE | ID: mdl-34801581

BACKGROUND: The pathogenesis of experimental diabetic cardiomyopathy may involve the activator protein 1 (AP-1) member, JunD. Using non-diabetic heart transplant (HTX) in recipients with diabetes, we examined the effects of the diabetic milieu (hyperglycemia and insulin resistance) on cardiac JunD expression over 12 months. Because sodium/glucose cotransporter-2 inhibitors (SGLT2i) significantly reverse high glucose-induced AP-1 binding in the proximal tubular cell, we investigated JunD expression in a subgroup of type 2 diabetic recipients receiving SGLT2i treatment. METHODS: We evaluated 77 first HTX recipients (40 and 37 patients with and without diabetes, respectively). Among the recipients with diabetes, 17 (45.9%) were receiving SGLT2i treatment. HTX recipients underwent standard clinical evaluation (metabolic status, echocardiography, coronary computed tomography angiography, and endomyocardial biopsy). In the biopsy samples, we evaluated JunD, insulin receptor substrates 1 and 2 (IRS1 and IRS2), peroxisome proliferator-activated receptor-γ (PPAR-γ), and ceramide levels using real-time polymerase chain reaction and immunofluorescence. The biopsy evaluations in this study were performed at 1-4 weeks (basal), 5-12 weeks (intermediate), and up to 48 weeks (final, end of 12-month follow-up) after HTX. RESULTS: There was a significant early and progressive increase in the cardiac expression of JunD/PPAR-γ and ceramide levels, along with a significant decrease in IRS1 and IRS2 in recipients with diabetes but not in those without diabetes. These molecular changes were blunted in patients with diabetes receiving SGLT2i treatment. CONCLUSION: Early pathogenesis in human diabetic cardiomyopathy is associated with JunD/PPAR-γ overexpression and lipid accumulation following HTX in recipients with diabetes. Remarkably, this phenomenon was reduced by concomitant therapy with SGLT2i, which acted directly on diabetic hearts.


Diabetic Cardiomyopathies , Heart/drug effects , Proto-Oncogene Proteins c-jun/genetics , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Adult , Biopsy , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/surgery , Diabetic Cardiomyopathies/drug therapy , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/physiopathology , Diabetic Cardiomyopathies/surgery , Female , Follow-Up Studies , Gene Expression/drug effects , Heart/physiology , Heart Transplantation , Humans , Lipid Metabolism/drug effects , Male , Middle Aged , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use
16.
Biochem Biophys Res Commun ; 586: 8-13, 2022 01 01.
Article En | MEDLINE | ID: mdl-34818584

AIM: To evaluate the effects of exercise training (ET) on cardiac extracellular matrix (ECM) proteins homeostasis and cardiac dysfunction in mice with diabetic cardiomyopathy. METHODS: Thirty-six male C57BL/6 mice were randomized into 3 groups for 8 weeks (12mice/group): Diabetic control-DC: Diabetes was induced by single streptozotocin injection (200 mg/kg i.p.); Diabetic exercise-DE: Diabetic mice underwent ET program on motorized-treadmill (6-times/week, 60min/session); Non-diabetic control-NDC: Vehicle-treated, sedentary, non-diabetic mice served as controls. Before euthanasia, all groups underwent transthoracic echocardiography (TTE). Post-mortem, left-ventricle (LV) samples were histologically analysed for ECM proteins (collagen, elastin), matrix metalloproteinases (MMPs) and their tissue inhibitors (TIMPs). RESULTS: DC group showed significantly higher cardiac contents of collagen and MMP-9 and lower elastic concentration than NDC (p < 0.001). The implementation of ET completely outweighed those diabetes-induced changes (DE vs NDC, p > 0.05). TIMP-1 levels significantly increased across all groups (DC: 18.98 ± 3.47%, DE: 24.24 ± 2.36%, NDC: 46.36 ± 5.91%; p < 0.05), while MMP-9/TIMP-1 ratio followed a reverse pattern. ET tended to increase MMP-2 concentrations versus DC (p = 0.055), but did not achieve non-diabetic levels (p < 0.05). TIMP-2 cardiac concentrations remained unaltered throughout the study (p > 0.05). Importantly, ET ameliorated both LV end-systolic internal diameter (LVESD) (p < 0.001) and the percentage of LV fractional shortening (FS%) (p = 0.006) compared to DC. Despite that favorable effect, the cardiac function level of DE group remained worse than NDC group (%FS: p = 0.002; LVESD: p < 0.001). CONCLUSION: Systemic ET may favorably change ECM proteins, MMP-9 and TIMP-1 cardiac concentrations in mice with diabetic cardiomyopathy. Those results were associated with partial improvement of echocardiography-assessed cardiac function, indicating a therapeutic effect of ET in diabetic cardiomyopathy.


Diabetes Mellitus, Experimental/enzymology , Diabetic Cardiomyopathies/enzymology , Extracellular Matrix/enzymology , Matrix Metalloproteinase 9/genetics , Physical Conditioning, Animal/physiology , Tissue Inhibitor of Metalloproteinase-1/genetics , Animals , Blood Glucose/metabolism , Collagen/genetics , Collagen/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/physiopathology , Diabetic Cardiomyopathies/chemically induced , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/physiopathology , Echocardiography , Elastin/genetics , Elastin/metabolism , Exercise Test , Extracellular Matrix/genetics , Gene Expression Regulation , Heart Ventricles/metabolism , Heart Ventricles/physiopathology , Male , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction , Streptozocin/administration & dosage , Tissue Inhibitor of Metalloproteinase-1/metabolism
17.
Cell Mol Biol (Noisy-le-grand) ; 67(3): 201-203, 2021 Nov 25.
Article En | MEDLINE | ID: mdl-34933708

Type 1 diabetes mellitus, also called insulin-dependent diabetes is associated with elevated blood glucose concentration arising from the inability of the pancreas to produce insulin. Diabetic cardiomyopathy is a major cause of death in diabetic patients. CircRNAs have been reported to participate in various human diseases, including diabetic cardiomyopathy. In this study, the regulation network of circRNA in type 1 diabetes mellitus was investigated. Streptozotocin treatment was implemented to induce type 1 diabetes mellitus in the mouse model, and echocardiography was implemented to detect the heart function of the type 1 diabetes mellitus mouse. Also, the qRT-PCR assay was used to identify the circRNA expression in type 1 diabetes mellitus mouse myocardial tissue. Findings showed that heart function of type 1 diabetes mellitus mouse was significantly damaged than control group mouse and cardiac hypertrophy in type 1 diabetes mellitus mouse, circRNAs were aberrantly regulated in type 1 diabetes mellitus mouse myocardial tissue. The following circRNAs were mmu_circ_0001560, mmu_circ_0001800, mmu_circ_0001801, mmu_circ_0002281 and mmu_circ_0000614 were expressed low in type 1 diabetes mellitus mouse myocardial tissue. In conclusion, type 1 diabetes mellitus caused alterations in the regulation network of circRNAs.


Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Type 1/genetics , Gene Expression Regulation , Gene Regulatory Networks , Myocardium/metabolism , RNA, Circular/genetics , Animals , Diabetic Cardiomyopathies/diagnosis , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/physiopathology , Disease Models, Animal , Humans , Male , Mice , Myocardium/cytology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Reverse Transcriptase Polymerase Chain Reaction
18.
Biochem Biophys Res Commun ; 584: 80-86, 2021 12 20.
Article En | MEDLINE | ID: mdl-34775284

The aim of the study was to explore different effects of exercise, metformin alone, or exercise combined with metformin on cardiovascular morphological and functional changes in early stage of type 2 diabetes mellitus. Eight-week-old diabetic db/db mice and BKS mice were recruited and exposed to three different treatments (exercise, metformin alone, or their combination) for 8 weeks. Metformin was administered intragastrically, and aerobic exercise was performed using treadmill with 7-12 m/min, 30-40 min/day, 5 days/week. In the combination group, aerobic exercise was carried out for 30 min after intragastric administration of metformin. The results showed that all three treatments improved cardiac fibrosis and aortic lipid deposition. Exercise intervention failed to alleviate myocardial hypertrophy, but it improved the declined heart rate and diastolic blood pressure in diabetic db/db mice. In contrast, metformin caused opposite effects in these mice. The combination of exercise and metformin had additive effects on glucose intolerance and insulin sensitivity rather than on the improvement of myocardial and aortic structure. In conclusion, metformin improved changes in the morphology and structure of the heart and aorta, while exercise alone or in combination with metformin demonstrated more advantages in cardiac functional reserve through the physiological hypertrophy of myocardium in diabetic db/db mice.


Diabetes Mellitus, Type 2/complications , Diabetic Cardiomyopathies/therapy , Exercise Therapy/methods , Metformin/pharmacology , Physical Conditioning, Animal/physiology , Treatment Outcome , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Body Weight/drug effects , Body Weight/physiology , Combined Modality Therapy , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/physiopathology , Glucose Intolerance/physiopathology , Glucose Intolerance/therapy , Hypoglycemic Agents/pharmacology , Insulin Resistance , Male , Mice , Time Factors
19.
Metabolism ; 125: 154910, 2021 12.
Article En | MEDLINE | ID: mdl-34627874

Heart failure and cardiovascular disorders represent the leading cause of death in diabetic patients. Here we present a systematic review of the main mechanisms underlying the development of diabetic cardiomyopathy. We also provide an excursus on the relative contribution of cardiomyocytes, fibroblasts, endothelial and smooth muscle cells to the pathophysiology of heart failure in diabetes. After having described the preclinical tools currently available to dissect the mechanisms of this complex disease, we conclude with a section on the most recent updates of the literature on clinical management.


Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 2/complications , Diabetic Cardiomyopathies/physiopathology , Heart Failure/physiopathology , Diabetic Cardiomyopathies/etiology , Heart Failure/etiology , Humans
20.
Cardiovasc Diabetol ; 20(1): 212, 2021 10 23.
Article En | MEDLINE | ID: mdl-34688280

BACKGROUND: Left ventricular (LV) involvement in diabetic cardiomyopathy has been reported; however, only limited data exist on right ventricular (RV) involvement. Therefore, our purpose was to investigate RV systolic dysfunction and its association with LV longitudinal myocardial dysfunction in patients with type 2 diabetes mellitus (T2DM) and preserved LV ejection fraction (LVEF). METHODS: We studied 177 T2DM patients with preserved LVEF and 79 age-, sex-, and LVEF-matched healthy volunteers. LV longitudinal myocardial function was assessed as global longitudinal strain (GLS), and RV systolic function was assessed as RV free-wall strain, and predefined cutoff values for subclinical dysfunction were set at GLS < 18% and RV free-wall strain < 20%, respectively. RESULTS: RV free-wall strain in T2DM patients was significantly lower than that in normal controls (19.3% ± 4.8% vs. 24.4% ± 5.1%; P < 0.0001). RV free-wall strain in T2DM patients and LV longitudinal dysfunction was similar compared to that in T2DM patients without (19.0 ± 4.5% vs. 19.6 ± 5.0%, P = 0.40). Furthermore, multivariate logistic regression analyses showed that GLS was independently associated with RV systolic dysfunction as well as mitral inflow E and mitral e' annular velocities ratio (odds ratio, 1.16; 95% confidence interval: 1.03-1.31; P < 0.05). Sequential logistic models evaluating the association of RV systolic dysfunction in T2DM patients showed an improvement in clinical variables (χ2 = 6.2) with the addition of conventional echocardiographic parameters (χ2 = 13.4, P < 0.001) and a further improvement with the addition of GLS (χ2 = 20.8, P < 0.001). CONCLUSION: RV subclinical systolic dysfunction was observed in T2DM patients with preserved LVEF and was associated with LV longitudinal myocardial dysfunction. Our findings may provide additional findings for the management of T2DM patients.


Diabetes Mellitus, Type 2/complications , Diabetic Cardiomyopathies/etiology , Ventricular Dysfunction, Left/etiology , Ventricular Dysfunction, Right/etiology , Ventricular Function, Left , Ventricular Function, Right , Aged , Asymptomatic Diseases , Cross-Sectional Studies , Diabetes Mellitus, Type 2/diagnosis , Diabetic Cardiomyopathies/diagnostic imaging , Diabetic Cardiomyopathies/physiopathology , Echocardiography , Female , Humans , Male , Middle Aged , Prognosis , Retrospective Studies , Risk Assessment , Risk Factors , Ventricular Dysfunction, Left/diagnostic imaging , Ventricular Dysfunction, Left/physiopathology , Ventricular Dysfunction, Right/diagnostic imaging , Ventricular Dysfunction, Right/physiopathology
...