Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.468
Filter
1.
Crit Rev Biomed Eng ; 52(6): 15-31, 2024.
Article in English | MEDLINE | ID: mdl-39093445

ABSTRACT

Cardiovascular and cerebrovascular disease (CCVD) is a complex disease with a long latency period, and the most effective diagnosis and treatment methods are risk assessment and preventive interventions before onset. According to traditional Chinese medicine (TCM), Zhu-Ye-Qing wine (ZYQW) has the effect of invigorating blood and removing blood stasis. However, whether ZYQW can improve the progression of CCVD has not been reported. This study aims to explore the possible mechanism of ZYQW on CCVD through network pharmacology, and finally 249 potential targets of ZYQW and 2080 potential targets of CCVD are obtained. The key targets mainly include MAPK3, TP53, RELA, MAPK1 and AKT1. The main KEGG pathways include TNF signaling pathway, lipid and atherosclerosis pathway signaling pathway. The components in ZYQW are identified by ultra-performance liquid chromatography-mass spectrometry (UHPLC-CQE-CQE-MS/MS). Through network pharmacology, molecular docking and molecular dynamics simulation, the potential key components and prevention mechanisms of ZYQW in the prevention of CCVD are determined. ZYQW may be an effective and safe health food for the prevention of CCVD, providing guidance and basis for the further development of medicinal foods.


Subject(s)
Cardiovascular Diseases , Cerebrovascular Disorders , Network Pharmacology , Wine , Wine/analysis , Humans , Cerebrovascular Disorders/prevention & control , Cardiovascular Diseases/prevention & control , Cardiovascular Diseases/metabolism , Signal Transduction/drug effects , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/chemistry , Molecular Docking Simulation , Medicine, Chinese Traditional , Molecular Dynamics Simulation
2.
Int J Mol Sci ; 25(15)2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39125657

ABSTRACT

Well-controlled type 1 diabetes (T1DM) is characterized by inflammation and endothelial dysfunction, thus constituting a suitable model of subclinical cardiovascular disease (CVD). miR-199b-5p overexpression in murine CVD has shown proatherosclerotic effects. We hypothesized that miR-199b-5p would be overexpressed in subclinical CVD yet downregulated following metformin therapy. Inflammatory and vascular markers were measured in 29 individuals with T1DM and 20 matched healthy controls (HCs). miR-199b-5p expression in CFU-Hill's colonies was analyzed from each study group, and correlations with inflammatory/vascular health indices were evaluated. Significant upregulation of miR-199b-5p was observed in T1DM, which was significantly downregulated by metformin. miR-199b-5p correlated positively with vascular endothelial growth factor-D and c-reactive protein (CRP: nonsignificant). ROC analysis determined miR-199b-5p to define subclinical CVD by discriminating between HCs and T1DM individuals. ROC analyses of HbA1c and CRP showed that the upregulation of miR-199b-5p in T1DM individuals defined subclinical CVD at HbA1c > 44.25 mmol and CRP > 4.35 × 106 pg/mL. Ingenuity pathway analysis predicted miR-199b-5p to inhibit the target genes SIRT1, ETS1, and JAG1. Metformin was predicted to downregulate miR-199b-5p via NFATC2 and STAT3 and reverse its downstream effects. This study validated the antiangiogenic properties of miR-199b-5p and substantiated miR-199b-5p overexpression as a biomarker of subclinical CVD. The downregulation of miR-199b-5p by metformin confirmed its cardio-protective effect.


Subject(s)
Cardiovascular Diseases , Metformin , MicroRNAs , MicroRNAs/genetics , MicroRNAs/metabolism , Metformin/pharmacology , Metformin/therapeutic use , Humans , Male , Female , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Adult , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/metabolism , Inflammation/genetics , Inflammation/drug therapy , Inflammation/metabolism , Sirtuin 1/metabolism , Sirtuin 1/genetics , C-Reactive Protein/metabolism , C-Reactive Protein/genetics , Middle Aged , Gene Expression Regulation/drug effects , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Jagged-1 Protein/metabolism , Jagged-1 Protein/genetics , Biomarkers , Case-Control Studies
3.
Cell Commun Signal ; 22(1): 389, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39103830

ABSTRACT

Modern human society is burdened with the pandemic of cardiovascular and metabolic diseases. Metrnl is a widely distributed secreted protein in the body, involved in regulating glucose and lipid metabolism and maintaining cardiovascular system homeostasis. In this review, we present the predictive and therapeutic roles of Metrnl in various cardiovascular and metabolic diseases, including atherosclerosis, ischemic heart disease, cardiac remodeling, heart failure, hypertension, chemotherapy-induced myocardial injury, diabetes mellitus, and obesity.


Subject(s)
Biomarkers , Cardiovascular Diseases , Metabolic Diseases , Humans , Metabolic Diseases/metabolism , Metabolic Diseases/drug therapy , Cardiovascular Diseases/metabolism , Biomarkers/metabolism , Animals
4.
Front Endocrinol (Lausanne) ; 15: 1335371, 2024.
Article in English | MEDLINE | ID: mdl-39109081

ABSTRACT

Objective: We compared peripheral blood (PBL) chemokine ligand/receptor profiles in children and adolescents with type 1 diabetes mellitus (T1D) or obesity (OB) (both involving inflammation and vascular complications) to identify their associations with cardiometabolic risk factors. Materials and methods: PBL samples from children and adolescents (12-18 years) included: healthy controls (n=29), patients with T1D (n=31) and OB subjects (n=34). Frequency of mononuclear cell populations and chemokine receptor expression (CCR2, CCR4, CXCR3, CXCR4) were determined by flow cytometry. Chemokine levels of CCL2, CCL5, CXCL10 and CXCL11 were measured by bead-based assay and CXCL12 by ELISA. Data were correlated with cardiovascular, metabolic and inflammatory parameters. Results: The proportion of CD14+ monocytes was higher in T1D, whereas the proportion of CD19+ B lymphocytes was higher and CD3+ T lymphocytes was lower in OB. The level of CCL2 was higher in T1D (241.0 (IQR 189.6-295.3) pg/mL in T1D vs 191.5 (IQR 158.0-254.7) pg/mL in control, p=0.033), CXCL11 was lower in OB (6.6 (IQR 4.9-7.7) pg/mL in OB vs 8.2 (IQR 6.9-11.3) pg/mL in control, p=0.018) and CXCL12 was lower in both diseases (2.0 (IQR 1.8-2.5) ng/mL in T1D, 2.1 (IQR 1.9-2.4) ng/mL in OB vs 2.4 (IQR 2.2-2.5) ng/mL in control, p=0.016). Numerous significant associations were found for chemokine ligand/receptor profiles and clinical data. Among these, we are suggesting the most important indicators of cardiometabolic risk in T1D: positive associations of CCR2+ monocytes with blood pressure and CCL12 levels with urine albumin-to-creatinine ratio (ACR), inverse association of CXCR3+ B lymphocytes with AST but positive with triglycerides; and OB: positive associations of CXCL12 levels with triglycerides and AST/ALT, inverse association of CCR4+ and CXCR3+ monocytes with ACR. Both diseases share positive associations for CCR4+ T lymphocytes and blood pressure, inverse associations of CXCR4+ subsets with ACR and CXCR3+ T lymphocytes with lipid profile. Conclusion: Significantly changed chemokine ligand/receptor profiles were found in both T1D and OB even at a young age. Although different associations with cardiometabolic risk factors indicate disease-specific changes, overlapping pattern was found for the associations between CCR4+ T lymphocytes and vascular inflammation, CXCR4+ subsets and albuminuria as well as CXCR3+ T lymphocytes and dyslipidemia. Thus, chemokine axes might present potential therapeutic targets for disease-related morbidity.


Subject(s)
Chemokines , Diabetes Mellitus, Type 1 , Humans , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/metabolism , Adolescent , Child , Male , Female , Chemokines/blood , Chemokines/metabolism , Biomarkers/blood , Cardiometabolic Risk Factors , Obesity/metabolism , Obesity/blood , Obesity/complications , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/blood , Case-Control Studies , Pediatric Obesity/blood , Pediatric Obesity/metabolism
5.
Int J Mol Sci ; 25(15)2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39125842

ABSTRACT

Anderson-Fabry disease (AFD), a genetic disorder caused by mutations in the α-galactosidase-A (GLA) gene, disrupts lysosomal function, leading to vascular complications. The accumulation of globotriaosylceramide (Gb3) in arterial walls triggers upregulation of adhesion molecules, decreases endothelial nitric oxide synthesis, and induces reactive oxygen species production. This cascade results in fibrotic thickening, endothelial dysfunction, hypercontractility, vasospasm, and a pro-thrombotic phenotype. AFD patients display increased intima-media thickness (IMT) and reduced flow-mediated dilation (FMD), indicating heightened cardiovascular risk. Nailfold capillaroscopy (NFC) shows promise in diagnosing and monitoring microcirculatory disorders in AFD, though it remains underexplored. Morphological evidence of AFD as a storage disorder can be demonstrated through electron microscopy and immunodetection of Gb3. Secondary pathophysiological disturbances at cellular, tissue, and organ levels contribute to the clinical manifestations, with prominent lysosomal inclusions observed in vascular, cardiac, renal, and neuronal cells. Chronic accumulation of Gb3 represents a state of ongoing toxicity, leading to increased cell turnover, particularly in vascular endothelial cells. AFD-related vascular pathology includes increased renin-angiotensin system activation, endothelial dysfunction, and smooth muscle cell proliferation, resulting in IMT increase. Furthermore, microvascular alterations, such as atypical capillaries observed through NFC, suggest early microvascular involvement. This review aims to unravel the complex interplay between inflammation, oxidative stress, and endothelial dysfunction in AFD, highlighting the potential connections between metabolic disturbances, oxidative stress, inflammation, and fibrosis in vascular and cardiac complications. By exploring novel cardiovascular risk factors and potential diagnostic tools, we can advance our understanding of these mechanisms, which extend beyond sphingolipid accumulation to include other significant contributors to disease pathogenesis. This comprehensive approach can pave the way for innovative therapeutic strategies and improved patient outcomes.


Subject(s)
Endothelium, Vascular , Fabry Disease , Inflammation , Oxidative Stress , Fabry Disease/complications , Fabry Disease/metabolism , Fabry Disease/physiopathology , Fabry Disease/pathology , Humans , Inflammation/pathology , Inflammation/metabolism , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/etiology , Cardiovascular Diseases/pathology , Animals , alpha-Galactosidase/metabolism , Heart Disease Risk Factors
6.
Int J Mol Sci ; 25(15)2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39126035

ABSTRACT

Lipid metabolism is a critical component in preserving homeostasis and health, and lipids are significant chemicals involved in energy metabolism in living things. With the growing interest in lipid metabolism in recent years, an increasing number of studies have demonstrated the close relationship between abnormalities in lipid metabolism and the development of numerous human diseases, including cancer, cardiovascular, neurological, and endocrine system diseases. Thus, understanding how aberrant lipid metabolism contributes to the development of related diseases and how it works offers a theoretical foundation for treating and preventing related human diseases as well as new avenues for the targeted treatment of related diseases. Therefore, we discuss the processes of aberrant lipid metabolism in various human diseases in this review, including diseases of the cardiovascular system, neurodegenerative diseases, endocrine system diseases (such as obesity and type 2 diabetes mellitus), and other diseases including cancer.


Subject(s)
Lipid Metabolism , Neoplasms , Neurodegenerative Diseases , Humans , Neoplasms/metabolism , Neoplasms/etiology , Neoplasms/pathology , Animals , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/pathology , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/etiology , Obesity/metabolism , Endocrine System Diseases/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/pathology , Lipid Metabolism Disorders/metabolism
7.
J Cardiovasc Pharmacol ; 84(2): 125-135, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39115715

ABSTRACT

ABSTRACT: Aneurysms are localized dilations of blood vessels, which can expand to 50% of the original diameter. They are more common in cardiovascular and cerebrovascular vessels. Rupture is one of the most dangerous complications. The pathophysiology of aneurysms is complex and diverse, often associated with progressive vessel wall dysfunction resulting from vascular smooth muscle cell death and abnormal extracellular matrix synthesis and degradation. Multiple studies have shown that long noncoding RNAs (lncRNAs) play a significant role in the progression of cardiovascular and cerebrovascular diseases. Therefore, it is necessary to find and summarize them. LncRNAs control gene expression and disease progression by regulating target mRNA or miRNA and are biomarkers for the diagnosis and prognosis of aneurysmal cardiovascular and cerebrovascular diseases. This review explores the role, mechanism, and clinical value of lncRNAs in aneurysms, providing new insights for a deeper understanding of the pathogenesis of cardiovascular and cerebrovascular aneurysms.


Subject(s)
Intracranial Aneurysm , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Phenotype , RNA, Long Noncoding , Humans , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Intracranial Aneurysm/genetics , Intracranial Aneurysm/pathology , Intracranial Aneurysm/metabolism , Intracranial Aneurysm/physiopathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Animals , Gene Expression Regulation , Aneurysm/genetics , Aneurysm/pathology , Aneurysm/metabolism , Cardiovascular Diseases/genetics , Cardiovascular Diseases/pathology , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/physiopathology , Signal Transduction
8.
Nat Commun ; 15(1): 6919, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39134547

ABSTRACT

Serum response factor (SRF) controls gene transcription in vascular smooth muscle cells (VSMCs) and regulates VSMC phenotypic switch from a contractile to a synthetic state, which plays a key role in the pathogenesis of cardiovascular diseases (CVD). It is not known how post-translational SUMOylation regulates the SRF activity in CVD. Here we show that Senp1 deficiency in VSMCs increased SUMOylated SRF and the SRF-ELK complex, leading to augmented vascular remodeling and neointimal formation in mice. Mechanistically, SENP1 deficiency in VSMCs increases SRF SUMOylation at lysine 143, reducing SRF lysosomal localization concomitant with increased nuclear accumulation and switching a contractile phenotype-responsive SRF-myocardin complex to a synthetic phenotype-responsive SRF-ELK1 complex. SUMOylated SRF and phospho-ELK1 are increased in VSMCs from coronary arteries of CVD patients. Importantly, ELK inhibitor AZD6244 prevents the shift from SRF-myocardin to SRF-ELK complex, attenuating VSMC synthetic phenotypes and neointimal formation in Senp1-deficient mice. Therefore, targeting the SRF complex may have a therapeutic potential for the treatment of CVD.


Subject(s)
Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Nuclear Proteins , Phenotype , Serum Response Factor , Sumoylation , Vascular Remodeling , Animals , Humans , Male , Mice , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Cardiovascular Diseases/genetics , Cysteine Endopeptidases/metabolism , Cysteine Endopeptidases/genetics , ets-Domain Protein Elk-1/metabolism , ets-Domain Protein Elk-1/genetics , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Neointima/metabolism , Neointima/pathology , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Serum Response Factor/metabolism , Serum Response Factor/genetics , Trans-Activators/metabolism , Trans-Activators/genetics
9.
Redox Biol ; 75: 103297, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39127015

ABSTRACT

Cardiovascular disease (CVD) is one of the leading causes of mortality in humans, and oxidative stress plays a pivotal role in disease progression. This phenomenon typically arises from weakening of the cellular antioxidant system or excessive accumulation of peroxides. This review focuses on a specialized form of oxidative stress-disulfide stress-which is triggered by an imbalance in the glutaredoxin and thioredoxin antioxidant systems within the cell, leading to the accumulation of disulfide bonds. The genesis of disulfide stress is usually induced by extrinsic pathological factors that disrupt the thiol-dependent antioxidant system, manifesting as sustained glutathionylation of proteins, formation of abnormal intermolecular disulfide bonds between cysteine-rich proteins, or irreversible oxidation of thiol groups to sulfenic and sulfonic acids. Disulfide stress not only precipitates the collapse of the antioxidant system and the accumulation of reactive oxygen species, exacerbating oxidative stress, but may also initiate cellular inflammation, autophagy, and apoptosis through a cascade of signaling pathways. Furthermore, this review explores the detrimental effects of disulfide stress on the progression of various CVDs including atherosclerosis, hypertension, myocardial ischemia-reperfusion injury, diabetic cardiomyopathy, cardiac hypertrophy, and heart failure. This review also proposes several potential therapeutic avenues to improve the future treatment of CVDs.


Subject(s)
Cardiovascular Diseases , Disulfides , Oxidative Stress , Humans , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/etiology , Cardiovascular Diseases/pathology , Disulfides/metabolism , Animals , Reactive Oxygen Species/metabolism , Signal Transduction , Antioxidants/metabolism , Oxidation-Reduction , Thioredoxins/metabolism
10.
Mol Med Rep ; 30(4)2024 Oct.
Article in English | MEDLINE | ID: mdl-39129301

ABSTRACT

Chitinase­3 like­protein­1 (CHI3L1), a glycoprotein belonging to the glycoside hydrolase family 18, binds to chitin; however, this protein lacks chitinase activity. Although CHI3L1 is not an enzyme capable of degrading chitin, it plays significant roles in abnormal glucose and lipid metabolism, indicating its involvement in metabolic disorders. In addition, CHI3L1 is considered a key player in inflammatory diseases, with clinical data suggesting its potential as a predictor of cardiovascular disease. CHI3L1 regulates the inflammatory response of various cell types, including macrophages, vascular smooth muscle cells and fibroblasts. In addition, CHI3L1 participates in vascular remodeling and fibrosis, contributing to the pathogenesis of cardiovascular disease. At present, research is focused on elucidating the role of CHI3L1 in cardiovascular disease. The present systematic review was conducted to comprehensively evaluate the effects of CHI3L1 on cardiovascular cells, and determine the potential implications in the occurrence and progression of cardiovascular disease. The present study may further the understanding of the involvement of CHI3L1 in cardiovascular pathology, demonstrating its potential as a therapeutic target or biomarker in the management of cardiovascular disease.


Subject(s)
Biomarkers , Cardiovascular Diseases , Chitinase-3-Like Protein 1 , Humans , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Chitinase-3-Like Protein 1/metabolism , Animals , Fibroblasts/metabolism
11.
Int Immunopharmacol ; 138: 112622, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-38971111

ABSTRACT

Dyslipidemia, characterized by higher serum concentrations of low-density lipoprotein cholesterol (LDL-C), very low-density lipoprotein cholesterol (VLDL-C), triglyceride (TG), and lower serum concentrations of high-density lipoprotein cholesterol (HDL-C), is confirmed as a hallmark of cardiovascular diseases (CVD), posing serious risks to the future health of humans. Aside from the role of HDL-C concentrations, the capacity of cholesterol efflux to HDL is being identified as an enssential messurement for the dyslipidemic morbidity. Through inducing the progression of reverse cholesterol transport (RCT), the HDL-related cholesterol efflux plays a vital role in atherosclerotic plaque formation. In addition, increasing results demonstrated that the relationships between cholesterol efflux and cardiovascular events might be influenced by multiple factors, such as atherosclerosis, diabetes, and, inflammatory diseases. These risk factors could affect the intracellular composition of HDL, which might subsqently influence the cholesterol efflux process induced by HDL particle. In the present comprehensive article, we summarize the latest findings which described the modulatory roles of HDL in cardiometabolic disorders and inflammatory related diseases, focusing on its capacity in mediating cholesterol efflux. Moreover, the potential mechanisms whereby HDL regulate the risk of cardiometabolic disorders or inflammatory related diseases, at least partly, via cholesterol efflux pathway, are also well-listed.


Subject(s)
Cardiovascular Diseases , Inflammation , Humans , Animals , Cardiovascular Diseases/metabolism , Inflammation/metabolism , Cholesterol, HDL/metabolism , Cholesterol, HDL/blood , Cholesterol/metabolism , Cholesterol/blood , Biological Transport , Dyslipidemias/metabolism , Risk Factors , Lipoproteins, HDL/metabolism , Lipoproteins, HDL/blood
12.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 46(3): 435-443, 2024 Jun.
Article in Chinese | MEDLINE | ID: mdl-38953268

ABSTRACT

With the continuous development of identification technologies such as mass spectrometry,omics,and antibody technology,post-translational modification (PTM) has demonstrated increasing potential in medical research.PTM as a novel chemical modification method provides new perspectives for the research on diseases.Succinylation as a novel modification has aroused the interest of more and more researchers.The available studies about succinylation mainly focus on a desuccinylase named sirtuin 5.This enzyme plays a key role in modification and has been preliminarily explored in cardiovascular studies.This paper summarizes the influencing factors and regulatory roles of succinylation and the links between succinylation and other PTMs and reviews the research progress of PTMs in the cardiovascular field,aiming to deepen the understanding about the role of this modification and give new insights to the research in this field.


Subject(s)
Cardiovascular Diseases , Lysine , Protein Processing, Post-Translational , Cardiovascular Diseases/metabolism , Humans , Lysine/metabolism , Succinic Acid/metabolism
13.
Nat Commun ; 15(1): 5729, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38977723

ABSTRACT

Risk prediction for subsequent cardiovascular events remains an unmet clinical issue in patients with coronary artery disease. We aimed to investigate prognostic metabolic biomarkers by considering both shared and distinct metabolic disturbance associated with the composite and individual cardiovascular events. Here, we conducted an untargeted metabolomics analysis for 333 incident cardiovascular events and 333 matched controls. The cardiovascular events were designated as cardiovascular death, myocardial infarction/stroke and heart failure. A total of 23 shared differential metabolites were associated with the composite of cardiovascular events. The majority were middle and long chain acylcarnitines. Distinct metabolic patterns for individual events were revealed, and glycerophospholipids alteration was specific to heart failure. Notably, the addition of metabolites to clinical markers significantly improved heart failure risk prediction. This study highlights the potential significance of plasma metabolites on tailed risk assessment of cardiovascular events, and strengthens the understanding of the heterogenic mechanisms across different events.


Subject(s)
Biomarkers , Coronary Artery Disease , Metabolomics , Humans , Coronary Artery Disease/blood , Male , Female , Middle Aged , Aged , Biomarkers/blood , Myocardial Infarction/blood , Carnitine/blood , Carnitine/analogs & derivatives , Carnitine/metabolism , Heart Failure/blood , Heart Failure/metabolism , Prognosis , Risk Assessment , Case-Control Studies , Stroke/blood , Stroke/metabolism , Metabolome , Cardiovascular Diseases/blood , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/metabolism , Risk Factors
14.
Stem Cell Res Ther ; 15(1): 205, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38982541

ABSTRACT

Vascular tissue engineering is a promising approach for regenerating damaged blood vessels and developing new therapeutic approaches for heart disease treatment. To date, different sources of cells have been recognized that offer assistance within the recovery of heart supply routes and veins with distinctive capacities and are compelling for heart regeneration. However, some challenges still remain that need to be overcome to establish the full potential application of these cells. In this paper, we review the different cell sources used for vascular tissue engineering, focusing on extraembryonic tissue-derived cells (ESCs), and elucidate their roles in cardiovascular disease. In addition, we highlight the intricate interplay between mechanical and biochemical factors in regulating mesenchymal stem cell (MSC) differentiation, offering insights into optimizing their application in vascular tissues.


Subject(s)
Cell Differentiation , Mesenchymal Stem Cells , Regeneration , Tissue Engineering , Humans , Tissue Engineering/methods , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Regeneration/physiology , Animals , Blood Vessels/cytology , Blood Vessels/physiology , Blood Vessels/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Cardiovascular Diseases/therapy , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology
15.
Int J Biochem Cell Biol ; 173: 106616, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38992790

ABSTRACT

Extracellular vesicles (EVs) are important mediators of intercellular communication within the cardiovascular system, playing essential roles in physiological homeostasis and contributing to the pathogenesis of various cardiovascular diseases (CVDs). However, their potential as diagnostic biomarkers and therapeutic agents in rare cardiovascular diseases, such as valvular heart disease (VHD) and cardiomyopathies, remains largely unexplored. This review comprehensively emphasizes recent advancements in extracellular vesicle research, explicitly highlighting their growing significance in diagnosing and potentially treating rare cardiovascular diseases, with a particular focus on valvular heart disease and cardiomyopathies. We highlight the potential of extracellular vesicle-based liquid biopsies as non-invasive tools for early disease detection and risk stratification, showcasing specific extracellular vesicle-associated biomarkers (proteins, microRNAs, lipids) with diagnostic and prognostic value. Furthermore, we discussed the therapeutic promise of extracellular vesicles derived from various sources, including stem cells and engineered extracellular vesicles, for cardiac repair and regeneration through their ability to modulate inflammation, promote angiogenesis, and reduce fibrosis. By integrating the findings and addressing critical knowledge gaps, this review aims to stimulate further research and innovation in extracellular vesicle-based diagnostics and therapeutics of cardiovascular disease.


Subject(s)
Cardiovascular Diseases , Extracellular Vesicles , Humans , Extracellular Vesicles/metabolism , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/therapy , Cardiovascular Diseases/metabolism , Biomarkers/metabolism , Animals
16.
Int J Mol Sci ; 25(13)2024 Jul 02.
Article in English | MEDLINE | ID: mdl-39000380

ABSTRACT

Endothelial dysfunction often precedes the development of cardiovascular diseases, including heart failure. The cardioprotective benefits of sodium-glucose cotransporter 2 inhibitors (SGLT2is) could be explained by their favorable impact on the endothelium. In this review, we summarize the current knowledge on the direct in vitro effects of SGLT2is on endothelial cells, as well as the systematic observations in preclinical models. Four putative mechanisms are explored: oxidative stress, nitric oxide (NO)-mediated pathways, inflammation, and endothelial cell survival and proliferation. Both in vitro and in vivo studies suggest that SGLT2is share a class effect on attenuating reactive oxygen species (ROS) and on enhancing the NO bioavailability by increasing endothelial nitric oxide synthase activity and by reducing NO scavenging by ROS. Moreover, SGLT2is significantly suppress inflammation by preventing endothelial expression of adhesion receptors and pro-inflammatory chemokines in vivo, indicating another class effect for endothelial protection. However, in vitro studies have not consistently shown regulation of adhesion molecule expression by SGLT2is. While SGLT2is improve endothelial cell survival under cell death-inducing stimuli, their impact on angiogenesis remains uncertain. Further experimental studies are required to accurately determine the interplay among these mechanisms in various cardiovascular complications, including heart failure and acute myocardial infarction.


Subject(s)
Sodium-Glucose Transporter 2 Inhibitors , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Humans , Animals , Oxidative Stress/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Nitric Oxide/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/prevention & control , Reactive Oxygen Species/metabolism , Nitric Oxide Synthase Type III/metabolism , Inflammation/metabolism , Inflammation/drug therapy
17.
Ann Clin Lab Sci ; 54(3): 419-422, 2024 May.
Article in English | MEDLINE | ID: mdl-39048165

ABSTRACT

OBJECTIVE: Familial hypercholesterolemia (FH) is characterized by elevated levels of low-density lipoprotein cholesterol (LDL-C) and cardiovascular disease (CVD). Although the role of LDL-C in FH has been studied, the contribution of high-density lipoproteins (HDL) to CVD in FH remains unknown. This study aimed at highlighting the role of HDL in FH. METHODS: HDL-specific phospholipid efflux (HDL-SPE) assay was developed to predict CVD risk. HDL-SPE was examined in FH patients (n=30) and compared with age- and sex-matched non-FH controls (n=60). RESULTS: FH patients had significantly lower HDL-SPE levels (0.90±0.12) than controls (1.12±0.10; p<0.05), despite similar HDL-cholesterol levels in both groups (FH: 57.9±18.7 mg/dl; controls: 57.1±13.8 mg/dl). These differences remained significant after adjusting for confounders. CONCLUSIONS: These findings suggest there may be dysfunctionality of HDL in FH.


Subject(s)
Hyperlipoproteinemia Type II , Lipoproteins, HDL , Phospholipids , Humans , Hyperlipoproteinemia Type II/blood , Hyperlipoproteinemia Type II/metabolism , Male , Female , Lipoproteins, HDL/metabolism , Lipoproteins, HDL/blood , Adult , Phospholipids/metabolism , Phospholipids/blood , Middle Aged , Case-Control Studies , Cholesterol, HDL/blood , Cholesterol, HDL/metabolism , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/blood
18.
Biomolecules ; 14(7)2024 Jul 12.
Article in English | MEDLINE | ID: mdl-39062555

ABSTRACT

Affecting approximately 25% of the global population, steatotic liver disease (SLD) poses a significant health concern. SLD ranges from simple steatosis to metabolic dysfunction-associated steatohepatitis and fibrosis with a risk of severe liver complications such as cirrhosis and hepatocellular carcinoma. SLD is associated with obesity, atherogenic dyslipidaemia, and insulin resistance, increasing cardiovascular risks. As such, identifying SLD is vital for cardiovascular disease (CVD) prevention and treatment. Bile acids (BAs) have critical roles in lipid digestion and are signalling molecules regulating glucose and lipid metabolism and influencing gut microbiota balance. BAs have been identified as critical mediators in cardiovascular health, influencing vascular tone, cholesterol homeostasis, and inflammatory responses. The cardio-protective or harmful effects of BAs depend on their concentration and composition in circulation. The effects of certain BAs occur through the activation of a group of receptors, which reduce atherosclerosis and modulate cardiac functions. Thus, manipulating BA receptors could offer new avenues for treating not only liver diseases but also CVDs linked to metabolic dysfunctions. In conclusion, this review discusses the intricate interplay between BAs, metabolic pathways, and hepatic and extrahepatic diseases. We also highlight the necessity for further research to improve our understanding of how modifying BA characteristics affects or ameliorates disease.


Subject(s)
Bile Acids and Salts , Cardiovascular Diseases , Humans , Bile Acids and Salts/metabolism , Cardiovascular Diseases/metabolism , Animals , Fatty Liver/metabolism , Lipid Metabolism , Gastrointestinal Microbiome
19.
Life Sci ; 352: 122911, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39002609

ABSTRACT

The concept of "type 3 diabetes" has emerged to define alterations in glucose metabolism that predispose individuals to the development of Alzheimer's disease (AD). Novel evidence suggests that changes in the insulin/insulin-like growth factor 1 (IGF-1)/growth hormone (GH) axis, which are characteristic of Diabetes Mellitus, are one of the major factors contributing to excessive amyloid-beta (Aß) production and neurodegenerative processes in AD. Moreover, molecular findings suggest that insulin resistance and dysregulated IGF-1 signaling promote atherosclerosis via endothelial dysfunction and a pro-inflammatory state. As the pathophysiological role of Aß1-40 in patients with cardiovascular disease has attracted attention due to its involvement in plaque formation and destabilization, it is of great interest to explore whether a paradigm similar to that in AD exists in the cardiovascular field. Therefore, this review aims to elucidate the intricate interplay between insulin resistance, IGF-1, and Aß1-40 in the cardiovascular system and assess the applicability of the type 3 diabetes concept. Understanding these relationships may offer novel therapeutic targets and diagnostic strategies to mitigate cardiovascular risk in patients with insulin resistance and dysregulated IGF-1 signaling.


Subject(s)
Amyloid beta-Peptides , Cardiovascular Diseases , Insulin Resistance , Insulin-Like Growth Factor I , Peptide Fragments , Humans , Insulin Resistance/physiology , Insulin-Like Growth Factor I/metabolism , Amyloid beta-Peptides/metabolism , Cardiovascular Diseases/metabolism , Peptide Fragments/metabolism , Animals , Alzheimer Disease/metabolism
20.
Cell ; 187(15): 3880-3884, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39059364

ABSTRACT

The future of healthcare for cardiovascular diseases holds immense promise, not only based in new discoveries in cardiac metabolism but also in translating them to solutions for critical challenges faced by society. Here, ten scientists share their insights, shedding light on the future that lies ahead for this field.


Subject(s)
Cardiovascular Diseases , Humans , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/therapy , Translational Research, Biomedical , Animals
SELECTION OF CITATIONS
SEARCH DETAIL