Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.550
Filter
1.
Nat Cardiovasc Res ; 3(10): 1249-1265, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39402206

ABSTRACT

Evidence linking the endothelium to cardiac injury in long coronavirus disease (COVID) is well documented, but the underlying mechanisms remain unknown. Here we show that cytokines released by endothelial cells (ECs) contribute to long-COVID-associated cardiac dysfunction. Using thrombotic vascular tissues from patients with long COVID and induced pluripotent stem cell-derived ECs (iPSC-ECs), we modeled endotheliitis and observed similar dysfunction and cytokine upregulation, notably CCL2. Cardiac organoids comprising iPSC-ECs and iPSC-derived cardiomyocytes showed cardiac dysfunction after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) exposure, driven by CCL2. Profiling of chromatin accessibility and gene expression at a single-cell resolution linked CCL2 to 'phenotype switching' and cardiac dysfunction, validated by high-throughput proteomics. Disease modeling of cardiac organoids and exposure of human ACE2 transgenic mice to SARS-CoV-2 spike proteins revealed that CCL2-induced oxidative stress promoted post-translational modification of cardiac proteins, leading to cardiac dysfunction. These findings suggest that EC-released cytokines contribute to cardiac dysfunction in long COVID, highlighting the importance of early vascular health monitoring in patients with long COVID.


Subject(s)
Angiotensin-Converting Enzyme 2 , COVID-19 , Chemokine CCL2 , Endothelial Cells , SARS-CoV-2 , Humans , COVID-19/metabolism , COVID-19/complications , Animals , Angiotensin-Converting Enzyme 2/metabolism , Angiotensin-Converting Enzyme 2/genetics , Chemokine CCL2/metabolism , Chemokine CCL2/genetics , Endothelial Cells/metabolism , Endothelial Cells/virology , Endothelial Cells/pathology , Mice, Transgenic , Mice , Male , Female , Induced Pluripotent Stem Cells/metabolism , Oxidative Stress , Post-Acute COVID-19 Syndrome , Middle Aged , Aged , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/virology , Myocytes, Cardiac/pathology , Organoids/metabolism , Spike Glycoprotein, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/genetics , Heart Diseases/metabolism , Heart Diseases/pathology , Heart Diseases/virology , Heart Diseases/etiology
2.
Front Immunol ; 15: 1452678, 2024.
Article in English | MEDLINE | ID: mdl-39301029

ABSTRACT

Systemic lupus erythematosus (SLE) is a prevalent autoimmune disease primarily characterized by the involvement of multiple systems and organs. Cardiovascular disease is the primary cause of mortality in patients with SLE, though the mechanisms underlying the increased cardiovascular risk in SLE patients remain unclear. Recent studies indicate that abnormal activation of programmed cell death (PCD) signaling and the crosstalk among various forms of cell death are critical in the immunopathogenesis of SLE. Furthermore, apoptosis, necroptosis, pyroptosis, NETosis, and ferroptosis are recognized as key cellular processes in the pathogenesis of SLE and are closely linked to cardiac involvement. This review uniquely explores the intricate crosstalk between apoptosis, necroptosis, and other cell death pathways, discussing their roles and interactions in the pathogenesis of cardiac involvement in SLE. Investigating the interplay between PCD signaling and cardiac involvement in SLE in understanding the disease's underlying mechanisms and offers opportunities for new therapeutic interventions. The integration of precision medicine and innovative strategies targeting these complex pathways holds promise for enhancing the treatment prospects of SLE with cardiac involvement.


Subject(s)
Lupus Erythematosus, Systemic , Signal Transduction , Lupus Erythematosus, Systemic/immunology , Humans , Animals , Apoptosis , Cell Death , Heart Diseases/etiology , Heart Diseases/immunology , Heart Diseases/pathology , Necroptosis/immunology , Ferroptosis
3.
Biochem Biophys Res Commun ; 733: 150623, 2024 Nov 12.
Article in English | MEDLINE | ID: mdl-39255619

ABSTRACT

After prolonged space operations, astronauts showed maladaptive atrophy within mostly left-ventricular myocardium, resulting in cardiac dysfunction. However, the mechanism of cardiac dysfunction under microgravity conditions is unclear, and the relevant prevention and treatment measures also need to be explored. Through simulating the microgravity environment with a tail suspension (TS) model, we found that long-term exposure to microgravity promotes aging of mouse hearts, which is closely related to cardiac dysfunction. The intravenous administration of adipose-derived mesenchymal stem cells (ADSCs) emerged preventive and therapeutic effect against myocardial senescence and the decline in cardiac function. Plasma metabolomics analysis suggests the loss of NAD+ in TS mice and motivated myocardial NAD + metabolism and utilization in ADSCs-treated mice, likely accounting for ADSCs' function. Oral administration of nicotinamide mononucleotide (NMN, a NAD + precursor) showed similar therapeutic effect to ADSCs treatment. Collectively, these data implicate the effect of ADSCs in microgravity-induced cardiac dysfunction and provide new therapeutic ideas for aging-related maladaptive cardiac remodeling.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Mice, Inbred C57BL , Myocardium , NAD , Weightlessness , Animals , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , NAD/metabolism , Weightlessness/adverse effects , Myocardium/metabolism , Myocardium/pathology , Mice , Mesenchymal Stem Cell Transplantation/methods , Male , Nicotinamide Mononucleotide/pharmacology , Nicotinamide Mononucleotide/metabolism , Hindlimb Suspension/adverse effects , Aging/metabolism , Cellular Senescence/drug effects , Heart Diseases/metabolism , Heart Diseases/etiology , Heart Diseases/pathology , Heart Diseases/therapy , Heart Diseases/prevention & control
4.
Cardiovasc Toxicol ; 24(11): 1139-1150, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39240427

ABSTRACT

Doxorubicin (Dox) has been limited in clinical application due to its cardiac toxicity that varies with the dose. This study aimed to explore how Rhein modulates Dox-induced myocardial toxicity. The general condition and echocardiographic changes of mice were observed to evaluate cardiac function and structure, with myocardial cell injury and apoptosis checked by TUNEL and HE staining. The ELISA assessed markers of myocardial damage and inflammation. The TCMSP and SwissTargetPrediction databases were used to retrieve Rhein's targets while GeneCards was used to find genes related to Dox-induced myocardial injury. Intersection genes were analyzed by Protein-Protein Interaction Networks. The core network genes underwent GO and KEGG enrichment analysis using R software. Western blot was used to detect protein expression. Compared to the Dox group, there was no remarkable difference in heart mass /body mass ratio in the Rhein+Dox group. However, heart mass/tibia length increased. Mice in the Rhein+Dox group had significantly increased LVEF, LVPWs, and LVFS compared to those in the Dox group. Myocardial cell damage, inflammation, and apoptosis significantly reduced in the Rhein+Dox group compared to the model group. Eleven core network genes were selected. Further, Rhein+Dox group showed significantly downregulated expression of p38/p-p38, HSP90AA1, c-Jun/p-c-Jun, c-Fos/p-c-Fos, Bax, and cleaved-caspase-3/caspase-3 while Bcl-2 expression significantly upregulated compared to the Dox group. The study suggests that Rhein mediates cardioprotection against Dox-induced myocardial injury, at least partly, by influencing multiple core genes in the MAPK signaling pathway to inhibit myocardial cell apoptosis.


Subject(s)
Anthraquinones , Apoptosis , Cardiotoxicity , Disease Models, Animal , Doxorubicin , HSP90 Heat-Shock Proteins , Mice, Inbred C57BL , Myocytes, Cardiac , Proto-Oncogene Proteins c-fos , Proto-Oncogene Proteins c-jun , Signal Transduction , p38 Mitogen-Activated Protein Kinases , Animals , Apoptosis/drug effects , Anthraquinones/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Male , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Proto-Oncogene Proteins c-jun/genetics , HSP90 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/genetics , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-fos/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Ventricular Function, Left/drug effects , Mice , Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/genetics , Heart Diseases/chemically induced , Heart Diseases/prevention & control , Heart Diseases/pathology , Heart Diseases/metabolism , Protein Interaction Maps
5.
Int J Mol Sci ; 25(17)2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39273209

ABSTRACT

The endothelium is a cell monolayer that lines vessels and separates tissues from blood flow. Endothelial cells (ECs) have a multitude of functions, including regulating blood flow and systemic perfusion through changes in vessel diameter. When an injury occurs, the endothelium is affected by altering its functions and structure, which leads to endothelial dysfunction, a characteristic of many vascular diseases. Understanding the role that the endothelium plays in pulmonary vascular and cardiopulmonary diseases, and exploring new therapeutic strategies is of utmost importance to advance clinically. Currently, there are several treatments able to improve patients' quality of life, however, none are effective nor curative. This review examines the critical role of the endothelium in the pulmonary vasculature, investigating the alterations that occur in ECs and their consequences for blood vessels and potential molecular targets to regulate its alterations. Additionally, we delve into promising non-pharmacological therapeutic strategies, such as exercise and diet. The significance of the endothelium in cardiopulmonary disorders is increasingly being recognized, making ECs a relevant target for novel therapies aimed at preserving their functional and structural integrity.


Subject(s)
Endothelial Cells , Endothelium, Vascular , Humans , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Endothelial Cells/metabolism , Animals , Lung Diseases/pathology , Lung Diseases/therapy , Lung Diseases/metabolism , Lung Diseases/physiopathology , Heart Diseases/metabolism , Heart Diseases/therapy , Heart Diseases/pathology , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Cardiovascular Diseases/therapy
6.
Redox Biol ; 76: 103340, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39250857

ABSTRACT

Acute kidney injury (AKI) and chronic kidney disease (CKD) are global health burdens with rising prevalence. Their bidirectional relationship with cardiovascular dysfunction, manifesting as cardio-renal syndromes (CRS) types 3 and 4, underscores the interconnectedness and interdependence of these vital organ systems. Both the kidney and the heart are critically reliant on mitochondrial function. This organelle is currently recognized as a hub in signaling pathways, with emphasis on the redox regulation mediated by glutathione (GSH). Mitochondrial dysfunction, including impaired bioenergetics, redox, and biogenesis pathways, are central to the progression of AKI to CKD and the development of CRS type 3 and 4. This review delves into the metabolic reprogramming and mitochondrial redox signaling and biogenesis alterations in AKI, CKD, and CRS. We examine the pathophysiological mechanisms involving GSH redox signaling and the AMP-activated protein kinase (AMPK)-sirtuin (SIRT)1/3-peroxisome proliferator-activated receptor-gamma coactivator (PGC-1α) axis in these conditions. Additionally, we explore the therapeutic potential of GSH synthesis inducers in mitigating these mitochondrial dysfunctions, as well as their effects on inflammation and the progression of CKD and CRS types 3 and 4.


Subject(s)
Energy Metabolism , Glutathione , Mitochondria , Signal Transduction , Humans , Mitochondria/metabolism , Glutathione/metabolism , Animals , Oxidation-Reduction , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Organelle Biogenesis , Heart Diseases/metabolism , Heart Diseases/etiology , Heart Diseases/pathology , Oxidative Stress
7.
J Transl Med ; 22(1): 839, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39267173

ABSTRACT

BACKGROUND: Myocardial fibrosis, a hallmark of heart disease, is closely associated with macrophages, yet the genetic pathophysiology remains incompletely understood. In this study, we utilized integrated single-cell transcriptomics and bulk RNA-seq analysis to investigate the relationship between macrophages and myocardial fibrosis across omics integration. METHODS: We examined and curated existing single-cell data from dilated cardiomyopathy (DCM), ischemic cardiomyopathy (ICM), myocardial infarction (MI), and heart failure (HF), and analyzed the integrated data using cell communication, transcription factor identification, high dimensional weighted gene co-expression network analysis (hdWGCNA), and functional enrichment to elucidate the drivers of macrophage polarization and the macrophage-to-myofibroblast transition (MMT). Additionally, we assessed the accuracy of single-cell data from the perspective of driving factors, cell typing, anti-fibrosis performance of left ventricular assist device (LVAD). Candidate drugs were screened using L1000FWD. RESULTS: All four heart diseases exhibit myocardial fibrosis, with only MI showing an increase in macrophage proportions. Macrophages participate in myocardial fibrosis through various fibrogenic molecules, especially evident in DCM and MI. Abnormal RNA metabolism and dysregulated transcription are significant drivers of macrophage-mediated fibrosis. Furthermore, profibrotic macrophages exhibit M1 polarization and increased MMT. In HF patients, those responding to LVAD therapy showed a significant decrease in driver gene expression, M1 polarization, and MMT. Drug repurposing identified cinobufagin as a potential therapeutic agent. CONCLUSION: Using integrated single-cell transcriptomics, we identified the drivers of macrophage-mediated myocardial fibrosis in four heart diseases and confirmed the therapeutic effect of LVAD on improving HF with single-cell accuracy, providing novel insights into the diagnosis and treatment of myocardial fibrosis.


Subject(s)
Fibrosis , Heart Diseases , Macrophages , Humans , Macrophages/metabolism , Heart Diseases/genetics , Heart Diseases/pathology , Single-Cell Analysis , Gene Regulatory Networks , Myocardium/pathology , Gene Expression Regulation , Genomics , Gene Expression Profiling
8.
Biomed Pharmacother ; 179: 117312, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39167843

ABSTRACT

Anthracyclines are broad-spectrum anticancer drugs, but their clinical use is limited due to their severe cardiotoxicity. Anthracycline-induced cardiotoxicity (AIC) remains a significant cause of heart disease-related mortality in many cancer survivors. The underlying mechanisms of AIC have been explored over the past few decades. Reactive oxygen species and drug-induced inhibition of topoisomerase II beta are well-studied mechanisms, with mitochondria being a prominently investigated organelle. Emerging mechanisms such as ferroptosis, Ca2+ overload, autophagy and inflammation mediators have been implicated in recent years. In this review, our goal is to summarize and update the roles of various mechanisms in AIC, focusing on different cellular levels and further explore promising therapeutic approaches targeting these organelles or pathways.


Subject(s)
Anthracyclines , Cardiotoxicity , Humans , Anthracyclines/adverse effects , Cardiotoxicity/etiology , Animals , Reactive Oxygen Species/metabolism , Autophagy/drug effects , Heart Diseases/chemically induced , Heart Diseases/pathology , Heart Diseases/metabolism , Antibiotics, Antineoplastic/adverse effects , Ferroptosis/drug effects , Antineoplastic Agents/adverse effects
9.
Cardiovasc Toxicol ; 24(11): 1287-1300, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39212842

ABSTRACT

Cardiovascular diseases are a rampant public health threat. Environmental contaminants, such as Cadmium (Cd), a toxic metal, are risk factors for cardiovascular diseases. Given that human exposure to Cd is increasing, there is a need for therapies to ameliorate Cd toxicity. Selenium (Se), an essential trace element, has been proposed to rescue the effects of Cd toxicity, with mixed effects. Se's narrow therapeutic window necessitates precise dosing to avoid toxicity. Here, we assessed the effects of various waterborne Cd and Se concentrations and sequences on cardiac function using zebrafish (Danio rerio). We showed that Cd induced pericardial edemas and modified heart rates in zebrafish larvae in a concentration-dependent manner. To identify the therapeutic range of Se for Cd-induced cardiotoxicity, zebrafish embryos were treated with 0, 10, 50, 100, 150, or 200 µg/L Se for 1-4 days prior to exposure to 2.5 and 5 µg/L Cd. We found that a 50 µg/L Se pre-treatment before 2.5 µg/L Cd, but not 5 µg/L Cd, reduced the prevalence of pericardial edemas and ameliorated Cd-induced bradycardia in zebrafish. Zebrafish exposed to 10 and 50 µg/L of Se for up to 4 days showed typical heart morphology, whereas other Se-exposed and control fish presented pericardial edemas. Longer Se pre-treatment durations led to fewer incidences of pericardial edemas. Overall, this study highlights the importance of optimizing Se concentrations and pre-treatment periods to harness its protective effects against Cd-induced cardiotoxicity. These findings provide insights into potential therapeutic strategies for reducing Cd-related cardiovascular damage in humans.


Subject(s)
Cardiotoxicity , Heart Diseases , Heart Rate , Zebrafish , Animals , Heart Rate/drug effects , Heart Diseases/chemically induced , Heart Diseases/prevention & control , Heart Diseases/metabolism , Heart Diseases/pathology , Heart Diseases/physiopathology , Dose-Response Relationship, Drug , Water Pollutants, Chemical/toxicity , Disease Models, Animal , Cadmium/toxicity , Selenium/pharmacology , Cytoprotection
10.
Biomed Pharmacother ; 178: 117287, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39137652

ABSTRACT

This study investigates the effects of inositol (INO) supplementation on cardiac changes caused by Li in mice. The study involved 4 groups of C57BL6 mice (n=10 each): (i) mice orally administered with Li2CO3 for 8 weeks, then 4 additional weeks without (Li_group) or (ii) with INO supplementation (Li_INOdelayed_group) (total of 12 weeks); (iii) mice given Li2CO3 and INO supplementation concurrently for 12 weeks (Li+INO_group); (iv) one group left untreated (C-group). The INO was administered as a mixture of myo-inositol and d-chiro-inositol (80:1) in drinking water. The mice were characterised for heart morphology, function, electrical activity, arrhythmogenic susceptibility, and multiorgan histopathology (heart, liver and kidney). Cardiomyocyte size, protein expression of key signalling pathways related to hypertrophy, and transcription levels of ion channel subunits and hypertrophy markers were evaluated in the ventricle tissue. The study found that INO supplementation reduced the Li-induced cardiac adverse effects, including systolic impairment and increased susceptibility to arrhythmias. The positive effect on arrhythmias might be attributed to the restored expression levels of the potassium channel subunit Kv 1.5. Additionally, INO improved cardiomyocyte hypertrophy, possibly by inhibiting the Li-induced activation of the ERK1/2 signalling pathway and by restoring the normal expression level of BNP, and alleviated injury in the liver and kidney. The effect was preventive if INO supplementation was taken concurrently with Li and therapeutic if INO was administered after Li-induced cardiac impairments were established. These results provide new insights into the cardioprotective effect of INO and suggest a potential treatment approach for Li-induced cardiac disease.


Subject(s)
Dietary Supplements , Inositol , Mice, Inbred C57BL , Animals , Male , Mice , Administration, Oral , Inositol/pharmacology , Inositol/administration & dosage , Lithium/administration & dosage , Lithium/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Myocytes, Cardiac/metabolism , Arrhythmias, Cardiac/chemically induced , Arrhythmias, Cardiac/prevention & control , Arrhythmias, Cardiac/drug therapy , Heart Diseases/chemically induced , Heart Diseases/prevention & control , Heart Diseases/pathology , Heart Diseases/drug therapy
11.
Biochim Biophys Acta Mol Basis Dis ; 1870(8): 167487, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39216649

ABSTRACT

Myopathy leads to skeletal and cardiac muscle degeneration which is a major cause of physical disability and heart failure. Despite the therapeutic advancement the prevalence of particularly cardiac diseases is rising at an alarming rate and novel therapeutic targets are required. Nicotinamide riboside kinase-2 (NRK-2 or NMRK2) is a muscle-specific ß1-integrin binding protein abundantly expressed in the skeletal muscle while only a trace amount is detected in the healthy cardiac muscle. The level in cardiac tissue is profoundly upregulated under pathogenic conditions such as ischemia and hypertension. NRK-2 was initially identified to regulate myoblast differentiation and to enhance the levels of NAD+, an important coenzyme that potentiates cellular energy production and stress resilience. Recent advancement has shown that NRK-2 critically regulates numerous cellular and molecular processes under pathogenic conditions to modulate the disease severity. Therefore, given its restricted expression in the cardiac and skeletal muscle, NRK-2 may serve as a unique therapeutic target. In this review, we provided a comprehensive overview of the diverse roles of NRK-2 played in different cardiac and muscular diseases and discussed the underlying molecular mechanisms in detail. Moreover, this review precisely examined how NRK-2 regulates metabolism in cardiac muscle, and how dysfunctional NRK-2 is associated with energetic deficit and impaired muscle function, manifesting various cardiac and skeletal muscle disease conditions.


Subject(s)
Muscle, Skeletal , Phosphotransferases (Alcohol Group Acceptor) , Humans , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Animals , Myocardium/metabolism , Myocardium/pathology , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Muscular Diseases/metabolism , Muscular Diseases/pathology , Muscular Diseases/genetics , Heart Diseases/metabolism , Heart Diseases/pathology
12.
Vascul Pharmacol ; 155: 107324, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38985581

ABSTRACT

Doxorubicin (DOX) is a highly effective chemotherapeutic agent whose clinical use is hindered by the onset of cardiotoxic effects, resulting in reduced ejection fraction within the first year from treatment initiation. Recently it has been demonstrated that DOX accumulates within mitochondria, leading to disruption of metabolic processes and energetic imbalance. We previously described that phosphoinositide 3-kinase γ (PI3Kγ) contributes to DOX-induced cardiotoxicity, causing autophagy inhibition and accumulation of damaged mitochondria. Here we intend to describe the maladaptive metabolic rewiring occurring in DOX-treated hearts and the contribution of PI3Kγ signalling to this process. Metabolomic analysis of DOX-treated WT hearts revealed an accumulation of TCA cycle metabolites due to a cycle slowdown, with reduced levels of pyruvate, unchanged abundance of lactate and increased Acetyl-CoA production. Moreover, the activity of glycolytic enzymes was upregulated, and fatty acid oxidation downregulated, after DOX, indicative of increased glucose oxidation. In agreement, oxygen consumption was increased in after pyruvate supplementation, with the formation of cytotoxic ROS rather than energy production. These metabolic changes were fully prevented in KD hearts. Interestingly, they failed to increase glucose oxidation in response to DOX even with autophagy inhibition, indicating that PI3Kγ likely controls the fuel preference after DOX through an autophagy-independent mechanism. In vitro experiments showed that inhibition of PI3Kγ inhibits pyruvate dehydrogenase (PDH), the key enzyme of Randle cycle regulating the switch from fatty acids to glucose usage, while decreasing DOX-induced mobilization of GLUT-4-carrying vesicles to the plasma membrane and limiting the ensuing glucose uptake. These results demonstrate that PI3Kγ promotes a maladaptive metabolic rewiring in DOX-treated hearts, through a two-pronged mechanism controlling PDH activation and GLUT-4-mediated glucose uptake.


Subject(s)
Cardiotoxicity , Doxorubicin , Energy Metabolism , Fatty Acids , Glucose , Oxidation-Reduction , Animals , Doxorubicin/toxicity , Glucose/metabolism , Fatty Acids/metabolism , Energy Metabolism/drug effects , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Glycolysis/drug effects , Autophagy/drug effects , Male , Signal Transduction/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Citric Acid Cycle/drug effects , Mice, Inbred C57BL , Heart Diseases/chemically induced , Heart Diseases/metabolism , Heart Diseases/pathology , Heart Diseases/prevention & control , Heart Diseases/physiopathology , Mitochondria, Heart/metabolism , Mitochondria, Heart/drug effects , Mitochondria, Heart/pathology , Mitochondria, Heart/enzymology , Mice, Knockout , Disease Models, Animal , Reactive Oxygen Species/metabolism , Glucose Transporter Type 4/metabolism , Antibiotics, Antineoplastic/toxicity , Antibiotics, Antineoplastic/adverse effects
14.
J Biochem Mol Toxicol ; 38(8): e23783, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39056209

ABSTRACT

Cyclophosphamide (CYP) is widely used to treat various types of cancer. In addition to the therapeutic properties of this drug, unfortunately, its side effects are still not fully understood. This study investigated the protective effect of curcumin (CURC) and berberine (BER) on CYP-induced cardiac damage. Thirty-six male rats were equally divided into the control, dimethyl sulfoxide (DMSO), CYP, CYP + CURC, CYP + BER and CYP + BER + CURC groups. Troponin-I, Creatine kinase-myocardial band (CK-MB), total cholesterol, triglyceride levels in serum samples, and reactive oxygen species (ROS), poly(ADP-ribose) polymerase-1 (PARP-1), and transient receptor potential melastatin 2 (TRPM2) channel levels in heart tissue were measured using an enzyme-linked immunoassay (ELISA) kit. In addition, histopathological examination and immunohistochemical investigation of the TRPM2 channel, fibroblast specific protein-1 (FSP1), transforming growth factor-beta- 1 (TGF-ß1) and α-smooth muscle actin (α-SMA) expressions were determined in heart tissue. The CYP group's troponin-I, total cholesterol, triglyceride, CK-MB, ROS, PARP-1 and TRPM2 channel levels were higher than in the other groups in the ELISA measurements (p < 0.05). In contrast, these parameters in the group treated with CURC and BER together with CYP were lower than in the CYP group (p < 0.05). Additionally, CUR and BER reduced CYP-induced pathological damage, TRPM2, FSP1, TGF-ß1 and α-SMA expressions. The data showed that CYP administration can cause cardiac damage by increasing the TRPM2 channel, TGF-ß1, FSP1 and α-SMA expression levels. Therefore, we concluded that CURC and BER administration following CYP application may be used as therapeutic agents to prevent CYP-induced cardiac damage.


Subject(s)
Berberine , Curcumin , Cyclophosphamide , Fibrosis , Myocardium , TRPM Cation Channels , Animals , TRPM Cation Channels/metabolism , Cyclophosphamide/toxicity , Cyclophosphamide/adverse effects , Male , Rats , Curcumin/pharmacology , Berberine/pharmacology , Myocardium/metabolism , Myocardium/pathology , Biomarkers/metabolism , Biomarkers/blood , Lipids/blood , Rats, Wistar , Heart Diseases/chemically induced , Heart Diseases/metabolism , Heart Diseases/pathology , Heart Diseases/prevention & control , Heart Diseases/drug therapy
15.
Stem Cell Res Ther ; 15(1): 219, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39026298

ABSTRACT

Epicardial epithelial-to-mesenchymal transition (EMT) plays a pivotal role in both heart development and injury response and involves dynamic cellular changes that are essential for cardiogenesis and myocardial repair. Specifically, epicardial EMT is a crucial process in which epicardial cells lose polarity, migrate into the myocardium, and differentiate into various cardiac cell types during development and repair. Importantly, following EMT, the epicardium becomes a source of paracrine factors that support cardiac growth at the last stages of cardiogenesis and contribute to cardiac remodeling after injury. As such, EMT seems to represent a fundamental step in cardiac repair. Nevertheless, endogenous EMT alone is insufficient to stimulate adequate repair. Redirecting and amplifying epicardial EMT pathways offers promising avenues for the development of innovative therapeutic strategies and treatment approaches for heart disease. In this review, we present a synthesis of recent literature highlighting the significance of epicardial EMT reactivation in adult heart disease patients.


Subject(s)
Epithelial-Mesenchymal Transition , Pericardium , Humans , Pericardium/metabolism , Pericardium/cytology , Animals , Heart Diseases/metabolism , Heart Diseases/pathology , Heart Diseases/therapy , Myocardium/metabolism , Myocardium/pathology , Cell Differentiation
16.
Int J Mol Sci ; 25(14)2024 Jul 12.
Article in English | MEDLINE | ID: mdl-39062885

ABSTRACT

BACKGROUND: Mitochondria play a crucial role in adapting to fluctuating energy demands, particularly in various heart diseases. This study investigates mitochondrial morphology near intercalated discs in left ventricular (LV) heart tissues, comparing samples from patients with sinus rhythm (SR), atrial fibrillation (AF), dilated cardiomyopathy (DCM), and ischemic cardiomyopathy (ICM). METHODS: Transmission electron microscopy was used to analyze mitochondria within 0-3.5 µm and 3.5-7 µm of intercalated discs in 9 SR, 10 AF, 9 DCM, and 8 ICM patient samples. Parameters included mean size in µm2 and elongation, count, percental mitochondrial area in the measuring frame, and a conglomeration score. RESULTS: AF patients exhibited higher counts of small mitochondria in the LV myocardium, resembling SR. DCM and ICM groups had fewer, larger, and often hydropic mitochondria. Accumulation rates and percental mitochondrial area were similar across groups. Significant positive correlations existed between other defects/size and hydropic mitochondria and between count/area and conglomeration score, while negative correlations between count and size/other defects and between hydropic mitochondria and count could be seen as well. CONCLUSION: Mitochondrial parameters in the LV myocardium of AF patients were similar to those of SR patients, while DCM and ICM displayed distinct changes, including a decrease in number, an increase in size, and compromised mitochondrial morphology. Further research is needed to fully elucidate the pathophysiological role of mitochondrial morphology in different heart diseases, providing deeper insights into potential therapeutic targets and interventions.


Subject(s)
Mitochondria, Heart , Humans , Male , Female , Pilot Projects , Middle Aged , Aged , Mitochondria, Heart/metabolism , Mitochondria, Heart/ultrastructure , Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Dilated/metabolism , Heart Diseases/metabolism , Heart Diseases/pathology , Microscopy, Electron, Transmission , Adult , Heart Ventricles/pathology , Heart Ventricles/metabolism , Heart Ventricles/ultrastructure , Atrial Fibrillation/metabolism , Atrial Fibrillation/pathology , Atrial Fibrillation/physiopathology , Myocardium/metabolism , Myocardium/pathology , Myocardium/ultrastructure
17.
J Ethnopharmacol ; 334: 118582, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39009325

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Radiation-induced heart disease (RIHD) is one of the most serious complications in patients receiving chest radiotherapy, partially offsetting its benefits. At present, there is a lack of effective treatments for RIHD. Ferroptosis is a newly discovered type of cell death that results from iron-dependent lipid peroxide accumulation. It was recently shown that irradiation generates severe ferroptosis, providing new insights for the treatment of RIHD. Abelmoschus manihot (L.) possesses excellent pharmacological properties and is widely used in treating various ischemic heart and brain diseases; however, its efficacy and mechanism in treating RIHD are unknown. AIM: This study aimed to investigate the efficacy and mechanism of total extracts from A. manihot (L.) (TEA) in treating RIHD. MATERIALS AND METHODS: C57BL/6 mice and H9C2 cells were exposed to irradiation to induce RIHD in vivo and in vitro, respectively. In vivo, we evaluated the protective effects of TEA (150 and 300 mg/kg) on RIHD. Body and heart weight changes of mice were calculated in each group, and malondialdehyde (MDA) level, glutathione/oxidized glutathione (GSH/GSSH) and nicotinamide adenine dinucleotide phosphate (NADPH/NADP+) ratios, western blot, heart histology, and immunohistochemistry were used to evaluate TEA effectiveness. We identified the potential mechanism of radiation-induced cardiomyocyte injury in H9C2 cells treated with small interfering RNA. We determined the effective dose of TEA (0.6 mg/mL) using a Cell Counting Kit-8 assay. Intracellular Fe2+ and lipid peroxidation levels were detected by Phen Green™ SK diacetate probe, BODIPY 581/591 C11 staining, and MDA, GSH, and NADPH kits, and the level of target protein was evaluated by immunofluorescence and western blot. RESULTS: Radiation inhibited system Xc-cystine (xCT)/glutathione peroxidase 4 (GPX4) expression and activity in cardiomyocytes in a time and dose-dependent manner. After silencing xCT/GPX4, MDA significantly increased and GSH/GSSH and NADPH/NADP+ ratios were reduced. xCT/GPX4 inhibition drove ferroptosis in radiation-induced H9C2 injury. Oxidative stress in H9C2 was significantly enhanced by irradiation, which also significantly increased NADPH oxidase 4 (NOX4) expression and inhibited nuclear factor E2-related factor 2 (Nrf2) expression in vivo and in vitro. Inhibition of xCT/GPX4 drove ferroptosis in radiation-induced H9C2 injury, which was aggravated by inactivation of Nrf2 and alleviated by inhibition of NOX4. Compared with the ionizing radiation-only group, TEA improved body weight loss, MDA levels, and histological changes induced by irradiation in mice hearts, and increased the ratio of GSH/GSSH and NADPH/NADP+in vivo; it also reduced lipid peroxidation and intracellular Fe2+ accumulation, restored MDA levels, and elevated the ratios of GSH/GSSH and NADPH/NADP+ in irradiation-injured H9C2 cells. TEA up-regulated Nrf2, xCT, and GPX4 expression and inhibited NOX4 expression in vivo and in vitro. CONCLUSIONS: Ferroptosis induced by redox imbalance mediated through the NOX4/xCT/GPX4 axis is a potential mechanism behind radiation-induced cardiomyocyte injury, and can be prevented by TEA.


Subject(s)
Abelmoschus , Ferroptosis , Mice, Inbred C57BL , Myocytes, Cardiac , NADPH Oxidase 4 , Phospholipid Hydroperoxide Glutathione Peroxidase , Plant Extracts , Animals , Ferroptosis/drug effects , Ferroptosis/radiation effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/radiation effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Plant Extracts/pharmacology , Mice , Male , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Abelmoschus/chemistry , NADPH Oxidase 4/metabolism , Cell Line , Oxidation-Reduction/drug effects , Rats , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Heart Diseases/prevention & control , Heart Diseases/etiology , Heart Diseases/pathology
18.
J Clin Invest ; 134(13)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38949027

ABSTRACT

Biological sex is an important modifier of physiology and influences pathobiology in many diseases. While heart disease is the number one cause of death worldwide in both men and women, sex differences exist at the organ and cellular scales, affecting clinical presentation, diagnosis, and treatment. In this Review, we highlight baseline sex differences in cardiac structure, function, and cellular signaling and discuss the contribution of sex hormones and chromosomes to these characteristics. The heart is a remarkably plastic organ and rapidly responds to physiological and pathological cues by modifying form and function. The nature and extent of cardiac remodeling in response to these stimuli are often dependent on biological sex. We discuss organ- and molecular-level sex differences in adaptive physiological remodeling and pathological cardiac remodeling from pressure and volume overload, ischemia, and genetic heart disease. Finally, we offer a perspective on key future directions for research into cardiac sex differences.


Subject(s)
Sex Characteristics , Ventricular Remodeling , Humans , Female , Male , Animals , Heart Diseases/pathology , Heart Diseases/metabolism , Heart Diseases/physiopathology , Heart Diseases/genetics , Gonadal Steroid Hormones/metabolism , Heart/physiopathology , Heart/physiology , Myocardium/pathology , Myocardium/metabolism
19.
Cardiovasc Pathol ; 73: 107670, 2024.
Article in English | MEDLINE | ID: mdl-38880163

ABSTRACT

Electron microscopy (EM) was a popular diagnostic tool in the 1970s and early 80s. With the adoption of newer, less expensive techniques, such as immunohistochemistry, the role of EM in diagnostic surgical pathology has dwindled substantially. Nowadays, even in academic centers, EM interpretation is relegated to renal pathologists and the handful of (aging) pathologists with experience using the technique. As such, EM interpretation is truly arcane-understood by few and mysterious to many. Nevertheless, there remain situations in which EM is the best or only ancillary test to ascertain a specific diagnosis. Thus, there remains a critical need for the younger generation of surgical pathologists to learn EM interpretation. Recognizing this need, cardiac EM was made the theme of the Cardiovascular Evening Specialty Conference at the 2023 United States and Canadian Academy of Pathology (USCAP) annual meeting in New Orleans, Louisiana. Each of the speakers contributed their part to this article, the purpose of which is to review EM as it pertains to myocardial tissue and provide illustrative examples of the spectrum of ultrastructural cardiac pathology seen in storage/metabolic diseases, cardiomyopathies, infiltrative disorders, and cardiotoxicities.


Subject(s)
Heart Diseases , Microscopy, Electron , Myocardium , Humans , Myocardium/pathology , Myocardium/ultrastructure , Heart Diseases/pathology , Predictive Value of Tests , Prognosis , Animals
20.
Cardiovasc Pathol ; 73: 107671, 2024.
Article in English | MEDLINE | ID: mdl-38906439

ABSTRACT

The potential of the pericardial space as a therapeutic delivery tool for cardiac fibrosis and heart failure (HF) treatment has yet to be elucidated. Recently, miRNAs and exosomes have been discovered to be present in human pericardial fluid (PF). Novel studies have shown characteristic human PF miRNA compositions associated with cardiac diseases and higher miRNA expressions in PF compared to peripheral blood. Five key studies found differentially expressed miRNAs in HF, angina pectoris, aortic stenosis, ventricular tachycardia, and congenital heart diseases with either atrial fibrillation or sinus rhythm. As miRNA-based therapeutics for cardiac fibrosis and HF showed promising results in several in vivo studies for multiple miRNAs, we hypothesize a potential role of miRNA-based therapeutics delivered through the pericardial cavity. This is underlined by the favorable results of the first phase 1b clinical trial in this emerging field. Presenting the first human miRNA antisense drug trial, inhibition of miR-132 by intravenous administration of a novel antisense oligonucleotide, CDR132L, established efficacy in reducing miR-132 in plasma samples in a dose-dependent manner. We screened the literature, provided an overview of the miRNAs and exosomes present in PF, and drew a connection to those miRNAs previously elucidated in cardiac fibrosis and HF. Further, we speculate about clinical implications and potential delivery methods.


Subject(s)
Exosomes , Fibrosis , MicroRNAs , Humans , Exosomes/genetics , Exosomes/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , Myocardium/pathology , Myocardium/metabolism , Heart Diseases/genetics , Heart Diseases/pathology , Heart Diseases/metabolism , Pericardial Fluid/metabolism , Pericardium/metabolism , Pericardium/pathology
SELECTION OF CITATIONS
SEARCH DETAIL