Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.316
Filter
1.
J Ethnopharmacol ; 336: 118751, 2025 Jan 10.
Article in English | MEDLINE | ID: mdl-39214192

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Huachansu Capsule (HCSc) is a simple enteric-coated capsule refined from the skin of the dried toad, a traditional medicinal herb. It has been used clinically for many years to treat a variety of malignant tumors with remarkable efficacy. To date, a number of main components of HCSc have been reported to be cardiotoxic, but the specific mechanism of cardiotoxicity is still unknown. AIM OF THE STUDY: The aim of this study was to elucidate the possible cardiotoxic symptoms caused by high-doses of HCSc and to further reveal the complex mechanisms by which it causes cardiotoxicity. MATERIALS AND METHODS: UPLC-Q-Exactive Orbitrap MS and network toxicology were used to identify and predict the potential toxic components, related signaling pathways. Then, we used acute and sub-acute toxicity experiments to reveal the apparent phenomenon of HCSc-induced cardiotoxicity. Finally, we combined transcriptomics and metabolomics to elucidate the potential mechanism of action, and verified the putative mechanism by molecular docking, RT-qPCR, and Western blot. RESULTS: We found 8 toad bufadienolides components may be induced cardiac toxicity HCSc main toxic components. Through toxicity experiments, we found that high dose of HCSc could increase a variety of blood routine indexes, five cardiac enzymes, heart failure indexes (BNP), troponin (cTnI and cTnT), heart rate and the degree of heart tissue damage, while low-dose of HCSc had no such changes. In addition, by molecular docking, found that 8 kinds of main toxic components and cAMP, AMPK, IL1ß, mTOR all can be a very good combination, especially in the cAMP. Meanwhile, RT-qPCR and Western blot results showed that HCSc could induce cardiotoxicity by regulating a variety of heart-related differential genes and activating the cAMP signaling pathway. CONCLUSIONS: In this study, network toxicology, transcriptomics and metabolomics were used to elucidate the complex mechanism of possible cardiotoxicity induced by high-dose HCSc. Animal experiments, molecular docking, Western blot and RT-qPCR experiments were also used to verify the above mechanism. These findings will inform further mechanistic studies and provide theoretical support for its safe clinical application.


Subject(s)
Cardiotoxicity , Metabolomics , Transcriptome , Animals , Metabolomics/methods , Male , Transcriptome/drug effects , Rats , Bufanolides/toxicity , Molecular Docking Simulation , Rats, Sprague-Dawley , Network Pharmacology , Capsules , Signal Transduction/drug effects , Gene Expression Profiling/methods , Anura
2.
BMC Cardiovasc Disord ; 24(1): 505, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39300335

ABSTRACT

BACKGROUND: The overall mortality and morbidity benefit in patients with heart failure with a reduced ejection fraction is greatest with a treatment combination of sacubitril/valsartan, beta-blockers, mineralocorticoid-receptor antagonists, and sodium-glucose transporter-2 inhibitors, termed the "fantastic four" or "quadruple therapy." The addition of vericiguat (an oral soluble guanylate cyclase stimulator) is believed to aid in managing worsening heart failure after quadruple therapy. Among childhood and young adult cancer survivors, cardiovascular complications that develop more than 10 years after anthracycline-based chemotherapy have a poor prognosis. Therefore, this study reports the efficacy of multidrug regimen based on quadruple therapy for worsening heart failure in cancer survivors with anthracycline-induced cardiomyopathy. CASE PRESENTATION: A survivor of cancer as a young adult who received high-dose anthracycline chemotherapy presented with acute decompensated heart failure 20 years post-chemotherapy and worsening heart failure 1.5 years after discharge. The patient showed signs of improvement after a step-wise introduction of carvedilol, empagliflozin, sacubitril/valsartan, ivabradine, and spironolactone for worsening heart failure. Vericiguat was accelerated owing to the risk of more severe cardiovascular events associated with ongoing aortic stenosis and the poor prognosis of anthracycline-induced cardiomyopathy. Heart failure symptoms continued to improve, with significant cardiac reverse remodeling, and the patient successfully underwent aortic valve replacement for severe aortic stenosis. CONCLUSIONS: Our case highlighted that multidrug treatment with add-on vericiguat and ivabradine based on quadruple therapy can potentially treat worsening heart failure in young adult cancer survivors with severe anthracycline-induced cardiomyopathy.


Subject(s)
Aminobutyrates , Anthracyclines , Biphenyl Compounds , Cancer Survivors , Cardiomyopathies , Cardiotoxicity , Drug Combinations , Drug Therapy, Combination , Heart Failure , Humans , Heart Failure/chemically induced , Heart Failure/diagnosis , Heart Failure/physiopathology , Heart Failure/drug therapy , Aminobutyrates/adverse effects , Aminobutyrates/therapeutic use , Anthracyclines/adverse effects , Treatment Outcome , Cardiomyopathies/chemically induced , Cardiomyopathies/physiopathology , Cardiomyopathies/diagnosis , Mineralocorticoid Receptor Antagonists/therapeutic use , Male , Adrenergic beta-Antagonists/therapeutic use , Disease Progression , Sodium-Glucose Transporter 2 Inhibitors/adverse effects , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Valsartan , Cardiovascular Agents/adverse effects , Cardiovascular Agents/administration & dosage , Cardiovascular Agents/therapeutic use , Ventricular Function, Left/drug effects , Young Adult , Adult
3.
Drug Des Devel Ther ; 18: 4089-4116, 2024.
Article in English | MEDLINE | ID: mdl-39286288

ABSTRACT

With the continuous refinement of therapeutic measures, the survival rate of tumor patients has been improving year by year, while cardiovascular complications related to cancer therapy have become increasingly prominent. Exploring the mechanism and prevention strategy of cancer therapy-related cardiovascular toxicity (CTR-CVT) remains one of the research hotspots in the field of Cardio-Oncology in recent years. Cardiotoxicity of anticancer drugs involves heart failure, myocarditis, hypertension, arrhythmias and vascular toxicity, mechanistically related to vascular endothelial dysfunction, ferroptosis, mitochondrial dysfunction and oxidative stress. To address the cardiotoxicity induced by different anticancer drugs, various therapeutic measures have been put in place, such as reducing the accumulation of anticancer drugs, shifting to drugs with less cardiotoxicity, using cardioprotective drugs, and early detection. Due to the very limited treatments available to ameliorate anticancer drugs-induced cardiotoxicity, a few innovations are being shifted from animal studies to human studies. Examples include mitochondrial transplantation. Mitochondrial transplantation has been proven to be effective in in vivo and in vitro experiments. Several recent studies have demonstrated that intercellular mitochondrial transfer can ameliorate doxorubicin(DOX)-induced cardiotoxicity, laying the foundation for innovative therapies in anticancer drugs-induced cardiotoxicity. In this review, we will discuss the current status of anticancer drugs-induced cardiotoxicity in terms of the pathogenesis and treatment, with a focus on mitochondrial transplantation, and we hope that this review will bring some inspiration to you.


Subject(s)
Antineoplastic Agents , Cardiotoxicity , Humans , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Animals , Neoplasms/drug therapy
4.
Hum Genomics ; 18(1): 92, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39218963

ABSTRACT

Per- and poly-fluoroalkyl substances (PFAS) are emerging contaminants of concern because of their wide use, persistence, and potential to be hazardous to both humans and the environment. Several PFAS have been designated as substances of concern; however, most PFAS in commerce lack toxicology and exposure data to evaluate their potential hazards and risks. Cardiotoxicity has been identified as a likely human health concern, and cell-based assays are the most sensible approach for screening and prioritization of PFAS. Human-induced pluripotent stem cell (iPSC)-derived cardiomyocytes are a widely used method to test for cardiotoxicity, and recent studies showed that many PFAS affect these cells. Because iPSC-derived cardiomyocytes are available from different donors, they also can be used to quantify human variability in responses to PFAS. The primary objective of this study was to characterize potential human cardiotoxic hazard, risk, and inter-individual variability in responses to PFAS. A total of 56 PFAS from different subclasses were tested in concentration-response using human iPSC-derived cardiomyocytes from 16 donors without known heart disease. Kinetic calcium flux and high-content imaging were used to evaluate biologically-relevant phenotypes such as beat frequency, repolarization, and cytotoxicity. Of the tested PFAS, 46 showed concentration-response effects in at least one phenotype and donor; however, a wide range of sensitivities were observed across donors. Inter-individual variability in the effects could be quantified for 19 PFAS, and risk characterization could be performed for 20 PFAS based on available exposure information. For most tested PFAS, toxicodynamic variability was within a factor of 10 and the margins of exposure were above 100. This study identified PFAS that may pose cardiotoxicity risk and have high inter-individual variability. It also demonstrated the feasibility of using a population-based human in vitro method to quantify population variability and identify cardiotoxicity risks of emerging contaminants.


Subject(s)
Cardiotoxicity , Fluorocarbons , Induced Pluripotent Stem Cells , Myocytes, Cardiac , Humans , Induced Pluripotent Stem Cells/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Cardiotoxicity/etiology , Fluorocarbons/toxicity , Environmental Pollutants/toxicity , Risk Assessment , Adult , Female , Male , Environmental Exposure/adverse effects
5.
Mol Med Rep ; 30(4)2024 Oct.
Article in English | MEDLINE | ID: mdl-39219269

ABSTRACT

Tyrosine kinase inhibitors (TKIs) offer targeted therapy for cancers but can cause severe cardiotoxicities. Determining their dose­dependent impact on cardiac function is required to optimize therapy and minimize adverse effects. The dose­dependent cardiotoxic effects of two TKIs, imatinib and ponatinib, were assessed in vitro using H9c2 cardiomyoblasts and in vivo using zebrafish embryos. In vitro, H9c2 cardiomyocyte viability, apoptosis, size, and surface area were evaluated to assess the impact on cellular health. In vivo, zebrafish embryos were analyzed for heart rate, blood flow velocity, and morphological malformations to determine functional and structural changes. Additionally, reverse transcription­quantitative PCR (RT­qPCR) was employed to measure the gene expression of atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP), established markers of cardiac injury. This comprehensive approach, utilizing both in vitro and in vivo models alongside functional and molecular analyses, provides a robust assessment of the potential cardiotoxic effects. TKI exposure decreased viability and surface area in H9c2 cells in a dose­dependent manner. Similarly, zebrafish embryos exposed to TKIs exhibited dose­dependent heart malformation. Both TKIs upregulated ANP and BNP expression, indicating heart injury. The present study demonstrated dose­dependent cardiotoxic effects of imatinib and ponatinib in H9c2 cells and zebrafish models. These findings emphasize the importance of tailoring TKI dosage to minimize cardiac risks while maintaining therapeutic efficacy. Future research should explore the underlying mechanisms and potential mitigation strategies of TKI­induced cardiotoxicities.


Subject(s)
Cardiotoxicity , Imatinib Mesylate , Imidazoles , Myocytes, Cardiac , Pyridazines , Zebrafish , Animals , Zebrafish/embryology , Imidazoles/toxicity , Pyridazines/adverse effects , Pyridazines/pharmacology , Pyridazines/toxicity , Imatinib Mesylate/toxicity , Imatinib Mesylate/adverse effects , Imatinib Mesylate/pharmacology , Cardiotoxicity/etiology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/toxicity , Protein Kinase Inhibitors/pharmacology , Cell Line , Natriuretic Peptide, Brain/metabolism , Natriuretic Peptide, Brain/genetics , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Cell Survival/drug effects , Apoptosis/drug effects , Myoblasts, Cardiac/drug effects , Myoblasts, Cardiac/metabolism , Rats
6.
Clin Exp Pharmacol Physiol ; 51(10): e13920, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39227014

ABSTRACT

This study aimed to investigate the effects and possible mechanisms of adenylate cyclase 1 (ADCY1) on pirarubicin-induced cardiomyocyte injury. HL-1 cells were treated with pirarubicin (THP) to induce intracellular toxicity, and the extent of damage to mouse cardiomyocytes was assessed using CCK-8, Edu, flow cytometry, ROS, ELISA, RT-qPCR and western blotting. THP treatment reduced the viability of HL-1 cells, inhibited proliferation, induced apoptosis and triggered oxidative stress. In addition, the RT-qPCR results revealed that ADCY1 expression was significantly elevated in HL-1 cells, and molecular docking showed a direct interaction between ADCY1 and THP. Western blotting showed that ADCY1, phospho-protein kinase A and GRIN2D expression were also significantly elevated. Knockdown of ADCY1 attenuated THP-induced cardiotoxicity, possibly by regulating the ADCY1/PKA/GRIN2D pathway.


Subject(s)
Adenylyl Cyclases , Cardiotoxicity , Doxorubicin , Gene Knockdown Techniques , Myocytes, Cardiac , Adenylyl Cyclases/metabolism , Adenylyl Cyclases/genetics , Animals , Mice , Cardiotoxicity/genetics , Doxorubicin/toxicity , Doxorubicin/pharmacology , Doxorubicin/analogs & derivatives , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Cell Line , Apoptosis/drug effects , Oxidative Stress/drug effects , Oxidative Stress/genetics , Molecular Docking Simulation , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity
7.
J Biochem Mol Toxicol ; 38(10): e23851, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39267350

ABSTRACT

Doxorubicin (Dox) is frequently employed as a chemotherapy agent for breast cancer. As the chemotherapy moves forward, breast cancer cells tend to develop resistance to Dox, besides that, Dox are also easy to cause cardiotoxicity related to cumulative dose. Therefore, how to potentiate the chemosensitivity of breast cancer cells to Dox while attenuating its cardiotoxicity has become a research hotspot. Tanshinone IIA (Tan IIA) is known for its anticancer activity as well as for its cardioprotective effects. In view of the aforementioned facts, we assessed whether Tan IIA possesses synergism and attenuation effect on Dox for breast cancer chemotherapy. Our studies in vitro indicated that, Tan IIA could potentiate the effect of Dox on breast cancer cells proliferation inhibition and apoptosis promotion by inhibiting ERK1/2 pathway, but interestingly, Tan IIA attenuated the cytotoxicity of Dox to myocardial cells by activating ERK1/2 pathway. Additionally, our studies in vivo also suggested that Tan IIA potentiated the chemotherapeutic effect of Dox against breast cancer while attenuating Dox-induced myocardial injury. Given that Tan IIA had a synergism and attenuation effect on Dox, we believed that Tan IIA can be used as an ideal drug in combination with Dox for breast cancer therapy.


Subject(s)
Abietanes , Breast Neoplasms , Cardiotoxicity , Doxorubicin , MAP Kinase Signaling System , Doxorubicin/adverse effects , Doxorubicin/pharmacology , Abietanes/pharmacology , Female , Humans , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , MAP Kinase Signaling System/drug effects , Animals , Cardiotoxicity/drug therapy , Cardiotoxicity/prevention & control , Cardiotoxicity/etiology , Drug Synergism , MCF-7 Cells , Mice , Apoptosis/drug effects , Cell Line, Tumor , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism
8.
Int J Mol Sci ; 25(17)2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39273594

ABSTRACT

This study was designed to examine the association between myocardial concentrations of the trace elements Cu, Fe, Mn, Mo, and Zn and the expression of mitochondrial unfolded protein response (UPRmt) elements and the age of patients who received heart transplantation or a left-ventricular assist device (ageHTx/LVAD). Inductively coupled plasma mass spectrometry was used to determine the concentration of Cu, Fe, Mn, Mo, and Zn in the myocardium of control subjects and patients undergoing heart transplantation or left-ventricular assist device (LVAD) implantation. We used ELISA to quantify the expression of UPRmt proteins and 4-Hydroxynonenal (4-HNE), which served as a marker of oxidative-stress-induced lipid peroxidation. Concentrations of Cu, Mn, Mo, and Zn were similar in the control and heart failure (HF) myocardium, while Fe showed a significant decrease in the HF group compared to the control. A higher cumulative concentration of Fe and Zn in the myocardium was associated with reduced ageHTx/LVAD, which was not observed for other combinations of trace elements or their individual effects. The trace elements Cu, Mn, and Zn showed positive correlations with several UPRmt proteins, while Fe had a negative correlation with UPRmt effector protease YME1L. None of the trace elements correlated with 4-HNE in the myocardium. The concentrations of the trace elements Mn and Zn were significantly higher in the myocardium of patients with dilated cardiomyopathy than in patients with ischemic cardiomyopathy. A higher cumulative concentration of Fe and Zn in the myocardium was associated with a younger age at which patients received heart transplantation or LVAD, potentially suggesting an acceleration of HF. A positive correlation between myocardial Cu, Mn, and Zn and the expression of UPRmt proteins and a negative correlation between myocardial Fe and YME1L expression suggest that these trace elements exerted their actions on the human heart by interacting with the UPRmt. An altered generation of oxidative stress was not an underlying mechanism of the observed changes.


Subject(s)
Iron , Unfolded Protein Response , Zinc , Humans , Zinc/metabolism , Zinc/analysis , Male , Iron/metabolism , Middle Aged , Female , Adult , Cardiotoxicity/etiology , Cardiotoxicity/metabolism , Oxidative Stress , Heart Failure/metabolism , Myocardium/metabolism , Aged , Heart Transplantation , Heart-Assist Devices/adverse effects , Aldehydes/metabolism
9.
Int J Mol Sci ; 25(17)2024 Sep 07.
Article in English | MEDLINE | ID: mdl-39273638

ABSTRACT

Doxorubicin (DOX) is a potent chemotherapeutic agent with well-documented dose-dependent cardiotoxicity. Regular exercise is recognized for its cardioprotective effects against DOX-induced cardiac inflammation, although the precise mechanisms remain incompletely understood. The activation of inflammasomes has been implicated in the pathogenesis and treatment of DOX-induced cardiotoxicity, with the nucleotide-binding domain-like receptor protein 3 (NLRP3) inflammasome emerging as a key mediator in cardiovascular inflammation. This study aimed to investigate the role of exercise in modulating the NLRP3 inflammasome to protect against DOX-induced cardiac inflammation. Male Sprague-Dawley rats were randomly assigned to receive a 10-day course of DOX or saline injections, with or without a preceding 10-week treadmill running regimen. Cardiovascular function and histological changes were subsequently evaluated. DOX-induced cardiotoxicity was characterized by cardiac atrophy, systolic dysfunction, and hypotension, alongside activation of the NLRP3 inflammasome. Our findings revealed that regular exercise preserved cardiac mass and hypertrophic indices and prevented DOX-induced cardiac dysfunction, although it did not fully preserve blood pressure. These results underscore the significant cardioprotective effects of exercise against DOX-induced cardiotoxicity. While regular exercise did not entirely prevent DOX-induced hypotension, our findings demonstrate that it confers protection against DOX-induced cardiotoxicity by suppressing NLRP3 inflammasome activation in the heart, underscoring its anti-inflammatory role. Further research should explore the temporal dynamics and interactions among exercise, pyroptosis, and other pathways in DOX-induced cardiotoxicity to enhance translational applications in cardiovascular medicine.


Subject(s)
Cardiomyopathies , Disease Models, Animal , Doxorubicin , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Physical Conditioning, Animal , Rats, Sprague-Dawley , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Doxorubicin/adverse effects , Rats , Male , Inflammasomes/metabolism , Cardiomyopathies/chemically induced , Cardiomyopathies/metabolism , Cardiotoxicity/metabolism , Cardiotoxicity/prevention & control
10.
Int J Mol Sci ; 25(17)2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39273682

ABSTRACT

Doxorubicin (DOX), a commonly used anticancer agent, causes cardiotoxicity that begins with the first dose and may progress to heart failure years after treatment. An inflammatory response associated with neutrophil recruitment has been recognized as a mechanism of DOX-induced cardiotoxicity. This study aimed to validate mRNA expression of the previously identified biomarkers of DOX-induced cardiotoxicity, PGLYRP1, CAMP, MMP9, and CEACAM8, and to assay their protein expression in the peripheral blood of breast cancer patients. Blood samples from 40 breast cancer patients treated with DOX-based chemotherapy were collected before and after the first chemotherapy cycle and > 2 years after treatment. The protein and gene expression of PGLYRP1/Tag7, CAMP/LL37, MMP9/gelatinase B, and CEACAM8/CD66b were determined using ELISA and reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Receiver operating characteristic (ROC) curve analysis was used to determine the diagnostic value of each candidate biomarker. Patients with cardiotoxicity (n = 20) had significantly elevated levels of PGLYRP1, CAMP, MMP9, and CEACAM8 at baseline, after the first dose of DOX-based chemotherapy, and at > 2 years after treatment relative to patients without cardiotoxicity (n = 20). The first dose of DOX induced significantly higher levels of all examined biomarkers in both groups of patients. At > 2 years post treatment, the levels of all but MMP9 dropped below the baseline. There was a good correlation between the expression of mRNA and the target proteins. We demonstrate that circulating levels of PGLYRP1, CAMP, MMP9, and CEACAM8 can predict the cardiotoxicity of DOX. This novel finding may be of value in the early identification of patients at risk for cardiotoxicity.


Subject(s)
Anthracyclines , Breast Neoplasms , Cardiotoxicity , Doxorubicin , Neutrophils , Humans , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/blood , Middle Aged , Neutrophils/metabolism , Anthracyclines/adverse effects , Cardiotoxicity/etiology , Cardiotoxicity/blood , Cardiotoxicity/diagnosis , Doxorubicin/adverse effects , Adult , Aged , Biomarkers, Tumor/blood , Cell Adhesion Molecules/blood , Cell Adhesion Molecules/genetics , Matrix Metalloproteinase 9/blood , Matrix Metalloproteinase 9/genetics , Biomarkers/blood , Antigens, CD/blood , Antigens, CD/genetics , Antigens, CD/metabolism , GPI-Linked Proteins
11.
Med Oncol ; 41(10): 244, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39259412

ABSTRACT

The likelihood of survival for cancer patients has greatly improved due to chemotherapy medicines. However, these antitumor agents might also have unfavorable effects on the cardiovascular system, which could result in sudden or gradual cardiac failure. The production of free radicals that result in oxidative stress appears to be the key mechanism by which chemotherapy-induced cardiotoxicity (CIC) happens. Reports suggest that the Sirtuin-1 (Sirt1)/Nuclear factor E2-associated factor 2 (Nrf2) signaling pathway has been considered an alternative path for counteracting cardiotoxicity by suppressing oxidative stress, inflammation, and apoptosis. This review concludes recent knowledge about CIC with a special focus on the anti-oxidative regulation properties of the Sirt1/Nrf2 pathway.


Subject(s)
Antineoplastic Agents , Cardiotoxicity , NF-E2-Related Factor 2 , Signal Transduction , Sirtuin 1 , Humans , Sirtuin 1/metabolism , NF-E2-Related Factor 2/metabolism , Cardiotoxicity/etiology , Signal Transduction/drug effects , Antineoplastic Agents/adverse effects , Oxidative Stress/drug effects , Animals , Neoplasms/drug therapy , Neoplasms/metabolism
12.
Hereditas ; 161(1): 31, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39243097

ABSTRACT

BACKGROUND: Previous clinical and basic studies have revealed that ginseng might have cardioprotective properties against anthracycline-induced cardiotoxicity (AIC). However, the underlying mechanism of ginseng action against AIC remains insufficiently understood. The aim of this study was to explore the related targets and pathways of ginseng against AIC using network pharmacology, molecular docking, cellular thermal shift assay (CETSA) and molecular dynamics (MD) simulations. RESULTS: Fourteen drug-disease common targets were identified. Enrichment analysis showed that the AGE-RAGE in diabetic complications, fluid shear stress and atherosclerosis, and TNF signaling pathway were potentially involved in the action of ginseng against AIC. Molecular docking demonstrated that the core components including Kaempferol, beta-Sitosterol, and Fumarine had notable binding activity with the three core targets CCNA2, STAT1, and ICAM1. Furthermore, the stable complex of STAT1 and Kaempferol with favorable affinity was further confirmed by CETSA and MD simulation. CONCLUSIONS: This study suggested that ginseng might exert their protective effects against AIC through the derived effector compounds beta-Sitosterol, Kaempferol and Fumarine by targeting CCNA2, STAT1, and ICAM1, and modulating AGE-RAGE in diabetic complications, fluid shear stress and atherosclerosis, and TNF signaling pathways.


Subject(s)
Anthracyclines , Cardiotoxicity , Molecular Docking Simulation , Molecular Dynamics Simulation , Network Pharmacology , Panax , Panax/chemistry , Anthracyclines/adverse effects , Anthracyclines/chemistry , Anthracyclines/toxicity , Humans , Sitosterols/pharmacology , Sitosterols/chemistry , STAT1 Transcription Factor/metabolism , STAT1 Transcription Factor/genetics , Kaempferols/pharmacology , Kaempferols/chemistry , Signal Transduction/drug effects
13.
Nat Commun ; 15(1): 7968, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39261481

ABSTRACT

Drug-induced gene expression profiles can identify potential mechanisms of toxicity. We focus on obtaining signatures for cardiotoxicity of FDA-approved tyrosine kinase inhibitors (TKIs) in human induced-pluripotent-stem-cell-derived cardiomyocytes, using bulk transcriptomic profiles. We use singular value decomposition to identify drug-selective patterns across cell lines obtained from multiple healthy human subjects. Cellular pathways affected by cardiotoxic TKIs include energy metabolism, contractile, and extracellular matrix dynamics. Projecting these pathways to published single cell expression profiles indicates that TKI responses can be evoked in both cardiomyocytes and fibroblasts. Integration of transcriptomic outlier analysis with whole genomic sequencing of our six cell lines enables us to correctly reidentify a genomic variant causally linked to anthracycline-induced cardiotoxicity and predict genomic variants potentially associated with TKI-induced cardiotoxicity. We conclude that mRNA expression profiles when integrated with publicly available genomic, pathway, and single cell transcriptomic datasets, provide multiscale signatures for cardiotoxicity that could be used for drug development and patient stratification.


Subject(s)
Cardiotoxicity , Gene Expression Profiling , Myocytes, Cardiac , Protein Kinase Inhibitors , Transcriptome , Humans , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/toxicity , Gene Expression Profiling/methods , Cardiotoxicity/genetics , Cardiotoxicity/etiology , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/drug effects , Cell Line , Single-Cell Analysis/methods , Fibroblasts/drug effects , Fibroblasts/metabolism
14.
Sci Rep ; 14(1): 21180, 2024 09 11.
Article in English | MEDLINE | ID: mdl-39261543

ABSTRACT

Minocycline (Min), as an antibiotic, possesses various beneficial properties such as anti-inflammatory, antioxidant, and anti-apoptotic effects. Despite these known qualities, the precise cardioprotective effect and mechanism of Min in protecting against sepsis-induced cardiotoxicity (SIC) remain unspecified. To address this, our study sought to assess the protective effects of Min on the heart. Lipopolysaccharide (LPS) was utilized to establish a cardiotoxicity model both in vivo and in vitro. Min was pretreated in the models. In the in vivo setting, evaluation of heart tissue histopathological injury was performed using hematoxylin and eosin (H&E) staining and TUNEL. Immunohistochemistry (IHC) was employed to evaluate the expression levels of NLRP3 and Caspase-1 in the heart tissue of mice. During in vitro experiments, the viability of H9c2 cells was gauged utilizing the CCK8 assay kit. Intracellular ROS levels in H9c2 cells were quantified using a ROS assay kit. Both in vitro and in vivo settings were subjected to measurement of oxidative stress indexes, encompassing glutathione (GSH), malondialdehyde (MDA), and superoxide dismutase (SOD) levels. Additionglly, myocardial injury markers like lactate dehydrogenase (LDH) and creatine kinase MB (CK-MB) activity were quantified using appropriate assay kits. Western blotting (WB) analysis was conducted to detect the expression levels of NOD-like receptor protein-3 (NLRP3), caspase-1, IL-18, and IL-1ß, alongside apoptosis-related proteins such as Bcl-2 and Bax, and antioxidant proteins including superoxide dismutase-1 (SOD-1) and antioxidant proteins including superoxide dismutase-1 (SOD-2), both in H9c2 cells and mouse heart tissues. In vivo, Min was effective in reducing LPS-induced inflammation in cardiac tissue, preventing cell damage and apoptosis in cardiomyocytes. The levels of LDH and CK-MB were significantly reduced with Min treatment. In vitro studies showed that Min improved the viability of H9C2 cells, reduced apoptosis, and decreased ROS levels in these cells. Further analysis indicated that Min decreased the protein levels of NLRP3, Caspase-1, IL-18, and IL-1ß, while increasing the levels of SOD-1 and SOD-2 both in vivo and in vitro. Min alleviates LPS-induced SIC by suppressing the NLRP3/Caspase-1 signalling pathway in vivo and in vitro.


Subject(s)
Cardiotoxicity , Caspase 1 , Lipopolysaccharides , Minocycline , NLR Family, Pyrin Domain-Containing 3 Protein , Signal Transduction , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Signal Transduction/drug effects , Lipopolysaccharides/toxicity , Caspase 1/metabolism , Cardiotoxicity/metabolism , Cardiotoxicity/drug therapy , Mice , Minocycline/pharmacology , Male , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Oxidative Stress/drug effects , Cell Line , Apoptosis/drug effects , Reactive Oxygen Species/metabolism , Mice, Inbred C57BL , Rats
15.
Kardiologiia ; 64(8): 3-12, 2024 Aug 31.
Article in Russian, English | MEDLINE | ID: mdl-39262348

ABSTRACT

The most important component of cardio-oncology is the assessment of the risk of development and diagnosis of cardiovascular toxicity of the antitumor therapy, the detection of which is largely based on visualization of the cardiovascular system. The article addresses up-to-date methods of non-invasive visualization of the heart and blood vessels, according to the 2022 European Society of Cardiology Clinical Guidelines on cardio-oncology. Also, the article discusses promising cardiovascular imaging techniques that are not yet included in the guidelines: assessment of coronary calcium using multislice computed tomography and positron emission computed tomography with 18F-labeled 2-deoxy-2-fluoro-d-glucose.


Subject(s)
Cardiotoxicity , Humans , Risk Assessment/methods , Cardiotoxicity/etiology , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Cardiovascular Diseases/diagnosis
16.
Toxicol Appl Pharmacol ; 491: 117082, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39218162

ABSTRACT

PURPOSE: Doxorubicin is an antibiotic drug used clinically to treat infectious diseases and tumors. Unfortunately, it is cardiotoxic. Autophagy is a cellular self-decomposition process that is essential for maintaining homeostasis in the internal environment. Accordingly, the present study was proposed to characterize the autophagy-related signatures of doxorubicin-induced cardiotoxicity. METHODS: Datasets related to doxorubicin-induced cardiotoxicity were retrieved by searching the GEO database and differentially expressed genes (DEGs) were identified. DEGs were taken to intersect with autophagy-related genes to obtain autophagy-related signatures, and Gene Ontology (GO)/Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis and protein-protein interaction (PPI) network were performed on them. Further, construction of miRNA-hub gene networks and identification of target drugs to reveal potential molecular mechanisms and therapeutic strategies. Animal models of doxorubicin-induced cardiotoxicity were constructed to validate differences in gene expression for autophagy-related signatures. RESULTS: PBMC and heart samples from the GSE37260 dataset were selected for analysis. There were 995 and 2357 DEGs in PBMC and heart samples, respectively, and they had 23 intersecting genes with autophagy-related genes. RT-qPCR confirmed the differential expression of 23 intersecting genes in doxorubicin-induced cardiotoxicity animal models in general agreement with the bioinformatics results. An autophagy-related signatures consisting of 23 intersecting genes is involved in mediating processes and pathways such as autophagy, oxidative stress, apoptosis, protein ubiquitination and phosphorylation. Moreover, Akt1, Hif1a and Mapk3 are hub genes in autophagy-associated signatures and their upstream miRNAs are mainly rno-miR-1188-5p, rno-miR-150-3p and rno-miR-326-3p, and their drugs are mainly CHEMBL55802, Carboxyamidotriazole and 3-methyladenine. CONCLUSION: This study identifies for the first-time autophagy-related signatures in doxorubicin's cardiotoxicity, which could provide potential molecular mechanisms and therapeutic strategies for doxorubicin-induced cardiotoxicity.


Subject(s)
Autophagy , Cardiotoxicity , Doxorubicin , Doxorubicin/toxicity , Autophagy/drug effects , Animals , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Male , Protein Interaction Maps , Antibiotics, Antineoplastic/toxicity , Gene Regulatory Networks/drug effects , Mice , Gene Expression Profiling/methods , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism
17.
Int J Mol Sci ; 25(17)2024 Aug 24.
Article in English | MEDLINE | ID: mdl-39273136

ABSTRACT

One of the many unresolved obstacles in the field of cardiovascular research is an uncompromising in vitro cardiac model. While primary cell sources from animal models offer both advantages and disadvantages, efforts over the past half-century have aimed to reduce their use. Additionally, obtaining a sufficient quantity of human primary cardiomyocytes faces ethical and legal challenges. As the practically unlimited source of human cardiomyocytes from induced pluripotent stem cells (hiPSC-CM) is now mostly resolved, there are great efforts to improve their quality and applicability by overcoming their intrinsic limitations. The greatest bottleneck in the field is the in vitro ageing of hiPSC-CMs to reach a maturity status that closely resembles that of the adult heart, thereby allowing for more appropriate drug developmental procedures as there is a clear correlation between ageing and developing cardiovascular diseases. Here, we review the current state-of-the-art techniques in the most realistic heart models used in disease modelling and toxicity evaluations from hiPSC-CM maturation through heart-on-a-chip platforms and in silico models to the in vitro models of certain cardiovascular diseases.


Subject(s)
Cardiotoxicity , Induced Pluripotent Stem Cells , Myocytes, Cardiac , Humans , Cardiotoxicity/etiology , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/metabolism , Animals , Cell Differentiation , Cardiovascular Diseases , Models, Cardiovascular
18.
Int J Mol Sci ; 25(17)2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39273564

ABSTRACT

Cellular Communication Network Factor 2 (CCN2) is a matricellular protein implicated in cell communication and microenvironmental signaling. Overexpression of CCN2 has been documented in various cardiovascular pathologies, wherein it may exert either deleterious or protective effects depending on the pathological context, thereby suggesting that its role in the cardiovascular system is not yet fully elucidated. In this study, we aimed to investigate the effects of Ccn2 gene deletion on the progression of acute cardiac injury induced by doxorubicin (DOX), a widely utilized chemotherapeutic agent. To this end, we employed conditional knockout (KO) mice for the Ccn2 gene (CCN2-KO), which were administered DOX and compared to DOX-treated wild-type (WT) control mice. Our findings demonstrated that the ablation of CCN2 ameliorated DOX-induced cardiac dysfunction, as evidenced by improvements in ejection fraction (EF) and fractional shortening (FS) of the left ventricle. Furthermore, DOX-treated CCN2-KO mice exhibited a significant reduction in the gene expression and activation of oxidative stress markers (Hmox1 and Nfe2l2/NRF2) relative to DOX-treated WT controls. Additionally, the deletion of Ccn2 markedly attenuated DOX-induced cardiac fibrosis. Collectively, these results suggest that CCN2 plays a pivotal role in the pathogenesis of DOX-mediated cardiotoxicity by modulating oxidative stress and fibrotic pathways. These findings provide a novel avenue for future investigations to explore the therapeutic potential of targeting CCN2 in the prevention of DOX-induced cardiac dysfunction.


Subject(s)
Connective Tissue Growth Factor , Doxorubicin , Fibrosis , Mice, Knockout , Oxidative Stress , Animals , Doxorubicin/adverse effects , Connective Tissue Growth Factor/metabolism , Connective Tissue Growth Factor/genetics , Mice , Oxidative Stress/drug effects , Gene Deletion , Male , Myocardium/metabolism , Myocardium/pathology , Mice, Inbred C57BL , Cardiotoxicity/genetics , Cardiotoxicity/metabolism
19.
Arq. bras. cardiol ; 121(9 supl.1): 279-279, set.2024. tab
Article in English | CONASS, Sec. Est. Saúde SP, SESSP-IDPCPROD, Sec. Est. Saúde SP | ID: biblio-1568600

ABSTRACT

Breast Cancer (BC) is one of the most common cancers diagnosed in population femmale and it has several subtypes, one of them being theexpressing human epidermal growth factor receptor 2 positive (HER2 +), one of the treatments for HER2+ breast cancer consists of chemotherapy plus trastuzumab deruxtecan. Several clinical trials have shown the effectiveness and safety of trastuzumabe deruxtecano in cancer patients, however, several Adverse Events (AEs) have been described and the decrease in left ventricular ejection has been singled out for more prominent analysis. Objective: We conducted a systematic review and meta-analysis to investigate the cardiovascular effects of Trastuzumab Deruxtecano and whether it can influence the appearance of reduced left ventricular ejection fraction.. METHODS: We performed a systematic search in Embase, PubMed and Cochrane databases for randomized controlled trials (RCTs) showed a decrease in left ventricular ejection fraction in patients using trastuzumab deruxtecan against Her-2-positive breast cancer compared to patients to used another's treatments against this disease. Mean difference (MD) with 95% confidence intervals (CI) were calculated using a random effects model. The heterogeneity was examined in the I2 statistic. P-values > 0.05 were considered statistically significant. The statistical analysis was carried out using R software version 4.2.3. RESULTS: A total of 3 RCTs were included, with a total of 1656 patients evaluated, 928 patients randomized to the use of Trastuzumab Deruxtecan and 728 patients to the use of other treatments according to medical choice, follow-up ranged from 10 to 38 months. There was a visible in the decrease in left ventricular ejection fraction, with a higher incidence in the group that used trastuzumab compared to the placebo group (RR: 5.73%; 95% CI 1.51 - 21.78; I2 33% ; P= 0.010466). Another important point is the discontinuation of treatment due to grade 2 adverse events, classified as reduced LVEF, where a higher incidence is seen in the group that used Trastuzumab Deruxtecan compared to the placebo group (RR 2.11%; 95% CI 1.54 - 2.89; P = 0.000003),7. CONCLUSION: In this meta-analysis, Trastuzumab Deruxtecan showed a relationship with a decrease in left ventricular ejection fraction, displaying the need for more studies to evaluate the cardiotoxicity of trastuzumab and its effects as a whole on the cardiovascular system.


Subject(s)
Therapeutics , Breast Neoplasms , Cardiovascular Diseases , Drug Therapy , Cardiotoxicity , Trastuzumab , Data Interpretation, Statistical , ErbB Receptors
20.
Arq. bras. cardiol ; 121(9 supl.1): 280-280, set.2024. tab
Article in English | CONASS, Sec. Est. Saúde SP, SESSP-IDPCPROD, Sec. Est. Saúde SP | ID: biblio-1568623

ABSTRACT

BACKGROUND: Contemporary understanding characterizes cardiotoxicity as a reduction in left ventricular ejection fraction (LVEF) by at least 10%, resulting in a final value below 53% in successive assessments. Nevertheless, breast cancer therapy can impact the cardiovascular system through various avenues. Cardiotoxicity is a known side effect of anthracycline chemotherapy, and the effectiveness of concomitant statin use in mitigating this risk is still unclear. PURPOSE: We aimed to evaluate the potential cardioprotective effects of statin exposure during anthracycline treatment. Our hypothesis posited that patients receiving statins during their treatment would experience a lesser decline in left ventricular ejection fraction (LVEF), lower levels of cholesterol and a reduced occurrence of cardiotoxicity compared to those not exposed to statins. METHODS: We performed a systematic review and meta-analysis of randomized controlled trials (RCTs) comparing statin versus placebo in patients undergoing anthracycline therapy. We searched PubMed, Embase and Cochrane for eligible trials. Mean differences (MDs) with 95% confidence intervals (CIs) were calculated using a random-effects model. Heterogeneity was examined with I2 statistics. P values of < 0.05 were considered statistically significant. Statistical analysis were performed using R software version 4.2.3. RESULTS: A total of 4 RCTs comprising 580 patients were included, of whom 281 were randomized to statins and 299 to placebo. The follow up period ranged from 2.5 to 24 months, with participant ages varying between 36 to 68.9 in the intervention group and 37.9 to 72 in the control group. Compared with placebo, statins were significantly associated with a higher left ventricular ejection fraction (MD 2.57%; 95% CI 1.05-4.08; p<0.001; I2=0%), reduction in left ventricular systolic end-volume (MD -4.5 mL; 95% CI -7.57 to -1.44; p<0.004; I2=0%) and diastolic end-volume (MD -6.08 mL; 95% CI -11.27 to -0.9; p<0.021; I2=0%), with a low heterogeneity value. Statins also showed important reduction of total cholesterol (MD -46.28 mg/dL; 95% CI -71.3 to -21.25; p<0.001; I2=89%) and LDL-C (MD -39.45 mg/dL; 95% CI -52.27 to -26.64; p<0.001; I2=84%). CONCLUSIONS: In this metaanalysis of RCTs, the use of statins showed a correlation with improved cardiovascular parameters, indicating their effectiveness in minimizing cardiotoxicity in breast cancer patients undergoing anthracycline chemotherapy


Subject(s)
Breast Neoplasms , Cardiovascular Diseases , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Data Interpretation, Statistical , Drug Therapy , Cardiotoxicity
SELECTION OF CITATIONS
SEARCH DETAIL