International Immunopharmacology, Год журнала: 2024, Номер 146, С. 113788 - 113788
Опубликована: Дек. 19, 2024
Язык: Английский
International Immunopharmacology, Год журнала: 2024, Номер 146, С. 113788 - 113788
Опубликована: Дек. 19, 2024
Язык: Английский
Microscopy Research and Technique, Год журнала: 2024, Номер 87(7), С. 1598 - 1614
Опубликована: Март 5, 2024
Abstract Cardiotoxicity induced by doxorubicin (Dox) is a major complication in cancer patients. Exosomes (Ex) derived from mesenchymal cells could be promising therapeutic for various heart diseases. This study investigated the role of Ex Dox‐induced cardiotoxicity and its mechanistic insights, using Sacubitril/valsartan (S/V) as reference drug widely recommended failure management. The involved 24 Wistar rats, divided into control, Dox, Dox + S/V, groups. rats were assessed cardiac enzymes, inflammatory oxidative stress markers. Immunohistochemical expression caspase‐1, nuclear factor erythroid 2‐related 2 (NrF2), E‐Cadherin, CD117/c‐kit, Platelet‐derived growth factor‐α (PDGFα) was evaluated. P53 Annexin V PCR. Histological examination performed hematoxylin eosin Sirius red stains. ameliorated adverse pathological changes significantly decreased enzymes also exerted antifibrotic antiapoptotic effect tissue. treatment improved NrF2 immunohistochemistry, up‐regulated E‐Cadherin immune expression, restored telocyte markers CD117/c‐kit PDGFα. can mitigate acting an anti‐inflammatory, antioxidant, antiapoptotic, agents, restoring telocytes modulating epithelial transition. Research Highlights exhibit positive CD90 CD105 whereas showing −ve CD 34 flow cytometry. restore immunohistochemical alleviate myocardial apoptosis, fibrosis.
Язык: Английский
Процитировано
1European Journal of Pharmaceutical Sciences, Год журнала: 2024, Номер 200, С. 106849 - 106849
Опубликована: Июль 9, 2024
Doxorubicin (DOX) is an anthracycline chemotherapy drug widely employed in the treatment of various cancers, known for its potent antineoplastic properties but often associated with dose-dependent cardiotoxicity, limiting clinical use. This review explores complex molecular details that determine heart-protective effectiveness carvedilol relation to cardiotoxicity caused by DOX. The harmful effects DOX on heart cells could include oxidative stress, DNA damage, iron imbalance, disruption autophagy, calcium apoptosis, dysregulation topoisomerase 2-beta, arrhythmogenicity, and inflammatory responses. carefully reveals how serves as a strong protective mechanism, strategically reducing each aspect cardiac damage Carvedilol's antioxidant capabilities involve neutralizing free radicals adjusting crucial enzymes. It skillfully manages balance, controls restores balance essential cellular stability. Moreover, anti-apoptotic are outlined through adjustment Bcl-2 family proteins activation Akt signaling pathway. medication also 2-beta reduces renin-angiotensin-aldosterone system, together offering thorough defense against induced These findings not only provide detailed understanding into mechanisms coordinate protection offer considerable potential creation targeted strategies intended relieve chemotherapy.
Язык: Английский
Процитировано
1Current Problems in Cardiology, Год журнала: 2024, Номер unknown, С. 102882 - 102882
Опубликована: Окт. 1, 2024
Язык: Английский
Процитировано
1Опубликована: Дек. 13, 2023
Doxorubicin (DOX) is an incessantly used chemotherapeutic drug that can cause detrimental dose-dependent effects such as cardiotoxicity and congestive heart failure. Hence, there a need to discover innovative therapeutic approaches counteract DOX-induced (DIC). MSC-Exos have shown reduce apoptosis cardiac fibrosis promote cardiomyocyte proliferation in myocardial infracted mice. However, the effect of on ameliorating pyroptosis has not been investigated. In this current study H9c2 were first exposed DOX stimulate pyroptosis, followed by subsequent treatment with MSC-Exos, further analysis performed through immunocytochemistry, western blotting, RT-PCR. Our data depicted post-treatment significantly (p<0.05) reduced HMGB1/TLR4 axis, inflammasome formation (NLRP3), pyroptotic markers (caspase-1, IL-1β, IL-18), executioner (GSDMD) DOX-treated cells. conclusion, our shows attenuates inflammation-induced vitro DIC model. findings indicate may serve promising intervention for mitigating DIC, they maintain capabilities MSCs while circumventing drawbacks associated traditional stem cell therapy.
Язык: Английский
Процитировано
2Biochemical Pharmacology, Год журнала: 2023, Номер 220, С. 116009 - 116009
Опубликована: Дек. 26, 2023
Язык: Английский
Процитировано
2Research Square (Research Square), Год журнала: 2024, Номер unknown
Опубликована: Фев. 2, 2024
Язык: Английский
Процитировано
0Journal of Inflammation Research, Год журнала: 2024, Номер Volume 17, С. 669 - 685
Опубликована: Фев. 1, 2024
Purpose: Doxorubicin-induced cardiotoxicity (DIC) is a severe side reaction in cancer chemotherapy that greatly impacts the wellbeing of patients.Currently, there still an insufficiency effective and reliable biomarkers field clinical practice for early detection DIC.This study aimed to determine validate potential diagnostic predictive values critical signatures DIC.Methods: We obtained high-throughput sequencing data from GEO database performed analysis visualization using R software, GO, KEGG Cytoscape.Machine learning methods weighted gene coexpression network (WGCNA) were used identify key genes model construction.Receiver operating characteristic (ROC) nomogram assess their values.A multiregulatory was built reveal possible regulatory relationships signatures.Cell-type identification by estimating relative subsets RNA transcript (CIBERSORT) investigate differential immune cell infiltration.Additionally, animal constructed relationship between identified DIC.Results: Among 3713 differentially expressed genes, three (CSGALNACT1, ZNF296 FANCB) identified.A ROC curves based on showed excellent performance.The TFs CREB1, EP300, FLI1, FOXA1, MAX, MAZ modulated genes.An infiltration indicated many cells (activated NK cells, M0 macrophages, activated dendritic neutrophils) might be related progression DIC.Furthermore, may various degrees correlation cells.RT-qPCR demonstrated mRNA expression CSGALNACT1 significantly upregulated, while FANCB downregulated DOX-treated cardiomyocytes vitro vivo. Conclusion:Our suggested CSGALNACT1, associated with also involved DIC.They occurrence DIC.
Язык: Английский
Процитировано
0F1000Research, Год журнала: 2024, Номер 13, С. 1210 - 1210
Опубликована: Окт. 11, 2024
Язык: Английский
Процитировано
0Natural Product Communications, Год журнала: 2024, Номер 19(11)
Опубликована: Ноя. 1, 2024
Background: Triple-negative breast cancer (TNBC) remains a significant clinical challenge due to its aggressive nature and lack of targeted therapies. In TNBC, inflammation contributes tumour progression by promoting cell proliferation, survival, migration. It may be triggered cytokines such as TNF-α or chemotherapeutic agents doxorubicin (DOX). Thus, identifying that suppress in TNBC is prime interest. Caffeic acid phenethyl ester (CAPE) naturally occurring anti-inflammatory compound exhibits selective cytotoxicity against some cells. However, impact on cytokine chemotherapy induced has not yet been explored. Aim: This study aimed investigate the effects CAPE TNF-α- DOX-induced metastatic markers TNBC. Methods: The DOX viability migration were assessed using MTT wound healing assays, respectively. effect expression pro-inflammatory cytokines, epithelial-mesenchymal transition (EMT) markers, invasion was evaluated RT-qPCR. Results: inhibited TNF-α-induced proliferation also suppressed upregulation NF-κB-regulated genes, IL-1B, IL-6, IL-8, TNF-α, corroborating evidence suppresses NF-κB signalling. Additionally, EMT TNF-α–stimulated DOX-treated cells downregulating vimentin VCAM1. Furthermore, reduced MMP2 MMP9. Conclusions: We demonstrated anticancer potential ability chemotherapy-induced inflammation. offers novel insights into role suppressing metastasis specifically context These findings underscore urgent need further possible adjuvant for treating patients with especially those undergoing chemotherapy.
Язык: Английский
Процитировано
0International Immunopharmacology, Год журнала: 2024, Номер 146, С. 113788 - 113788
Опубликована: Дек. 19, 2024
Язык: Английский
Процитировано
0