Добірка наукової літератури з теми "Dox-Induced Cardiotoxicity"

Оформте джерело за APA, MLA, Chicago, Harvard та іншими стилями

Оберіть тип джерела:

Ознайомтеся зі списками актуальних статей, книг, дисертацій, тез та інших наукових джерел на тему "Dox-Induced Cardiotoxicity".

Біля кожної праці в переліку літератури доступна кнопка «Додати до бібліографії». Скористайтеся нею – і ми автоматично оформимо бібліографічне посилання на обрану працю в потрібному вам стилі цитування: APA, MLA, «Гарвард», «Чикаго», «Ванкувер» тощо.

Також ви можете завантажити повний текст наукової публікації у форматі «.pdf» та прочитати онлайн анотацію до роботи, якщо відповідні параметри наявні в метаданих.

Статті в журналах з теми "Dox-Induced Cardiotoxicity":

1

Sumneang, Natticha, Pongpan Tanajak, and Thura Tun Oo. "Toll-like Receptor 4 Inflammatory Perspective on Doxorubicin-Induced Cardiotoxicity." Molecules 28, no. 11 (May 24, 2023): 4294. http://dx.doi.org/10.3390/molecules28114294.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Doxorubicin (Dox) is one of the most frequently used chemotherapeutic drugs in a variety of cancers, but Dox-induced cardiotoxicity diminishes its therapeutic efficacy. The underlying mechanisms of Dox-induced cardiotoxicity are still not fully understood. More significantly, there are no established therapeutic guidelines for Dox-induced cardiotoxicity. To date, Dox-induced cardiac inflammation is widely considered as one of the underlying mechanisms involved in Dox-induced cardiotoxicity. The Toll-like receptor 4 (TLR4) signaling pathway plays a key role in Dox-induced cardiac inflammation, and growing evidence reports that TLR4-induced cardiac inflammation is strongly linked to Dox-induced cardiotoxicity. In this review, we outline and address all the available evidence demonstrating the involvement of the TLR4 signaling pathway in different models of Dox-induced cardiotoxicity. This review also discusses the effect of the TLR4 signaling pathway on Dox-induced cardiotoxicity. Understanding the role of the TLR4 signaling pathway in Dox-induced cardiac inflammation might be beneficial for developing a potential therapeutic strategy for Dox-induced cardiotoxicity.
2

Ammar, El-Sayed M., Shehta A. Said, Ghada M. Suddek, and Sally L. El-Damarawy. "Amelioration of doxorubicin-induced cardiotoxicity by deferiprone in rats." Canadian Journal of Physiology and Pharmacology 89, no. 4 (April 2011): 269–76. http://dx.doi.org/10.1139/y11-020.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
The therapeutic usefulness of doxorubicin (Dox), an anthracycline antibiotic used as an anticancer agent, is limited by its cardiotoxicity. Dox-induced cardiotoxicity is mainly attributed to accumulation of reactive oxygen species and interaction of Dox with cellular iron metabolism. The present study investigated the effects of the iron chelator deferiprone (Def) against Dox-induced cardiotoxicity in rats. Dox (15 mg/kg) was injected intraperitoneally as a single dose, and Def (10 mg/kg) was administered orally for 10 days. Dox showed cardiotoxicity as evidenced by increased heart rate, elevated ST segment, prolonged QTc interval, and increased T wave amplitude. In addition, Dox enhanced aconitine cardiotoxicity by decreasing its dose, producing ventricular tachycardia. Administration of Def significantly attenuated Dox-induced electrocardiographic changes. Cardiotoxicity of Dox was confirmed biochemically by a significant elevation in serum creatine kinase-MB and lactate dehydrogenase activities as well as by myocardial malondialdehyde and reduced glutathione contents. Moreover, Dox caused a significant decrease in myocardial superoxide dismutase activity. Administration of Def significantly attenuated the biochemical changes. These results suggest that Def might be a potential cardioprotective agent against Dox-induced cardiotoxicity.
3

Hamaamin, Karmand Salih, and Tavga Ahmed Aziz. "Doxorubicin-Induced Cardiotoxicity: Mechanisms and Management." Al-Rafidain Journal of Medical Sciences ( ISSN: 2789-3219 ) 3 (December 10, 2022): 87–97. http://dx.doi.org/10.54133/ajms.v3i.90.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Unfortunately, anticancer medications are extremely harmful to normal cells. Doxorubicin (DOX) is a highly cardiotoxic medication that can result in cardiomyopathy. The most significant mechanism for DOX-induced cardiotoxicity is oxidative stress, while other pathways have also been put forth. This review aims to highlight the mechanisms of DOX-induced cardiotoxicity and the most updated managements. Trustworthy sites such as Google Scholar, PubMed, and Research Gate were used to find the most updated articles. Many titles have been used for searching, such as "doxorubicin and cardiotoxicity," "cardioprotection," and "cardio-protective effects of phytochemicals." Preprints, review articles, and research with meta-analyses were disregarded. Three pathways, including oxidative stress, mitochondrial damage, and calcium excess, were responsible for DOX-induced cardiotoxicity. Cardiotoxicity may be partially caused by cell death, activation of the ubiquitin-ligase-proteasome system, and changes in its gene expression brought on by DOX. In the instance of DOX cardiotoxicity, medications and nutraceuticals with antioxidants and iron chelating properties have been found to have cardio-protective benefits. In conclusion, doxorubicin-treated cancer patients have been linked to cardiotoxicity, making cardioprotection extremely important in these patients. All of the mechanisms included in this review's discussion might provide light on fresh approaches to the treatment and/or prevention of DOX-induced cardiotoxicity.
4

Zheng, Dong, Yi Zhang, Ming Zheng, Ting Cao, Grace Wang, Lulu Zhang, Rui Ni, et al. "Nicotinamide riboside promotes autolysosome clearance in preventing doxorubicin-induced cardiotoxicity." Clinical Science 133, no. 13 (July 2019): 1505–21. http://dx.doi.org/10.1042/cs20181022.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Abstract Doxorubicin (DOX) is widely used as a first-line chemotherapeutic drug for various malignancies. However, DOX causes severe cardiotoxicity, which limits its clinical uses. Oxidative stress is one of major contributors to DOX-induced cardiotoxicity. While autophagic flux serves as an important defense mechanism against oxidative stress in cardiomyocytes, recent studies have demonstrated that DOX induces the blockage of autophagic flux, which contributes to DOX cardiotoxicity. The present study investigated whether nicotinamide riboside (NR), a precursor of nicotinamide adenine dinucleotide (NAD)+, prevents DOX cardiotoxicity by improving autophagic flux. We report that administration of NR elevated NAD+ levels, and reduced cardiac injury and myocardial dysfunction in DOX-injected mice. These protective effects of NR were recapitulated in cultured cardiomyocytes upon DOX treatment. Mechanistically, NR prevented the blockage of autophagic flux, accumulation of autolysosomes, and oxidative stress in DOX-treated cardiomyocytes, the effects of which were associated with restoration of lysosomal acidification. Furthermore, inhibition of lysosomal acidification or SIRT1 abrogated these protective effects of NR during DOX-induced cardiotoxicity. Collectively, our study shows that NR enhances autolysosome clearance via the NAD+/SIRT1 signaling, thereby preventing DOX-triggered cardiotoxicity.
5

Zhang, Wei, Zhixing Fan, Fengyuan Wang, Lin Yin, Jinchun Wu, Dengke Li, Siwei Song, Xi Wang, Yanhong Tang, and Congxin Huang. "Tubeimoside I Ameliorates Doxorubicin-Induced Cardiotoxicity by Upregulating SIRT3." Oxidative Medicine and Cellular Longevity 2023 (January 14, 2023): 1–23. http://dx.doi.org/10.1155/2023/9966355.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Cardiotoxicity linked to doxorubicin (DOX) is primarily caused by inflammation, oxidative stress, and apoptosis. The role of tubeimoside I (TBM) in DOX-induced cardiotoxicity remains ambiguous, despite growing evidence that it could reduce inflammation, oxidative stress, and apoptosis in various diseases. This study was designed to investigate the role of TBM in DOX-induced cardiotoxicity and uncover the underlying mechanisms. H9c2 cell line and C57BL/6 mice were used to construct an in vitro and in vivo model of DOX-induced myocardial injury, respectively. We observed that DOX treatment provoked inflammation, oxidative stress, and cardiomyocyte apoptosis, which were significantly alleviated by TBM administration. Mechanistically, TBM attenuated DOX-induced downregulation of sirtuin 3 (SIRT3), and SIRT3 inhibition abrogated the beneficial effects of TBM both in vitro and in vivo. In conclusion, TBM eased inflammation, oxidative stress, and apoptosis in DOX-induced cardiotoxicity by increasing the expression of SIRT3, suggesting that it holds great promise for treating DOX-induced cardiac injury.
6

Mao, Jin Ning, Ai Jun Li, Liang Ping Zhao, Lan Gao, Wei Ting Xu, Xiao Su Hong, Wen Ping Jiang, and Jian Chang Chen. "PEG-PLGA Nanoparticles Entrapping Doxorubicin Reduced Doxorubicin-Induced Cardiotoxicity in Rats." Advanced Materials Research 912-914 (April 2014): 263–68. http://dx.doi.org/10.4028/www.scientific.net/amr.912-914.263.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Aim:Doxorubicin-induced cardiotoxicity limited its clinical utilization in oncology. In this study, Dox was entrapped into PEG-PLGA Nanoparticles, cardiotoxicity of Dox or PEG-PLGA-Dox was investigated in rats. Materials and methods :PEG-PLGA-Dox was prepared via modified single emulsion method. Its characterization including size, Drug loading capacity (DLC), entrapment efficiency (EE) were estimated. The cardiotoxicity of PEG-PLGA-Dox was assessed on SD rats via echocardiography and biochemical indicators compare to free Dox and physical sodium. Results:The average diameter of PEG-PLGA-Dox is around 200 nm, with DLC about 10%.After administered PEG-PLGA-Dox, the ratio of heart weight to body weight decreased not as significant as Dox group, level of serum parameters and echocardiography parameter also decreased little compared to the Dox group. Conclusions:After entrapped into PEG-PLGA nanoparticle, Dox-induced cardiotoxicity was reduced significantly.
7

Maneechote, Chayodom, Siriporn C. Chattipakorn, and Nipon Chattipakorn. "Recent Advances in Mitochondrial Fission/Fusion-Targeted Therapy in Doxorubicin-Induced Cardiotoxicity." Pharmaceutics 15, no. 4 (April 7, 2023): 1182. http://dx.doi.org/10.3390/pharmaceutics15041182.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Doxorubicin (DOX) has been recognized as one of the most effective chemotherapies and extensively used in the clinical settings of human cancer. However, DOX-mediated cardiotoxicity is known to compromise the clinical effectiveness of chemotherapy, resulting in cardiomyopathy and heart failure. Recently, accumulation of dysfunctional mitochondria via alteration of the mitochondrial fission/fusion dynamic processes has been identified as a potential mechanism underlying DOX cardiotoxicity. DOX-induced excessive fission in conjunction with impaired fusion could severely promote mitochondrial fragmentation and cardiomyocyte death, while modulation of mitochondrial dynamic proteins using either fission inhibitors (e.g., Mdivi-1) or fusion promoters (e.g., M1) can provide cardioprotection against DOX-induced cardiotoxicity. In this review, we focus particularly on the roles of mitochondrial dynamic pathways and the current advanced therapies in mitochondrial dynamics-targeted anti-cardiotoxicity of DOX. This review summarizes all the novel insights into the development of anti-cardiotoxic effects of DOX via the targeting of mitochondrial dynamic pathways, thereby encouraging and guiding future clinical investigations to focus on the potential application of mitochondrial dynamic modulators in the setting of DOX-induced cardiotoxicity.
8

Kitakata, Hiroki, Jin Endo, Hidehiko Ikura, Hidenori Moriyama, Kohsuke Shirakawa, Yoshinori Katsumata, and Motoaki Sano. "Therapeutic Targets for DOX-Induced Cardiomyopathy: Role of Apoptosis vs. Ferroptosis." International Journal of Molecular Sciences 23, no. 3 (January 26, 2022): 1414. http://dx.doi.org/10.3390/ijms23031414.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Doxorubicin (DOX) is the most widely used anthracycline anticancer agent; however, its cardiotoxicity limits its clinical efficacy. Numerous studies have elucidated the mechanisms underlying DOX-induced cardiotoxicity, wherein apoptosis has been reported as the most common final step leading to cardiomyocyte death. However, in the past two years, the involvement of ferroptosis, a novel programmed cell death, has been proposed. The purpose of this review is to summarize the historical background that led to each form of cell death, focusing on DOX-induced cardiotoxicity and the molecular mechanisms that trigger each form of cell death. Furthermore, based on this understanding, possible therapeutic strategies to prevent DOX cardiotoxicity are outlined. DNA damage, oxidative stress, intracellular signaling, transcription factors, epigenetic regulators, autophagy, and metabolic inflammation are important factors in the molecular mechanisms of DOX-induced cardiomyocyte apoptosis. Conversely, the accumulation of lipid peroxides, iron ion accumulation, and decreased expression of glutathione and glutathione peroxidase 4 are important in ferroptosis. In both cascades, the mitochondria are an important site of DOX cardiotoxicity. The last part of this review focuses on the significance of the disruption of mitochondrial homeostasis in DOX cardiotoxicity.
9

da Cunha Menezes Souza, Leonardo, Meng Chen, Yuji Ikeno, Daisy Maria Fávero Salvadori, and Yidong Bai. "The implications of mitochondria in doxorubicin treatment of cancer in the context of traditional and modern medicine." Traditional Medicine and Modern Medicine 03, no. 04 (December 2020): 239–54. http://dx.doi.org/10.1142/s2575900020300076.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Doxorubicin (DOX) is an antibiotic anthracycline extensively used in the treatment of different malignancies, such as breast cancer, lymphomas and leukemias. The cardiotoxicity induced by DOX is one of the most important pathophysiological events that limit its clinical application. Accumulating evidence highlights mitochondria as a central role in this process. Modulation of mitochondrial functions as therapeutic strategy for DOX-induced cardiotoxicity has thus attracted much attention. In particular, emerging studies investigated the potential of natural mitochondria-targeting compounds from Traditional Chinese Medicine (TCM) as adjunct or alternative treatment for DOX-induced toxicity. This review summarizes studies about the mechanisms of DOX-induced cardiotoxicity, evidencing the importance of mitochondria and presenting TCM treatment alternatives for DOX-induced cardiomyopathy.
10

Jiang, Lai, Yanping Gong, Yida Hu, Yangyang You, Jiawu Wang, Zhetao Zhang, Zeyuan Wei, and Chaoliang Tang. "Peroxiredoxin-1 Overexpression Attenuates Doxorubicin-Induced Cardiotoxicity by Inhibiting Oxidative Stress and Cardiomyocyte Apoptosis." Oxidative Medicine and Cellular Longevity 2020 (July 29, 2020): 1–11. http://dx.doi.org/10.1155/2020/2405135.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Background. Previous research has shown that peroxiredoxin 1 (Prdx1) is an important modulator of physiological and pathophysiological cardiovascular events. This study is aimed at investigating the role and underlying mechanism of Prdx1 in doxorubicin- (DOX-) induced cardiotoxicity. Cardiac-specific expression of Prdx1 was induced in mice, and the mice received a single dose of DOX (15 mg/kg) to generate cardiotoxicity. First, our study demonstrated that Prdx1 expression was upregulated in the heart and in cardiomyocytes after DOX treatment. Second, we provided direct evidence that Prdx1 overexpression ameliorated DOX-induced cardiotoxicity by attenuating oxidative stress and cardiomyocyte apoptosis. Mechanistically, we found that DOX treatment increased the phosphorylation level of apoptosis signal-regulating kinase-1 (ASK1) and the downstream protein p38 in the heart and in cardiomyocytes, and these effects were decreased by Prdx1 overexpression. In contrast, inhibiting Prdx1 promoted DOX-induced cardiac injury via the ASK1/p38 pathway. These results suggest that Prdx1 may be an effective therapeutic option to prevent DOX-induced cardiotoxicity.

Дисертації з теми "Dox-Induced Cardiotoxicity":

1

Mammadova, Aynura. "The role of MEIS inhibitors in cardiac regeneration and protection." Electronic Thesis or Diss., Strasbourg, 2024. http://www.theses.fr/2024STRAJ006.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Анотація:
Le gène homeobox de type TALE MEIS1 a été identifié comme un facteur critique dans le contrôle de l'arrêt du cycle cellulaire des cardiomyocytes et pouvant ainsi représenter une cible thérapeutique attrayante. Nos dernières recherches ont révélé le potentiel de la suppression de MEIS1 dans la promotion de la régénération des cardiomyocytes. Des expériences effectuées avec des cardiomyocytes néonataux ont montré que deux petites molécules innovantes, MEISi-1 et MEISi-2, ont amélioré la prolifération (cellules Ph3+TnnT) et la cytokinèse (cellules AuroraB+TnnT) de ces cellules. La suppression de l'activité de MEIS1 a entraîné une diminution de l'expression de ces gènes cibles et des inhibiteurs des kinases dépendantes des cyclines. De plus, cette recherche s'est également intéressée à la différentiation de cellules souches pluripotentes induites humaines (hiPSCs) en cardiomyocytes pour examiner l'impact de la suppression de MEIS1, sans modification de leur viabilité. Fait intéressant, un traitement à court et à long terme avec MEISi des hiPSCs a conduit à une élévation significative de l'expression des gènes spécifiques cardiaques essentiels, influençant notablement le mésoderme cardiaque et les cellules progénitrices, et positionnant les inhibiteurs de MEIS1 comme des modulateurs cruciaux de l'expression génique cardiaque. Nos résultats indiquent que les inhibiteurs de MEIS peuvent offrir une protection contre les effets cardiotoxiques aigus de la doxorubicine (DOX) chez les rats Wistar, comme en témoigne la structure préservée du tissu cardiaque et les niveaux inchangés de fibrose ou de collagène. Les analyses qPCR ont en outre confirmé la surexpression des gènes des progéniteurs cardiaques et un équilibre dans l'expression génique anti-apoptotique et liée aux ROS, suggérant un rôle protecteur des inhibiteurs de MEIS contre les dommages induits par la DOX sans influencer la fibrose. Ces résultats soulignent le potentiel thérapeutique des inhibiteurs de MEIS en cardiologie régénérative, suggérant leur utilité dans l'amélioration du renouvellement des cardiomyocytes et offrant une protection contre la cardiotoxicité
The TALE-type homeobox gene MEIS1 has been identified as a critical factor in controlling the cell cycle arrest of cardiomyocytes, presenting itself as an attractive target for therapy. Our latest investigations have revealed the potential of MEIS1 suppression to promote the regeneration of cardiomyocytes. Further experiments with neonatal cardiomyocytes showed that two innovative small molecules, MEISi-1 and MEISi-2, enhanced the proliferation (Ph3+TnnT cells) and cytokinesis (AuroraB+TnnT cells) of these cells. Suppressing MEIS1 activity resulted in the diminished expression of its target genes and the inhibitors of cyclin-dependent kinases. Additionally, this research extended to cultivating human induced pluripotent stem cells (hiPSCs) into cardiomyocytes to examine the impact of MEIS1 suppression, which notably did not compromise their viability. Intriguingly, short-term and long-term treatment with MEISi in hiPSCs led to significant elevation in essential cardiac-specific gene expression, notably influencing cardiac mesoderm and progenitor cells, and positioning MEIS1 inhibitors as crucial modulators of cardiac gene expression. Our findings indicate that MEIS inhibitors can provide protection against the acute cardiotoxic effects of doxorubicin (DOX) in Wistar rats, as evidenced by the maintained structure of cardiac tissue and unchanged levels of fibrosis or collagen. qPCR analyses further confirmed the upregulation of cardiac progenitor genes and a balance in anti-apoptotic and ROS-related gene expression, hinting at the protective role of MEIS inhibitors against DOX-induced damage without influencing fibrosis. These results highlight the therapeutic potential of MEIS inhibitors in regenerative cardiology, suggesting their utility in enhancing cardiomyocyte renewal and offering protection against cardiotoxicity

Тези доповідей конференцій з теми "Dox-Induced Cardiotoxicity":

1

Tao, Rong-Hua, Masato Kobayashi, Fei Wang, Yuanzheng Yang, and Eugenie S. Kleinerman. "Abstract 5422: Dissecting the mechanism of exercise-mediated protection of Dox-induced cardiotoxicity." In Proceedings: AACR Annual Meeting 2020; April 27-28, 2020 and June 22-24, 2020; Philadelphia, PA. American Association for Cancer Research, 2020. http://dx.doi.org/10.1158/1538-7445.am2020-5422.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.

До бібліографії