1
|
Fonseka O, Gare SR, Chen X, Zhang J, Alatawi NH, Ross C, Liu W. Molecular Mechanisms Underlying Heart Failure and Their Therapeutic Potential. Cells 2025; 14:324. [PMID: 40072053 PMCID: PMC11899429 DOI: 10.3390/cells14050324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/07/2025] [Accepted: 02/17/2025] [Indexed: 03/15/2025] Open
Abstract
Heart failure (HF) is a prominent fatal cardiovascular disorder afflicting 3.4% of the adult population despite the advancement of treatment options. Therefore, a better understanding of the pathogenesis of HF is essential for exploring novel therapeutic strategies. Hypertrophy and fibrosis are significant characteristics of pathological cardiac remodeling, contributing to HF. The mechanisms involved in the development of cardiac remodeling and consequent HF are multifactorial, and in this review, the key underlying mechanisms are discussed. These have been divided into the following categories thusly: (i) mitochondrial dysfunction, including defective dynamics, energy production, and oxidative stress; (ii) cardiac lipotoxicity; (iii) maladaptive endoplasmic reticulum (ER) stress; (iv) impaired autophagy; (v) cardiac inflammatory responses; (vi) programmed cell death, including apoptosis, pyroptosis, and ferroptosis; (vii) endothelial dysfunction; and (viii) defective cardiac contractility. Preclinical data suggest that there is merit in targeting the identified pathways; however, their clinical implications and outcomes regarding treating HF need further investigation in the future. Herein, we introduce the molecular mechanisms pivotal in the onset and progression of HF, as well as compounds targeting the related mechanisms and their therapeutic potential in preventing or rescuing HF. This, therefore, offers an avenue for the design and discovery of novel therapies for the treatment of HF.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Wei Liu
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; (O.F.); (S.R.G.); (X.C.); (J.Z.); (N.H.A.)
| |
Collapse
|
2
|
Guo L, Chen W, Yue J, Gao M, Zhang J, Huang Y, Xiong H, Li X, Wang Y, Yuan Y, Chen L, Fei F, Xu R. Unlocking the potential of LHPP: Inhibiting glioma growth and cell cycle via the MDM2/p53 pathway. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167509. [PMID: 39277057 DOI: 10.1016/j.bbadis.2024.167509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 08/31/2024] [Accepted: 09/07/2024] [Indexed: 09/17/2024]
Abstract
The recurrence of glioma after treatment has remained an intractable problem for many years. Recently, numerous studies have explored the pivotal role of the mouse double minute 2 (MDM2)/p53 pathway in cancer treatment. Lysine phosphate phosphohistidine inorganic pyrophosphate phosphatase (LHPP), a newly discovered tumor suppressor, has been confirmed in numerous studies on tumors, but its role in glioma remains poorly understood. Expression matrices in The Cancer Genome Atlas (TCGA) and Chinese Glioma Genome Atlas (CGGA) databases were analyzed using gene set enrichment analysis (GSEA), revealing significant alterations in the p53 pathway among glioma patients with high LHPP expression. The overexpression of LHPP in glioma cells resulted in a reduction in cell proliferation, migration, and invasive ability, as well as an increase in apoptosis and alterations to the cell cycle. The present study has identified a novel inhibitory mechanism of LHPP against glioma, both in vivo and in vitro. The results demonstrate that LHPP exerts anti-glioma effects via the MDM2/p53 pathway. These findings may offer a new perspective for the treatment of glioma in the clinic.
Collapse
Affiliation(s)
- Lili Guo
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Wenjin Chen
- Department of Neurosurgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jiong Yue
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Mingjun Gao
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Jin Zhang
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yukai Huang
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Huan Xiong
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Xinda Li
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yangyang Wang
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Ying Yuan
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Longyi Chen
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| | - Fan Fei
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| | - Ruxiang Xu
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
3
|
Borlak J, Ciribilli Y, Bisio A, Selvaraj S, Inga A, Oh JH, Spanel R. The Abl1 tyrosine kinase is a key player in doxorubicin-induced cardiomyopathy and its p53/p73 cell death mediated signaling differs in atrial and ventricular cardiomyocytes. J Transl Med 2024; 22:845. [PMID: 39285385 PMCID: PMC11403941 DOI: 10.1186/s12967-024-05623-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/16/2024] [Indexed: 09/20/2024] Open
Abstract
BACKGROUND Doxorubicin is an important anticancer drug, however, elicits dose-dependently cardiomyopathy. Given its mode of action, i.e. topoisomerase inhibition and DNA damage, we investigated genetic events associated with cardiomyopathy and searched for mechanism-based possibilities to alleviate cardiotoxicity. We treated rats at clinically relevant doses of doxorubicin. Histopathology and transmission electron microscopy (TEM) defined cardiac lesions, and transcriptomics unveiled cardiomyopathy-associated gene regulations. Genomic-footprints revealed critical components of Abl1-p53-signaling, and EMSA-assays evidenced Abl1 DNA-binding activity. Gene reporter assays confirmed Abl1 activity on p53-targets while immunohistochemistry/immunofluorescence microscopy demonstrated Abl1, p53&p73 signaling. RESULTS Doxorubicin treatment caused dose-dependently toxic cardiomyopathy, and TEM evidenced damaged mitochondria and myofibrillar disarray. Surviving cardiomyocytes repressed Parkin-1 and Bnip3-mediated mitophagy, stimulated dynamin-1-like dependent mitochondrial fission and induced anti-apoptotic Bag1 signaling. Thus, we observed induced mitochondrial biogenesis. Transcriptomics discovered heterogeneity in cellular responses with minimal overlap between treatments, and the data are highly suggestive for distinct cardiomyocyte (sub)populations which differed in their resilience and reparative capacity. Genome-wide footprints revealed Abl1 and p53 enriched binding sites in doxorubicin-regulated genes, and we confirmed Abl1 DNA-binding activity in EMSA-assays. Extraordinarily, Abl1 signaling differed in the heart with highly significant regulations of Abl1, p53 and p73 in atrial cardiomyocytes. Conversely, in ventricular cardiomyocytes, Abl1 solely-modulated p53-signaling that was BAX transcription-independent. Gene reporter assays established Abl1 cofactor activity for the p53-reporter PG13-luc, and ectopic Abl1 expression stimulated p53-mediated apoptosis. CONCLUSIONS The tyrosine kinase Abl1 is of critical importance in doxorubicin induced cardiomyopathy, and we propose its inhibition as means to diminish risk of cardiotoxicity.
Collapse
Affiliation(s)
- Jürgen Borlak
- Centre for Pharmacology and Toxicology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| | - Yari Ciribilli
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Alessandra Bisio
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Saravanakumar Selvaraj
- Centre for Pharmacology and Toxicology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Alberto Inga
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Jung-Hwa Oh
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, Republic of Korea
| | - Reinhard Spanel
- Centre for Pharmacology and Toxicology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| |
Collapse
|
4
|
He X, Cantrell AC, Williams QA, Gu W, Chen Y, Chen JX, Zeng H. p53 Acetylation Exerts Critical Roles in Pressure Overload-Induced Coronary Microvascular Dysfunction and Heart Failure in Mice. Arterioscler Thromb Vasc Biol 2024; 44:826-842. [PMID: 38328937 PMCID: PMC10978286 DOI: 10.1161/atvbaha.123.319601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 01/25/2024] [Indexed: 02/09/2024]
Abstract
BACKGROUND Coronary microvascular dysfunction (CMD) has been shown to contribute to cardiac hypertrophy and heart failure (HF) with preserved ejection fraction. At this point, there are no proven treatments for CMD. METHODS We have shown that histone acetylation may play a critical role in the regulation of CMD. By using a mouse model that replaces lysine with arginine at residues K98, K117, K161, and K162R of p53 (p534KR), preventing acetylation at these sites, we test the hypothesis that acetylation-deficient p534KR could improve CMD and prevent the progression of hypertensive cardiac hypertrophy and HF. Wild-type and p534KR mice were subjected to pressure overload by transverse aortic constriction to induce cardiac hypertrophy and HF. RESULTS Echocardiography measurements revealed improved cardiac function together with a reduction of apoptosis and fibrosis in p534KR mice. Importantly, myocardial capillary density and coronary flow reserve were significantly improved in p534KR mice. Moreover, p534KR upregulated the expression of cardiac glycolytic enzymes and Gluts (glucose transporters), as well as the level of fructose-2,6-biphosphate; increased PFK-1 (phosphofructokinase 1) activity; and attenuated cardiac hypertrophy. These changes were accompanied by increased expression of HIF-1α (hypoxia-inducible factor-1α) and proangiogenic growth factors. Additionally, the levels of SERCA-2 were significantly upregulated in sham p534KR mice, as well as in p534KR mice after transverse aortic constriction. In vitro, p534KR significantly improved endothelial cell glycolytic function and mitochondrial respiration and enhanced endothelial cell proliferation and angiogenesis. Similarly, acetylation-deficient p534KR significantly improved coronary flow reserve and rescued cardiac dysfunction in SIRT3 (sirtuin 3) knockout mice. CONCLUSIONS Our data reveal the importance of p53 acetylation in coronary microvascular function, cardiac function, and remodeling and may provide a promising approach to improve hypertension-induced CMD and to prevent the transition of cardiac hypertrophy to HF.
Collapse
Affiliation(s)
- Xiaochen He
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, School of Medicine, Jackson, MS, 39216, USA
| | - Aubrey C Cantrell
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, School of Medicine, Jackson, MS, 39216, USA
| | - Quinesha A Williams
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, School of Medicine, Jackson, MS, 39216, USA
| | - Wei Gu
- Department of Pathology & Cell Biology, Columbia University, Institute for Cancer Genetics, New York, NY 10032, USA
| | - Yingjie Chen
- Department of Physiology & Biophysics, University of Mississippi Medical Center, School of Medicine, Jackson, MS, 39216, USA
| | - Jian-Xiong Chen
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, School of Medicine, Jackson, MS, 39216, USA
| | - Heng Zeng
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, School of Medicine, Jackson, MS, 39216, USA
| |
Collapse
|
5
|
Hu Z. Exploring the mechanism of curcumin in the treatment of doxorubicin-induced cardiotoxicity based on network pharmacology and molecular docking technology. Medicine (Baltimore) 2024; 103:e36593. [PMID: 38363942 PMCID: PMC10869047 DOI: 10.1097/md.0000000000036593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 11/21/2023] [Indexed: 02/18/2024] Open
Abstract
Doxorubicin (DOX) is one of the most effective chemotherapeutic agents. However, the nonselective effect leads to serious cardiotoxicity risk in clinical use. Curcumin is a well-known dietary polyphenol that showed a protective effect against the cardiotoxic effect of DOX. This study aimed to assess the role of curcumin in protection against DOX-induced cardiotoxicity. Potential compound and disease targets were obtained from relevant databases, and common targets were screened. Protein-protein interaction (PPI) was used to predict the core targets. Gene ontology (GO) bioprocess analysis and Kyoto encyclopedia of genes and genome enrichment analysis enriched the possible biological processes (BP), cellular components, molecular function, and signaling pathways involved. Finally, the binding of curcumin to target proteins was evaluated through molecular docking. The docking score verified the reliability of the prediction results. In total, 205 curcumin and 700 disease targets were identified. A topological analysis of the PPI network revealed 10 core targets including TP53, tumor necrosis factor-alpha (TNF), AKT1, vascular endothelial growth factor A (VEGFA), prostaglandin-endoperoxide synthase 2 (PTGS2), signal transducer and activator of the transcription 3 (STAT3), HIF1A, MYC, epidermal growth factor receptor (EGFR), and CASP3 (Caspase-3). Furthermore, the enrichment analyses indicated that the effects of curcumin were mediated by genes related to oxidation, inflammation, toxification, cell proliferation, migration, apoptosis, wounding, metabolism, proteolysis, and the signaling pathway of calcium (Ca2+). Molecular docking showed that curcumin could bind with the target proteins with strong molecular force, exhibiting good docking activity. Curcumin has a multi-cardioprotective effect by modulating the core targets' expression in DOX-induced cardiotoxicity. This study elucidated the key target proteins and provided a theoretical basis for further exploring curcumin in the prevention and treatment of DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Zhen Hu
- Department of Electrocardiography, Wuhan No.1 Hospital, Wuhan, China
| |
Collapse
|
6
|
Yu W, Xu H, Sun Z, Du Y, Sun S, Abudureyimu M, Zhang M, Tao J, Ge J, Ren J, Zhang Y. TBC1D15 deficiency protects against doxorubicin cardiotoxicity via inhibiting DNA-PKcs cytosolic retention and DNA damage. Acta Pharm Sin B 2023; 13:4823-4839. [PMID: 38045047 PMCID: PMC10692480 DOI: 10.1016/j.apsb.2023.09.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 07/28/2023] [Accepted: 08/09/2023] [Indexed: 12/05/2023] Open
Abstract
Clinical application of doxorubicin (DOX) is heavily hindered by DOX cardiotoxicity. Several theories were postulated for DOX cardiotoxicity including DNA damage and DNA damage response (DDR), although the mechanism(s) involved remains to be elucidated. This study evaluated the potential role of TBC domain family member 15 (TBC1D15) in DOX cardiotoxicity. Tamoxifen-induced cardiac-specific Tbc1d15 knockout (Tbc1d15CKO) or Tbc1d15 knockin (Tbc1d15CKI) male mice were challenged with a single dose of DOX prior to cardiac assessment 1 week or 4 weeks following DOX challenge. Adenoviruses encoding TBC1D15 or containing shRNA targeting Tbc1d15 were used for Tbc1d15 overexpression or knockdown in isolated primary mouse cardiomyocytes. Our results revealed that DOX evoked upregulation of TBC1D15 with compromised myocardial function and overt mortality, the effects of which were ameliorated and accentuated by Tbc1d15 deletion and Tbc1d15 overexpression, respectively. DOX overtly evoked apoptotic cell death, the effect of which was alleviated and exacerbated by Tbc1d15 knockout and overexpression, respectively. Meanwhile, DOX provoked mitochondrial membrane potential collapse, oxidative stress and DNA damage, the effects of which were mitigated and exacerbated by Tbc1d15 knockdown and overexpression, respectively. Further scrutiny revealed that TBC1D15 fostered cytosolic accumulation of the cardinal DDR element DNA-dependent protein kinase catalytic subunit (DNA-PKcs). Liquid chromatography-tandem mass spectrometry and co-immunoprecipitation denoted an interaction between TBC1D15 and DNA-PKcs at the segment 594-624 of TBC1D15. Moreover, overexpression of TBC1D15 mutant (∆594-624, deletion of segment 594-624) failed to elicit accentuation of DOX-induced cytosolic retention of DNA-PKcs, DNA damage and cardiomyocyte apoptosis by TBC1D15 wild type. However, Tbc1d15 deletion ameliorated DOX-induced cardiomyocyte contractile anomalies, apoptosis, mitochondrial anomalies, DNA damage and cytosolic DNA-PKcs accumulation, which were canceled off by DNA-PKcs inhibition or ATM activation. Taken together, our findings denoted a pivotal role for TBC1D15 in DOX-induced DNA damage, mitochondrial injury, and apoptosis possibly through binding with DNA-PKcs and thus gate-keeping its cytosolic retention, a route to accentuation of cardiac contractile dysfunction in DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Wenjun Yu
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan 430071, China
| | - Haixia Xu
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Zhe Sun
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Yuxin Du
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Shiqun Sun
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Miyesaier Abudureyimu
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
- Cardiovascular Department, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Shanghai 200030, China
| | - Mengjiao Zhang
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Jun Tao
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510000, China
| | - Junbo Ge
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Jun Ren
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Yingmei Zhang
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| |
Collapse
|
7
|
Al-Masri A. Apoptosis and long non-coding RNAs: Focus on their roles in Heart diseases. Pathol Res Pract 2023; 251:154889. [PMID: 38238070 DOI: 10.1016/j.prp.2023.154889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 01/23/2024]
Abstract
Heart disease is one of the principal death reasons around the world and there is a growing requirement to discover novel healing targets that have the potential to avert or manage these illnesses. On the other hand, apoptosis is a strongly controlled, cell removal procedure that has a crucial part in numerous cardiac problems, such as reperfusion injury, MI (myocardial infarction), consecutive heart failure, and inflammation of myocardium. Completely comprehending the managing procedures of cell death signaling is critical as it is the primary factor that influences patient mortality and morbidity, owing to cardiomyocyte damage. Indeed, the prevention of heart cell death appears to be a viable treatment approach for heart illnesses. According to current researches, a number of long non-coding RNAs cause the heart cells death via different methods that are embroiled in controlling the activity of transcription elements, the pathways that signals transmission within cells, small miRNAs, and the constancy of proteins. When there is too much cell death in the heart, it can cause problems like reduced blood flow, heart damage after restoring blood flow, heart disease in diabetics, and changes in the heart after reduced blood flow. Therefore, studying how lncRNAs control apoptosis could help us find new treatments for heart diseases. In this review, we present recent discoveries about how lncRNAs are involved in causing cell death in different cardiovascular diseases.
Collapse
Affiliation(s)
- Abeer Al-Masri
- Department of Physiology, College of Medicine, King Saud University, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
8
|
Wang H, Yu W, Wang Y, Wu R, Dai Y, Deng Y, Wang S, Yuan J, Tan R. p53 contributes to cardiovascular diseases via mitochondria dysfunction: A new paradigm. Free Radic Biol Med 2023; 208:846-858. [PMID: 37776918 DOI: 10.1016/j.freeradbiomed.2023.09.036] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/21/2023] [Accepted: 09/28/2023] [Indexed: 10/02/2023]
Abstract
Cardiovascular diseases (CVDs) are leading causes of global mortality; however, their underlying mechanisms remain unclear. The tumor suppressor factor p53 has been extensively studied for its role in cancer and is also known to play an important role in regulating CVDs. Abnormal p53 expression levels and modifications contribute to the occurrence and development of CVDs. Additionally, mounting evidence underscores the critical involvement of mitochondrial dysfunction in CVDs. Notably, studies indicate that p53 abnormalities directly correlate with mitochondrial dysfunction and may even interact with each other. Encouragingly, small molecule inhibitors targeting p53 have exhibited remarkable effects in animal models of CVDs. Moreover, therapeutic strategies aimed at mitochondrial-related molecules and mitochondrial replacement therapy have demonstrated their advantageous potential. Therefore, targeting p53 or mitochondria holds immense promise as a pioneering therapeutic approach for combating CVDs. In this comprehensive review, we delve into the mechanisms how p53 influences mitochondrial dysfunction, including energy metabolism, mitochondrial oxidative stress, mitochondria-induced apoptosis, mitochondrial autophagy, and mitochondrial dynamics, in various CVDs. Furthermore, we summarize and discuss the potential significance of targeting p53 or mitochondria in the treatment of CVDs.
Collapse
Affiliation(s)
- Hao Wang
- School of Clinical Medicine, Xuzhou Medical University, Xuzhou, 221004, China
| | - Wei Yu
- School of Clinical Medicine, Xuzhou Medical University, Xuzhou, 221004, China
| | - Yibo Wang
- School of Clinical Medicine, Xuzhou Medical University, Xuzhou, 221004, China
| | - Ruihao Wu
- School of Clinical Medicine, Xuzhou Medical University, Xuzhou, 221004, China
| | - Yifei Dai
- School of Stomatology, Xuzhou Medical University, Xuzhou, 221004, China
| | - Ye Deng
- School of Stomatology, Xuzhou Medical University, Xuzhou, 221004, China
| | - Shijun Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200433, China.
| | - Jinxiang Yuan
- The Collaborative Innovation Center, Jining Medical University, Jining, 272000, China.
| | - Rubin Tan
- Department of Physiology, Basic Medical School, Xuzhou Medical University, Xuzhou, 221004, China.
| |
Collapse
|
9
|
Gambardella J, Santulli G, Fiordelisi A, Cerasuolo FA, Wang X, Prevete N, Sommella E, Avvisato R, Buonaiuto A, Altobelli GG, Rinaldi L, Chiuso F, Feliciello A, Dal Piaz F, Campiglia P, Ciccarelli M, Morisco C, Sadoshima J, Iaccarino G, Sorriento D. Infiltrating macrophages amplify doxorubicin-induced cardiac damage: role of catecholamines. Cell Mol Life Sci 2023; 80:323. [PMID: 37819449 PMCID: PMC10567889 DOI: 10.1007/s00018-023-04922-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/12/2023] [Accepted: 08/13/2023] [Indexed: 10/13/2023]
Abstract
BACKGROUND The functional contribution of non-myocyte cardiac cells, such as inflammatory cells, in the setup of heart failure in response to doxorubicin (Dox) is recently becoming of growing interest. OBJECTIVES The study aims to evaluate the role of macrophages in cardiac damage elicited by Dox treatment. METHODS C57BL/6 mice were treated with one intraperitoneal injection of Dox (20 mg/kg) and followed up for 5 days by cardiac ultrasounds (CUS), histological, and flow cytometry evaluations. We also tested the impact of Dox in macrophage-depleted mice. Rat cardiomyoblasts were directly treated with Dox (D-Dox) or with a conditioned medium from cultured murine macrophages treated with Dox (M-Dox). RESULTS In response to Dox, macrophage infiltration preceded cardiac damage. Macrophage depletion prevents Dox-induced damage, suggesting a key role of these cells in promoting cardiotoxicity. To evaluate the crosstalk between macrophages and cardiac cells in response to DOX, we compared the effects of D-Dox and M-Dox in vitro. Cell vitality was lower in cardiomyoblasts and apoptosis was higher in response to M-Dox compared with D-Dox. These events were linked to p53-induced mitochondria morphology, function, and autophagy alterations. We identify a mechanistic role of catecholamines released by Dox-activated macrophages that lead to mitochondrial apoptosis of cardiac cells through β-AR stimulation. CONCLUSIONS Our data indicate that crosstalk between macrophages and cardiac cells participates in cardiac damage in response to Dox.
Collapse
Affiliation(s)
- Jessica Gambardella
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
- Department of Medicine (Cardiology) and Department of Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Montefiore University Hospital, New York, USA
| | - Gaetano Santulli
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
- Department of Medicine (Cardiology) and Department of Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Montefiore University Hospital, New York, USA
- Hypertension Research Center (CIRIAPA), Federico II University, Naples, Italy
| | - Antonella Fiordelisi
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | | | - Xujun Wang
- Department of Medicine (Cardiology) and Department of Molecular Pharmacology, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Montefiore University Hospital, New York, USA
| | - Nella Prevete
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
- Institute of Experimental Endocrinology and Oncology (IEOS), CNR, Naples, Italy
| | - Eduardo Sommella
- Department of Pharmacy, University of Salerno, Fisciano (Salerno), Italy
| | - Roberta Avvisato
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Antonietta Buonaiuto
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | | | - Laura Rinaldi
- Department of Molecular Medicine and Medical Biotechnology, Federico II University, Naples, Italy
| | - Francesco Chiuso
- Department of Molecular Medicine and Medical Biotechnology, Federico II University, Naples, Italy
| | - Antonio Feliciello
- Department of Molecular Medicine and Medical Biotechnology, Federico II University, Naples, Italy
| | - Fabrizio Dal Piaz
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno (Salerno), Baronissi, Italy
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Fisciano (Salerno), Italy
| | - Michele Ciccarelli
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno (Salerno), Baronissi, Italy
| | - Carmine Morisco
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Guido Iaccarino
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
- Hypertension Research Center (CIRIAPA), Federico II University, Naples, Italy
| | - Daniela Sorriento
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy.
- Hypertension Research Center (CIRIAPA), Federico II University, Naples, Italy.
| |
Collapse
|
10
|
Samir R, Hassan EA, Saber AA, Haneen DSA, Saleh EM. Seaweed Sargassum aquifolium extract ameliorates cardiotoxicity induced by doxorubicin in rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:58226-58242. [PMID: 36977879 PMCID: PMC10163098 DOI: 10.1007/s11356-023-26259-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 02/28/2023] [Indexed: 05/08/2023]
Abstract
Doxorubicin (DOX) is a potent anticancer drug with adverse cardiotoxic effects. Alginates are multifunctional biopolymers and polyelectrolytes derived from the cell walls of brown seaweeds. They are nontoxic, biocompatible, and biodegradable, and hence, utilized in several biomedical and pharmaceutical applications. Here, we investigated the potential cardioprotective effect of thermally treated sodium alginate (TTSA), which was extracted and purified from the seaweed Sargassum aquifolium, in treating acute DOX cardiotoxicity and apoptotic pathways in rats. UV-visible spectroscopy, Fourier-transform infrared, and nuclear magnetic resonance (1H-NMR) spectroscopy techniques were used to characterize TTSA. CK-MB and AST levels in sera samples were determined. The expression levels of Erk-2 (MAPK-1) and iNOS genes were investigated by quantitative real-time polymerase chain reaction (qRT-PCR). The protein expression levels of Erk-2, anti-apoptotic p53, and caspase-3 were analyzed using western blotting and ELISA. For the in vivo studies, sixty rats were randomly divided equally into six groups and treated with DOX, followed by TTSA. We revealed that treatment with TTSA, which has low molecular weight and enhanced antioxidant properties, improved DOX-mediated cardiac dysfunction and alleviated DOX-induced myocardial apoptosis. Furthermore, TTSA exhibited a cardioprotective effect against DOX-induced cardiac toxicity, indicated by the increased expression of MAPK-1 (Erk2) and iNOS genes, which are implicated in the adaptive responses regulating DOX-induced myocardial damage. Moreover, TTSA significantly (p < 0.05) suppressed caspase-3 and upregulated anti-apoptotic protein p53 expression. TTSA also rebalanced the cardiomyocyte redox potential by significantly (p < 0.05) increasing the levels of endogenous antioxidant enzymes, including catalase and superoxide dismutase. Our findings suggest that TTSA, particularly at a dose of 400 mg/kg b.w., is a potential prophylactic supplement for treating acute DOX-linked cardiotoxicity.
Collapse
Affiliation(s)
- Rania Samir
- Biochemistry Department, Faculty of Science, Ain Shams University, Abbassia, Cairo, 11566 Egypt
| | - Ekrami A. Hassan
- Biochemistry Department, Faculty of Science, Ain Shams University, Abbassia, Cairo, 11566 Egypt
| | - Abdullah A. Saber
- Botany Department, Faculty of Science, Ain Shams University, Abbassia Square, Cairo, 11566 Egypt
| | - David S. A. Haneen
- Chemistry Department, Faculty of Science, Ain Shams University, Abbassia, Cairo, 11566 Egypt
| | - Eman M. Saleh
- Biochemistry Department, Faculty of Science, Ain Shams University, Abbassia, Cairo, 11566 Egypt
| |
Collapse
|
11
|
He X, Cantrell AC, Williams QA, Gu W, Chen Y, Chen JX, Zeng H. P53 Acetylation Exerts Critical Roles In Pressure Overload Induced Coronary Microvascular Dysfunction and Heart Failure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.08.527691. [PMID: 36798200 PMCID: PMC9934706 DOI: 10.1101/2023.02.08.527691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Coronary microvascular dysfunction (CMD) has been shown to contribute to cardiac hypertrophy and heart failure with preserved ejection fraction. At this point, there are no proven treatments for CMD. We have shown that histone acetylation may play a critical role in the regulation of CMD. By using a mouse model that replaces lysine with arginine at residues K98/117/161/162R of p53 (p534KR), preventing acetylation at these sites, we test the hypothesis that acetylation-deficient p534KR could improve coronary microvascular dysfunction and prevent the progression of hypertensive cardiac hypertrophy and heart failure. Wild-type (WT) and p534KR mice were subjected to pressure overload (PO) by transverse aortic constriction to induce cardiac hypertrophy and heart failure (HF). Echocardiography measurements revealed improved cardiac function together with reduction of apoptosis and fibrosis in p534KR mice. Importantly, myocardial capillary density and coronary flow reserve (CFR) were significantly improved in p534KR mice. Moreover, p534KR upregulated the expression of cardiac glycolytic enzymes and glucose transporters, as well as the level of fructose-2,6-biphosphate; increased PFK-1 activity; and attenuated cardiac hypertrophy. These changes were accompanied by increased expression of HIF-1α and proangiogenic growth factors. Additionally, the levels of SERCA-2 were significantly upregulated in sham p534KR mice as well as in p534KR mice after TAC. In vitro, p534KR significantly improved endothelial cell (EC) glycolytic function and mitochondrial respiration, and enhanced EC proliferation and angiogenesis. Similarly, acetylation-deficient p534KR significantly improved CFR and rescued cardiac dysfunction in SIRT3 KO mice. Our data reveal the importance of p53 acetylation in coronary microvascular function, cardiac function, and remodeling, and may provide a promising approach to improve hypertension-induced coronary microvascular dysfunction (CMD) and to prevent the transition of cardiac hypertrophy to heart failure.
Collapse
Affiliation(s)
- Xiaochen He
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, School of Medicine, Jackson, MS, 39216, USA
| | - Aubrey C Cantrell
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, School of Medicine, Jackson, MS, 39216, USA
| | - Quinesha A Williams
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, School of Medicine, Jackson, MS, 39216, USA
| | - Wei Gu
- Department of Pathology & Cell Biology, Columbia University, Institute for Cancer Genetics, New York, NY 10032, USA
| | - Yingjie Chen
- Department of Physiology & Biophysics, University of Mississippi Medical Center, School of Medicine, Jackson, MS, 39216, USA
| | - Jian-Xiong Chen
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, School of Medicine, Jackson, MS, 39216, USA
| | - Heng Zeng
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, School of Medicine, Jackson, MS, 39216, USA
| |
Collapse
|
12
|
Nemoto H, Umemura M, Suzuki F, Nagasako A, Nagao K, Hidaka Y, Nakakaji R, Uchida K, Suzuki S, Masuda M, Ishikawa Y. Store-operated calcium entry via ORAI1 regulates doxorubicin-induced apoptosis and prevents cardiotoxicity in cardiac fibroblasts. PLoS One 2022; 17:e0278613. [PMID: 36472998 PMCID: PMC9725120 DOI: 10.1371/journal.pone.0278613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 11/18/2022] [Indexed: 12/12/2022] Open
Abstract
Despite exhibiting cardiotoxicity, doxorubicin (DOX) is widely used for cancer treatments. Cardiac fibroblasts (CFs) are important in the pathogenesis of heart failure. This necessitates the study of the effect of DOX on CFs. The impairment of calcium (Ca2+) homeostasis is a common mechanism of heart failure. Store-operated Ca2+ entry (SOCE) is a receptor-regulated Ca2⁺ entry pathway that maintains calcium balance by sensing reduced calcium stores in the endoplasmic reticulum. ORAI1, a calcium channel protein and the most important component of SOCE, is highly expressed in human cardiac fibroblasts (HCFs). It is upregulated in CFs from failing ventricles. However, whether ORAI1 in HCFs is increased and/or plays a role in DOX-induced cardiotoxicity remains unknown. In this study, we aimed to elucidate the relationship between ORAI1/SOCE and DOX-induced heart failure. Induction of apoptosis by DOX was characterized in HCFs. Apoptosis and cell cycle analyses were performed by fluorescence-activated cell sorting (FACS). Reactive oxygen species (ROS) production was measured using fluorescence. YM-58483 was used as an ORAI1/SOCE inhibitor. ORAI1-knockdown cells were established by RNA interference. In vivo experiments were performed by intraperitoneally injecting YM-58483 and DOX into mice. We first demonstrated that DOX significantly increased the protein expression level of p53 in HCFs by western blotting. FACS analysis revealed that DOX increased early apoptosis and induced cell cycle arrest in the G2 phase in fibroblasts. DOX also increased ROS production. DOX significantly increased the expression level of ORAI1 in CFs. Both YM-58483 and ORAI1 gene knockdown attenuated DOX-induced apoptosis. Similarly, YM-58483 attenuated cell cycle arrest in the G2 phase, and ORAI1 knockdown attenuated DOX-induced ROS production in HCFs. In the animal experiment, YM-58483 attenuated DOX-induced apoptosis. In HCFs, ORAI1/SOCE regulates p53 expression and plays an important role in DOX-induced cardiotoxicity. ORAI1 may serve as a new target for preventing DOX-induced heart failure.
Collapse
Affiliation(s)
- Hiroko Nemoto
- Cardiovascular Research Institute, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
- Department of Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Masanari Umemura
- Cardiovascular Research Institute, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
- * E-mail: (MU); (YI)
| | - Fumina Suzuki
- Cardiovascular Research Institute, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Akane Nagasako
- Cardiovascular Research Institute, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Kagemichi Nagao
- Cardiovascular Research Institute, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Yuko Hidaka
- Cardiovascular Research Institute, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Rina Nakakaji
- Cardiovascular Research Institute, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Keiji Uchida
- Department of Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Shinichi Suzuki
- Department of Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Munetaka Masuda
- Department of Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Yoshihiro Ishikawa
- Cardiovascular Research Institute, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
- * E-mail: (MU); (YI)
| |
Collapse
|
13
|
Seara FAC, Kasai-Brunswick TH, Nascimento JHM, Campos-de-Carvalho AC. Anthracycline-induced cardiotoxicity and cell senescence: new therapeutic option? Cell Mol Life Sci 2022; 79:568. [PMID: 36287277 PMCID: PMC11803035 DOI: 10.1007/s00018-022-04605-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/30/2022] [Accepted: 10/17/2022] [Indexed: 11/30/2022]
Abstract
Anthracyclines are chemotherapeutic drugs widely used in the frontline of cancer treatment. The therapeutic mechanisms involve the stabilization of topoisomerase IIα, DNA, and the anthracycline molecule in a ternary complex that is recognized as DNA damage. Redox imbalance is another vital source of oxidative DNA damage. Together, these mechanisms lead to cytotoxic effects in neoplastic cells. However, anthracycline treatment can elicit cardiotoxicity and heart failure despite the therapeutic benefits. Topoisomerase IIβ and oxidative damage in cardiac cells have been the most reported pathophysiological mechanisms. Alternatively, cardiac cells can undergo stress-induced senescence when exposed to anthracyclines, a state primarily characterized by cell cycle arrest, organelle dysfunction, and a shift to senescence-associated secretory phenotype (SASP). The SASP can propagate senescence to neighboring cells in an ongoing process that leads to the accumulation of senescent cells, promoting cellular dysfunction and extracellular matrix remodeling. Therefore, the accumulation of senescent cardiac cells is an emerging pathophysiological mechanism associated with anthracycline-induced cardiotoxicity. This paradigm also raises the potential for therapeutic approaches to clear senescent cells in treating anthracycline-induced cardiotoxicity (i,e, senolytic therapies).
Collapse
Affiliation(s)
- Fernando A C Seara
- Departament of Physiological Sciences, Institute of Health and Biological Sciences, Federal Rural University of Rio de Janeiro, Seropédica, Brazil
- Multicenter Graduate Program of Physiological Sciences, Brazilian Society of Physiology, Rio de Janeiro, Brazil
| | - Tais H Kasai-Brunswick
- National Centre of Structural Biology and Bioimaging, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Science and Technology Institute in Regenerative Medicine, Rio de Janeiro, Brazil
| | - Jose H M Nascimento
- Laboratory of Cellular and Molecular Cardiology, Health Sciences Building, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Room G2-053, 373 Carlos Chagas Filho Avenue, Rio de Janeiro, RJ, 21941-590, Brazil
- National Science and Technology Institute in Regenerative Medicine, Rio de Janeiro, Brazil
| | - Antonio C Campos-de-Carvalho
- National Centre of Structural Biology and Bioimaging, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
- Laboratory of Cellular and Molecular Cardiology, Health Sciences Building, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Room G2-053, 373 Carlos Chagas Filho Avenue, Rio de Janeiro, RJ, 21941-590, Brazil.
- National Science and Technology Institute in Regenerative Medicine, Rio de Janeiro, Brazil.
| |
Collapse
|
14
|
AlAsmari AF, Ali N, Alharbi M, Alqahtani F, Alasmari F, Almoqbel D, AlSwayyed M, Alshammari A, Alanazi MM, Alhoshani A, Al-Harbi NO. Geraniol Ameliorates Doxorubicin-Mediated Kidney Injury through Alteration of Antioxidant Status, Inflammation, and Apoptosis: Potential Roles of NF-κB and Nrf2/Ho-1. Nutrients 2022; 14:nu14081620. [PMID: 35458182 PMCID: PMC9031157 DOI: 10.3390/nu14081620] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/05/2022] [Accepted: 04/07/2022] [Indexed: 12/10/2022] Open
Abstract
Doxorubicin-mediated kidney impairment is a serious problem in cancer treatment. Accordingly, this work investigated the ability of geraniol to modulate doxorubicin-induced kidney damage using a rat model. Rats were randomly assigned to four groups: control, doxorubicin (20 mg/kg, intraperitoneal, i.p.), doxorubicin plus 100 mg/kg of geraniol, and doxorubicin plus 200 mg/kg of geraniol. A single doxorubicin injection triggered kidney impairment, as evidenced by the altered serum creatinine, blood urea nitrogen, and albumin values; it also caused histological changes in the kidney architecture. Additionally, doxorubicin enhanced lipid peroxidation while lowering reduced glutathione, catalase activity, and the expression of glutathione peroxidase and superoxide dismutase. Interestingly, pre-treatment with geraniol rescued doxorubicin-induced alterations in kidney antioxidant parameters, enzymatic activity, and the expression of inflammatory and apoptosis-mediating gene and proteins. Moreover, prophylactic treatment with geraniol preserved most kidney histological characteristics in a dose-dependent manner. These findings support that geraniol could protect against doxorubicin-mediated kidney dysfunction. However, further research is needed to clarify the mechanisms of geraniol’s protective effects against doxorubicin-mediated kidney dysfunction.
Collapse
Affiliation(s)
- Abdullah F. AlAsmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (M.A.); (F.A.); (F.A.); (D.A.); (A.A.); (M.M.A.); (A.A.); (N.O.A.-H.)
- Correspondence: ; Tel.: +966-114677180
| | - Nemat Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (M.A.); (F.A.); (F.A.); (D.A.); (A.A.); (M.M.A.); (A.A.); (N.O.A.-H.)
| | - Metab Alharbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (M.A.); (F.A.); (F.A.); (D.A.); (A.A.); (M.M.A.); (A.A.); (N.O.A.-H.)
| | - Faleh Alqahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (M.A.); (F.A.); (F.A.); (D.A.); (A.A.); (M.M.A.); (A.A.); (N.O.A.-H.)
| | - Fawaz Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (M.A.); (F.A.); (F.A.); (D.A.); (A.A.); (M.M.A.); (A.A.); (N.O.A.-H.)
| | - Daad Almoqbel
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (M.A.); (F.A.); (F.A.); (D.A.); (A.A.); (M.M.A.); (A.A.); (N.O.A.-H.)
| | - Mohammed AlSwayyed
- Department of Pathology, College of Medicine, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Abdulrahman Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (M.A.); (F.A.); (F.A.); (D.A.); (A.A.); (M.M.A.); (A.A.); (N.O.A.-H.)
| | - Mohammed M. Alanazi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (M.A.); (F.A.); (F.A.); (D.A.); (A.A.); (M.M.A.); (A.A.); (N.O.A.-H.)
| | - Ali Alhoshani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (M.A.); (F.A.); (F.A.); (D.A.); (A.A.); (M.M.A.); (A.A.); (N.O.A.-H.)
| | - Naif O. Al-Harbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (M.A.); (F.A.); (F.A.); (D.A.); (A.A.); (M.M.A.); (A.A.); (N.O.A.-H.)
| |
Collapse
|
15
|
Xiao M, Tang Y, Wang J, Lu G, Niu J, Wang J, Li J, Liu Q, Wang Z, Huang Z, Guo Y, Gao T, Zhang X, Yue S, Gu J. A new FGF1 variant protects against adriamycin-induced cardiotoxicity via modulating p53 activity. Redox Biol 2022; 49:102219. [PMID: 34990928 PMCID: PMC8743227 DOI: 10.1016/j.redox.2021.102219] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 12/05/2021] [Accepted: 12/17/2021] [Indexed: 12/17/2022] Open
Abstract
A cumulative and progressively developing cardiomyopathy induced by adriamycin (ADR)-based chemotherapy is a major obstacle for its clinical application. However, there is a lack of safe and effective method to protect against ADR-induced cardiotoxicity. Here, we found that mRNA and protein levels of FGF1 were decreased in ADR-treated mice, primary cardiomyocytes and H9c2 cells, suggesting the potential effect of FGF1 to protect against ADR-induced cardiotoxicity. Then, we showed that treatment with a FGF1 variant (FGF1ΔHBS) with reduced proliferative potency significantly prevented ADR-induced cardiac dysfunction as well as ADR-associated cardiac inflammation, fibrosis, and hypertrophy. The mechanistic study revealed that apoptosis and oxidative stress, the two vital pathological factors in ADR-induced cardiotoxicity, were largely alleviated by FGF1ΔHBS treatment. Furthermore, the inhibitory effects of FGF1ΔHBS on ADR-induced apoptosis and oxidative stress were regulated by decreasing p53 activity through upregulation of Sirt1-mediated p53 deacetylation and enhancement of murine double minute 2 (MDM2)-mediated p53 ubiquitination. Upregulation of p53 expression or cardiac specific-Sirt1 knockout (Sirt1-CKO) almost completely abolished FGF1ΔHBS-induced protective effects in cardiomyocytes. Based on these findings, we suggest that FGF1ΔHBS may be a potential therapeutic agent against ADR-induced cardiotoxicity. Cardiac expression of FGF1 were decreased by ADR treatment. FGF1ΔHBS prevented ADR-induced cardiac structural abnormalities and dysfunction. FGF1ΔHBS inhibited ADR-induced oxidative stress and apoptosis by deacetylating p53. Deacetylated p53 induced by FGF1ΔHBS accelerated the ubiquitination of p53 by MDM2.
Collapse
Affiliation(s)
- Mengjie Xiao
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Yufeng Tang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, 250014, China
| | - Jie Wang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Guangping Lu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Jianlou Niu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Jie Wang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Jiahao Li
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Qingbo Liu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Zhaoyun Wang
- Department of Neurosurgical Intensive Care Unit & Emergency Neurosurgery, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Zhifeng Huang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Yuanfang Guo
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Ting Gao
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Xiaohui Zhang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Shouwei Yue
- Rehabilitation Center, Qilu Hospital, Cheelo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Junlian Gu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
16
|
Wu L, Sowers JR, Zhang Y, Ren J. OUP accepted manuscript. Cardiovasc Res 2022; 119:691-709. [PMID: 35576480 DOI: 10.1093/cvr/cvac080] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/21/2022] [Accepted: 04/25/2022] [Indexed: 11/13/2022] Open
Abstract
Cardiovascular diseases (CVDs) arise from a complex interplay among genomic, proteomic, and metabolomic abnormalities. Emerging evidence has recently consolidated the presence of robust DNA damage in a variety of cardiovascular disorders. DNA damage triggers a series of cellular responses termed DNA damage response (DDR) including detection of DNA lesions, cell cycle arrest, DNA repair, cellular senescence, and apoptosis, in all organ systems including hearts and vasculature. Although transient DDR in response to temporary DNA damage can be beneficial for cardiovascular function, persistent activation of DDR promotes the onset and development of CVDs. Moreover, therapeutic interventions that target DNA damage and DDR have the potential to attenuate cardiovascular dysfunction and improve disease outcome. In this review, we will discuss molecular mechanisms of DNA damage and repair in the onset and development of CVDs, and explore how DDR in specific cardiac cell types contributes to CVDs. Moreover, we will highlight the latest advances regarding the potential therapeutic strategies targeting DNA damage signalling in CVDs.
Collapse
Affiliation(s)
- Lin Wu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - James R Sowers
- Diabetes and Cardiovascular Research Center, University of Missouri Columbia, Columbia, MO 65212, USA
| | - Yingmei Zhang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Jun Ren
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai 200032, China
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
17
|
Shao D, Zhai P, Hu C, Mukai R, Sciarretta S, Del Re D, Sadoshima J. Lats2 promotes heart failure by stimulating p53-mediated apoptosis during pressure overload. Sci Rep 2021; 11:23469. [PMID: 34873220 PMCID: PMC8648781 DOI: 10.1038/s41598-021-02846-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 11/18/2021] [Indexed: 12/25/2022] Open
Abstract
The Hippo pathway plays a wide variety of roles in response to stress in the heart. Lats2, a component of the Hippo pathway, is phosphorylated by Mst1/2 and, in turn, phosphorylates YAP, causing inactivation of YAP. Lats2 stimulates apoptosis and negatively affects hypertrophy in cardiomyocytes. However, the role of Lats2 during cardiac stress is poorly understood in vivo. Lats2 is activated in the mouse heart in response to transverse aortic constriction (TAC). We used systemic Lats2 +/- mice to elucidate the role of endogenous Lats2. Cardiac hypertrophy and dysfunction induced by 4 weeks of TAC were attenuated in Lats2 +/- mice, and interstitial fibrosis and apoptosis were suppressed. Although TAC upregulated the Bcl-2 family proapoptotic (Bax and Bak) and anti-apoptotic (Bcl-2 and Bcl-xL) molecules in non-transgenic mice, TAC-induced upregulation of Bax and Bak was alleviated and that of Bcl-2 was enhanced in Lats2 +/- mice. TAC upregulated p53, but this upregulation was abolished in Lats2 +/- mice. Lats2-induced increases in apoptosis and decreases in survival in cardiomyocytes were inhibited by Pifithrin-α, a p53 inhibitor, suggesting that Lats2 stimulates apoptosis via a p53-dependent mechanism. In summary, Lats2 is activated by pressure overload, thereby promoting heart failure by stimulating p53-dependent mechanisms of cell death.
Collapse
Affiliation(s)
- Dan Shao
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, 185 S Orange Ave, MSB G609, Newark, NJ, 07103, USA
| | - Peiyong Zhai
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, 185 S Orange Ave, MSB G609, Newark, NJ, 07103, USA
| | - Chengchen Hu
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, 185 S Orange Ave, MSB G609, Newark, NJ, 07103, USA
| | - Risa Mukai
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, 185 S Orange Ave, MSB G609, Newark, NJ, 07103, USA
| | - Sebastiano Sciarretta
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy.,IRCCS Neuromed, Pozzilli, IS, Italy
| | - Dominic Del Re
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, 185 S Orange Ave, MSB G609, Newark, NJ, 07103, USA
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, 185 S Orange Ave, MSB G609, Newark, NJ, 07103, USA.
| |
Collapse
|
18
|
Nishi M, Wang PY, Hwang PM. Cardiotoxicity of Cancer Treatments: Focus on Anthracycline Cardiomyopathy. Arterioscler Thromb Vasc Biol 2021; 41:2648-2660. [PMID: 34587760 PMCID: PMC8551037 DOI: 10.1161/atvbaha.121.316697] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 09/16/2021] [Indexed: 12/19/2022]
Abstract
Significant progress has been made in developing new treatments and refining the use of preexisting ones against cancer. Their successful use and the longer survival of cancer patients have been associated with reports of new cardiotoxicities and the better characterization of the previously known cardiac complications. Immunotherapies with monoclonal antibodies against specific cancer-promoting genes, chimeric antigen receptor T cells, and immune checkpoint inhibitors have been developed to fight cancer cells, but they can also show off-target effects on the heart. Some of these cardiotoxicities are thought to be due to nonspecific immune activation and inflammatory damage. Unlike immunotherapy-associated cardiotoxicities which are relatively new entities, there is extensive literature on anthracycline-induced cardiomyopathy. Here, we provide a brief overview of the cardiotoxicities of immunotherapies for the purpose of distinguishing them from anthracycline cardiomyopathy. This is especially relevant as the expansion of oncological treatments presents greater diagnostic challenges in determining the cause of cardiac dysfunction in cancer survivors with a history of multiple cancer treatments including anthracyclines and immunotherapies administered concurrently or serially over time. We then provide a focused review of the mechanisms proposed to underlie the development of anthracycline cardiomyopathy based on experimental data mostly in mouse models. Insights into its pathogenesis may stimulate the development of new strategies to identify patients who are susceptible to anthracycline cardiomyopathy while permitting low cardiac risk patients to receive optimal treatment for their cancer.
Collapse
Affiliation(s)
- Masahiro Nishi
- Cardiovascular Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Ping-Yuan Wang
- Cardiovascular Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Paul M Hwang
- Cardiovascular Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
19
|
Acute and Delayed Doxorubicin-Induced Myocardiotoxicity Associated with Elevation of Cardiac Biomarkers, Depletion of Cellular Antioxidant Enzymes, and Several Histopathological and Ultrastructural Changes. Life (Basel) 2021; 11:life11090880. [PMID: 34575029 PMCID: PMC8467687 DOI: 10.3390/life11090880] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/17/2021] [Accepted: 08/24/2021] [Indexed: 12/13/2022] Open
Abstract
Doxorubicin (DOX; Adricin) is an anthracycline antibiotic, which is an efficient anticancer chemotherapeutic agent that targets many types of adult and pediatric tumors, such as breast cancer, leukemia, and lymphomas. However, use of DOX is limited due to its cardiotoxic effects. This study sequentially investigated the mechanistic pathways of the cardiotoxic process of DOX in rats at different post-treatment periods using cumulative dose, which is used in therapeutic regimes. In this regard, 56 male albino rats were used for the experiment. The experimental animals were divided into seven groups (n = 8/group) based on dose and sacrifice schedule as follows: G1 (2 mg/kg body weight [BW] and sacrificed at day 4), G2 (4 mg/kg BW and sacrificed at day 8), G3 (6 mg/kg BW and sacrificed at day 15), G4 (8 mg/kg BW and sacrificed at day 30), G5 (10 mg/kg BW and sacrificed at day 60), G6 (10 mg/kg BW and sacrificed at day 90), and G7 (10 mg/kg BW and sacrificed at day 120). As expected, G1, G2, and G3-treated groups revealed features of acute toxic myocarditis associated with degenerative and necrotic changes in myocytes, mitochondrial damage, elevation of cardiac biomarkers, and depletion of cellular antioxidant enzymes. However, these changes increased in severity with subsequent treatment with the same dose until reaching a cumulative dose of 10 mg/kg BW for 30 d. Furthermore, after a cumulative dose of 10 mg/kg BW with a withdrawal period of 2–3 months, various predominant changes in chronicity were reported, such as disorganization and atrophy of myocytes, condensation and atrophy of mitochondria, degranulation of mast cells, and fibrosis with occasional focal necrosis, indicating incomplete elimination of DOX and/or its metabolites. Altogether, these data provide interesting observations associated with the cardiotoxic process of DOX in rats that would help understand the accompanying changes underlying the major toxic effects of the drug. Future research is suggested to explore more about the dose-dependent mechanisms of such induced toxicity of DOX that would help determine the proper doses and understand the resulting cardiomyopathy.
Collapse
|
20
|
Ali N, AlAsmari AF, Imam F, Ahmed MZ, Alqahtani F, Alharbi M, AlSwayyed M, AlAsmari F, Alasmari M, Alshammari A, Fantoukh OI, Alanazi MM. Protective effect of diosmin against doxorubicin-induced nephrotoxicity. Saudi J Biol Sci 2021; 28:4375-4383. [PMID: 34354422 PMCID: PMC8324953 DOI: 10.1016/j.sjbs.2021.04.030] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/27/2021] [Accepted: 04/08/2021] [Indexed: 12/12/2022] Open
Abstract
Dox induces kidney damage. Dox leads to a decrease in antioxidant defense mechanism. Diosmin administration restores antioxidant properties.
Doxorubicin (Dox) is an anthracycline antibiotic that is primarily used for treating various solid tumors including that of pulmonary, ovary, breast, uterine, cervix, and several blood cancers. However, nephrotoxicity associated with Dox treatment limits its clinical use. Administration of Dox in combination with compounds exhibiting antioxidant properties are being used to minimize the side effects of Dox. Diosmin is a flavonoid glycoside with numerous beneficial properties that is found in the pericarp of many citrus fruits. Diosmin has demonstrated antioxidant, anti-inflammatory, and anti-apoptotic effects in response to various insults, although the exact mechanism remains unknown. Therefore, this study was designed to evaluate the effect of diosmin in preventing kidney damage in response to Dox treatment. Male Wistar rats were randomly divided into four groups: control group, Dox group (20 mg/kg, i.p.), Dox plus low-dose diosmin group (100 mg/kg orally), and Dox plus high-dose diosmin group (200 mg/kg orally). A single intraperitoneal injection of Dox resulted in kidney damage as evidenced by significant alterations in kidney markers, histological abnormalities, and the attenuation of antioxidant defense mechanisms (GSH, SOD, and CAT). Moreover, Dox treatment significantly altered the expression of oxidative stress, inflammatory, and anti-apoptotic protein markers. Diosmin pretreatment alleviated Dox-induced nephrotoxicity by ameliorating the antioxidant mechanism, decreasing inflammation and apoptosis, and restoring kidney architecture. In conclusion, our results indicate that diosmin is a promising therapeutic agent for the prevention of nephrotoxicity associated with DOX.
Collapse
Affiliation(s)
- Nemat Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
- Corresponding author at: Dept. of Pharmacology & Toxicology, College of Pharmacy, Building 23, second floor, P.O. Box 2457, Riyadh 11451, Saudi Arabia.
| | - Abdullah F. AlAsmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Faisal Imam
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammad Z. Ahmed
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Faleh Alqahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Metab Alharbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed AlSwayyed
- Department of Pathology, College of Medicine, King Saud University, Riyadh 11451, Saudi Arabia
| | - Fawaz AlAsmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdulrahman Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Omer I. Fantoukh
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed M. Alanazi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
21
|
Ramani S, Park S. HSP27 role in cardioprotection by modulating chemotherapeutic doxorubicin-induced cell death. J Mol Med (Berl) 2021; 99:771-784. [PMID: 33728476 DOI: 10.1007/s00109-021-02048-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 02/05/2021] [Accepted: 02/09/2021] [Indexed: 01/19/2023]
Abstract
The common phenomenon expected from any anti-cancer drug in use is to kill the cancer cells without any side effects to non-malignant cells. Doxorubicin is an anthracycline derivative anti-cancer drug active over different types of cancers with anti-cancer activity but attributed to unintended cytotoxicity and genotoxicity triggering mitogenic signals inducing apoptosis. Administration of doxorubicin tends to both acute and chronic toxicity resulting in cardiomyopathy (left ventricular dysfunction) and congestive heart failure (CHF). Cardiotoxicity is prevented through administration of different cardioprotectants along with the drug. This review elaborates on mechanism of drug-mediated cardiotoxicity and attenuation principle by different cardioprotectants, with a focus on Hsp27 as cardioprotectant by prevention of drug-induced oxidative stress, cell survival pathways with suppression of intrinsic cell death. In conclusion, Hsp27 may offer an exciting/alternating cardioprotectant, with a wider study being need of the hour, specifically on primary cell line and animal models in conforming its cardioprotectant behaviour.
Collapse
Affiliation(s)
- Sivasubramanian Ramani
- Department of Food Science and Biotechnology, Sejong University, 209 Neungdong-ro, Seoul, 05006, South Korea
| | - Sungkwon Park
- Department of Food Science and Biotechnology, Sejong University, 209 Neungdong-ro, Seoul, 05006, South Korea.
| |
Collapse
|
22
|
Men H, Cai H, Cheng Q, Zhou W, Wang X, Huang S, Zheng Y, Cai L. The regulatory roles of p53 in cardiovascular health and disease. Cell Mol Life Sci 2021; 78:2001-2018. [PMID: 33179140 PMCID: PMC11073000 DOI: 10.1007/s00018-020-03694-6] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/22/2020] [Accepted: 10/29/2020] [Indexed: 02/06/2023]
Abstract
Cardiovascular disease (CVD) remains the leading cause of mortality globally, so further investigation is required to identify its underlying mechanisms and potential targets for its prevention. The transcription factor p53 functions as a gatekeeper, regulating a myriad of genes to maintain normal cell functions. It has received a great deal of research attention as a tumor suppressor. In the past three decades, evidence has also shown a regulatory role for p53 in the heart. Basal p53 is essential for embryonic cardiac development; it is also necessary to maintain normal heart architecture and physiological function. In pathological cardiovascular circumstances, p53 expression is elevated in both patient samples and animal models. Elevated p53 plays a regulatory role via anti-angiogenesis, pro-programmed cell death, metabolism regulation, and cell cycle arrest regulation. This largely promotes the development of CVDs, particularly cardiac remodeling in the infarcted heart, hypertrophic cardiomyopathy, dilated cardiomyopathy, and diabetic cardiomyopathy. Roles for p53 have also been found in atherosclerosis and chemotherapy-induced cardiotoxicity. However, it has different roles in cardiomyocytes and non-myocytes, even in the same model. In this review, we describe the different effects of p53 in cardiovascular physiological and pathological conditions, in addition to potential CVD therapies targeting p53.
Collapse
Affiliation(s)
- Hongbo Men
- Department of Cardiovascular Disease, First Hospital of Jilin University, Jilin University, Changchun, 130021, China
- Department of Pediatrics, Pediatric Research Institute, University of Louisville, Louisville, KY, 40202, USA
| | - He Cai
- Department of Cardiovascular Disease, First Hospital of Jilin University, Jilin University, Changchun, 130021, China
| | - Quanli Cheng
- Department of Cardiovascular Disease, First Hospital of Jilin University, Jilin University, Changchun, 130021, China
| | - Wenqian Zhou
- Department of Cardiovascular Disease, First Hospital of Jilin University, Jilin University, Changchun, 130021, China
- Department of Pediatrics, Pediatric Research Institute, University of Louisville, Louisville, KY, 40202, USA
| | - Xiang Wang
- Department of Cardiovascular Disease, First Hospital of Jilin University, Jilin University, Changchun, 130021, China
- Department of Pediatrics, Pediatric Research Institute, University of Louisville, Louisville, KY, 40202, USA
| | - Shan Huang
- Department of Cardiovascular Disease, First Hospital of Jilin University, Jilin University, Changchun, 130021, China
- Department of Pediatrics, Pediatric Research Institute, University of Louisville, Louisville, KY, 40202, USA
| | - Yang Zheng
- Department of Cardiovascular Disease, First Hospital of Jilin University, Jilin University, Changchun, 130021, China.
| | - Lu Cai
- Department of Pediatrics, Pediatric Research Institute, University of Louisville, Louisville, KY, 40202, USA.
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, 40202, USA.
| |
Collapse
|
23
|
Ovics P, Regev D, Baskin P, Davidor M, Shemer Y, Neeman S, Ben-Haim Y, Binah O. Drug Development and the Use of Induced Pluripotent Stem Cell-Derived Cardiomyocytes for Disease Modeling and Drug Toxicity Screening. Int J Mol Sci 2020; 21:E7320. [PMID: 33023024 PMCID: PMC7582587 DOI: 10.3390/ijms21197320] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/23/2020] [Accepted: 09/27/2020] [Indexed: 12/19/2022] Open
Abstract
: Over the years, numerous groups have employed human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) as a superb human-compatible model for investigating the function and dysfunction of cardiomyocytes, drug screening and toxicity, disease modeling and for the development of novel drugs for heart diseases. In this review, we discuss the broad use of iPSC-CMs for drug development and disease modeling, in two related themes. In the first theme-drug development, adverse drug reactions, mechanisms of cardiotoxicity and the need for efficient drug screening protocols-we discuss the critical need to screen old and new drugs, the process of drug development, marketing and Adverse Drug reactions (ADRs), drug-induced cardiotoxicity, safety screening during drug development, drug development and patient-specific effect and different mechanisms of ADRs. In the second theme-using iPSC-CMs for disease modeling and developing novel drugs for heart diseases-we discuss the rationale for using iPSC-CMs and modeling acquired and inherited heart diseases with iPSC-CMs.
Collapse
Affiliation(s)
- Paz Ovics
- Department of Physiology, Biophysics and Systems Biology, The Rappaport Institute, Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel; (P.O.); (D.R.); (P.B.); (M.D.); (Y.S.); (S.N.)
| | - Danielle Regev
- Department of Physiology, Biophysics and Systems Biology, The Rappaport Institute, Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel; (P.O.); (D.R.); (P.B.); (M.D.); (Y.S.); (S.N.)
| | - Polina Baskin
- Department of Physiology, Biophysics and Systems Biology, The Rappaport Institute, Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel; (P.O.); (D.R.); (P.B.); (M.D.); (Y.S.); (S.N.)
| | - Mor Davidor
- Department of Physiology, Biophysics and Systems Biology, The Rappaport Institute, Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel; (P.O.); (D.R.); (P.B.); (M.D.); (Y.S.); (S.N.)
| | - Yuval Shemer
- Department of Physiology, Biophysics and Systems Biology, The Rappaport Institute, Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel; (P.O.); (D.R.); (P.B.); (M.D.); (Y.S.); (S.N.)
| | - Shunit Neeman
- Department of Physiology, Biophysics and Systems Biology, The Rappaport Institute, Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel; (P.O.); (D.R.); (P.B.); (M.D.); (Y.S.); (S.N.)
| | - Yael Ben-Haim
- Institute of Molecular and Clinical Sciences, St. George’s University of London, London SW17 0RE, UK;
- Cardiology Clinical Academic Group, St. George’s University Hospitals NHS Foundation Trust, London SW17 0QT, UK
| | - Ofer Binah
- Department of Physiology, Biophysics and Systems Biology, The Rappaport Institute, Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel; (P.O.); (D.R.); (P.B.); (M.D.); (Y.S.); (S.N.)
| |
Collapse
|
24
|
Sachinidis A. Cardiotoxicity and Heart Failure: Lessons from Human-Induced Pluripotent Stem Cell-Derived Cardiomyocytes and Anticancer Drugs. Cells 2020; 9:cells9041001. [PMID: 32316481 PMCID: PMC7226145 DOI: 10.3390/cells9041001] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/14/2020] [Accepted: 04/15/2020] [Indexed: 12/17/2022] Open
Abstract
Human-induced pluripotent stem cells (hiPSCs) are discussed as disease modeling for optimization and adaptation of therapy to each individual. However, the fundamental question is still under debate whether stem-cell-based disease modeling and drug discovery are applicable for recapitulating pathological processes under in vivo conditions. Drug treatment and exposure to different chemicals and environmental factors can initiate diseases due to toxicity effects in humans. It is well documented that drug-induced cardiotoxicity accelerates the development of heart failure (HF). Until now, investigations on the understanding of mechanisms involved in HF by anticancer drugs are hindered by limitations of the available cellular models which are relevant for human physiology and by the fact that the clinical manifestation of HF often occurs several years after its initiation. Recently, we identified similar genomic biomarkers as observed by HF after short treatment of hiPSCs-derived cardiomyocytes (hiPSC-CMs) with different antitumor drugs such as anthracyclines and etoposide (ETP). Moreover, we identified common cardiotoxic biological processes and signal transduction pathways which are discussed as being crucial for the survival and function of cardiomyocytes and, therefore, for the development of HF. In the present review, I discuss the applicability of the in vitro cardiotoxicity test systems as modeling for discovering preventive mechanisms/targets against cardiotoxicity and, therefore, for novel HF therapeutic concepts.
Collapse
Affiliation(s)
- Agapios Sachinidis
- Faculty of Medicine, Institute of Neurophysiology, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany;
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Str. 21, 50931 Cologne, Germany
| |
Collapse
|
25
|
Ibrahim KM, Mantawy EM, Elanany MM, Abdelgawad HS, Khalifa NM, Hussien RH, El-Agroudy NN, El-Demerdash E. Protection from doxorubicin-induced nephrotoxicity by clindamycin: novel antioxidant, anti-inflammatory and anti-apoptotic roles. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2020; 393:739-748. [PMID: 31853613 DOI: 10.1007/s00210-019-01782-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 11/28/2019] [Indexed: 12/12/2022]
Abstract
This study was performed to examine whether clindamycin could protect against doxorubicin (DOX)-induced acute nephrotoxicity, and if so, what molecular mechanisms responsible for this protective effect. Male albino rats were pretreated with clindamycin once per day for 5 consecutive days at a dose of 300 mg/kg, i.p, then received a single dose of DOX (15 mg/kg; i.p) on the 5th day. DOX-induced marked renal injury as indicated by the presence of inflammatory cell infiltration, congestion, and edema accompanied by elevation in serum levels of creatinine and urea. These effects were alleviated by clindamycin pretreatment. DOX caused glutathione depletion and reduction in level of the antioxidant enzyme, catalase. Pretreatment with clindamycin markedly prohibited DOX-induced oxidative damage in renal tissue. Moreover, DOX provoked inflammatory responses in renal tissues as confirmed by increased expressions of NF-κB and COX-2 which were significantly reduced by clindamycin pretreatment. Besides, DOX-triggered apoptotic cascades in renal tissues as evidenced by elevated expression of pro-apoptotic proteins; Bax and cytochrome c, enhancing activity of caspase-3 enzyme whereas reducing the expression of anti-apoptotic Bcl-2 protein. Clindamycin pretreatment counteracts these apoptotic effects of DOX. Summarily, our results provide an evidence for the first time that clindamycin has a potential protective action against DOX-induced acute nephrotoxicity through inhibiting oxidative stress, inflammatory cascades, and apoptotic tissue injury.
Collapse
Affiliation(s)
- Kamilia M Ibrahim
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Badr University, Cairo, Egypt
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Eman M Mantawy
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mona M Elanany
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Hend S Abdelgawad
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Nuha M Khalifa
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Rada H Hussien
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Nermeen N El-Agroudy
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
- Medizinische klinik III, Universitätsklinikum Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Ebtehal El-Demerdash
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
26
|
Sun F, Wang J, Wu X, Yang CS, Zhang J. Selenium nanoparticles act as an intestinal p53 inhibitor mitigating chemotherapy-induced diarrhea in mice. Pharmacol Res 2019; 149:104475. [PMID: 31593755 DOI: 10.1016/j.phrs.2019.104475] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 09/21/2019] [Accepted: 10/01/2019] [Indexed: 12/27/2022]
Abstract
Selenium, at high-dose levels approaching its toxicity, protects tissues from dose-limiting toxicities of many cancer chemotherapeutics without compromising their therapeutic effects on tumors, there by allowing the delivery of higher chemotherapeutic doses to achieve increased cure rate. In this regard, selenium nanoparticles (SeNPs), which show the lowest toxicity among extensively investigated selenium compounds including methylselenocysteine and selenomethionine, are more promising for application. The key issue remains to be resolved is whether low-toxicity SeNPs possess a selective protective mechanism. p53 or p53-regulated thrombospondin-1 has each been confirmed to be an appropriate target for therapeutic suppression to reduce side effects of anticancer therapy. The present study demonstrated that SeNPs transiently suppressed the expression of many intestinal p53-associated genes in healthy mice. SeNPs did not interfere with tumor-suppressive effect of nedaplatin, a cisplatin analogue; however, effectively reduced nedaplatin-evoked diarrhea. Nedaplatin-induced diarrhea was associated with activation of intestinal p53 and high expression of intestinal thrombospondin-1. The preventive effect of SeNPs on nedaplatin-induced diarrhea was correlated with a powerful concomitant suppression of p53 and thrombospondin-1. Moreover, the high-dose SeNPs used in the present study did not suppress growth nor caused liver and kidney injuries as well as alterations of hematological parameters in healthy mice. Overall, the present study reveals that chemotherapeutic selectivity conferred by SeNPs involves a dual suppression of two well-documented targets, the p53 and thrombospondin-1, providing mechanistic and pharmacologic insights on low-toxicity SeNPs as a potential chemoprotectant for mitigating chemotherapy-induced diarrhea.
Collapse
Affiliation(s)
- Feng Sun
- Laboratory of Redox Biology, School of Tea & Food Science, State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei, Anhui, China
| | - Jiajia Wang
- Laboratory of Redox Biology, School of Tea & Food Science, State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei, Anhui, China
| | - Ximing Wu
- Laboratory of Redox Biology, School of Tea & Food Science, State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei, Anhui, China
| | - Chung S Yang
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Jinsong Zhang
- Laboratory of Redox Biology, School of Tea & Food Science, State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei, Anhui, China.
| |
Collapse
|
27
|
Zhang YJ, Huang H, Liu Y, Kong B, Wang G. MD-1 Deficiency Accelerates Myocardial Inflammation and Apoptosis in Doxorubicin-Induced Cardiotoxicity by Activating the TLR4/MAPKs/Nuclear Factor kappa B (NF-κB) Signaling Pathway. Med Sci Monit 2019; 25:7898-7907. [PMID: 31636246 PMCID: PMC6820359 DOI: 10.12659/msm.919861] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 10/02/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Myocardial apoptosis and inflammation play important roles in doxorubicin (DOX)-caused cardiotoxicity. Our prior studies have characterized the effects of myeloid differentiation protein 1(MD-1) in pathological cardiac remodeling and myocardial ischemia/reperfusion (I/R) injury, but its participations and potential molecular mechanisms in DOX-caused cardiotoxicity remain unknown. MATERIAL AND METHODS In the present study, MD-1 knockout mice were generated, and a single intraperitoneal injection of DOX (15 mg/kg) was performed to elicit DOX-induced cardiotoxicity. Cardiac function, histological change, mitochondrial structure, myocardial death, apoptosis, inflammation, and molecular alterations were measured systemically. RESULTS The results showed that the protein and mRNA levels of MD-1 were dramatically downregulated in DOX-treated cardiomyocytes. DOX insult markedly accelerated cardiac dysfunction and injury, followed by enhancements of apoptosis and inflammation, all of which were further aggravated in MD-1 knockout mice. Mechanistically, the TLR4/MAPKs/NF-kappaB pathways, which were over-activated in MD-1-deficient mice, were significantly increased in DOX-damaged cardiomyocytes. Moreover, the abolishment of TLR4 or NF-kappaB via a specific inhibitor exerted protective effects against the adverse effects of MD-1 loss on DOX-caused cardiotoxicity. CONCLUSIONS Collectively, these findings suggest that MD-1 is a novel target for the treatment of DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Ying-Jun Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China
- Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei, P.R. China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, P.R. China
| | - He Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China
- Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei, P.R. China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, P.R. China
| | - Yu Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China
- Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei, P.R. China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, P.R. China
| | - Bin Kong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China
- Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei, P.R. China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, P.R. China
| | - Guangji Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China
- Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei, P.R. China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, P.R. China
| |
Collapse
|
28
|
Al-Taee H, Azimullah S, Meeran MN, Alaraj Almheiri MK, Al Jasmi RA, Tariq S, AB Khan M, Adeghate E, Ojha S. β-caryophyllene, a dietary phytocannabinoid attenuates oxidative stress, inflammation, apoptosis and prevents structural alterations of the myocardium against doxorubicin-induced acute cardiotoxicity in rats: An in vitro and in vivo study. Eur J Pharmacol 2019; 858:172467. [DOI: 10.1016/j.ejphar.2019.172467] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 06/12/2019] [Accepted: 06/14/2019] [Indexed: 12/12/2022]
|
29
|
Das B, Pal B, Bhuyan R, Li H, Sarma A, Gayan S, Talukdar J, Sandhya S, Bhuyan S, Gogoi G, Gouw AM, Baishya D, Gotlib JR, Kataki AC, Felsher DW. MYC Regulates the HIF2α Stemness Pathway via Nanog and Sox2 to Maintain Self-Renewal in Cancer Stem Cells versus Non-Stem Cancer Cells. Cancer Res 2019; 79:4015-4025. [PMID: 31266772 DOI: 10.1158/0008-5472.can-18-2847] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 04/08/2019] [Accepted: 06/17/2019] [Indexed: 12/19/2022]
Abstract
Cancer stem cells (CSC) maintain both undifferentiated self-renewing CSCs and differentiated, non-self-renewing non-CSCs through cellular division. However, molecular mechanisms that maintain self-renewal in CSCs versus non-CSCs are not yet clear. Here, we report that in a transgenic mouse model of MYC-induced T-cell leukemia, MYC, maintains self-renewal in Sca1+ CSCs versus Sca-1- non-CSCs. MYC preferentially bound to the promoter and activated hypoxia-inducible factor-2α (HIF2α) in Sca-1+ cells only. Furthermore, the reprogramming factors, Nanog and Sox2, facilitated MYC regulation of HIF2α in Sca-1+ versus Sca-1- cells. Reduced expression of HIF2α inhibited the self-renewal of Sca-1+ cells; this effect was blocked through suppression of ROS by N-acetyl cysteine or the knockdown of p53, Nanog, or Sox2. Similar results were seen in ABCG2+ CSCs versus ABCG2- non-CSCs from primary human T-cell lymphoma. Thus, MYC maintains self-renewal exclusively in CSCs by selectively binding to the promoter and activating the HIF2α stemness pathway. Identification of this stemness pathway as a unique CSC determinant may have significant therapeutic implications. SIGNIFICANCE: These findings show that the HIF2α stemness pathway maintains leukemic stem cells downstream of MYC in human and mouse T-cell leukemias. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/79/16/4015/F1.large.jpg.
Collapse
Affiliation(s)
- Bikul Das
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, California. .,Department of Cancer and Stem Cell Biology, KaviKrishna Laboratory, Guwahati Biotech Park, Indian Institute of Technology, Guwahati, Assam, India.,Department of Experimental Therapeutics, Thoreau Laboratory for Global Health, M2D2, University of Massachusetts, Lowell, Massachusetts.,Department of Immunology and Infectious Diseases, Forsyth Institute, Cambridge, Massachusetts
| | - Bidisha Pal
- Department of Cancer and Stem Cell Biology, KaviKrishna Laboratory, Guwahati Biotech Park, Indian Institute of Technology, Guwahati, Assam, India.,Department of Experimental Therapeutics, Thoreau Laboratory for Global Health, M2D2, University of Massachusetts, Lowell, Massachusetts.,Department of Immunology and Infectious Diseases, Forsyth Institute, Cambridge, Massachusetts
| | - Rashmi Bhuyan
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, California.,Department of Cancer and Stem Cell Biology, KaviKrishna Laboratory, Guwahati Biotech Park, Indian Institute of Technology, Guwahati, Assam, India.,Department of Experimental Therapeutics, Thoreau Laboratory for Global Health, M2D2, University of Massachusetts, Lowell, Massachusetts.,Department of Immunology and Infectious Diseases, Forsyth Institute, Cambridge, Massachusetts
| | - Hong Li
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, California.,Department of Experimental Therapeutics, Thoreau Laboratory for Global Health, M2D2, University of Massachusetts, Lowell, Massachusetts.,Department of Immunology and Infectious Diseases, Forsyth Institute, Cambridge, Massachusetts
| | - Anupam Sarma
- Department of Cancer and Stem Cell Biology, KaviKrishna Laboratory, Guwahati Biotech Park, Indian Institute of Technology, Guwahati, Assam, India.,Dr. B. Borooah Cancer Institute, Guwahati, Assam, India
| | - Sukanya Gayan
- Department of Cancer and Stem Cell Biology, KaviKrishna Laboratory, Guwahati Biotech Park, Indian Institute of Technology, Guwahati, Assam, India.,Department of Experimental Therapeutics, Thoreau Laboratory for Global Health, M2D2, University of Massachusetts, Lowell, Massachusetts.,Department of Immunology and Infectious Diseases, Forsyth Institute, Cambridge, Massachusetts
| | - Joyeeta Talukdar
- Department of Cancer and Stem Cell Biology, KaviKrishna Laboratory, Guwahati Biotech Park, Indian Institute of Technology, Guwahati, Assam, India
| | - Sorra Sandhya
- Department of Cancer and Stem Cell Biology, KaviKrishna Laboratory, Guwahati Biotech Park, Indian Institute of Technology, Guwahati, Assam, India
| | - Seema Bhuyan
- Department of Cancer and Stem Cell Biology, KaviKrishna Laboratory, Guwahati Biotech Park, Indian Institute of Technology, Guwahati, Assam, India
| | - Gayatri Gogoi
- Department of Cancer and Stem Cell Biology, KaviKrishna Laboratory, Guwahati Biotech Park, Indian Institute of Technology, Guwahati, Assam, India.,Department of Immunology and Infectious Diseases, Forsyth Institute, Cambridge, Massachusetts.,Department of Pathology, Assam Medical College, Dibrugarh, Assam, India
| | - Arvin M Gouw
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, California
| | - Debabrat Baishya
- Department of Cancer and Stem Cell Biology, KaviKrishna Laboratory, Guwahati Biotech Park, Indian Institute of Technology, Guwahati, Assam, India.,Department of Bioengineering and Technology, Gauhati University, Guwahati, Assam, India
| | - Jason R Gotlib
- Division of Hematology, Stanford Cancer Institute, Stanford, California
| | - Amal C Kataki
- Dr. B. Borooah Cancer Institute, Guwahati, Assam, India
| | - Dean W Felsher
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, California.
| |
Collapse
|
30
|
Zheng WF, Zhang SY, Ma HF, Chang XW, Wang H. C1qTNF-related protein-6 protects against doxorubicin-induced cardiac injury. J Cell Biochem 2019; 120:10748-10755. [PMID: 30719766 DOI: 10.1002/jcb.28366] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 11/29/2018] [Indexed: 12/11/2022]
Abstract
The clinical use of doxorubicin (DOX) is limited by its toxic effect. However, there is no specific drug that can prevent DOX-related cardiac injury. C1qTNF-related protein-6 (CTRP6) is a newly identified adiponectin paralog with many protective functions on metabolism and cardiovascular diseases. However, little is known about the effect of CTRP6 on DOX-induced cardiac injury. The present study aimed to investigate whether CTRP6 could protect against DOX-related cardiotoxicity. To induce acute cardiotoxicity, the mice were intraperitoneally injected with a single dose of DOX (15 mg/kg). Cardiomyocyte-specific CTRP6 overexpression was achieved using an adenoassociated virus system at 4 weeks before DOX injection. The data in our study demonstrated that CTRP6 messenger RNA and protein expression were decreased in DOX-treated hearts. CTRP6 attenuated cardiac atrophy induced by DOX injection and inhibited cardiac apoptosis and improved cardiac function in vivo. CTRP6 also promoted the activation of protein kinase B (AKT/PKB) signaling pathway in DOX-treated mice. CTRP6 prevented cardiomyocytes from DOX-induced apoptosis and activated the AKT pathway in vitro. CTRP6 lost its protection against DOX-induced cardiac injury in mice with AKT inhibition. In conclusion, CTRP6 protected the heart from DOX-cardiotoxicity and improves cardiac function via activation of the AKT signaling pathway.
Collapse
Affiliation(s)
- Wei-Feng Zheng
- Department of Cardiology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan, China
| | - Shou-Yan Zhang
- Department of Cardiology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan, China
| | - Hui-Fang Ma
- Department of Cardiology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan, China
| | - Xue-Wei Chang
- Department of Cardiology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan, China
| | - Hao Wang
- Department of Cardiology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan, China
| |
Collapse
|
31
|
Gogiraju R, Bochenek ML, Schäfer K. Angiogenic Endothelial Cell Signaling in Cardiac Hypertrophy and Heart Failure. Front Cardiovasc Med 2019; 6:20. [PMID: 30895179 PMCID: PMC6415587 DOI: 10.3389/fcvm.2019.00020] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 02/14/2019] [Indexed: 12/30/2022] Open
Abstract
Endothelial cells are, by number, one of the most abundant cell types in the heart and active players in cardiac physiology and pathology. Coronary angiogenesis plays a vital role in maintaining cardiac vascularization and perfusion during physiological and pathological hypertrophy. On the other hand, a reduction in cardiac capillary density with subsequent tissue hypoxia, cell death and interstitial fibrosis contributes to the development of contractile dysfunction and heart failure, as suggested by clinical as well as experimental evidence. Although the molecular causes underlying the inadequate (with respect to the increased oxygen and energy demands of the hypertrophied cardiomyocyte) cardiac vascularization developing during pathological hypertrophy are incompletely understood. Research efforts over the past years have discovered interesting mediators and potential candidates involved in this process. In this review article, we will focus on the vascular changes occurring during cardiac hypertrophy and the transition toward heart failure both in human disease and preclinical models. We will summarize recent findings in transgenic mice and experimental models of cardiac hypertrophy on factors expressed and released from cardiomyocytes, pericytes and inflammatory cells involved in the paracrine (dys)regulation of cardiac angiogenesis. Moreover, we will discuss major signaling events of critical angiogenic ligands in endothelial cells and their possible disturbance by hypoxia or oxidative stress. In this regard, we will particularly highlight findings on negative regulators of angiogenesis, including protein tyrosine phosphatase-1B and tumor suppressor p53, and how they link signaling involved in cell growth and metabolic control to cardiac angiogenesis. Besides endothelial cell death, phenotypic conversion and acquisition of myofibroblast-like characteristics may also contribute to the development of cardiac fibrosis, the structural correlate of cardiac dysfunction. Factors secreted by (dysfunctional) endothelial cells and their effects on cardiomyocytes including hypertrophy, contractility and fibrosis, close the vicious circle of reciprocal cell-cell interactions within the heart during pathological hypertrophy remodeling.
Collapse
Affiliation(s)
- Rajinikanth Gogiraju
- Center for Cardiology, Cardiology I, Translational Vascular Biology, University Medical Center Mainz, Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany.,Center for Translational Vascular Biology, University Medical Center Mainz, Mainz, Germany.,Deutsches Zentrum für Herz-Kreislauf-Forschung e.V., Partner Site RheinMain (Mainz), Mainz, Germany
| | - Magdalena L Bochenek
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany.,Center for Translational Vascular Biology, University Medical Center Mainz, Mainz, Germany.,Deutsches Zentrum für Herz-Kreislauf-Forschung e.V., Partner Site RheinMain (Mainz), Mainz, Germany
| | - Katrin Schäfer
- Center for Cardiology, Cardiology I, Translational Vascular Biology, University Medical Center Mainz, Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany.,Center for Translational Vascular Biology, University Medical Center Mainz, Mainz, Germany.,Deutsches Zentrum für Herz-Kreislauf-Forschung e.V., Partner Site RheinMain (Mainz), Mainz, Germany
| |
Collapse
|
32
|
β-Caryophyllene, a natural bicyclic sesquiterpene attenuates doxorubicin-induced chronic cardiotoxicity via activation of myocardial cannabinoid type-2 (CB 2) receptors in rats. Chem Biol Interact 2019; 304:158-167. [PMID: 30836069 DOI: 10.1016/j.cbi.2019.02.028] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 02/19/2019] [Accepted: 02/26/2019] [Indexed: 11/20/2022]
Abstract
The cannabinoid type 2 receptor (CB2) has recently emerged as an important therapeutic target for cancer as well as cardiovascular diseases. The CB2 receptor downregulation has been reported in solid tumors and cardiovascular diseases, therefore the CB2 receptor activation has been considered as a viable strategy for chemotherapy as well as cardioprotection. Doxorubicin (DOX) is an important drug that continues to be the mainstay of chemotherapy in solid tumors, leukemia, and lymphoma. However, the use of DOX is often limited due to its lethal cardiotoxicity. Considering the role of CB2 receptors in cardiovascular diseases and cancer, the activation of CB2 receptors may protect against DOX-induced chronic cardiotoxicity in rats. In the present study, we investigated the cardioprotective effect of a selective CB2 receptor agonist; β-Caryophyllene (BCP), a natural bicyclic sesquiterpene, against DOX-induced chronic cardiotoxicity in rats. AM630, a CB2 receptor antagonist was administered as a pharmacological challenge prior to BCP treatment to demonstrate CB2 receptor mediated cardioprotective mechanism of BCP. DOX (2.5 mg/kg) was injected intraperitoneally once a week for five weeks to induce chronic cardiotoxicity in rats. BCP was also injected into rats six days a week for a total duration of five weeks. DOX induced a significant decline in cardiac function and oxidative stress evidenced by the depletion of antioxidant enzymes, glutathione, and increased lipid peroxidation. DOX also triggered activation of nuclear factor kappa B (NF-κB) and increased the levels of pro-inflammatory cytokines (TNF-α, IL-6, and IL-1β) and expression of the inflammatory mediators (iNOS and COX-2) in the heart. Furthermore, DOX also upregulated the expression of pro-apoptotic markers such as Bax, p53, cleaved PARP, active caspase-3 and downregulated anti-apoptotic marker Bcl-2 in the myocardium. BCP treatment exerted significant cardioprotective effect by salvaging the heart tissues, improving cardiac function, mitigating oxidative stress, inflammation, and apoptosis. The histological and ultrastructural studies also appear in line with our findings of biochemical and molecular parameters. The CB2 receptor-mediated cardioprotective mechanism was further confirmed by the abrogation of the beneficial effects of BCP with prior administration of the CB2 receptor antagonist; AM630. Our study revealed the novel mechanism of BCP in cardioprotection against DOX-induced chronic cardiotoxicity by the activation of CB2 receptors.
Collapse
|
33
|
Park AM, Tsunoda I, Yoshie O. Heat shock protein 27 promotes cell cycle progression by down-regulating E2F transcription factor 4 and retinoblastoma family protein p130. J Biol Chem 2018; 293:15815-15826. [PMID: 30166342 PMCID: PMC6187643 DOI: 10.1074/jbc.ra118.003310] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 08/23/2018] [Indexed: 02/05/2023] Open
Abstract
Heat shock protein 27 (HSP27) protects cells under stress. Here, we demonstrate that HSP27 also promotes cell cycle progression of MRC-5 human lung fibroblast cells. Serum starvation for 24 h induced G1 arrest in these cells, and upon serum refeeding, the cells initiated cell cycle progression accompanied by an increase in HSP27 protein levels. HSP27 levels peaked at 12 h, and transcriptional up-regulation of six G2/M-related genes (CCNA2, CCNB1, CCNB2, CDC25C, CDCA3, and CDK1) peaked at 24-48 h. siRNA-mediated HSP27 silencing in proliferating MRC-5 cells induced G2 arrest coinciding with down-regulation of these six genes. Of note, the promoters of all of these genes have the cell cycle-dependent element and/or the cell cycle gene-homology region. These promoter regions are known to be bound by the E2F family proteins (E2F-1 to E2F-8) and retinoblastoma (RB) family proteins (RB1, p107, and p130), among which E2F-4 and p130 were strongly up-regulated in HSP27-knockdown cells. E2F-4 or p130 knockdown concomitant with the HSP27 knockdown rescued MRC-5 cells from G2 arrest and up-regulated the six cell cycle genes. Moreover, we observed cellular senescence in MRC-5 cells on day 3 after the HSP27 knockdown, as evidenced by increased senescence-associated β-gal activity and up-regulated inflammatory cytokines. The cellular senescence was also suppressed by the concomitant knockdown of E2F-4/HSP27 or p130/HSP27. Our findings indicate that HSP27 promotes cell cycle progression of MRC-5 cells by suppressing expression of the transcriptional repressors E2F-4 and p130.
Collapse
Affiliation(s)
- Ah-Mee Park
- From the Department of Microbiology, Kindai University Faculty of Medicine, Osakasayama, Osaka 589-8511, Japan and
| | - Ikuo Tsunoda
- From the Department of Microbiology, Kindai University Faculty of Medicine, Osakasayama, Osaka 589-8511, Japan and
| | - Osamu Yoshie
- From the Department of Microbiology, Kindai University Faculty of Medicine, Osakasayama, Osaka 589-8511, Japan and
- the Health and Kampo Institute, 1-11-10 Murasakiyama, Sendai, Miyagi 981-3205, Japan
| |
Collapse
|
34
|
Indole-3-Carbinol (I3C) enhances the sensitivity of murine breast adenocarcinoma cells to doxorubicin (DOX) through inhibition of NF-κβ, blocking angiogenesis and regulation of mitochondrial apoptotic pathway. Chem Biol Interact 2018; 290:19-36. [DOI: 10.1016/j.cbi.2018.05.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 05/06/2018] [Accepted: 05/14/2018] [Indexed: 12/21/2022]
|
35
|
Nemade H, Chaudhari U, Acharya A, Hescheler J, Hengstler JG, Papadopoulos S, Sachinidis A. Cell death mechanisms of the anti-cancer drug etoposide on human cardiomyocytes isolated from pluripotent stem cells. Arch Toxicol 2018; 92:1507-1524. [PMID: 29397400 PMCID: PMC5882643 DOI: 10.1007/s00204-018-2170-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 01/31/2018] [Indexed: 12/14/2022]
Abstract
Etoposide (ETP) and anthracyclines are applied for wide anti-cancer treatments. However, the ETP-induced cardiotoxicity remains to be a major safety issue and the underlying cardiotoxic mechanisms are not well understood. This study is aiming to unravel the cardiotoxicity profile of ETP in comparison to anthracyclines using physiologically relevant human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs). Using xCELLigence real-time cell analyser (RTCA), we found that single high dose of ETP induces irreversible increase in hPSC-CMs beating rate and decrease in beating amplitude. We also identified 58 deregulated genes consisting of 33 upregulated and 25 downregulated genes in hPSC-CMs after ETP treatment. Gene ontology (GO) and pathway analysis showed that most upregulated genes are enriched in GO categories like positive regulation of apoptotic process, regulation of cell death, and mitochondria organization, whereas most downregulated genes were enriched in GO categories like cytoskeletal organization, muscle contraction, and Ca2+ ion homeostasis. Moreover, we also found upregulation in 5 miRNAs (has-miR-486-3p, has-miR-34c-5p, has-miR-4423-3p, has-miR-182-5p, and has-miR-139-5p) which play role in muscle contraction, arginine and proline metabolism, and hypertrophic cardiomyopathy (HCM). Immunostaining and transmission electron microscopy also confirmed the cytoskeletal and mitochondrial damage in hPSC-CMs treated with ETP, as well as noticeable alterations in intracellular calcium handling and mitochondrial membrane potential were also observed. The apoptosis inhibitor, Pifithrin-α, found to protect hPSC-CMs from ETP-induced cardiotoxicity, whereas hPSC-CMs treated with ferroptosis inhibitor, Liproxstatin-1, showed significant recovery in hPSC-CMs functional properties like beating rate and amplitude after ETP treatment. We suggest that the damage to mitochondria is a major contributing factor involved in ETP-induced cardiotoxicity and the activation of the p53-mediated ferroptosis pathway by ETP is likely the critical pathway in ETP-induced cardiotoxicity. We also conclude that the genomic biomarkers identified in this study will significantly contribute to develop and predict potential cardiotoxic effects of novel anti-cancer drugs in vitro.
Collapse
Affiliation(s)
- Harshal Nemade
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne (UKK), Robert-Koch-Str. 39, 50931, Cologne, Germany
| | - Umesh Chaudhari
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne (UKK), Robert-Koch-Str. 39, 50931, Cologne, Germany
| | - Aviseka Acharya
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne (UKK), Robert-Koch-Str. 39, 50931, Cologne, Germany
| | - Jürgen Hescheler
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne (UKK), Robert-Koch-Str. 39, 50931, Cologne, Germany
| | - Jan Georg Hengstler
- Leibniz Research Centre for Working Environment and Human Factors, Technical University of Dortmund (IfADo), 44139, Dortmund, Germany
| | - Symeon Papadopoulos
- Center of Physiology and Pathophysiology, Institute of Vegetative Physiology, University of Cologne, Robert-Koch-Str. 39, 50931, Cologne, Germany
| | - Agapios Sachinidis
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne (UKK), Robert-Koch-Str. 39, 50931, Cologne, Germany.
| |
Collapse
|
36
|
Chakraborti S, Pramanick A, Saha S, Roy SS, Chaudhuri AR, Das M, Ghosh S, Stewart A, Maity B. Atypical G Protein β5 Promotes Cardiac Oxidative Stress, Apoptosis, and Fibrotic Remodeling in Response to Multiple Cancer Chemotherapeutics. Cancer Res 2017; 78:528-541. [DOI: 10.1158/0008-5472.can-17-1280] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 09/08/2017] [Accepted: 11/13/2017] [Indexed: 11/16/2022]
|
37
|
Molecular mechanism of doxorubicin-induced cardiomyopathy - An update. Eur J Pharmacol 2017; 818:241-253. [PMID: 29074412 DOI: 10.1016/j.ejphar.2017.10.043] [Citation(s) in RCA: 392] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 10/11/2017] [Accepted: 10/20/2017] [Indexed: 12/27/2022]
Abstract
Doxorubicin is utilized for anti-neoplastic treatment for several decades. The utility of this drug is limited due to its side effects. Generally, doxorubicin toxicity is originated from the myocardium and then other organs are also ruined. The mechanism of doxorubicin is intercalated with the DNA and inhibits topoisomerase 2. There are various signalling mechanisms involved in doxorubicin cardiotoxicity. First and foremost, the doxorubicin-induced cardiotoxicity is due to oxidative stress. Cardiac mitochondrial damage is supposed after few hours following the revelation of doxorubicin. This has led important new uses for the mechanism of doxorubicin-induced cardiotoxicity and novel avenues of investigation to determine better pharmacotherapies and interventions for the impediment of cardiotoxicity. The idea of this review is to bring up to date the recent findings of the mechanism of doxorubicin cardiomyopathies such as calcium dysregulation, endoplasmic reticulum stress, impairment of progenitor cells, activation of immune, ubiquitous system and some other parameters.
Collapse
|
38
|
Anderson R, Richardson GD, Passos JF. Mechanisms driving the ageing heart. Exp Gerontol 2017; 109:5-15. [PMID: 29054534 DOI: 10.1016/j.exger.2017.10.015] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 10/16/2017] [Indexed: 01/07/2023]
Abstract
Cardiovascular disease (CVD) is the leading cause of death globally. One of the main risk factors for CVD is age, however the biological processes that occur in the heart during ageing are poorly understood. It is therefore important to understand the fundamental mechanisms driving heart ageing to enable the development of preventions and treatments targeting these processes. Cellular senescence is often described as the irreversible cell-cycle arrest which occurs in somatic cells. Emerging evidence suggests that cellular senescence plays a key role in heart ageing, however the cell-types involved and the underlying mechanisms are not yet elucidated. In this review we discuss the current understanding of how mechanisms known to contribute to senescence impact on heart ageing and CVD. Finally, we evaluate recent data suggesting that targeting senescent cells may be a viable therapy to counteract the ageing of the heart.
Collapse
Affiliation(s)
- Rhys Anderson
- The Randall Division, King's College London, London, UK; Ageing Research Laboratories, Newcastle University Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK; Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK.
| | - Gavin D Richardson
- Cardiovascular Research Centre, Institute for Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - João F Passos
- Ageing Research Laboratories, Newcastle University Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK; Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
39
|
Chen YX, Zhu DY, Yin JH, Yin WJ, Zhang YL, Ding H, Yu XW, Mei J, Gao YS, Zhang CQ. The protective effect of PFTα on alcohol-induced osteonecrosis of the femoral head. Oncotarget 2017; 8:100691-100707. [PMID: 29246013 PMCID: PMC5725055 DOI: 10.18632/oncotarget.19160] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 06/29/2017] [Indexed: 12/11/2022] Open
Abstract
Epidemiologic studies have shown alcohol plays a pivotal role in the development of osteonecrosis of the femoral head (ONFH). The aim of this study was to explore the underlying mechanism of alcohol-induced ONFH and the protective effect of pifithrin-α (PFTα). In vitro, we found ethanol treatment significantly activated p53, suppressed Wnt/β-catenin signaling and inhibited osteogenic-related proteins. Furthermore, by separating the cytoplasmic and nuclear proteins, we found ethanol inhibited osteogenesis by impairing the accumulation of β-catenin in both the cytoplasm and nucleus in human bone mesenchymal stem cells (hBMSCs), which resulted from activating glycogen synthase kinase-3β (GSK-3β). Therefore, PFTα, a p53 inhibitor, was introduced in this study to block the ethanol-triggered activation of p53 in hBMSCs and alcohol-induced ONFH in a rat model. In vivo, we established alcohol-induced ONFH in rats and investigated the protective effect of PFTα. Hematoxylin & eosin (H&E) staining combined with TdT-mediated dUTP nick end labeling (TUNEL), cleaved caspase-3 immunohistochemical staining, and micro-CT images revealed substantial ONFH in the alcohol-administered rats, whereas significantly less osteonecrosis developed in the rats injected with PFTα. Osteogenic-related proteins, including osteocalcin, osteopontin and collagen I, were significantly decreased in the alcohol-administered rats, whereas these results were reversed in the PFTα-injected rats. Fluorochrome labeling similarly showed that alcohol significantly reduced the osteogenic activity in the rat femoral head, which was blocked by the injection of PFTα. In conclusion, PFTα had an antagonistic effect against the effects of ethanol on hBMSCs and could be a clinical strategy to prevent the development of alcohol-induced ONFH.
Collapse
Affiliation(s)
- Yi-Xuan Chen
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Dao-Yu Zhu
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Jun-Hui Yin
- Institute of Microsurgery on Extremities, Shanghai 200233, China
| | - Wen-Jing Yin
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Yue-Lei Zhang
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Hao Ding
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Xiao-Wei Yu
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Jiong Mei
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - You-Shui Gao
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Chang-Qing Zhang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.,Institute of Microsurgery on Extremities, Shanghai 200233, China
| |
Collapse
|
40
|
Huang L, Zhang Q, Dai L, Shen X, Chen W, Cai K. Phenylboronic acid-modified hollow silica nanoparticles for dual-responsive delivery of doxorubicin for targeted tumor therapy. Regen Biomater 2017; 4:111-124. [PMID: 30792886 PMCID: PMC6371689 DOI: 10.1093/rb/rbw045] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 11/22/2016] [Accepted: 12/03/2016] [Indexed: 01/01/2023] Open
Abstract
This work reports a multifunctional nanocarrier based on hollow mesoporous silica nanoparticles (HMSNs) for targeting tumor therapy. Doxorubicin (DOX) was loaded into HMSNs and blocked with cytochrome C conjugated lactobionic acid (CytC-LA) via redox-cleavable disulfide bonds and pH-disassociation boronate ester bonds as intermediate linkers. The CytC-LA was used both as sealing agent and targeting motif. A series of characterizations demonstrated the successful construction of the drug delivery system. The system demonstrated pH and redox dual-responsive drug release behavior in vitro. The DOX loading HMSNs system displayed a good biocompatibility, which could be specifically endocytosed by HepG2 cells and led to high cytotoxicity against tumor cells by inducing cell apoptosis. In vivo data (tumor volume, tumor weight, terminal deoxynucleotidyl transferase dUTP nick end labeling and hematoxylin and eosin staining) proved that the system could deliver DOX to tumor site with high efficiency and inhibit tumor growth with minimal toxic side effect.
Collapse
Affiliation(s)
| | | | | | | | | | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing 400044, P. R. China
| |
Collapse
|
41
|
Grant MKO, Seelig DM, Sharkey LC, Zordoky BN. Sex-dependent alteration of cardiac cytochrome P450 gene expression by doxorubicin in C57Bl/6 mice. Biol Sex Differ 2017; 8:1. [PMID: 28078076 PMCID: PMC5219702 DOI: 10.1186/s13293-016-0124-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 12/16/2016] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND There is inconclusive evidence about the role of sex as a risk factor for doxorubicin (DOX)-induced cardiotoxicity. Recent experimental studies have shown that adult female rats are protected against DOX-induced cardiotoxicity. However, the mechanisms of this sexual dimorphism are not fully elucidated. We have previously demonstrated that DOX alters the expression of several cytochrome P450 (CYP) enzymes in the hearts of male rats. Nevertheless, the sex-dependent effect of DOX on the expression of CYP enzymes is still not known. Therefore, in the present study, we determined the effect of acute DOX exposure on the expression of CYP genes in the hearts of both male and female C57Bl/6 mice. METHODS Acute DOX cardiotoxicity was induced by a single intraperitoneal injection of 20 mg/kg DOX in male and female adult C57Bl/6 mice. Cardiac function was assessed 5 days after DOX exposure by trans-thoracic echocardiography. Mice were euthanized 1 day or 6 days after DOX or saline injection. Thereafter, the hearts were harvested and weighed. Heart sections were evaluated for pathological lesions. Total RNA was extracted and expression of natriuretic peptides, inflammatory and apoptotic markers, and CYP genes was measured by real-time PCR. RESULTS Adult female C57Bl/6 mice were protected from acute DOX-induced cardiotoxicity as they show milder pathological lesions, less inflammation, and faster recovery from DOX-induced apoptosis and DOX-mediated inhibition of beta-type natriuretic peptide. Acute DOX exposure altered the gene expression of multiple CYP genes in a sex-dependent manner. In 24 h, DOX exposure caused male-specific induction of Cyp1b1 and female-specific induction of Cyp2c29 and Cyp2e1. CONCLUSIONS Acute DOX exposure causes sex-dependent alteration of cardiac CYP gene expression. Since cardiac CYP enzymes metabolize several endogenous compounds to biologically active metabolites, sex-dependent alteration of CYP genes may play a role in the sexual dimorphism of acute DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Marianne K O Grant
- Department of Experimental and Clinical Pharmacology, University of Minnesota, 308 Harvard St S.E, Minneapolis, MN 55455 USA
| | - Davis M Seelig
- Veterinary Clinical Sciences Department, University of Minnesota, 1352 Boyd Ave, St. Paul, MN 55108 USA
| | - Leslie C Sharkey
- Veterinary Clinical Sciences Department, University of Minnesota, 1352 Boyd Ave, St. Paul, MN 55108 USA
| | - Beshay N Zordoky
- Department of Experimental and Clinical Pharmacology, University of Minnesota, 308 Harvard St S.E, Minneapolis, MN 55455 USA
| |
Collapse
|
42
|
Myricitrin Protects against Doxorubicin-Induced Cardiotoxicity by Counteracting Oxidative Stress and Inhibiting Mitochondrial Apoptosis via ERK/P53 Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:6093783. [PMID: 27703489 PMCID: PMC5039279 DOI: 10.1155/2016/6093783] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 05/08/2016] [Accepted: 06/05/2016] [Indexed: 12/20/2022]
Abstract
Doxorubicin (Dox) is one of the most effective and widely used anthracycline antineoplastic antibiotics. Unfortunately, the use of Dox is limited by its cumulative and dose-dependent cardiac toxicity. Myricitrin, a natural flavonoid which is isolated from the ground bark of Myrica rubra, has recently been found to have a strong antioxidative effect. This study aimed to evaluate the possible protective effect of myricitrin against Dox-induced cardiotoxicity and the underlying mechanisms. An in vivo investigation in SD rats demonstrated that myricitrin significantly reduced the Dox-induced myocardial damage, as indicated by the decreases in the cardiac index, amelioration of heart pathological injuries, and decreases in the serum cardiac enzyme levels. In addition, in vitro studies showed that myricitrin effectively reduced the Dox-induced cell toxicity. Further study showed that myricitrin exerted its function by counteracting oxidative stress and increasing the activities of antioxidant enzymes. Moreover, myricitrin suppressed the myocardial apoptosis induced by Dox, as indicated by decreases in the activation of caspase-3 and the numbers of TUNEL-positive cells, maintenance of the mitochondrial membrane potential, and increase in the Bcl-2/Bax ratio. Further mechanism study revealed that myricitrin-induced suppression of myocardial apoptosis relied on the ERK/p53-mediated mitochondrial apoptosis pathway.
Collapse
|
43
|
Mazelin L, Panthu B, Nicot AS, Belotti E, Tintignac L, Teixeira G, Zhang Q, Risson V, Baas D, Delaune E, Derumeaux G, Taillandier D, Ohlmann T, Ovize M, Gangloff YG, Schaeffer L. mTOR inactivation in myocardium from infant mice rapidly leads to dilated cardiomyopathy due to translation defects and p53/JNK-mediated apoptosis. J Mol Cell Cardiol 2016; 97:213-25. [DOI: 10.1016/j.yjmcc.2016.04.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 04/05/2016] [Accepted: 04/12/2016] [Indexed: 10/21/2022]
|
44
|
Wang L, Zhang TP, Zhang Y, Bi HL, Guan XM, Wang HX, Wang X, Du J, Xia YL, Li HH. Protection against doxorubicin-induced myocardial dysfunction in mice by cardiac-specific expression of carboxyl terminus of hsp70-interacting protein. Sci Rep 2016; 6:28399. [PMID: 27323684 PMCID: PMC4914971 DOI: 10.1038/srep28399] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 06/01/2016] [Indexed: 01/06/2023] Open
Abstract
Carboxyl terminus of Hsp70-interacting protein (CHIP) is a critical ubiquitin ligase/cochaperone to reduce cardiac oxidative stress, inflammation, cardiomyocyte apoptosis and autophage etc. However, it is unclear whether overexpression of CHIP in the heart would exert protective effects against DOX-induced cardiomyopathy. Cardiac-specific CHIP transgenic (CHIP-TG) mice and the wild-type (WT) littermates were treated with DOX or saline. DOX-induced cardiac atrophy, dysfunction, inflammation, oxidative stress and cardiomyocyte apoptosis were significantly attenuated in CHIP-TG mice. CHIP-TG mice also showed higher survival rate than that of WT mice (40% versus 10%) after 10-day administration of DOX. In contrast, knockdown of CHIP by siRNA in vitro further enhanced DOX-induced cardiotoxic effects. Global gene microarray assay revealed that after DOX-treatment, differentially expressed genes between WT and CHIP-TG mice were mainly involved in apoptosis, atrophy, immune/inflammation and oxidative stress. Mechanistically, CHIP directly promotes ubiquitin-mediated degradation of p53 and SHP-1, which results in activation of ERK1/2 and STAT3 pathways thereby ameliorating DOX-induced cardiac toxicity.
Collapse
Affiliation(s)
- Lei Wang
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Tian-Peng Zhang
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Yuan Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Baotou Medical College, Baotou 014060, China
| | - Hai-Lian Bi
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xu-Min Guan
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Hong-Xia Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Xia Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Jie Du
- Beijing AnZhen Hospital the Key Laboratory of Remodeling-Related Cardiovascular Diseases, Capital Medical University, Beijing 100029, China
| | - Yun-Long Xia
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Hui-Hua Li
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.,Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian 116044, China
| |
Collapse
|
45
|
Zeng F, Yu X, Sherry JP, Dixon B, Duncker BP, Bols NC. The p53 inhibitor, pifithrin-α, disrupts microtubule organization, arrests growth, and induces polyploidy in the rainbow trout gill cell line, RTgill-W1. Comp Biochem Physiol C Toxicol Pharmacol 2016; 179:1-10. [PMID: 26291498 DOI: 10.1016/j.cbpc.2015.08.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 08/04/2015] [Accepted: 08/10/2015] [Indexed: 11/16/2022]
Abstract
Pifithrin-α (PFT-α) blocks p53-dependent transcription and is an example of the many drugs being developed to target the p53 pathway in humans that could be released into the environment with potential impacts on aquatic animals if they were to become successful pharmaceuticals. In order to understand how p53 drugs might act on fish, the effects of PFT-α on rainbow trout gill epithelial cell line, RTgill-W1, were studied. PFT-α was not cytotoxic to RTgill-W1 in cultures with or without fetal bovine serum (FBS), but at 5.25μg/ml, PFT-α completely arrested proliferation. When FBS was present, PFT-α increased the number of polyploid cells over 12days. Those results suggest that like in mammals, p53 appears to regulate ploidy in fish. However, several effects were seen that have not been observed with mammalian cells. PFT-α caused a transient rise in the mitotic index and a disruption in cytoskeletal microtubules. These results suggest that in fish cells PFT-α affects microtubules either directly through an off-target action on tubulin or indirectly through an on-target action on p53-regulated transcription.
Collapse
Affiliation(s)
- Fanxing Zeng
- Department of Biology, University of Waterloo, Waterloo, ON, Canada N2L 3G1
| | - Xiang Yu
- Department of Biology, University of Waterloo, Waterloo, ON, Canada N2L 3G1
| | - James P Sherry
- Aquatic Contaminants Research Division, Environment Canada, Burlington, ON, Canada L7R 4A6
| | - Brian Dixon
- Department of Biology, University of Waterloo, Waterloo, ON, Canada N2L 3G1
| | - Bernard P Duncker
- Department of Biology, University of Waterloo, Waterloo, ON, Canada N2L 3G1
| | - Niels C Bols
- Department of Biology, University of Waterloo, Waterloo, ON, Canada N2L 3G1.
| |
Collapse
|
46
|
Baicalein alleviates doxorubicin-induced cardiotoxicity via suppression of myocardial oxidative stress and apoptosis in mice. Life Sci 2016; 144:8-18. [DOI: 10.1016/j.lfs.2015.11.018] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 10/30/2015] [Accepted: 11/19/2015] [Indexed: 01/20/2023]
|
47
|
Zheng T, Lu Y. SIRT1 Protects Human Lens Epithelial Cells Against Oxidative Stress by Inhibiting p53-Dependent Apoptosis. Curr Eye Res 2015; 41:1068-1075. [DOI: 10.3109/02713683.2015.1093641] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
48
|
Zhao WJ, Wei SN, Zeng XJ, Xia YL, Du J, Li HH. Gene expression profiling identifies the novel role of immunoproteasome in doxorubicin-induced cardiotoxicity. Toxicology 2015; 333:76-88. [DOI: 10.1016/j.tox.2015.04.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 04/13/2015] [Accepted: 04/14/2015] [Indexed: 02/02/2023]
|
49
|
Saeed NM, El-Naga RN, El-Bakly WM, Abdel-Rahman HM, Salah ElDin RA, El-Demerdash E. Epigallocatechin-3-gallate pretreatment attenuates doxorubicin-induced cardiotoxicity in rats: A mechanistic study. Biochem Pharmacol 2015; 95:145-155. [PMID: 25701654 DOI: 10.1016/j.bcp.2015.02.006] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 02/10/2015] [Accepted: 02/12/2015] [Indexed: 12/24/2022]
Abstract
Doxorubicin (DOX) is a widely used chemotherapeutic agent however its clinical use is limited by cumulative cardiotoxicity. Epigallocatechin-3-gallate (EGCG), a main catechin in green tea, possesses a potent antioxidant, anti-apoptotic and anticancer properties. The current study aimed to investigate the potential protective effect of EGCG against DOX-induced cardiotoxicity. Firstly the potential cardioprotective dose of EGCG was screened at different doses (10, 20 and 40 mg/kg/day) against a single dose of DOX (15 mg/kg; i.p.). EGCG protected against DOX-induced ECG changes, leakage of cardiac enzymes (creatine kinase isoenzyme-MB, and lactate dehydrogenase) and histopathological changes. The dose of 40 mg/kg EGCG was selected for further assessment to address the EGCG cardioprotective mechanisms. EGCG was given orally 3 times/week for 4 consecutive weeks and DOX (2.5 mg/kg; i.p.) 3 times/week on the last 2 weeks. EGCG significantly ameliorated oxidative stress injury evoked by DOX as evidenced by inhibition of reduced glutathione depletion and lipid peroxidation as well as elevation of antioxidant enzyme activities. DOX caused down-regulation of ErbB2 expression while EGCG pretreatment significantly increased ErbB2 expression indicating its effect on pro-survival pathway. Furthermore, DOX provoked apoptotic responses evidenced by increasing the expression of nuclear factor kappa-B, tumor suppressor protein p53, calpain 2, caspases 3 and 12. Additionally basal level of Hsp70 was reduced in DOX-intoxicated group. EGCG pretreatment significantly ameliorated these apoptotic signals indicating its anti-inflammatory and anti-apoptotic actions. In conclusion, EGCG possesses cardioprotective action against DOX-induced cardiotoxicity by suppressing oxidative stress, inflammation and apoptotic signals as well as activation of pro-survival pathways.
Collapse
Affiliation(s)
- Noha M Saeed
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Reem N El-Naga
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Wesam M El-Bakly
- Department of Pharmacology & Therapeutics, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Hanaa M Abdel-Rahman
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Rania A Salah ElDin
- Department of Anatomy, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Ebtehal El-Demerdash
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
50
|
Ali SA, Zaitone SA, Moustafa YM. Boswellic acids synergize antitumor activity and protect against the cardiotoxicity of doxorubicin in mice bearing Ehrlich's carcinoma. Can J Physiol Pharmacol 2015; 93:695-708. [PMID: 26230640 DOI: 10.1139/cjpp-2014-0524] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This study aimed to test whether boswellic acids add to the antitumor effects of doxorubicin against solid tumors of Ehrlich's ascites carcinoma (EAC) grown in mice, and to investigate the protective effects of boswellic acids against doxorubicin-induced cardiotoxicity. Sixty-four female Swiss albino mice bearing EAC solid tumors were distributed among 8 groups as follows: group 1, EAC control group; group 2, doxorubicin treatment group [mice were injected with doxorubicin (6 mg·(kg body mass)(-1)·week(-1)) for 3 weeks]; groups 3-5, these mice were treated with boswellic acids (125, 250, or 500 mg·kg(-1)·day(-1)), respectively; groups 6-8, these mice were treated with a combination of doxorubicin and boswellic acids (125, 250, or 500 mg·kg(-1)·day(-1)), respectively, for 3 weeks. The results indicated that boswellic acids synergized the antitumor activity of doxorubicin. Doxorubicin-treated mice showed elevated serum activities of lactate dehydrogenase and creatine kinase isoenzyme MB as well as cardiac malondialdehyde. Further, decreases in cardiac levels of reduced glutathione, superoxide dismutase, and catalase activities were observed. These effects were accompanied by an increase in cardiac expression of caspase 3. Thus, treatment with boswellic acids attenuated doxorubicin-evoked disturbances in the above-mentioned parameters, highlighting antioxidant and antiapoptotic activities. Therefore, boswellic acids could be potential candidates for ameliorating the cardiotoxicity of doxorubicin.
Collapse
Affiliation(s)
- Shimaa A Ali
- a Suez Canal Authority hospital, Ismailia, Egypt
| | - Sawsan A Zaitone
- b Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Yasser M Moustafa
- b Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|