1
|
Feng P, Yang F, Zang D, Bai D, Xu L, Fu Y, You R, Liu T, Yang X. Deciphering the roles of cellular and extracellular non-coding RNAs in chemotherapy-induced cardiotoxicity. Mol Cell Biochem 2025; 480:2177-2199. [PMID: 39485641 PMCID: PMC11961477 DOI: 10.1007/s11010-024-05143-5] [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: 08/23/2024] [Accepted: 10/18/2024] [Indexed: 11/03/2024]
Abstract
Chemotherapy-induced cardiotoxicity is a major adverse effect, driven by multiple factors in its pathogenesis. Notably, RNAs have emerged as significant contributors in both cancer and heart failure (HF). RNAs carry genetic and metabolic information that mirrors the current state of cells, making them valuable as potential biomarkers and therapeutic tools for diagnosing, predicting, and treating a range of diseases, including cardiotoxicity. Over 97% of the genome is transcribed into non-coding RNAs (ncRNAs), including ribosomal RNA (rRNAs), transfer RNAs (tRNAs), and newly identified microRNAs (miRNAs), circular RNAs (circRNAs), and long non-coding RNAs (lncRNAs). NcRNAs function not only within their originating cells but also in recipient cells by being transported through extracellular compartments, referred to as extracellular RNAs (exRNAs). Since ncRNAs were identified as key regulators of gene expression, numerous studies have highlighted their significance in both cancer and cardiovascular diseases. Nevertheless, the role of ncRNAs in cardiotoxicity remains not fully elucidated. The study aims to review the existing knowledge on ncRNAs in Cardio-Oncology and explore the potential of ncRNA-based biomarkers and therapies. These investigations could advance the clinical application of ncRNA research, improving early detection and mitigating of chemotherapy-induced cardiotoxicity.
Collapse
Affiliation(s)
- Pan Feng
- Baoji Hospital of Traditional Chinese Medicine, Baoji, 721000, China
| | - Fan Yang
- Guang'an Men Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Dongmei Zang
- Fangshan Hospital Beijing University of Chinese Medicine, Beijing, 102400, China
| | - Dapeng Bai
- Fangshan Hospital Beijing University of Chinese Medicine, Beijing, 102400, China
| | - Liyan Xu
- Fangshan Hospital Beijing University of Chinese Medicine, Beijing, 102400, China
| | - Yueyun Fu
- Fangshan Hospital Beijing University of Chinese Medicine, Beijing, 102400, China
| | - Ranran You
- Fangshan Hospital Beijing University of Chinese Medicine, Beijing, 102400, China
| | - Tao Liu
- Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710068, China.
| | - Xinyu Yang
- Fangshan Hospital Beijing University of Chinese Medicine, Beijing, 102400, China.
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
2
|
Adamcova M, Parova H, Lencova-Popelova O, Kollarova-Brazdova P, Baranova I, Slavickova M, Stverakova T, Mikyskova PS, Mazurova Y, Sterba M. Cardiac miRNA expression during the development of chronic anthracycline-induced cardiomyopathy using an experimental rabbit model. Front Pharmacol 2024; 14:1298172. [PMID: 38235109 PMCID: PMC10791979 DOI: 10.3389/fphar.2023.1298172] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 12/06/2023] [Indexed: 01/19/2024] Open
Abstract
Background: Anthracycline cardiotoxicity is a well-known complication of cancer treatment, and miRNAs have emerged as a key driver in the pathogenesis of cardiovascular diseases. This study aimed to investigate the expression of miRNAs in the myocardium in early and late stages of chronic anthracycline induced cardiotoxicity to determine whether this expression is associated with the severity of cardiac damage. Method: Cardiotoxicity was induced in rabbits via daunorubicin administration (daunorubicin, 3 mg/kg/week; for five and 10 weeks), while the control group received saline solution. Myocardial miRNA expression was first screened using TaqMan Advanced miRNA microfluidic card assays, after which 32 miRNAs were selected for targeted analysis using qRT-PCR. Results: The first subclinical signs of cardiotoxicity (significant increase in plasma cardiac troponin T) were observed after 5 weeks of daunorubicin treatment. At this time point, 10 miRNAs (including members of the miRNA-34 and 21 families) showed significant upregulation relative to the control group, with the most intense change observed for miRNA-1298-5p (29-fold change, p < 0.01). After 10 weeks of daunorubicin treatment, when a further rise in cTnT was accompanied by significant left ventricle systolic dysfunction, only miR-504-5p was significantly (p < 0.01) downregulated, whereas 10 miRNAs were significantly upregulated relative to the control group; at this time-point, the most intense change was observed for miR-34a-5p (76-fold change). Strong correlations were found between the expression of multiple miRNAs (including miR-34 and mir-21 family and miR-1298-5p) and quantitative indices of toxic damage in both the early and late phases of cardiotoxicity development. Furthermore, plasma levels of miR-34a-5p were strongly correlated with the myocardial expression of this miRNA. Conclusion: To the best of our knowledge, this is the first study that describes alterations in miRNA expression in the myocardium during the transition from subclinical, ANT-induced cardiotoxicity to an overt cardiotoxic phenotype; we also revealed how these changes in miRNA expression are strongly correlated with quantitative markers of cardiotoxicity.
Collapse
Affiliation(s)
| | - Helena Parova
- Department of Clinical Biochemistry and Diagnostics, Faculty of Medicine in Hradec Kralove and University Hospital Hradec Kralove, Hradec Kralove, Czechia
| | | | | | - Ivana Baranova
- Department of Clinical Biochemistry and Diagnostics, Faculty of Medicine in Hradec Kralove and University Hospital Hradec Kralove, Hradec Kralove, Czechia
| | - Marcela Slavickova
- Department of Clinical Biochemistry and Diagnostics, Faculty of Medicine in Hradec Kralove and University Hospital Hradec Kralove, Hradec Kralove, Czechia
| | - Tereza Stverakova
- Department of Clinical Biochemistry and Diagnostics, Faculty of Medicine in Hradec Kralove and University Hospital Hradec Kralove, Hradec Kralove, Czechia
| | - Petra Sauer Mikyskova
- Department of Clinical Biochemistry and Diagnostics, Faculty of Medicine in Hradec Kralove and University Hospital Hradec Kralove, Hradec Kralove, Czechia
| | - Yvona Mazurova
- Department of Histology and Embryology, Charles University in Prague, Hradec Kralove, Czechia
| | | |
Collapse
|
3
|
Atteia HH. MicroRNAs in Anticancer Drugs Hepatotoxicity: From Pathogenic Mechanism and Early Diagnosis to Therapeutic Targeting by Natural Products. Curr Pharm Biotechnol 2024; 25:1791-1806. [PMID: 38178678 DOI: 10.2174/0113892010282155231222071903] [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: 09/13/2023] [Revised: 11/11/2023] [Accepted: 11/24/2023] [Indexed: 01/06/2024]
Abstract
Patients receiving cancer therapies experience severe adverse effects, including hepatotoxicity, even at therapeutic doses. Consequently, monitoring patients on cancer therapy for hepatic functioning is necessary to avoid permanent liver damage. Several pathways of anticancer drug-induced hepatotoxicity involve microRNAs (miRNAs) via targeting mRNAs. These short and non-coding RNAs undergo rapid modulation in non-targeted organs due to cancer therapy insults. Recently, there has been an interest for miRNAs as useful and promising biomarkers for monitoring toxicity since they have conserved sequences across species and are cellular-specific, stable, released during injury, and simple to analyze. Herein, we tried to review the literature handling miRNAs as mediators and biomarkers of anticancer drug-induced hepatotoxicity. Natural products and phytochemicals are suggested as safe and effective candidates in treating cancer. There is also an attempt to combine anticancer drugs with natural compounds to enhance their efficiencies and reduce systemic toxicities. We also discussed natural products protecting against chemotherapy hepatotoxicity via modulating miRNAs, given that miRNAs have pathogenic and diagnostic roles in chemotherapy-induced hepatotoxicity and that many natural products can potentially regulate their expression. Future studies should integrate these findings into clinical trials by formulating suitable therapeutic dosages of natural products to target miRNAs involved in anticancer drug hepatotoxicity.
Collapse
Affiliation(s)
- Hebatallah Husseini Atteia
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Sharkia, 44519, Egypt
| |
Collapse
|
4
|
Zare N, Dana N, Mosayebi A, Vaseghi G, Javanmard SH. Evaluation of Expression Level of miR-3135b-5p in Blood Samples of Breast Cancer Patients Experiencing Chemotherapy-Induced Cardiotoxicity. Indian J Clin Biochem 2023; 38:536-540. [PMID: 37746544 PMCID: PMC10516830 DOI: 10.1007/s12291-022-01075-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 07/12/2022] [Indexed: 11/26/2022]
Abstract
The efficacy of chemotherapeutics in the treatment of breast cancer is limited by cardiotoxicity, which could lead to irreversible heart failure. The evaluation of miRNA levels as a vital biomarker could predict cardiotoxicity induced by chemotherapy. According to our previous meta-analysis study on patients with heart failure, we found that miR-3135b had a significant increase in patients with heart failure. Therefore, the present study aimed to evaluate the expression level of miR-3135b in the blood sample of patients experiencing chemotherapy-induced cardiotoxicity. Blood samples were collected from breast cancer patients or breast cancer patients who had received chemotherapy and had not experienced any chemotherapy-induced cardiotoxicity (N = 37, control group) and breast cancer patients experiencing chemotherapy-induced cardiotoxicity after chemotherapy (N = 33). The expression level of miR-3135b was evaluated using real-time polymerase chain reaction (RT-PCR). The 2-ΔCt values of miR-3135b were compared between two groups. We observed a significant increase in the expression level of miR-3135b between patients experiencing chemotherapy-induced cardiotoxicity and the control group (P = 0.0001). Besides, the ejection fraction parameter was correlated with the expression level of miR-3135b (r = 0.5 and P = 0.0001). To sum up, miR-3135b might be useful as a promising circulating biomarker in predicting cardiotoxicity induced by chemotherapy. However, more studies are needed to validate miR-3135b as a biomarker for the diagnosis of chemotherapy-induced cardiotoxicity. Supplementary Information The online version contains supplementary material available at 10.1007/s12291-022-01075-3.
Collapse
Affiliation(s)
- Nasrin Zare
- School of Medicine, Najafabad Branch, Islamic Azad University, Najafabad, Iran
- Clinical Research Development Centre, Najafabad branch, Islamic Azad university, Najafabad, Iran
| | - Nasim Dana
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Azam Mosayebi
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Golnaz Vaseghi
- Interventional Cardiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shaghayegh Haghjooy Javanmard
- Department of Physiology, Applied Physiology Research Center, Cardiovascular Research Institute , Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Physiology, Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Hezar jerib Avenue, Isfahan, Iran
| |
Collapse
|
5
|
Kuang Z, Wu J, Tan Y, Zhu G, Li J, Wu M. MicroRNA in the Diagnosis and Treatment of Doxorubicin-Induced Cardiotoxicity. Biomolecules 2023; 13:biom13030568. [PMID: 36979503 PMCID: PMC10046787 DOI: 10.3390/biom13030568] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/12/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Doxorubicin (DOX), a broad-spectrum chemotherapy drug, is widely applied to the treatment of cancer; however, DOX-induced cardiotoxicity (DIC) limits its clinical therapeutic utility. However, it is difficult to monitor and detect DIC at an early stage using conventional detection methods. Thus, sensitive, accurate, and specific methods of diagnosis and treatment are important in clinical practice. MicroRNAs (miRNAs) belong to non-coding RNAs (ncRNAs) and are stable and easy to detect. Moreover, miRNAs are expected to become biomarkers and therapeutic targets for DIC; thus, there are currently many studies focusing on the role of miRNAs in DIC. In this review, we list the prominent studies on the diagnosis and treatment of miRNAs in DIC, explore the feasibility and difficulties of using miRNAs as diagnostic biomarkers and therapeutic targets, and provide recommendations for future research.
Collapse
Affiliation(s)
- Ziyu Kuang
- Oncology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
- Graduate School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jingyuan Wu
- Oncology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
- Graduate School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Ying Tan
- Oncology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Guanghui Zhu
- Oncology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
- Graduate School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jie Li
- Oncology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Min Wu
- Cardiovascular Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| |
Collapse
|
6
|
Rudolf K, Rudolf E. Increased Intracellular Free Zinc Has Pleiotropic Effects on Doxorubicin-Induced Cytotoxicity in hiPCS-CMs Cells. Int J Mol Sci 2023; 24:ijms24054518. [PMID: 36901950 PMCID: PMC10003200 DOI: 10.3390/ijms24054518] [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: 12/01/2022] [Revised: 02/15/2023] [Accepted: 02/21/2023] [Indexed: 03/02/2023] Open
Abstract
(1) the mechanisms and outcomes of doxorubicin (DOX)-dependent toxicity upon changed intracellular zinc (Zn) concentrations in the cardiomyocytes obtained from human-induced pluripotent stem cells (hiPCS-CMs) were investigated; (2) cells exposed to the DOX were pretreated or cotreated with zinc pyrythione (ZnPyr) and various cellular endpoints and mechanisms were analyzed via cytometric methods; (3) both DOX concentrations (0.3 and 1 µM) induced a concentration-dependent loss of viability, an activation of autophagy, cell death, and the appearance of senescence. These phenotypes were preceded by an oxidative burst, DNA damage, and a loss of mitochondrial and lysosomal integrity. Furthermore, in DOX-treated cells, proinflammatory and stress kinase signaling (in particular, JNK and ERK) were upregulated upon the loss of free intracellular Zn pools. Increased free Zn concentrations proved to have both inhibitory and stimulatory effects on the investigated DOX-related molecular mechanisms, as well as on signaling pathways on the resulting cell fates; and (4) free intracellular Zn pools, their status, and their elevation might have, in a specific context, a pleiotropic impact upon DOX-dependent cardiotoxicity.
Collapse
|
7
|
Kawano I, Adamcova M. MicroRNAs in doxorubicin-induced cardiotoxicity: The DNA damage response. Front Pharmacol 2022; 13:1055911. [PMID: 36479202 PMCID: PMC9720152 DOI: 10.3389/fphar.2022.1055911] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 11/11/2022] [Indexed: 10/17/2023] Open
Abstract
Doxorubicin (DOX) is a chemotherapeutic drug widely used for cancer treatment, but its use is limited by cardiotoxicity. Although free radicals from redox cycling and free cellular iron have been predominant as the suggested primary pathogenic mechanism, novel evidence has pointed to topoisomerase II inhibition and resultant genotoxic stress as the more fundamental mechanism. Recently, a growing list of microRNAs (miRNAs) has been implicated in DOX-induced cardiotoxicity (DIC). This review summarizes miRNAs reported in the recent literature in the context of DIC. A particular focus is given to miRNAs that regulate cellular responses downstream to DOX-induced DNA damage, especially p53 activation, pro-survival signaling pathway inhibition (e.g., AMPK, AKT, GATA-4, and sirtuin pathways), mitochondrial dysfunction, and ferroptosis. Since these pathways are potential targets for cardioprotection against DOX, an understanding of how miRNAs participate is necessary for developing future therapies.
Collapse
Affiliation(s)
| | - Michaela Adamcova
- Department of Physiology, Faculty of Medicine in Hradec Kralove, Charles University in Prague, Hradec Kralove, Czechia
| |
Collapse
|
8
|
Liu X, Tian R, Tao H, Wu J, Yang L, Zhang Y, Meng X. The cardioprotective potentials and the involved mechanisms of phenolic acids in drug-induced cardiotoxicity. Eur J Pharmacol 2022; 936:175362. [DOI: 10.1016/j.ejphar.2022.175362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/22/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
|
9
|
MicroRNA-4732-3p Is Dysregulated in Breast Cancer Patients with Cardiotoxicity, and Its Therapeutic Delivery Protects the Heart from Doxorubicin-Induced Oxidative Stress in Rats. Antioxidants (Basel) 2022; 11:antiox11101955. [PMID: 36290678 PMCID: PMC9599023 DOI: 10.3390/antiox11101955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/24/2022] [Accepted: 09/26/2022] [Indexed: 11/17/2022] Open
Abstract
Anthracycline-induced cardiotoxicity is the most severe collateral effect of chemotherapy originated by an excess of oxidative stress in cardiomyocytes that leads to cardiac dysfunction. We assessed clinical data from patients with breast cancer receiving anthracyclines and searched for discriminating microRNAs between patients that developed cardiotoxicity (cases) and those that did not (controls), using RNA sequencing and regression analysis. Serum levels of 25 microRNAs were differentially expressed in cases versus controls within the first year after anthracycline treatment, as assessed by three different regression models (elastic net, Robinson and Smyth exact negative binomial test and random forest). MiR-4732-3p was the only microRNA identified in all regression models and was downregulated in patients that experienced cardiotoxicity. MiR-4732-3p was also present in neonatal rat cardiomyocytes and cardiac fibroblasts and was modulated by anthracycline treatment. A miR-4732-3p mimic was cardioprotective in cardiac and fibroblast cultures, following doxorubicin challenge, in terms of cell viability and ROS levels. Notably, administration of the miR-4732-3p mimic in doxorubicin-treated rats preserved cardiac function, normalized weight loss, induced angiogenesis, and decreased apoptosis, interstitial fibrosis and cardiac myofibroblasts. At the molecular level, miR-4732-3p regulated genes of TGFβ and Hippo signaling pathways. Overall, the results indicate that miR-4732-3p is a novel biomarker of cardiotoxicity that has therapeutic potential against anthracycline-induced heart damage.
Collapse
|
10
|
Sun W, Xu J, Wang L, Jiang Y, Cui J, Su X, Yang F, Tian L, Si Z, Xing Y. Non-coding RNAs in cancer therapy-induced cardiotoxicity: Mechanisms, biomarkers, and treatments. Front Cardiovasc Med 2022; 9:946137. [PMID: 36082126 PMCID: PMC9445363 DOI: 10.3389/fcvm.2022.946137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 07/28/2022] [Indexed: 02/06/2023] Open
Abstract
As a result of ongoing breakthroughs in cancer therapy, cancer patients' survival rates have grown considerably. However, cardiotoxicity has emerged as the most dangerous toxic side effect of cancer treatment, negatively impacting cancer patients' prognosis. In recent years, the link between non-coding RNAs (ncRNAs) and cancer therapy-induced cardiotoxicity has received much attention and investigation. NcRNAs are non-protein-coding RNAs that impact gene expression post-transcriptionally. They include microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs). In several cancer treatments, such as chemotherapy, radiotherapy, and targeted therapy-induced cardiotoxicity, ncRNAs play a significant role in the onset and progression of cardiotoxicity. This review focuses on the mechanisms of ncRNAs in cancer therapy-induced cardiotoxicity, including apoptosis, mitochondrial damage, oxidative stress, DNA damage, inflammation, autophagy, aging, calcium homeostasis, vascular homeostasis, and fibrosis. In addition, this review explores potential ncRNAs-based biomarkers and therapeutic strategies, which may help to convert ncRNAs research into clinical practice in the future for early detection and improvement of cancer therapy-induced cardiotoxicity.
Collapse
Affiliation(s)
- Wanli Sun
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Juping Xu
- The Second People's Hospital of Jiaozuo, Jiaozuo, China
| | - Li Wang
- Department of Breast Surgery, Xingtai People's Hospital, Xingtai, China
| | - Yuchen Jiang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jingrun Cui
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xin Su
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fan Yang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Li Tian
- Beijing University of Chinese Medicine, Beijing, China
| | - Zeyu Si
- The First Clinical Medical College of Shaanxi University of Chinese Medicine, Taiyuan, China
- Zeyu Si
| | - Yanwei Xing
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Yanwei Xing
| |
Collapse
|
11
|
Zhang J, Xie B, Tang Y, Zhou B, Wang Q, Ge Q, Zhou Y, Gu T. Downregulation of miR-34c-5p alleviates chronic intermittent hypoxia-induced myocardial damage by targeting sirtuin 1. J Biochem Mol Toxicol 2022; 36:e23164. [PMID: 35848756 DOI: 10.1002/jbt.23164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 05/28/2022] [Accepted: 07/01/2022] [Indexed: 11/09/2022]
Abstract
Numerous microRNAs (miRs) are abnormally expressed in response to hypoxia-induced myocardial damage. Herein, miR-34c-5p as a potential pharmaco-target was investigated in a mouse model of chronic intermittent hypoxia (CIH)-induced myocardial damage. A mouse model of myocardial damage was established using CIH with 7% or 21% O2 alternating 60 s for 12 h/day, 21% O2 for 12 h/day. AntagomiR-34c-5p (20 nM/0.1 ml; once a week for 12 weeks) was used as a miR-34c-5p inhibitor in a mouse model with tail-vein injection. In another experiment, mice were administrated with Sirt1 activator SRT1720 (50 mg/kg/day) by intraperitoneal injection. Gene Expression Omnibus database showed a significant upregulation of miR-34c-5p expression in the ischemic myocardium of male mice. In CIH-stimulated mice, miR-34c-5p expression was also significantly increased compared with normal mice. Treatment of antagomiR-34c-5p significantly restrained CIH-triggered myocardial apoptosis. After administration of antagomiR-34c-5p or Sirt1 activator SRT1720, cardiac hypertrophy and oxidative stress were attenuated in CIH-stimulated mice. We also found sirtuin 1 (Sirt1) as a direct target of miR-34c-5p, which was able to mediate Sirt1 protein expression in cardiomyocytes. AntagomiR-34c-5p injection markedly elevated Sirt1 protein expression in CIH-stimulated mice. AntagomiR-34c-5p or Sirt1 activator SRT1720 administration exhibited the antioxidative activity and cardioprotective roles in CIH-stimulated mice.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Cardiology, Chengdu First People's Hospital, Chengdu, Sichuan, China
| | - Bo Xie
- Department of Cardiology, Chengdu First People's Hospital, Chengdu, Sichuan, China
| | - Yanrong Tang
- Department of Cardiology, Chengdu First People's Hospital, Chengdu, Sichuan, China
| | - Bo Zhou
- Department of Cardiology, Chengdu First People's Hospital, Chengdu, Sichuan, China
| | - Qiong Wang
- Department of Cardiology, Chengdu First People's Hospital, Chengdu, Sichuan, China
| | - Qing Ge
- Department of Cardiology, Chengdu First People's Hospital, Chengdu, Sichuan, China
| | - Yufei Zhou
- Department of Cardiology, Chengdu First People's Hospital, Chengdu, Sichuan, China
| | - Tongqing Gu
- School of Foreign Languages, Chengdu University of Information Technology, Chengdu, Sichuan, China
| |
Collapse
|
12
|
MiRNAs and circRNAs for the Diagnosis of Anthracycline-Induced Cardiotoxicity in Breast Cancer Patients: A Narrative Review. J Pers Med 2022; 12:jpm12071059. [PMID: 35887556 PMCID: PMC9315470 DOI: 10.3390/jpm12071059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 06/25/2022] [Indexed: 11/28/2022] Open
Abstract
Breast cancer (BC) is the most frequent type of female cancer with increasing incidence in recent years. Doxorubicin (DOX) is an important backbone chemotherapy in BC, responsible for cardiotoxicity (CTX) in about 9% of treated women within the first year. Biomarkers of early CTX diagnosis are essential to avoid complicated DOX-related cardiac diseases. Traditional serum biomarkers are either poorly sensitive with transient elevation, and even absent if investigated outside their diagnostic window, or arise only in late-stage CTX. Emerging biomarkers such as non-coding RNA (ncRNA) have been recently investigated in DOX-related CTX. In our review, we revised the role of microRNAs, the most studied type of ncRNA, both in animal and human models, highlighting the interesting but often contrasting results. Moreover, we reviewed a novel class of ncRNA, circular RNA (circRNA), focusing on their modulatory mechanisms also involving microRNAs. MicroRNA and circRNA are players in a wide homeostatic balance with their perturbation representing a possible compensation for DOX damage. Further studies are required to assess the modalities of early detection of their variation in BC patients suffering from heart disease induced by DOX treatment.
Collapse
|
13
|
Tonry C, Russel-Hallinan A, McCune C, Collier P, Harbinson M, Dixon L, Watson CJ. Circulating biomarkers for management of cancer therapeutics related cardiac dysfunction. Cardiovasc Res 2022; 119:710-728. [PMID: 35640873 PMCID: PMC10153425 DOI: 10.1093/cvr/cvac087] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 05/09/2022] [Accepted: 05/15/2022] [Indexed: 11/14/2022] Open
Abstract
Cancer therapeutics related cardiac dysfunction (CTRCD) has emerged as a major cause of morbidity and mortality in cancer survivors. Effective clinical management of CTRCD is impeded by a lack of sensitive diagnostic and prognostic strategies. Circulating molecular markers could potentially address this need as they are often indicative of cardiac stress before cardiac damage can be detected clinically. A growing understanding of the underlying physiological mechanisms for CTRCD has inspired research efforts to identify novel pathophysiologically-relevant biomarkers that may also guide development of cardio-protective therapeutic approaches. The purpose of this review is to evaluate current circulating biomarkers of cardiac stress and their potential role in diagnosis and management of CTRCD. We also discuss some emerging avenues for CTRCD-focused biomarker investigations.
Collapse
Affiliation(s)
- Claire Tonry
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Northern Ireland, United Kingdom
| | - Adam Russel-Hallinan
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Northern Ireland, United Kingdom
| | - Claire McCune
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Northern Ireland, United Kingdom
| | | | | | | | - Chris J Watson
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Northern Ireland, United Kingdom
| |
Collapse
|
14
|
Pantazi D, Tselepis AD. Cardiovascular toxic effects of antitumor agents: Pathogenetic mechanisms. Thromb Res 2022; 213 Suppl 1:S95-S102. [DOI: 10.1016/j.thromres.2021.12.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/06/2021] [Accepted: 12/16/2021] [Indexed: 02/08/2023]
|
15
|
L'Abbate S, Chianca M, Fabiani I, Del Franco A, Giannoni A, Vergaro G, Grigoratos C, Kusmic C, Passino C, D'Alessandra Y, Burchielli S, Emdin M, Cardinale DM. In Vivo Murine Models of Cardiotoxicity Due to Anticancer Drugs: Challenges and Opportunities for Clinical Translation. J Cardiovasc Transl Res 2022; 15:1143-1162. [PMID: 35312959 DOI: 10.1007/s12265-022-10231-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/04/2022] [Indexed: 12/13/2022]
Abstract
Modern therapeutic approaches have led to an improvement in the chances of surviving a diagnosis of cancer. However, this may come with side effects, with patients experiencing adverse cardiovascular events or exacerbation of underlying cardiovascular disease related to their cancer treatment. Rodent models of chemotherapy-induced cardiotoxicity are useful to define pathophysiological mechanisms of cardiac damage and to identify potential therapeutic targets. The key mechanisms involved in cardiotoxicity induced by specific different antineoplastic agents are summarized in this state-of-the-art review, as well as the rodent models of cardiotoxicity by different classes of anticancer drugs, along with the strategies tested for primary and secondary cardioprotection. Current approaches for early detection of cardiotoxicity in preclinical studies with a focus on the application of advanced imaging modalities and biomarker strategies are also discussed. Potential applications of cardiotoxicity modelling in rodents are illustrated in relation to the advancements of promising research topics of cardiotoxicity. Created with BioRender.com.
Collapse
Affiliation(s)
- Serena L'Abbate
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Michela Chianca
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Iacopo Fabiani
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy.
| | - Annamaria Del Franco
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy.,Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Alberto Giannoni
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy.,Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Giuseppe Vergaro
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy.,Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | | | | | - Claudio Passino
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy.,Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Yuri D'Alessandra
- Cardiovascular Proteomics Unit, Centro Cardiologico Monzino I.R.C.C.S., Milan, Italy
| | | | - Michele Emdin
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy.,Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Daniela Maria Cardinale
- Cardioncology Unit, Cardiology Division, European Institute of Oncology, I.R.C.C.S., Milan, Italy
| |
Collapse
|
16
|
Schofield AL, Brown JP, Brown J, Wilczynska A, Bell C, Glaab WE, Hackl M, Howell L, Lee S, Dear JW, Remes M, Reeves P, Zhang E, Allmer J, Norris A, Falciani F, Takeshita LY, Seyed Forootan S, Sutton R, Park BK, Goldring C. Systems analysis of miRNA biomarkers to inform drug safety. Arch Toxicol 2021; 95:3475-3495. [PMID: 34510227 PMCID: PMC8492583 DOI: 10.1007/s00204-021-03150-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 08/23/2021] [Indexed: 02/06/2023]
Abstract
microRNAs (miRNAs or miRs) are short non-coding RNA molecules which have been shown to be dysregulated and released into the extracellular milieu as a result of many drug and non-drug-induced pathologies in different organ systems. Consequently, circulating miRs have been proposed as useful biomarkers of many disease states, including drug-induced tissue injury. miRs have shown potential to support or even replace the existing traditional biomarkers of drug-induced toxicity in terms of sensitivity and specificity, and there is some evidence for their improved diagnostic and prognostic value. However, several pre-analytical and analytical challenges, mainly associated with assay standardization, require solutions before circulating miRs can be successfully translated into the clinic. This review will consider the value and potential for the use of circulating miRs in drug-safety assessment and describe a systems approach to the analysis of the miRNAome in the discovery setting, as well as highlighting standardization issues that at this stage prevent their clinical use as biomarkers. Highlighting these challenges will hopefully drive future research into finding appropriate solutions, and eventually circulating miRs may be translated to the clinic where their undoubted biomarker potential can be used to benefit patients in rapid, easy to use, point-of-care test systems.
Collapse
Affiliation(s)
- Amy L Schofield
- MRC Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, University of Liverpool, Sherrington Buildings, Ashton Street, Liverpool, L69 3GE, UK
| | - Joseph P Brown
- MRC Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, University of Liverpool, Sherrington Buildings, Ashton Street, Liverpool, L69 3GE, UK
| | - Jack Brown
- MRC Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, University of Liverpool, Sherrington Buildings, Ashton Street, Liverpool, L69 3GE, UK
| | - Ania Wilczynska
- bit.bio, Babraham Research Campus, The Dorothy Hodgkin Building, Cambridge, CB22 3FH, UK
| | - Catherine Bell
- CVRM Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Warren E Glaab
- Merck & Co., Inc, 770 Sumneytown Pike, West Point, PA, 19486, USA
| | | | - Lawrence Howell
- GlaxoSmithKline (GSK), Stevenage, Greater Cambridge Area, UK
| | - Stephen Lee
- ABHI, 1 Duchess St, 4th Floor, Suite 2, London, W1W 6AN, UK
| | - James W Dear
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Mika Remes
- Genomics EMEA, QIAGEN Aarhus, Prismet, Silkeborgvej 2, 8000, Aarhus C, Denmark
| | - Paul Reeves
- Arcis Biotechnology Limited, Suite S07, Techspace One, Sci-tech Daresbury, Keckwick Lane, Daresbury, Warrington, WA4 4AB, UK
| | - Eunice Zhang
- Wolfson Centre for Personalised Medicine, Department of Pharmacology and Therapeutics, University of Liverpool, Crown Street, Liverpool, L69 3BX, UK
| | - Jens Allmer
- Applied Bioinformatics, Bioscience, Wageningen University and Research, Droevendaalsesteeg 4, 6708 PB, Wageningen, The Netherlands
| | - Alan Norris
- MRC Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, University of Liverpool, Sherrington Buildings, Ashton Street, Liverpool, L69 3GE, UK
| | - Francesco Falciani
- Computational Biology Facility, MerseyBio, University of Liverpool, Crown Street, Liverpool, L69 7ZB, UK
| | - Louise Y Takeshita
- Computational Biology Facility, MerseyBio, University of Liverpool, Crown Street, Liverpool, L69 7ZB, UK
| | - Shiva Seyed Forootan
- MRC Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, University of Liverpool, Sherrington Buildings, Ashton Street, Liverpool, L69 3GE, UK
| | - Robert Sutton
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Biosciences Building, Crown Street, Liverpool, L69 7BE, UK
| | - B Kevin Park
- MRC Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, University of Liverpool, Sherrington Buildings, Ashton Street, Liverpool, L69 3GE, UK
| | - Chris Goldring
- MRC Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, University of Liverpool, Sherrington Buildings, Ashton Street, Liverpool, L69 3GE, UK.
| |
Collapse
|
17
|
Rawat PS, Jaiswal A, Khurana A, Bhatti JS, Navik U. Doxorubicin-induced cardiotoxicity: An update on the molecular mechanism and novel therapeutic strategies for effective management. Biomed Pharmacother 2021; 139:111708. [PMID: 34243633 DOI: 10.1016/j.biopha.2021.111708] [Citation(s) in RCA: 427] [Impact Index Per Article: 106.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 12/06/2022] Open
Abstract
Doxorubicin (Dox) is a secondary metabolite of the mutated strain of Streptomyces peucetius var. Caesius and belongs to the anthracyclines family. The anti-cancer activity of Dox is mainly exerted through the DNA intercalation and inhibiting topoisomerase II enzyme in fast-proliferating tumors. However, Dox causes cumulative and dose-dependent cardiotoxicity, which results in increased risks of mortality among cancer patients and thus limiting its wide clinical applications. There are several mechanisms has been proposed for doxorubicin-induced cardiotoxicity and oxidative stress, free radical generation and apoptosis are most widely reported. Apart from this, other mechanisms are also involved in Dox-induced cardiotoxicity such as impaired mitochondrial function, a perturbation in iron regulatory protein, disruption of Ca2+ homeostasis, autophagy, the release of nitric oxide and inflammatory mediators and altered gene and protein expression that involved apoptosis. Dox also causes downregulation of DNA methyltransferase 1 (DNMT1) enzyme activity which leads to a reduction in the DNA methylation process. This hypomethylation causes dysregulation in the mitochondrial genes like peroxisome proliferator-activated receptor-gamma coactivator (PGC)-1-alpha (PGC-1α), nuclear respiratory factor 1 (NRF-1) and mitochondrial transcription factor A (TFAM) unit in the heart. Apart from DNA methylation, Dox treatment also alters the micro RNAs levels and histone deacetylase (HDAC) activity. Therefore, in the current review, we have provided a detailed update on the current understanding of the pathological mechanisms behind the well-known Dox-induced cardiotoxicity. Further, we have provided some of the most plausible pharmacological strategies which have been tested against Dox-induced cardiotoxicity.
Collapse
Affiliation(s)
- Pushkar Singh Rawat
- Department of Pharmacology, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Aiswarya Jaiswal
- Department of Pharmacology, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Amit Khurana
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science, PVNRTVU, Rajendranagar, Hyderabad 500030, Telangana, India; Centre for Biomedical Engineering (CBME), Indian Institute of Technology (IIT), Delhi 110016, India.
| | - Jasvinder Singh Bhatti
- Department of human genetics and molecular medicine, School of health sciences, Central University of Punjab, Bathinda 151401, Punjab, India.
| | - Umashanker Navik
- Department of Pharmacology, Central University of Punjab, Bathinda, Punjab, 151401, India.
| |
Collapse
|
18
|
Chen Y, Xu Y, Deng Z, Wang Y, Zheng Y, Jiang W, Jiang L. MicroRNA expression profiling involved in doxorubicin-induced cardiotoxicity using high-throughput deep-sequencing analysis. Oncol Lett 2021; 22:560. [PMID: 34093775 PMCID: PMC8170198 DOI: 10.3892/ol.2021.12821] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 03/09/2021] [Indexed: 12/31/2022] Open
Abstract
MicroRNAs (miRNAs/miRs) are sensitive biomarkers and endogenous repressors of gene expression by decreasing mRNA stability and interfering with mRNA translation. Despite a number of investigations revealing the dysregulation of miRNA expression associated with cardiotoxicity induced by doxorubicin (Dox), perturbation of miRNAs directly resulting from Dox at early stage in cardiomyocytes and the target gene interaction remain largely unknown. In the present study, high-throughput deep-sequencing was used to analyze changes in global miRNA expression in H9c2 cardiomyocytes exposed to 5 µg/ml Dox for 0, 12 or 24 h. Compared with the 0-h time point, the expression levels of 386 unique miRNAs were altered. Based on miRNA expression and fold-change, the target genes of 76 selected miRNAs were further analyzed using gene interaction networks and pathway enrichment analysis. These miRNAs were involved in the regulation of different pathways, whose functions included apoptosis, cell proliferation, extracellular matrix remodeling, oxidative stress and lipid metabolism. These differentially expressed miRNAs included let-7 family, miR-29b-3p, miR-378-3/5p, miR-351-3p, miR-664-3p, miR-455-3p, miR-298-3p, miR-702-5p, miR-128-1-5p, miR-671 and miR-421-5p. The present data indicated that global wide miRNA profiling in Dox-induced cardiomyocytes may provide a novel mechanistic insight into understanding Dox-induced heart failure and cardiotoxicity, as well as novel biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Ying Chen
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| | - Yingjie Xu
- Department of Cardiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| | - Zhoufeng Deng
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| | - Yin Wang
- Department of Cardiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| | - Ying Zheng
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200336, P.R. China
| | - Weihua Jiang
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| | - Li Jiang
- Department of Cardiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| |
Collapse
|
19
|
Machackova T, Vychytilova-Faltejskova P, Souckova K, Laga R, Androvič L, Mixová G, Slaby O. Barriers in systemic delivery and preclinical testing of synthetic microRNAs in animal models: an experimental study on miR-215-5p mimic. Physiol Res 2021; 70:481-487. [PMID: 33982582 DOI: 10.33549/physiolres.934571] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Mus musculus is the most commonly used animal model in microRNA research; however, little is known about the endogenous miRNome of the animals used in the miRNA-targeting preclinical studies with the human xenografts. In the presented study, we evaluated the NOD/SCID gamma mouse model for the preclinical study of systemic miR-215-5p substitution with a semitelechelic poly[N-(2-hydroxypropyl)-methacrylamide]-based carrier conjugated with miR-215-5p-mimic via a reductively degradable disulfide bond. Murine mmu-miR-215-5p and human hsa-miR-215-5p have a high homology of mature sequences with only one nucleotide substitution. Due to the high homology of hsa-miR-215-5p and mmu-hsa-miR-215-5p, a similar expression in human and NOD/SCID gamma mice was expected. Expression of mmu-miR-215 in murine organs did not indicate tissue-specific expression and was highly expressed in all examined tissues. All animals included in the study showed a significantly higher concentration of miR-215-5p in the blood plasma compared to human blood plasma, where miR-215-5p is on the verge of a reliable detection limit. However, circulating mmu-miR-215-5p did not enter the human xenograft tumors generated with colorectal cancer cell lines since the levels of miR-215-5p in control tumors remained notably lower compared to those originally transfected with miR-215-5p. Finally, the systemic administration of polymer-miR-215-5p-mimic conjugate to the tail vein did not increase miR-215-5p in NOD/SCID gamma mouse blood plasma, organs, and subcutaneous tumors. It was impossible to distinguish hsa-miR-215-5p and mmu-miR-215-5p in the murine blood and organs due to the high expression of endogenous mmu-miR-215-5p. In conclusion, the examination of endogenous tissue and circulating miRNome of an experimental animal model of choice might be necessary for future miRNA studies focused on the systemic delivery of miRNA-based drugs conducted in the animal models.
Collapse
Affiliation(s)
- T Machackova
- Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic.
| | | | | | | | | | | | | |
Collapse
|
20
|
Circulating high-sensitivity troponin T and microRNAs as markers of myocardial damage during childhood leukaemia treatment. Pediatr Res 2021; 89:1245-1252. [PMID: 32634817 DOI: 10.1038/s41390-020-1049-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 05/23/2020] [Accepted: 06/23/2020] [Indexed: 11/08/2022]
Abstract
BACKGROUND We investigated whether plasma high-sensitivity cardiac troponin T (hs-cTnT) and circulating heart-associated microRNA (miRs) are increased in children with leukaemias during anthracycline-based chemotherapeutic treatment. METHODS In vitro human pluripotent stem cell (hPSC)-derived cardiomyocyte model showed that miR-1, miR-133a, miR-208a, miR-208b, and miR-499 are released from cells into culture medium in a time- and dose-dependent manner on doxorubicin exposure. Left ventricular (LV) myocardial deformation and circulating heart-associated miRs and plasma hs-cTnT during and after completion of chemotherapy were determined in 40 children with newly diagnosed acute leukaemia. RESULTS Significant reduction of LV global longitudinal strain and strain rates were found within 1 week after completion of anthracycline therapy in the induction phase of treatment (all p < 0.05). Hs-cTnT level peaked and miR-1 increased significantly at this time point. Log-transformed hs-cTnT correlated negatively with LV global systolic longitudinal strain (r = -0.38, p < 0.001). Receiver operating characteristic analysis revealed that area under the curve for changes in plasma hs-cTnT from baseline and plasma miR-1 levels in detecting a reduction in ≥20% of global longitudinal strain were respectively 0.62 (95% CI 0.38-0.87) and 0.62 (95% CI 0.40-0.84). CONCLUSION Plasma hs-cTnT and circulating miR-1 may be useful markers of myocardial damage during chemotherapy in children with leukaemias. IMPACT Heart-associated miRNAs including miR-1, miR-133a, miR-208a, miR-208b,and miR-499 were increased in the culture medium upon exposure of hPSC-derived cardiomyocytes to doxorubicin. Only miR-1 increased significantly during anthracycline-based therapy in paediatric leukaemic patients. In paediatric leukaemic patients, plasma hs-cTnT and circulating level of miR-1 showed the most significant increase within 1 week after completion of anthracycline therapy in the induction treatment phase. The study provides the first evidence of progressive increase in circulating miR-1 and plasma hs-cTnT levels during the course of anthracycline-based therapy in children with leukaemias, with hs-cTnT level also associated with changes in LV myocardial deformation.
Collapse
|
21
|
Fa HG, Chang WG, Zhang XJ, Xiao DD, Wang JX. Noncoding RNAs in doxorubicin-induced cardiotoxicity and their potential as biomarkers and therapeutic targets. Acta Pharmacol Sin 2021; 42:499-507. [PMID: 32694762 PMCID: PMC8114921 DOI: 10.1038/s41401-020-0471-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 07/03/2020] [Indexed: 02/07/2023]
Abstract
Anthracyclines, such as doxorubicin (DOX), are well known for their high efficacy in treating multiple cancers, but their clinical usage is limited due to their potential to induce fatal cardiotoxicity. Such detrimental effects significantly impact the overall physical condition or even induce the morbidity and mortality of cancer survivors. Therefore, it is extremely important to understand the mechanisms of DOX-induced cardiotoxicity to develop methods for the early detection of cytotoxicity and therapeutic applications. Studies have shown that many molecular events are involved in DOX-induced cardiotoxicity. However, the precise mechanisms are still not completely understood. Recently, noncoding RNAs (ncRNAs) have been extensively studied in a diverse range of regulatory roles in cellular physiological and pathological processes. With respect to their roles in DOX-induced cardiotoxicity, microRNAs (miRNAs) are the most widely studied, and studies have focused on the regulatory roles of long noncoding RNAs (lncRNAs) and circular RNAs (circRNAs), which have been shown to have significant functions in the cardiovascular system. Recent discoveries on the roles of ncRNAs in DOX-induced cardiotoxicity have prompted extensive interest in exploring candidate ncRNAs for utilization as potential therapeutic targets and/or diagnostic biomarkers. This review presents the frontier studies on the roles of ncRNAs in DOX-induced cardiotoxicity, addresses the possibility and prospects of using ncRNAs as diagnostic biomarkers or therapeutic targets, and discusses the possible reasons for related discrepancies and limitations of their use.
Collapse
|
22
|
Tantawy M, Pamittan FG, Singh S, Gong Y. Epigenetic Changes Associated With Anthracycline-Induced Cardiotoxicity. Clin Transl Sci 2021; 14:36-46. [PMID: 32770710 PMCID: PMC7877852 DOI: 10.1111/cts.12857] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 07/11/2020] [Indexed: 12/24/2022] Open
Abstract
Advances in cancer treatment have significantly improved the survival of patients with cancer, but, unfortunately, many of these treatments also have long-term complications. Cancer treatment-related cardiotoxicities are becoming a significant clinical problem that a new discipline, Cardio-Oncology, was established to advance the cardiovascular care of patients with growing cancer populations. Anthracyclines are a class of chemotherapeutic agents used to treat many cancers in adults and children. Their clinical use is limited by anthracycline-induced cardiotoxicity (AIC), which can lead to heart failure. Early-onset cardiotoxicity appears within a year of treatment, whereas late-onset cardiotoxicity occurs > 1 year and even up to decades after treatment completion. The pathophysiology of AIC was hypothesized to be caused by generation of reactive oxygen species that lead to lipid peroxidation, defective mitochondrial biogenesis, and DNA damage of the cardiomyocytes. The accumulation of anthracycline metabolites was also proposed to cause mitochondrial damage and the induction of cardiac cell apoptosis, which induces arrhythmias, contractile dysfunction, and cardiomyocyte death. This paper will provide a general overview of cardiotoxicity focusing on the effect of anthracyclines and their epigenetic molecular mechanisms on cardiotoxicity.
Collapse
Affiliation(s)
- Marwa Tantawy
- Department of Pharmacotherapy and Translational ResearchCollege of PharmacyUniversity of FloridaGainesvilleFloridaUSA
- Center for Pharmacogenomics and Precision MedicineCollege of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Frances G. Pamittan
- Department of Pharmacotherapy and Translational ResearchCollege of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | | | - Yan Gong
- Department of Pharmacotherapy and Translational ResearchCollege of PharmacyUniversity of FloridaGainesvilleFloridaUSA
- Center for Pharmacogenomics and Precision MedicineCollege of PharmacyUniversity of FloridaGainesvilleFloridaUSA
- UF Health Cancer CenterGainesvilleFloridaUSA
| |
Collapse
|
23
|
The role of metabolic diseases in cardiotoxicity associated with cancer therapy: What we know, what we would know. Life Sci 2020; 255:117843. [PMID: 32464123 DOI: 10.1016/j.lfs.2020.117843] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/16/2020] [Accepted: 05/22/2020] [Indexed: 12/12/2022]
Abstract
Metabolic diseases, such as obesity and type 2 diabetes, are known risk factors for cardiovascular (CV) diseases. Thus, patients with those comorbidities could be at increased risk of experiencing cardiotoxicity related to treatment with Anthracyclines and the other new generation targeted anticancer drugs. However, investigations addressing the mechanisms underlying the development of CV complications and poor outcome in such cohort of patients are still few and controversial. Given the importance of a personalized approach against chemotherapy-induced cardiomyopathy, this review summarizes our current knowledge on the pathophysiology of chemotherapy-induced cardiomyopathy and its association with obesity and type 2 diabetes. Along with clinical evidences, future perspectives of preclinical research around this field and its role in addressing important open questions, including the development of more proactive strategies for prevention, and treatment of cardiotoxicity during and after chemotherapy in the presence of metabolic diseases, is also presented.
Collapse
|
24
|
Sala V, Della Sala A, Hirsch E, Ghigo A. Signaling Pathways Underlying Anthracycline Cardiotoxicity. Antioxid Redox Signal 2020; 32:1098-1114. [PMID: 31989842 DOI: 10.1089/ars.2020.8019] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Significance: The cardiac side effects of hematological treatments are a major issue of the growing population of cancer survivors, often affecting patient survival even more than the tumor for which the treatment was initially prescribed. Among the most cardiotoxic drugs are anthracyclines (ANTs), highly potent antitumor agents, which still represent a mainstay in the treatment of hematological and solid tumors. Unfortunately, diagnosis, prevention, and treatment of cardiotoxicity are still unmet clinical needs, which call for a better understanding of the molecular mechanism behind the pathology. Recent Advances: This review article will discuss recent findings on the pathomechanisms underlying the cardiotoxicity of ANTs, spanning from DNA and mitochondrial damage to calcium homeostasis, autophagy, and apoptosis. Special emphasis will be given to the role of reactive oxygen species and their interplay with major signaling pathways. Critical Issues: Although new promising therapeutic targets and new drugs have started to be identified, their efficacy has been mainly proven in preclinical studies and requires clinical validation. Future Directions: Future studies are awaited to confirm the relevance of recently uncovered targets, as well as to identify new druggable pathways, in more clinically relevant models, including, for example, human induced pluripotent stem cell-derived cardiomyocytes.
Collapse
Affiliation(s)
- Valentina Sala
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Angela Della Sala
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| |
Collapse
|
25
|
Balasubramanian S, Gunasekaran K, Sasidharan S, Jeyamanickavel Mathan V, Perumal E. MicroRNAs and Xenobiotic Toxicity: An Overview. Toxicol Rep 2020; 7:583-595. [PMID: 32426239 PMCID: PMC7225592 DOI: 10.1016/j.toxrep.2020.04.010] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/13/2020] [Accepted: 04/19/2020] [Indexed: 12/27/2022] Open
Abstract
The advent of new technologies has paved the rise of various chemicals that are being employed in industrial as well as consumer products. This leads to the accumulation of these xenobiotic compounds in the environment where they pose a serious threat to both target and non-target species. miRNAs are one of the key epigenetic mechanisms that have been associated with toxicity by modulating the gene expression post-transcriptionally. Here, we provide a comprehensive view on miRNA biogenesis, their mechanism of action and, their possible role in xenobiotic toxicity. Further, we review the recent in vitro and in vivo studies involved in xenobiotic exposure induced miRNA alterations and the mRNA-miRNA interactions. Finally, we address the challenges associated with the miRNAs in toxicological studies.
Collapse
Key Words
- ADAMTS9, A disintegrin and metalloproteinase with thrombospondin motifs 9
- AHR, Aryl Hydrocarbon Receptor
- AMPK, Adenosine Monophosphate-activated protein kinase
- ARRB1, Arrestin beta 1
- Ag, Silver
- Al2O3, Aluminium oxide
- Au, Gold
- Aβ, Amyloid Beta
- BCB, Blood-cerebrospinal fluid barrier
- BNIP3−3, BCL2/adenovirus E1B 19 kDa protein-interacting protein 3
- BaP, Benzo[a]pyrene
- Biomarkers
- CCNB1, Cyclin B1
- CDC25A, M-phase inducer phosphatase 1
- CDC25C, M-phase inducer phosphatase 3
- CDK, Cyclin-dependent Kinase
- CDK1, Cyclin-dependent kinase 1
- CDK6, Cyclin-dependent kinase 6
- CDKN1b, Cyclin-dependent kinase Inhibitor 1B
- CEC, Contaminants of Emerging Concern
- COPD, Chronic obstructive pulmonary disease
- COX2, Cyclooxygenase-2
- CTGF, Connective Tissue Growth Factor
- DGCR8, DiGeorge syndrome chromosomal [or critical] region 8
- DNA, Deoxy ribonucleic acid
- DON, Deoxynivalenol
- ER, Endoplasmic Reticulum
- Environment
- Epigenetics
- Fadd, Fas-associated protein with death domain
- GTP, Guanosine triphosphate
- Gene regulation
- Grp78/BIP, Binding immunoglobulin protein
- HSPA1A, Heat shock 70 kDa protein 1
- Hpf, Hours post fertilization
- IL-6, Interleukin 6
- IL1R1, Interleukin 1 receptor, type 1
- LIN28B, Lin-28 homolog B
- LRP-1-, Low density lipoprotein receptor-related protein 1
- MAPK, Mitogen Activated Protein Kinase
- MC-LR, Microcystin-Leucine Arginine
- MC-RR, Microcystin-Arginine Arginine
- MRE, MicroRNA Response Elements
- Mn, Manganese
- NASH, Non-alcoholic steatohepatitis
- NET1, Neuroepithelial Cell Transforming 1
- NF- ҡB, Nuclear Factor kappa-light-chain-enhancer of activated B cells
- NFKBAP, NFKB Activating protein-1
- NMDAR, N-methyl-d-aspartate receptor
- NPs, Nanoparticles
- Non-coding RNAs
- Nrf2, Nuclear factor erythroid 2-related factor 2
- PDCD4, Programmed cell death protein 4
- PFAS, Poly-fluoroalkyl substances
- PM2.5, Particulate Matter2.5
- RISC, RNA-induced silencing complex
- RNA, Ribonucleic acid
- RNAi, RNA interference
- RNase III, Ribonuclease III
- SEMA6D, Semaphorin-6D
- SOLiD, Sequencing by Oligonucleotide Ligation and Detection
- SPIONs, Superparamagnetic Iron Oxide Nanoparticles
- SiO2, Silicon dioxide
- TCDD, 2,3,7,8-Tetrachlorodibenzodioxin
- TNF-α, Tumor necrosis factor – alpha
- TP53, Tumor protein 53
- TRBP, Transactivation Response RNA Binding Protein
- Toxicity
- UTR, Untranslated region
- WHO, World Health Organization
- Wnt, Wingless-related integration site
- ZEA, Zearalanone
- Zn, Zinc
- bcl2l11, B-cell lymphoma-2-like protein 11
- ceRNA, Competing endogenous RNA
- lncRNAs, Long non-coding RNA
- mRNA, Messenger RNA
- miRNA, MicroRNA
- qRT-PCR, quantitative Real Time-Polymerase Chain Reaction
- ripk 1, Receptor-interacting serine/threonine-protein kinase 1
Collapse
Affiliation(s)
| | - Kanmani Gunasekaran
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, 641 046, India
| | - Saranyadevi Sasidharan
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, 641 046, India
| | | | - Ekambaram Perumal
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, 641 046, India
| |
Collapse
|
26
|
Chiba K, Ishizaka T, Yoshimatsu Y, Mikamoto K, Maeda Y, Iguchi T, Shirai M, Yamaguchi T, Goto K, Sakurai K, Tamai S, Kataoka H, Hasegawa M, Mori K. Comprehensive analysis of cardiac function, blood biomarkers and histopathology for milrinone-induced cardiotoxicity in cynomolgus monkeys. J Pharmacol Toxicol Methods 2020; 103:106870. [PMID: 32353509 DOI: 10.1016/j.vascn.2020.106870] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 04/13/2020] [Accepted: 04/22/2020] [Indexed: 01/08/2023]
Abstract
The objective of this study was to elucidate the underlying cardiotoxic mechanism of milrinone, a cAMP phosphodiesterase 3 inhibitor, by evaluating cardiac functions, blood biomarkers including cardiac troponin I (cTnI), microRNAs (miR-1, miR-133a and miR-499a) and various endogenous metabolites, and histopathology in conscious cynomolgus monkeys. Milrinone at doses of 0, 3 and 30 mg/kg were orally administered to monkeys (n = 3-4/group), and the endpoints were evaluated 1 to 24 h post-dosing. Milrinone caused myocardial injuries characterized by myocardial degeneration/necrosis, cell infiltration and hemorrhage 24 h after drug administration. Cardiac functional analysis revealed that milrinone dose-dependently increased the maximum upstroke velocity of the left ventricular pressure and heart rate, and decreased the QA interval and systemic blood pressure 1-4 h post-dosing, being associated with pharmacological action of the drug. In the blood biomarker analysis, only plasma cTnI was dose-dependently increased 4-7 h after drug administration, suggesting that cTnI is the most sensitive biomarker for early detection of milrinone-induced myocardial injuries. In the metabolomics analysis, high dose of milrinone induced transient changes in lipid metabolism, amino acid utilization and oxidative stress, together with the pharmacological action of increased cAMP and lipolysis 1 h post-dosing before the myocardial injuries were manifested by increased cTnI levels. Taken together, milrinone showed acute positive inotropic and multiple metabolic changes including excessive pharmacological actions, resulting in myocardial injuries. Furthermore, a comprehensive analysis of cardiac functions, blood biomarkers and histopathology can provide more appropriate information for overall assessment of preclinical cardiovascular safety.
Collapse
Affiliation(s)
- Katsuyoshi Chiba
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan.
| | - Tomomichi Ishizaka
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Yu Yoshimatsu
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Kei Mikamoto
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Yu Maeda
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Takuma Iguchi
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Makoto Shirai
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Takashi Yamaguchi
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Koichi Goto
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Ken Sakurai
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Satoshi Tamai
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Hiroko Kataoka
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Miki Hasegawa
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Kazuhiko Mori
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 1-16-13 Kita-Kasai, Edogawa-ku, Tokyo 134-8630, Japan
| |
Collapse
|
27
|
Feng Y, Chen X, Ding W, Ma J, Zhang B, Li X. MicroRNA-16 participates in the cell cycle alteration of HepG2 cells induced by MC-LR. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 192:110295. [PMID: 32066005 DOI: 10.1016/j.ecoenv.2020.110295] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/30/2020] [Accepted: 02/01/2020] [Indexed: 05/26/2023]
Abstract
Microcystin-LR (MC-LR) is a cyclic hepatotoxin produced by cyanobacteria in freshwater, and chronic MC-LR exposure could induce human hepatitis if consumed in drinking water. In recent years, many studies have indicated that microRNAs participate in the hepatotoxicity of MC-LR. The purpose of this study was to investigate the potential function of miR-16 in the hepatocellular toxicity and cell cycle alteration induced by MC-LR in human hepatocellular carcinoma (HepG2) cells after treatment with 10 μM MC-LR. The result of flow cytometry detection showed that a low concentration of MC-LR (10 μM) failed to induce apoptosis but promoted cell cycle G1/S transition in HepG2 cells. In addition, the expression of apoptosis-related genes was suppressed after MC-LR exposure. These results confirm that MC-LR exposure at a low dose can promote the proliferation of HepG2 cells. Furthermore, we also found that microRNA-16 (miR-16) expression was suppressed in HepG2 cells following MC-LR exposure. Hence, we overexpressed miR-16 in HepG2 cells and treated them with MC-LR, and the results showed that miR-16 overexpression induced an increase in the G0/G1 phase and a decrease in the S phase cell cycle populations in HepG2 cells, suggesting that miR-16 can inhibit the cell proliferation of HepG2 cells. In conclusion, our results suggest that miR-16 may play a vital role in the cell cycle alteration of HepG2 cells after MC-LR exposure.
Collapse
Affiliation(s)
- Yiyi Feng
- College of Life Science, Henan Normal University, Xinxiang, Henan, 453007, China
| | - Xi Chen
- College of Life Science, Henan Normal University, Xinxiang, Henan, 453007, China
| | - Weikai Ding
- College of Life Science, Henan Normal University, Xinxiang, Henan, 453007, China
| | - Junguo Ma
- College of Life Science, Henan Normal University, Xinxiang, Henan, 453007, China
| | - Bangjun Zhang
- College of Life Science, Henan Normal University, Xinxiang, Henan, 453007, China
| | - Xiaoyu Li
- College of Life Science, Henan Normal University, Xinxiang, Henan, 453007, China.
| |
Collapse
|
28
|
MicroRNAs in Cancer Treatment-Induced Cardiotoxicity. Cancers (Basel) 2020; 12:cancers12030704. [PMID: 32192047 PMCID: PMC7140035 DOI: 10.3390/cancers12030704] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/12/2020] [Accepted: 03/13/2020] [Indexed: 12/20/2022] Open
Abstract
Cancer treatment has made significant progress in the cure of different types of tumors. Nevertheless, its clinical use is limited by unwanted cardiotoxicity. Aside from the conventional chemotherapy approaches, even the most newly developed, i.e., molecularly targeted therapy and immunotherapy, exhibit a similar frequency and severity of toxicities that range from subclinical ventricular dysfunction to severe cardiomyopathy and, ultimately, congestive heart failure. Specific mechanisms leading to cardiotoxicity still remain to be elucidated. For instance, oxidative stress and DNA damage are considered key players in mediating cardiotoxicity in different treatments. microRNAs (miRNAs) act as key regulators in cell proliferation, cell death, apoptosis, and cell differentiation. Their dysregulation has been associated with adverse cardiac remodeling and toxicity. This review provides an overview of the cardiotoxicity induced by different oncologic treatments and potential miRNAs involved in this effect that could be used as possible therapeutic targets.
Collapse
|
29
|
Blockade of L-type Ca 2+ channel attenuates doxorubicin-induced cardiomyopathy via suppression of CaMKII-NF-κB pathway. Sci Rep 2019; 9:9850. [PMID: 31285514 PMCID: PMC6614470 DOI: 10.1038/s41598-019-46367-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 06/27/2019] [Indexed: 12/30/2022] Open
Abstract
Ca2+/calmodulin-dependent protein kinase II (CaMKII) and nuclear factor-kappa B (NF-κB) play crucial roles in pathogenesis of doxorubicin (DOX)-induced cardiomyopathy. Their activities are regulated by intracellular Ca2+. We hypothesized that blockade of L-type Ca2+ channel (LTCC) could attenuate DOX-induced cardiomyopathy by regulating CaMKII and NF-κB. DOX activated CaMKII and NF-κB through their phosphorylation and increased cleaved caspase 3 in cardiomyocytes. Pharmacological blockade or gene knockdown of LTCC by nifedipine or small interfering RNA, respectively, suppressed DOX-induced phosphorylation of CaMKII and NF-κB and apoptosis in cardiomyocytes, accompanied by decreasing intracellular Ca2+ concentration. Autocamtide 2-related inhibitory peptide (AIP), a selective CaMKII inhibitor, inhibited DOX-induced phosphorylation of NF-κB and cardiomyocyte apoptosis. Inhibition of NF-κB activity by ammonium pyrrolidinedithiocarbamate (PDTC) suppressed DOX-induced cardiomyocyte apoptosis. DOX-treatment (18 mg/kg via intravenous 3 injections over 1 week) increased phosphorylation of CaMKII and NF-κB in mouse hearts. Nifedipine (10 mg/kg/day) significantly suppressed DOX-induced phosphorylation of CaMKII and NF-κB and cardiomyocyte injury and apoptosis in mouse hearts. Moreover, it attenuated DOX-induced left ventricular dysfunction and dilatation. Our findings suggest that blockade of LTCC attenuates DOX-induced cardiomyocyte apoptosis via suppressing intracellular Ca2+ elevation and activation of CaMKII-NF-κB pathway. LTCC blockers might be potential therapeutic agents against DOX-induced cardiomyopathy.
Collapse
|
30
|
Hanousková B, Skála M, Brynychová V, Zárybnický T, Skarková V, Kazimírová P, Vernerová A, Souček P, Skálová L, Pudil R, Matoušková P. Imatinib-induced changes in the expression profile of microRNA in the plasma and heart of mice-A comparison with doxorubicin. Biomed Pharmacother 2019; 115:108883. [PMID: 31004989 DOI: 10.1016/j.biopha.2019.108883] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 03/13/2019] [Accepted: 04/11/2019] [Indexed: 10/27/2022] Open
Abstract
Cardiotoxicity is a serious adverse reaction to cancer chemotherapy and may lead to critical heart damage. Imatinib mesylate (IMB), a selective tyrosine kinase inhibitor, is sometimes accompanied by severe cardiovascular complications. To minimize risk, early biomarkers of such complications are of utmost importance. At the present time, microRNAs (miRNAs) are intensively studied as potential biomarkers of many pathological processes. Many miRNAs appear to be specific in some tissues, including the heart. In the present study we have explored the potential of specific miRNAs to be early markers of IMB-induced cardiotoxicity. Doxorubicin (DOX), an anthracycline with well-known cardiotoxicity, was used for comparison. NMRI mice were treated with IMB or DOX for nine days in doses corresponding to the highest recommended doses in oncological patients, following which plasmatic levels of miRNAs were analyzed in miRNA microarrays and selected cardio-specific miRNAs were quantified using qPCR. The plasmatic level of miR-1a, miR-133a, miR-133b, miR-339, miR-7058, miR-6236 and miR-6240 were the most different between the IMB-treated and control mice. Interestingly, most of the miRNAs affected by DOX were also affected by IMB showing the same trends. Concerning selected microRNAs in the hearts of individual mice, only miR-34a was significantly increased after DOX treatment, and only miR-205 was significantly decreased after IMB and DOX treatment. However, no changes in any miRNA expression correlated with the level of troponin T, a classical marker of heart injury.
Collapse
Affiliation(s)
- Barbora Hanousková
- Faculty of Pharmacy, Charles University, Heyrovského 1203, Hradec Králové, Czech Republic
| | - Mikuláš Skála
- Faculty of Medicine in Hradec Králové, Charles University, Šimkova 870, Hradec Králové, Czech Republic; Department of Pulmology, University Hospital in Hradec Králové, Hradec Králové, Czech Republic
| | - Veronika Brynychová
- The National Institute of Public Health, Šrobárova 48, Praha 10, Czech Republic; Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655/76, Plzeň, Czech Republic
| | - Tomáš Zárybnický
- Faculty of Pharmacy, Charles University, Heyrovského 1203, Hradec Králové, Czech Republic
| | - Veronika Skarková
- Faculty of Medicine in Hradec Králové, Charles University, Šimkova 870, Hradec Králové, Czech Republic
| | - Petra Kazimírová
- Faculty of Medicine in Hradec Králové, Charles University, Šimkova 870, Hradec Králové, Czech Republic
| | - Andrea Vernerová
- Faculty of Pharmacy, Charles University, Heyrovského 1203, Hradec Králové, Czech Republic
| | - Pavel Souček
- The National Institute of Public Health, Šrobárova 48, Praha 10, Czech Republic; Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655/76, Plzeň, Czech Republic
| | - Lenka Skálová
- Faculty of Pharmacy, Charles University, Heyrovského 1203, Hradec Králové, Czech Republic
| | - Radek Pudil
- Faculty of Medicine in Hradec Králové, Charles University, Šimkova 870, Hradec Králové, Czech Republic
| | - Petra Matoušková
- Faculty of Pharmacy, Charles University, Heyrovského 1203, Hradec Králové, Czech Republic.
| |
Collapse
|
31
|
Abstract
Cardiotoxicity is a well-known side effect of doxorubicin (DOX), but the mechanisms leading to this phenomenon are still not completely clear. Prediction of drug-induced dysfunction onset is difficult and is still largely based on detection of cardiac troponin (cTn), a circulating marker of heart damage. In the last years, several investigations focused on the possible involvement of microRNAs (miRNAs) in DOX-induced toxicity in vitro, with contrasting results. Recently, several groups employed animal models to mimic patient’s condition, investigate the biological pathways perturbed by DOX, and identify diagnostic markers of cardiotoxicity. We reviewed the results from several studies investigating cardiac miRNAs expression in rodent models of DOX-treatment. We also discussed the data from two publications indicating the possible use of circulating miRNA as biomarkers of DOX-induced cardiotoxicity. Unfortunately, limited information was derived from these studies, as selection methods of candidate-miRNAs and heterogeneity in cardiotoxicity assessment greatly hampered the novelty and robustness of the findings. Nevertheless, at least one circulating miRNA, miR-1, showed a good potential as early biomarker of drug-mediated cardiac dysfunction onset. The use of animal models to investigate DOX-induced cardiotoxicity surely helps narrowing the gap between basic research and clinical practice. Despite this, several issues, including selection of relevant miRNAs and less-than-optimal assessment of cardiotoxicity, greatly limited the results obtained so far. Nonetheless, the association of patients-based studies with the use of preclinical models may be the key to address the many unanswered questions regarding the pathophysiology and early detection of cardiotoxicity.
Collapse
|
32
|
Ruggeri C, Gioffré S, Chiesa M, Buzzetti M, Milano G, Scopece A, Castiglioni L, Pontremoli M, Sironi L, Pompilio G, Colombo GI, D'Alessandra Y. A Specific Circulating MicroRNA Cluster Is Associated to Late Differential Cardiac Response to Doxorubicin-Induced Cardiotoxicity In Vivo. DISEASE MARKERS 2018; 2018:8395651. [PMID: 30627229 PMCID: PMC6304816 DOI: 10.1155/2018/8395651] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 09/25/2018] [Accepted: 10/03/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Cardiotoxicity is a detrimental side effect of the anticancer drug doxorubicin (DOX), characterized by progressive heart dysfunction. Circulating microRNAs (miRNAs) are recognized as potential biomarkers of cardiac disease; thus, we aimed to investigate their association with late cardiotoxicity in an animal model of disease. METHODS Twenty C57BL/6 female mice were administered with 24 mg/kg cumulative dose of DOX or saline during 2 weeks, followed by a recovery period of one month (T42). Echocardiography was performed at baseline and at T42, and plasma samples were collected at T42. The selection of all miRNAs of interest was conducted by literature overview and by screening, followed by RT-qPCR validation. Results. The analysis of cardiac function at T42 evidenced five DOX-treated animals indistinguishable (NoTox) from controls (CTRLs), while four presented heart impairment (Tox). Our analyses identified eight dysfunction-associated plasma miRNAs. In particular, seven miRNAs were found downregulated in comparison to CTRLs, miR-1-3p, miR-122-5p, miR-127-3p, miR-133a-3p, miR-215-5p, miR-455-3-p, and miR-499a-5p. Conversely, miR-34a-5p showed increased levels in Tox plasma samples. Noteworthy, we determined a cluster composed of miR-1-3p, miR-34a-5p, miR-133a-3p, and miR-499a-5p that distinguished with high-accuracy Tox from NoTox mice. CONCLUSION This is the first study indicating that, similarly to what is observed in patients, DOX-administered animals present a differential cardiac response to treatment. Moreover, our results indicate the presence of specific plasma miRNAs whose expression reflect the presence of cardiac dysfunction in response to drug-induced injury.
Collapse
Affiliation(s)
- Clarissa Ruggeri
- Immunology and Functional Genomics Unit, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Sonia Gioffré
- Immunology and Functional Genomics Unit, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Mattia Chiesa
- Immunology and Functional Genomics Unit, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Marta Buzzetti
- Immunology and Functional Genomics Unit, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Giuseppina Milano
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino IRCCS, Milan, Italy
- Laboratory of Cardiovascular Research, Department of Surgery and Anesthesiology, University Hospital of Lausanne, Lausanne, Switzerland
| | - Alessandro Scopece
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Laura Castiglioni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Marta Pontremoli
- Immunology and Functional Genomics Unit, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Luigi Sironi
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
- Unit of Cardio- and Cerebrovascular Research: Experimental Models and In Vivo Imaging, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Giulio Pompilio
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Gualtiero I. Colombo
- Immunology and Functional Genomics Unit, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Yuri D'Alessandra
- Immunology and Functional Genomics Unit, Centro Cardiologico Monzino IRCCS, Milan, Italy
| |
Collapse
|
33
|
MicroRNAs in the diagnosis and prevention of drug-induced cardiotoxicity. Arch Toxicol 2018; 93:1-9. [DOI: 10.1007/s00204-018-2356-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 11/14/2018] [Indexed: 01/12/2023]
|
34
|
Abstract
Recent advances in cancer prevention and management have led to an exponential increase of cancer survivors worldwide. Regrettably, cardiovascular disease has risen in the aftermath as one of the most devastating consequences of cancer therapies. In this work, we define cancer therapeutics-induced cardiotoxicity as the direct or indirect cardiovascular injury or injurious effect caused by cancer therapies. We describe four progressive stages of this condition and four corresponding levels of prevention, each having a specific goal, focus, and means of action. We subsequently unfold this didactic framework, surveying mechanisms of cardiotoxicity, risk factors, cardioprotectants, biomarkers, and diagnostic imaging modalities. Finally, we outline the most current evidence-based recommendations in this area according to multidisciplinary expert consensus guidelines.
Collapse
Affiliation(s)
- J. Emanuel Finet
- Section of Heart Failure and Transplantation Medicine, Robert and Suzanne Tomsich Department of Cardiovascular Medicine, and Sydell and Arnold Miller Family Heart and Vascular Institute, Cleveland Clinic, Cleveland, USA
| | - W. H. Wilson Tang
- Section of Heart Failure and Transplantation Medicine, Robert and Suzanne Tomsich Department of Cardiovascular Medicine, and Sydell and Arnold Miller Family Heart and Vascular Institute, Cleveland Clinic, Cleveland, USA
- Cleveland Clinic Lerner College of Medicine at Case Western Reserve University; Center for Clinical Genomics; Cleveland Clinic, Cleveland, USA
| |
Collapse
|
35
|
The role of cardiac biomarkers in cardio-oncology. Curr Probl Cancer 2018; 42:375-385. [PMID: 30126650 DOI: 10.1016/j.currproblcancer.2018.06.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 06/30/2018] [Indexed: 11/24/2022]
Abstract
Major advances in cancer therapy have resulted in an unprecedented improvement in patient survival for many cancers. Partially as a consequence of increased longevity, it has become evident that the cardiovascular system is frequently impacted by chemotherapy, radiation, and particularly certain targeted therapy. Cardiotoxicity continues to be a dose-limiting side effect of many chemotherapeutic agents including the anthracyclines. Early identification of patients at increased risk of cardiotoxicity or detecting cardiac injury at the earliest point will allow for initiation of cardio-protective strategies and hopefully prevent or reduce the need to modify life-saving cancer therapies. Biomarkers offer a potential solution to this clinical challenge because they may assist us in early identification of subclinical cardiac disease. In this review, we will summarize studies conducted evaluating the use of cardiac troponins (TnI, TnT) and natriuretic peptides, BNP and NT-proBNP. Other biomarkers for cardiotoxicity that have been at least preliminarily investigated, including myeloperoxidase and microRNAs, will be discussed as an exploratory tool for evidence of cardiac dysfunction during cancer treatment.
Collapse
|
36
|
Dai Y, Lu H, Wang S, Chang S, Li C, Huang Z, Zhang F, Yang H, Shen Y, Chen Z, Qian J, Ge J. MicroRNA-216b actively modulates diabetic angiopathy through inverse regulation on FZD5. Gene 2018; 658:129-135. [PMID: 29477872 DOI: 10.1016/j.gene.2018.02.050] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Revised: 02/14/2018] [Accepted: 02/21/2018] [Indexed: 12/01/2022]
Abstract
BACKGROUND In this work, we examined the angiogenic function of microRNA-216b in an in vitro rat diabetic model of myocardial microvascular endothelial cells (MMECs). METHODS MMECs were extracted from Wistar rats (MMEC(WI)) or diabetic Goto-Kakizaki (GK) rats (MMEC(GK)) and cultured in vitro. QRT-PCR was applied to compare miR-216b between MMEC(WI) and MMEC(GK). MiR-216b was downregulated in MMEC(GK). Its effects on angiogenic development, including invasion and proliferation, were evaluated. In MMEC(GK), putative miR-216b downstream target gene, frizzled class receptor 5 (FZD5), was evaluated by dual-luciferase reporter, qRT-PCR and western blot assays, respectively. FZD5 was further downregulated in MMEC(GK) with stable miR-216b downregulation to evaluate its functional role in regulating diabetic angiogenesis. RESULTS MiR-216b was markedly overexpressed in MMEC(GK). MiR-216b downregulation significantly enhanced angiogenesis in MMEC(GK) by promoting invasion and proliferation. FZD5 was inversely upregulated in miR-216b-downregulated MMEC(GK). Subsequently, FZD5 downregulation suppressed angiogenic development, by inhibiting invasion and proliferation in miR-216b-downregulated MMEC(GK). CONCLUSION MicroRNA-216b was overexposed in diabetic MMECs and its downregulation may actively enhance angiogenesis in diabetic angiopathy through inverse regulation on FZD5.
Collapse
Affiliation(s)
- Yuxiang Dai
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Hao Lu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Shen Wang
- Department of Cardiology, Xinhua Hospital of Zhejiang Province, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310003, China
| | - Shufu Chang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Chenguang Li
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zheyong Huang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Feng Zhang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Hongbo Yang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yi Shen
- Department of Geratology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zhangwei Chen
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Juying Qian
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Junbo Ge
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
37
|
Circulating miR-1 as a potential biomarker of doxorubicin-induced cardiotoxicity in breast cancer patients. Oncotarget 2018; 8:6994-7002. [PMID: 28052002 PMCID: PMC5351685 DOI: 10.18632/oncotarget.14355] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 12/07/2016] [Indexed: 01/01/2023] Open
Abstract
Cardiotoxicity is associated with the chronic use of doxorubicin leading to cardiomyopathy and heart failure. Identification of cardiotoxicity-specific miRNA biomarkers could provide clinicians with a valuable prognostic tool. The aim of the study was to evaluate circulating levels of miRNAs in breast cancer patients receiving doxorubicin treatment and to correlate with cardiac function. This is an ancillary study from “Carvedilol Effect on Chemotherapy-induced Cardiotoxicity” (CECCY trial), which included 56 female patients (49.9±3.3 years of age) from the placebo arm. Enrolled patients were treated with doxorubicin followed by taxanes. cTnI, LVEF, and miRNAs were measured periodically. Circulating levels of miR-1, -133b, -146a, and -423-5p increased during the treatment whereas miR-208a and -208b were undetectable. cTnI increased from 6.6±0.3 to 46.7±5.5 pg/mL (p<0.001), while overall LVEF tended to decrease from 65.3±0.5 to 63.8±0.9 (p=0.053) over 12 months. Ten patients (17.9%) developed cardiotoxicity showing a decrease in LVEF from 67.2±1.0 to 58.8±2.7 (p=0.005). miR-1 was associated with changes in LVEF (r=-0.531, p<0.001). In a ROC curve analysis miR-1 showed an AUC greater than cTnI to discriminate between patients who did and did not develop cardiotoxicity (AUC = 0.851 and 0.544, p= 0.0016). Our data suggest that circulating miR-1 might be a potential new biomarker of doxorubicin-induced cardiotoxicity in breast cancer patients.
Collapse
|
38
|
Piegari E, Russo R, Cappetta D, Esposito G, Urbanek K, Dell'Aversana C, Altucci L, Berrino L, Rossi F, De Angelis A. MicroRNA-34a regulates doxorubicin-induced cardiotoxicity in rat. Oncotarget 2018; 7:62312-62326. [PMID: 27694688 PMCID: PMC5308729 DOI: 10.18632/oncotarget.11468] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 07/26/2016] [Indexed: 12/22/2022] Open
Abstract
New strategies to prevent and early detect the cardiotoxic effects of the anticancer drug doxorubicin (DOXO) are required. MicroRNAs emerged as potential diagnostic, therapeutic and prognostic approaches in cardiovascular diseases. MiR-34a has a role in cardiac dysfunction and ageing and is involved in several cellular processes associated with DOXO cardiotoxicity. Our in vitro and in vivo results indicated that after DOXO exposure the levels of miR-34a are enhanced in cardiac cells, including Cardiac Progenitor Cells (CPCs). Since one of the determining event responsible for the initiation and evolution of the DOXO toxicity arises at the level of the CPC compartment, we evaluated if miR-34a pharmacological inhibition in these cells ameliorates the detrimental aftermath of the drug. AntimiR-34a has beneficial consequences on vitality, proliferation, apoptosis and senescence of DOXO-treated rat CPC. These effects are mediated by an increase of prosurvival miR-34a targets Bcl-2 and SIRT1, accompanied by a decrease of acetylated-p53 and p16INK4a. Importantly, miR-34a silencing also reduces the release of this miRNA from DOXO-exposed rCPCs, decreasing its negative paracrine effects on other rat cardiac cells. In conclusion, the silencing of miR-34a could represent a future therapeutic option for cardioprotection in DOXO toxicity and at the same time, it could be considered as a circulating biomarker for anthracycline-induced cardiac damage.
Collapse
Affiliation(s)
- Elena Piegari
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Naples, Italy
| | - Rosa Russo
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Naples, Italy
| | - Donato Cappetta
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Naples, Italy
| | - Grazia Esposito
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Naples, Italy
| | - Konrad Urbanek
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Naples, Italy
| | | | - Lucia Altucci
- Institute of Genetics and Biophysics, IGB 'Adriano Buzzati-Traverso', Naples, Italy.,Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Liberato Berrino
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Naples, Italy
| | - Francesco Rossi
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Naples, Italy
| | - Antonella De Angelis
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Naples, Italy
| |
Collapse
|
39
|
miR-21-5p as a potential biomarker of inflammatory infiltration in the heart upon acute drug-induced cardiac injury in rats. Toxicol Lett 2018; 286:31-38. [PMID: 29355689 DOI: 10.1016/j.toxlet.2018.01.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 01/12/2018] [Accepted: 01/15/2018] [Indexed: 01/09/2023]
Abstract
Investigation of genomic changes in cardiotoxicity can provide novel biomarkers and insights into molecular mechanisms of drug-induced cardiac injury (DICI). The main objective of this study was to identify and characterize dysregulated microRNAs (miRNAs) in the heart associated with cardiotoxicity. Wistar rats were dosed once with either isoproterenol (1.5 mg/kg, i.p), allylamine (100 mg/kg, p.o.) or the respective vehicle controls. Heart tissue was collected at 24 h, 48 h and 72 h post-drug administration and used for histopathological assessment, miRNA profiling, immunohistochemical analysis and in situ hybridization. Multiplex analysis of 68 miRNAs in the heart revealed a significant upregulation of several miRNAs (miR-19a-3p, miR-142-3p, miR-155-5p, miR-208b-3p, miR-21-5p) after isoproterenol and one miRNA (miR-21-5p) after allylamine administration. Localization of miR-21-5p was specific to inflammatory cell infiltrates in the heart after both treatments. Immunohistochemical analysis of Stat3, a known miR-21-5p regulator, also confirmed its upregulation in cardiomyocytes and inflammatory cell infiltrates. The toxicity signatures based on miRNA networks, identified in vivo, can potentially be used as mechanistic biomarkers as well as to study cardiotoxicity in vitro in order to develop sensitive tools for early hazard identification and risk assessment.
Collapse
|
40
|
Frères P, Bouznad N, Servais L, Josse C, Wenric S, Poncin A, Thiry J, Moonen M, Oury C, Lancellotti P, Bours V, Jerusalem G. Variations of circulating cardiac biomarkers during and after anthracycline-containing chemotherapy in breast cancer patients. BMC Cancer 2018; 18:102. [PMID: 29378531 PMCID: PMC5789542 DOI: 10.1186/s12885-018-4015-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 01/22/2018] [Indexed: 11/21/2022] Open
Abstract
Background Over time, the chance of cure after the diagnosis of breast cancer has been increasing, as a consequence of earlier diagnosis, improved diagnostic procedures and more effective treatment options. However, oncologists are concerned by the risk of long term treatment side effects, including congestive heart failure (CHF). Methods In this study, we evaluated innovative circulating cardiac biomarkers during and after anthracycline-based neoadjuvant chemotherapy (NAC) in breast cancer patients. Levels of cardiac-specific troponins T (cTnT), N-terminal natriuretic peptides (NT-proBNP), soluble ST2 (sST2) and 10 circulating microRNAs (miRNAs) were measured. Results Under chemotherapy, we observed an elevation of cTnT and NT-proBNP levels, but also the upregulation of sST2 and of 4 CHF-related miRNAs (miR-126-3p, miR-199a-3p, miR-423-5p, miR-34a-5p). The elevations of cTnT, NT-proBNP, sST2 and CHF-related miRNAs were poorly correlated, suggesting that these molecules could provide different information. Conclusions Circulating miRNA and sST2 are potential biomarkers of the chemotherapy-related cardiac dysfunction (CRCD). Nevertheless, further studies and long-term follow-up are needed in order to evaluate if these new markers may help to predict CRCD and to identify the patients at risk to later develop CHF. Electronic supplementary material The online version of this article (10.1186/s12885-018-4015-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Pierre Frères
- Department of Medical Oncology, University Hospital (CHU) and University of Liège, Liège, Belgium.,Laboratory of Human Genetics, GIGA Research, University Hospital (CHU) and University of Liège, Liège, Belgium
| | - Nassim Bouznad
- GIGA Cardiovascular Sciences, Department of Cardiology, Heart Valve Clinic, University Hospital (CHU) and University of Liège, Liège, Belgium
| | - Laurence Servais
- GIGA Cardiovascular Sciences, Department of Cardiology, Heart Valve Clinic, University Hospital (CHU) and University of Liège, Liège, Belgium
| | - Claire Josse
- Laboratory of Human Genetics, GIGA Research, University Hospital (CHU) and University of Liège, Liège, Belgium
| | - Stéphane Wenric
- Laboratory of Human Genetics, GIGA Research, University Hospital (CHU) and University of Liège, Liège, Belgium
| | - Aurélie Poncin
- Department of Medical Oncology, University Hospital (CHU) and University of Liège, Liège, Belgium.,Laboratory of Human Genetics, GIGA Research, University Hospital (CHU) and University of Liège, Liège, Belgium
| | - Jérôme Thiry
- Laboratory of Human Genetics, GIGA Research, University Hospital (CHU) and University of Liège, Liège, Belgium
| | - Marie Moonen
- Gruppo Villa Maria Care and Research, Anthea Hospital, Bari, Italy
| | - Cécile Oury
- GIGA Cardiovascular Sciences, Department of Cardiology, Heart Valve Clinic, University Hospital (CHU) and University of Liège, Liège, Belgium
| | - Patrizio Lancellotti
- GIGA Cardiovascular Sciences, Department of Cardiology, Heart Valve Clinic, University Hospital (CHU) and University of Liège, Liège, Belgium.,Gruppo Villa Maria Care and Research, Anthea Hospital, Bari, Italy
| | - Vincent Bours
- Laboratory of Human Genetics, GIGA Research, University Hospital (CHU) and University of Liège, Liège, Belgium
| | - Guy Jerusalem
- Department of Medical Oncology, University Hospital (CHU) and University of Liège, Liège, Belgium.
| |
Collapse
|
41
|
Cappetta D, Rossi F, Piegari E, Quaini F, Berrino L, Urbanek K, De Angelis A. Doxorubicin targets multiple players: A new view of an old problem. Pharmacol Res 2018; 127:4-14. [DOI: 10.1016/j.phrs.2017.03.016] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 03/16/2017] [Accepted: 03/16/2017] [Indexed: 01/22/2023]
|
42
|
Berridge BR, Schultze AE, Heyen JR, Searfoss GH, Sarazan RD. Technological Advances in Cardiovascular Safety Assessment Decrease Preclinical Animal Use and Improve Clinical Relevance. ILAR J 2017; 57:120-132. [PMID: 28053066 DOI: 10.1093/ilar/ilw028] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 10/09/2016] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular (CV) safety liabilities are significant concerns for drug developers and preclinical animal studies are predominately where those liabilities are characterized before patient exposures. Steady progress in technology and laboratory capabilities is enabling a more refined and informative use of animals in those studies. The application of surgically implantable and telemetered instrumentation in the acute assessment of drug effects on CV function has significantly improved historical approaches that involved anesthetized or restrained animals. More chronically instrumented animals and application of common clinical imaging assessments like echocardiography and MRI extend functional and in-life structural assessments into the repeat-dose setting. A growing portfolio of circulating CV biomarkers is allowing longitudinal and repeated measures of cardiac and vascular injury and dysfunction better informing an understanding of temporal pathogenesis and allowing earlier detection of undesirable effects. In vitro modeling systems of the past were limited by their lack of biological relevance to the in vivo human condition. Advances in stem cell technology and more complex in vitro modeling platforms are quickly creating more opportunity to supplant animals in our earliest assessments for liabilities. Continuing improvement in our capabilities in both animal and nonanimal modeling should support a steady decrease in animal use for primary liability identification and optimize the translational relevance of the animal studies we continue to do.
Collapse
Affiliation(s)
- Brian R Berridge
- Brian R. Berridge, DVM, PhD, is a Senior GSK Fellow and Head of Worldwide Animal Research Strategy at GlaxoSmithKline in King of Prussia, Pennsylvania. A. Eric Schultze, DVM, PhD, is a Senior Research Advisor-Pathologist at Lilly Research Laboratories in Indianapolis, Indiana. Jon R. Heyen, MS, is a Senior Principal Scientist at Pfizer in La Jolla, California. George H. Searfoss, MS, is a Consultant Toxicologist at Lilly Research Laboratories in Indianapolis, Indiana. R. Dustan Sarazan, DVM, PhD, is a cardiovascular consultant currently residing in Rhinelander, Wisconsin
| | - A Eric Schultze
- Brian R. Berridge, DVM, PhD, is a Senior GSK Fellow and Head of Worldwide Animal Research Strategy at GlaxoSmithKline in King of Prussia, Pennsylvania. A. Eric Schultze, DVM, PhD, is a Senior Research Advisor-Pathologist at Lilly Research Laboratories in Indianapolis, Indiana. Jon R. Heyen, MS, is a Senior Principal Scientist at Pfizer in La Jolla, California. George H. Searfoss, MS, is a Consultant Toxicologist at Lilly Research Laboratories in Indianapolis, Indiana. R. Dustan Sarazan, DVM, PhD, is a cardiovascular consultant currently residing in Rhinelander, Wisconsin
| | - Jon R Heyen
- Brian R. Berridge, DVM, PhD, is a Senior GSK Fellow and Head of Worldwide Animal Research Strategy at GlaxoSmithKline in King of Prussia, Pennsylvania. A. Eric Schultze, DVM, PhD, is a Senior Research Advisor-Pathologist at Lilly Research Laboratories in Indianapolis, Indiana. Jon R. Heyen, MS, is a Senior Principal Scientist at Pfizer in La Jolla, California. George H. Searfoss, MS, is a Consultant Toxicologist at Lilly Research Laboratories in Indianapolis, Indiana. R. Dustan Sarazan, DVM, PhD, is a cardiovascular consultant currently residing in Rhinelander, Wisconsin
| | - George H Searfoss
- Brian R. Berridge, DVM, PhD, is a Senior GSK Fellow and Head of Worldwide Animal Research Strategy at GlaxoSmithKline in King of Prussia, Pennsylvania. A. Eric Schultze, DVM, PhD, is a Senior Research Advisor-Pathologist at Lilly Research Laboratories in Indianapolis, Indiana. Jon R. Heyen, MS, is a Senior Principal Scientist at Pfizer in La Jolla, California. George H. Searfoss, MS, is a Consultant Toxicologist at Lilly Research Laboratories in Indianapolis, Indiana. R. Dustan Sarazan, DVM, PhD, is a cardiovascular consultant currently residing in Rhinelander, Wisconsin
| | - R Dustan Sarazan
- Brian R. Berridge, DVM, PhD, is a Senior GSK Fellow and Head of Worldwide Animal Research Strategy at GlaxoSmithKline in King of Prussia, Pennsylvania. A. Eric Schultze, DVM, PhD, is a Senior Research Advisor-Pathologist at Lilly Research Laboratories in Indianapolis, Indiana. Jon R. Heyen, MS, is a Senior Principal Scientist at Pfizer in La Jolla, California. George H. Searfoss, MS, is a Consultant Toxicologist at Lilly Research Laboratories in Indianapolis, Indiana. R. Dustan Sarazan, DVM, PhD, is a cardiovascular consultant currently residing in Rhinelander, Wisconsin
| |
Collapse
|
43
|
The Positive Effects of Exercise in Chemotherapy-Related Cardiomyopathy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1000:103-129. [DOI: 10.1007/978-981-10-4304-8_8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
44
|
Renu K, Abilash V, Tirupathi Pichiah P, Syeda TA, Arunachalam S. Adriamycin-induced cardiomyopathy can serve as a model for diabetic cardiomyopathy – a hypothesis. Asian Pac J Trop Biomed 2017. [DOI: 10.1016/j.apjtb.2017.09.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
45
|
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: 394] [Impact Index Per Article: 49.3] [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
|
46
|
Activation of miR-34a-5p/Sirt1/p66shc pathway contributes to doxorubicin-induced cardiotoxicity. Sci Rep 2017; 7:11879. [PMID: 28928469 PMCID: PMC5605522 DOI: 10.1038/s41598-017-12192-y] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 09/06/2017] [Indexed: 02/07/2023] Open
Abstract
The molecular mechanisms underlying anthracyclines-induced cardiotoxicity have not been well elucidated. MiRNAs were revealed dysregulated in the myocardium and plasma of rats received Dox treatment. MicroRNA-34a-5p (miR-34a-5p) was verified increased in the myocardium and plasma of Dox-treated rats, but was reversed in rats received Dox plus DEX treatments. Human miR-34a-5p was also observed increased in the plasma of patients with diffuse large B-cell lymphoma after 9- and 16-week epirubicin therapy. Up-regulation of miR-34a-5p was observed in Dox-induced rat cardiomyocyte H9c2 cells. MiR-34a-5p could augment Bax expression, but inhibited Bcl-2 expression, along with the increases of the activated caspase-3 and mitochondrial potentials in H9C2 cells. MiR-34a-5p was verified to modulate Sirt1 expression post-transcriptionally. In parallel to Sirt1 siRNA, miR-34a-5p could enhance p66shc expression, accompanied by increases of Bax and the activated caspase-3 and a decrease of Bcl-2 in H9c2 cells. Moreover, enforced expression of Sirt1 alleviated Dox-induced apoptosis of H9c2 cells, with suppressing levels of p66shc, Bax, the activated caspase-3 and miR-34a-5p, and enhancing Bcl-2 expression. Therefore, miR-34a-5p enhances cardiomyocyte apoptosis by targeting Sirt1, activation of miR-34a-5p/Sirt1/p66shc pathway contributes to Dox-induced cardiotoxicity, and blockage of this pathway represents a potential cardioprotective effect against anthracyclines.
Collapse
|
47
|
Razavi-Azarkhiavi K, Jaafari MR, Abnous K, Razavi BM, Jafarian AH, Hassani FV, Shirani K, Karimi G. The Cardiotoxic Mechanism of Doxorubicin (DOX) and Pegylated Liposomal DOX in Mice Bearing C-26 Colon Carcinoma: a Study Focused on microRNA Role for Toxicity Assessment of New Formulations. Pharm Res 2017; 34:1849-1856. [PMID: 28560697 DOI: 10.1007/s11095-017-2194-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 05/23/2017] [Indexed: 01/25/2023]
Abstract
PURPOSE MicroRNAs (miRs) are a group of small non-coding RNAs that regulate transcriptional or post-transcriptional gene expression. The aim of the present study was to investigate the role of miR -1, -21 and -145 and their targets in cardiotoxicity-induced by DOX and pegylated liposomal DOX. METHODS BALB/c mice subjected to subcutaneous injection of C-26 tumor cells. Eight days after tumor inoculation, animals were divided into 6 groups: control, liposome, DOX (6 and 9 mg/kg) and PL-DOX (6 and 9 mg/kg). The formulations were administered one time per week for four weeks. 24 h after the last injection, mice were sacrificed; blood and heart samples were taken. Western blot analysis was done on protein extracts to investigate the expression of cardiac caspase-3, -8, Bax, Bcl2, Programmed cell death 4 (PDCD4) and BCL2/Adenovirus E1B 19 kDa Interacting Protein 3 (BNIP3). The expression levels of miR -1, -21 and -145 were also evaluated by quantitative real-time PCR. RESULTS Mice treated with both DOX formulations showed a marked inhibition in tumor growth. Western blot analysis indicated that the expression level of cardiac caspase-3, caspase-8, Bax and BNIP3 were up-regulated due to DOX injection (9 mg/kg). Exposure of mice with DOX resulted in a significant increase in cardiac miR-1 and miR-21 expression level. PL-DOX treatment did not change the proteins and miRs expression. CONCLUSION The results suggest that miR -1, -21 and -145 may involve in cardiotoxicity induced by DOX. Evaluation of miRs signaling pathways might be of potential value for toxicity assessment of new formulations. Graphical Abstract The cardiotoxic mechanism of doxorubicin (DOX) and pegylated liposomal DOX (PL-DOX).
Collapse
Affiliation(s)
- Kamal Razavi-Azarkhiavi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Biotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Nanotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, School of Pharmacy, Mashhad University of Medical Sciences, P. O. Box, 1365-91775, I.R., Mashhad, Iran
| | - Bibi Marjan Razavi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Hossein Jafarian
- Cancer Molecular Research Center, Ghaem Hospital, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Faezeh Vahdati Hassani
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Kobra Shirani
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gholamreza Karimi
- Pharmaceutical Research Center, School of Pharmacy, Mashhad University of Medical Sciences, P. O. Box, 1365-91775, I.R., Mashhad, Iran.
| |
Collapse
|
48
|
Diagnóstico y prevención de la cardiotoxicidad inducida por fármacos antineoplásicos: de la imagen a las tecnologías «ómicas». Rev Esp Cardiol 2017. [DOI: 10.1016/j.recesp.2016.12.032] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
49
|
Leger KJ, Leonard D, Nielson D, de Lemos JA, Mammen PPA, Winick NJ. Circulating microRNAs: Potential Markers of Cardiotoxicity in Children and Young Adults Treated With Anthracycline Chemotherapy. J Am Heart Assoc 2017; 6:JAHA.116.004653. [PMID: 28377429 PMCID: PMC5532993 DOI: 10.1161/jaha.116.004653] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Background Biomarkers for early detection of anthracycline (AC)‐induced cardiotoxicity may allow cardioprotective intervention before irreversible damage. Circulating microRNAs (miRNAs) are promising biomarkers of cardiovascular disease, however, have not been studied in the setting of AC‐induced cardiotoxicity. This study aimed to identify AC‐induced alterations in plasma miRNA expression in children and correlate expression with markers of cardiac injury. Methods and Results Candidate plasma profiling of 24 miRNAs was performed in 33 children before and after a cycle of AC (n=24) or noncardiotoxic chemotherapy (n=9). Relative miRNA changes between the pre‐ and postcycle time points (6, 12, and 24 hours) were determined within each treatment group and compared across groups. Plasma miRNA expression patterns were further explored with respect to AC dose and high‐sensitivity troponin T. Greater chemotherapy‐induced dysregulation was observed in this panel of candidate, cardiac‐related plasma miRNAs in patients receiving anthracyclines compared with those receiving noncardiotoxic chemotherapy (24‐hour MANOVA; P=0.024). Specifically, plasma miRs‐29b and ‐499 were upregulated 6 to 24 hours post‐AC, and their postchemotherapy expression significantly correlated with AC dose. Patients with acute cardiomyocyte injury (high‐sensitivity troponin T increase ≥5 ng/L from baseline) demonstrated higher expression of miR‐29b and miR‐499 post‐AC compared with those without. Conclusions In this pilot study, cardiac‐related plasma miRNAs are dysregulated following ACs. Plasma miR‐29b and ‐499 are acutely elevated post‐AC, with dose response relationships observed with anthracycline dose and markers of cardiac injury. Further evaluation of miRNAs may provide mechanistic insight into AC‐induced cardiotoxicity and yield biomarkers to facilitate earlier intervention to mitigate cardiotoxicity.
Collapse
Affiliation(s)
- Kasey J Leger
- Department of Pediatrics, Seattle Children's Hospital, University of Washington School of Medicine, Seattle, WA
| | - David Leonard
- Department of Clinical Research, Children's Medical Center, Dallas, TX
| | - Danelle Nielson
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX
| | - James A de Lemos
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX
| | - Pradeep P A Mammen
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX
- Heart Failure, Ventricular Assist Device & Heart Transplant Program, University of Texas Southwestern Medical Center, Dallas, TX
| | - Naomi J Winick
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
50
|
Srikanthan K, Klug R, Tirona M, Thompson E, Visweshwar H, Puri N, Shapiro J, Sodhi K. Creating a Biomarker Panel for Early Detection of Chemotherapy Related Cardiac Dysfunction in Breast Cancer Patients. ACTA ACUST UNITED AC 2017. [PMID: 28642833 DOI: 10.4172/2155-9880.1000507] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cardiotoxicity is an important issue for breast cancer patients receiving anthracycline-trastuzumab therapy in the adjuvant setting. Studies show that 3-36% of patients receiving anthracyclines and/or trastuzumab experience chemotherapy related cardiac dysfunction (CRCD) and approximately 17% of patients must stop chemotherapy due to the consequences of CRCD. There is currently no standardized, clinically verified way to detect CRCD early, but common practices include serial echocardiography and troponin measurements, which can be timely, costly, and not always available in areas where health care resources are scarce. Furthermore, detection of CRCD, before there is any echocardiographic evidence of dysfunction or clinical symptoms present, would allow maximal benefit of chemotherapy and minimize cardiac complications. Creating a panel of serum biomarkers would allow for more specificity and sensitivity in the early detection of CRCD, which would be easy to implement and cost effective in places with limited health care. Based on a review of the literature, we propose creating a biomarker panel consisting of topoisomerase 2β, serum troponin T/I, myeloperoxidase, NT-proBNP, miR-208b, miR-34a, and miR-150 in breast cancer patients receiving anthracyclines and/or trastuzumab to detect CRCD before any signs of overt cardiotoxicity are apparent.
Collapse
Affiliation(s)
- Krithika Srikanthan
- Department of Internal Medicine, Marshall University Joan C Edwards School of Medicine, Huntington, WV, USA
| | - Rebecca Klug
- Department of Surgery, Marshall University Joan C Edwards School of Medicine, Huntington, WV, USA
| | - Maria Tirona
- Division of Hematology and Oncology, Department of Internal Medicine, Marshall University Joan C Edwards School of Medicine, Huntington, WV, USA
| | - Ellen Thompson
- Department of Cardiology, Marshall University Joan C Edwards School of Medicine, Huntington, WV, USA
| | - Haresh Visweshwar
- Department of Internal Medicine, Marshall University Joan C Edwards School of Medicine, Huntington, WV, USA
| | - Nitin Puri
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, 43614, USA
| | - Joseph Shapiro
- Department of Internal Medicine, Marshall University Joan C Edwards School of Medicine, Huntington, WV, USA
| | - Komal Sodhi
- Department of Surgery, Marshall University Joan C Edwards School of Medicine, Huntington, WV, USA
| |
Collapse
|