1
|
Disner GR, Fernandes TADM, Nishiyama-Jr MY, Lima C, Wincent E, Lopes-Ferreira M. TnP and AHR-CYP1A1 Signaling Crosstalk in an Injury-Induced Zebrafish Inflammation Model. Pharmaceuticals (Basel) 2024; 17:1155. [PMID: 39338318 PMCID: PMC11435205 DOI: 10.3390/ph17091155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 08/21/2024] [Accepted: 08/29/2024] [Indexed: 09/30/2024] Open
Abstract
Aryl Hydrocarbon Receptor (AHR) signaling is crucial for regulating the biotransformation of xenobiotics and physiological processes like inflammation and immunity. Meanwhile, Thalassophryne nattereri Peptide (TnP), a promising anti-inflammatory candidate from toadfish venom, demonstrates therapeutic effects through immunomodulation. However, its influence on AHR signaling remains unexplored. This study aimed to elucidate TnP's molecular mechanisms on the AHR-cytochrome P450, family 1 (CYP1) pathway upon injury-induced inflammation in wild-type (WT) and Ahr2-knockdown (KD) zebrafish larvae through transcriptomic analysis and Cyp1a reporters. TnP, while unable to directly activate AHR, potentiated AHR activation by the high-affinity ligand 6-Formylindolo [3,2-b]carbazole (FICZ), implying a role as a CYP1A inhibitor, confirmed by in vitro studies. This interplay suggests TnP's ability to modulate the AHR-CYP1 complex, prompting investigations into its influence on biotransformation pathways and injury-induced inflammation. Here, the inflammation model alone resulted in a significant response on the transcriptome, with most differentially expressed genes (DEGs) being upregulated across the groups. Ahr2-KD resulted in an overall greater number of DEGs, as did treatment with the higher dose of TnP in both WT and KD embryos. Genes related to oxidative stress and inflammatory response were the most apparent under inflamed conditions for both WT and KD groups, e.g., Tnfrsf1a, Irf1b, and Mmp9. TnP, specifically, induces the expression of Hspa5, Hsp90aa1.2, Cxcr3.3, and Mpeg1.2. Overall, this study suggests an interplay between TnP and the AHR-CYP1 pathway, stressing the inflammatory modulation through AHR-dependent mechanisms. Altogether, these results may offer new avenues in novel therapeutic strategies, such as based on natural bioactive molecules, harnessing AHR modulation for targeted and sustained drug effects in inflammatory conditions.
Collapse
Affiliation(s)
- Geonildo Rodrigo Disner
- Immunoregulation Unit, Laboratory of Applied Toxinology (CeTICS/FAPESP), Butantan Institute, São Paulo 05585-000, Brazil
- Unit of System Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77 Solna, Sweden
| | - Thales Alves de Melo Fernandes
- Nucleus of Bioinformatics and Computational Biology, Laboratory of Applied Toxinology, Butantan Institute, São Paulo 05585-000, Brazil
| | - Milton Yutaka Nishiyama-Jr
- Nucleus of Bioinformatics and Computational Biology, Laboratory of Applied Toxinology, Butantan Institute, São Paulo 05585-000, Brazil
| | - Carla Lima
- Immunoregulation Unit, Laboratory of Applied Toxinology (CeTICS/FAPESP), Butantan Institute, São Paulo 05585-000, Brazil
| | - Emma Wincent
- Unit of System Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77 Solna, Sweden
| | - Monica Lopes-Ferreira
- Immunoregulation Unit, Laboratory of Applied Toxinology (CeTICS/FAPESP), Butantan Institute, São Paulo 05585-000, Brazil
| |
Collapse
|
2
|
Yadav P, Singh SK, Datta S, Verma S, Verma A, Rakshit A, Bali A, Bhatti JS, Khurana A, Navik U. Therapeutic potential and pharmacological mechanism of visnagin. JOURNAL OF INTEGRATIVE MEDICINE 2024; 22:399-412. [PMID: 38797603 DOI: 10.1016/j.joim.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 04/10/2024] [Indexed: 05/29/2024]
Abstract
Visnagin is a furanochromone and one of the most important compound in the Ammi visnaga (L.) Lam (a synonym of Visnaga daucoides Gaertn.) plant, which is used to cure various ailments. Many investigations into the bioactive properties of visnagin have been studied to date. The literature on visnagin demonstrates its biological properties, including anti-inflammatory, anti-diabetic, and beneficial effects in cardiovascular and renal diseases. Moreover, visnagin improves sperm quality parameters, stimulates steroidogenesis, and increases serum gonadotropins and testosterone levels, while decreasing pro-inflammatory cytokines, oxidative damage, genomic instability, and it modulates apoptosis. Thus, visnagin has emerged as an exciting lead for further research, owing to its potential in various unmet clinical needs. The current review summarized its basic structure, pharmacokinetics, and pharmacological effects, focusing on its mechanisms of action. The review will help to understand the potential of visnagin as an alternative treatment strategy for several diseases and provide insight into research topics that need further exploration for visnagin's safe clinical use. Please cite this article as: Yadav P, Singh SK, Datta S, Verma S, Verma A, Rakshit A, Bali A, Bhatti JS, Khurana A, Navik U. Therapeutic potential and pharmacological mechanism of visnagin. J Integr Med. 2024; 22(4): 399-412.
Collapse
Affiliation(s)
- Poonam Yadav
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Sumeet Kumar Singh
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Sayantap Datta
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204-5000, USA
| | - Saloni Verma
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Aarti Verma
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Arnab Rakshit
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Anjana Bali
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Jasvinder Singh Bhatti
- Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Amit Khurana
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India.
| | - Umashanker Navik
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India.
| |
Collapse
|
3
|
Obeidat O, Obeidat A, Obeidat A, Ismail MF. Visnagin: A novel cardioprotective agent against anthracycline toxicity (Review). MEDICINE INTERNATIONAL 2024; 4:37. [PMID: 38799005 PMCID: PMC11117031 DOI: 10.3892/mi.2024.161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 04/30/2024] [Indexed: 05/29/2024]
Abstract
Doxorubicin (DOX), a cornerstone of cancer chemotherapy, is marred by its dose-dependent cardiotoxicity, leading to cardiomyopathy and heart failure. The epidemiology of DOX-related cardiotoxicity highlights its cumulative, progressive nature, with a significant impact on the health of patients. The pathophysiological mechanisms involve mitochondrial dysfunction, oxidative stress and disrupted calcium homeostasis in cardiomyocytes. Despite the search for effective cardioprotective strategies, current treatments offer limited efficacy. Visnagin emerges as a potential solution, known for its vasodilatory and anti-inflammatory properties, and recent studies suggest its cardioprotective efficacy against DOX-induced cardiotoxicity through mitochondrial protection, the modulation of key signaling pathways and the inhibition of apoptosis. The present review aimed to provide a comprehensive overview of the mechanisms of action of visnagin, as well as to provide experimental evidence, and potential integration into cancer treatment regimens, highlighting its promise as a novel therapeutic agent for managing cardiotoxicity in patients undergoing anthracycline chemotherapy.
Collapse
Affiliation(s)
- Omar Obeidat
- Graduate Medical Education Program, College of Medicine, University of Central Florida, Orlando, FL 32816, USA
- Internal Medicine Residency Program, HCA Florida North Florida Hospital, Gainesville, FL 32605, USA
| | - Ali Obeidat
- Division of Cardiology, Department of Internal Medicine, Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Abedallah Obeidat
- Division of Cardiology, Department of Internal Medicine, Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Mohamed F. Ismail
- Graduate Medical Education Program, College of Medicine, University of Central Florida, Orlando, FL 32816, USA
- Internal Medicine Residency Program, HCA Florida North Florida Hospital, Gainesville, FL 32605, USA
| |
Collapse
|
4
|
Liu J, Curtin C, Lall R, Lane S, Wieke J, Ariza A, Sejour L, Vlachos I, Zordoky BN, Peterson RT, Asnani A. Inhibition of Cyp1a Protects Mice against Anthracycline Cardiomyopathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.10.588915. [PMID: 38645084 PMCID: PMC11030370 DOI: 10.1101/2024.04.10.588915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Background Anthracyclines such as doxorubicin (Dox) are highly effective anti-tumor agents, but their use is limited by dose-dependent cardiomyopathy and heart failure. Our laboratory previously reported that induction of cytochrome P450 family 1 (Cyp1) enzymes contributes to acute Dox cardiotoxicity in zebrafish and in mice, and that potent Cyp1 inhibitors prevent cardiotoxicity. However, the role of Cyp1 enzymes in chronic Dox cardiomyopathy, as well as the mechanisms underlying cardioprotection associated with Cyp1 inhibition, have not been fully elucidated. Methods The Cyp1 pathway was evaluated using a small molecule Cyp1 inhibitor in wild-type (WT) mice, or Cyp1-null mice ( Cyp1a1/1a2 -/- , Cyp1b1 -/- , and Cyp1a1/1a2/1b1 -/- ). Low-dose Dox was administered by serial intraperitoneal or intravenous injections, respectively. Expression of Cyp1 isoforms was measured by RT-qPCR, and myocardial tissue was isolated from the left ventricle for RNA sequencing. Cardiac function was evaluated by transthoracic echocardiography. Results In WT mice, Dox treatment was associated with a decrease in Cyp1a2 and increase in Cyp1b1 expression in the heart and in the liver. Co-treatment of WT mice with Dox and the novel Cyp1 inhibitor YW-130 protected against cardiac dysfunction compared to Dox treatment alone. Cyp1a1/1a2 -/- and Cyp1a1/1a2/1b1 -/- mice were protected from Dox cardiomyopathy compared to WT mice. Male, but not female, Cyp1b1 -/- mice had increased cardiac dysfunction following Dox treatment compared to WT mice. RNA sequencing of myocardial tissue showed upregulation of Fundc1 and downregulation of Ccl21c in Cyp1a1/1a2 -/- mice treated with Dox, implicating changes in mitophagy and chemokine-mediated inflammation as possible mechanisms of Cyp1a-mediated cardioprotection. Conclusions Taken together, this study highlights the potential therapeutic value of Cyp1a inhibition in mitigating anthracycline cardiomyopathy.
Collapse
|
5
|
Sheng SY, Li JM, Hu XY, Wang Y. Regulated cell death pathways in cardiomyopathy. Acta Pharmacol Sin 2023; 44:1521-1535. [PMID: 36914852 PMCID: PMC10374591 DOI: 10.1038/s41401-023-01068-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 02/20/2023] [Indexed: 03/16/2023]
Abstract
Heart disease is a worldwide health menace. Both intractable primary and secondary cardiomyopathies contribute to malignant cardiac dysfunction and mortality. One of the key cellular processes associated with cardiomyopathy is cardiomyocyte death. Cardiomyocytes are terminally differentiated cells with very limited regenerative capacity. Various insults can lead to irreversible damage of cardiomyocytes, contributing to progression of cardiac dysfunction. Accumulating evidence indicates that majority of cardiomyocyte death is executed by regulating molecular pathways, including apoptosis, ferroptosis, autophagy, pyroptosis, and necroptosis. Importantly, these forms of regulated cell death (RCD) are cardinal features in the pathogenesis of various cardiomyopathies, including dilated cardiomyopathy, diabetic cardiomyopathy, sepsis-induced cardiomyopathy, and drug-induced cardiomyopathy. The relevance between abnormity of RCD with adverse outcome of cardiomyopathy has been unequivocally evident. Therefore, there is an urgent need to uncover the molecular and cellular mechanisms for RCD in order to better understand the pathogenesis of cardiomyopathies. In this review, we summarize the latest progress from studies on RCD pathways in cardiomyocytes in context of the pathogenesis of cardiomyopathies, with particular emphasis on apoptosis, necroptosis, ferroptosis, autophagy, and pyroptosis. We also elaborate the crosstalk among various forms of RCD in pathologically stressed myocardium and the prospects of therapeutic applications targeted to various cell death pathways.
Collapse
Affiliation(s)
- Shu-Yuan Sheng
- Department of Cardiology, Zhejiang University School of Medicine, Second Affiliated Hospital, Hangzhou, 310009, China
| | - Jia-Min Li
- Department of Cardiology, Zhejiang University School of Medicine, Second Affiliated Hospital, Hangzhou, 310009, China
| | - Xin-Yang Hu
- Department of Cardiology, Zhejiang University School of Medicine, Second Affiliated Hospital, Hangzhou, 310009, China
| | - Yibin Wang
- Department of Cardiology, Zhejiang University School of Medicine, Second Affiliated Hospital, Hangzhou, 310009, China.
- Signature Program in Cardiovascular and Metabolic Diseases, DukeNUS Medical School and National Heart Center of Singapore, Singapore, Singapore.
| |
Collapse
|
6
|
Wu Y, Shi JH, Zhu GH, Xiong Y, Gong JH, Wei HZ, Guo ZB, Dai ZR, Sun XB, Ge GB. Discovery of 4'-trifluoromethylchalcones as novel, potent and selective hCYP1B1 inhibitors without concomitant AhR activation. Eur J Med Chem 2023; 258:115552. [PMID: 37315474 DOI: 10.1016/j.ejmech.2023.115552] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/27/2023] [Accepted: 06/07/2023] [Indexed: 06/16/2023]
Abstract
Human cytochrome P450 1B1 (hCYP1B1), an extrahepatic cytochrome P450 enzyme over-expressed in various tumors, has been validated as a promising target for preventing and treating cancers. Herein, two series of chalcone derivatives were synthesized to discover potent hCYP1B1 inhibitors without AhR agonist effect. Structure-activity relationship (SAR) studies demonstrated that 4'-trifluoromethyl on the B-ring strongly enhanced the anti-hCYP1B1 effects, identifying A9 as a promising lead compound. Further SAR analysis on A9 derivatives (modified A-ring of 4'-trifluoromethylchalcone) showed that introducing 2-methoxyl improved the anti-hCYP1B1 effect and selectivity, while introducing a methoxyl at the C-4 site was beneficial for avoiding AhR activation. Ultimately, five 4'-trifluoromethyl chalcones were identified as potent hCYP1B1 inhibitors (IC50 < 10 nM), while B18 exhibits the most potent anti-hCYP1B1 effect (IC50 = 3.6 nM), suitable metabolic stability and good cell-permeability. B18 also acted as an AhR antagonist and could down-regulate hCYP1B1 in living systems. Mechanistic studies showed that B18 potently inhibited hCYP1B1 in a competitive inhibition manner (Ki = 3.92 nM), while docking simulations revealed that B18 could tightly bind to the catalytic cavity of hCYP1B1 mainly via hydrophobic and hydrogen-bonding interactions. Furthermore, B18 could potently inhibit hCYP1B1 in living cells and showed remarkable anti-migration ability on MFC-7 cells. Taken together, this study deciphered the SARs of chalcones as hCYP1B1 inhibitors and provided several potent hCYP1B1 inhibitors as promising candidates for the development of more efficacious anti-migration agents.
Collapse
Affiliation(s)
- Yue Wu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jin-Hui Shi
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Guang-Hao Zhu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yuan Xiong
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jia-Hao Gong
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Hui-Zhen Wei
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zhao-Bin Guo
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zi-Ru Dai
- Ministry of Education, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100193, China.
| | - Xiao-Bo Sun
- Ministry of Education, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100193, China
| | - Guang-Bo Ge
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
7
|
Moossavi M, Lu X, Herrmann J, Xu X. Molecular mechanisms of anthracycline induced cardiotoxicity: Zebrafish come into play. Front Cardiovasc Med 2023; 10:1080299. [PMID: 36970353 PMCID: PMC10036604 DOI: 10.3389/fcvm.2023.1080299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 02/22/2023] [Indexed: 03/12/2023] Open
Abstract
Anthracyclines are among the most potent chemotherapeutics; however, cardiotoxicity significantly restricts their use. Indeed, anthracycline-induced cardiotoxicity (AIC) fares among the worst types of cardiomyopathy, and may only slowly and partially respond to standard heart failure therapies including β-blockers and ACE inhibitors. No therapy specifically designed to treat anthracycline cardiomyopathy at present, and neither is it known if any such strategy could be developed. To address this gap and to elucidate the molecular basis of AIC with a therapeutic goal in mind, zebrafish has been introduced as an in vivo vertebrate model about a decade ago. Here, we first review our current understanding of the basic molecular and biochemical mechanisms of AIC, and then the contribution of zebrafish to the AIC field. We summarize the generation of embryonic zebrafish AIC models (eAIC) and their use for chemical screening and assessment of genetic modifiers, and then the generation of adult zebrafish AIC models (aAIC) and their use for discovering genetic modifiers via forward mutagenesis screening, deciphering spatial-temporal-specific mechanisms of modifier genes, and prioritizing therapeutic compounds via chemical genetic tools. Several therapeutic target genes and related therapies have emerged, including a retinoic acid (RA)-based therapy for the early phase of AIC and an autophagy-based therapy that, for the first time, is able to reverse cardiac dysfunction in the late phase of AIC. We conclude that zebrafish is becoming an important in vivo model that would accelerate both mechanistic studies and therapeutic development of AIC.
Collapse
Affiliation(s)
- Maryam Moossavi
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Xiaoguang Lu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Joerg Herrmann
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Xiaolei Xu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Correspondence: Xiaolei Xu
| |
Collapse
|
8
|
Liu J, Lane S, Lall R, Russo M, Farrell L, Debreli Coskun M, Curtin C, Araujo-Gutierrez R, Scherrer-Crosbie M, Trachtenberg BH, Kim J, Tolosano E, Ghigo A, Gerszten RE, Asnani A. Circulating hemopexin modulates anthracycline cardiac toxicity in patients and in mice. SCIENCE ADVANCES 2022; 8:eadc9245. [PMID: 36563141 PMCID: PMC9788780 DOI: 10.1126/sciadv.adc9245] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 11/29/2022] [Indexed: 05/28/2023]
Abstract
Anthracyclines such as doxorubicin (Dox) are effective chemotherapies, but their use is limited by cardiac toxicity. We hypothesized that plasma proteomics in women with breast cancer could identify new mechanisms of anthracycline cardiac toxicity. We measured changes in 1317 proteins in anthracycline-treated patients (n = 30) and replicated key findings in a second cohort (n = 31). An increase in the heme-binding protein hemopexin (Hpx) 3 months after anthracycline initiation was associated with cardiac toxicity by echocardiography. To assess the functional role of Hpx, we administered Hpx to wild-type (WT) mice treated with Dox and observed improved cardiac function. Conversely, Hpx-/- mice demonstrated increased Dox cardiac toxicity compared to WT mice. Initial mechanistic studies indicate that Hpx is likely transported to the heart by circulating monocytes/macrophages and that Hpx may mitigate Dox-induced ferroptosis to confer cardioprotection. Together, these observations suggest that Hpx induction represents a compensatory response during Dox treatment.
Collapse
Affiliation(s)
- Jing Liu
- Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Sarah Lane
- Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Rahul Lall
- Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Michele Russo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, "Guido Tarone," University of Torino, Torino, Italy
| | - Laurie Farrell
- Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Melis Debreli Coskun
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, Lowell, MA, USA
| | - Casie Curtin
- Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Raquel Araujo-Gutierrez
- Division of Advanced Heart Failure and Transplantation, Houston Methodist Heart and Vascular Center, Houston, TX, USA
| | - Marielle Scherrer-Crosbie
- Division of Cardiovascular Diseases, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Barry H. Trachtenberg
- Division of Advanced Heart Failure and Transplantation, Houston Methodist Heart and Vascular Center, Houston, TX, USA
| | - Jonghan Kim
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, Lowell, MA, USA
| | - Emanuela Tolosano
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, "Guido Tarone," University of Torino, Torino, Italy
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, "Guido Tarone," University of Torino, Torino, Italy
| | - Robert E. Gerszten
- Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Aarti Asnani
- Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
9
|
Chen HH, Khatun Z, Wei L, Mekkaoui C, Patel D, Kim SJW, Boukhalfa A, Enoma E, Meng L, Chen YI, Kaikkonen L, Li G, Capen DE, Sahu P, Kumar ATN, Blanton RM, Yuan H, Das S, Josephson L, Sosnovik DE. A nanoparticle probe for the imaging of autophagic flux in live mice via magnetic resonance and near-infrared fluorescence. Nat Biomed Eng 2022; 6:1045-1056. [PMID: 35817962 PMCID: PMC9492651 DOI: 10.1038/s41551-022-00904-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 02/23/2022] [Indexed: 01/18/2023]
Abstract
Autophagy-the lysosomal degradation of cytoplasmic components via their sequestration into double-membraned autophagosomes-has not been detected non-invasively. Here we show that the flux of autophagosomes can be measured via magnetic resonance imaging or serial near-infrared fluorescence imaging of intravenously injected iron oxide nanoparticles decorated with cathepsin-cleavable arginine-rich peptides functionalized with the near-infrared fluorochrome Cy5.5 (the peptides facilitate the uptake of the nanoparticles by early autophagosomes, and are then cleaved by cathepsins in lysosomes). In the heart tissue of live mice, the nanoparticles enabled quantitative measurements of changes in autophagic flux, upregulated genetically, by ischaemia-reperfusion injury or via starvation, or inhibited via the administration of a chemotherapeutic or the antibiotic bafilomycin. In mice receiving doxorubicin, pre-starvation improved cardiac function and overall survival, suggesting that bursts of increased autophagic flux may have cardioprotective effects during chemotherapy. Autophagy-detecting nanoparticle probes may facilitate the further understanding of the roles of autophagy in disease.
Collapse
Affiliation(s)
- Howard H Chen
- Molecular Cardiology Research Institute, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA.
- A.A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Zehedina Khatun
- Molecular Cardiology Research Institute, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA
| | - Lan Wei
- Molecular Cardiology Research Institute, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA
| | - Choukri Mekkaoui
- A.A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Dakshesh Patel
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sally Ji Who Kim
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Asma Boukhalfa
- Molecular Cardiology Research Institute, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA
| | - Efosa Enoma
- Molecular Cardiology Research Institute, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA
| | - Lin Meng
- Molecular Cardiology Research Institute, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA
| | - Yinching I Chen
- A.A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Leena Kaikkonen
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Guoping Li
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Diane E Capen
- Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Parul Sahu
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Anand T N Kumar
- A.A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Robert M Blanton
- Molecular Cardiology Research Institute, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA
| | - Hushan Yuan
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Saumya Das
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Lee Josephson
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - David E Sosnovik
- A.A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
10
|
Abstract
Heart disease is the leading cause of death worldwide. Despite decades of research, most heart pathologies have limited treatments, and often the only curative approach is heart transplantation. Thus, there is an urgent need to develop new therapeutic approaches for treating cardiac diseases. Animal models that reproduce the human pathophysiology are essential to uncovering the biology of diseases and discovering therapies. Traditionally, mammals have been used as models of cardiac disease, but the cost of generating and maintaining new models is exorbitant, and the studies have very low throughput. In the last decade, the zebrafish has emerged as a tractable model for cardiac diseases, owing to several characteristics that made this animal popular among developmental biologists. Zebrafish fertilization and development are external; embryos can be obtained in high numbers, are cheap and easy to maintain, and can be manipulated to create new genetic models. Moreover, zebrafish exhibit an exceptional ability to regenerate their heart after injury. This review summarizes 25 years of research using the zebrafish to study the heart, from the classical forward screenings to the contemporary methods to model mutations found in patients with cardiac disease. We discuss the advantages and limitations of this model organism and introduce the experimental approaches exploited in zebrafish, including forward and reverse genetics and chemical screenings. Last, we review the models used to induce cardiac injury and essential ideas derived from studying natural regeneration. Studies using zebrafish have the potential to accelerate the discovery of new strategies to treat cardiac diseases.
Collapse
Affiliation(s)
- Juan Manuel González-Rosa
- Cardiovascular Research Center, Massachusetts General Hospital Research Institute, Harvard Medical School, Charlestown, MA
| |
Collapse
|
11
|
Vuong JT, Stein-Merlob AF, Cheng RK, Yang EH. Novel Therapeutics for Anthracycline Induced Cardiotoxicity. Front Cardiovasc Med 2022; 9:863314. [PMID: 35528842 PMCID: PMC9072636 DOI: 10.3389/fcvm.2022.863314] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/14/2022] [Indexed: 01/04/2023] Open
Abstract
Anthracyclines remain an essential component of the treatment of many hematologic and solid organ malignancies, but has important implications on cardiovascular disease. Anthracycline induced cardiotoxicity (AIC) ranges from asymptomatic LV dysfunction to highly morbid end- stage heart failure. As cancer survivorship improves, the detection and treatment of AIC becomes more crucial to improve patient outcomes. Current treatment modalities for AIC have been largely extrapolated from treatment of conventional heart failure, but developing effective therapies specific to AIC is an area of growing research interest. This review summarizes the current evidence behind the use of neurohormonal agents, dexrazoxane, and resynchronization therapy in AIC, evaluates the clinical outcomes of advanced therapy and heart transplantation in AIC, and explores future horizons for treatment utilizing gene therapy, stem cell therapy, and mechanism-specific targets.
Collapse
Affiliation(s)
- Jacqueline T. Vuong
- Department of Medicine, Ronald Reagan UCLA Medical Center, Los Angeles, CA, United States
| | - Ashley F. Stein-Merlob
- Division of Cardiology, Department of Medicine, Ronald Reagan UCLA Medical Center, Los Angeles, CA, United States
| | - Richard K. Cheng
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, United States
| | - Eric H. Yang
- Division of Cardiology, Department of Medicine, Ronald Reagan UCLA Medical Center, Los Angeles, CA, United States
- UCLA Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- *Correspondence: Eric H. Yang,
| |
Collapse
|
12
|
Pharmacological assessment of zebrafish-based cardiotoxicity models. Biomed Pharmacother 2022; 148:112695. [DOI: 10.1016/j.biopha.2022.112695] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/14/2022] [Accepted: 02/02/2022] [Indexed: 01/03/2023] Open
|
13
|
Bowley G, Kugler E, Wilkinson R, Lawrie A, van Eeden F, Chico TJA, Evans PC, Noël ES, Serbanovic-Canic J. Zebrafish as a tractable model of human cardiovascular disease. Br J Pharmacol 2022; 179:900-917. [PMID: 33788282 DOI: 10.1111/bph.15473] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/18/2021] [Accepted: 03/24/2021] [Indexed: 12/17/2022] Open
Abstract
Mammalian models including non-human primates, pigs and rodents have been used extensively to study the mechanisms of cardiovascular disease. However, there is an increasing desire for alternative model systems that provide excellent scientific value while replacing or reducing the use of mammals. Here, we review the use of zebrafish, Danio rerio, to study cardiovascular development and disease. The anatomy and physiology of zebrafish and mammalian cardiovascular systems are compared, and we describe the use of zebrafish models in studying the mechanisms of cardiac (e.g. congenital heart defects, cardiomyopathy, conduction disorders and regeneration) and vascular (endothelial dysfunction and atherosclerosis, lipid metabolism, vascular ageing, neurovascular physiology and stroke) pathologies. We also review the use of zebrafish for studying pharmacological responses to cardiovascular drugs and describe several features of zebrafish that make them a compelling model for in vivo screening of compounds for the treatment cardiovascular disease. LINKED ARTICLES: This article is part of a themed issue on Preclinical Models for Cardiovascular disease research (BJP 75th Anniversary). To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v179.5/issuetoc.
Collapse
Affiliation(s)
- George Bowley
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
- Bateson Centre, University of Sheffield, Sheffield, UK
| | - Elizabeth Kugler
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
- Bateson Centre, University of Sheffield, Sheffield, UK
- Institute of Ophthalmology, Faculty of Brain Sciences, University College London, London, UK
| | - Rob Wilkinson
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Allan Lawrie
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Freek van Eeden
- Bateson Centre, University of Sheffield, Sheffield, UK
- Department of Biomedical Science, University of Sheffield, Sheffield, UK
| | - Tim J A Chico
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
- Bateson Centre, University of Sheffield, Sheffield, UK
| | - Paul C Evans
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
- Bateson Centre, University of Sheffield, Sheffield, UK
| | - Emily S Noël
- Bateson Centre, University of Sheffield, Sheffield, UK
- Department of Biomedical Science, University of Sheffield, Sheffield, UK
| | - Jovana Serbanovic-Canic
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
- Bateson Centre, University of Sheffield, Sheffield, UK
| |
Collapse
|
14
|
Wang Y, Lu X, Wang X, Qiu Q, Zhu P, Ma L, Ma X, Herrmann J, Lin X, Wang W, Xu X. atg7-Based Autophagy Activation Reverses Doxorubicin-Induced Cardiotoxicity. Circ Res 2021; 129:e166-e182. [PMID: 34384247 PMCID: PMC8484060 DOI: 10.1161/circresaha.121.319104] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Yong Wang
- Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
- Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoguang Lu
- Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
- Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoping Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Qi Qiu
- Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
- Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Institute of Clinical Pharmacology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Ping Zhu
- Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
- Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Lin Ma
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xiao Ma
- Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
- Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Joerg Herrmann
- Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Xueying Lin
- Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
- Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Wei Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaolei Xu
- Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
- Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
15
|
Qin Y, Guo T, Wang Z, Zhao Y. The role of iron in doxorubicin-induced cardiotoxicity: recent advances and implication for drug delivery. J Mater Chem B 2021; 9:4793-4803. [PMID: 34059858 DOI: 10.1039/d1tb00551k] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
As an anthracycline antibiotic, doxorubicin (DOX) is one of the most potent and widely used chemotherapeutic agents for treating various types of tumors. Unfortunately, the clinical application of this drug results in severe side effects, particularly dose-dependent cardiotoxicity. There are multiple mechanisms involved with the cardiotoxicity caused by DOX, among which intracellular iron homeostasis plays an essential role based on a recent discovery. In this mini-review, we summarize the clinical features and symptoms of DOX-dependent cardiotoxicity, discuss the correlation between iron and cardiotoxicity, and highlight the involvement of iron-dependent ferroptotic cell death therein. Recent advances in this topic will aid the development of novel DOX delivery systems with reduced adverse effects, and expand the clinical application of anthracycline.
Collapse
Affiliation(s)
- Yan Qin
- School of Pharmaceutical Science & Technology, Tianjin Key Laboratory for Modern Drug Delivery & High Efficiency, and Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, China.
| | | | | | | |
Collapse
|
16
|
Ajarem JS, Hegazy AK, Allam GA, Allam AA, Maodaa SN, Mahmoud AM. Effect of Visnagin on Altered Steroidogenesis and Spermatogenesis, and Testicular Injury Induced by the Heavy Metal Lead. Comb Chem High Throughput Screen 2021; 24:758-766. [PMID: 32957877 DOI: 10.2174/1386207323999200918124639] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/02/2020] [Accepted: 07/03/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Lead (Pb) is an environmental pollutant causing serious health problems, including impairment of reproduction. Visnagin (VIS) is a furanochromone with promising antioxidant and anti-inflammatory effects; however, its protective efficacy against Pb toxicity has not been investigated. OBJECTIVE This study evaluated the protective effect of VIS on Pb reproductive toxicity, impaired steroidogenesis and spermatogenesis, oxidative stress and inflammation. METHODS Rats received VIS (30 or 60 mg/kg) and 50 mg/kg lead acetate for 3 weeks and blood and testes samples were collected. RESULTS Pb intoxication impaired the pituitary-testicular axis (PTA) manifested by the decreased serum levels of gonadotropins and testosterone. Pb decreased sperm count, motility and viability, increased sperm abnormalities, and downregulated the steroidogenesis markers StAR, CYP17A1, 3β-HSD and 17β-HSD in the testis of rats. VIS significantly increased serum gonadotropins and testosterone, alleviated sperm parameters and upregulated steroidogenesis. In addition, VIS decreased pro-inflammatory cytokines, testicular lipid peroxidation and DNA fragmentation, downregulated Bax, and enhanced antioxidants and Bcl-2. CONCLUSION These results demonstrate the protective effect of VIS against Pb reproductive toxicity in rats. VIS improved serum gonadotropins and testosterone, enhanced steroidogenesis and spermatogenesis, and attenuated oxidative injury, inflammation and apoptosis. Therefore, VIS is a promising candidate for the protection against Pb-induced reproduction impairment.
Collapse
Affiliation(s)
- Jamaan S Ajarem
- Department of Zoology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ahmad K Hegazy
- Department of Botany & Microbiology, College of Science, King Saud University, Riyadh11451, Saudi Arabia
| | - Gamal A Allam
- Immunology Section, Department of Microbiology, College of Medicine, Taif University, Taif 21974, Saudi Arabia
| | - Ahmed A Allam
- Department of Zoology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Saleh N Maodaa
- Department of Zoology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ayman M Mahmoud
- Department of Zoology, Faculty of Science, Beni-Suef University, Beni-Suef 62514, Egypt
| |
Collapse
|
17
|
Kim CW, Choi KC. Effects of anticancer drugs on the cardiac mitochondrial toxicity and their underlying mechanisms for novel cardiac protective strategies. Life Sci 2021; 277:119607. [PMID: 33992675 DOI: 10.1016/j.lfs.2021.119607] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 04/29/2021] [Accepted: 05/04/2021] [Indexed: 12/12/2022]
Abstract
Mitochondria are organelles that play a pivotal role in the production of energy in cells, and vital to the maintenance of cellular homeostasis due to the regulation of many biochemical processes. The heart contains a lot of mitochondria because those muscles require a lot of energy to keep supplying blood through the circulatory system, implying that the energy generated from mitochondria is highly dependent. Thus, cardiomyocytes are sensitive to mitochondrial dysfunction and are likely to be targeted by mitochondrial toxic drugs. It has been reported that some anticancer drugs caused unwanted toxicity to mitochondria. Mitochondrial dysfunction is related to aging and the onset of many diseases, such as obesity, diabetes, cancer, cardiovascular and neurodegenerative diseases. Mitochondrial toxic mechanisms can be mainly explained concerning reactive oxygen species (ROS)/redox status, calcium homeostasis, and endoplasmic reticulum stress (ER) stress signaling. The toxic mechanisms of many anticancer drugs have been revealed, but more studying and understanding of the mechanisms of drug-induced mitochondrial toxicity is required to develop mitochondrial toxicity screening system as well as novel cardioprotective strategies for the prevention of cardiac disorders of drugs. This review focuses on the cardiac mitochondrial toxicity of commonly used anticancer drugs, i.e., doxorubicin, mitoxantrone, cisplatin, arsenic trioxide, and cyclophosphamide, and their possible chemopreventive agents that can prevent or alleviate cardiac mitochondrial toxicity.
Collapse
Affiliation(s)
- Cho-Won Kim
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea.
| |
Collapse
|
18
|
Yu X, Guo L, Deng X, Yang F, Tian Y, Liu P, Xu F, Zhang Z, Huang Y. Attenuation of doxorubicin-induced oxidative damage in rat brain by regulating amino acid homeostasis with Astragali Radix. Amino Acids 2021; 53:893-901. [PMID: 33945017 DOI: 10.1007/s00726-021-02992-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 04/21/2021] [Indexed: 01/19/2023]
Abstract
The nervous system disorders caused by doxorubicin (DOX) are among the severe adverse effects that dramatically reduce the quality of life of cancer survivors. Astragali Radix (AR), a popular herbal drug and dietary supplement, is believed to help treat brain diseases by reducing oxidative stress and maintaining metabolic homeostasis. Here we show the protective effects of AR against DOX-induced oxidative damage in rat brain via regulating amino acid homeostasis. By constructing a clinically relevant low-dose DOX-induced toxicity rat model, we first performed an untargeted metabolomics analysis to discover specific metabolic features in the brain after DOX treatment and AR co-treatment. It was found that the amino acid (AA) metabolism pathways altered most significantly. To accurately characterize the brain AA profile, we established a sensitive, fast, and reproducible hydrophilic interaction chromatography-tandem mass spectrometry method for the simultaneous quantification of 22 AAs. The targeted analysis further confirmed the changes of AAs between different groups of rat brain. Specifically, the levels of six AAs, including glutamate, glycine, serine, alanine, citrulline, and ornithine, correlated (Pearson |r| > 0.47, p < 0.05) with the brain oxidative damage that was caused by DOX and rescued by AR. These findings present that AAs are among the regulatory targets of DOX-induced brain toxicity, and AR is a promising therapeutic agent for it.
Collapse
Affiliation(s)
- Xinyue Yu
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing, 210009, China.,Department of Pharmaceutical Analysis, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Linling Guo
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing, 210009, China.,Department of Pharmaceutical Analysis, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Xiaoying Deng
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing, 210009, China
| | - Fang Yang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing, 210009, China
| | - Yuan Tian
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing, 210009, China.,Department of Pharmaceutical Analysis, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Peifang Liu
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Fengguo Xu
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing, 210009, China.,Department of Pharmaceutical Analysis, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Zunjian Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing, 210009, China.
| | - Yin Huang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing, 210009, China. .,Department of Pharmaceutical Analysis, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
19
|
Asnani A, Moslehi JJ, Adhikari BB, Baik AH, Beyer AM, de Boer RA, Ghigo A, Grumbach IM, Jain S, Zhu H. Preclinical Models of Cancer Therapy-Associated Cardiovascular Toxicity: A Scientific Statement From the American Heart Association. Circ Res 2021; 129:e21-e34. [PMID: 33934611 DOI: 10.1161/res.0000000000000473] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Although cardiovascular toxicity from traditional chemotherapies has been well recognized for decades, the recent explosion of effective novel targeted cancer therapies with cardiovascular sequelae has driven the emergence of cardio-oncology as a new clinical and research field. Cardiovascular toxicity associated with cancer therapy can manifest as a broad range of potentially life-threatening complications, including heart failure, arrhythmia, myocarditis, and vascular events. Beyond toxicology, the intersection of cancer and heart disease has blossomed to include discovery of genetic and environmental risk factors that predispose to both. There is a pressing need to understand the underlying molecular mechanisms of cardiovascular toxicity to improve outcomes in patients with cancer. Preclinical cardiovascular models, ranging from cellular assays to large animals, serve as the foundation for mechanistic studies, with the ultimate goal of identifying biologically sound biomarkers and cardioprotective therapies that allow the optimal use of cancer treatments while minimizing toxicities. Given that novel cancer therapies target specific pathways integral to normal cardiovascular homeostasis, a better mechanistic understanding of toxicity may provide insights into fundamental pathways that lead to cardiovascular disease when dysregulated. The goal of this scientific statement is to summarize the strengths and weaknesses of preclinical models of cancer therapy-associated cardiovascular toxicity, to highlight overlapping mechanisms driving cancer and cardiovascular disease, and to discuss opportunities to leverage cardio-oncology models to address important mechanistic questions relevant to all patients with cardiovascular disease, including those with and without cancer.
Collapse
|
20
|
CYP1B1 as a therapeutic target in cardio-oncology. Clin Sci (Lond) 2021; 134:2897-2927. [PMID: 33185690 PMCID: PMC7672255 DOI: 10.1042/cs20200310] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/12/2020] [Accepted: 10/28/2020] [Indexed: 02/06/2023]
Abstract
Cardiovascular complications have been frequently reported in cancer patients and survivors, mainly because of various cardiotoxic cancer treatments. Despite the known cardiovascular toxic effects of these treatments, they are still clinically used because of their effectiveness as anti-cancer agents. In this review, we discuss the growing body of evidence suggesting that inhibition of the cytochrome P450 1B1 enzyme (CYP1B1) can be a promising therapeutic strategy that has the potential to prevent cancer treatment-induced cardiovascular complications without reducing their anti-cancer effects. CYP1B1 is an extrahepatic enzyme that is expressed in cardiovascular tissues and overexpressed in different types of cancers. A growing body of evidence is demonstrating a detrimental role of CYP1B1 in both cardiovascular diseases and cancer, via perturbed metabolism of endogenous compounds, production of carcinogenic metabolites, DNA adduct formation, and generation of reactive oxygen species (ROS). Several chemotherapeutic agents have been shown to induce CYP1B1 in cardiovascular and cancer cells, possibly via activating the Aryl hydrocarbon Receptor (AhR), ROS generation, and inflammatory cytokines. Induction of CYP1B1 is detrimental in many ways. First, it can induce or exacerbate cancer treatment-induced cardiovascular complications. Second, it may lead to significant chemo/radio-resistance, undermining both the safety and effectiveness of cancer treatments. Therefore, numerous preclinical studies demonstrate that inhibition of CYP1B1 protects against chemotherapy-induced cardiotoxicity and prevents chemo- and radio-resistance. Most of these studies have utilized phytochemicals to inhibit CYP1B1. Since phytochemicals have multiple targets, future studies are needed to discern the specific contribution of CYP1B1 to the cardioprotective and chemo/radio-sensitizing effects of these phytochemicals.
Collapse
|
21
|
Lane S, More LA, Asnani A. Zebrafish Models of Cancer Therapy-Induced Cardiovascular Toxicity. J Cardiovasc Dev Dis 2021; 8:jcdd8020008. [PMID: 33499052 PMCID: PMC7911266 DOI: 10.3390/jcdd8020008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/11/2021] [Accepted: 01/20/2021] [Indexed: 12/13/2022] Open
Abstract
Purpose of review: Both traditional and novel cancer therapies can cause cardiovascular toxicity in patients. In vivo models integrating both cardiovascular and cancer phenotypes allow for the study of on- and off-target mechanisms of toxicity arising from these agents. The zebrafish is the optimal whole organism model to screen for cardiotoxicity in a high throughput manner, while simultaneously assessing the role of cardiotoxicity pathways on the cancer therapy’s antitumor effect. Here we highlight established zebrafish models of human cardiovascular disease and cancer, the unique advantages of zebrafish to study mechanisms of cancer therapy-associated cardiovascular toxicity, and finally, important limitations to consider when using the zebrafish to study toxicity. Recent findings: Cancer therapy-associated cardiovascular toxicities range from cardiomyopathy with traditional agents to arrhythmias and thrombotic complications associated with newer targeted therapies. The zebrafish can be used to identify novel therapeutic strategies that selectively protect the heart from cancer therapy without affecting antitumor activity. Advances in genome editing technology have enabled the creation of several transgenic zebrafish lines valuable to the study of cardiovascular and cancer pathophysiology. Summary: The high degree of genetic conservation between zebrafish and humans, as well as the ability to recapitulate cardiotoxic phenotypes observed in patients with cancer, make the zebrafish an effective model to study cancer therapy-associated cardiovascular toxicity. Though this model provides several key benefits over existing in vitro and in vivo models, limitations of the zebrafish model include the early developmental stage required for most high-throughput applications.
Collapse
Affiliation(s)
- Sarah Lane
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; (S.L.); (L.A.M.)
| | - Luis Alberto More
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; (S.L.); (L.A.M.)
| | - Aarti Asnani
- CardioVascular Institute, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; (S.L.); (L.A.M.)
- Harvard Medical School, Boston, MA 02115, USA
- Correspondence:
| |
Collapse
|
22
|
Hoeger CW, Turissini C, Asnani A. Doxorubicin Cardiotoxicity: Pathophysiology Updates. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2020. [DOI: 10.1007/s11936-020-00842-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
23
|
Khodadust R, Alpsoy A, Ünsoy G, GÜndÜz U. Poly (I:C)- and doxorubicin-loaded magnetic dendrimeric nanoparticles affect the apoptosis-related gene expressions in MCF-7 cells. ACTA ACUST UNITED AC 2020; 44:133-144. [PMID: 32922121 PMCID: PMC7478132 DOI: 10.3906/biy-1912-71] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Use of nanoparticles as drug carrier vectors has great potential to circumvent the limitations associated with chemotherapy, including drug resistance and destructive side effects. For this purpose, magnetic generation 4 dendrimeric nanoparticles were prepared to carry chemotherapeutic agent doxorubicin (G4-DOX) and immune modulator polyinosinic:polycytidylic acid [Poly(I:C)]. As previously reported, DOX and Poly(I:C) was loaded onto G4 nanoparticles (PIC-G4-DOX). Cellular internalization study using confocal microscopy demonstrated high levels of cellular internalization of PIC-G4-DOX nanoparticles by MCF-7 cells. This resulted in higher efficacy of PIC-G4-DOX nanoparticles in killing MCF-7 breast cancer cells. Alteration in the expression levels of selected genes was determined by RT-qPCR analyses. Proapoptotic NOXA, PUMA, and BAX genes were upregulated, and SURVIVIN, APOLLON, and BCL-2 genes were downregulated, indicating the cell-killing effectiveness of PIC-G4-DOX nanoparticles. Gene expression analysis provided some insights into the possible molecular mechanisms on cytotoxicity of DOX and Poly(I:C) delivered through G4 magnetic nanoparticles. The results demonstrated that PIC-G4-DOX can be useful for targeted delivery affecting apoptotic pathways, resulting in an advanced degree of cancer-cell–killing. They are promising for targeting cancer-cells because of their stability, biocompatibility, higher internalization, and toxicity.
Collapse
Affiliation(s)
- Rouhollah Khodadust
- Department of Biotechnology, Middle East Technical University, Ankara Turkey.,Department of Biotechnology, Hamidiye Health Science Institute, University of Health Science-Turkey, İstanbul Turkey
| | - Aktan Alpsoy
- Department of Biological Sciences, Middle East Technical University, Ankara Turkey
| | - Gözde Ünsoy
- Department of Biotechnology, Middle East Technical University, Ankara Turkey
| | - Ufuk GÜndÜz
- Department of Biotechnology, Middle East Technical University, Ankara Turkey.,Department of Biological Sciences, Middle East Technical University, Ankara Turkey
| |
Collapse
|
24
|
Fu HR, Li XS, Zhang YH, Feng BB, Pan LH. Visnagin ameliorates myocardial ischemia/reperfusion injury through the promotion of autophagy and the inhibition of apoptosis. Eur J Histochem 2020; 64. [PMID: 32909422 PMCID: PMC7482183 DOI: 10.4081/ejh.2020.3131] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 05/30/2020] [Indexed: 01/31/2023] Open
Abstract
Visnagin is a furanochromone and one of the main compounds of Ammi visnaga L. that had been used to treat nephrolithiasis in Ancient Egypt. Nowadays, visnagin was widely used to treat angina pectoris, urolithiasis and hypertriglyceridemia. The potential mechanisms of visnagin involved in inflammation and cardiovascular disease were also identified. But the protective effect of visnagin on myocardial ischemia/reperfusion injury has not been confirmed. Our aim was, for the first time, to investigate the potential protective effect of visnagin on cardiac function after myocardial ischemia-reperfusion injury in a rat model, and to identify its underlying mechanism involving the inhibition of apoptosis and induction of autophagy. Thirty SD rats were randomly divided into sham group, ischemia/reperfusion group (IR), ischemia/reperfusion with visnagin (IR + visnagin) group. Myocardial ischemia/Reperfusion injury model was established. Hemodynamic measurements and echocardiography were used to analyze cardiac function, TUNEL staining and caspase activity, LC3 dots were detected with immunofluorescence staining, LC3 expression was evaluated by western blot analysis, transmission electron microscopy (TEM) was used to detect autophagosomes. Compared with the sham group and visnagin group, the cardiac dysfunction, LC3II, autophagy flow in the IR+ visnagin group increased significantly (P<0.01), but the activity of caspase-3 and caspase-9 and the apoptotic in the IR + visnagin group decreased significantly (P<0.01). In conclusion, visnagin may play a protective role in ischemia/reperfusion injury by inducing autophagy and reducing apoptosis.
Collapse
Affiliation(s)
- Hai-Rong Fu
- Department of Basic Medicine, Three Gorges Medical College, Chongqing.
| | - Xiao-Shan Li
- Department of Basic Medicine, Three Gorges Medical College, Chongqing.
| | - Yong-Hui Zhang
- Department of Basic Medicine, Three Gorges Medical College, Chongqing.
| | - Bin-Bin Feng
- Department of Pharmacy, Three Gorges Medical College, Chongqing.
| | - Lian-Hong Pan
- Center for Natural Anti-tumor Medicine Engineering, Three Gorges Medical College, Chongqing.
| |
Collapse
|
25
|
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
|
26
|
Raber I, Asnani A. Cardioprotection in cancer therapy: novel insights with anthracyclines. Cardiovasc Res 2020; 115:915-921. [PMID: 30726931 DOI: 10.1093/cvr/cvz023] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 12/12/2018] [Accepted: 01/25/2019] [Indexed: 12/23/2022] Open
Abstract
Anthracyclines are a class of antineoplastic agents that remain critical to modern-day cancer treatment. However, their propensity to cause cardiotoxic effects limits their use and can cause increased morbidity and mortality among patients with cancer. Currently available methods to minimize the impact of anthracycline cardiotoxicity have not been widely successful. While it is largely accepted that the generation of oxygen radicals contributes to the development of anthracycline cardiotoxicity, the exact mechanisms of cardiomyocyte injury remain unclear. In this review, we discuss the current state of basic and translational research on the cardiotoxic mechanisms of anthracyclines that have led to the discovery of new therapeutic targets. Pending validation in patient populations, these recent advances have the potential to be translated into clinical approaches that will minimize anthracycline cardiotoxicity and improve outcomes in cancer survivors.
Collapse
Affiliation(s)
- Inbar Raber
- Department of Internal Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Aarti Asnani
- Harvard Medical School, Boston, MA, USA.,CardioVascular Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
27
|
Coumarins as Modulators of the Keap1/Nrf2/ARE Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:1675957. [PMID: 32377290 PMCID: PMC7196981 DOI: 10.1155/2020/1675957] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/14/2020] [Accepted: 04/06/2020] [Indexed: 12/30/2022]
Abstract
The Keap1/Nrf2/ARE system is a central defensive mechanism against oxidative stress which plays a key role in the pathogenesis and progression of many diseases. Nrf2 is a redox-sensitive transcription factor controlling a variety of downstream antioxidant and cytodefensive genes. Nrf2 has a powerful anti-inflammatory activity mediated via modulating NF-κB. Therefore, pharmacological activation of Nrf2 is a promising therapeutic strategy for the treatment/prevention of several diseases that are underlined by both oxidative stress and inflammation. Coumarins are natural products with promising pharmacological activities, including antioxidant, anticancer, antimicrobial, and anti-inflammatory efficacies. Coumarins are found in many plants, fungi, and bacteria and have been widely used as complementary and alternative medicines. Some coumarins have shown an ability to activate Nrf2 signaling in different cells and animal models. The present review compiles the research findings of seventeen coumarin derivatives of plant origin (imperatorin, visnagin, urolithin B, urolithin A, scopoletin, esculin, esculetin, umbelliferone, fraxetin, fraxin, daphnetin, anomalin, wedelolactone, glycycoumarin, osthole, hydrangenol, and isoimperatorin) as antioxidant and anti-inflammatory agents, emphasizing the role of Nrf2 activation in their pharmacological activities. Additionally, molecular docking simulations were utilized to investigate the potential binding mode of these coumarins with Keap1 as a strategy to disrupt Keap1/Nrf2 protein-protein interaction and activate Nrf2 signaling.
Collapse
|
28
|
Lam PY, Kutchukian P, Anand R, Imbriglio J, Andrews C, Padilla H, Vohra A, Lane S, Parker DL, Cornella Taracido I, Johns DG, Beerens M, MacRae CA, Caldwell JP, Sorota S, Asnani A, Peterson RT. Cyp1 Inhibition Prevents Doxorubicin-Induced Cardiomyopathy in a Zebrafish Heart-Failure Model. Chembiochem 2020; 21:1905-1910. [PMID: 32003101 DOI: 10.1002/cbic.201900741] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Indexed: 12/19/2022]
Abstract
Doxorubicin is a highly effective chemotherapy agent used to treat many common malignancies. However, its use is limited by cardiotoxicity, and cumulative doses exponentially increase the risk of heart failure. To identify novel heart failure treatment targets, a zebrafish model of doxorubicin-induced cardiomyopathy was previously established for small-molecule screening. Using this model, several small molecules that prevent doxorubicin-induced cardiotoxicity both in zebrafish and in mouse models have previously been identified. In this study, exploration of doxorubicin cardiotoxicity is expanded by screening 2271 small molecules from a proprietary, target-annotated tool compound collection. It is found that 120 small molecules can prevent doxorubicin-induced cardiotoxicity, including 7 highly effective compounds. Of these, all seven exhibited inhibitory activity towards cytochrome P450 family 1 (CYP1). These results are consistent with previous findings, in which visnagin, a CYP1 inhibitor, also prevents doxorubicin-induced cardiotoxicity. Importantly, genetic mutation of cyp1a protected zebrafish against doxorubicin-induced cardiotoxicity phenotypes. Together, these results provide strong evidence that CYP1 is an important contributor to doxorubicin-induced cardiotoxicity and highlight the CYP1 pathway as a candidate therapeutic target for clinical cardioprotection.
Collapse
Affiliation(s)
- Pui-Ying Lam
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, 30 S 2000 E, Salt Lake City, UT, 84112, USA
| | - Peter Kutchukian
- Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, MA, 02115, USA
| | - Rajan Anand
- Merck & Co., Inc, 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Jason Imbriglio
- Merck & Co., Inc, 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | | | - Hugo Padilla
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, 30 S 2000 E, Salt Lake City, UT, 84112, USA
| | - Anita Vohra
- CardioVascular Institute, Beth Israel Deaconess Medical Center, and, Harvard Medical School, Boston, MA, 02115, USA
| | - Sarah Lane
- CardioVascular Institute, Beth Israel Deaconess Medical Center, and, Harvard Medical School, Boston, MA, 02115, USA
| | - Dann L Parker
- Merck & Co., Inc, 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | | | - Douglas G Johns
- Merck & Co., Inc, 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Manu Beerens
- Department of Cardiovascular Medicine, Genetics and Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Calum A MacRae
- Department of Cardiovascular Medicine, Genetics and Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - John P Caldwell
- Merck & Co., Inc, 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Steve Sorota
- Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, MA, 02115, USA
| | - Aarti Asnani
- CardioVascular Institute, Beth Israel Deaconess Medical Center, and, Harvard Medical School, Boston, MA, 02115, USA
| | - Randall T Peterson
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, 30 S 2000 E, Salt Lake City, UT, 84112, USA
| |
Collapse
|
29
|
Grant MK, Abdelgawad IY, Lewis CA, Zordoky BN. Sexual Dimorphism in Doxorubicin-induced Systemic Inflammation: Implications for Hepatic Cytochrome P450 Regulation. Int J Mol Sci 2020; 21:ijms21041279. [PMID: 32074957 PMCID: PMC7072970 DOI: 10.3390/ijms21041279] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/03/2020] [Accepted: 02/11/2020] [Indexed: 12/20/2022] Open
Abstract
Doxorubicin (DOX) is an effective chemotherapeutic agent used to treat a wide variety of malignancies. In addition to its multi-organ toxicity, DOX treatment has been shown to induce systemic inflammation in patients and experimental animals. Inflammation alters the expression of hepatic cytochrome P450 (CYP) enzymes, which play important roles in drug metabolism and DOX-induced toxicity. Significant sex differences have been reported in DOX-induced toxicity; however, sex differences in DOX-induced systemic inflammation and the potential effects on hepatic CYP expression have not been determined. In the current work, male and female C57Bl/6 mice were administered DOX (20 mg/kg by intraperitoneal injection), and groups of mice were sacrificed 24 and 72 h after DOX administration. DOX elicited a systemic inflammatory response in both male and female mice, but the inflammatory response was stronger in male mice. DOX altered the expression of hepatic CYP isoforms in a sex-dependent manner. Most notably, inhibition of Cyp2c29 and Cyp2e1 was stronger in male than in female mice, which paralleled the sex differences in systemic inflammation. Therefore, sex differences in DOX-induced systemic inflammation may lead to sexually dimorphic drug interactions, in addition to contributing to the previously reported sexual dimorphism in specific DOX-induced organ toxicity.
Collapse
|
30
|
Chen J, Ding Y, Chen M, Gau J, Jen N, Nahal C, Tu S, Chen C, Zhou S, Chang CC, Lyu J, Xu X, Hsiai TK, Packard RRS. Displacement analysis of myocardial mechanical deformation (DIAMOND) reveals segmental susceptibility to doxorubicin-induced injury and regeneration. JCI Insight 2019; 4:125362. [PMID: 30996130 PMCID: PMC6538350 DOI: 10.1172/jci.insight.125362] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 02/27/2019] [Indexed: 12/27/2022] Open
Abstract
Zebrafish are increasingly utilized to model cardiomyopathies and regeneration. Current methods evaluating cardiac function have known limitations, fail to reliably detect focal mechanics, and are not readily feasible in zebrafish. We developed a semiautomated, open-source method - displacement analysis of myocardial mechanical deformation (DIAMOND) - for quantitative assessment of 4D segmental cardiac function. We imaged transgenic embryonic zebrafish in vivo using a light-sheet fluorescence microscopy system with 4D cardiac motion synchronization. Our method permits the derivation of a transformation matrix to quantify the time-dependent 3D displacement of segmental myocardial mass centroids. Through treatment with doxorubicin, and by chemically and genetically manipulating the myocardial injury-activated Notch signaling pathway, we used DIAMOND to demonstrate that basal ventricular segments adjacent to the atrioventricular canal display the highest 3D displacement and are also the most susceptible to doxorubicin-induced injury. Thus, DIAMOND provides biomechanical insights into in vivo segmental cardiac function scalable to high-throughput research applications.
Collapse
Affiliation(s)
- Junjie Chen
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences
| | - Yichen Ding
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine
| | - Michael Chen
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences
| | - Jonathan Gau
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine
| | - Nelson Jen
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine
| | - Chadi Nahal
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences
| | - Sally Tu
- Department of Neuroscience, College of Letters and Science, University of California, Los Angeles, California, USA
| | - Cynthia Chen
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences
| | - Steve Zhou
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine
| | - Chih-Chiang Chang
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences
| | - Jintian Lyu
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences
| | - Xiaolei Xu
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Tzung K. Hsiai
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine
| | | |
Collapse
|
31
|
Pasari LP, Khurana A, Anchi P, Aslam Saifi M, Annaldas S, Godugu C. Visnagin attenuates acute pancreatitis via Nrf2/NFκB pathway and abrogates associated multiple organ dysfunction. Biomed Pharmacother 2019; 112:108629. [PMID: 30798137 DOI: 10.1016/j.biopha.2019.108629] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 01/13/2019] [Accepted: 01/28/2019] [Indexed: 12/14/2022] Open
Abstract
Acute pancreatitis (AP) is an exocrine dysfunction of the pancreas where oxidative stress and inflammatory cytokines play a key role in induction and progression of the disease. Studies have demonstrated that antioxidant phytochemicals have been effective in improving pancreatitis condition, but there are no clinically approved drugs till date. Our study aims to assess the preventive activity of visnagin, a novel phytochemical isolated from Ammi visnaga against cerulein induced AP. Male Swiss albino mice were divided into six groups (n = 6, each group) comprising of normal control, cerulein control, seven day pre-treatment with visnagin at three dose levels; visnagin low dose (10 mg/kg), visnagin mid dose (30 mg/kg), visnagin high dose (60 mg/kg) and visnagin control (60 mg/kg). AP was induced by six injections of cerulein (50 μg/kg, i.p.) on the 7th day and the animals were sacrificed after 6 h of last cerulein dose. Various markers of pancreatic function, oxidative stress and inflammation were assessed. Visnagin was found to be effective in reducing plasma amylase and lipase levels, reduced cerulein induced oxidative stress. Visnagin dose dependently decreased the expression of IL-1β, IL-6, TNF-α and IL-17. It attenuated the levels of nuclear p65-NFκB. Visnagin improved the antioxidant defence by improving Nrf2 expression and halted pancreatic inflammation by suppressing NFκB and nitrotyrosine expression in the acinar cells. Further, it attenuated the expression of markers of multiple organ dysfunction syndrome and reduced inflammatory cytokines in lungs and intestine. Cumulatively, these findings indicate that visnagin has substantial potential to prevent cerulein induced AP.
Collapse
Affiliation(s)
- Lakshmi Priya Pasari
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana State, India
| | - Amit Khurana
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana State, India
| | - Pratibha Anchi
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana State, India
| | - Mohd Aslam Saifi
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana State, India
| | - Shivaraju Annaldas
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana State, India
| | - Chandraiah Godugu
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana State, India.
| |
Collapse
|
32
|
Abstract
PURPOSE OF REVIEW In this article, we review current and emerging approaches to biomarker discovery to facilitate early diagnosis of cancer therapy-associated cardiovascular toxicity. RECENT FINDINGS Although small studies have demonstrated an association between established biomarkers of cardiac injury (troponins and brain natriuretic peptide) and acute or subacute cardiotoxicity, there is insufficient evidence to support their use in routine clinical care. Preclinical studies to define the molecular mechanisms of cardiotoxicity, as well as the use of unbiased "omics" techniques in small patient cohorts, have yielded promising candidate biomarkers that have the potential to enrich current risk stratification algorithms. New biomarkers of cardiotoxicity have the potential to improve patient outcomes in cardio-oncology. Further studies are needed to assess the clinical relevance of molecular mechanisms described in animal models. Similarly, findings from "omics" platforms require validation in large patient cohorts before they can be incorporated into everyday practice.
Collapse
|