1
|
Tungalag T, Kang HS, Yang DK. Sinapic Acid Ameliorates Doxorubicin-Induced Cardiotoxicity in H9c2 Cardiomyoblasts by Inhibiting Oxidative Stress Through Activation of the Nrf2 Signaling Pathway. Antioxidants (Basel) 2025; 14:337. [PMID: 40227457 PMCID: PMC11939272 DOI: 10.3390/antiox14030337] [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: 01/09/2025] [Revised: 03/07/2025] [Accepted: 03/12/2025] [Indexed: 04/15/2025] Open
Abstract
The use of doxorubicin (Dox) is restricted because of its cardiotoxicity, which poses a significant mortality risk for cancer patients, despite being a highly effective antibiotic for treating various types of cancer. Therefore, identifying substances or developing preventive strategies against Dox-induced cardiotoxicity is crucial. This study was conducted to determine whether sinapic acid (SA), a phenolic compound with a range of pharmacological effects, could protect against Dox-induced cardiotoxicity in H9c2 cardiomyoblasts. To investigate the preventive effect of SA, H9c2 cardiomyoblasts treated with Dox were pretreated with SA at various concentrations. SA effectively rescued the cells from Dox-induced cardiotoxicity. Additionally, SA significantly reduced oxidative stress by inhibiting mitochondrial dysfunction and endoplasmic reticulum stress. SA also suppressed the expression of MAPK proteins. As for the underlying mechanism of SA's protective effect against Dox-induced cardiotoxicity, SA activated nuclear factor erythroid-2-related factor (Nrf2) by facilitating its movement from the cytosol to the nucleus and increasing the expression of its target antioxidative genes. In summary, this study demonstrated that SA protects H9c2 cardiomyoblasts from Dox-induced cardiotoxicity by inhibiting oxidative stress by the activation of Nrf2-related signaling pathway. Our findings enhance the development of therapeutic strategies to mitigate cardiac toxicity caused by Dox, highlighting the potential antioxidant effect of SA in Dox-treated H9c2 cardiomyoblasts.
Collapse
Affiliation(s)
- Tsendsuren Tungalag
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Jeonbuk National University, Iksan 54596, Jeollabuk-do, Republic of Korea;
| | - Hyung-Sub Kang
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Jeonbuk National University, Iksan 54596, Jeollabuk-do, Republic of Korea;
| | - Dong Kwon Yang
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Jeonbuk National University, Iksan 54596, Jeollabuk-do, Republic of Korea;
- Biosafety Research Institute and College of Veterinary Medicine, Jeonbuk National University, Iksan 54596, Jeollabuk-do, Republic of Korea
| |
Collapse
|
2
|
Tiwari V, Salgar S, Jorvekar SB, Kumbhar BM, Arava SK, Borkar RM, Banerjee SK. Doxorubicin-induced phosphorylation of lamin A/C enhances DNMT1 and activates cardiomyocyte death via suppressing GATA-4 and Bcl-xL in rat heart. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167692. [PMID: 39864225 DOI: 10.1016/j.bbadis.2025.167692] [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: 08/25/2024] [Revised: 01/21/2025] [Accepted: 01/21/2025] [Indexed: 01/28/2025]
Abstract
Cardiotoxic effect of Doxorubicin (Dox) limits its clinical application. Previously, we reported that Dox induces phosphorylation of lamin A/C (pS22 lamin A/C), increased nuclear size, damage to the nuclear membrane, and cell death. However, the activation of signalling pathway during this event remains elusive, and it is unclear whether increased phospho-lamin A/C activates the cell death pathway in heart. Here, we demonstrated that Dox-induced lamin A/C phosphorylation causes apoptotic cell death. Increased levels of reactive oxygen species (ROS), DNA methylation and apoptosis markers (Bax, Bid, caspase 3 and caspase 9) were observed in Dox-exposed H9c2 cells. Nuclear membrane damage due to increased pS22 lamin A/C causes increased DNMT1 and DNA methylation followed by reduced expression of GATA-4 and Bcl-xL in Dox-treated H9c2 cells and rat hearts. Further, increased mRNA expression of DNMT1 and reduced expression of GATA-4 and Bcl-xL was observed in H9c2 cells after knocking down of lamin A/C expression. Previously, we reported that N-acetylcysteine improves lamin A/C levels and maintain nuclear membrane integrity. Similarly, in this study Astaxanthin (Ast), a membrane-specific antioxidant, reduces the expression of DNMT1 and phospho-lamin A/C levels; increases mRNA expression of GATA-4 and Bcl-xL; reduces ROS levels and DNA leakage in Dox-treated H9c2 cells and rat hearts. Ast also improves the cardiac structure and function in Dox-treated rats. In conclusion, Dox exposure in cardiomyoblasts and hearts causes cell death by increasing the pS22 lamin A/C, DNA methylation and reducing the expression GATA-4 and Bcl-xL. This study provides a novel pathway for Dox-induced cardiotoxicity and a possible therapeutic approach to reduce it.
Collapse
Affiliation(s)
- Vikas Tiwari
- Department of Biotechnology, National institute of Pharmaceutical Education and Research (NIPER), Guwahati, India
| | - Sanjay Salgar
- Department of Biotechnology, National institute of Pharmaceutical Education and Research (NIPER), Guwahati, India
| | - Sachin B Jorvekar
- Department of Pharmaceutical Analysis, National institute of Pharmaceutical Education and Research (NIPER), Guwahati, India
| | - Bhagyashri Manoj Kumbhar
- Department of Biotechnology, National institute of Pharmaceutical Education and Research (NIPER), Guwahati, India
| | - Sudheer K Arava
- Department of Pathology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Roshan M Borkar
- Department of Pharmaceutical Analysis, National institute of Pharmaceutical Education and Research (NIPER), Guwahati, India
| | - Sanjay K Banerjee
- Department of Biotechnology, National institute of Pharmaceutical Education and Research (NIPER), Guwahati, India.
| |
Collapse
|
3
|
Rezgui A, Tachour RA, Layaida H, Derguine R, Hab FZ, Benmanseur A, Matougui B, Agred R, Sobhi W. Doxorubicin inhibits SIRT2 and NF-kB p65 phosphorylation in Brest cell-line cancer. Biochem Biophys Res Commun 2025; 743:151162. [PMID: 39689645 DOI: 10.1016/j.bbrc.2024.151162] [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: 09/04/2024] [Revised: 12/04/2024] [Accepted: 12/09/2024] [Indexed: 12/19/2024]
Abstract
Doxorubicin (DOXO) is a widely used anti-cancer agent, yet the precise mechanism underlying the induction of tumor cell death remains unclear. This study aimed to elucidate new mechanisms by which doxorubicin induces apoptosis in the EMT6 mouse breast carcinoma cell line. The role of doxorubicin was assessed using the XTT assay. The assessment of oxidative stress markers, alongside the analysis of SIRT2 and NF-κB p65 (RelA) phosphorylation inhibition, was conducted. In silico studies, including density functional theory (DFT) calculations and molecular docking simulations, were employed to characterize the molecular interactions between doxorubicin and SIRT2. Additionally, doxorubicin was assessed for its capacity to modulate gene expression and associated pathways using multiple bioinformatics tools and web-based platforms. Our finding indicates that Doxorubicin induced apoptosis in EMT6 cells with an IC50 of 8,32 μM. At lower concentrations, doxorubicin enhances the oxidative balance and promotes cell viability. At high concentrations, doxorubicin inhibits SIRT2. Furthermore, an experimental investigation revealed that doxorubicin inhibits RelA phosphorylation. The results also showed that doxorubicin modulated the expression of 19 genes involved in different pathways and several transcription factors. The results of implementing the gene set with SIRT2 and RELA consolidated the experimental results. In conclusion, Doxorubicin was observed to induce EMT6 apoptosis through the inhibition of SIRT2 and RelA proteins. The outcomes of both experimental and bioinformatic studies provide a novel perspective on the biological effects of doxorubicin and underscore the potential of inhibiting the SIRT2-RelA axis as a promising biological target for cancer therapy.
Collapse
Affiliation(s)
- Abdelmalek Rezgui
- Biotechnology Research Center (CRBt), Ali Mendjli, Constantine, 25000, Algeria
| | - Rechda Amel Tachour
- Laboratory of Plant Biotechnology and Ethnobotany, Faculty of Nature and Life Sciences, University of Bejaia, Bejaia, 6000, Algeria
| | - Houdhaifa Layaida
- Laboratoire d'Electrochimie des Matériaux Moléulaires et des Complexes (LEMMC), Faculté de Technologie, Université Ferhat Abbas- Sétif 1, Setif, Algeria
| | - Rania Derguine
- Laboratory of Applied Biochemistry, Faculty of Nature and Life Sciences, University of Ferhat Abbas Setif-1, Setif, 19000, Algeria
| | - Fatma Zahra Hab
- Laboratory of Plant Biotechnology and Ethnobotany, Faculty of Nature and Life Sciences, University of Bejaia, Bejaia, 6000, Algeria
| | - Anfel Benmanseur
- Laboratory of Plant Biotechnology and Ethnobotany, Faculty of Nature and Life Sciences, University of Bejaia, Bejaia, 6000, Algeria
| | - Brahim Matougui
- Biotechnology Research Center (CRBt), Ali Mendjli, Constantine, 25000, Algeria
| | - Rym Agred
- Biotechnology Research Center (CRBt), Ali Mendjli, Constantine, 25000, Algeria
| | - Widad Sobhi
- Biotechnology Research Center (CRBt), Ali Mendjli, Constantine, 25000, Algeria.
| |
Collapse
|
4
|
Chen W, Byun J, Kang HC, Lee HS, Lee JY, Kwon YJ, Cho YY. Karyoptosis as a novel type of UVB-induced regulated cell death. Free Radic Res 2024; 58:796-810. [PMID: 39625813 DOI: 10.1080/10715762.2024.2433986] [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: 08/01/2024] [Revised: 11/14/2024] [Accepted: 11/20/2024] [Indexed: 12/28/2024]
Abstract
Karyoptosis is a type of regulated cell death (RCD) characterized by explosive nuclear rupture caused by a loss of nuclear membrane integrity, resulting in the release of genomic DNA and other nuclear components into the cytosol and extracellular environment. The mechanism underlying karyoptosis involves a delicate balance between the following forces: the expansion force exerted by the tightly packed DNA in the nucleus, the resistance provided by the nuclear lamina at the inner nuclear membrane (INM), and the tensile force from the cytoskeleton that helps position the nucleus at the center of the cytoplasm, allowing it to remain maximally expanded. In addition, CREB3, a type II integral membrane protein with DNA-binding ability, tethers chromatin to the INM, providing a tightening force through chromatin interactions that prevent nuclear membrane rupture. UVB radiation can trigger this process, inducing CREB3-FL cleavage and producing CREB3-CF. Therefore, UVB acts as an intrinsic factor in the induction of karyoptosis. Importantly, biochemical analysis of RCD markers shows that karyoptosis is distinct from other forms of cell death, such as apoptosis, autophagy, necroptosis, and pyroptosis. This review explores the mechanisms involved in maintaining nuclear membrane integrity and the role of CREB3 in triggering karyoptosis and provides brief suggestions on the potential implications for targeting cancer cells.
Collapse
Affiliation(s)
- Weidong Chen
- BK21-Four, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, South Korea
| | - Jiin Byun
- BK21-Four, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, South Korea
| | - Han Chang Kang
- College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, South Korea
| | - Hye Suk Lee
- BK21-Four, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, South Korea
| | - Joo Young Lee
- BK21-Four, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, South Korea
| | - Young Jik Kwon
- College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, South Korea
- Department of Pharmaceutical Sciences, University of California, Irvine, CA, USA
| | - Yong-Yeon Cho
- BK21-Four, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, South Korea
| |
Collapse
|
5
|
Das D, Jothimani G, Banerjee A, Duttaroy AK, Pathak S. The cardioprotective effects of Fruitflow® against Doxorubicin-induced toxicity in rat cardiomyoblast cells H9c2 (2-1) and high-fat diet-induced dyslipidemia and pathological alteration in cardiac tissue of Wistar Albino rats. Biomed Pharmacother 2024; 180:117607. [PMID: 39471653 DOI: 10.1016/j.biopha.2024.117607] [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: 08/12/2024] [Revised: 09/17/2024] [Accepted: 10/21/2024] [Indexed: 11/01/2024] Open
Abstract
BACKGROUND Natural compounds offer promising targets for cardioprotection, which could lead to enhanced clinical outcomes. We aimed to determine the cardioprotective effects of Fruitflow®, a water-soluble tomato extract known for its anti-platelet effects in doxorubicin-induced toxicity in rat cardiomyoblast cell line pathological alteration in heart tissue of high fat-fed Wistar Albino rats. METHODS The cardioprotective effect of Fruitflow® was investigated using H9c2 (2-1) cells (rat cardiomyoblast cell line) and high-fat diet-fed Wistar Albino rats. We evaluated morphological changes, cell proliferation, cell migration, antioxidant activity, cell cycle progression, and mitochondrial membrane potential after the Fruitflow® treatment in the Doxorubicin-injured H9c2 (2-1) cell line. We studied lipid profiles, inflammation, oxidative stress, and cardiac function regulatory enzyme activity in the rat model. RESULTS Fruitflow® dose-dependently stimulated cell proliferation and migration in Doxorubicin-injured H9c2 (2-1) cells, potentially promoting cardiac regeneration and supporting tissue repair. Fruitflow® modulated the cell cycle, improved mitochondrial function, and reduced oxidative stress. Furthermore, it significantly improved lipid profiles and enzyme activities and reduced inflammation and oxidative stress in high-fat-fed rats. Fruitflow® also modulated the expression of genes involved in cardiac remodeling, mitochondrial biogenesis, inflammation, and vascular function. CONCLUSION Our findings suggest Fruitflow® may have cardioprotective effects, making it a potential treatment option for cardiac ailments. Larger-scale clinical trials were recommended further to determine the efficacy and safety of Fruitflow® as a potential therapeutic agent for cardiac diseases, potentially in combination with other cardioprotective medications.
Collapse
Affiliation(s)
- Diptimayee Das
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Tamil Nadu 603103, India
| | - Ganesan Jothimani
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Tamil Nadu 603103, India
| | - Antara Banerjee
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Tamil Nadu 603103, India
| | - Asim K Duttaroy
- Department of Nutrition, Institute of Medical Sciences, Faculty of Medicine, University of Oslo, Norway.
| | - Surajit Pathak
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Tamil Nadu 603103, India.
| |
Collapse
|
6
|
Shi X, Cao Y, Wang H, Zhao Q, Yan C, Li S, Jing L. Vaccarin Ameliorates Doxorubicin-Induced Cardiotoxicity via Inhibition of p38 MAPK Mediated Mitochondrial Dysfunction. J Cardiovasc Transl Res 2024; 17:1155-1171. [PMID: 38886316 PMCID: PMC11519163 DOI: 10.1007/s12265-024-10525-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 05/16/2024] [Indexed: 06/20/2024]
Abstract
Doxorubicin is a frequently used chemotherapeutic agent for treating various malignancies. However, it leads to severe cardiotoxic side effects, such as heart failure, and elevates the risk of sudden cardiac death among cancer patients. While oxidative stress has been identified as the primary cause of doxorubicin-induced cardiotoxicity, therapeutic antioxidant approaches have yielded unsatisfactory outcomes. The aim of this study is to explore the therapeutic potential of vaccarin, an active flavonoid glycoside extracted from traditional Chinese herbal agent Semen Vaccariae, in doxorubicin-induced cardiotoxicity. We observed that vaccarin significantly ameliorates doxorubicin-induced heart dysfunction in mouse model and suppresses oxidative stress mediated cell apoptosis via specifically inhibiting the activation of p38 MAPK pathway. In vitro, we observed that vaccarin alleviates doxorubicin-induced mitochondrial membrane depolarization and ROS generation in H9c2 cell, but the p38 MAPK agonist anisomycin reverses these effects. Our findings provide a promising natural antioxidant to protect against DOX-induced cardiotoxicity.
Collapse
MESH Headings
- Animals
- p38 Mitogen-Activated Protein Kinases/metabolism
- p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
- Doxorubicin/toxicity
- Mitochondria, Heart/drug effects
- Mitochondria, Heart/enzymology
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/pathology
- Cardiotoxicity
- Oxidative Stress/drug effects
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/pathology
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/metabolism
- Apoptosis/drug effects
- Disease Models, Animal
- Cell Line
- Male
- Antioxidants/pharmacology
- Membrane Potential, Mitochondrial/drug effects
- Mice, Inbred C57BL
- Reactive Oxygen Species/metabolism
- Signal Transduction/drug effects
- Heart Diseases/chemically induced
- Heart Diseases/prevention & control
- Heart Diseases/pathology
- Heart Diseases/metabolism
- Heart Diseases/enzymology
- Rats
- Ventricular Function, Left/drug effects
- Glycosides/pharmacology
- Protein Kinase Inhibitors/pharmacology
Collapse
Affiliation(s)
- Xin Shi
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang Qu, Harbin, 150001, Heilongjiang, China
| | - Yang Cao
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang Qu, Harbin, 150001, Heilongjiang, China
| | - Hongyu Wang
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang Qu, Harbin, 150001, Heilongjiang, China
| | - Qi Zhao
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang Qu, Harbin, 150001, Heilongjiang, China
| | - Cong Yan
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang Qu, Harbin, 150001, Heilongjiang, China
| | - Shengzhu Li
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang Qu, Harbin, 150001, Heilongjiang, China
| | - Ling Jing
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang Qu, Harbin, 150001, Heilongjiang, China.
| |
Collapse
|
7
|
Chen JK, Ramesh S, Islam MN, Shibu MA, Kuo CH, Hsieh DJY, Lin SZ, Kuo WW, Huang CY, Ho TJ. Ohwia caudata inhibits doxorubicin-induced cardiotoxicity by regulating mitochondrial dynamics via the IGF-IIR/p-Drp1/PARP signaling pathway. Biotechnol Appl Biochem 2024; 71:1181-1194. [PMID: 38837810 DOI: 10.1002/bab.2620] [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: 03/25/2024] [Accepted: 05/16/2024] [Indexed: 06/07/2024]
Abstract
The most effective drug, doxorubicin (DOX), is widely used worldwide for clinical application as an anticancer drug. DOX-induced cytotoxicity is characterized by mitochondrial dysfunction. There is no alternative treatment against DOX-induced cardiac damage despite intensive research in the present decades. Ohwia caudata has emerged as a potential herbal remedy that prevents from DOX-induced cytotoxicity owing to its pharmacological action of sustaining mitochondrial dynamics by attenuating oxidative stress and inducing cellular longevity. However, its underlying mechanisms are unknown. The novel treatment provided here depends on new evidence from DOX-treated H9c2 cells, which significantly enhanced insulin-like growth factor (IGF) II receptor (IGF-IIR) pathways that activated calcineurin and phosphorylated dynamin-related protein 1 (p-Drp1) at ser616 (p-Drp1[ser616]); cells undergo apoptosis due to these factors, which translocate to mitochondria and disrupt their function and integrity, and in terms of herbal medicine treatment, which significantly blocked these phenomena. Thus, our findings indicate that maintaining integrity of mitochondria is an essential element in lowering DOX-induced cytotoxicity, which further emphasizes that our herbal medicine can successfully block IGF-IIR pathways and could potentially act as an alternative mechanism in terms of cardioprotective against doxorubicin.
Collapse
Affiliation(s)
- Jhong-Kuei Chen
- Department of Chinese Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| | - Samiraj Ramesh
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Department of Research and Innovation, Institute of Biotechnology, Saveetha School of Engineering (SSE), Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, India
| | - Md Nazmul Islam
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | | | - Chia-Hua Kuo
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei, Taiwan
| | - Dennis Jine-Yuan Hsieh
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Shinn-Zong Lin
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, College of Life Sciences, China Medical University, Taichung, Taiwan
- Ph.D. Program for Biotechnology Industry, China Medical University, Taichung, Taiwan
- School of Pharmacy, China Medical University, Taichung, Taiwan
| | - Chih-Yang Huang
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan
- Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien, Taiwan
| | - Tsung-Jung Ho
- Department of Chinese Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
- School of Post-Baccalaureate Chinese Medicine, College of Medicine, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
8
|
Tiwari V, Gupta P, Malladi N, Salgar S, Banerjee SK. Doxorubicin induces phosphorylation of lamin A/C and loss of nuclear membrane integrity: A novel mechanism of cardiotoxicity. Free Radic Biol Med 2024; 218:94-104. [PMID: 38582228 DOI: 10.1016/j.freeradbiomed.2024.04.212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/02/2024] [Accepted: 04/04/2024] [Indexed: 04/08/2024]
Abstract
Lamin A/C, essential inner nuclear membrane proteins, have been linked to progeria, a disease of accelerated aging, and many other diseases, which include cardiac disorder. Lamin A/C mutation and its phosphorylation are associated with altering nuclear shape and size. The role of lamin A/C in regulating normal cardiac function was reported earlier. In the present study, we hypothesized that Doxorubicin (Dox) may alter total lamin A/C expression and phosphorylation, thereby taking part in cardiac injury. An in vitro cellular injury model was generated with Dox (0.1-10.0 μM) treatment on cardiomyoblast cells (H9c2) to prove our hypothesis. Increased size and irregular (ameboid) nucleus shape were observed in H9c2 cells after Dox treatment. Similarly, we have observed a significant increase in cell death on increasing the Dox concentration. The expression of lamin A/C and its phosphorylation at serine 22 significantly decreased and increased, respectively in H9c2 cells and rat hearts after Dox exposure. Phosphorylation led to depolymerization of the lamin A/C in the inner nuclear membrane and was evidenced by their presence throughout the nucleoplasm as observed by immunocytochemistry techniques. Thinning and perforation on the walls of the nuclear membrane were observed in Dox-treated H9c2 cells. LMNA-overexpression in H9c2 protected the cells from Dox-induced cell death, reversing all changes described above. Further, improvement of lamin A/C levels was observed in Dox-treated H9c2 cells when treated with Purvalanol A, a CDK1 inhibitor and N-acetylcysteine, an antioxidant. The study provides new insight regarding Dox-induced cardiac injury with the involvement of lamin A/C and alteration of inner nuclear membrane structure.
Collapse
Affiliation(s)
- Vikas Tiwari
- National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India.
| | - Paras Gupta
- National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India.
| | - Navya Malladi
- National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India.
| | - Sanjay Salgar
- National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India.
| | - Sanjay K Banerjee
- National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India.
| |
Collapse
|
9
|
Mah CK, Ahmed N, Lopez NA, Lam DC, Pong A, Monell A, Kern C, Han Y, Prasad G, Cesnik AJ, Lundberg E, Zhu Q, Carter H, Yeo GW. Bento: a toolkit for subcellular analysis of spatial transcriptomics data. Genome Biol 2024; 25:82. [PMID: 38566187 PMCID: PMC11289963 DOI: 10.1186/s13059-024-03217-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 03/14/2024] [Indexed: 04/04/2024] Open
Abstract
The spatial organization of molecules in a cell is essential for their functions. While current methods focus on discerning tissue architecture, cell-cell interactions, and spatial expression patterns, they are limited to the multicellular scale. We present Bento, a Python toolkit that takes advantage of single-molecule information to enable spatial analysis at the subcellular scale. Bento ingests molecular coordinates and segmentation boundaries to perform three analyses: defining subcellular domains, annotating localization patterns, and quantifying gene-gene colocalization. We demonstrate MERFISH, seqFISH + , Molecular Cartography, and Xenium datasets. Bento is part of the open-source Scverse ecosystem, enabling integration with other single-cell analysis tools.
Collapse
Affiliation(s)
- Clarence K Mah
- Division of Medical Genetics, Department of Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Sanford Stem Cell Institute Innovation Center, La Jolla, CA, USA
| | - Noorsher Ahmed
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Sanford Stem Cell Institute Innovation Center, La Jolla, CA, USA
| | - Nicole A Lopez
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Dylan C Lam
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Sanford Stem Cell Institute Innovation Center, La Jolla, CA, USA
- Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Avery Pong
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Alexander Monell
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Colin Kern
- Center for Epigenomics, University of California San Diego, La Jolla, CA, USA
| | - Yuanyuan Han
- Center for Epigenomics, University of California San Diego, La Jolla, CA, USA
| | - Gino Prasad
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Anthony J Cesnik
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Emma Lundberg
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Department of Pathology, Stanford University, Stanford, CA, USA
- Chan-Zuckerberg Biohub, San Francisco, CA, USA
| | - Quan Zhu
- Center for Epigenomics, University of California San Diego, La Jolla, CA, USA
| | - Hannah Carter
- Division of Medical Genetics, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Gene W Yeo
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA.
- Sanford Stem Cell Institute Innovation Center, La Jolla, CA, USA.
- Stem Cell Program, University of California San Diego, La Jolla, CA, USA.
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
10
|
Belger C, Abrahams C, Imamdin A, Lecour S. Doxorubicin-induced cardiotoxicity and risk factors. IJC HEART & VASCULATURE 2024; 50:101332. [PMID: 38222069 PMCID: PMC10784684 DOI: 10.1016/j.ijcha.2023.101332] [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] [Received: 07/24/2023] [Revised: 12/14/2023] [Accepted: 12/16/2023] [Indexed: 01/16/2024]
Abstract
Doxorubicin (DOX) is an anthracycline antibiotic widely used as a chemotherapeutic agent to treat solid tumours and hematologic malignancies. Although useful in the treatment of cancers, the benefit of DOX is limited due to its cardiotoxic effect that is observed in a large number of patients. In the literature, there is evidence that the presence of various factors may increase the risk of developing DOX-induced cardiotoxicity. A better understanding of the role of these different factors in DOX-induced cardiotoxicity may facilitate the choice of the therapeutic approach in cancer patients suffering from various cardiovascular risk factors. In this review, we therefore discuss the latest findings in both preclinical and clinical research suggesting a link between DOX-induced cardiotoxicity and various risk factors including sex, age, ethnicity, diabetes, dyslipidaemia, obesity, hypertension, cardiovascular disease and co-medications.
Collapse
Affiliation(s)
| | | | - Aqeela Imamdin
- Cardioprotection Group, Cape Heart Institute, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Sandrine Lecour
- Cardioprotection Group, Cape Heart Institute, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
11
|
Tuo H, Li W, Zhao W, Zhao J, Li D, Jin L. Shikonin alleviates doxorubicin-induced cardiotoxicity via Mst1/Nrf2 pathway in mice. Sci Rep 2024; 14:924. [PMID: 38195835 PMCID: PMC10776756 DOI: 10.1038/s41598-024-51675-7] [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: 09/03/2023] [Accepted: 01/08/2024] [Indexed: 01/11/2024] Open
Abstract
Doxorubicin (DOX) is a popular and potent anticancer drug, but its cardiotoxicity limits its clinical application. Shikonin has a wide range of biological functions, including antioxidant and anti-inflammatory effects. The aim of this study was to investigate the effects of shikonin on DOX-induced cardiac injury and to identify the underlying mechanisms. Mice receiving shikonin showed reduced cardiac injury response and enhanced cardiac function after DOX administration. Shikonin significantly attenuated DOX-induced oxidative damage, inflammation accumulation and cardiomyocyte apoptosis. Shikonin protects against DOX-induced cardiac injury by inhibiting Mammalian sterile 20-like kinase 1 (Mst1) and oxidative stress and activating the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway. In conclusion, shikonin alleviates DOX-induced cardiotoxicity by inhibiting Mst1 and activating Nrf2. Shikonin may be used to treat DOX-induced cardiac injury.
Collapse
Affiliation(s)
- Hu Tuo
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wenjing Li
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wei Zhao
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Juan Zhao
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Danni Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lin Jin
- Department of Orthopedics, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China.
| |
Collapse
|
12
|
Lv L, Shi Y, Deng Z, Xu J, Ye Z, He J, Chen G, Yu X, Wu J, Huang X, Li G. A polymeric nanocarrier that eradicates breast cancer stem cells and delivers chemotherapeutic drugs. Biomater Res 2023; 27:133. [PMID: 38102651 PMCID: PMC10722842 DOI: 10.1186/s40824-023-00465-9] [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: 09/19/2023] [Accepted: 11/20/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Drug nanocarriers can markedly reduce the toxicities and side effects of encapsulated chemotherapeutic drugs in the clinic. However, these drug nanocarriers have little effect on eradicating breast cancer stem cells (BCSCs). Although compounds that can inhibit BCSCs have been reported, these compounds are difficult to use as carriers for the widespread delivery of conventional chemotherapeutic drugs. METHODS Herein, we synthesize a polymeric nanocarrier, hyaluronic acid-block-poly (curcumin-dithiodipropionic acid) (HA-b-PCDA), and explore the use of HA-b-PCDA to simultaneously deliver chemotherapeutic drugs and eradicate BCSCs. RESULTS Based on molecular docking and molecular dynamics studies, HA-b-PCDA delivers 35 clinical chemotherapeutic drugs. To further verify the drug deliver ability of HA-b-PCDA, doxorubicin, paclitaxel, docetaxel, gemcitabine and camptothecin are employed as model drugs to prepare nanoparticles. These drug-loaded HA-b-PCDA nanoparticles significantly inhibit the proliferation and stemness of BCSC-enriched 4T1 mammospheres. Moreover, doxorubicin-loaded HA-b-PCDA nanoparticles efficiently inhibit tumor growth and eradicate approximately 95% of BCSCs fraction in vivo. Finally, HA-b-PCDA eradicates BCSCs by activating Hippo and inhibiting the JAK2/STAT3 pathway. CONCLUSION HA-b-PCDA is a polymeric nanocarrier that eradicates BCSCs and potentially delivers numerous clinical chemotherapeutic drugs.
Collapse
Affiliation(s)
- Li Lv
- Department of Pharmacy, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Yonghui Shi
- Department of Pharmacy, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Zhicheng Deng
- Shenshan Medical Center, Memorial Hospital of Sun Yat-Sen University, Shanwei, Guangdong, 516600, China
| | - Jiajia Xu
- School of Pharmacy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Zicong Ye
- Department of Pharmacy, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Jianxiong He
- Department of Pharmacy, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Guanghui Chen
- Department of Pharmacy, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Xiaoxia Yu
- Department of Pharmacy, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Junyan Wu
- Department of Pharmacy, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China.
| | - Xingzhen Huang
- School of Pharmacy, Guangxi Medical University, Nanning, Guangxi, 530021, China.
| | - Guocheng Li
- Department of Pharmacy, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China.
- Shenshan Medical Center, Memorial Hospital of Sun Yat-Sen University, Shanwei, Guangdong, 516600, China.
| |
Collapse
|
13
|
Foglio E, D’Avorio E, Vitiello L, Masuelli L, Bei R, Pacifici F, Della-Morte D, Mirabilii S, Ricciardi MR, Tafuri A, Garaci E, Russo MA, Tafani M, Limana F. Doxorubicin-Induced Cardiac Senescence Is Alleviated Following Treatment with Combined Polyphenols and Micronutrients through Enhancement in Mitophagy. Cells 2023; 12:2605. [PMID: 37998340 PMCID: PMC10670650 DOI: 10.3390/cells12222605] [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: 06/28/2023] [Revised: 10/29/2023] [Accepted: 11/07/2023] [Indexed: 11/25/2023] Open
Abstract
Oxidative stress and impaired mitophagy are the hallmarks of cardiomyocyte senescence. Specifically, a decrease in mitophagic flux leads to the accumulation of damaged mitochondria and the development of senescence through increased ROS and other mediators. In this study, we describe the preventive role of A5+, a mix of polyphenols and other micronutrients, in doxorubicin (DOXO)-induced senescence of H9C2 cells. Specifically, H9C2 cells exposed to DOXO showed an increase in the protein expression proteins of senescence-associated genes, p21 and p16, and a decrease in the telomere binding factors TRF1 and TRF2, indicative of senescence induction. Nevertheless, A5+ pre-treatment attenuated the senescent-like cell phenotype, as evidenced by inhibition of all senescent markers and a decrease in SA-β-gal staining in DOXO-treated H9C2 cells. Importantly, A5+ restored the LC3 II/LC3 I ratio, Parkin and BNIP3 expression, therefore rescuing mitophagy, and decreased ROS production. Further, A5+ pre-treatment determined a ripolarization of the mitochondrial membrane and improved basal respiration. A5+-mediated protective effects might be related to its ability to activate mitochondrial SIRT3 in synergy with other micronutrients, but in contrast with SIRT4 activation. Accordingly, SIRT4 knockdown in H9C2 cells further increased MnSOD activity, enhanced mitophagy, and reduced ROS generation following A5+ pre-treatment and DOXO exposure compared to WT cells. Indeed, we demonstrated that A5+ protects H9C2 cells from DOXO-induced senescence, establishing a new specific role for A5+ in controlling mitochondrial quality control by restoring SIRT3 activity and mitophagy, which provided a molecular basis for the development of therapeutic strategies against cardiomyocyte senescence.
Collapse
Affiliation(s)
- Eleonora Foglio
- Technoscience, Parco Scientifico e Tecnologico Pontino, 04100 Latina, Italy
| | - Erica D’Avorio
- Department of Human Sciences and Quality of Life Promotion, San Raffaele University, 00166 Rome, Italy (F.P.); (D.D.-M.); (E.G.); (M.A.R.)
| | | | - Laura Masuelli
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (L.M.); (M.T.)
| | - Roberto Bei
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy;
| | - Francesca Pacifici
- Department of Human Sciences and Quality of Life Promotion, San Raffaele University, 00166 Rome, Italy (F.P.); (D.D.-M.); (E.G.); (M.A.R.)
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - David Della-Morte
- Department of Human Sciences and Quality of Life Promotion, San Raffaele University, 00166 Rome, Italy (F.P.); (D.D.-M.); (E.G.); (M.A.R.)
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy
- Department of Neurology, Evelyn F. McKnight Brain Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Simone Mirabilii
- Hematology, Department of Clinical and Molecular Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00161 Rome, Italy; (S.M.); (M.R.R.); (A.T.)
| | - Maria Rosaria Ricciardi
- Hematology, Department of Clinical and Molecular Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00161 Rome, Italy; (S.M.); (M.R.R.); (A.T.)
| | - Agostino Tafuri
- Hematology, Department of Clinical and Molecular Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00161 Rome, Italy; (S.M.); (M.R.R.); (A.T.)
| | - Enrico Garaci
- Department of Human Sciences and Quality of Life Promotion, San Raffaele University, 00166 Rome, Italy (F.P.); (D.D.-M.); (E.G.); (M.A.R.)
| | - Matteo Antonio Russo
- Department of Human Sciences and Quality of Life Promotion, San Raffaele University, 00166 Rome, Italy (F.P.); (D.D.-M.); (E.G.); (M.A.R.)
- IRCCS San Raffaele Roma, 00166 Rome, Italy;
| | - Marco Tafani
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (L.M.); (M.T.)
| | - Federica Limana
- Department of Human Sciences and Quality of Life Promotion, San Raffaele University, 00166 Rome, Italy (F.P.); (D.D.-M.); (E.G.); (M.A.R.)
- Laboratory of Cellular and Molecular Pathology, IRCCS San Raffaele Roma, 00166 Rome, Italy
| |
Collapse
|
14
|
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
|
15
|
Arrigo E, Gilardi S, Muratori L, Raimondo S, Mancardi D. Biological effects of sub-lethal doses of glyphosate and AMPA on cardiac myoblasts. Front Physiol 2023; 14:1165868. [PMID: 37168227 PMCID: PMC10164986 DOI: 10.3389/fphys.2023.1165868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/10/2023] [Indexed: 05/13/2023] Open
Abstract
Introduction: Glyphosate is the active compound of different non-selective herbicides, being the most used agriculture pesticide worldwide. Glyphosate and AMPA (one of its main metabolites) are common pollutants of water, soil, and food sources such as crops. They can be detected in biological samples from both exposed workers and general population. Despite glyphosate acts as inhibitor of the shikimate pathway, present only in plants and some microorganisms, its safety in mammals is still debated. Acute glyphosate intoxications are correlated to cardiovascular/neuronal damages, but little is known about the effects of the chronic exposure. Methods: We evaluated the direct biological effects of different concentrations of pure glyphosate/AMPA on a rat-derived cell line of cardiomyoblasts (H9c2) in acute (1-2 h) or sub-chronic (24-48 h) settings. We analyzed cell viability/morphology, ROS production and mitochondrial dynamics. Results: Acute exposure to high doses (above 10 mM) of glyphosate and AMPA triggers immediate cytotoxic effects: reduction in cell viability, increased ROS production, morphological alterations and mitochondrial function. When exposed to lower glyphosate concentrations (1 μM-1 mM), H9c2 cells showed only a slight variation in cell viability and ROS production, while mitochondrial dynamic was unvaried. Moreover, the phenotype was completely restored after 48 h of treatment. Surprisingly, the sub-chronic (48 h) treatment with low concentrations (1 μM-1 mM) of AMPA led to a late cytotoxic response, reflected in a reduction in H9c2 viability. Conclusion: The comprehension of the extent of human exposure to these molecules remains pivotal to have a better critical view of the available data.
Collapse
Affiliation(s)
- Elisa Arrigo
- Department of Clinical and Biological Sciences, University of Torino, Turin, Italy
- *Correspondence: Daniele Mancardi, ; Elisa Arrigo,
| | - Sara Gilardi
- Department of Clinical and Biological Sciences, University of Torino, Turin, Italy
| | - Luisa Muratori
- Department of Clinical and Biological Sciences, University of Torino, Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Torino, Turin, Italy
| | - Stefania Raimondo
- Department of Clinical and Biological Sciences, University of Torino, Turin, Italy
- Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Torino, Turin, Italy
| | - Daniele Mancardi
- Department of Clinical and Biological Sciences, University of Torino, Turin, Italy
- *Correspondence: Daniele Mancardi, ; Elisa Arrigo,
| |
Collapse
|
16
|
Hsieh PL, Chu PM, Cheng HC, Huang YT, Chou WC, Tsai KL, Chan SH. Dapagliflozin Mitigates Doxorubicin-Caused Myocardium Damage by Regulating AKT-Mediated Oxidative Stress, Cardiac Remodeling, and Inflammation. Int J Mol Sci 2022; 23:ijms231710146. [PMID: 36077544 PMCID: PMC9456438 DOI: 10.3390/ijms231710146] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/27/2022] [Accepted: 09/01/2022] [Indexed: 11/21/2022] Open
Abstract
Doxorubicin (Dox) is a commonly used anthracycline chemotherapy with a side effect of cardiotoxicity, which may increase the risk of heart failure for cancer patients. Although various studies have demonstrated the cardioprotective property of dapagliflozin (DAPA), a sodium-glucose cotransporter 2 inhibitor, the detailed mechanism underlying its effect on Dox-induced cardiomyopathy is still limited. In this study, we showed that DAPA induced the activation of AKT/PI3K signaling in cardiac myoblast H9c2 cells following Dox treatment, leading to the upregulation of antioxidant HO-1, NQO1, and SOD, as well as an improved mitochondrial dysfunction via Nrf2. In addition, the reduced oxidative stress resulted in the downregulation of hypertrophy (ANP and BNP) and fibrosis (phospho-Smad3, collagen I, fibronectin, and α-SMA) markers. Furthermore, the inflammatory IL-8 concentration was inhibited after DAPA, possibly through PI3K/AKT/Nrf2/p38/NF-κB signaling. Moreover, our results were validated in vivo, and echocardiography results suggested an improved cardiac function in DAPA-receiving rats. In summary, we demonstrated that the administration of DAPA could mitigate the Dox-elicited cardiotoxicity by reducing oxidative stress, mitochondrial dysfunction, fibrosis, hypertrophy, and inflammation via PI3K/AKT/Nrf2 signaling.
Collapse
Affiliation(s)
- Pei-Ling Hsieh
- Department of Anatomy, School of Medicine, China Medical University, Taichung 404, Taiwan
| | - Pei-Ming Chu
- Department of Anatomy, School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
| | - Hui-Ching Cheng
- Department of Physical Therapy, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Yu-Ting Huang
- Department of Physical Therapy, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Wan-Ching Chou
- Department of Physical Therapy, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Kun-Ling Tsai
- Department of Physical Therapy, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
- Institute of Allied Health Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
- Correspondence: (K.-L.T.); (S.-H.C.)
| | - Shih-Hung Chan
- Department of Internal Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan 704, Taiwan
- Correspondence: (K.-L.T.); (S.-H.C.)
| |
Collapse
|
17
|
Afrin H, Huda MN, Islam T, Oropeza BP, Alvidrez E, Abir MI, Boland T, Turbay D, Nurunnabi M. Detection of Anticancer Drug-Induced Cardiotoxicity Using VCAM1-Targeted Nanoprobes. ACS APPLIED MATERIALS & INTERFACES 2022; 14:37566-37576. [PMID: 35939041 PMCID: PMC9994100 DOI: 10.1021/acsami.2c13019] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Chemotherapy-induced cardiac toxicity is an undesirable yet very common effect that increases the risk of death and reduce the quality of life of individuals undergoing chemotherapy. However, no feasible methods and techniques are available to monitor and detect the degree of cardiotoxicity at an early stage. Therefore, in this project, we aim to develop a fluorescent nanoprobe to image the toxicity within the cardiac tissue induced by an anticancer drug. We have observed that vascular cell adhesion molecule 1 (VCAM1) protein alone with collagen was overly expressed within the heart, when an animal was treated with doxorubicin (DOX), because of inflammation in the epithelial cells. We hypothesize that developing a VCAM1-targeted peptide-based (VHPKQHRGGSKGC) fluorescent nanoprobe can detect and visualize the affected heart. In this regard, we prepared a poly(lactic-co-glycolic acid) (PLGA) nanoparticle linked with VCAM1 peptide and rhodamine B (PLGA-VCAM1-RhB). Selective binding and higher accumulation of the PLGA-VCAM1-RhB nanoprobes were detected in DOX-treated human cardiomyocyte cells (HCMs) compared to the untreated cells. For in vivo studies, DOX (5 mg/kg) was injected via the tail vein once in two weeks for 6 weeks (3 injection total). PLGA-VCAM1-RhB and PLGA-RhB were injected via the tail vein after 1 week of the last dose of DOX, and images were taken 4 h after administration. A higher fluorescent signal of PLGA-RhB-VCAM-1 (48.62% ± 12.79%) was observed in DOX-treated animals compared to the untreated control PLGA-RhB (10.61% ± 4.90) within the heart, indicating the specificity and targeting ability of PLGA-VCAM1-RhB to the inflamed tissues. The quantified fluorescence intensity of the homogenized cardiac tissue of PLGA-RhB-VCAM1 showed 156% higher intensity than the healthy control group. We conclude that PLGA-VCAM1-RhB has the potential to bind inflamed cardiac cells, thereby detecting DOX-induced cardiotoxicity and damaged heart at an early stage.
Collapse
Affiliation(s)
- Humayra Afrin
- Environmental Science and Engineering, University of Texas at El Paso, El Paso, Texas 79965, United States
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, Texas 79902, United States
| | - Md Nurul Huda
- Environmental Science and Engineering, University of Texas at El Paso, El Paso, Texas 79965, United States
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, Texas 79902, United States
| | - Tamanna Islam
- Environmental Science and Engineering, University of Texas at El Paso, El Paso, Texas 79965, United States
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, Texas 79902, United States
| | - Beu P Oropeza
- Biomedical Engineering, College of Engineering, University of Texas at El Paso, El Paso, Texas 79965, United States
| | - Efren Alvidrez
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, Texas 79902, United States
- Aerospace Center (cSETR), University of Texas at El Paso, El Paso, Texas 79965, United States
| | - Muhammad I Abir
- Aerospace Center (cSETR), University of Texas at El Paso, El Paso, Texas 79965, United States
| | - Thomas Boland
- Biomedical Engineering, College of Engineering, University of Texas at El Paso, El Paso, Texas 79965, United States
| | - David Turbay
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, Texas 79902, United States
| | - Md Nurunnabi
- Environmental Science and Engineering, University of Texas at El Paso, El Paso, Texas 79965, United States
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, Texas 79902, United States
- Biomedical Engineering, College of Engineering, University of Texas at El Paso, El Paso, Texas 79965, United States
- Aerospace Center (cSETR), University of Texas at El Paso, El Paso, Texas 79965, United States
| |
Collapse
|
18
|
Goyal SG, Dhar A. Downregulation of nesfatin-1 expression in acute kidney injury in vivo in wistar rats and in vitro in cultured cells. Life Sci 2022; 305:120762. [PMID: 35787996 DOI: 10.1016/j.lfs.2022.120762] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/19/2022] [Accepted: 06/28/2022] [Indexed: 10/17/2022]
Abstract
AIMS Acute kidney injury (AKI) is a debilitating condition followed by sudden kidney damage or failure within hours or days of its occurrence. AKI is characterized by rapid increase in serum creatinine/BUN and decrease in urine output. Nesfatin-1 is an endogenous peptide reported to possess anorexic, antioxidant and anti-apoptotic properties. Although few clinical studies have shown altered nesfatin-1 levels in hemodialysis patients, however, there are no reports investigating the distribution and expression pattern of nesfatin-1 in AKI. MATERIALS AND METHODS Nesfatin-1 expression was determined in different disease induced models of AKI by immunoblotting, immunofluorescence and RT-PCR. Gene markers of oxidative stress and inflammation were determined by RT-PCR. The expression of different markers of AKI was measured by assay kits and RT-PCR analysis. KEY FINDINGS There was a significant increase in serum levels of creatinine and BUN in AKI rats followed by significant increase in KIM-1 in the kidneys. Significant decrease in nesfatin-1 expression along with increased expression of IL-1β, TNF-α and decreased expression of SOD and catalase was observed in doxorubicin and cisplatin induced AKI rats. However, SOD and catalase expression were upregulated in glycerol induced AKI rats. Moreover, in vitro treatment of renal NRK-52E epithelial cells with nesfatin-1 reversed the changes induced by doxorubicin. SIGNIFICANCE Our study reports for the first time, nesfatin-1 expression is decreased in kidneys of different models of AKI induced rats as well as cultured NRK-52E renal epithelial cells. Further studies are required to understand the possible molecular mechanism and therapeutic potential of nesfatin-1 in acute kidney injury.
Collapse
Affiliation(s)
- Srashti Gopal Goyal
- Department of Pharmacy, Birla Institute of Technology and Sciences (BITS) Pilani, Hyderabad Campus, Jawahar Nagar, Shameerpet, Hyderabad, Telangana 500078, India
| | - Arti Dhar
- Department of Pharmacy, Birla Institute of Technology and Sciences (BITS) Pilani, Hyderabad Campus, Jawahar Nagar, Shameerpet, Hyderabad, Telangana 500078, India.
| |
Collapse
|
19
|
Assessing Drug-Induced Mitochondrial Toxicity in Cardiomyocytes: Implications for Preclinical Cardiac Safety Evaluation. Pharmaceutics 2022; 14:pharmaceutics14071313. [PMID: 35890211 PMCID: PMC9319223 DOI: 10.3390/pharmaceutics14071313] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/09/2022] [Accepted: 06/14/2022] [Indexed: 02/07/2023] Open
Abstract
Drug-induced cardiotoxicity not only leads to the attrition of drugs during development, but also contributes to the high morbidity and mortality rates of cardiovascular diseases. Comprehensive testing for proarrhythmic risks of drugs has been applied in preclinical cardiac safety assessment for over 15 years. However, other mechanisms of cardiac toxicity have not received such attention. Of them, mitochondrial impairment is a common form of cardiotoxicity and is known to account for over half of cardiovascular adverse-event-related black box warnings imposed by the U.S. Food and Drug Administration. Although it has been studied in great depth, mitochondrial toxicity assessment has not yet been incorporated into routine safety tests for cardiotoxicity at the preclinical stage. This review discusses the main characteristics of mitochondria in cardiomyocytes, drug-induced mitochondrial toxicities, and high-throughput screening strategies for cardiomyocytes, as well as their proposed integration into preclinical safety pharmacology. We emphasize the advantages of using adult human primary cardiomyocytes for the evaluation of mitochondrial morphology and function, and the need for a novel cardiac safety testing platform integrating mitochondrial toxicity and proarrhythmic risk assessments in cardiac safety evaluation.
Collapse
|
20
|
Pluronic F127/Doxorubicin microemulsions: Preparation, characterization, and toxicity evaluations. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2021.117028] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
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
|
Zhou W, Ouyang J, Hu N, Li G, Wang H. Protective Effect of Two Alkaloids from Hippophae rhamnoides Linn. against Doxorubicin-Induced Toxicity in H9c2 Cardiomyoblasts. Molecules 2021; 26:molecules26071946. [PMID: 33808398 PMCID: PMC8037594 DOI: 10.3390/molecules26071946] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 03/24/2021] [Accepted: 03/27/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Doxorubicin (Dox) is one of the most frequently prescribed anti-cancer drugs. However, clinical application with Dox is limited due to its potentially fatal cumulative cardiotoxicity. N-p-coumaroyl-4-aminobutan-1-ol (alk-A), an organic amide alkaloid and hippophamide (alk-B), a rare pyridoindole alkaloid were successfully obtained by purification and separation of seabuckthorn seed residue in our previous research. This study was undertaken to investigate the protective effect of alk-A and alk-B against Dox-induced embryonic rat cardiac cells (H9c2 cells) apoptosis. METHODS H9c2 cells were treated with Dox (2.5 µM) in the presence of alk-A and alk-B (10, 20, and 40 µM) and incubated for 24 h. RESULTS It was shown that pretreatment of the H9c2 cells with alk-A and alk-B significantly reduced Dox-induced apoptosis. Alk-A and alk-B both inhibited reactive oxygen species (ROS) production and suppressed cleaved-caspase-3 protein expression and the activation of JNK (Jun N-terminal kinases), as well as increasing ATP levels, favoring mitochondrial mitofusin protein expression, and relieving damage to mitochondrial DNA. CONCLUSIONS These results suggest that alk-A and alk-B can inhibit Dox-induced apoptosis in H9C2 cardiac muscle cells via inhibition of cell apoptosis and improvement of mitochondrial function, while alk-B showed more protection. Alk-B could be a potential candidate agent for protecting against cardiotoxicity in Dox-exposed patients.
Collapse
Affiliation(s)
- Wenna Zhou
- Department of Life Sciences and Health, QiuZhen College, Huzhou University, Huzhou 313000, China;
| | - Jian Ouyang
- CAS Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Xining 810008, China; (J.O.); (N.H.)
- Huzhou Plateau Biological Resource Centre of Innovation, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Huzhou 313000, China
| | - Na Hu
- CAS Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Xining 810008, China; (J.O.); (N.H.)
| | - Gang Li
- Center for Mitochondria and Healthy Aging, College of Life Sciences, Yantai University, Yantai 264005, China
- Correspondence: (G.L.); (H.W.); Tel.: +86-136-7865-9123 (G.L.); +86-139-9738-4106 (H.W.)
| | - Honglun Wang
- CAS Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Xining 810008, China; (J.O.); (N.H.)
- Huzhou Plateau Biological Resource Centre of Innovation, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Huzhou 313000, China
- Correspondence: (G.L.); (H.W.); Tel.: +86-136-7865-9123 (G.L.); +86-139-9738-4106 (H.W.)
| |
Collapse
|
23
|
Afrostyrax lepidophyllus Mildbr. and Monodora myristica (Gaertn.) Dunal Extracts Decrease Doxorubicin Cytotoxicity on H9c2 Cardiomyoblasts. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:8858165. [PMID: 33688366 PMCID: PMC7920721 DOI: 10.1155/2021/8858165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 01/25/2021] [Accepted: 02/05/2021] [Indexed: 11/17/2022]
Abstract
Materials and Methods Bark extracts of these plants (1 and 25 µg/mL) were added 3 hours before coincubating H9c2 cardiomyoblasts with Dox (0.5 and 1 µM) for 24 hours more. We measured cell mass and metabolic viability, mitochondrial transmembrane potential, superoxide anion content, and activity-like of caspase-3 and caspase-9 following treatment with the extracts and/or Dox. Also, selenium and vitamin C contents were measured in the plant extracts. Results The results confirmed that Dox treatment decreased cell mass, mitochondrial membrane potential and metabolic viability, increased mitochondrial superoxide anion, and stimulated caspase-3 and caspase-9-like activities. Pretreatment of the cells with the plant extracts significantly inhibited Dox cytotoxicity, with more significant results at the higher concentration. Measurements of selenium and vitamin C in the extracts revealed higher concentration of both when compared with other Cameroonian spices. Conclusion Both extracts of A. lepidophyllus and M. myristica were effective against Dox-induced cytotoxicity, most likely due to their content in antioxidants.
Collapse
|
24
|
Anthracycline-induced cardiomyopathy: cellular and molecular mechanisms. Clin Sci (Lond) 2021; 134:1859-1885. [PMID: 32677679 DOI: 10.1042/cs20190653] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 07/02/2020] [Accepted: 07/03/2020] [Indexed: 02/06/2023]
Abstract
Despite the known risk of cardiotoxicity, anthracyclines are widely prescribed chemotherapeutic agents. They are broadly characterized as being a robust effector of cellular apoptosis in rapidly proliferating cells through its actions in the nucleus and formation of reactive oxygen species (ROS). And, despite the early use of dexrazoxane, no effective treatment strategy has emerged to prevent the development of cardiomyopathy, despite decades of study, suggesting that much more insight into the underlying mechanism of the development of cardiomyopathy is needed. In this review, we detail the specific intracellular activities of anthracyclines, from the cell membrane to the sarcoplasmic reticulum, and highlight potential therapeutic windows that represent the forefront of research into the underlying causes of anthracycline-induced cardiomyopathy.
Collapse
|
25
|
Human Amnion Membrane Proteins Prevent Doxorubicin-Induced Oxidative Stress Injury and Apoptosis in Rat H9c2 Cardiomyocytes. Cardiovasc Toxicol 2021; 20:370-379. [PMID: 32086724 DOI: 10.1007/s12012-020-09564-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Doxorubicin (DOX) is widely used as an effective chemotherapy agent in cancer treatment. Cardiac toxicity in cancer treatment with DOX demand urgent attention and no effective treatment has been established for DOX-induced cardiomyopathy. It has been well documented that human amniotic membrane proteins (AMPs), extracted from amnion membrane (AM), have antioxidant, anti-apoptotic, and cytoprotective properties. Therefore, in this study, we aimed to investigate the protective effects of AMPs against cardiotoxicity induced by DOX in cultured rat cardiomyocyte cells (H9c2). DOX-induced cell injury was evaluated using multi-parametric assay including thiazolyl blue tetrazolium bromide (MTT), the release of lactic dehydrogenase (LDH), intracellular Ca2+ , reactive oxygen species (ROS) levels, cellular antioxidant status, mitochondrial membrane potential (ΔΨm), malondialdehyde (MDA), and NF-κB p65 DNA-binding activity. Moreover, expression profiling of apoptosis-related genes (P53, Bcl-2, and Bax) and Annexin V by flow cytometry were used for cell apoptosis detection. It was shown that AMPs pretreatment inhibited the cell toxicity induced by DOX. AMPs effectively attenuated the increased levels of LDH, Ca2+ , ROS, and MDA and also simultaneously elevated the ΔΨm and antioxidant status such as superoxide dismutase (SOD) and Catalase (CAT) in pretreated H9c2 cardiomyocytes. Besides, the activity of NF-kB p65 was reduced and the p53 and Bax protein levels were inhibited in these myocardial cells subjected to DOX. These findings provide the first evidence that AMPs potently suppressed DOX-induced toxicity in cardiomyocytes through inhibition of oxidative stress and apoptosis. Thus, AMPs can be a potential therapeutic agent against DOX cardiotoxicity.
Collapse
|
26
|
Nandi S, Kale N, Patil A, Banerjee S, Patil Y, Khandare J. A graphene-sandwiched DNA nano-system: regulation of intercalated doxorubicin for cellular localization. NANOSCALE ADVANCES 2020; 2:5746-5759. [PMID: 36133866 PMCID: PMC9417510 DOI: 10.1039/d0na00575d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 10/05/2020] [Indexed: 06/16/2023]
Abstract
Control of the sub-cellular localization of nanoparticles (NPs) with enhanced drug-loading capacity, employing graphene oxide (GO), iron oxide (Fe3O4) NPs and sandwiched deoxyribonucleic acid (DNA) bearing intercalated anticancer drug doxorubicin (DOX) has been investigated in this work. The nanosystems G-DNA-DOX-Fe3O4 and Fe3O4-DNA-DOX differentially influence serum protein binding and deliver DOX to lysosomal compartments of cervical cancer (HeLa) cells with enhanced retention. Stern-Volmer plots describing BSA adsorption on the nanosystems demonstrated the quenching constants, K sv for G-DNA-DOX-Fe3O4 and Fe3O4-DNA-DOX (0.025 mL μg-1 and 0.0103 mL μg-1 respectively). Nuclear DOX intensity, measured at 24 h, was ∼2.0 fold higher for Fe3O4-DNA-DOX in HeLa cells. Parallelly, the cytosol displayed ∼2.2 fold higher DOX intensity for Fe3O4-DNA-DOX compared to G-DNA-DOX-Fe3O4. Fe3O4-DNA-DOX was more efficacious in the cytotoxic effect than G-DNA-DOX-Fe3O4 (viability of treated cells: 33% and 49% respectively). The DNA:nanosystems demonstrated superior cytotoxicity compared to mole-equivalent free DOX administration. The results implicate DNA:DOX NPs in influencing the cellular uptake mechanism and were critically subject to cellular localization. Furthermore, cell morphology analysis evidenced maximum deformation attributed to free-DOX with 34% increased cell roundness, 63% decreased cell area and ∼1.9 times increased nuclear-to-cytoplasmic (N/C) ratio after 24 h. In the case of Fe3O4-DNA-DOX, the N/C ratio increased 1.2 times and a maximum ∼37% decrease in NSA was noted suggesting involvement of non-canonical cytotoxic pathways. In conclusion, the study makes a case for designing nanosystems with controlled and regulated sub-cellular localization to potentially exploit secondary cytotoxic pathways, in addition to optimized drug-loading for enhanced anticancer efficacy and reduced adverse effects.
Collapse
Affiliation(s)
- Semonti Nandi
- MAEER's Maharashtra Institute of Pharmacy Kothrud Pune 411038 India
- School of Pharmacy, Dr Vishwanath Karad MIT World Peace University MIT Campus, S. No. 124, Paud Road, Kothrud Pune 411038 India
| | - Narendra Kale
- MAEER's Maharashtra Institute of Pharmacy Kothrud Pune 411038 India
- School of Pharmacy, Dr Vishwanath Karad MIT World Peace University MIT Campus, S. No. 124, Paud Road, Kothrud Pune 411038 India
| | - Ashwini Patil
- MAEER's Maharashtra Institute of Pharmacy Kothrud Pune 411038 India
- School of Pharmacy, Dr Vishwanath Karad MIT World Peace University MIT Campus, S. No. 124, Paud Road, Kothrud Pune 411038 India
| | - Shashwat Banerjee
- Maharashtra Institute of Medical Education and Research Medical College Talegaon Dabhade Pune 410507 India
| | - Yuvraj Patil
- Maharashtra Institute of Medical Education and Research Medical College Talegaon Dabhade Pune 410507 India
| | - Jayant Khandare
- School of Pharmacy, Dr Vishwanath Karad MIT World Peace University MIT Campus, S. No. 124, Paud Road, Kothrud Pune 411038 India
- School of Consciousness, Dr Vishwanath Karad MIT World Peace University MIT Campus, S. No. 124, Paud Road, Kothrud Pune 411038 India
| |
Collapse
|
27
|
Kale N, Nandi S, Patil A, Patil Y, Banerjee S, Khandare J. Nanocarrier anticancer drug-conjugates cause higher cellular deformations: culpable for mischief. Biomater Sci 2020; 8:5729-5738. [PMID: 32940277 DOI: 10.1039/d0bm00923g] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Here we report nanocarrier-anticancer drug conjugates culpable for cellular deformations, critically evidenced through image-based analysis as a measure of karyoplasmic ratio (KR) and nuclear surface area (NSA). Multiwalled carbon nanotubes (MWCNTs) were coordinated additionally with Fe3O4 nanoparticles (NPs) to evaluate the symbiotic influence, and further conjugated to Dox for evaluating the cellular kinetics and for measuring cell deformations. Cellular entry kinetics of the CNT (CNT-Dox and CNT-Cys-Fe3O4-Dox) nanocarriers and their efficiency in nuclear localization were evaluated using cervical cancer (HeLa) cells. Of note, the Dox-bound nanocarriers showed significantly enhanced cell toxicity over the free form of the drug. CNT-Dox and CNT-Cys-Fe3O4-Dox influx occurred within 4 hours, while maximum cellular retention of Dox was observed for CNT-Dox at 24 h. However, the highest KR (∼0.51) was observed for CNT-Dox within 8 hours indicating similar cellular deformations using nanocarrier anticancer drug-conjugates to that of free Dox (KR ∼0.50) at 4 hours. In addition, we observed increased NSA at 4 h in Dox treatment whereas in the case of the Dox conjugated nanocarrier, increased NSA was noted at 8 h treatment. At 8 h exposure of HeLa cells with Dox conjugates, we observed that the cells fall into distinct regions of the morphospace with respect to KR and NSA. Conclusively, nano delivery systems considered for clinical and biomedical translations must take into account the possible negative influences imparting higher cellular deformations and secondary adverse effects over the free form of the drug.
Collapse
Affiliation(s)
- Narendra Kale
- MAEER's Maharashtra Institute of Pharmacy, Kothrud, Pune 411038, India
| | - Semonti Nandi
- MAEER's Maharashtra Institute of Pharmacy, Kothrud, Pune 411038, India
| | - Ashwini Patil
- MAEER's Maharashtra Institute of Pharmacy, Kothrud, Pune 411038, India
| | - Yuvraj Patil
- Maharashtra Institute of Medical Education and Research, Talegaon Dabhade, Pune 410507, India.
| | - Shashwat Banerjee
- Maharashtra Institute of Medical Education and Research, Talegaon Dabhade, Pune 410507, India.
| | - Jayant Khandare
- School of Pharmacy, Dr. Vishwanath Karad Maharashtra Institute of Technology-World Peace University, Kothrud, Pune 411038, India. and School of Consciousness, Dr. Vishwanath Karad Maharashtra Institute of Technology-World Peace University, Kothrud, Pune 411038, India
| |
Collapse
|
28
|
Peptidomics Analysis Reveals Peptide PDCryab1 Inhibits Doxorubicin-Induced Cardiotoxicity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:7182428. [PMID: 33110475 PMCID: PMC7582065 DOI: 10.1155/2020/7182428] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 09/01/2020] [Accepted: 09/20/2020] [Indexed: 12/14/2022]
Abstract
Doxorubicin (DOX) is limited due to dose-dependent cardiotoxicity. Peptidomics is an emerging field of proteomics that has attracted much attention because it can be used to study the composition and content of endogenous peptides in various organisms. Endogenous peptides participate in various biological processes and are important sources of candidates for drug development. To explore peptide changes related to DOX-induced cardiotoxicity and to find peptides with cardioprotective function, we compared the expression profiles of peptides in the hearts of DOX-treated and control mice by mass spectrometry. The results showed that 236 differential peptides were identified upon DOX treatment, of which 22 were upregulated and 214 were downregulated. Next, we predicted that 31 peptides may have cardioprotective function by conducting bioinformatics analysis on the domains of each precursor protein, the predicted score of peptide biological activity, and the correlation of each peptide with cardiac events. Finally, we verified that a peptide (SPFYLRPPSF) from Cryab can inhibit cardiomyocyte apoptosis, reduce the production of reactive oxygen species, improve cardiac function, and ameliorate myocardial fibrosis in vitro and vivo. In conclusion, our results showed that the expression profiles of peptides in cardiac tissue change significantly upon DOX treatment and that these differentially expressed peptides have potential cardioprotective functions. Our study suggests a new direction for the treatment of DOX-induced cardiotoxicity.
Collapse
|
29
|
Doxorubicin Cytotoxicity in Differentiated H9c2 Cardiomyocytes: Evidence for Acute Mitochondrial Superoxide Generation. Cardiovasc Toxicol 2020; 21:152-161. [PMID: 32910361 DOI: 10.1007/s12012-020-09606-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 08/31/2020] [Indexed: 10/23/2022]
Abstract
Although a mitochondrial redox-cycling superoxide-generating mechanism for the cardiotoxicity of doxorubicin was suggested from experiments with isolated mitochondria, its occurrence and contribution to cytotoxicity in intact cardiomyocytes is not fully established. Therefore, we determined the immediate and delayed effects of doxorubicin on the generation of reactive oxygen species (ROS) and cytotoxicity in differentiated H9c2 cardiomyocytes. Although relatively short incubations (3 or 6 h) with 1 or 5 µM doxorubicin did not acutely decrease cell survival, exposure to 5 µM doxorubicin for 3 h was sufficient to cause a significant delayed decrease in cell survival after an additional 24 h without doxorubicin. Mitochondrial superoxide generation was observed to increase within 30 min of incubation with 5 µM doxorubicin. Increased intracellular ROS generation, decreased mitochondrial metabolic activity, and decreased mitochondrial membrane potential (MMP) were observed after more extended periods (6-12 h). Overall, these observations support that the toxicity of doxorubicin to differentiated cardiomyocytes involves acute mitochondrial superoxide generation with subsequent intracellular ROS generation, mitochondrial dysfunction, and cell death.
Collapse
|
30
|
New tirapazamine derivatives protect cardiomyocytes from doxorubicin toxicity. CURRENT ISSUES IN PHARMACY AND MEDICAL SCIENCES 2020. [DOI: 10.2478/cipms-2020-0001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Doxorubicin cardiotoxicity is caused by various mechanisms, most importantly by oxidative stress originating in the mitochondria. Tirapazamine is a hypoxia-activated anticancer experimental drug. Both drugs in normoxia conditions undergo univalent reduction, thus tirapazamine may compete with doxorubicin in univalent reduction enzyme uptake. Herein, tirapazamine derivatives consisted of drug molecules and alkyl chain-connected triphenylphosphine cations that bring about an accumulation in mitochondria. The aim of this study was to evaluate the interaction of newly synthesized tirapazamine derivatives with doxorubicin in rat cardiomyocytes via an vitro model. In the work, H9C2 cells were incubated with combinations of doxorubicin, tirapazamine and seven variants of tirapazamine derivatives. After 24 hours, cell viability was assessed using MTT assay and the results were confirmed by microscopic observation. Tirapazamine in all tested concentrations did not revealed significant protective activity to cardiomyocytes treated with doxorubicine. However, tirapazamine derivatives diminished the cytotoxic effect of doxorubicin regardless of concentration and alkyl chain length. Tirapazamine derivatives have shown protective effects in relation to cardiomyocytes treated with doxorubicin and the mechanism of this phenomenon must be confirmed.
Collapse
|
31
|
Effects of grapeseed extract on doxorubicin-induced cardiotoxicity in rats. Herz 2020; 46:103-108. [PMID: 31970462 DOI: 10.1007/s00059-019-04888-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 09/23/2019] [Accepted: 12/12/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND Doxorubicin (DX) is used for the treatment of many types of cancer; however, a side effect of this agent is cardiotoxicity, which may lead to cardiomyopathy or cardiac failure. Oxidative stress is thought to play a major role in the development of cardiotoxic effects. Proanthocyanidins found in grapeseed (GS) extract may inhibit chemically induced lipid peroxidation and apoptosis caused by oxidative stress. We aimed to investigate the cardioprotective effects of GS extract against DX-induced cardiotoxicity. METHODS A total of 28 male Sprague Dawley rats were grouped to receive: (a) standard nutrition (n = 7); (b) standard nutrition with an additional dose of 10 mg/kg DX (n = 7); (c) standard nutrition plus 100 mg/kg/day of GS (n = 7); (d) standard nutrition with 100 mg/kg/day of GS plus a single dose of 10 mg/kg DX. After 35 days the rats were decapitated and blood samples were taken for biochemical testing. Cardiac tissue samples were prepared for microscopy and histopathological evaluation. RESULTS Rats in the DX group exhibited significant elevations in biomarkers such as troponin and NT-proBNP as well as in oxidative stress markers compared with all other groups. Histopathological examination corroborated these findings by demonstrating significant and severe structural injury in the cardiac tissue of DX rates. Moreover, rats in the DX + GS group had significantly lower cardiac injury than rats in the DX group according to both biochemical (troponin and NT-proBNP) and histopathological analyses. Serum malondialdehyde levels (a marker of oxidative stress) in the DX + GS rats were significantly lower than in the DX rats. CONCLUSION Our findings suggest that GS may reduce the severity of DX-induced cardiotoxicity and thus has the potential to prevent cardiac injury in this setting.
Collapse
|
32
|
Catanzaro MP, Weiner A, Kaminaris A, Li C, Cai F, Zhao F, Kobayashi S, Kobayashi T, Huang Y, Sesaki H, Liang Q. Doxorubicin-induced cardiomyocyte death is mediated by unchecked mitochondrial fission and mitophagy. FASEB J 2019; 33:11096-11108. [PMID: 31291545 PMCID: PMC6766652 DOI: 10.1096/fj.201802663r] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 06/17/2019] [Indexed: 02/06/2023]
Abstract
Doxorubicin (Dox) is a widely used antineoplastic agent that can cause heart failure. Dox cardiotoxicity is closely associated with mitochondrial damage. Mitochondrial fission and mitophagy are quality control mechanisms that normally help maintain a pool of healthy mitochondria. However, unchecked mitochondrial fission and mitophagy may compromise the viability of cardiomyocytes, predisposing them to cell death. Here, we tested this possibility by using Dox-treated H9c2 cardiac myoblast cells expressing either the mitochondria-targeted fluorescent protein MitoDsRed or the novel dual-fluorescent mitophagy reporter mt-Rosella. Dox induced mitochondrial fragmentation as shown by reduced form factor, aspect ratio, and mean mitochondrial size. This effect was abolished by short interference RNA-mediated knockdown of dynamin-related protein 1 (DRP1), a major regulator of fission. Importantly, DRP1 knockdown decreased cell death as indicated by the reduced number of propidium iodide-positive cells and the cleavage of caspase-3 and poly (ADP-ribose) polymerase. Moreover, DRP1-deficient mice were protected from Dox-induced cardiac damage, strongly supporting a role for DRP1-dependent mitochondrial fragmentation in Dox cardiotoxicity. In addition, Dox accelerated mitophagy flux, which was attenuated by DRP1 knockdown, as assessed by the mitophagy reporter mt-Rosella, suggesting the necessity of mitochondrial fragmentation in Dox-induced mitophagy. Knockdown of parkin, a positive regulator of mitophagy, dramatically diminished Dox-induced cell death, whereas overexpression of parkin had the opposite effect. Together, these results suggested that Dox cardiotoxicity was mediated, at least in part, by the increased mitochondrial fragmentation and accelerated mitochondrial degradation by the lysosome. Strategies that limit mitochondrial fission and mitophagy in the physiologic range may help reduce Dox cardiotoxicity.-Catanzaro, M. P., Weiner, A., Kaminaris, A., Li, C., Cai, F., Zhao, F., Kobayashi, S., Kobayashi, T., Huang, Y., Sesaki, H., Liang, Q. Doxorubicin-induced cardiomyocyte death is mediated by unchecked mitochondrial fission and mitophagy.
Collapse
Affiliation(s)
- Michael P. Catanzaro
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, New York, USA
| | - Ashley Weiner
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, New York, USA
| | - Amanda Kaminaris
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, New York, USA
| | - Cairong Li
- Clinical Medical College, Hubei University of Science and Technology, Xianning, China
| | - Fei Cai
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, China
| | - Fengyi Zhao
- Department of Endocrinology, The First Affiliated Hospital, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Satoru Kobayashi
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, New York, USA
| | - Tamayo Kobayashi
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, New York, USA
| | - Yuan Huang
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, New York, USA
| | - Hiromi Sesaki
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Qiangrong Liang
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, New York, USA
| |
Collapse
|
33
|
The anti-cancer drug doxorubicin induces substantial epigenetic changes in cultured cardiomyocytes. Chem Biol Interact 2019; 313:108834. [PMID: 31545955 DOI: 10.1016/j.cbi.2019.108834] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/17/2019] [Accepted: 09/20/2019] [Indexed: 02/07/2023]
Abstract
The anthracycline doxorubicin (DOX) is widely used in cancer therapy with the limitation of cardiotoxicity leading to the development of congestive heart failure. DOX-induced oxidative stress and changes of the phosphoproteome as well as epigenome were described but the exact mechanisms of the adverse long-term effects are still elusive. Here, we tested the impact of DOX treatment on cell death, oxidative stress parameters and expression profiles of proteins involved in epigenetic pathways in a cardiomyocyte cell culture model. Markers of oxidative stress, apoptosis and expression of proteins involved in epigenetic processes were assessed by immunoblotting in cultured rat myoblasts (H9c2) upon treatment with DOX (1 or 5 μM for 24 or 48 h) in adherent viable and detached apoptotic cells. The apoptosis markers cleaved caspase-3 and fractin as well as oxidative stress markers 3-nitrotyrosine and malondialdehyde were dose-dependently increased by DOX treatment. Histone deacetylases (SIRT1 and HDAC2), histone lysine demethylases (KDM3A and LSD1) and histone lysine methyltransferases (SET7 and SMYD1) were significantly regulated by DOX treatment with generation of cleaved protein fragments and posttranslational modifications. Overall, we found significant decrease in histone 3 acetylation in DOX-treated cells. DOX treatment of cultured cardiomyocyte precursor cells causes severe cell death by apoptosis associated with cellular oxidative stress. In addition, significant regulation of proteins involved in epigenetic processes and changes in global histone 3 acetylation were observed. However, the significance and clinical impact of these changes remain elusive.
Collapse
|
34
|
Mendez-Encinas MA, Carvajal-Millan E, Rascón-Chu A, Astiazarán-García H, Valencia-Rivera DE, Brown-Bojorquez F, Alday E, Velazquez C. Arabinoxylan-Based Particles: In Vitro Antioxidant Capacity and Cytotoxicity on a Human Colon Cell Line. MEDICINA (KAUNAS, LITHUANIA) 2019; 55:E349. [PMID: 31284672 PMCID: PMC6681090 DOI: 10.3390/medicina55070349] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 06/29/2019] [Accepted: 07/04/2019] [Indexed: 01/03/2023]
Abstract
Background and objectives: Arabinoxylans (AX) can gel and exhibit antioxidant capacity. Previous studies have demonstrated the potential application of AX microspheres as colon-targeted drug carriers. However, the cytotoxicity of AX gels has not been investigated so far. Therefore, the aim of the present study was to prepare AX-based particles (AXM) by coaxial electrospraying method and to investigate their antioxidant potential and cytotoxicity on human colon cells. Materials and Methods: The gelation of AX was studied by monitoring the storage (G') and loss (G'') moduli. The morphology of AXM was evaluated using optical and scanning electron microscopy (SEM). The in vitro antioxidant activity of AX before and after gelation was measured using the 2,2'-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) (ABTS+), 2,2-diphenyl-1-picrylhydrazyl (DPPH) and ferric reducing antioxidant power (FRAP) methods. In addition, the effect of AX and AXM on the proliferation of human colon cells (CCD 841 CoN) was evaluated using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Results: The final G' and G'' values for AX gels were 293 and 0.31 Pa, respectively. AXM presented spherical shape and rough surface with a three-dimensional and porous network. The swelling ratio and mesh size of AXM were 35 g water/g AX and 27 nm, respectively. Gelation decreased the antioxidant activity of AX by 61-64 %. AX and AXM did not affect proliferation or show any toxic effect on the normal human colon cell line CCD 841 CoN. Conclusion: The results indicate that AXM could be promising biocompatible materials with antioxidant activity.
Collapse
Affiliation(s)
- Mayra A Mendez-Encinas
- Biopolymers, Research Center for Food and Development (CIAD), Hermosillo, Sonora 83304, Mexico
| | | | - Agustín Rascón-Chu
- Biotechnology, Research Center for Food and Development (CIAD), Hermosillo, Sonora 83304, Mexico
| | | | - Dora E Valencia-Rivera
- Department of Chemical Biological and Agropecuary Sciences, University of Sonora, Caborca, Sonora 83621, Mexico
| | | | - Efrain Alday
- Department of Chemistry-Biology, University of Sonora, Hermosillo, Sonora 83000, Mexico
| | - Carlos Velazquez
- Department of Chemistry-Biology, University of Sonora, Hermosillo, Sonora 83000, Mexico
| |
Collapse
|
35
|
Adamcova M, Skarkova V, Seifertova J, Rudolf E. Cardiac Troponins are Among Targets of Doxorubicin-Induced Cardiotoxicity in hiPCS-CMs. Int J Mol Sci 2019; 20:ijms20112638. [PMID: 31146354 PMCID: PMC6600382 DOI: 10.3390/ijms20112638] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/21/2019] [Accepted: 05/27/2019] [Indexed: 01/11/2023] Open
Abstract
Modern diagnostic strategies for early recognition of cancer therapeutics-related cardiac dysfunction involve cardiac troponins measurement. Still, the role of other markers of cardiotoxicity is still unclear. The present study was designed to investigate dynamics of response of human cardiomyocytes derived from induced pluripotent stem cells (hiPCS-CMs) to doxorubicin with the special emphasis on their morphological changes in relation to expression and organization of troponins. The hiPCS-CMs were treated with doxorubicin concentrations (1 and 0.3 µM) for 48 h and followed for next up to 6 days. Exposure of hiPCS-CMs to 1 µM doxorubicininduced suppression of both cardiac troponin T (cTnT) and cardiac troponin I (cTnI) gene expression. Conversely, lower 0.3 µM doxorubicin concentration produced no significant changes in the expression of aforementioned genes. However, the intracellular topography, arrangement, and abundance of cardiac troponin proteins markedly changed after both doxorubicin concentrations. In particular, at 48 h of treatment, both cTnT and cTnI bundles started to reorganize, with some of them forming compacted shapes extending outwards and protruding outside the cells. At later intervals (72 h and onwards), the whole troponin network collapsed and became highly disorganized following, to some degree, overall changes in the cellular shape. Moreover, membrane permeability of cardiomyocytes was increased, and intracellular mitochondrial network rearranged and hypofunctional. Together, our results demonstrate complex effects of clinically relevant doxorubicin concentrations on hiPCS-CM cells including changes in cTnT and cTnI, but also in other cellular compartments contributing to the overall cytotoxicity of this class of cytostatics.
Collapse
Affiliation(s)
- Michaela Adamcova
- Department of Physiology, Faculty of Medicine in Hradec Kralove, Charles University in Prague, Simkova 870, 500 03 Hradec Kralove, Czech Republic.
| | - Veronika Skarkova
- Department of Biology, Faculty of Medicine in Hradec Kralove, Charles University in Prague, Zborovská 2089, 500 03 Hradec Kralove, Czech Republic.
| | - Jitka Seifertova
- Department of Biology, Faculty of Medicine in Hradec Kralove, Charles University in Prague, Zborovská 2089, 500 03 Hradec Kralove, Czech Republic.
| | - Emil Rudolf
- Department of Biology, Faculty of Medicine in Hradec Kralove, Charles University in Prague, Zborovská 2089, 500 03 Hradec Kralove, Czech Republic.
| |
Collapse
|
36
|
Reis-Mendes A, Carvalho F, Remião F, Sousa E, Bastos MDL, Costa VM. The Main Metabolites of Fluorouracil + Adriamycin + Cyclophosphamide (FAC) Are Not Major Contributors to FAC Toxicity in H9c2 Cardiac Differentiated Cells. Biomolecules 2019; 9:98. [PMID: 30862114 PMCID: PMC6468772 DOI: 10.3390/biom9030098] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 02/21/2019] [Accepted: 03/01/2019] [Indexed: 02/07/2023] Open
Abstract
In the clinical practice, the combination of 5-fluorouracil (5-FU) + Adriamycin (also known as doxorubicin, DOX) + cyclophosphamide (CYA) (known as FAC) is used to treat breast cancer. The FAC therapy, however, carries some serious risks, namely potential cardiotoxic effects, although the mechanisms are still unclear. In the present study, the role of the main metabolites regarding FAC-induced cardiotoxicity was assessed at clinical relevant concentrations. Seven-day differentiated H9c2 cells were exposed for 48 h to the main metabolites of FAC, namely the metabolite of 5-FU, α-fluoro-β-alanine (FBAL, 50 or 100 μM), of DOX, doxorubicinol (DOXOL, 0.2 or 1 μM), and of CYA, acrolein (ACRO, 1 or 10 μM), as well as to their combination. The parent drugs (5-FU 50 μM, DOX 1 μM, and CYA 50 μM) were also tested isolated or in combination with the metabolites. Putative cytotoxicity was evaluated through phase contrast microscopy, Hoechst staining, membrane mitochondrial potential, and by two cytotoxicity assays: the reduction of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) and the neutral red (NR) lysosomal incorporation. The metabolite DOXOL was more toxic than FBAL and ACRO in the MTT and NR assays. When in combination, neither FBAL nor ACRO increased DOXOL-induced cytotoxicity. No nuclear condensation was observed for any of the tested combinations; however, a significant mitochondrial potential depolarization after FBAL 100 μM + DOXOL 1 μM + ACRO 10 μM or FBAL 100 μM + DOXOL 1 μM exposure was seen at 48 h. When tested alone DOX 1 μM was more cytotoxic than all the parent drugs and metabolites in both the cytotoxicity assays performed. These results demonstrated that DOXOL was the most toxic of all the metabolites tested; nonetheless, the metabolites do not seem to be the major contributors to FAC-induced cardiotoxicity in this cardiac model.
Collapse
Affiliation(s)
- Ana Reis-Mendes
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - Félix Carvalho
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - Fernando Remião
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - Emília Sousa
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemistry, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
- CIIMAR⁻Interdisciplinary Centre of Marine and Environmental Research, 4450-208 Porto, Portugal.
| | - Maria de Lourdes Bastos
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - Vera Marisa Costa
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
37
|
Lee BH, Suresh S, Ekpenyong A. Fluorescence intensity modulation of CdSe/ZnS quantum dots assesses reactive oxygen species during chemotherapy and radiotherapy for cancer cells. JOURNAL OF BIOPHOTONICS 2019; 12:e201800172. [PMID: 30315626 DOI: 10.1002/jbio.201800172] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 09/02/2018] [Accepted: 10/11/2018] [Indexed: 06/08/2023]
Abstract
Quantum dots (QDs) are semiconductor nanoparticles ranging in size from 2 to 10 nm. QDs are increasingly being developed for biomedical imaging, targeted drug delivery and green energy technology. These have led to much research on QD interactions with various physical, chemical and biological systems. For biological systems, research has focused on the biocompatibility/cytotoxicity of QDs in the context of imaging/therapy. However, there is a paucity of work on how biological systems and bioactive molecules might be used to alter the optoelectronic properties of QDs. Here, it is shown that these properties can be altered by reactive oxygen species (ROS) from chemotherapeutic media and biological cells following controlled changes in cellular activities. Using CdSe/ZnS core-shell QDs, spectroscopic analysis of optically excited QDs with HL60, K562 and T98G cancer cell lines is performed. Our results show statistically significant (P < 0.0001) modulation of the fluorescence emission spectra of the QDs due to the ROS produced by common chemotherapeutic drugs, daunorubicin and doxorubicin and by cells following chemotherapy/radiotherapy. This optical modulation, in addition to assessing ROS generation, will possibly enhance applications of QDs in simultaneous diagnostic imaging and nanoparticle-mediated drug delivery as well as simultaneous ROS assessment and radiosensitization for improved outcomes in cancer treatments. Reactive molecular species produced by biological cells and chemotherapeutic drugs can create electric fields that alter the photophysical properties of QDs, and this can be used for concurrent monitoring of cellular activities, while inducing changes in those cellular activities.
Collapse
Affiliation(s)
- Bong H Lee
- Department of Physics, Creighton University, Omaha, Nebraska
| | - Sindhuja Suresh
- Department of Computer Science, Creighton University, Omaha, Nebraska
| | | |
Collapse
|
38
|
Zhang G, Qiao J, Liu X, Liu Y, Wu J, Huang L, Ji D, Guan Q. Interactions of Self-Assembled Bletilla S triata Polysaccharide Nanoparticles with Bovine Serum Albumin and Biodistribution of Its Docetaxel-Loaded Nanoparticles. Pharmaceutics 2019; 11:E43. [PMID: 30669500 PMCID: PMC6358745 DOI: 10.3390/pharmaceutics11010043] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 01/12/2019] [Accepted: 01/15/2019] [Indexed: 01/01/2023] Open
Abstract
: Amphiphilic copolymers of stearic acid (SA)-modified Bletilla striata polysaccharides (BSPs-SA) with three different degrees of substitution (DSs) were synthesized. The effects of DS values on the properties of BSPs-SA nanoparticles were evaluated. Drug state, cytotoxicity, and histological studies were carried out. The affinity ability of bovine serum albumin (BSA) and the BSPs-SA nanoparticles was also characterized utilizing ultraviolet and fluorescence spectroscopy. Besides, the bioavailability and tissue distribution of docetaxel (DTX)-loaded BSPs-SA nanoparticles were also assessed. The results demonstrated that the DS increase of the hydrophobic stearic acid segment increased the negative charge, encapsulation efficiency, and drug-loading capacity while decreasing the critical aggregation concentration value as well as the release rate of docetaxel from the nanoparticles. Docetaxel was encapsulated in nanoparticles at the small molecules or had an amorphous status. The inhibitory capability of DTX-loaded BSPs-SA nanoparticles against 4T1 tumor cells was superior to that of Duopafei®. The ultraviolet and fluorescence results exhibited a strong binding affinity between BSPs-SA nanoparticles and bovine serum albumin, but the conformation of bovine serum albumin was not altered. Additionally, the area under the concentration⁻time curve (AUC₀⁻∞) of DTX-loaded BSPs-SA nanoparticles was about 1.42-fold higher compared with Duopafei® in tumor-bearing mice. Docetaxel levels of DTX-loaded BSPs-SA nanoparticles in some organs changed, and more docetaxel accumulated in the liver, spleen, and the tumor compared with Duopafei®. The experimental results provided a theoretical guidance for further applications of BSPs-SA conjugates as nanocarriers for delivering anticancer drugs.
Collapse
Affiliation(s)
- Guangyuan Zhang
- Department of Pharmaceutics, School of Pharmacy, Jilin University, Changchun 130012, China.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Mei C, Chao CW, Lin CW, Li ST, Wu KH, Yang KC, Yu J. Three-dimensional spherical gelatin bubble-based scaffold improves the myotube formation of H9c2 myoblasts. Biotechnol Bioeng 2019; 116:1190-1200. [PMID: 30636318 DOI: 10.1002/bit.26917] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 11/19/2018] [Accepted: 01/06/2019] [Indexed: 01/09/2023]
Abstract
Microenvironmental factors including physical and chemical cues can regulate stem cells as well as terminally differentiated cells to modulate their biological function and differentiation. However, one of the physical cues, the substrate's dimensionality, has not been studied extensively. In this study, the flow-focusing method with a microfluidic device was used to generate gelatin bubbles to fabricate highly ordered three-dimensional (3D) scaffolds. Rat H9c2 myoblasts were seeded into the 3D gelatin bubble-based scaffolds and compared to those grown on 2D gelatin-coating substrates to demonstrate the influences of spatial cues on cell behaviors. Relative to cells on the 2D substrates, the H9c2 myoblasts were featured by a good survival and normal mitochondrial activity but slower cell proliferation within the 3D scaffolds. The cortical actin filaments of H9c2 cells were localized close to the cell membrane when cultured on the 2D substrates, while the F-actins distributed uniformly and occupied most of the cell cytoplasm within the 3D scaffolds. H9c2 myoblasts fused as multinuclear myotubes within the 3D scaffolds without any induction but cells cultured on the 2D substrates had a relatively lower fusion index even differentiation medium was provided. Although there was no difference in actin α 1 and myosin heavy chain 1, H9c2 cells had a higher myogenin messenger RNA level in the 3D scaffolds than those of on the 2D substrates. This study reveals that the dimensionality influences differentiation and fusion of myoblasts.
Collapse
Affiliation(s)
- Chieh Mei
- Department of Chemical Engineering, College of Engineering, National Taiwan University, Taipei, Taiwan
| | - Chih-Wei Chao
- Department of Chemical Engineering, College of Engineering, National Taiwan University, Taipei, Taiwan
| | - Che-Wei Lin
- Department of Chemical Engineering, College of Engineering, National Taiwan University, Taipei, Taiwan
| | - Shing Tak Li
- Department of Chemical Engineering, College of Engineering, National Taiwan University, Taipei, Taiwan
| | - Kuan-Han Wu
- Department of Chemical Engineering, College of Engineering, National Taiwan University, Taipei, Taiwan
| | - Kai-Chiang Yang
- School of Dental Technology, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan.,Laboratory of Organ and Tissue Reconstruction, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Jiashing Yu
- Department of Chemical Engineering, College of Engineering, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
40
|
Pereira-Oliveira M, Reis-Mendes A, Carvalho F, Remião F, Bastos MDL, Costa VM. Doxorubicin Is Key for the Cardiotoxicity of FAC (5-Fluorouracil + Adriamycin + Cyclophosphamide) Combination in Differentiated H9c2 Cells. Biomolecules 2019; 9:21. [PMID: 30634681 PMCID: PMC6358964 DOI: 10.3390/biom9010021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 01/03/2019] [Indexed: 12/17/2022] Open
Abstract
Currently, a common therapeutic approach in cancer treatment encompasses a drug combination to attain an overall better efficacy. Unfortunately, it leads to a higher incidence of severe side effects, namely cardiotoxicity. This work aimed to assess the cytotoxicity of doxorubicin (DOX, also known as Adriamycin), 5-fluorouracil (5-FU), cyclophosphamide (CYA), and their combination (5-Fluorouracil + Adriamycin + Cyclophosphamide, FAC) in H9c2 cardiac cells, for a better understanding of the contribution of each drug to FAC-induced cardiotoxicity. Differentiated H9c2 cells were exposed to pharmacological relevant concentrations of DOX (0.13⁻5 μM), 5-FU (0.13⁻5 μM), CYA (0.13⁻5 μM) for 24 or 48 h. Cells were also exposed to FAC mixtures (0.2, 1 or 5 μM of each drug and 50 μM 5-FU + 1 μM DOX + 50 μM CYA). DOX was the most cytotoxic drug, followed by 5-FU and lastly CYA in both cytotoxicity assays (reduction of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) and neutral red (NR) uptake). Concerning the equimolar combination with 1 or 5 μM, FAC caused similar cytotoxicity to DOX alone. Even in the presence of higher concentrations of 5-FU and CYA (50 μM 5-FU + 1 μM DOX + 50 μM CYA), 1 μM DOX was still a determinant for the cardiotoxicity observed in the cytotoxicity assays, phase contrast morphological evaluation, and mitochondrial potential depolarization evaluation. To the best of our knowledge, this was the first in vitro work with this combination regimen, DOX being the most toxic drug and key to the toxicity of FAC.
Collapse
Affiliation(s)
- Maria Pereira-Oliveira
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Ana Reis-Mendes
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Félix Carvalho
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Fernando Remião
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Maria de Lourdes Bastos
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Vera Marisa Costa
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| |
Collapse
|
41
|
Ferreira LL, Cunha-Oliveira T, Veloso CD, Costa CF, Wallace KB, Oliveira PJ. Single nanomolar doxorubicin exposure triggers compensatory mitochondrial responses in H9c2 cardiomyoblasts. Food Chem Toxicol 2018; 124:450-461. [PMID: 30557669 DOI: 10.1016/j.fct.2018.12.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 12/08/2018] [Accepted: 12/13/2018] [Indexed: 12/31/2022]
Abstract
Dose-dependent and cumulative cardiotoxicity associated with doxorubicin (DOX) is the main limitation of anticancer therapy. Pediatric cancer survivors are particularly vulnerable, and no effective prevention measures are available. The aim of the present study was to investigate the persistent effects of nanomolar DOX concentrations and determine whether a pretreatment would induce mitochondrial adaptations in H9c2 cardiomyoblasts. H9c2 cells were incubated with DOX (10 and 25 nM) for 24 h, followed by 9 days of recovery in drug-free medium. We found that the sub-therapeutic DOX treatment induced persistent hypertrophy and dose-dependent cell cycle arrest in G2/M. Glycolytic activity, indirectly based on extracellular acidification rate, and basal respiration were significantly decreased in DOX-treated cells compared to controls, although both groups showed similar maximal respiration. Additionally, nanomolar DOX pretreatment resulted in upregulation of mitochondrial DNA transcripts accompanied by a decrease in DNA methyltransferase 1 (DNMT1) and global methylation levels. Finally, the pretreatment with DOX ameliorated H9c2 cells resistance against a subsequent exposure to DOX. These results suggest that nanomolar DOX pretreatment induced a beneficial and possibly epigenetic-based mitochondrial adaptation, raising the possibility that an early sub-therapeutic DOX treatment can be used as a preconditioning and protective approach during anticancer therapies.
Collapse
Affiliation(s)
- Luciana L Ferreira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, UC Biotech Building, Biocant Park, 3060-197, Cantanhede, Portugal.
| | - Teresa Cunha-Oliveira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, UC Biotech Building, Biocant Park, 3060-197, Cantanhede, Portugal.
| | - Caroline D Veloso
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, UC Biotech Building, Biocant Park, 3060-197, Cantanhede, Portugal.
| | - Cláudio F Costa
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, UC Biotech Building, Biocant Park, 3060-197, Cantanhede, Portugal.
| | - Kendall B Wallace
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN, 55812, USA.
| | - Paulo J Oliveira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, UC Biotech Building, Biocant Park, 3060-197, Cantanhede, Portugal.
| |
Collapse
|
42
|
Zuo YH, Han QB, Dong GT, Yue RQ, Ren XC, Liu JX, Liu L, Luo P, Zhou H. Panax ginseng Polysaccharide Protected H9c2 Cardiomyocyte From Hypoxia/Reoxygenation Injury Through Regulating Mitochondrial Metabolism and RISK Pathway. Front Physiol 2018; 9:699. [PMID: 29962955 PMCID: PMC6013582 DOI: 10.3389/fphys.2018.00699] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 05/18/2018] [Indexed: 12/13/2022] Open
Abstract
Background and Objective: Ischemic heart disease (IHD) has been the major issue of public health. Panax ginseng (ginseng) has been verified as an effective traditional Chinese medicines and exerted cardioprotective effect. This study aimed to investigate the polysaccharide fraction of ginseng on hypoxia/reoxygenation (H/R) injury in cardiomyocytes and the underlying mechanisms. Methods: Ginseng was extracted by ethanol and fractionated by high-speed counter current chromatography (HSCCC) and column separation. The cardioprotective effect was evaluated in H9c2 cardiomyocytes underwent H/R treatment. The cell viability, apoptosis and mitochondrial respiration were examined. Results: An acid polysaccharides fraction of ginseng (AP1) was identified the most effective fraction in protecting cardiomyocytes from H/R injury. AP1 restored the mitochondrial function by maintaining mitochondrial membrane potential (MMP), blocking the release of cytochrome C, and increasing the ATP generation and oxygen consumption rate (OCR) of cardiomyocytes. Meanwhile, AP1 induced the expression of glucocorticoid receptor (GR) and estrogen receptor (ER) which further activated reperfusion injury salvage kinase (RISK) pathway. Finally, AP1 increased nitric oxide (NO) production and regulated endothelial function by increasing endothelial NO synthase (eNOS) expression and decreasing inducible NOS (iNOS) expression in H/R injury. Conclusion: The results suggested that AP1 exerted a protective effect in myocardial H/R injury mainly through maintaining myocardial mitochondrial function, thereby inhibiting myocardial H/R caused apoptosis and increasing the expressions of GR and ER, which in turn mediated the activation of RISK pathway and eNOS-dependent mechanism to resist the reperfusion injury.
Collapse
Affiliation(s)
- Yi-Han Zuo
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Quan-Bin Han
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Geng-Ting Dong
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Rui-Qi Yue
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Xue-Cong Ren
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Jian-Xin Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China.,College of Pharmacy, Hunan University of Medicine, Huaihua, China
| | - Liang Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Pei Luo
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Hua Zhou
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China.,International Institute of Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
43
|
Cunha-Oliveira T, Ferreira LL, Coelho AR, Deus CM, Oliveira PJ. Doxorubicin triggers bioenergetic failure and p53 activation in mouse stem cell-derived cardiomyocytes. Toxicol Appl Pharmacol 2018; 348:1-13. [PMID: 29653124 DOI: 10.1016/j.taap.2018.04.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 04/06/2018] [Accepted: 04/08/2018] [Indexed: 01/28/2023]
Abstract
Doxorubicin (DOX) is a widely used anticancer drug that could be even more effective if its clinical dosage was not limited because of delayed cardiotoxicity. Beating stem cell-derived cardiomyocytes are a preferred in vitro model to further uncover the mechanisms of DOX-induced cardiotoxicity. Our objective was to use cultured induced-pluripotent stem cell(iPSC)-derived mouse cardiomyocytes (Cor.At) to investigate the effects of DOX on cell and mitochondrial metabolism, as well as on stress responses. Non-proliferating and beating Cor.At cells were treated with 0.5 or 1 μM DOX for 24 h, and morphological, functional and biochemical changes associated with mitochondrial bioenergetics, DNA-damage response and apoptosis were measured. Both DOX concentrations decreased ATP levels and SOD2 protein levels and induced p53-dependent caspase activation. However, differential effects were observed for the two DOX concentrations. The highest concentration induced a high degree of apoptosis, with increased nuclear apoptotic morphology, PARP-1 cleavage and decrease of some OXPHOS protein subunits. At the lowest concentration, DOX increased the expression of p53 target transcripts associated with mitochondria-dependent apoptosis and decreased transcripts related with DNA-damage response and glycolysis. Interestingly, cells treated with 0.5 μM DOX presented an increase in PDK4 transcript levels, accompanied by an increase in phospho-PDH and decreased PDH activity. This was accompanied by an apparent decrease in basal and maximal oxygen consumption rates (OCR) and in basal extracellular acidification rate (ECAR). Cells pre-treated with the PDK inhibitor dichloroacetate (DCA), with the aim of restoring PDH activity, partially recovered OCR and ECAR. The results suggest that the higher DOX concentration mainly induces p53-dependent apoptosis, whereas for the lower DOX concentration the cardiotoxic effects involve bioenergetic failure, unveiling PDH as a possible therapeutic target to decrease DOX cardiotoxicity.
Collapse
Affiliation(s)
- Teresa Cunha-Oliveira
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra, UC-Biotech Building, Biocant Park, Cantanhede, Portugal.
| | - Luciana L Ferreira
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra, UC-Biotech Building, Biocant Park, Cantanhede, Portugal
| | - Ana Raquel Coelho
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra, UC-Biotech Building, Biocant Park, Cantanhede, Portugal; Institute for Interdisciplinary Research (I.I.I.), University of Coimbra, 3030-789 Coimbra, Portugal
| | - Cláudia M Deus
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra, UC-Biotech Building, Biocant Park, Cantanhede, Portugal; Institute for Interdisciplinary Research (I.I.I.), University of Coimbra, 3030-789 Coimbra, Portugal
| | - Paulo J Oliveira
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra, UC-Biotech Building, Biocant Park, Cantanhede, Portugal; Institute for Interdisciplinary Research (I.I.I.), University of Coimbra, 3030-789 Coimbra, Portugal
| |
Collapse
|
44
|
Detecting the functional complexities between high-density lipoprotein mimetics. Biomaterials 2018; 170:58-69. [PMID: 29653287 DOI: 10.1016/j.biomaterials.2018.04.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 04/03/2018] [Accepted: 04/05/2018] [Indexed: 01/05/2023]
Abstract
High-density lipoprotein (HDL) is a key regulator of lipid homeostasis through its native roles like reverse cholesterol transport. The reconstitution of this natural nanoparticle (NP) has become a nexus between nanomedicine and multi-disease therapies, for which a major portion of HDL functionality is attributed to its primary scaffolding protein, apolipoprotein A1 (apoA1). ApoA1-mimetic peptides were formulated as cost-effective alternatives to apoA1-based therapies; reverse-4F (r4F) is one such peptide used as part of a nanoparticle platform. While similarities between r4F- and apoA1-based HDL-mimetic nanoparticles have been identified, key functional differences native to HDL have remained undetected. In the present study, we executed a multidisciplinary approach to uncover these differences by exploring the form, function, and medical applicability of engineered HDL-mimetic NPs (eHNPs) made from r4F (eHNP-r4F) and from apoA1 (eHNP-A1). Comparative analyses of the eHNPs through computational molecular dynamics (MD), advanced microfluidic NP synthesis and screening technologies, and in vivo animal model studies extracted distinguishable eHNP characteristics: the eHNPs share identical structural and compositional characteristics with distinct differences in NP stability and organization; eHNP-A1 could more significantly stimulate anti-inflammatory responses characteristic of the scavenger receptor class B type 1 (SR-B1) mediated pathways; and eHNP-A1 could outperform eHNP-r4F in the delivery of a model hydrophobic drug to an in vivo tumor. The biomimetic microfluidic technologies and MD simulations uniquely enabled our comparative analysis through which we determined that while eHNP-r4F is a capable NP with properties mimicking natural eHNP-A1, challenges remain in reconstituting the full functionality of NPs naturally derived from humans.
Collapse
|
45
|
Marques-Aleixo I, Santos-Alves E, Oliveira PJ, Moreira PI, Magalhães J, Ascensão A. The beneficial role of exercise in mitigating doxorubicin-induced Mitochondrionopathy. Biochim Biophys Acta Rev Cancer 2018; 1869:189-199. [PMID: 29408395 DOI: 10.1016/j.bbcan.2018.01.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 01/09/2018] [Accepted: 01/11/2018] [Indexed: 01/07/2023]
Abstract
Doxorubicin (DOX) is a widely used antineoplastic agent for a wide range of cancers, including hematological malignancies, soft tissue sarcomas and solid tumors. However, DOX exhibits a dose-related toxicity that results in life-threatening cardiomyopathy. In addition to the heart, there is evidence that DOX toxicity extends to other organs. This general toxicity seems to be related to mitochondrial network structural, molecular and functional impairments. Several countermeasures for these negative effects have been proposed, being physical exercise, not only one of the most effective non-pharmacologic strategy but also widely recommended as booster against cancer-related fatigue. It is widely accepted that mitochondria are critical sensors of tissue functionality, both modulated by DOX and exercise. Therefore, this review focuses on the current understanding of the mitochondrial-mediated mechanisms underlying the protective effect of exercise against DOX-induced toxicity, not only limited to the cardiac tissue, but also in other tissues such as skeletal muscle, liver and brain. We here analyze recent developments regarding the beneficial effects of exercise targeting mitochondrial responsive phenotypes against redox changes, mitochondrial bioenergetics, apoptotic, dynamics and quality control signalling affected by DOX treatment.
Collapse
Affiliation(s)
- I Marques-Aleixo
- CIAFEL - Research Centre in Physical Activity, Health and Leisure, Portugal; LAMETEX - Laboratory of Exercise and Metabolism; Faculty of Psychology, Education and Sport, University Lusófona of Porto, Portugal.
| | - E Santos-Alves
- CIAFEL - Research Centre in Physical Activity, Health and Leisure, Portugal; LAMETEX - Laboratory of Exercise and Metabolism; Departament de Biologia Cellular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, Spain
| | - P J Oliveira
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, UC Biotech Building, Biocant Park, Cantanhede, Portugal
| | - P I Moreira
- CNC - Centre for Neuroscience and Cell Biology, University of Coimbra, Portugal; Institute of Physiology, Faculty of Medicine, University of Coimbra, Portugal
| | - J Magalhães
- CIAFEL - Research Centre in Physical Activity, Health and Leisure, Portugal; LAMETEX - Laboratory of Exercise and Metabolism; Faculty of Sport, University of Porto, Portugal
| | - A Ascensão
- CIAFEL - Research Centre in Physical Activity, Health and Leisure, Portugal; LAMETEX - Laboratory of Exercise and Metabolism; Faculty of Sport, University of Porto, Portugal
| |
Collapse
|
46
|
Bishop S, Liu SJ. Cardioprotective action of the aqueous extract of Terminalia arjuna bark against toxicity induced by doxorubicin. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2017; 36:210-216. [PMID: 29157817 DOI: 10.1016/j.phymed.2017.10.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 08/03/2017] [Accepted: 10/10/2017] [Indexed: 06/07/2023]
Abstract
BACKGROUND The aqueous extract of Terminalia arjuna (TA) bark (TAAqE) has been shown to have a direct inotropic effect on ventricular myocytes. Active constituents of TAAqE contain various flavonoids and proanthocyanidins, some of which are known to have antioxidant activities. Whether TAAqE affords a cardioprotective action against oxidative stress (OS) remains unclear. PURPOSE Increased OS is one of the major mechanisms underlying cardiotoxicity induced by doxorubicin (DOX), a commonly-used anticancer agent. The aim of the present study was to investigate potential cardioprotective effect of TAAqE against DOX-induced OS and cardiac dysfunction. METHODS OS and cytotoxicity were induced by 1 µM DOX for 24 h in H9c2 cells, a cardiac tissue-derived cell line, and left ventricular (LV) dysfunction was induced by intrapleural injection of DOX (accumulative 20 mg/kg body weight) to mice. Cellular oxidative levels and morphology were assessed using microscopy and oxidative-sensitive fluorescent dyes with and without co-treatment with TAAqE. LV function was monitored weekly with echocardiography. RESULTS TAAqE reduced OS and preserved mitochondria and cell growth of H9c2 cells against DOX treatment. TAAqE (in drinking water) attenuated the decreased LV function and altered myocardial structure caused by DOX treatment. CONCLUSION TAAqE exerts a protective action against cardiotoxicity caused by DOX in part via suppression of OS. Thus, TAAqE is a promising cardiotonic in adjuvant cancer chemotherapy.
Collapse
Affiliation(s)
- Sarah Bishop
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Shi J Liu
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA; Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| |
Collapse
|
47
|
Zhou B, Tan J, Zhang C, Wu Y. Neuroprotective effect of polysaccharides from Gastrodia elata blume against corticosterone‑induced apoptosis in PC12 cells via inhibition of the endoplasmic reticulum stress‑mediated pathway. Mol Med Rep 2017; 17:1182-1190. [PMID: 29115511 DOI: 10.3892/mmr.2017.7948] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 09/18/2017] [Indexed: 11/05/2022] Open
Abstract
Depression is a common mental health disorder and is the leading cause of disability worldwide. Gastrodia elata (G. elata) was demonstrated to exhibit a neuroprotective effect in the authors' previous study. The present study investigated the effect of polysaccharides from G. elata (GEP) on PC12 cell apoptosis induced by corticosterone (CORT) and its possible underlying mechanisms. PC12 cells were treated with 200 µM CORT in the absence or presence of different concentrations of GEP for 48 h. Then, cell viability was measured by CCK‑8 assay. The lactate dehydrogenase (LDH) leakage was quantified using an LDH assay kit. The apoptosis degree of the PC12 cells and the morphology was measured by DAPI staining. Subsequently, intracellular ROS level was detected by using DCFH‑DA method, the morphology staining of the endoplasmic reticulum in PC12 cells was determined using the cationic probe, and levels of five proteins involved in apoptosis, i.e., glucose‑regulated protein, 78k Da (GRP78), X‑box binding protein 1 (XBP‑1), growth arrest‑ and DNA damage‑inducible gene 153 (GADD153), caspase 9 and caspase 12 were determined by western blotting. The results demonstrated that treatment with 1,000 µg/ml GEP prior to 200 µM CORT exposure significantly protected the PC12 cells from CORT‑induced cell apoptosis, and reduced levels of LDH leakage and intracellular reactive oxygen species. In addition, pretreatment with GEP inhibited the activation of GRP78, X‑BP‑1, GADD153, caspase 9 and caspase 12. These findings suggested that GEP exhibited a neuroprotective effect against CORT‑induced apoptosis in PC12 cells, and the underlying molecular mechanisms were dependent on inhibition of the endoplasmic reticulum stress‑mediated pathway. This provides novel insight into the effect of GEP when used for the treatment of diseases of the nervous system.
Collapse
Affiliation(s)
- Benhong Zhou
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jun Tan
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Chan Zhang
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Yue Wu
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
48
|
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
|
49
|
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
|
50
|
Priya LB, Baskaran R, Huang CY, Padma VV. Neferine ameliorates cardiomyoblast apoptosis induced by doxorubicin: possible role in modulating NADPH oxidase/ROS-mediated NFκB redox signaling cascade. Sci Rep 2017; 7:12283. [PMID: 28947826 PMCID: PMC5612945 DOI: 10.1038/s41598-017-12060-9] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 08/29/2017] [Indexed: 12/04/2022] Open
Abstract
Doxorubicin (DOX) mediated cardiomyopathy is a major challenge in cancer chemotherapy. Redox-cycling of doxorubicin by flavoenzymes makes the heart more vulnerable to oxidative stress leading to cardiac dysfunction. The present study evaluates the role of neferine, a bisbenzylisoquinoline alkaloid, in curbing the molecular consequences of DOX-exposure in H9c2 cardiomyoblasts. Neferine pre-treatment increased cell viability upon DOX-exposure. DOX activates NADPH oxidase subunits, (p22phox, p47phox, gp91phox) as the primary event followed by peak in [Ca2+]i accumulation by 2 h, ROS by 3 h and activated ERK1/2 and p38 MAPKinases, time dependently along with the activation and translocation of NFκB and up-regulated COX2 and TNF-α expressions. Neferine pre-treatment modulated NADPH oxidase/ROS system, inhibited MAPKinases and NFκB activation, reduced sub G1 cell population and concomitantly increased cyclin D1 expression reducing DOX-mediated apoptosis. The study demonstrates for the first time, the molecular sequential events behind DOX toxicity and the mechanism of protection offered by neferine with specific relevance to NADPH oxidase system, MAPKinases, inflammation and apoptosis in H9c2 cells. Our data suggests the use of neferine as a new approach in pharmacological interventions against cardiovascular disorders as secondary complications.
Collapse
Affiliation(s)
- Lohanathan Bharathi Priya
- Translational Research Laboratory, Department of Biotechnology, School of Biotechnology and Genetic Engineering, Bharathiar University, Coimbatore, 641046, Tamil Nadu, India
| | - Rathinasamy Baskaran
- National Institute of Cancer Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | - Chih-Yang Huang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
- Graduate Institute of Chinese Medical Science, China Medical University, Taichung, Taiwan
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan
| | - Viswanadha Vijaya Padma
- Translational Research Laboratory, Department of Biotechnology, School of Biotechnology and Genetic Engineering, Bharathiar University, Coimbatore, 641046, Tamil Nadu, India.
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan.
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan.
| |
Collapse
|