1
|
Feng W, Wang Q, Tan Y, Qiao J, Liu Q, Yang B, Yang S, Cui L. Early detection of anthracycline-induced cardiotoxicity. Clin Chim Acta 2025; 565:120000. [PMID: 39401650 DOI: 10.1016/j.cca.2024.120000] [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: 09/14/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024]
Abstract
Although anthracyclines are important anticancer agents, their use is limited due to various adverse effects, particularly cardiac toxicity. Mechanisms underlying anthracycline-induced cardiotoxicity (AIC) are complex. Given the irreplaceable role of anthracyclines in treatment of malignancies and other serious diseases, early monitoring of AIC is paramount. In recent years, multiple studies have investigated various biomarkers for early detection of AIC. Currently, the two most common are cardiac troponin and B-type natriuretic peptide. In addition, a range of other molecules, including RNAs, myeloperoxidase (MPO), C-reactive protein (CRP), various genes, and others, also play roles in AIC prediction. Unfortunately, current research indicates a need to validate their sensitivity and specificity of these biomarkers especially in large study populations. In this review, we summarize the mechanisms and potential biomarkers of AIC, although some remain preliminary.
Collapse
Affiliation(s)
- Weimin Feng
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Qingchen Wang
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Yuan Tan
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Jiao Qiao
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Qi Liu
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Boxin Yang
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Shuo Yang
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Liyan Cui
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| |
Collapse
|
2
|
Sahoo S, Kumari S, Pulipaka S, Chandra Y, Kotamraju S. SIRT1 promotes doxorubicin-induced breast cancer drug resistance and tumor angiogenesis via regulating GSH-mediated redox homeostasis. Mol Carcinog 2024; 63:2291-2304. [PMID: 39136605 DOI: 10.1002/mc.23809] [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: 04/30/2024] [Revised: 07/04/2024] [Accepted: 08/02/2024] [Indexed: 11/16/2024]
Abstract
Sirtuin 1 (SIRT1), a member of histone deacetylase III family, plays a pivotal role in mediating chemoresistance in several cancers, including breast cancer. However, the molecular mechanism by which the deregulated SIRT1 promotes doxorubicin (Dox) resistance is still elusive. Here, we showed that the cell proliferation rates and invasive properties of MDA-MB-231 breast cancer cells were increased from low- to high-Dox-resistant cells. In agreement, severe combined immunodeficiency disease (SCID) mice bearing labeled MDA-MB-231high Dox-Res cells showed significantly higher tumor growth, angiogenesis, and metastatic ability than parental MDA-MB-231 cells. Interestingly, the levels of SIRT1 and glutathione (GSH) were positively correlated with the degree of Dox-resistance. Dox-induced SIRT1 promoted NRF2 nuclear translocation with an accompanying increase in the antioxidant response element promotor activity and GSH levels. In contrast, inhibition of SIRT1 by EX527 greatly reversed these events. More so, Dox-resistance-induced pro-proliferative, proangiogenic, and invasive effects were obviated with depletion of either SIRT1 or GSH. Together, Dox-induced SIRT1 promotes dysregulation of redox homeostasis leading to breast cancer chemoresistance, tumor aggressiveness, angiogenesis, and metastasis.
Collapse
Affiliation(s)
- Shashikanta Sahoo
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Sunita Kumari
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Sriravali Pulipaka
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Yogesh Chandra
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
| | - Srigiridhar Kotamraju
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| |
Collapse
|
3
|
Scalise M, Cianflone E, Quercia C, Pagano L, Chiefalo A, Stincelli A, Torella A, Puccio B, Santamaria G, Guzzi HP, Veltri P, De Angelis A, Urbanek K, Ellison-Hughes GM, Torella D, Marino F. Senolytics rejuvenate aging cardiomyopathy in human cardiac organoids. Mech Ageing Dev 2024; 223:112007. [PMID: 39622416 DOI: 10.1016/j.mad.2024.112007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/19/2024] [Accepted: 11/21/2024] [Indexed: 12/10/2024]
Abstract
BACKGROUND Human cardiac organoids closely replicate the architecture and function of the human heart, offering a potential accurate platform for studying cellular and molecular features of aging cardiomyopathy. Senolytics have shown potential in addressing age-related pathologies but their potential to reverse aging-related human cardiomyopathy remains largely unexplored. METHODS We employed human iPSC-derived cardiac organoids (hCOs/hCardioids) to model doxorubicin(DOXO)-induced cardiomyopathy in an aged context. hCardioids were treated with DOXO and subsequently with a combination of two senolytics: dasatinib (D) and quercetin (Q). RESULTS DOXO-treated hCardioids exhibited significantly increased oxidative stress, DNA damage (pH2AX), cellular senescence (p16INK4A) and decreased cell proliferation associated with a senescence-associated secretory phenotype (SASP). DOXO-treated hCardioids were considerably deprived of cardiac progenitors and displayed reduced cardiomyocyte proliferation as well as contractility. These distinctive aging-associated characteristics were confirmed by global RNA-sequencing analysis. Treatment with D+Q reversed these effects, reducing oxidative stress and senescence markers, alleviating SASP, and restoring hCardioids viability and function. Additionally, senolytics replenished cardiac progenitors and reversed the cardiomyocyte proliferation deficit. CONCLUSIONS Doxorubicin triggers an age-associated phenotype in hCardioids reliably modelling the main cellular and molecular features of aging cardiomyopathy. Senescence is a key mechanism of the aged-hCOs phenotype as senolytics rejuvenated aged-hCardioids restoring their structure and function while reverting the age-associated regenerative deficit.
Collapse
Affiliation(s)
- Mariangela Scalise
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro 88100, Italy; Centre for Human and Applied Physiological, School of Basic and Medical Biosciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Eleonora Cianflone
- Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro 88100, Italy.
| | - Claudia Quercia
- Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro 88100, Italy
| | - Loredana Pagano
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro 88100, Italy
| | - Antonio Chiefalo
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro 88100, Italy
| | - Antonio Stincelli
- Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro 88100, Italy
| | - Annalaura Torella
- Department of Experimental Medicine, University of Campania "L. Vanvitelli", Naples 80138, Italy
| | - Barbara Puccio
- Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro 88100, Italy
| | - Gianluca Santamaria
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro 88100, Italy
| | - Hiram P Guzzi
- Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro 88100, Italy
| | - Pierangelo Veltri
- DIMES Department of Informatics, Modeling, Electronics and Systems, UNICAL, Rende, Cosenza, Italy
| | - Antonella De Angelis
- Department of Experimental Medicine, University of Campania "L. Vanvitelli", Naples 80138, Italy
| | - Konrad Urbanek
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", and CEINGE-Advanced Biotechnologies, Naples 80131, Italy
| | - Georgina M Ellison-Hughes
- Centre for Human and Applied Physiological, School of Basic and Medical Biosciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Daniele Torella
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro 88100, Italy.
| | - Fabiola Marino
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro 88100, Italy
| |
Collapse
|
4
|
Liu X, Guo Y, Huang Y, Wang Q, Huang Y, Lei Y, Liu Z, Zhang L. GPX4 allosteric activators inhibit ferroptosis and exert myocardial protection in doxorubicin-induced myocardial injury mouse model. Eur J Med Chem 2024; 277:116721. [PMID: 39096818 DOI: 10.1016/j.ejmech.2024.116721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 08/05/2024]
Abstract
Ferroptosis is a unique type of non-apoptotic form of cell death characterized by increased lipid hydroperoxide levels. It has relevance for a number of pathological conditions including multiple organ injuries and degenerative diseases. GPX4 plays an important role in ferroptosis by repairing lipid hydroperoxides. Based on the reported allosteric sites, we obtained the GPX4 allosteric activator hit compound A9 through virtual screening. A9 can bind to GPX4 and prevent RSL3-induced lipid peroxidation production in HT-1080 cells. In addition, A9 can specifically rescue erastin-induced cell death. Further chemical modification and structure-activity relationship studies afforded the optimized compound C3. C3 showed the activity of alleviating myocardial injury in the doxorubicin-induced myocardial injury mouse model. This study demonstrated that inhibiting ferroptosis by activating GPX4 is expected to be a potential solution to treat myocardial injury.
Collapse
Affiliation(s)
- Xiaoang Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Yi Guo
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Yusong Huang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Qian Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Yanwen Huang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Yizhi Lei
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Zhenming Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Liangren Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
| |
Collapse
|
5
|
Więckowska M, Cichon N, Szelenberger R, Gorniak L, Bijak M. Ochratoxin A and Its Role in Cancer Development: A Comprehensive Review. Cancers (Basel) 2024; 16:3473. [PMID: 39456567 PMCID: PMC11506779 DOI: 10.3390/cancers16203473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/05/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Ochratoxin A (OTA) is widely recognized for its broad spectrum of toxic effects and is classified as a potential human carcinogen, placed in group 2B by the International Agency for Research on Cancer (IARC). Its presence in food and beverages poses a significant health hazard. Extensive research has documented the efficient absorption and distribution of OTA throughout the body via the bloodstream and tissues, underscoring the associated health risk. Additionally, ongoing studies aim to clarify the link between OTA exposure and carcinogenesis. The obtained results indicate a strong correlation between OTA and renal cell carcinoma (RCC), with potential associations with other malignancies, including hepatocellular carcinoma (HCC), gallbladder cancer (GBC), and squamous cell carcinoma (SCC). OTA is implicated in oxidative stress, lipid peroxidation, apoptosis, DNA damage, adduct formation, miRNA deregulation, and distributions in the cell cycle, all of which may contribute to carcinogenesis. Conclusions: Despite significant research efforts, the topic remains inexhaustible and requires further investigation. The obtained results do not yield definitive conclusions, potentially due to species-specific differences in the animal models used and challenges in extrapolating these results to humans. In our review, we delve deeper into the potential mechanisms underlying OTA-induced carcinogenesis and discuss existing limitations, providing directions for future research.
Collapse
Affiliation(s)
| | - Natalia Cichon
- Biohazard Prevention Centre, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland; (M.W.); (R.S.); (L.G.); (M.B.)
| | | | | | | |
Collapse
|
6
|
Pal M, Bera A, Masarkar N, Upadhyay A, Mukherjee S, Roy M. Targeted Chemo-Phototherapy in Red Light with Novel Doxorubicin and Iron(III) Complex-Functionalized Gold Nanoconjugate (Dox-Fe@FA-AuNPs). Chem Asian J 2024; 19:e202400616. [PMID: 38923831 DOI: 10.1002/asia.202400616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 06/28/2024]
Abstract
The anticancer efficacy of doxorubicin, an anthracycline-based and FDA-approved chemotherapeutic drug, is significantly hindered by acquired chemoresistance and severe side effects despite its potent anticancer properties. To overcome these challenges, we developed an innovative therapeutic formulation that integrates targeted chemotherapy and phototherapy within a single platform using gold nanoparticles (AuNPs). This novel nanoconjugate, designated as Dox-Fe@FA-AuNPs, is co-functionalized with folic acid, doxorubicin, and an iron(III)-phenolate/carboxylate complex, enabling cancer-specific drug activation. Here, we report the synthesis, characterization, and comprehensive physico-chemical and biological evaluations of Dox-Fe@FA-AuNPs. The nanoconjugate exhibited excellent solubility, stability, and enhanced cellular uptake in folate receptor-positive cancer cells. The nanoconjugate was potently cytotoxic against HeLa and MDA-MB-231 cancer cells (HeLa: 105.5±16.52 μg mL-1; MDA-MB-231: 112.0±12.31 μg mL-1; MDA-MB-231 (3D): 156.31±19.35 μg mL-1) while less cytotoxic to the folate(-) cancer cells (MCF-7, A549 and HepG2). The cytotoxicity was attributed to the pH-dependent release of doxorubicin, which preferentially occurs in the acidic tumor microenvironment. Additionally, under red light irradiation, the nanoconjugate generated ROS, inducing caspase-3/7-dependent apoptosis with a photo-index (PI) >50, and inhibited cancer cell migration. Our findings underscore the potential of Dox-Fe@FA-AuNPs as a highly effective and sustainable platform for targeted chemo-phototherapy.
Collapse
Affiliation(s)
- Maynak Pal
- Department of Chemistry, National Institute of Technology Manipur, Langol, 795004, Imphal West, Manipur
| | - Arpan Bera
- Department of Inorganic and Physical Chemistry, Indian Institute of Science Bangalore, Bangalore, 560012, Karnataka
| | - Neha Masarkar
- Department of Biochemistry, AIIMS Bhopal, Saket Nagar, Bhopal, Madhya Pradesh
| | - Aarti Upadhyay
- Department of Inorganic and Physical Chemistry, Indian Institute of Science Bangalore, Bangalore, 560012, Karnataka
| | - Sukhes Mukherjee
- Department of Biochemistry, AIIMS Bhopal, Saket Nagar, Bhopal, Madhya Pradesh
| | - Mithun Roy
- Department of Chemistry, National Institute of Technology Manipur, Langol, 795004, Imphal West, Manipur
- Department of Chemistry, National Institute of Technology Agartala, Jirania, 799046, Tripura West
| |
Collapse
|
7
|
Xie L, Liu H, Zhang K, Pan Y, Chen M, Xue X, Wan G. Exploring the molecular mechanism of ginseng against anthracycline-induced cardiotoxicity based on network pharmacology, molecular docking and molecular dynamics simulation. Hereditas 2024; 161:31. [PMID: 39243097 PMCID: PMC11378563 DOI: 10.1186/s41065-024-00334-y] [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: 06/02/2024] [Accepted: 09/01/2024] [Indexed: 09/09/2024] Open
Abstract
BACKGROUND Previous clinical and basic studies have revealed that ginseng might have cardioprotective properties against anthracycline-induced cardiotoxicity (AIC). However, the underlying mechanism of ginseng action against AIC remains insufficiently understood. The aim of this study was to explore the related targets and pathways of ginseng against AIC using network pharmacology, molecular docking, cellular thermal shift assay (CETSA) and molecular dynamics (MD) simulations. RESULTS Fourteen drug-disease common targets were identified. Enrichment analysis showed that the AGE-RAGE in diabetic complications, fluid shear stress and atherosclerosis, and TNF signaling pathway were potentially involved in the action of ginseng against AIC. Molecular docking demonstrated that the core components including Kaempferol, beta-Sitosterol, and Fumarine had notable binding activity with the three core targets CCNA2, STAT1, and ICAM1. Furthermore, the stable complex of STAT1 and Kaempferol with favorable affinity was further confirmed by CETSA and MD simulation. CONCLUSIONS This study suggested that ginseng might exert their protective effects against AIC through the derived effector compounds beta-Sitosterol, Kaempferol and Fumarine by targeting CCNA2, STAT1, and ICAM1, and modulating AGE-RAGE in diabetic complications, fluid shear stress and atherosclerosis, and TNF signaling pathways.
Collapse
Affiliation(s)
- Lin Xie
- Department of Oncology, Institute of Medicine and Nursing, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Hanze Liu
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, 39 Chaoyang Road, Shiyan, Hubei, 442000, China
| | - Ke Zhang
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, 39 Chaoyang Road, Shiyan, Hubei, 442000, China
| | - Yijun Pan
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, 39 Chaoyang Road, Shiyan, Hubei, 442000, China
| | - Mengyao Chen
- Department of Oncology, Institute of Medicine and Nursing, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Xiangyue Xue
- Department of Oncology, Institute of Medicine and Nursing, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Guoxing Wan
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, 39 Chaoyang Road, Shiyan, Hubei, 442000, China.
| |
Collapse
|
8
|
Matusik K, Kamińska K, Sobiborowicz-Sadowska A, Borzuta H, Buczma K, Cudnoch-Jędrzejewska A. The significance of the apelinergic system in doxorubicin-induced cardiotoxicity. Heart Fail Rev 2024; 29:969-988. [PMID: 38990214 PMCID: PMC11306362 DOI: 10.1007/s10741-024-10414-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/27/2024] [Indexed: 07/12/2024]
Abstract
Cancer is the leading cause of death worldwide, and the number of cancer-related deaths is expected to increase. Common types of cancer include skin, breast, lung, prostate, and colorectal cancers. While clinical research has improved cancer therapies, these treatments often come with significant side effects such as chronic fatigue, hair loss, and nausea. In addition, cancer treatments can cause long-term cardiovascular complications. Doxorubicin (DOX) therapy is one example, which can lead to decreased left ventricle (LV) echocardiography (ECHO) parameters, increased oxidative stress in cellular level, and even cardiac fibrosis. The apelinergic system, specifically apelin and its receptor, together, has shown properties that could potentially protect the heart and mitigate the damages caused by DOX anti-cancer treatment. Studies have suggested that stimulating the apelinergic system may have therapeutic benefits for heart damage induced by DOX. Further research in chronic preclinical models is needed to confirm this hypothesis and understand the mechanism of action for the apelinergic system. This review aims to collect and present data on the effects of the apelinergic system on doxorubicin-induced cardiotoxicity.
Collapse
Affiliation(s)
- Katarzyna Matusik
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Katarzyna Kamińska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland.
| | - Aleksandra Sobiborowicz-Sadowska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Hubert Borzuta
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Kasper Buczma
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Agnieszka Cudnoch-Jędrzejewska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
9
|
Neefjes J, Gurova K, Sarthy J, Szabó G, Henikoff S. Chromatin as an old and new anticancer target. Trends Cancer 2024; 10:696-707. [PMID: 38825423 PMCID: PMC11479676 DOI: 10.1016/j.trecan.2024.05.005] [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: 01/13/2024] [Revised: 05/08/2024] [Accepted: 05/13/2024] [Indexed: 06/04/2024]
Abstract
Recent genome-wide analyses identified chromatin modifiers as one of the most frequently mutated classes of genes across all cancers. However, chemotherapies developed for cancers involving DNA damage remain the standard of care for chromatin-deranged malignancies. In this review we address this conundrum by establishing the concept of 'chromatin damage': the non-genetic damage to protein-DNA interactions induced by certain small molecules. We highlight anthracyclines, a class of chemotherapeutic agents ubiquitously applied in oncology, as an example of overlooked chromatin-targeting agents. We discuss our current understanding of this phenomenon and explore emerging chromatin-damaging agents as a basis for further studies to maximize their impact in modern cancer treatment.
Collapse
Affiliation(s)
- Jacques Neefjes
- Department of Cell and Chemical Biology and Oncode Institute, LUMC, Einthovenweg 20, 2333, ZC, Leiden, The Netherlands
| | - Katerina Gurova
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY, 14263, USA.
| | - Jay Sarthy
- Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Research Institute, 1920 Terry Ave, Seattle, WA 98109, USA
| | - Gábor Szabó
- Faculty of Medicine, Department of Biophysics and Cell Biology, University of Debrecen, Debrecen, Egyetem tér 1, 4032, Hungary
| | - Steven Henikoff
- Basic Sciences Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA 98109, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| |
Collapse
|
10
|
Cesca BA, Caverzan MD, Lamberti MJ, Ibarra LE. Enhancing Therapeutic Approaches in Glioblastoma with Pro-Oxidant Treatments and Synergistic Combinations: In Vitro Experience of Doxorubicin and Photodynamic Therapy. Int J Mol Sci 2024; 25:7525. [PMID: 39062770 PMCID: PMC11277534 DOI: 10.3390/ijms25147525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/27/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
Glioblastoma (GBM) is an aggressive brain cancer characterized by significant molecular and cellular heterogeneity, which complicates treatment efforts. Current standard therapies, including surgical resection, radiation, and temozolomide (TMZ) chemotherapy, often fail to achieve long-term remission due to tumor recurrence and resistance. A pro-oxidant environment is involved in glioma progression, with oxidative stress contributing to the genetic instability that leads to gliomagenesis. Evaluating pro-oxidant therapies in brain tumors is crucial due to their potential to selectively target and eradicate cancer cells by exploiting the elevated oxidative stress levels inherent in these malignant cells, thereby offering a novel and effective strategy for overcoming resistance to conventional therapies. This study investigates the therapeutic potential of doxorubicin (DOX) and photodynamic therapy (PDT) with Me-ALA, focusing on their effects on redox homeostasis. Basal ROS levels and antioxidant gene expression (NFE2L2, CAT, GSR) were quantitatively assessed across GBM cell lines, revealing significant variability probably linked to genetic differences. DOX and PDT treatments, both individually and in combination, were analyzed for their efficacy in inducing oxidative stress and cytotoxicity. An in silico analysis further explored the relationship between gene mutations and oxidative stress in GBM patients, providing insights into the molecular mechanisms underlying treatment responses. Our findings suggest that pro-oxidant therapies, such as DOX and PDT in combination, could selectively target GBM cells, highlighting a promising avenue for improving therapeutic outcomes in GBM.
Collapse
Affiliation(s)
- Bruno Agustín Cesca
- Departamento de Biología Molecular, Facultad de Ciencias Exactas, Fisicoquímicas y Naturales, Universidad Nacional de Rio Cuarto, Rio Cuarto X5800BIA, Argentina; (B.A.C.); (M.J.L.)
| | - Matías Daniel Caverzan
- Departamento de Patología Animal, Facultad de Agronomía y Veterinaria, Universidad Nacional de Rio Cuarto, Rio Cuarto X5800BIA, Argentina;
- Instituto de Investigaciones en Tecnologías Energéticas y Materiales Avanzados (IITEMA), Universidad Nacional de Rio Cuarto (UNRC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Rio Cuarto X5800BIA, Argentina
| | - María Julia Lamberti
- Departamento de Biología Molecular, Facultad de Ciencias Exactas, Fisicoquímicas y Naturales, Universidad Nacional de Rio Cuarto, Rio Cuarto X5800BIA, Argentina; (B.A.C.); (M.J.L.)
- Instituto de Biotecnología Ambiental y Salud (INBIAS), Universidad Nacional de Rio Cuarto (UNRC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Rio Cuarto X5800BIA, Argentina
| | - Luis Exequiel Ibarra
- Departamento de Biología Molecular, Facultad de Ciencias Exactas, Fisicoquímicas y Naturales, Universidad Nacional de Rio Cuarto, Rio Cuarto X5800BIA, Argentina; (B.A.C.); (M.J.L.)
- Instituto de Biotecnología Ambiental y Salud (INBIAS), Universidad Nacional de Rio Cuarto (UNRC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Rio Cuarto X5800BIA, Argentina
| |
Collapse
|
11
|
Vitale R, Marzocco S, Popolo A. Role of Oxidative Stress and Inflammation in Doxorubicin-Induced Cardiotoxicity: A Brief Account. Int J Mol Sci 2024; 25:7477. [PMID: 39000584 PMCID: PMC11242665 DOI: 10.3390/ijms25137477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/01/2024] [Accepted: 07/06/2024] [Indexed: 07/16/2024] Open
Abstract
Cardiotoxicity is the main side effect of several chemotherapeutic drugs. Doxorubicin (Doxo) is one of the most used anthracyclines in the treatment of many tumors, but the development of acute and chronic cardiotoxicity limits its clinical usefulness. Different studies focused only on the effects of long-term Doxo administration, but recent data show that cardiomyocyte damage is an early event induced by Doxo after a single administration that can be followed by progressive functional decline, leading to overt heart failure. The knowledge of molecular mechanisms involved in the early stage of Doxo-induced cardiotoxicity is of paramount importance to treating and/or preventing it. This review aims to illustrate several mechanisms thought to underlie Doxo-induced cardiotoxicity, such as oxidative and nitrosative stress, inflammation, and mitochondrial dysfunction. Moreover, here we report data from both in vitro and in vivo studies indicating new therapeutic strategies to prevent Doxo-induced cardiotoxicity.
Collapse
Affiliation(s)
| | | | - Ada Popolo
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (R.V.); (S.M.)
| |
Collapse
|
12
|
Song JH, Kim MS, Lee SH, Hwang JT, Park SH, Park SW, Jeon SB, Lee RR, Lee J, Choi HK. Hydroethanolic extract of Cirsium setidens ameliorates doxorubicin-induced cardiotoxicity by AMPK-PGC-1α-SOD-mediated mitochondrial protection. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 129:155633. [PMID: 38640859 DOI: 10.1016/j.phymed.2024.155633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 04/02/2024] [Accepted: 04/11/2024] [Indexed: 04/21/2024]
Abstract
BACKGROUND Doxorubicin (DOX) is an effective anticancer agent. However, the clinical outcomes of DOX-based therapies are severely hampered by their significant cardiotoxicity. PURPOSE We investigated the beneficial effects of an ethanol extract of Cirsium setidens (CSE) on DOX-induced cardiomyotoxicity (DICT). METHODS UPLC-TQ/MS analysis was used to identify CSE metabolite profiles. H9c2 rat cardiomyocytes and MDA-MB-231 human breast cancer cells were used to evaluate the effects of CSE on DICT-induced cell death. To elucidate the mechanism underlying it, AMP-activated protein kinase (AMPK), peroxisome proliferator-activated receptor gamma co-activator l-alpha (PGC1-α), nuclear respiratory factor 1 (NRF1), NRF2, superoxide dismutase (SOD1), and SOD2 expression was detected using western blot analysis. The oxygen consumption rate (OCR), cellular ROS, and mitochondrial membrane potential were measured. Finally, we confirmed the cardioprotective effect of CSE against DICT in both C57BL/6 mice and human induced pluripotent stem cell-derived cardiomyocytes (hiPSCCMs) by observing various parameters, such as electrophysiological changes, cardiac fibrosis, and cardiac cell death. RESULTS Chlorogenic acid and nicotiflorin were the major compounds in CSE. Our data demonstrated that CSE blocked DOX-induced cell death of H9c2 cells without hindrance of its apoptotic effects on MDA-MB-231 cells. DOX-induced defects of OCR and mitochondrial membrane potential were recovered in a CSE through upregulation of the AMPK-PGC1-α-NRF1 signaling pathway. CSE accelerated NRF1 translocation to the nucleus, increased SOD activity, and consequently blocked apoptosis in H9c2 cells. In mice treated with 400 mg/kg CSE for 4 weeks, electrocardiogram data, creatine kinase and lactate dehydrogenase levels in the serum, and cardiac fibrosis, were improved. Moreover, various electrophysiological features indicative of cardiac function were significantly enhanced following the CSE treatment of hiPSCCMs. CONCLUSION Our findings demonstrate CSE that ameliorates DICT by protecting mitochondrial dysfunction via the AMP- PGC1α-NRF1 axis, underscoring the therapeutic potential of CSE and its underlying molecular pathways, setting the stage for future investigations into its clinical applications.
Collapse
Affiliation(s)
- Ji-Hye Song
- Korea Food Research Institute, Jeollabukdo 55365, South Korea
| | - Min-Sun Kim
- Korea Food Research Institute, Jeollabukdo 55365, South Korea
| | - Seung-Hyun Lee
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, South Korea; Institution of Genetic Science, Yonsei University College of Medicine, Seodaemun-gu, Seoul 03722, Republic of Korea; Division of Cardiology, Department of Medicine, Johns Hopkins University, MD, 21205, Baltimore, USA
| | - Jin-Taek Hwang
- Korea Food Research Institute, Jeollabukdo 55365, South Korea
| | - Soo-Hyun Park
- Korea Food Research Institute, Jeollabukdo 55365, South Korea
| | - Sahng Wook Park
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, South Korea; Institution of Genetic Science, Yonsei University College of Medicine, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Sae-Bom Jeon
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Ru-Ri Lee
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Jangho Lee
- Korea Food Research Institute, Jeollabukdo 55365, South Korea.
| | - Hyo-Kyoung Choi
- Korea Food Research Institute, Jeollabukdo 55365, South Korea.
| |
Collapse
|
13
|
Mohsenizadeh SA, Rajaeinejad M, Khoshfetrat M, Arefizadeh R, Mousavi SH, Mosaed R, Kazemi-Galougahi MH, Jalaeikhoo H, Faridfar A, Nikandish M, Alavi-Moghadam S, Arjmand B. Anthracycline-Induced Cardiomyopathy in Cancer Survivors: Management and Long-Term Implications. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024. [PMID: 38842787 DOI: 10.1007/5584_2024_804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
Recent advancements in personalized treatments, such as anthracycline chemotherapy, coupled with timely diagnoses, have contributed to a decrease in cancer-specific mortality rates and an improvement in cancer prognosis. Anthracyclines, a potent class of antibiotics, are extensively used as anticancer medications to treat a broad spectrum of tumors. Despite these advancements, a considerable number of cancer survivors face increased risks of treatment complications, particularly the cardiotoxic effects of chemotherapeutic drugs like anthracyclines. These effects can range from subclinical manifestations to severe consequences such as irreversible heart failure and death, highlighting the need for effective management of chemotherapy side effects for improved cancer care outcomes. Given the lack of specific treatments, early detection of subclinical cardiac events post-anthracycline therapy and the implementation of preventive strategies are vital. An interdisciplinary approach involving cardiovascular teams is crucial for the prevention and efficient management of anthracycline-induced cardiotoxicity. Various factors, such as age, gender, duration of treatment, and comorbidities, should be considered significant risk factors for developing chemotherapy-related cardiotoxicity. Tools such as electrocardiography, echocardiography, nuclear imaging, magnetic resonance imaging, histopathologic evaluations, and serum biomarkers should be appropriately used for the early detection of anthracycline-related cardiotoxicity. Furthermore, understanding the underlying biological mechanisms is key to developing preventive measures and personalized treatment strategies to mitigate anthracycline-induced cardiotoxicity. Exploring specific cardiotoxic mechanisms and identifying genetic variations can offer fresh perspectives on innovative, personalized treatments. This chapter aims to discuss cardiomyopathy following anthracycline therapy, with a focus on molecular mechanisms, preventive strategies, and emerging treatments.
Collapse
Affiliation(s)
| | - Mohsen Rajaeinejad
- AJA Cancer Epidemiology Research and Treatment Center (AJA-CERTC), AJA University of Medical Sciences, Tehran, Iran
| | - Mehran Khoshfetrat
- Department of Cardiology, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Reza Arefizadeh
- Department of Cardiology, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Seyed Hossein Mousavi
- Department of Cardiology, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Reza Mosaed
- Toxicology Research Center, AJA University of Medical Sciences, Tehran, Iran
- Student Research Committee, AJA University of Medical Sciences, Tehran, Iran
| | | | - Hasan Jalaeikhoo
- AJA Cancer Epidemiology Research and Treatment Center (AJA-CERTC), AJA University of Medical Sciences, Tehran, Iran
| | - Ali Faridfar
- AJA Cancer Epidemiology Research and Treatment Center (AJA-CERTC), AJA University of Medical Sciences, Tehran, Iran
| | - Mohsen Nikandish
- AJA Cancer Epidemiology Research and Treatment Center (AJA-CERTC), AJA University of Medical Sciences, Tehran, Iran
| | - Sepideh Alavi-Moghadam
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
14
|
Liu J, Hu X, Yu G, Wang Q, Gu L, Shen J, Zhao Q, Sun H, Wang S, Guo Z, Zhao Y, Ma H. Doxorubicin-based ENO1 targeted drug delivery strategy enhances therapeutic efficacy against colorectal cancer. Biochem Pharmacol 2024; 224:116220. [PMID: 38641307 DOI: 10.1016/j.bcp.2024.116220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/12/2024] [Accepted: 04/16/2024] [Indexed: 04/21/2024]
Abstract
Alpha-enolase (ENO1), a multifunctional protein with carcinogenic properties, has emerged as a promising cancer biomarker because of its differential expression in cancer and normal cells. On the basis of this characteristic, we designed a cell-targeting peptide that specifically targets ENO1 and connected it with the drug doxorubicin (DOX) by aldehyde-amine condensation. A surface plasmon resonance (SPR) assay showed that the affinity for ENO1 was stronger (KD = 2.5 µM) for the resulting cell-targeting drug, DOX-P, than for DOX. Moreover, DOX-P exhibited acid-responsive capabilities, enabling precise release at the tumor site under the guidance of the homing peptide and alleviating DOX-induced cardiotoxicity. An efficacy experiment confirmed that, the targeting ability of DOX-P toward ENO1 demonstrated superior antitumor activity against colorectal cancer than that of DOX, while reducing its toxicity to cardiomyocytes. Furthermore, in vivo metabolic distribution results indicated low accumulation of DOX-P in nontumor sites, further validating its targeting ability. These results showed that the ENO1-targeted DOX-P peptide has great potential for application in targeted drug-delivery systems for colorectal cancer therapy.
Collapse
Affiliation(s)
- Jun Liu
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing 100700, China
| | - Xiaoyu Hu
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing 100700, China
| | - Guanghao Yu
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing 100700, China
| | - Qingrong Wang
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing 100700, China
| | - Liwei Gu
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing 100700, China
| | - Jianying Shen
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing 100700, China
| | - Qinghe Zhao
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing 100700, China
| | - Hao Sun
- Nanjing Agricultural University, Nanjing 210009, China
| | - Shi Wang
- Nanjing Agricultural University, Nanjing 210009, China
| | - Zhongyuan Guo
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing 100700, China; College of Pharmacy, Henan University of Chinese Medicine, Henan Zhengzhou 450046, China
| | - Yu Zhao
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing 100700, China.
| | - Hai Ma
- China Academy of Chinese Medical Sciences, Institute of Chinese Materia Medica, Beijing 100700, China.
| |
Collapse
|
15
|
Golonko A, Olichwier AJ, Szklaruk A, Paszko A, Świsłocka R, Szczerbiński Ł, Lewandowski W. Apigenin's Modulation of Doxorubicin Efficacy in Breast Cancer. Molecules 2024; 29:2603. [PMID: 38893482 PMCID: PMC11174085 DOI: 10.3390/molecules29112603] [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: 04/05/2024] [Revised: 05/16/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Apigenin, a naturally derived flavonoid, is increasingly being acknowledged for its potential therapeutic applications, especially in oncology. This research explores apigenin's capacity to modulate cancer cell viability, emphasizing its roles beyond its minimal antioxidant activity attributed to its basic molecular structure devoid of hydroxyl groups. We investigated apigenin's effects on two breast cancer cell lines, estrogen-dependent MCF-7 and non-estrogen-dependent MDA-MB-231 cells. Our findings reveal that apigenin exerts a dose-dependent cytotoxic and anti-migratory impact on these cells. Interestingly, both apigenin and doxorubicin-a standard chemotherapeutic agent-induced lipid droplet accumulation in a dose-dependent manner in MDA-MB-231 cells. This phenomenon was absent in MCF-7 cells and not evident when doxorubicin and apigenin were used concurrently, suggesting distinct cellular responses to these treatments that imply that their synergistic effects might be mediated through mechanisms unrelated to lipid metabolism. A further chemoinformatics analysis indicated that apigenin and doxorubicin might interact primarily at the level of ATP-binding cassette (ABC) transporter proteins, with potential indirect influences from the AKT and MYC signaling pathways. These results highlight the importance of understanding the nuanced interactions between apigenin and conventional chemotherapeutic drugs, as they could lead to more effective strategies for cancer treatment. This study underscores apigenin's potential as a modulator of cancer cell dynamics through mechanisms independent of its direct antioxidant effects, thereby contributing to the development of flavonoid-based adjunct therapies in cancer management.
Collapse
Affiliation(s)
- Aleksandra Golonko
- Clinical Research Centre, Medical University of Bialystok, 15-089 Bialystok, Poland
| | - Adam Jan Olichwier
- Clinical Research Centre, Medical University of Bialystok, 15-089 Bialystok, Poland
| | - Agata Szklaruk
- Clinical Research Centre, Medical University of Bialystok, 15-089 Bialystok, Poland
| | - Adam Paszko
- Clinical Research Centre, Medical University of Bialystok, 15-089 Bialystok, Poland
| | - Renata Świsłocka
- Department of Chemistry, Biology and Biotechnology, Bialystok University of Technology, 15-351 Bialystok, Poland
| | - Łukasz Szczerbiński
- Clinical Research Centre, Medical University of Bialystok, 15-089 Bialystok, Poland
| | - Włodzimierz Lewandowski
- Department of Chemistry, Biology and Biotechnology, Bialystok University of Technology, 15-351 Bialystok, Poland
| |
Collapse
|
16
|
Li S, Niu W, Wang C, Zhao J, Zhang N, Yin Y, Jia M, Cui L. Exploring Anthracycline-Induced Cardiotoxicity from the Perspective of Protein Quality Control. Rev Cardiovasc Med 2024; 25:213. [PMID: 39076322 PMCID: PMC11270093 DOI: 10.31083/j.rcm2506213] [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: 11/03/2023] [Revised: 12/26/2023] [Accepted: 01/15/2024] [Indexed: 07/31/2024] Open
Abstract
Anthracyclines are effective anticancer drugs; however, their use is restricted because of their dose-dependent, time-dependent and irreversible myocardial toxicity. The mechanism of anthracycline cardiotoxicity has been widely studied but remains unclear. Protein quality control is crucial to the stability of the intracellular environment and, ultimately, to the heart because cardiomyocytes are terminally differentiated. Two evolutionarily conserved mechanisms, autophagy, and the ubiquitin-proteasome system, synergistically degrade misfolded proteins and remove defective organelles. Recent studies demonstrated the importance of these mechanisms. Further studies will reveal the detailed metabolic pathway and metabolic control of the protein quality control mechanism integrated into anthracycline-induced cardiotoxicity. This review provides theoretical support for clinicians in the application and management of anthracyclines.
Collapse
Affiliation(s)
- Shanshan Li
- Department of Laboratory Medicine, Peking University Third Hospital, 100191 Beijing, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Peking University Third Hospital, 100191 Beijing, China
- Department of Clinical Laboratory, Peking University People’s Hospital, 100041 Beijing, China
| | - Weihua Niu
- Department of Clinical Laboratory, Peking University People’s Hospital, 100041 Beijing, China
| | - Chunyan Wang
- Department of Clinical Laboratory, Peking University People’s Hospital, 100041 Beijing, China
| | - Jie Zhao
- Department of Clinical Laboratory, Peking University People’s Hospital, 100041 Beijing, China
| | - Na Zhang
- Department of Clinical Laboratory, Peking University People’s Hospital, 100041 Beijing, China
| | - Yue Yin
- Department of Clinical Laboratory, Peking University People’s Hospital, 100041 Beijing, China
| | - Mei Jia
- Department of Clinical Laboratory, Peking University People’s Hospital, 100041 Beijing, China
| | - Liyan Cui
- Department of Laboratory Medicine, Peking University Third Hospital, 100191 Beijing, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Peking University Third Hospital, 100191 Beijing, China
| |
Collapse
|
17
|
Tonon CR, Monte MG, Balin PS, Fujimori ASS, Ribeiro APD, Ferreira NF, Vieira NM, Cabral RP, Okoshi MP, Okoshi K, Zornoff LAM, Minicucci MF, Paiva SAR, Gomes MJ, Polegato BF. Liraglutide Pretreatment Does Not Improve Acute Doxorubicin-Induced Cardiotoxicity in Rats. Int J Mol Sci 2024; 25:5833. [PMID: 38892020 PMCID: PMC11172760 DOI: 10.3390/ijms25115833] [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: 03/15/2024] [Revised: 05/17/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
Doxorubicin is an effective drug for cancer treatment; however, cardiotoxicity limits its use. Cardiotoxicity pathophysiology is multifactorial. GLP-1 analogues have been shown to reduce oxidative stress and inflammation. In this study, we evaluated the effect of pretreatment with liraglutide on doxorubicin-induced acute cardiotoxicity. A total of 60 male Wistar rats were allocated into four groups: Control (C), Doxorubicin (D), Liraglutide (L), and Doxorubicin + Liraglutide (DL). L and DL received subcutaneous injection of liraglutide 0.6 mg/kg daily, while C and D received saline for 2 weeks. Afterwards, D and DL received a single intraperitoneal injection of doxorubicin 20 mg/kg; C and L received an injection of saline. Forty-eight hours after doxorubicin administration, the rats were subjected to echocardiogram, isolated heart functional study, and euthanasia. Liraglutide-treated rats ingested significantly less food and gained less body weight than animals that did not receive the drug. Rats lost weight after doxorubicin injection. At echocardiogram and isolated heart study, doxorubicin-treated rats had systolic and diastolic function impairment. Myocardial catalase activity was statistically higher in doxorubicin-treated rats. Myocardial protein expression of tumor necrosis factor alpha (TNF-α), phosphorylated nuclear factor-κB (p-NFκB), troponin T, and B-cell lymphoma 2 (Bcl-2) was significantly lower, and the total NFκB/p-NFκB ratio and TLR-4 higher in doxorubicin-treated rats. Myocardial expression of OPA-1, MFN-2, DRP-1, and topoisomerase 2β did not differ between groups (p > 0.05). In conclusion, doxorubicin-induced cardiotoxicity is accompanied by decreased Bcl-2 and phosphorylated NFκB and increased catalase activity and TLR-4 expression. Liraglutide failed to improve acute doxorubicin-induced cardiotoxicity in rats.
Collapse
Affiliation(s)
- Carolina R. Tonon
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (M.G.M.); (P.S.B.); (A.S.S.F.); (A.P.D.R.); (N.F.F.); (N.M.V.); (R.P.C.); (M.P.O.); (K.O.); (L.A.M.Z.); (M.F.M.); (S.A.R.P.); (B.F.P.)
| | - Marina G. Monte
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (M.G.M.); (P.S.B.); (A.S.S.F.); (A.P.D.R.); (N.F.F.); (N.M.V.); (R.P.C.); (M.P.O.); (K.O.); (L.A.M.Z.); (M.F.M.); (S.A.R.P.); (B.F.P.)
| | - Paola S. Balin
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (M.G.M.); (P.S.B.); (A.S.S.F.); (A.P.D.R.); (N.F.F.); (N.M.V.); (R.P.C.); (M.P.O.); (K.O.); (L.A.M.Z.); (M.F.M.); (S.A.R.P.); (B.F.P.)
| | - Anderson S. S. Fujimori
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (M.G.M.); (P.S.B.); (A.S.S.F.); (A.P.D.R.); (N.F.F.); (N.M.V.); (R.P.C.); (M.P.O.); (K.O.); (L.A.M.Z.); (M.F.M.); (S.A.R.P.); (B.F.P.)
| | - Ana Paula D. Ribeiro
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (M.G.M.); (P.S.B.); (A.S.S.F.); (A.P.D.R.); (N.F.F.); (N.M.V.); (R.P.C.); (M.P.O.); (K.O.); (L.A.M.Z.); (M.F.M.); (S.A.R.P.); (B.F.P.)
| | - Natália F. Ferreira
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (M.G.M.); (P.S.B.); (A.S.S.F.); (A.P.D.R.); (N.F.F.); (N.M.V.); (R.P.C.); (M.P.O.); (K.O.); (L.A.M.Z.); (M.F.M.); (S.A.R.P.); (B.F.P.)
| | - Nayane M. Vieira
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (M.G.M.); (P.S.B.); (A.S.S.F.); (A.P.D.R.); (N.F.F.); (N.M.V.); (R.P.C.); (M.P.O.); (K.O.); (L.A.M.Z.); (M.F.M.); (S.A.R.P.); (B.F.P.)
| | - Ronny P. Cabral
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (M.G.M.); (P.S.B.); (A.S.S.F.); (A.P.D.R.); (N.F.F.); (N.M.V.); (R.P.C.); (M.P.O.); (K.O.); (L.A.M.Z.); (M.F.M.); (S.A.R.P.); (B.F.P.)
| | - Marina P. Okoshi
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (M.G.M.); (P.S.B.); (A.S.S.F.); (A.P.D.R.); (N.F.F.); (N.M.V.); (R.P.C.); (M.P.O.); (K.O.); (L.A.M.Z.); (M.F.M.); (S.A.R.P.); (B.F.P.)
| | - Katashi Okoshi
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (M.G.M.); (P.S.B.); (A.S.S.F.); (A.P.D.R.); (N.F.F.); (N.M.V.); (R.P.C.); (M.P.O.); (K.O.); (L.A.M.Z.); (M.F.M.); (S.A.R.P.); (B.F.P.)
| | - Leonardo A. M. Zornoff
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (M.G.M.); (P.S.B.); (A.S.S.F.); (A.P.D.R.); (N.F.F.); (N.M.V.); (R.P.C.); (M.P.O.); (K.O.); (L.A.M.Z.); (M.F.M.); (S.A.R.P.); (B.F.P.)
| | - Marcos F. Minicucci
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (M.G.M.); (P.S.B.); (A.S.S.F.); (A.P.D.R.); (N.F.F.); (N.M.V.); (R.P.C.); (M.P.O.); (K.O.); (L.A.M.Z.); (M.F.M.); (S.A.R.P.); (B.F.P.)
| | - Sergio A. R. Paiva
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (M.G.M.); (P.S.B.); (A.S.S.F.); (A.P.D.R.); (N.F.F.); (N.M.V.); (R.P.C.); (M.P.O.); (K.O.); (L.A.M.Z.); (M.F.M.); (S.A.R.P.); (B.F.P.)
| | - Mariana J. Gomes
- Department of Kinesiology and Sport Management, Texas A&M University, College Station, TX 77843, USA;
| | - Bertha F. Polegato
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (M.G.M.); (P.S.B.); (A.S.S.F.); (A.P.D.R.); (N.F.F.); (N.M.V.); (R.P.C.); (M.P.O.); (K.O.); (L.A.M.Z.); (M.F.M.); (S.A.R.P.); (B.F.P.)
| |
Collapse
|
18
|
Nagy A, Börzsei D, Hoffmann A, Török S, Veszelka M, Almási N, Varga C, Szabó R. A Comprehensive Overview on Chemotherapy-Induced Cardiotoxicity: Insights into the Underlying Inflammatory and Oxidative Mechanisms. Cardiovasc Drugs Ther 2024:10.1007/s10557-024-07574-0. [PMID: 38492161 DOI: 10.1007/s10557-024-07574-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/05/2024] [Indexed: 03/18/2024]
Abstract
While oncotherapy has made rapid progress in recent years, side effects of anti-cancer drugs and treatments have also come to the fore. These side effects include cardiotoxicity, which can cause irreversible cardiac damages with long-term morbidity and mortality. Despite the continuous in-depth research on anti-cancer drugs, an improved knowledge of the underlying mechanisms of cardiotoxicity are necessary for early detection and management of cardiac risk. Although most reviews focus on the cardiotoxic effect of a specific individual chemotherapeutic agent, the aim of our review is to provide comprehensive insight into various agents that induced cardiotoxicity and their underlying mechanisms. Characterization of these mechanisms are underpinned by research on animal models and clinical studies. In order to gain insight into these complex mechanisms, we emphasize the role of inflammatory processes and oxidative stress on chemotherapy-induced cardiac changes. A better understanding and identification of the interplay between chemotherapy and inflammatory/oxidative processes hold some promise to prevent or at least mitigate cardiotoxicity-associated morbidity and mortality among cancer survivors.
Collapse
Affiliation(s)
- András Nagy
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary
| | - Denise Börzsei
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary
| | - Alexandra Hoffmann
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary
| | - Szilvia Török
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary
| | - Médea Veszelka
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary
| | - Nikoletta Almási
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary
| | - Csaba Varga
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary
| | - Renáta Szabó
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép Fasor 52, 6726, Szeged, Hungary.
| |
Collapse
|
19
|
Wongsawatkul O, Buachan P, Jaisin Y, Busarakumtragul P, Chainakul S, Watanapokasin R, Prachayasittikul V, Prachayasittikul S, Ruchirawat S, Prachayasittikul V. Effects of barakol from Cassia siamea on neuroblastoma SH-SY5Y cell line: A potential combined therapy with doxorubicin. Heliyon 2024; 10:e24694. [PMID: 38318050 PMCID: PMC10839565 DOI: 10.1016/j.heliyon.2024.e24694] [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: 05/12/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 02/07/2024] Open
Abstract
Management of neuroblastoma is challenging because of poor response to drugs, chemotherapy resistance, high relapse, and treatment failures. Doxorubicin is a potent anticancer drug commonly used for neuroblastoma treatment. However, doxorubicin induces considerable toxicities, particularly those caused by oxidative-related damage. To minimize drug-induced adverse effects, the combined use of anticancer drugs with natural-derived compounds possessing antioxidant properties has become an interesting treatment strategy. Barakol is a major compound found in Cassia siamea, an edible plant with antioxidant and anticancer properties. Therefore, barakol could potentially be used in combination with doxorubicin to synergize the anticancer effect, while minimizing the oxidative-related toxicities. Herein, the potential of barakol (0.0043-43.0 μM) to synergize the anticancer effect of low-dose doxorubicin (0.5 and 1.0 μM) was investigated. Results indicated that barakol could enhance the cytotoxic effect of low-dose doxorubicin by affecting the cell viability of the treated cells. Furthermore, the co-treatment with barakol and low-dose doxorubicin decreased the levels of intracellular ROS when compared with the control. Moreover, the antimetastatic effect of the barakol itself was studied through its ability to inhibit metalloproteinase-3 (MMP-3) activity and prevent cell migration. Results revealed that the barakol inhibited MMP-3 activity and prevented cell migration in time- and dose-dependent manners. Additionally, barakol was a non-cytotoxic agent against the normal tested cell line (MRC-5), which suggested its selectivity and safety. Taken together, barakol could be a promising compound to be further developed for combination treatment with low-dose doxorubicin to improve therapeutic effectiveness but decrease drug-induced toxicities. The inhibitory effects of barakol on MMP-3 activity and cancer cell migration also supported its potential to be developed as an antimetastatic agent.
Collapse
Affiliation(s)
- Orapin Wongsawatkul
- Department of Pharmacology, Faculty of Medicine, Srinakharinwirot University, Bangkok, 10110, Thailand
| | - Paiwan Buachan
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand
| | - Yamaratee Jaisin
- Department of Pharmacology, Faculty of Medicine, Srinakharinwirot University, Bangkok, 10110, Thailand
| | - Panaree Busarakumtragul
- Department of Physiology, Faculty of Medicine, Srinakharinwirot University, Bangkok, 10110, Thailand
| | - Sunan Chainakul
- Department of Chemistry, Faculty of Science, Srinakharinwirot University, Bangkok, 10110, Thailand
| | - Ramida Watanapokasin
- Department of Biochemistry, Faculty of Medicine, Srinakharinwirot University, Bangkok, 10110, Thailand
| | - Veda Prachayasittikul
- Center for Research Innovation and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand
| | - Supaluk Prachayasittikul
- Center for Research Innovation and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand
| | - Somsak Ruchirawat
- Laboratory of Medicinal Chemistry, Chulabhorn Research Institute, Bangkok, 10210, Thailand
- Program in Chemical Sciences, Chulabhorn Graduate Institute, Bangkok, 10210, Thailand
- Center of Excellence on Environmental Health and Toxicology (EHT), Commission on Higher Education, Ministry of Education, Bangkok, 10400, Thailand
| | - Virapong Prachayasittikul
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand
| |
Collapse
|
20
|
Oraiopoulou ME, Tzamali E, Psycharakis SE, Tzedakis G, Makatounakis T, Manolitsi K, Drakos E, Vakis AF, Zacharakis G, Papamatheakis J, Sakkalis V. The Temozolomide-Doxorubicin paradox in Glioblastoma in vitro-in silico preclinical drug-screening. Sci Rep 2024; 14:3759. [PMID: 38355655 PMCID: PMC10866941 DOI: 10.1038/s41598-024-53684-y] [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: 07/25/2023] [Accepted: 02/03/2024] [Indexed: 02/16/2024] Open
Abstract
Adjuvant Temozolomide is considered the front-line Glioblastoma chemotherapeutic treatment; yet not all patients respond. Latest trends in clinical trials usually refer to Doxorubicin; yet it can lead to severe side-effects if administered in high doses. While Glioblastoma prognosis remains poor, little is known about the combination of the two chemotherapeutics. Patient-derived spheroids were generated and treated with a range of Temozolomide/Doxorubicin concentrations either as monotherapy or in combination. Optical microscopy was used to monitor the growth pattern and cell death. Based on the monotherapy experiments, we developed a probabilistic mathematical framework in order to describe the drug-induced effect at the single-cell level and simulate drug doses in combination assuming probabilistic independence. Doxorubicin was found to be effective in doses even four orders of magnitude less than Temozolomide in monotherapy. The combination therapy doses tested in vitro were able to lead to irreversible growth inhibition at doses where monotherapy resulted in relapse. In our simulations, we assumed both drugs are anti-mitotic; Temozolomide has a growth-arrest effect, while Doxorubicin is able to cumulatively cause necrosis. Interestingly, under no mechanistic synergy assumption, the in silico predictions underestimate the in vitro results. In silico models allow the exploration of a variety of potential underlying hypotheses. The simulated-biological discrepancy at certain doses indicates a supra-additive response when both drugs are combined. Our results suggest a Temozolomide-Doxorubicin dual chemotherapeutic scheme to both disable proliferation and increase cytotoxicity against Glioblastoma.
Collapse
Affiliation(s)
- Mariam-Eleni Oraiopoulou
- Institute of Computer Science (ICS), Foundation for Research and Technology-Hellas (FORTH), Heraklion, Greece
- Cancer Research UK - Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Eleftheria Tzamali
- Institute of Computer Science (ICS), Foundation for Research and Technology-Hellas (FORTH), Heraklion, Greece
| | - Stylianos E Psycharakis
- Institute of Electronic Structure and Laser (IESL), Foundation for Research and Technology Hellas (FORTH), Heraklion, Greece
- School of Medicine, University of Crete, Heraklion, Greece
| | - Georgios Tzedakis
- Institute of Computer Science (ICS), Foundation for Research and Technology-Hellas (FORTH), Heraklion, Greece
| | - Takis Makatounakis
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology Hellas (FORTH), Heraklion, Greece
| | - Katina Manolitsi
- University General Hospital of Heraklion (PAGNI), Heraklion, Greece
| | - Elias Drakos
- School of Medicine, University of Crete, Heraklion, Greece
- University General Hospital of Heraklion (PAGNI), Heraklion, Greece
| | - Antonis F Vakis
- School of Medicine, University of Crete, Heraklion, Greece
- University General Hospital of Heraklion (PAGNI), Heraklion, Greece
| | - Giannis Zacharakis
- Institute of Electronic Structure and Laser (IESL), Foundation for Research and Technology Hellas (FORTH), Heraklion, Greece
| | - Joseph Papamatheakis
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology Hellas (FORTH), Heraklion, Greece
- Department of Biology, University of Crete, Heraklion, Greece
| | - Vangelis Sakkalis
- Institute of Computer Science (ICS), Foundation for Research and Technology-Hellas (FORTH), Heraklion, Greece.
| |
Collapse
|
21
|
Fan Z, Tong Y, Yang Z, Wang S, Huang T, Yang D, Ni Q, Zhang M, Li D, Yang M, Fan X. Inhibitor PF-04691502 works as a senolytic to regulate cellular senescence. Exp Gerontol 2024; 186:112359. [PMID: 38184267 DOI: 10.1016/j.exger.2024.112359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/24/2023] [Accepted: 01/02/2024] [Indexed: 01/08/2024]
Abstract
Aging is a gradual process of natural change that occurs after reaching sexual maturity. It is also a known risk factor for many chronic diseases. Recent research has shown that senolytics can extend the lifespans and health spans of model organisms, and they have also been demonstrated effective in treating age-related diseases. In this study, we conducted a high-throughput screening of 156 drugs that targeted the PI3K/AKT/mTOR pathway to identify potential senolytic medications. Among these drugs, PF-04691502 was selected for further investigation to understand its molecular mechanism of action. Our findings indicate that PF-04691502, a dual inhibitor of PI3K/AKT and mTOR, specifically eliminates senescent cells. It reduces the expression levels of key markers of cellular senescence, such as SA-β-Gal, senescence-associated secretory phenotypes (SASPs) and p16INK4a. Additionally, PF-04691502 inhibits the phosphorylation of S6K and AKT, leading to the apoptosis of senescent cells. These results suggest that PF-04691502 holds promise as a new senolytic drug. This paper provides important insights into the potential application of PF-04691502 in the study of cell senescence.
Collapse
Affiliation(s)
- Ziqiang Fan
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Yingdong Tong
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Ziyue Yang
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Shuai Wang
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Tiantian Huang
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China
| | - Deying Yang
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Qingyong Ni
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Mingwang Zhang
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Diyan Li
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China; Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, 610106 Chengdu, China
| | - Mingyao Yang
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China.
| | - Xiaolan Fan
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, Sichuan 611130, PR China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China.
| |
Collapse
|
22
|
Yun C, Kim SH, Kwon D, Byun MR, Chung KW, Lee J, Jung YS. Doxorubicin Attenuates Free Fatty Acid-Induced Lipid Accumulation via Stimulation of p53 in HepG2 Cells. Biomol Ther (Seoul) 2024; 32:94-103. [PMID: 38148555 PMCID: PMC10762281 DOI: 10.4062/biomolther.2023.200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 12/28/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is characterized by excessive accumulation of fat in the liver, and there is a global increase in its incidence owing to changes in lifestyle and diet. Recent findings suggest that p53 is involved in the development of non-alcoholic fatty liver disease; however, the association between p53 expression and the disease remains unclear. Doxorubicin, an anticancer agent, increases the expression of p53. Therefore, this study aimed to investigate the role of doxorubicin-induced p53 upregulation in free fatty acid (FFA)-induced intracellular lipid accumulation. HepG2 cells were pretreated with 0.5 μg/mL of doxorubicin for 12 h, followed by treatment with FFA (0.5 mM) for 24 h to induce steatosis. Doxorubicin pretreatment upregulated p53 expression and downregulated the expression of endoplasmic reticulum stress- and lipid synthesis-associated genes in the FFA -treated HepG2 cells. Additionally, doxorubicin treatment upregulated the expression of AMP-activated protein kinase, a key modulator of lipid metabolism. Notably, siRNA-targeted p53 knockdown reversed the effects of doxorubicin in HepG2 cells. Moreover, doxorubicin treatment suppressed FFA -induced lipid accumulation in HepG2 spheroids. Conclusively, these results suggest that doxorubicin possesses potential application for the regulation of lipid metabolism by enhance the expression of p53 an in vitro NAFLD model.
Collapse
Affiliation(s)
- Chawon Yun
- Department of Pharmacy, College of Pharmacy, Research Institute for Drug Development, Pusan National University, Busan 46241, Republic of Korea
| | - Sou Hyun Kim
- Department of Pharmacy, College of Pharmacy, Research Institute for Drug Development, Pusan National University, Busan 46241, Republic of Korea
| | - Doyoung Kwon
- College of Pharmacy, Jeju Research Institute of Pharmaceutical Sciences, Jeju National University, Jeju 63243, Republic of Korea
| | - Mi Ran Byun
- College of Pharmacy, Daegu Catholic University, Gyeongsan 38430, Republic of Korea
| | - Ki Wung Chung
- Department of Pharmacy, College of Pharmacy, Research Institute for Drug Development, Pusan National University, Busan 46241, Republic of Korea
| | - Jaewon Lee
- Department of Pharmacy, College of Pharmacy, Research Institute for Drug Development, Pusan National University, Busan 46241, Republic of Korea
| | - Young-Suk Jung
- Department of Pharmacy, College of Pharmacy, Research Institute for Drug Development, Pusan National University, Busan 46241, Republic of Korea
| |
Collapse
|
23
|
Kim MS, Choi HK, Park SH, Lee JI, Lee J. Poncirus trifoliata Aqueous Extract Protects Cardiomyocytes against Doxorubicin-Induced Toxicity through Upregulation of NAD(P)H Dehydrogenase Quinone Acceptor Oxidoreductase 1. Molecules 2023; 28:8090. [PMID: 38138580 PMCID: PMC10745630 DOI: 10.3390/molecules28248090] [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: 10/29/2023] [Revised: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
Doxorubicin (DOX), an anthracycline-based chemotherapeutic agent, is widely used to treat various types of cancer; however, prolonged treatment induces cardiomyotoxicity. Although studies have been performed to overcome DOX-induced cardiotoxicity (DICT), no effective method is currently available. This study investigated the effects and potential mechanisms of Poncirus trifoliata aqueous extract (PTA) in DICT. Changes in cell survival were assessed in H9c2 rat cardiomyocytes and MDA-MB-231 human breast cancer cells. The C57BL/6 mice were treated with DOX to induce DICT in vivo, and alterations in electrophysiological characteristics, serum biomarkers, and histological features were examined. The PTA treatment inhibited DOX-induced decrease in H9c2 cell viability but did not affect the MDA-MB-231 cell viability. Additionally, the PTA restored the abnormal heart rate, R-R interval, QT interval, and ST segment and inhibited the decrease in serum cardiac and hepatic toxicity indicators in the DICT model. Moreover, the PTA administration protected against myocardial fibrosis and apoptosis in the heart tissue of mice with DICT. PTA treatment restored DOX-induced decrease in the expression of NAD(P)H dehydrogenase quinone acceptor oxidoreductase 1 in a PTA concentration-dependent manner. In conclusion, the PTA inhibitory effect on DICT is attributable to its antioxidant properties, suggesting the potential of PTA as a phytotherapeutic agent for DICT.
Collapse
Affiliation(s)
| | | | | | | | - Jangho Lee
- Korea Food Research Institute, Wanju 55365, Republic of Korea; (M.-S.K.); (H.-K.C.); (S.-H.P.); (J.-I.L.)
| |
Collapse
|
24
|
Soliman AG, Mahmoud B, Eldin ZE, El-Shahawy AAG, Abdel-Gabbar M. Optimized synthesis characterization and protective activity of quercetin and quercetin–chitosan nanoformula against cardiotoxicity that was induced in male Wister rats via anticancer agent: doxorubicin. Cancer Nanotechnol 2023. [DOI: 10.1186/s12645-023-00158-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023] Open
Abstract
AbstractThe study’s goal was to look into the protective properties of quercetin (QU) in natural form and QU nanoparticles-loaded chitosan nanoparticles (QU-CHSNPs) against cardiotoxicity. The ionotropic gelation approach was adopted to form QU-CHSNPs. The characterizations were performed using advanced techniques. In vitro, the release profile of QU was studied. Cardiotoxicity was induced by doxorubicin (DOX) and protected via concurrent administration of QU and QU-CHSNPs. The heart's preventive effects of QU and QU-CHSNPs were manifested by a decrease in elevated serum activities of cardiac enzymes, as well as an improvement in the heart's antioxidant defence system and histological changes. The findings substantiated QU-CHSNPs' structure with an entrapment efficiency of 92.56%. The mean of the zeta size distribution was 150 nm, the real average particle size was 50 nm, and the zeta potential value was − 27.9 mV, exhibiting low physical stability. The percent of the free QU-cumulative release was about 70% after 12 h, and QU-CHSNPs showed a 49% continued release with a pattern of sustained release, reaching 98% after 48 h. And as such, QU and QU-CHSNPs restrained the induced cardiotoxicity of DOX in male Wistar rats, with the QU-CHSNPs being more efficient.
Collapse
|
25
|
Zhang Y, Zhang D, Meng T, Tian P, Chen J, Liu A, Zheng Y, Su G. SGK1 is involved in doxorubicin-induced chronic cardiotoxicity and dysfunction through activation of the NFκB pathway. Int Immunopharmacol 2023; 125:111151. [PMID: 37948859 DOI: 10.1016/j.intimp.2023.111151] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/25/2023] [Accepted: 10/29/2023] [Indexed: 11/12/2023]
Abstract
Breast cancer is the predominant cancer among women worldwide, and chemotherapeutic agents, such as doxorubicin (DOX), have the potential to significantly prolong survival, albeit at the cost of inducing severe cardiovascular toxicity. Inflammation has emerged as a crucial biological process contributing to the remodeling of cardiovascular toxicity. The role of serum glucocorticoid kinase 1 (SGK1) in various inflammatory diseases has been extensively investigated. Here, we studied the molecular mechanisms underlying the function of SGK1 in DOX-induced cardiotoxicity in HL-1 cardiomyocyte cell lines and in a tumor-bearing mouse model. SGK1 was upregulated in the DOX-induced cardiotoxicity model, accompanied by increased levels of inflammatory factors. Furthermore, inhibition of SGK1 suppresses the phosphorylation of nuclear factor-kappa B (NFκB) in cardiomyocytes, which inhibits the production of inflammatory factors and apoptosis of cardiomyocytes, and has cardioprotective effects. Simultaneously, small interfering RNA targeting SGK1 inhibited the proliferation of breast cancer cells. Conversely, overexpression of SGK1 increases the phosphorylation of NFκB and aggravates myocardial injury. In conclusion, our study demonstrates that SGK1 promotes DOX-induced cardiac inflammation and apoptosis by promoting NFκB activity. Our results indicate that inhibiting SGK1 might be an effective treatment strategy that can provide both tumor-killing and cardioprotective functions. Further in vivo research is needed to fully elucidate the effects and mechanisms of combination therapy with SGK1 inhibitors and DOX in breast cancer treatment.
Collapse
Affiliation(s)
- Yu Zhang
- Jinan Central Hospital, Shandong University, Jinan, Shandong, People's Republic of China
| | - Dan Zhang
- Jinan Central Hospital, Jinan, Shandong, People's Republic of China
| | - Tingting Meng
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China
| | - Peng Tian
- Jinan Central Hospital, Shandong University, Jinan, Shandong, People's Republic of China
| | - Jianlin Chen
- Research Center of Translational Medicine, Jinan Central Hospital, Weifang Medical University, Weifang, Shandong, People's Republic of China
| | - Anbang Liu
- Jinan Central Hospital, Jinan, Shandong, People's Republic of China
| | - Yan Zheng
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China.
| | - Guohai Su
- Jinan Central Hospital, Shandong University, Jinan, Shandong, People's Republic of China; Jinan Central Hospital, Jinan, Shandong, People's Republic of China; Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China.
| |
Collapse
|
26
|
Casagrande N, Borghese C, Avanzo M, Aldinucci D. In Doxorubicin-Adapted Hodgkin Lymphoma Cells, Acquiring Multidrug Resistance and Improved Immunosuppressive Abilities, Doxorubicin Activity Was Enhanced by Chloroquine and GW4869. Cells 2023; 12:2732. [PMID: 38067159 PMCID: PMC10706762 DOI: 10.3390/cells12232732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 11/27/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
Classical Hodgkin lymphoma (cHL) is a highly curable disease (70-80%), even though long-term toxicities, drug resistance, and predicting clinical responses to therapy are major challenges in cHL treatment. To solve these problems, we characterized two cHL cell lines with acquired resistance to doxorubicin, KM-H2dx and HDLM-2dx (HRSdx), generated from KM-H2 and HDLM-2 cells, respectively. HRSdx cells developed cross-resistance to vinblastine, bendamustin, cisplatin, dacarbazine, gemcitabine, brentuximab vedotin (BV), and γ-radiation. Both HDLM-2 and HDLM-2dx cells had intrinsic resistance to BV but not to the drug MMAE. HDLM-2dx acquired cross-resistance to caelyx. HRSdx cells had in common decreased CD71, CD80, CD54, cyt-ROS, HLA-DR, DDR1, and CD44; increased Bcl-2, CD58, COX2, CD26, CCR5, and invasive capability; increased CCL5, TARC, PGE2, and TGF-β; and the capability of hijacking monocytes. In HRSdx cells less sensitive to DNA damage and oxidative stress, the efflux drug transporters MDR1 and MRP1 were not up-regulated, and doxorubicin accumulated in the cytoplasm rather than in the nucleus. Both the autophagy inhibitor chloroquine and extracellular vesicle (EV) release inhibitor GW4869 enhanced doxorubicin activity and counteracted doxorubicin resistance. In conclusion, this study identifies common modulated antigens in HRSdx cells, the associated cross-resistance patterns, and new potential therapeutic options to enhance doxorubicin activity and overcome resistance.
Collapse
Affiliation(s)
- Naike Casagrande
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (C.B.); (D.A.)
| | - Cinzia Borghese
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (C.B.); (D.A.)
| | - Michele Avanzo
- Department of Medical Physics, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy;
| | - Donatella Aldinucci
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (C.B.); (D.A.)
| |
Collapse
|
27
|
Alhowail AH, Eggert M, Bloemer J, Pinky PD, Woodie L, Bhattacharya S, Bhattacharya D, Buabeid MA, Smith B, Dhanasekaran M, Piazza G, Reed MN, Escobar M, Arnold RD, Suppiramaniam V. Phenyl-2-aminoethyl selenide ameliorates hippocampal long-term potentiation and cognitive deficits following doxorubicin treatment. PLoS One 2023; 18:e0294280. [PMID: 37948406 PMCID: PMC10637675 DOI: 10.1371/journal.pone.0294280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 10/30/2023] [Indexed: 11/12/2023] Open
Abstract
Chemotherapy-induced memory loss ("chemobrain") can occur following treatment with the widely used chemotherapeutic agent doxorubicin (DOX). However, the mechanisms through which DOX induces cognitive dysfunction are not clear, and there are no commercially available therapies for its treatment or prevention. Therefore, the aim of this study was to determine the therapeutic potential of phenyl-2-aminoethyl selenide (PAESe), an antioxidant drug previously demonstrated to reduce cardiotoxicity associated with DOX treatment, against DOX-induced chemobrain. Four groups of male athymic NCr nude (nu/nu) mice received five weekly tail-vein injections of saline (Control group), 5 mg/kg of DOX (DOX group), 10 mg/kg PAESe (PAESe group), or 5 mg/kg DOX and 10 mg/kg PAESe (DOX+PAESe group). Spatial memory was evaluated using Y-maze and novel object location tasks, while synaptic plasticity was assessed through the measurement of field excitatory postsynaptic potentials from the Schaffer collateral circuit. Western blot analyses were performed to assess hippocampal protein and phosphorylation levels. In this model, DOX impaired synaptic plasticity and memory, and increased phosphorylation of protein kinase B (Akt) and extracellular-regulated kinase (ERK). Co-administration of PAESe reduced Akt and ERK phosphorylation and ameliorated the synaptic and memory deficits associated with DOX treatment.
Collapse
Affiliation(s)
- Ahmad H. Alhowail
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, United States of America
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah, Kingdom of Saudi Arabia
| | - Matthew Eggert
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, United States of America
| | - Jenna Bloemer
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, United States of America
| | - Priyanka D. Pinky
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, United States of America
| | - Lauren Woodie
- Department of Nutrition, Dietetics and Hospitality Management, College of Human Sciences, Auburn University, Auburn, AL, United States of America
| | - Subhrajit Bhattacharya
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, United States of America
| | - Dwipayan Bhattacharya
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, United States of America
| | - Manal A. Buabeid
- College of Pharmacy and Health Sciences, Ajman University, Ajman, UAE
| | - Bruce Smith
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States of America
| | - Muralikrishnan Dhanasekaran
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, United States of America
- Center for Neuroscience Initiative, Auburn University, Auburn, AL, United States of America
| | - Gary Piazza
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, United States of America
| | - Miranda N. Reed
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, United States of America
- Center for Neuroscience Initiative, Auburn University, Auburn, AL, United States of America
| | - Martha Escobar
- Department of Psychology, Oakland University, Rochester, MI, United States of America
| | - Robert D. Arnold
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, United States of America
- Center for Neuroscience Initiative, Auburn University, Auburn, AL, United States of America
| | - Vishnu Suppiramaniam
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, United States of America
- Center for Neuroscience Initiative, Auburn University, Auburn, AL, United States of America
- Department of Molecular and Cellular Biology, College of Science and Mathematics, Kennesaw State University, Kennesaw, Georgia
| |
Collapse
|
28
|
Deszcz I. Stem Cell-Based Therapy and Cell-Free Therapy as an Alternative Approach for Cardiac Regeneration. Stem Cells Int 2023; 2023:2729377. [PMID: 37954462 PMCID: PMC10635745 DOI: 10.1155/2023/2729377] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 06/21/2023] [Accepted: 10/10/2023] [Indexed: 11/14/2023] Open
Abstract
The World Health Organization reports that cardiovascular diseases (CVDs) represent 32% of all global deaths. The ineffectiveness of conventional therapies in CVDs encourages the development of novel, minimally invasive therapeutic strategies for the healing and regeneration of damaged tissue. The self-renewal capacity, multilineage differentiation, lack of immunogenicity, and immunosuppressive properties of mesenchymal stem cells (MSCs) make them a promising option for CVDs. However, growing evidence suggests that myocardial regeneration occurs through paracrine factors and extracellular vesicle (EV) secretion, rather than through differentiation into cardiomyocytes. Research shows that stem cells secrete or surface-shed into their culture media various cytokines, chemokines, growth factors, anti-inflammatory factors, and EVs, which constitute an MSC-conditioned medium (MSC-CM) or the secretome. The use of MSC-CM enhances cardiac repair through resident heart cell differentiation, proliferation, scar mass reduction, a decrease in infarct wall thickness, and cardiac function improvement comparable to MSCs without their side effects. This review highlights the limitations and benefits of therapies based on stem cells and their secretome as an innovative treatment of CVDs.
Collapse
Affiliation(s)
- Iwona Deszcz
- Department of Immunopathology and Molecular Biology, Wroclaw Medical University, Borowska 211, 50-556, Wroclaw, Poland
| |
Collapse
|
29
|
Lin L, Lyu J, Zhou X. Editorial for "Serial Cardiovascular MRI for Quantification of the Dynamics of Anthracycline-Induced Subclinical Myocardial Injury". J Magn Reson Imaging 2023; 58:1542-1543. [PMID: 36905196 DOI: 10.1002/jmri.28666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 02/16/2023] [Accepted: 02/16/2023] [Indexed: 03/12/2023] Open
Affiliation(s)
- Ling Lin
- Department of Radiology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Cardio Vascular Imaging Group, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Junhong Lyu
- Department of Radiology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Xuhui Zhou
- Department of Radiology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
30
|
Emori C, Boucher Z, Bolcun-Filas E. CHEK2 signaling is the key regulator of oocyte survival after chemotherapy. SCIENCE ADVANCES 2023; 9:eadg0898. [PMID: 37862420 PMCID: PMC10588956 DOI: 10.1126/sciadv.adg0898] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 09/06/2023] [Indexed: 10/22/2023]
Abstract
Cancer treatments can damage the ovarian follicle reserve, leading to primary ovarian insufficiency and infertility among survivors. Checkpoint kinase 2 (CHEK2) deficiency prevents elimination of oocytes in primordial follicles in female mice exposed to radiation and preserves their ovarian function and fertility. Here, we demonstrate that CHEK2 also coordinates the elimination of oocytes after exposure to standard-of-care chemotherapy drugs. CHEK2 activates two downstream targets-TAp63 and p53-which direct oocyte elimination. CHEK2 knockout or pharmacological inhibition preserved ovarian follicle reserve after radiation and chemotherapy. However, the lack of specificity for CHEK2 among available inhibitors limits their potential for clinical development. These findings demonstrate that CHEK2 is a master regulator of the ovarian cellular response to damage caused by radiation and chemotherapy and warrant the development of selective inhibitors specific to CHEK2 as a potential avenue for ovario-protective treatments.
Collapse
Affiliation(s)
- Chihiro Emori
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 5650871, Japan
| | - Zachary Boucher
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | | |
Collapse
|
31
|
Sun Y, Guo D, Yue S, Zhou M, Wang D, Chen F, Wang L. Afzelin protects against doxorubicin-induced cardiotoxicity by promoting the AMPKα/SIRT1 signaling pathway. Toxicol Appl Pharmacol 2023; 477:116687. [PMID: 37703929 DOI: 10.1016/j.taap.2023.116687] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 09/15/2023]
Abstract
BACKGROUND Doxorubicin (DOX), a chemotherapeutic drug, could relieve the progressions of various diseases. However, its clinical application is limited due to its cardiotoxicity. This study aimed to investigate the effects of afzelin (a flavonol glycoside found in Houttuynia cordata) on the cardiotoxicity induced by DOX. METHODS In ex-vivo, H9C2 cells were incubated with 20, 40, or 80 μM afzelin for 12 h, followed by the treatment with 1 μM DOX for 12 h. In vivo, C57BL/6 J mice were intraperitoneally injected with 4 mg/kg/day DOX on days 1, 7, and 14. Meanwhile, starting from day 1, mice were intragastrically administrated with 5 mg/kg/day or 10 mg/kg/day afzelin for 20 days. The cardiac function of mice was evaluated by detecting hemodynamic parameters using the M-mode echocardiography. RESULTS DOX decreased the cell survival rate, and elevated apoptotic rate, as well as induced the oxidative stress and mitochondrial dysfunction in H9C2 cells. All these changes were alleviated by afzelin treatment in a concentration-dependent manner. The results were further proven by the mitigation of cardiac injury in vivo, as evidenced by the elevation of fractional shortening, heart weight/tibia length, and the rate of the increase/decrease of left ventricular pressure in mice subjected to DOX-induced cardiotoxicity. Furthermore, afzelin upregulated the expression of p-AMP-activated protein kinase alpha (AMPKα) and sirtuin1 (SIRT1). Dorsomorphin (an AMPKα inhibitor) abrogated the anti-cardiotoxicity effects of afzelin in H9C2 cells induced by DOX. CONCLUSION Afzelin protected against DOX-induced cardiotoxicity by promoting the AMPKα/SIRT1 signaling pathway.
Collapse
Affiliation(s)
- Yixin Sun
- Department of Ultrasound, Harbin Medical University Cancer Hospital, 150 Haping Road, Nangang District, Harbin 150081, Heilongjiang, China
| | - Danyang Guo
- Department of Ultrasound, the Sixth Affiliated Hospital of Harbin Medical University, 57 Youyi Road, Daoli District, Harbin 150076, Heilongjiang, China
| | - Saiding Yue
- Department of Nephrology, Harbin Jing-En Nephrology Hospital, 11 Xiangbin Road, Xiangfang District, Harbin 150036, Heilongjiang, China
| | - Mingyan Zhou
- Department of Ultrasound, Harbin Medical University Cancer Hospital, 150 Haping Road, Nangang District, Harbin 150081, Heilongjiang, China
| | - Dongxu Wang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, 150 Haping Road, Nangang District, Harbin 150081, Heilongjiang, China
| | - Fengjiao Chen
- Department of Ultrasound, Harbin Medical University Cancer Hospital, 150 Haping Road, Nangang District, Harbin 150081, Heilongjiang, China
| | - Lingling Wang
- Department of Ultrasound, the Sixth Affiliated Hospital of Harbin Medical University, 998 Aiying Avenue, Songbei District, Harbin 150027, Heilongjiang, China.
| |
Collapse
|
32
|
Umemura M, Nakakaji R, Ishikawa Y. Physiological functions of calcium signaling via Orai1 in cancer. J Physiol Sci 2023; 73:21. [PMID: 37759164 DOI: 10.1186/s12576-023-00878-0] [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: 08/01/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023]
Abstract
Intracellular calcium (Ca2+) signaling regulates many cellular functions, including cell proliferation and migration, in both normal cells and cancer cells. Store-operated Ca2+ entry (SOCE) is a major mechanism by which Ca2+ is imported from the extracellular space to the intracellular space, especially in nonexcitable cells. Store-operated Ca2+ entry (SOCE) is also a receptor-regulated Ca2+ entry pathway that maintains Ca2+ homeostasis by sensing reduced Ca2+ levels in the endoplasmic reticulum (ER). In general, the activation of G protein-coupled receptors (GPCRs) or immunoreceptors, such as T-cell, B-cell and Fc receptors, results in the production of inositol 1,4,5-trisphosphate (IP3). IP3 binds to IP3 receptors located in the ER membrane. The, IP3 receptors in the ER membrane trigger a rapid and transient release of Ca2+ from the ER store. The resulting depletion of ER Ca2+ concentrations is sensed by the EF-hand motif of stromal interaction molecule (STIM), i.e., calcium sensor, which then translocates to the plasma membrane (PM). STIM interacts with Orai Ca2+ channel subunits (also known as CRACM1) on the PM, leading to Ca2+ influx from the extracellular space to increase intracellular Ca2+ concentrations. The physiological functions of Orai and STIM have been studied mainly with respect to their roles in the immune system. Based on numerous previous studies, Orai channels (Orai1, Orai2 and Orai3 channels) control Ca2+ release-activated Ca2+ (CRAC) currents and contribute to SOCE currents in other types of cells, including various cancer cells. There are many reports that Orai1 is involved in cell proliferation, migration, metastasis, apoptosis and epithelial-mesenchymal transition (EMT) in various cancers. We previously found that Orai1 plays important roles in cell apoptosis and migration in melanoma. Recently, we reported novel evidence of Orai1 in human oral squamous cell carcinoma (OSCC) cells and human cardiac fibroblasts (HCFs). In this review, we present multiple physiological functions of Orai1 in various cancer cells and cardiac fibroblasts, including our findings.
Collapse
Affiliation(s)
- Masanari Umemura
- Cardiovascular Research Institute, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan.
| | - Rina Nakakaji
- Cardiovascular Research Institute, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Yoshihiro Ishikawa
- Cardiovascular Research Institute, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan.
| |
Collapse
|
33
|
Hsieh DJY, Tsai BCK, Barik P, Shibu MA, Kuo CH, Kuo WW, Lin PY, Shih CY, Lin SZ, Ho TJ, Huang CY. Human adipose-derived stem cells preconditioned with a novel herbal formulation Jing Shi attenuate doxorubicin-induced cardiac damage. Aging (Albany NY) 2023; 15:9167-9181. [PMID: 37708248 PMCID: PMC10522400 DOI: 10.18632/aging.205026] [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: 05/15/2023] [Accepted: 08/21/2023] [Indexed: 09/16/2023]
Abstract
Pathological cardiac hypertrophy is a considerable contributor to global disease burden. Chinese herbal medicine (CHM) has been used to treat cardiovascular diseases since antiquity. Enhancing stem cell-mediated recovery through CHM represents a promising approach for protection against doxorubicin (Dox)-induced cardiac hypertrophy. Herein, we investigated whether human adipose-derived stem cells (hADSCs) preconditioned with novel herbal formulation Jing Si (JS) improved protective ability of stem cells against doxorubicin-induced cardiac damage. The effect of JS on hADSC viability and migration capacity was determined via MTT and migration assays, respectively. Co-culture of hADSC or JS-preconditioned hADSCs with H9c2 cells was analyzed with immunoblot, flow cytometry, TUNEL staining, LC3B staining, F-actin staining, and MitoSOX staining. The in vivo study was performed M-mode echocardiography after the treatment of JS and JS-preconditioned hADSCs by using Sprague Dawley (SD) rats. Our results indicated that JS at doses below 100 μg/mL had less cytotoxicity in hADSC and JS-preconditioned hADSCs exhibited better migration. Our results also revealed that DOX enhanced apoptosis, cardiac hypertrophy, and mitochondrial reactive oxygen species in DOX-challenged H9c2 cells, while H9c2 cells co-cultured with JS-preconditioned hADSCs alleviated these effects. It also enhanced the expression of autophagy marker LC3B, mTOR and CHIP in DOX-challenged H9c2 cells after co-culture with JS-preconditioned hADSCs. In Dox-challenged rats, the ejection fraction and fractional shortening improved in DOX-challenged SD rats exposed to JS-preconditioned hADSCs. Taken together, our data indicate that JS-preconditioned stem cells exhibit a cardioprotective capacity both in vitro and in vivo, highlighting the value of this therapeutic approach for regenerative therapy.
Collapse
Affiliation(s)
- 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
| | - Bruce Chi-Kang Tsai
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Parthasarathi Barik
- 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
- Department of Kinesiology and Health Science, College of William and Mary, Williamsburg, USA
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
- Ph.D. Program for Biotechnology Industry, China Medical University, Taichung, Taiwan
| | - Pi-Yu Lin
- Buddhist Compassion Relief Tzu Chi Foundation, Hualien, Taiwan
| | | | - Shinn-Zong Lin
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Hualien, Taiwan
- Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Tsung-Jung Ho
- Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Department of Chinese Medicine, Hualien Tzu Chi Hospital, Hualien, Taiwan
- School of Post-Baccalaureate Chinese Medicine, College of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Chih-Yang Huang
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, 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
- Graduate Institute of Basic Medical Science, China Medical University, Taichung City, Taiwan
| |
Collapse
|
34
|
Wang T, Shi S, Shi Y, Jiang P, Hu G, Ye Q, Shi Z, Yu K, Wang C, Fan G, Zhao S, Ma H, Chang ACY, Li Z, Bian Q, Lin CP. Chemical-induced phase transition and global conformational reorganization of chromatin. Nat Commun 2023; 14:5556. [PMID: 37689690 PMCID: PMC10492836 DOI: 10.1038/s41467-023-41340-4] [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: 01/27/2023] [Accepted: 08/29/2023] [Indexed: 09/11/2023] Open
Abstract
Chemicals or drugs can accumulate within biomolecular condensates formed through phase separation in cells. Here, we use super-resolution imaging to search for chemicals that induce phase transition within chromatin at the microscale. This microscopic screening approach reveals that adriamycin (doxorubicin) - a widely used anticancer drug that is known to interact with chromatin - specifically induces visible local condensation and global conformational change of chromatin in cancer and primary cells. Hi-C and ATAC-seq experiments systematically and quantitatively demonstrate that adriamycin-induced chromatin condensation is accompanied by weakened chromatin interaction within topologically associated domains, compartment A/B switching, lower chromatin accessibility, and corresponding transcriptomic changes. Mechanistically, adriamycin complexes with histone H1 and induces phase transition of H1, forming fibrous aggregates in vitro. These results reveal a phase separation-driven mechanism for a chemotherapeutic drug.
Collapse
Affiliation(s)
- Tengfei Wang
- School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
| | - Shuxiang Shi
- School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
- Lingang Laboratory, 200031, Shanghai, China
| | - Yuanyuan Shi
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200125, Shanghai, China
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Peipei Jiang
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200125, Shanghai, China
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Ganlu Hu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 201210, Shanghai, China
| | - Qinying Ye
- School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
| | - Zhan Shi
- School of Physical Science and Technology, ShanghaiTech University, 201210, Shanghai, China
| | - Kexin Yu
- School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
- iHuman Institute, ShanghaiTech University, 201010, Shanghai, China
| | - Chenguang Wang
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200125, Shanghai, China
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
- Department of Cardiology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guoping Fan
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 201210, Shanghai, China
| | - Suwen Zhao
- School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
- iHuman Institute, ShanghaiTech University, 201010, Shanghai, China
| | - Hanhui Ma
- School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
| | - Alex C Y Chang
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200125, Shanghai, China
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
- Department of Cardiology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhi Li
- School of Physical Science and Technology, ShanghaiTech University, 201210, Shanghai, China
| | - Qian Bian
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200125, Shanghai, China.
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
| | - Chao-Po Lin
- School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China.
| |
Collapse
|
35
|
Uche N, Dai Q, Lai S, Kolander K, Thao M, Schibly E, Sendaydiego X, Zielonka J, Benjamin IJ. Carvedilol Phenocopies PGC-1α Overexpression to Alleviate Oxidative Stress, Mitochondrial Dysfunction and Prevent Doxorubicin-Induced Toxicity in Human iPSC-Derived Cardiomyocytes. Antioxidants (Basel) 2023; 12:1585. [PMID: 37627583 PMCID: PMC10451268 DOI: 10.3390/antiox12081585] [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: 07/05/2023] [Revised: 07/30/2023] [Accepted: 08/01/2023] [Indexed: 08/27/2023] Open
Abstract
Doxorubicin (DOX), one of the most effective and widely used anticancer drugs, has the major limitation of cancer treatment-related cardiotoxicity (CTRTOX) in the clinic. Reactive oxygen species (ROS) generation and mitochondrial dysfunction are well-known consequences of DOX-induced injury to cardiomyocytes. This study aimed to explore the mitochondrial functional consequences and associated mechanisms of pretreatment with carvedilol, a ß-blocking agent known to exert protection against DOX toxicity. When disease modeling was performed using cultured rat cardiac muscle cells (H9c2 cells) and human iPSC-derived cardiomyocytes (iPSC-CMs), we found that prophylactic carvedilol mitigated not only the DOX-induced suppression of mitochondrial function but that the mitochondrial functional readout of carvedilol-pretreated cells mimicked the readout of cells overexpressing the major regulator of mitochondrial biogenesis, PGC-1α. Carvedilol pretreatment reduces mitochondrial oxidants, decreases cell death in both H9c2 cells and human iPSC-CM and maintains the cellular 'redox poise' as determined by sustained expression of the redox sensor Keap1 and prevention of DOX-induced Nrf2 nuclear translocation. These results indicate that, in addition to the already known ROS-scavenging effects, carvedilol has a hitherto unrecognized pro-reducing property against the oxidizing conditions induced by DOX treatment, the sequalae of DOX-induced mitochondrial dysfunction and compromised cell viability. The novel findings of our preclinical studies suggest future trial design of carvedilol prophylaxis, such as prescreening for redox state, might be an alternative strategy for preventing oxidative stress writ large in lieu of the current lack of clinical evidence for ROS-scavenging agents.
Collapse
Affiliation(s)
- Nnamdi Uche
- Cardiovascular Center, Department of Physiology, Medical College of Wisconsin, 8701 W Watertown Plank Road, Milwaukee, WI 53226, USA;
| | - Qiang Dai
- Cardiovascular Center, Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, 8701 W Watertown Plank Road, Milwaukee, WI 53226, USA; (Q.D.); (S.L.); (K.K.); (M.T.); (E.S.); (X.S.)
| | - Shuping Lai
- Cardiovascular Center, Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, 8701 W Watertown Plank Road, Milwaukee, WI 53226, USA; (Q.D.); (S.L.); (K.K.); (M.T.); (E.S.); (X.S.)
| | - Kurt Kolander
- Cardiovascular Center, Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, 8701 W Watertown Plank Road, Milwaukee, WI 53226, USA; (Q.D.); (S.L.); (K.K.); (M.T.); (E.S.); (X.S.)
| | - Mai Thao
- Cardiovascular Center, Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, 8701 W Watertown Plank Road, Milwaukee, WI 53226, USA; (Q.D.); (S.L.); (K.K.); (M.T.); (E.S.); (X.S.)
| | - Elizabeth Schibly
- Cardiovascular Center, Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, 8701 W Watertown Plank Road, Milwaukee, WI 53226, USA; (Q.D.); (S.L.); (K.K.); (M.T.); (E.S.); (X.S.)
| | - Xavier Sendaydiego
- Cardiovascular Center, Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, 8701 W Watertown Plank Road, Milwaukee, WI 53226, USA; (Q.D.); (S.L.); (K.K.); (M.T.); (E.S.); (X.S.)
| | - Jacek Zielonka
- Free Radical Laboratory, Department of Biophysics, Medical College of Wisconsin, 8701 W Watertown Plank Road, Milwaukee, WI 53226, USA;
| | - Ivor J. Benjamin
- Cardiovascular Center, Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, 8701 W Watertown Plank Road, Milwaukee, WI 53226, USA; (Q.D.); (S.L.); (K.K.); (M.T.); (E.S.); (X.S.)
| |
Collapse
|
36
|
Mazevet M, Belhadef A, Ribeiro M, Dayde D, Llach A, Laudette M, Belleville T, Mateo P, Gressette M, Lefebvre F, Chen J, Bachelot-Loza C, Rucker-Martin C, Lezoualch F, Crozatier B, Benitah JP, Vozenin MC, Fischmeister R, Gomez AM, Lemaire C, Morel E. EPAC1 inhibition protects the heart from doxorubicin-induced toxicity. eLife 2023; 12:e83831. [PMID: 37551870 PMCID: PMC10484526 DOI: 10.7554/elife.83831] [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: 09/29/2022] [Accepted: 08/03/2023] [Indexed: 08/09/2023] Open
Abstract
Anthracyclines, such as doxorubicin (Dox), are widely used chemotherapeutic agents for the treatment of solid tumors and hematologic malignancies. However, they frequently induce cardiotoxicity leading to dilated cardiomyopathy and heart failure. This study sought to investigate the role of the exchange protein directly activated by cAMP (EPAC) in Dox-induced cardiotoxicity and the potential cardioprotective effects of EPAC inhibition. We show that Dox induces DNA damage and cardiomyocyte cell death with apoptotic features. Dox also led to an increase in both cAMP concentration and EPAC1 activity. The pharmacological inhibition of EPAC1 (with CE3F4) but not EPAC2 alleviated the whole Dox-induced pattern of alterations. When administered in vivo, Dox-treated WT mice developed a dilated cardiomyopathy which was totally prevented in EPAC1 knock-out (KO) mice. Moreover, EPAC1 inhibition potentiated Dox-induced cell death in several human cancer cell lines. Thus, EPAC1 inhibition appears as a potential therapeutic strategy to limit Dox-induced cardiomyopathy without interfering with its antitumoral activity.
Collapse
Affiliation(s)
| | | | | | | | | | - Marion Laudette
- Institut des Maladies Metaboliques et Cardiovasculaires - I2MC, INSERM, Université de ToulouseToulouseFrance
| | - Tiphaine Belleville
- Innovations Thérapeutiques en Hémostase - UMR-S 1140, INSERM, Faculté de Pharmacie, Université Paris Descartes, Sorbonne Paris CitéParisFrance
| | | | | | | | - Ju Chen
- Basic Cardiac Research UCSD School of Medicine La JollaSan DiegoUnited States
| | - Christilla Bachelot-Loza
- Innovations Thérapeutiques en Hémostase - UMR-S 1140, INSERM, Faculté de Pharmacie, Université Paris Descartes, Sorbonne Paris CitéParisFrance
| | - Catherine Rucker-Martin
- Faculté de Médecine, Université Paris-SaclayLe Kremlin BicêtreFrance
- Inserm UMR_S 999, Hôpital Marie LannelongueLe Plessis RobinsonFrance
| | - Frank Lezoualch
- Institut des Maladies Metaboliques et Cardiovasculaires - I2MC, INSERM, Université de ToulouseToulouseFrance
| | | | | | | | | | | | - Christophe Lemaire
- Université Paris-SaclayOrsayFrance
- Université Paris-Saclay, UVSQ, InsermOrsayFrance
| | | |
Collapse
|
37
|
Patintingan CG, Louisa M, Juniantito V, Arozal W, Hanifah S, Wanandi SI, Thandavarayan R. Moringa oleifera Leaves Extract Ameliorates Doxorubicin-Induced Cardiotoxicity via Its Mitochondrial Biogenesis Modulatory Activity in Rats. J Exp Pharmacol 2023; 15:307-319. [PMID: 37525636 PMCID: PMC10387274 DOI: 10.2147/jep.s413256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 07/12/2023] [Indexed: 08/02/2023] Open
Abstract
Background Doxorubicin, an anthracycline class of anticancer, is an effective chemotherapeutic agent with serious adverse effects, mainly cardiotoxicity. Several possible causes of doxorubicin cardiotoxicity are increased oxidative stress, nucleic acid and protein synthesis inhibition, cardiomyocyte apoptosis, and mitochondrial biogenesis disruptions. Moringa oleifera (MO), a naturally derived medicine, is known for its antioxidative properties and activity in alleviating mitochondrial dysfunction. To determine the potency and possible cardioprotective mechanism of MO leaves aqueous extract via the mitochondrial biogenesis pathway in doxorubicin-induced rats. Methods Twenty-four Sprague-Dawley rats were divided into four groups of six. The first group was normal rats; the second group was treated with doxorubicin 4 mg/kg BW intraperitoneally once weekly for four weeks; the third and fourth groups were treated with doxorubicin 4 mg/kg BW intraperitoneally once weekly, and MO leaves extract at 200 mg/kg BW or 400 mg/kg BW orally daily, for four weeks. At the end of the fourth week, blood and cardiac tissues were obtained and analyzed for cardiac biomarkers, mitochondrial DNA copy number, mRNA expressions of peroxisome-activated receptor-gamma coactivator-1 alpha (PGC-1α), the nuclear factor erythroid 2-related factor 2 (Nrf2), superoxide dismutase 2 (SOD2), caspase 3, the activity of glutathione peroxidase (GPx), levels of 8-hydroxy-2-deoxyguanosine (8-OH-dG), and malondialdehyde. Results MO leaves extract was shown to decrease biomarkers of cardiac damage (LDH and CK-MB), malondialdehyde levels, and GPx activity. These changes align with the reduction of mRNA expressions of caspase-3, the increase of mRNA expressions of PGC-1α and Nrf2, and the elevation of mitochondrial DNA copy number. MO leaves extracts did not influence the mRNA expressions of superoxide dismutase 2 (SOD2) or the levels of 8-OH-dG. Conclusion Moringa oleifera leaves extract ameliorates doxorubicin-induced cardiotoxicity by reducing apoptosis and restoring gene expression of PGC-1α and Nrf2, a key regulator in mitochondrial biogenesis.
Collapse
Affiliation(s)
| | - Melva Louisa
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Vetnizah Juniantito
- Department of Veterinary Clinic Reproduction and Pathology, Faculty of Veterinary Medicine, Agriculture Institute of Bogor, Bogor, Indonesia
| | - Wawaimuli Arozal
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Silmi Hanifah
- Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Septelia Inawati Wanandi
- Department of Biochemistry and Molecular Biology, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
| | - Rajarajan Thandavarayan
- Department of Cardiovascular Sciences Houston Methodist Research Institute, Houston, TX, USA
| |
Collapse
|
38
|
Voufo RA, Kouotou AE, Tatah NJ, TeTo G, Gueguim C, Ngondé CME, Njiguet Tepa AG, Gabin A, Amazia F, Yembeau NL, Kouanfack C, Anatole PC. Relation between interleukin-6 concentrations and oxidative status of HIV infected patients with /or at risk of Kaposi disease in Yaounde. Virol J 2023; 20:165. [PMID: 37491285 PMCID: PMC10369812 DOI: 10.1186/s12985-023-02109-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 06/26/2023] [Indexed: 07/27/2023] Open
Abstract
OBJECTIVE To investigate the relation between interleukin-6 concentration and oxidative status of HIV infected patients with or at risk of Kaposi's disease in Yaoundé. METHODS We conducted a two-months cross-sectional study on 87 consenting HIV infected patients followed at the Day Hospital of the Yaoundé Central Hospital. Serum/plasma obtained after centrifugation of blood collected in dry/EDTA tubes was used for the determination of Human Herpes Virus-8 antigen (HHV-8) and Interleukin-6 (IL-6) by the ELISA technique, and that of oxidative stress markers: Malondialdehyde (MDA) reduced Glutathione (GSH) and total antioxidant capacity by spectrophotometry. RESULTS Subjects belonging to the [40-50[year-old age group were mainly represented in our study population with 43.7%. The average age was 44.6 ± 10.4 years with extremes ranging from 26 to 72 years. The sex ratio was 0.24. Our population was mainly represented by people infected with HIV type I (90.8%) and 3.4% had developed clinical signs of Kaposi's disease. The prevalence of the HHV-8 antigen was 57.5%. Immune and oxidative parameters did not vary with age, sex and therapeutic line. We noted a significant increase in IL-6 concentrations in patients positive to the HHV-8 antigen for IL-6 concentrations < 37 (P = 0.005; CI= [0.40; 0.59]. MDA and GSH concentrations increased significantly with the HHV-8 infection (P < 0.0001; CI= [0.40; 0.59] and P < 0.0001; CI= [13.30;21.45], respectively). Total antioxidant capacity (FRAP) decreased significantly with HHV-8 infection (P = 0.004; CI= [-69.18; -13.78]). We noted a significant increase in MDA concentrations in patients taking their ARVs irregularly, (P < 0.0001). CONCLUSION Our study showed a weak positive correlation between IL-6 and MDA, a strong negative correlation between FRAP and MDA and a strong positive correlation between MDA and GSH highlighting the association of these few markers of oxidative stress and Il-6 to the risk of Kaposi's disease.
Collapse
Affiliation(s)
- Roger Ahouga Voufo
- Institute of Medical Research and Medicinal Plants Studies (IMPM), Yaounde, Cameroon
- Department of Biochemistry, Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaounde, Cameroon
| | - Armand Emmanuel Kouotou
- Department of Internal Medicine Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaounde, Cameroon
| | - Nchinda Jones Tatah
- Department of Biochemistry, Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaounde, Cameroon
| | - Georges TeTo
- Centre International de Référence Chantal Biya (CICRB), Yaounde, Cameroon
| | - Cédric Gueguim
- Department of Biochemistry, Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaounde, Cameroon
| | | | | | - Arnaud Gabin
- Department of Biochemistry, Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaounde, Cameroon
| | - Falmata Amazia
- Department of Biochemistry, Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaounde, Cameroon
| | - Natacha Lena Yembeau
- Department of Biochemistry, Faculty of Science, University of Dschang, Dschang, Cameroon
| | - Charles Kouanfack
- Faculty of Medicine and pharmaceutical Sciences, University of Dschang, Dschang, Cameroon
| | - Pieme Constant Anatole
- Department of Biochemistry, Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaounde, Cameroon.
| |
Collapse
|
39
|
Reinal I, Ontoria-Oviedo I, Selva M, Casini M, Peiró-Molina E, Fambuena-Santos C, Climent AM, Balaguer J, Cañete A, Mora J, Raya Á, Sepúlveda P. Modeling Cardiotoxicity in Pediatric Oncology Patients Using Patient-Specific iPSC-Derived Cardiomyocytes Reveals Downregulation of Cardioprotective microRNAs. Antioxidants (Basel) 2023; 12:1378. [PMID: 37507917 PMCID: PMC10376252 DOI: 10.3390/antiox12071378] [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: 06/04/2023] [Revised: 06/25/2023] [Accepted: 06/29/2023] [Indexed: 07/30/2023] Open
Abstract
Anthracyclines are widely used in the treatment of many solid cancers, but their efficacy is limited by cardiotoxicity. As the number of pediatric cancer survivors continues to rise, there has been a concomitant increase in people living with anthracycline-induced cardiotoxicity. Accordingly, there is an ongoing need for new models to better understand the pathophysiological mechanisms of anthracycline-induced cardiac damage. Here we generated induced pluripotent stem cells (iPSCs) from two pediatric oncology patients with acute cardiotoxicity induced by anthracyclines and differentiated them to ventricular cardiomyocytes (hiPSC-CMs). Comparative analysis of these cells (CTX hiPSC-CMs) and control hiPSC-CMs revealed that the former were significantly more sensitive to cell injury and death from the anthracycline doxorubicin (DOX), as measured by viability analysis, cleaved caspase 3 expression, oxidative stress, genomic and mitochondrial damage and sarcomeric disorganization. The expression of several mRNAs involved in structural integrity and inflammatory response were also differentially affected by DOX. Functionally, optical mapping analysis revealed higher arrythmia complexity after DOX treatment in CTX iPSC-CMs. Finally, using a panel of previously identified microRNAs associated with cardioprotection, we identified lower levels of miR-22-3p, miR-30b-5p, miR-90b-3p and miR-4732-3p in CTX iPSC-CMs under basal conditions. Our study provides valuable phenotype information for cellular models of cardiotoxicity and highlights the significance of using patient-derived cardiomyocytes for studying the associated pathogenic mechanisms.
Collapse
Affiliation(s)
- Ignacio Reinal
- Regenerative Medicine and Heart Transplantation Unit, Health Research Institute Hospital la Fe, 46026 Valencia, Spain
| | - Imelda Ontoria-Oviedo
- Regenerative Medicine and Heart Transplantation Unit, Health Research Institute Hospital la Fe, 46026 Valencia, Spain
| | - Marta Selva
- Regenerative Medicine and Heart Transplantation Unit, Health Research Institute Hospital la Fe, 46026 Valencia, Spain
| | - Marilù Casini
- Regenerative Medicine and Heart Transplantation Unit, Health Research Institute Hospital la Fe, 46026 Valencia, Spain
| | - Esteban Peiró-Molina
- Regenerative Medicine and Heart Transplantation Unit, Health Research Institute Hospital la Fe, 46026 Valencia, Spain
- Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain
| | | | - Andreu M Climent
- ITACA Institute, Universitat Politècnica de València, 46026 Valencia, Spain
| | - Julia Balaguer
- Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain
- Transtational Research in Cancer Unit-Pediatric Oncology, Health Research Institute Hospital La Fe, 46026 Valencia, Spain
| | - Adela Cañete
- Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain
- Transtational Research in Cancer Unit-Pediatric Oncology, Health Research Institute Hospital La Fe, 46026 Valencia, Spain
- Department of Pediatrics, University of Valencia, 46010 Valencia, Spain
| | - Jaume Mora
- Oncology Service, Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
| | - Ángel Raya
- Regenerative Medicine Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, 08908 Barcelona, Spain
- Program for Clinical Translation of Regenerative Medicine in Catalonia-P-[CMRC], L'Hospitalet de Llobregat, 08908 Barcelona, Spain
- Centro de Investigación Biomédica en Red Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Carlos III Institute of Health, 28029 Madrid, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
| | - Pilar Sepúlveda
- Regenerative Medicine and Heart Transplantation Unit, Health Research Institute Hospital la Fe, 46026 Valencia, Spain
- Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Carlos III Institute of Health, 28029 Madrid, Spain
- Department of Pathology, University of Valencia, 46010 Valencia, Spain
| |
Collapse
|
40
|
Salerno N, Scalise M, Marino F, Filardo A, Chiefalo A, Panuccio G, Torella M, De Angelis A, De Rosa S, Ellison-Hughes GM, Urbanek K, Viglietto G, Torella D, Cianflone E. A Mouse Model of Dilated Cardiomyopathy Produced by Isoproterenol Acute Exposure Followed by 5-Fluorouracil Administration. J Cardiovasc Dev Dis 2023; 10:225. [PMID: 37367390 DOI: 10.3390/jcdd10060225] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 05/19/2023] [Accepted: 05/20/2023] [Indexed: 06/28/2023] Open
Abstract
Appropriate dilated cardiomyopathy (DCM) animal models are highly desirable considering the pathophysiological and clinical heterogeneity of DCM. Genetically modified mice are the most widely and intensively utilized research animals for DCM. However, to translate discoveries from basic science into new and personalized medical applications, research in non-genetically based DCM models remains a key issue. Here, we characterized a mouse model of non-ischemic DCM induced by a stepwise pharmacologic regime of Isoproterenol (ISO) high dose bolus followed by a low dose systemic injection of the chemotherapy agent, 5-Fluorouracil (5-FU). C57BL/6J mice were injected with ISO and, 3 days after, were randomly assigned to saline or 5-FU. Echocardiography and a strain analysis show that ISO + 5FU in mice induces progressive left ventricular (LV) dilation and reduced systolic function, along with diastolic dysfunction and a persistent global cardiac contractility depression through 56 days. While mice treated with ISO alone recover anatomically and functionally, ISO + 5-FU causes persistent cardiomyocyte death, ensuing in cardiomyocyte hypertrophy through 56 days. ISO + 5-FU-dependent damage was accompanied by significant myocardial disarray and fibrosis along with exaggerated oxidative stress, tissue inflammation and premature cell senescence accumulation. In conclusions, a combination of ISO + 5FU produces anatomical, histological and functional cardiac alterations typical of DCM, representing a widely available, affordable, and reproducible mouse model of this cardiomyopathy.
Collapse
Affiliation(s)
- Nadia Salerno
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Mariangela Scalise
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Fabiola Marino
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Andrea Filardo
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Antonio Chiefalo
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Giuseppe Panuccio
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Michele Torella
- Department of Experimental Medicine, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Antonella De Angelis
- Department of Experimental Medicine, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Salvatore De Rosa
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Georgina M Ellison-Hughes
- Centre for Human and Applied Physiological Sciences, School of Basic and Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Konrad Urbanek
- Department of Molecular Medicine and Medical Biotechnology, Federico II University, 88121 Naples, Italy
| | - Giuseppe Viglietto
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Daniele Torella
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Eleonora Cianflone
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| |
Collapse
|
41
|
Montalvo RN, Boeno FP, Dowllah IM, Moritz CEJ, Nguyen BL, Doerr V, Bomkamp MP, Smuder AJ. Exercise and Doxorubicin Modify Markers of Iron Overload and Cardiolipin Deficiency in Cardiac Mitochondria. Int J Mol Sci 2023; 24:ijms24097689. [PMID: 37175395 PMCID: PMC10177936 DOI: 10.3390/ijms24097689] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/10/2023] [Accepted: 04/19/2023] [Indexed: 05/15/2023] Open
Abstract
Doxorubicin (DOX) is a chemotherapeutic agent highly effective at limiting cancer progression. Despite the efficacy of this anticancer drug, the clinical use of DOX is limited due to cardiotoxicity. The cardiac mitochondria are implicated as the primary target of DOX, resulting in inactivation of electron transport system complexes, oxidative stress, and iron overload. However, it is established that the cardiac mitochondrial subpopulations reveal differential responses to DOX exposure, with subsarcolemmal (SS) mitochondria demonstrating redox imbalance and the intermyofibrillar (IMF) mitochondria showing reduced respiration. In this regard, exercise training is an effective intervention to prevent DOX-induced cardiac dysfunction. Although it is clear that exercise confers mitochondrial protection, it is currently unknown if exercise training mitigates DOX cardiac mitochondrial toxicity by promoting beneficial adaptations to both the SS and IMF mitochondria. To test this, SS and IMF mitochondria were isolated from sedentary and exercise-preconditioned female Sprague Dawley rats exposed to acute DOX treatment. Our findings reveal a greater effect of exercise preconditioning on redox balance and iron handling in the SS mitochondria of DOX-treated rats compared to IMF, with rescue of cardiolipin synthase 1 expression in both subpopulations. These results demonstrate that exercise preconditioning improves mitochondrial homeostasis when combined with DOX treatment, and that the SS mitochondria display greater protection compared to the IMF mitochondria. These data provide important insights into the molecular mechanisms that are in part responsible for exercise-induced protection against DOX toxicity.
Collapse
Affiliation(s)
- Ryan N Montalvo
- Department of Applied Physiology & Kinesiology, University of Florida, Gainesville, FL 32611, USA
| | - Franccesco P Boeno
- Department of Applied Physiology & Kinesiology, University of Florida, Gainesville, FL 32611, USA
| | - Imtiaz M Dowllah
- Department of Applied Physiology & Kinesiology, University of Florida, Gainesville, FL 32611, USA
| | - Cesar E Jacintho Moritz
- Department of Applied Physiology & Kinesiology, University of Florida, Gainesville, FL 32611, USA
| | - Branden L Nguyen
- Department of Applied Physiology & Kinesiology, University of Florida, Gainesville, FL 32611, USA
| | - Vivian Doerr
- Department of Applied Physiology & Kinesiology, University of Florida, Gainesville, FL 32611, USA
| | - Matthew P Bomkamp
- Department of Applied Physiology & Kinesiology, University of Florida, Gainesville, FL 32611, USA
| | - Ashley J Smuder
- Department of Applied Physiology & Kinesiology, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
42
|
Kaur G, Wang X, Li X, Ong H, He X, Cai C. Overexpression of GREM1 Improves the Survival Capacity of Aged Cardiac Mesenchymal Progenitor Cells via Upregulation of the ERK/NRF2-Associated Antioxidant Signal Pathway. Cells 2023; 12:1203. [PMID: 37190112 PMCID: PMC10136744 DOI: 10.3390/cells12081203] [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: 03/13/2023] [Revised: 04/11/2023] [Accepted: 04/17/2023] [Indexed: 05/17/2023] Open
Abstract
Ischemic heart disease is the leading cause of mortality in the United States. Progenitor cell therapy can restore myocardial structure and function. However, its efficacy is severely limited by cell aging and senescence. Gremlin-1 (GREM1), a member of the bone morphogenetic protein antagonist family, has been implicated in cell proliferation and survival. However, GREM1's role in cell aging and senescence has never been investigated in human cardiac mesenchymal progenitor cells (hMPCs). Therefore, this study assessed the hypothesis that overexpression of GREM1 rejuvenates the cardiac regenerative potential of aging hMPCs to a youthful stage and therefore allows better capacity for myocardial repair. We recently reported that a subpopulation of hMPCs with low mitochondrial membrane potential can be sorted from right atrial appendage-derived cells in patients with cardiomyopathy and exhibit cardiac reparative capacity in a mouse model of myocardial infarction. In this study, lentiviral particles were used to overexpress GREM1 in these hMPCs. Protein and mRNA expression were assessed through Western blot and RT-qPCR. FACS analysis for Annexin V/PI staining and lactate dehydrogenase assay were used to assess cell survival. It was observed that cell aging and cell senescence led to a decrease in GREM1 expression. In addition, overexpression of GREM1 led to a decrease in expression of senescence genes. Overexpression of GREM1 led to no significant change in cell proliferation. However, GREM1 appeared to have an anti-apoptotic effect, with an increase in survival and decrease in cytotoxicity evident in GREM1-overexpressing hMPCs. Overexpressing GREM1 also induced cytoprotective properties by decreasing reactive oxidative species and mitochondrial membrane potential. This result was associated with increased expression of antioxidant proteins, such as SOD1 and catalase, and activation of the ERK/NRF2 survival signal pathway. Inhibition of ERK led to a decrease in GREM1-mediated rejuvenation in terms of cell survival, which suggests that an ERK-dependent pathway may be involved. Taken altogether, these results indicate that overexpression of GREM1 can allow aging hMPCs to adopt a more robust phenotype with improved survival capacity, which is associated with an activated ERK/NRF2 antioxidant signal pathway.
Collapse
Affiliation(s)
- Gurleen Kaur
- Department of Molecular and Cellular Physiology, Department of Medicine, Albany Medical College, Albany, NY 12208, USA; (G.K.); (X.W.); (X.L.)
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Xiaoliang Wang
- Department of Molecular and Cellular Physiology, Department of Medicine, Albany Medical College, Albany, NY 12208, USA; (G.K.); (X.W.); (X.L.)
- Division of Surgical Sciences, Department of Surgery, University of Virginia, Charlottesville, VA 22903, USA
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (H.O.); (X.H.)
| | - Xiuchun Li
- Department of Molecular and Cellular Physiology, Department of Medicine, Albany Medical College, Albany, NY 12208, USA; (G.K.); (X.W.); (X.L.)
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (H.O.); (X.H.)
| | - Hannah Ong
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (H.O.); (X.H.)
| | - Xiangfei He
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (H.O.); (X.H.)
| | - Chuanxi Cai
- Department of Molecular and Cellular Physiology, Department of Medicine, Albany Medical College, Albany, NY 12208, USA; (G.K.); (X.W.); (X.L.)
- Division of Surgical Sciences, Department of Surgery, University of Virginia, Charlottesville, VA 22903, USA
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (H.O.); (X.H.)
| |
Collapse
|
43
|
Cicek B, Hacimuftuoglu A, Yeni Y, Danisman B, Ozkaraca M, Mokhtare B, Kantarci M, Spanakis M, Nikitovic D, Lazopoulos G, Tsarouhas K, Tsatsakis A, Taghizadehghalehjoughi A. Chlorogenic Acid Attenuates Doxorubicin-Induced Oxidative Stress and Markers of Apoptosis in Cardiomyocytes via Nrf2/HO-1 and Dityrosine Signaling. J Pers Med 2023; 13:jpm13040649. [PMID: 37109035 PMCID: PMC10140899 DOI: 10.3390/jpm13040649] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/26/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
(1) Background: Doxorubicin (DOX) is extensively used for cancer treatments; however, its clinical application is limited because of its cardiotoxic adverse effects. A combination of DOX and agents with cardioprotective properties is an effective strategy to ameliorate DOX-related cardiotoxicity. Polyphenolic compounds are ideal for the investigation of novel cardioprotective agents. Chlorogenic acid (CGA), an essential dietary polyphenol found in plants, has been previously reported to exert antioxidant, cardioprotective, and antiapoptotic properties. The current research evaluated CGA's in vivo cardioprotective properties in DOX-induced cardiotoxicity and the probable mechanisms underlying this protection. (2) Methods: CGA's cardioprotective properties were investigated in rats that were treated with CGA (100 mg/kg, p.o.) for fourteen days. The experimental model of cardiotoxicity was induced with a single intraperitoneal (15 mg/kg i.p.) injection of DOX on the 10th day. (3) Results: Treatment with CGA significantly improved the DOX-caused altered cardiac damage markers (LDH, CK-MB, and cTn-T), and a marked improvement in cardiac histopathological features accompanied this. DOX downregulated the expression of Nrf2/HO-1 signaling pathways, and the CGA reversed this effect. Consistently, caspase-3, an apoptotic-related marker, and dityrosine expression were suppressed, while Nrf2 and HO-1 expressions were elevated in the cardiac tissues of DOX-treated rats after treatment with the CGA. Furthermore, the recovery was confirmed by the downregulation of 8-OHdG and dityrosine (DT) expressions in immunohistochemical findings. (4) Conclusions: CGA demonstrated a considerable cardioprotective effect against DOX-induced cardiotoxicity. One of the possible mechanisms for these protective properties was the upregulation of the Nrf2/HO-1-dependent pathway and the downregulation of DT, which may ameliorate oxidative stress and cardiomyocyte apoptosis. These findings suggest that CGA may be cardioprotective, particularly in patients receiving DOX-based chemotherapy.
Collapse
Affiliation(s)
- Betul Cicek
- Department of Physiology, Faculty of Medicine, Erzincan Binali Yildirim University, 24100 Erzincan, Turkey
| | - Ahmet Hacimuftuoglu
- Department of Medical Pharmacology, Faculty of Medicine, Ataturk University, 25240 Erzurum, Turkey
| | - Yesim Yeni
- Department of Medical Pharmacology, Faculty of Medicine, Malatya Turgut Ozal University, 44210 Malatya, Turkey
| | - Betul Danisman
- Department of Biophysics, Faculty of Medicine, Ataturk University, 25240 Erzurum, Turkey
| | - Mustafa Ozkaraca
- Department of Pathology, Faculty of Veterinary, Sivas Cumhuriyet University, 58140 Sivas, Turkey
| | - Behzad Mokhtare
- Department of Pathology, Faculty of Veterinary, Sivas Cumhuriyet University, 58140 Sivas, Turkey
| | - Mecit Kantarci
- Department of Radiology, Faculty of Medicine, Ataturk University, 25240 Erzurum, Turkey
| | - Marios Spanakis
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Dragana Nikitovic
- Dragana Nikitovic, Laboratory of Histology-Embryology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Georgios Lazopoulos
- Department of Cardiac Surgery, University General Hospital of Heraklion, Medical School, University of Crete, 71003 Heraklion, Greece
| | | | - Aristidis Tsatsakis
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Ali Taghizadehghalehjoughi
- Department of Medical Pharmacology, Faculty of Medicine, Bilecik Seyh Edebali University, 11230 Bilecik, Turkey
| |
Collapse
|
44
|
Zhang XJ, Li L, Wang AL, Guo HX, Zhao HP, Chi RF, Xu HY, Yang LG, Li B, Qin FZ, Wang JP. GSK2795039 prevents RIP1-RIP3-MLKL-mediated cardiomyocyte necroptosis in doxorubicin-induced heart failure through inhibition of NADPH oxidase-derived oxidative stress. Toxicol Appl Pharmacol 2023; 463:116412. [PMID: 36764612 DOI: 10.1016/j.taap.2023.116412] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 01/19/2023] [Accepted: 01/31/2023] [Indexed: 02/11/2023]
Abstract
Doxorubicin (DOX), which is widely used for the treatment of cancer, induces cardiomyopathy associated with NADPH oxidase-derived reactive oxygen species. GSK2795039 is a novel small molecular NADPH oxidase 2 (Nox2) inhibitor. In this study, we investigated whether GSK2795039 prevents receptor-interacting protein kinase 1 (RIP1)-RIP3-mixed lineage kinase domain-like protein (MLKL)-mediated cardiomyocyte necroptosis in DOX-induced heart failure through NADPH oxidase inhibition. Eight-week old mice were randomly divided into 4 groups: control, GSK2795039, DOX and DOX plus GSK2795039. H9C2 cardiomyocytes were treated with DOX and GSK2795039. In DOX-treated mice, the survival rate was reduced, left ventricular (LV) end-systolic dimension was increased and LV fractional shortening was decreased, and these alterations were attenuated by the GSK2795039 treatment. GSK2795039 inhibited not only myocardial NADPH oxidase subunit gp91phox (Nox2) protein, but also p22phox, p47phox and p67phox proteins and prevented oxidative stress 8-hydroxy-2'-deoxyguanosine levels in DOX-treated mice. RIP3 protein and phosphorylated RIP1 (p-RIP1), p-RIP3 and p-MLKL proteins, reflective of their respective kinase activities, markers of necroptosis, were markedly increased in DOX-treated mice, and the increases were prevented by GSK2795039. GSK2795039 prevented the increases in serum lactate dehydrogenase and myocardial fibrosis in DOX-treated mice. Similarly, in DOX-treated cardiomyocytes, GSK2795039 improved cell viability, attenuated apoptosis and necrosis and prevented the increases in p-RIP1, p-RIP3 and p-MLKL expression. In conclusion, GSK2795039 prevents RIP1-RIP3-MLKL-mediated cardiomyocyte necroptosis through inhibition of NADPH oxidase-derived oxidative stress, leading to the improvement of myocardial remodeling and function in DOX-induced heart failure. These findings suggest that GSK2795039 may have implications for the treatment of DOX-induced cardiomyopathy.
Collapse
Affiliation(s)
- Xiao-Juan Zhang
- The Second Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan 030001, Shanxi, PR China; Shanxi Province Cardiovascular Hospital, Taiyuan 030024, Shanxi, PR China
| | - Lu Li
- Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
| | - Ai-Ling Wang
- The Second Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
| | - Hong-Xia Guo
- The Second Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
| | - Hui-Ping Zhao
- The Second Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
| | - Rui-Fang Chi
- The Second Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
| | - Hui-Yu Xu
- Shanxi Medical University, Taiyuan 030001, Shanxi, PR China; Shanxi Province Cardiovascular Hospital, Taiyuan 030024, Shanxi, PR China
| | - Li-Guo Yang
- The Second Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan 030001, Shanxi, PR China
| | - Bao Li
- The Second Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan 030001, Shanxi, PR China.
| | - Fu-Zhong Qin
- The Second Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan 030001, Shanxi, PR China.
| | - Jia-Pu Wang
- Shanxi Province Cardiovascular Hospital, Taiyuan 030024, Shanxi, PR China
| |
Collapse
|
45
|
Chen J, Zhang M, Zou H, Aniagu S, Jiang Y, Chen T. PM2.5 induces mitochondrial dysfunction via AHR-mediated cyp1a1 overexpression during zebrafish heart development. Toxicology 2023; 487:153466. [PMID: 36841371 DOI: 10.1016/j.tox.2023.153466] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 02/14/2023] [Accepted: 02/22/2023] [Indexed: 02/27/2023]
Abstract
Accumulating evidence suggests an association between maternal PM2.5 exposure and congenital heart diseases, but the underlying mechanisms remain unclear. We previously reported that PM2.5 induces cardiac malformations in zebrafish embryos via the aryl hydrocarbon receptor (AHR) pathway, which mediates the generation of reactive oxygen species (ROS). Since mitochondria are not only the main source of ROS but also sensitive to oxidative damage, we hypothesize that mitochondria may play an important role in the cardiac developmental toxicity of PM2.5. In this study, we demonstrated that extractable organic matter (EOM) from PM2.5 caused mitochondrial dysfunction in the heart of zebrafish embryos, including increased mitochondrial ROS (mtROS) levels, mitochondrial permeability transition pore (mPTP) opening, mitochondrial membrane potential (MMP) collapse, reduced mitochondrial ATP levels, and decreased expression levels of the mRNAs encoding mitochondrial proteins, which were attenuated by either pharmacological or genetic inhibition of AHR. We further demonstrated that improving mitochondrial function by inhibiting mPTP opening with Cyclosporin A suppressed the EOM-induced intracellular ROS and mtROS generation, MMP collapse, intrinsic apoptosis, and heart defects. Moreover, the EOM-induced mPTP opening was counteracted by inhibiting mtROS with mitoquinone mesylate (MitoQ). Supplementation with MitoQ also attenuated the EOM-induced mitochondrial dysfunction, apoptosis and heart defects. Additionally, knockdown of cyp1a1 but not cyp1b1 attenuated the EOM-induced mtROS generation and heart defects. Taken together, this study indicates that PM2.5 triggers mtROS generation via AHR-mediated cyp1a1 overexpression, which then causes mPTP opening and mitochondrial dysfunction, leading to apoptosis and heart defects.
Collapse
Affiliation(s)
- Jin Chen
- Suzhou Medical College of Soochow University, Suzhou, China; Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
| | - Mingxuan Zhang
- Suzhou Medical College of Soochow University, Suzhou, China; Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
| | - Hongmei Zou
- Suzhou Medical College of Soochow University, Suzhou, China; Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
| | - Stanley Aniagu
- Toxicology, Risk Assessment, and Research Division, Texas Commission on Environmental Quality, 12015 Park 35 Cir, Austin, TX, USA
| | - Yan Jiang
- Suzhou Medical College of Soochow University, Suzhou, China.
| | - Tao Chen
- Suzhou Medical College of Soochow University, Suzhou, China; Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China.
| |
Collapse
|
46
|
Shi S, Chen Y, Luo Z, Nie G, Dai Y. Role of oxidative stress and inflammation-related signaling pathways in doxorubicin-induced cardiomyopathy. Cell Commun Signal 2023; 21:61. [PMID: 36918950 PMCID: PMC10012797 DOI: 10.1186/s12964-023-01077-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 02/12/2023] [Indexed: 03/16/2023] Open
Abstract
Doxorubicin (DOX) is a powerful and commonly used chemotherapeutic drug, used alone or in combination in a variety of cancers, while it has been found to cause serious cardiac side effects in clinical application. More and more researchers are trying to explore the molecular mechanisms of DOX-induced cardiomyopathy (DIC), in which oxidative stress and inflammation are considered to play a significant role. This review summarizes signaling pathways related to oxidative stress and inflammation in DIC and compounds that exert cardioprotective effects by acting on relevant signaling pathways, including the role of Nrf2/Keap1/ARE, Sirt1/p66Shc, Sirt1/PPAR/PGC-1α signaling pathways and NOS, NOX, Fe2+ signaling in oxidative stress, as well as the role of NLRP3/caspase-1/GSDMD, HMGB1/TLR4/MAPKs/NF-κB, mTOR/TFEB/NF-κB pathways in DOX-induced inflammation. Hence, we attempt to explain the mechanisms of DIC in terms of oxidative stress and inflammation, and to provide a theoretical basis or new idea for further drug research on reducing DIC. Video Abstract.
Collapse
Affiliation(s)
- Saixian Shi
- Department of Pharmacy, Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Luzhou, 646000, Sichuan Province, China.,School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Ye Chen
- Department of Pharmacy, Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Luzhou, 646000, Sichuan Province, China.,School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Zhijian Luo
- Department of Ultrasound, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Guojun Nie
- The First Outpatient Department of People's Liberation Army Western Theater General Hospital, Chengdu, 610000, Sichuan Province, China
| | - Yan Dai
- Department of Pharmacy, Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Luzhou, 646000, Sichuan Province, China.
| |
Collapse
|
47
|
Shamseddine A, Patel SH, Chavez V, Moore ZR, Adnan M, Di Bona M, Li J, Dang CT, Ramanathan LV, Oeffinger KC, Liu JE, Steingart RM, Piersigilli A, Socci ND, Chan AT, Yu AF, Bakhoum SF, Schmitt AM. Innate immune signaling drives late cardiac toxicity following DNA-damaging cancer therapies. J Exp Med 2023; 220:213768. [PMID: 36534085 PMCID: PMC9767651 DOI: 10.1084/jem.20220809] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 09/15/2022] [Accepted: 11/22/2022] [Indexed: 12/24/2022] Open
Abstract
Late cardiac toxicity is a potentially lethal complication of cancer therapy, yet the pathogenic mechanism remains largely unknown, and few treatment options exist. Here we report DNA-damaging agents such as radiation and anthracycline chemotherapies inducing delayed cardiac inflammation following therapy due to activation of cGAS- and STING-dependent type I interferon signaling. Genetic ablation of cGAS-STING signaling in mice inhibits DNA damage-induced cardiac inflammation, rescues late cardiac functional decline, and prevents death from cardiac events. Treatment with a STING antagonist suppresses cardiac interferon signaling following DNA-damaging therapies and effectively mitigates cardiac toxicity. These results identify a therapeutically targetable, pathogenic mechanism for one of the most vexing treatment-related toxicities in cancer survivors.
Collapse
Affiliation(s)
- Achraf Shamseddine
- Division of Translational Oncology, Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Suchit H. Patel
- Division of Translational Oncology, Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Radiation Oncology, Mary Bird Perkins Cancer Center, Baton Rouge, LA, USA
| | - Valery Chavez
- Division of Translational Oncology, Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zachary R. Moore
- Division of Translational Oncology, Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mutayyaba Adnan
- Division of Translational Oncology, Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Melody Di Bona
- Division of Translational Oncology, Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jun Li
- Division of Translational Oncology, Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Chau T. Dang
- Breast Medicine Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lakshmi V. Ramanathan
- Clinical Chemistry Service, Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kevin C. Oeffinger
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Jennifer E. Liu
- Cardiology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Richard M. Steingart
- Cardiology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alessandra Piersigilli
- Laboratory of Comparative Pathology, Rockefeller University, Weill Cornell Medicine and Memorial Sloan-Kettering Cancer Center, New York, NY, USA
- Takeda Development Center Americas, Drug Safety Research Evaluation, Cambridge, MA, USA
| | - Nicholas D. Socci
- Marie-Josee & Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Angel T. Chan
- Cardiology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anthony F. Yu
- Cardiology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Samuel F. Bakhoum
- Division of Translational Oncology, Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Adam M. Schmitt
- Division of Translational Oncology, Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Correspondence to Adam M. Schmitt:
| |
Collapse
|
48
|
Nisa FY, Rahman MA, Rafi MKJ, Khan MAN, Sultana F, Majid M, Hossain MA, Deen JI, Mannan M, Saha S, Tangpong J, Choudhury TR. Biosynthesized magnesium oxide nanoparticles from Tamarindus indica seed attenuate doxorubicin-induced cardiotoxicity by regulating biochemical indexes and linked genes. BIOMATERIALS ADVANCES 2023; 146:213291. [PMID: 36709628 DOI: 10.1016/j.bioadv.2023.213291] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 06/09/2023]
Abstract
The phytochemicals of Tamarindus indica seed hydroalcoholic extract were exploited as a biocatalyst for the sustainable synthesis of magnesium oxide nanoparticles (MgO-NPs). This research investigated the cardioprotective effects of biosynthesized magnesium oxide nanoparticle (MgO-NPs). The biosynthesized seed MgO-NPs were characterized by ultraviolet-visible spectroscopy (UV-Vis), X-ray diffraction (XRD), scanning electron microscopy (SEM) with energy-dispersive X-ray diffraction (EDX), and Fourier-transform infrared spectroscopy (FT-IR). These methodological approaches demonstrated their capacity to synthesize crystalline and aggregated MgO-NPs with a size average of 13.38 ± 0.16 nm. The biogenic MgO-NPs were found to have a significant quantity of total phenolic contents (TPC) and total flavonoid contents (TFC), indicating the existence of phenol and flavonoid-like components. The biogenic MgO-NPs demonstrated a significant free radical scavenging effects compared to different standards as measured by the inhibition of free radicals produced in 2,2-diphenyl-1-picrylhydrazyl (DPPH), 2,2-azinobis-(3-ethylbenzothiazoline-6-sulfonate) (ABTS•+), and Nitric oxide (NO) scavenging methods; they also exhibited higher ferric ion reducing capacity in FRAP assay. Moreover, they were found to be non-toxic in cytotoxic assessment. Pretreatment of Wistar Albino rats with seed MgO-NPs resulted in a significant reduction of cardiac biomarkers, i.e., cardiac Troponin-I (cTnI), creatine kinase (CK-MB), and aspartate aminotransferase (AST). The seed MgO-NPs were more successful in reducing lipid levels. The results of the mRNA expression analysis showed that seed MgO-NPs efficiently reduced the expression of the apoptotic genes p53 and Caspase-3 while restoring the expected levels of SOD gene expression. The histopathological observations were primarily focused on the disruption of cardiac fibers and myofibrillar disintegration, which are consistent with the biochemical findings. Therefore, our research suggests that MgO-NPs derived from the seeds of Tamarindus indica as a powerful antioxidant; the administration may be effective in protecting the heart from DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Fatema Yasmin Nisa
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong 4331, Bangladesh
| | - Md Atiar Rahman
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong 4331, Bangladesh; School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand.
| | - Md Khalid Juhani Rafi
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong 4331, Bangladesh
| | - Md Asif Nadim Khan
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong 4331, Bangladesh
| | - Farjana Sultana
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong 4331, Bangladesh
| | - Mumtahina Majid
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong 4331, Bangladesh
| | - Md Altaf Hossain
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong 4331, Bangladesh
| | - Jobaier Ibne Deen
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong 4331, Bangladesh
| | - Md Mannan
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong 4331, Bangladesh
| | - Srabonti Saha
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong 4331, Bangladesh.
| | - Jitbanjong Tangpong
- School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand.
| | - Tasrina Rabia Choudhury
- Analytical Chemistry Laboratory, Chemistry Division, Atomic Energy Centre Dhaka, Bangladesh Atomic Energy Commission, Bangladesh
| |
Collapse
|
49
|
Doxorubicin-An Agent with Multiple Mechanisms of Anticancer Activity. Cells 2023; 12:cells12040659. [PMID: 36831326 PMCID: PMC9954613 DOI: 10.3390/cells12040659] [Citation(s) in RCA: 137] [Impact Index Per Article: 137.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/15/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
Doxorubicin (DOX) constitutes the major constituent of anti-cancer treatment regimens currently in clinical use. However, the precise mechanisms of DOX's action are not fully understood. Emerging evidence points to the pleiotropic anticancer activity of DOX, including its contribution to DNA damage, reactive oxygen species (ROS) production, apoptosis, senescence, autophagy, ferroptosis, and pyroptosis induction, as well as its immunomodulatory role. This review aims to collect information on the anticancer mechanisms of DOX as well as its influence on anti-tumor immune response, providing a rationale behind the importance of DOX in modern cancer therapy.
Collapse
|
50
|
Jiao Q, Liu B, Xu X, Huang T, Cao B, Wang L, Wang Q, Du A, Li J, Zhou B, Wang T. Biodegradable porous polymeric drug as a drug delivery system: alleviation of doxorubicin-induced cardiotoxicity via passive targeted release. RSC Adv 2023; 13:5444-5456. [PMID: 36793291 PMCID: PMC9923820 DOI: 10.1039/d2ra07410a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 02/04/2023] [Indexed: 02/15/2023] Open
Abstract
Doxorubicin (DOX) is an effective chemotherapeutic drug developed against a broad range of cancers, and its clinical applications are greatly restricted by the side effects of severe cardiotoxicity during tumour treatment. Herein, the DOX-loaded biodegradable porous polymeric drug, namely, Fc-Ma-DOX, which was stable in the circulation, but easy to compose in the acidic medium, was used as the drug delivery system avoiding the indiscriminate release of DOX. Fc-Ma was constructed via the copolymerization of 1,1'-ferrocenecarbaldehyde with d-mannitol (Ma) through the pH-sensitive acetal bonds. Echocardiography, biochemical parameters, pathological examination, and western blot results showed that DOX treatment caused increased myocardial injury and oxidative stress damage. In contrast, treatment with Fc-Ma-DOX significantly reduced myocardial injury and oxidative stress by DOX treatment. Notably, in the Fc-Ma-DOX treatment group, we observed a significant decrease in the uptake of DOX by H9C2 cells and a significant decrease in reactive oxygen species (ROS) production.
Collapse
Affiliation(s)
- Qiuhong Jiao
- Department of Cardiology, Affiliated Hospital of Weifang Medical University Weifang 261031 Shandong China
| | - Baoting Liu
- Department of Cardiology, Affiliated Hospital of Weifang Medical University Weifang 261031 Shandong China
| | - Xiufeng Xu
- Department of Geriatrics, Affiliated Hospital of Weifang Medical UniversityWeifang 261031ShandongChina
| | - Tao Huang
- Department of Cardiology, Affiliated Hospital of Weifang Medical University Weifang 261031 Shandong China
| | - Bufan Cao
- Department of Cardiology, Affiliated Hospital of Weifang Medical University Weifang 261031 Shandong China
| | - Lide Wang
- Department of Cardiology, Affiliated Hospital of Weifang Medical University Weifang 261031 Shandong China
| | - Qingguo Wang
- Department of Cardiology, Affiliated Hospital of Weifang Medical University Weifang 261031 Shandong China
| | - Ailing Du
- Department of Cardiology, Affiliated Hospital of Weifang Medical University Weifang 261031 Shandong China
| | - Jingtian Li
- Department of Cardiology, Affiliated Hospital of Weifang Medical University Weifang 261031 Shandong China
| | - Baolong Zhou
- School of Pharmacy, Weifang Medical University Weifang 261031 Shandong China
| | - Tao Wang
- Department of Cardiology, Affiliated Hospital of Weifang Medical University Weifang 261031 Shandong China
| |
Collapse
|