1
|
Tan N, Luo H, Li W, Ling G, Wei Y, Wang W, Wang Y. The dual function of autophagy in doxorubicin-induced cardiotoxicity: Mechanism and natural products. Semin Cancer Biol 2025; 109:83-90. [PMID: 39827930 DOI: 10.1016/j.semcancer.2025.01.004] [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: 12/08/2024] [Revised: 01/08/2025] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
Doxorubicin (DOX) is an anthracycline antitumor drug discovered in 1969, which can care for leukemia, breast cancer, lymphoma, and sarcoma. However, cardiotoxicity induced by DOX seriously limits its clinical value. The etiopathogenesis and therapeutic strategies are not unified. Autophagy is a critical mechanism in the progression of DOX-induced cardiotoxicity (DIC), autophagy intervention is a potential therapeutic strategy for DIC. Natural product has been considered as a complementary and alternative approach to treat cardiovascular disease. In this review, we summarize the pathology of autophagy in DIC and the natural products for DIC therapy.
Collapse
Affiliation(s)
- Nannan Tan
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; Anhui university of Chinese medicine, Hefei 230012, China
| | - Hanwen Luo
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Weili Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Guanjing Ling
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yan Wei
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Wei Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Beijing Key Laboratory of TCM Syndrome and Formula, Beijing University of Chinese Medicine, Beijing 100029, China; Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing 100029, China.
| | - Yong Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
2
|
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
|
3
|
Qi XM, Zhang WZ, Zuo YQ, Qiao YB, Zhang YL, Ren JH, Li QS. Nrf2/NRF1 signaling activation and crosstalk amplify mitochondrial biogenesis in the treatment of triptolide-induced cardiotoxicity using calycosin. Cell Biol Toxicol 2024; 41:2. [PMID: 39707073 PMCID: PMC11662064 DOI: 10.1007/s10565-024-09969-z] [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: 04/23/2024] [Accepted: 12/10/2024] [Indexed: 12/23/2024]
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) regulates both oxidative stress and mitochondrial biogenesis. Our previous study reported the cardioprotection of calycosin against triptolide toxicity through promoting mitochondrial biogenesis by activating nuclear respiratory factor 1 (NRF1), a coregulatory effect contributed by Nrf2 was not fully elucidated. This work aimed at investigating the involvement of Nrf2 in mitochondrial protection and elucidating Nrf2/NRF1 signaling crosstalk on amplifying the detoxification of calycosin. Results indicated that calycosin inhibited cardiomyocytes apoptosis and F-actin depolymerization following triptolide exposure. Cardiac contraction was improved by calycosin through increasing both fractional shortening (FS%) and ejection fraction (EF%). This enhanced contractile capacity of heart was benefited from mitochondrial protection reflected by ultrastructure improvement, augment in mitochondrial mass and ATP production. NRF1 overexpression in cardiomyocytes increased mitochondrial mass and DNA copy number, whereas NRF1 knockdown mitigated calycosin-mediated enhancement in mitochondrial mass. For nuclear Nrf2, it was upregulated by calycosin in a way of disrupting Nrf2-Keap1 (Kelch-like ECH associated protein 1) interaction, followed by inhibiting ubiquitination and degradation. The involvement of Nrf2 in mitochondrial protection was validated by the results that both Nrf2 knockdown and Nrf2 inhibitor blocked the calycosin effects on mitochondrial biogenesis and respiration. In the case of calycosin treatment, its effect on NRF1 and Nrf2 upregulations were respectively blocked by PGCα/Nrf2 and NRF1 knockdown, indicative of the mutual regulation between Nrf2 and NRF1. Accordingly, calycosin activated Nrf2/NRF1 and the signaling crosstalk, leading to mitochondrial biogenesis amplification, which would become a novel mechanism of calycosin against triptolide-induced cardiotoxicity.
Collapse
Affiliation(s)
- Xiao-Ming Qi
- Shanxi Key Laboratory of Innovative Drug for the Treatment of Serious Diseases Basing On the Chronic Inflammation, College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, Shanxi Province, China
| | - Wei-Zheng Zhang
- Shanxi Key Laboratory of Innovative Drug for the Treatment of Serious Diseases Basing On the Chronic Inflammation, College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, Shanxi Province, China
| | - Yu-Qin Zuo
- Shanxi Key Laboratory of Innovative Drug for the Treatment of Serious Diseases Basing On the Chronic Inflammation, College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, Shanxi Province, China
| | - Yuan-Biao Qiao
- Shanxi Key Laboratory of Innovative Drug for the Treatment of Serious Diseases Basing On the Chronic Inflammation, College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, Shanxi Province, China
| | - Yuan-Lin Zhang
- Shanxi Key Laboratory of Innovative Drug for the Treatment of Serious Diseases Basing On the Chronic Inflammation, College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, Shanxi Province, China
| | - Jin-Hong Ren
- Shanxi Key Laboratory of Innovative Drug for the Treatment of Serious Diseases Basing On the Chronic Inflammation, College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, Shanxi Province, China
| | - Qing-Shan Li
- Shanxi Key Laboratory of Innovative Drug for the Treatment of Serious Diseases Basing On the Chronic Inflammation, College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, Shanxi Province, China.
| |
Collapse
|
4
|
Han Q, Shi J, Yu Y, Yuan H, Guo Y, Liu X, Xue Y, Li Y. Calycosin alleviates ferroptosis and attenuates doxorubicin-induced myocardial injury via the Nrf2/SLC7A11/GPX4 signaling pathway. Front Pharmacol 2024; 15:1497733. [PMID: 39600362 PMCID: PMC11589554 DOI: 10.3389/fphar.2024.1497733] [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: 09/17/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
Background Heart failure is primarily characterized by damage to the structure and function of the heart. Ferroptosis represents a form of programmed cell death, and studies indicate that it constitutes one of the primary mechanisms underlying cardiomyocyte death in heart failure. Calycosin, a natural compound derived from astragalus, exhibits various pharmacological properties, including anti-ferroptosis, antioxidant effects, and cardiovascular protection. Nonetheless, the specific role of Calycosin in the treatment of ferroptosis in heart failure remains poorly understood. Objective This study aims to elucidate the regulatory effect of Calycosin on ferroptosis and its influence on the treatment mechanisms of heart failure through in vivo and in vitro experiments. Methods A rat model of heart failure was induced using doxorubicin, and the cardiac function was evaluated through cardiac ultrasound examination and NT-Pro BNP detection. Myocardial injury was assessed using H&E staining and Masson staining. The extent of mitochondrial damage was evaluated through transmission electron microscopy. Concurrently, the level of ferroptosis was analyzed by measuring ferroptosis markers, including MDA, ferrous ions, the GSH/GSSG ratio, and GPX4 activity. Subsequently, the molecular mechanism by which Calycosin exerts its therapeutic effects in heart failure was investigated through immunofluorescence and Western blotting. Finally, H9c2 cardiomyocytes were treated with doxorubicin to simulate myocardial injury, and the mechanism by which Calycosin mediates its effects in the treatment of heart failure was further verified through Nrf2 gene silencing. Results Calycosin significantly improves cardiac function in rats, reduces serum NT-Pro BNP levels, and alleviates myocardial cell damage. Additionally, it significantly decreases the levels of ferroptosis in myocardial tissue, as confirmed through transmission electron microscopy and the assessment of ferroptosis markers, including MDA, ferrous ions, GSH, and GPX4 activity. At the molecular level, Calycosin exerts its effects by activating the Nrf2/SLC7A11/GPX4 signaling pathway, evidenced by the upregulation of Nrf2, SLC7A11, GPX4, GSS, and GCL protein expression. This process substantially enhances the antioxidant capacity of rat myocardial tissue and effectively suppresses ferroptosis in myocardial cells. The results obtained from both in vivo and in vitro experiments are consistent. Notably, when Nrf2 is silenced, the protective effect of Calycosin on the myocardium is markedly diminished. Conclusion Calycosin effectively treats doxorubicin-induced cardiac injury, and its therapeutic effect is likely closely associated with the activation of the Nrf2/SLC7A11/GPX4 signaling pathway and the inhibition of ferroptosis in myocardial cells. Consequently, Calycosin, as a promising compound against doxorubicin-induced cardiotoxicity, warrants further investigation.
Collapse
Affiliation(s)
- Quancheng Han
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jingle Shi
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yiding Yu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Huajing Yuan
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yonghong Guo
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiujuan Liu
- Department of Cardiology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yitao Xue
- Department of Cardiology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yan Li
- Department of Cardiology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
5
|
Zhang Y, Chen Z, Chen L, Dong Q, Yang DH, Zhang Q, Zeng J, Wang Y, Liu X, Cui Y, Li M, Luo X, Zhou C, Ye M, Li L, He Y. Astragali radix (Huangqi): a time-honored nourishing herbal medicine. Chin Med 2024; 19:119. [PMID: 39215362 PMCID: PMC11363671 DOI: 10.1186/s13020-024-00977-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Astragali radix (AR, namded Huangqi in Chinese) is the dried root of Astragalus membranaceus (Fisch.) Bge. var. mongholicus (Bge.) Hsiao or Astragalus membranaceus (Fisch.) Bge. As a widely used ethnomedicine, the biological activities of AR include immunomodulatory, anti-hyperglycemic, anti-oxidant, anti-aging, anti-inflammatory, anti-viral, anti-tumor, cardioprotective, and anti-diabetic effects, with minimum side effects. Currently, it is known that polysaccharides, saponins, and flavonoids are the indispensable components of AR. In this review, we will elaborate the research advancements of AR on ethnobotany, ethnopharmacological practices, phytochemicals, pharmacological activities, clinical uses, quality control, production developments, and toxicology. The information is expected to assist clinicians and scientists in developing useful therapeutic medicines with minimal systemic side effects.
Collapse
Affiliation(s)
- Yuyu Zhang
- School of Food and Bioengineering, Xihua University, Chengdu, 610039, China
| | - Zhejie Chen
- Institute of Molecular Medicine (IMM), Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Liping Chen
- School of Comprehensive Health Management, Xihua University, Chengdu, 610039, China
| | - Qin Dong
- School of Food and Bioengineering, Xihua University, Chengdu, 610039, China
| | - Dong-Hua Yang
- New York College of Traditional Chinese Medicine, Mineola, NY, 11501, USA
| | - Qi Zhang
- Pengzhou Hospital of Traditional Chinese Medicine, Pengzhou, 611930, China
| | - Jing Zeng
- School of Food and Bioengineering, Xihua University, Chengdu, 610039, China
| | - Yang Wang
- School of Food and Bioengineering, Xihua University, Chengdu, 610039, China
| | - Xiao Liu
- School of Food and Bioengineering, Xihua University, Chengdu, 610039, China
| | - Yuan Cui
- School of Food and Bioengineering, Xihua University, Chengdu, 610039, China
| | - Minglong Li
- School of Food and Bioengineering, Xihua University, Chengdu, 610039, China
| | - Xiao Luo
- Chengdu Institute for Drug Control, NMPA Key Laboratory for Quality Monitoring and Evaluation of Traditional Chinese Medicine, Chengdu, 610045, China
| | - Chongjian Zhou
- HuBei Guizhenyuan Chinese Herbal Medicine Co.Ltd., Hong'an, 438400, China
| | - Mingzhu Ye
- School of Food and Bioengineering, Xihua University, Chengdu, 610039, China
| | - Ling Li
- School of Food and Bioengineering, Xihua University, Chengdu, 610039, China.
- Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China.
| | - Yuxin He
- School of Food and Bioengineering, Xihua University, Chengdu, 610039, China.
| |
Collapse
|
6
|
L’Abbate S, Kusmic C. The Protective Effect of Flavonoids in the Diet on Autophagy-Related Cardiac Impairment. Nutrients 2024; 16:2207. [PMID: 39064651 PMCID: PMC11279826 DOI: 10.3390/nu16142207] [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: 06/25/2024] [Revised: 07/07/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
The compounds known as flavonoids, commonly found in fruits, vegetables, legumes, medicinal herbs, chocolate, and coffee and tea beverages, have been extensively researched for their impact on cardiovascular health. Flavonoids, with their demonstrated potential, have shown promising effects in regulating blood vessel function and apoptotic processes, as well as in improving lipid profiles. While their powerful antioxidant properties were initially thought to be the main reason behind these effects, recent studies have uncovered new insights into the positive effects of flavonoids on cardiovascular health, and researchers have now identified several signaling pathways and mechanisms that also play a role. Of particular interest are the studies that have highlighted the role of autophagy in maintaining the physiological functions of cardiomyocytes and protecting them from harm. Recent publications have linked the dysregulation of autophagic processes with the development of cardiomyopathies, heart failure, and other cardiovascular diseases. This review aims to present the latest, novel findings from preclinical research regarding the potential beneficial effects of flavonoids on various heart conditions associated with altered autophagy processes.
Collapse
Affiliation(s)
| | - Claudia Kusmic
- Istituto di Fisiologia Clinica, Consiglio Nazionale delle Ricerche (CNR), 56124 Pisa, Italy;
| |
Collapse
|
7
|
Huang Y, Gu W, Qin Z, Jin Y. Bromuconazole exposure induces cardiac dysfunction by upregulating the expression LEF1. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 933:173113. [PMID: 38735319 DOI: 10.1016/j.scitotenv.2024.173113] [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: 01/26/2024] [Revised: 04/18/2024] [Accepted: 05/08/2024] [Indexed: 05/14/2024]
Abstract
With the wide application of bromuconazole (BRO), a kind of triazole fungicide, the environmental problems caused by BRO have been paid more and more attention. In this study, adult male zebrafish were exposed to environmental related concentration and the maximum non-lethal concentration for zebrafish larvae (0,50 ng/L and 7.5 mg/L) for 7 days, respectively. Zebrafish exposed to BRO exhibited a significant reduction in body length and an increase in fatness index, indicating adverse physiological changes. Notably, the exposed zebrafish showed enlarged heart ventricular volumes and thinner heart walls. Transcriptome analysis of heart samples showed that BRO exposure mainly affected pathways related to cardiac energy metabolism. In addition, the amount of ATP in the heart tissue was correspondingly reduced, and the expression levels of genes related to controlling ion balance and myosin synthesis in the heart were also altered. The study extended its findings to the rat cardiomyocytes (H9C2), where similar cardiotoxic effects including changes in transcription of genes related to energy metabolism and heart function were also observed, suggesting a potential universal mechanism of BRO-induced cardiotoxicity. In a doxorubicin (DOX) induced larval zebrafish heart failure model, the expression of lymphoid enhancer-binding factor 1(LEF1), a key gene in the Wnt/β-catenin signaling pathway, was significantly increased in larval zebrafish and adult fish heart tissues and cardiomyocytes, suggesting that LEF1 might play an important role in BRO-induced cardiotoxicity. Taken together, BRO exposure could interfere with cardiac function and metabolic capacity by abnormal activation the expression of LEF1. The study emphasized the urgent need for monitoring and regulating BRO due to its harmful effects on the hearts of aquatic organisms.
Collapse
Affiliation(s)
- Yilin Huang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Weijie Gu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Zhen Qin
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Yuanxiang Jin
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China.
| |
Collapse
|
8
|
Lai PF, Mahendran R, Tsai BCK, Lu CY, Kuo CH, Lin KH, Lu SY, Wu YL, Chang YM, Kuo WW, Huang CY. Calycosin Enhances Heat Shock Related-Proteins in H9c2 Cells to Modulate Survival and Apoptosis against Heat Shock. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:1173-1193. [PMID: 38938156 DOI: 10.1142/s0192415x24500472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
Abstract
Heat shock proteins (HSPs), which function as chaperones, are activated in response to various environmental stressors. In addition to their role in diverse aspects of protein production, HSPs protect against harmful protein-related stressors. Calycosin exhibits numerous beneficial properties. This study aims to explore the protective effects of calycosin in the heart under heat shock and determine its underlying mechanism. H9c2 cells, western blot, TUNEL staining, flow cytometry, and immunofluorescence staining were used. The time-dependent effects of heat shock analyzed using western blot revealed increased HSP expression for up to 2[Formula: see text]h, followed by protein degradation after 4[Formula: see text]h. Hence, a heat shock damage duration of 4[Formula: see text]h was chosen for subsequent investigations. Calycosin administered post-heat shock demonstrated dose-dependent recovery of cell viability. Under heat shock conditions, calycosin prevented the apoptosis of H9c2 cells by upregulating HSPs, suppressing p-JNK, enhancing Bcl-2 activation, and inhibiting cleaved caspase 3. Calycosin also inhibited Fas/FasL expression and activated cell survival markers (p-PI3K, p-ERK, p-Akt), indicating their cytoprotective properties through PI3K/Akt activation and JNK inhibition. TUNEL staining and flow cytometry confirmed that calycosin reduced apoptosis. Moreover, calycosin reversed the inhibitory effects of quercetin on HSF1 and Hsp70 expression, illustrating its role in enhancing Hsp70 expression through HSF1 activation during heat shock. Immunofluorescence staining demonstrated HSF1 translocation to the nucleus following calycosin treatment, emphasizing its cytoprotective effects. In conclusion, calycosin exhibits pronounced protective effects against heat shock-induced damages by modulating HSP expression and regulating key signaling pathways to promote cell survival in H9c2 cells.
Collapse
Affiliation(s)
- Pei-Fang Lai
- Department of Emergency Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan
- Department of Medicine, Tzu Chi University, Hualien 970, Taiwan
| | - Ramasamy Mahendran
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan
| | - Bruce Chi-Kang Tsai
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan
| | - Cheng-You Lu
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 402, Taiwan
| | - Chia-Hua Kuo
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei 111, Taiwan
- Institute of Sports Sciences, University of Taipei, Taipei 111, Taiwan
- School of Physical Education and Sports Science, Soochow University, Suzhou 215021, China
- Department of Kinesiology and Health, College of William and Mary, Williamsburg, VA 23185, USA
| | - Kuan-Ho Lin
- Department of Emergency Medicine, China Medical University Hospital, Taichung 404, Taiwan
- College of Medicine, China Medical University, Taichung 406, Taiwan
| | - Shang-Yeh Lu
- College of Medicine, China Medical University, Taichung 406, Taiwan
- Division of Cardiovascular Medicine, Department of Internal, Medicine China Medical University Hospital, Taichung 404, Taiwan
| | - Yu-Ling Wu
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan
| | - Yung-Ming Chang
- The School of Chinese Medicine for Post-Baccalaureate, I-Shou University, 840, Kaohsiung, Taiwan
- Chinese Medicine Department, E-DA Hospital, Kaohsiung, 824, Taiwan
- 1PT Biotechnology Co., Ltd., Taichung 433, Taiwan
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, College of Life Sciences, China Medical University, Taichung 406, Taiwan
- Ph.D. Program for Biotechnology Industry, China Medical University, Taichung 406, Taiwan
- School of Pharmacy, China Medical University, Taichung 406, Taiwan
| | - Chih-Yang Huang
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 406, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung 413, Taiwan
- Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien 970, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404, Taiwan
| |
Collapse
|
9
|
van Doorn ECH, Amesz JH, Sadeghi AH, de Groot NMS, Manintveld OC, Taverne YJHJ. Preclinical Models of Cardiac Disease: A Comprehensive Overview for Clinical Scientists. Cardiovasc Eng Technol 2024; 15:232-249. [PMID: 38228811 PMCID: PMC11116217 DOI: 10.1007/s13239-023-00707-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] [Received: 03/02/2023] [Accepted: 12/19/2023] [Indexed: 01/18/2024]
Abstract
For recent decades, cardiac diseases have been the leading cause of death and morbidity worldwide. Despite significant achievements in their management, profound understanding of disease progression is limited. The lack of biologically relevant and robust preclinical disease models that truly grasp the molecular underpinnings of cardiac disease and its pathophysiology attributes to this stagnation, as well as the insufficiency of platforms that effectively explore novel therapeutic avenues. The area of fundamental and translational cardiac research has therefore gained wide interest of scientists in the clinical field, while the landscape has rapidly evolved towards an elaborate array of research modalities, characterized by diverse and distinctive traits. As a consequence, current literature lacks an intelligible and complete overview aimed at clinical scientists that focuses on selecting the optimal platform for translational research questions. In this review, we present an elaborate overview of current in vitro, ex vivo, in vivo and in silico platforms that model cardiac health and disease, delineating their main benefits and drawbacks, innovative prospects, and foremost fields of application in the scope of clinical research incentives.
Collapse
Affiliation(s)
- Elisa C H van Doorn
- Translational Cardiothoracic Surgery Research Lab, Department of Cardiothoracic Surgery, Erasmus Medical Center, Rotterdam, The Netherlands
- Translational Electrophysiology Laboratory, Department of Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Jorik H Amesz
- Translational Cardiothoracic Surgery Research Lab, Department of Cardiothoracic Surgery, Erasmus Medical Center, Rotterdam, The Netherlands
- Translational Electrophysiology Laboratory, Department of Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Amir H Sadeghi
- Translational Cardiothoracic Surgery Research Lab, Department of Cardiothoracic Surgery, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Natasja M S de Groot
- Translational Electrophysiology Laboratory, Department of Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Yannick J H J Taverne
- Translational Cardiothoracic Surgery Research Lab, Department of Cardiothoracic Surgery, Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
10
|
Zhu P, Ren Q, Zhang R, Zhang L, Xia X, Zheng C, Ye T. Exploring the effects of calycosin on anthracycline-induced cardiotoxicity: a network pharmacology, molecular docking, and experimental study. Front Cardiovasc Med 2024; 11:1286620. [PMID: 38576421 PMCID: PMC10991710 DOI: 10.3389/fcvm.2024.1286620] [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: 08/31/2023] [Accepted: 03/11/2024] [Indexed: 04/06/2024] Open
Abstract
Background Chemotherapy with anthracyclines can cause cardiotoxicity, possibly leading to stopping treatment in some cancer patients. In cardio-oncology research, preventing and minimizing anthracycline-induced cardiotoxicity (AIC) is a hot issue. For the treatment of AIC, calycosin (CA), an isoflavone component in astragali radix (AR), has become a research focus. However, the elaborate mechanisms of calycosin treating AIC remain to be unrevealed. Aim of the study To explore the effects of CA on AIC through multiple dimensions concerning network pharmacology, molecular docking, and experimental evaluations. Methods The study evaluated calycosin's potential targets and mechanisms for treating AIC using network pharmacology and molecular docking. The candidate genes/targets of CA and AIC were screened using the online-available database. Protein-protein interactions (PPI) between the common targets were constructed using the STRING platform, and the results were then visualized using Cytoscape. Molecular docking was used to evaluate the strength of the binding force between CA and the common targets. The possible pharmacological mechanisms of CA were explained by pathway enrichment and GSEA. Subsequently, the candidate targets were identified in vitro experiments. Results Network pharmacology effectively discovered the CA's multitarget intervention in AIC, including TNF, ABCC1, TOP2A, ABCB1, and XDH. CA binds to the ATP-binding cassette subfamily B member 1(ABCB1) had the highest binding energy (-7.5 kcal/mol) according to the molecular docking analysis and was selected and visualized for subsequent analysis. In vitro experiments showed that ABCB1 exhibited significant time-curve changes under different doses of doxorubicin (DOX) compared with DMSO control experiments. The anti-AIC pharmacological mechanism of CA were revealed by highlighting the biological processes of oxidative stress (OR) and inflammation. Conclusions We employed a practicable bioinformatics method to connect network and molecular docking to determine the calycosin's therapeutic mechanism against AIC and identified some bioinformatics results in in vitro experiments. The results presented show that CA may represent an encouraging treatment for AIC.
Collapse
Affiliation(s)
- Peng Zhu
- Department of Hepatobiliary Surgery, Wuhan No.1 Hospital, Wuhan, China
| | - Qianqian Ren
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Ruizhi Zhang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Licai Zhang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Xiangwen Xia
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Chuansheng Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Tianhe Ye
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| |
Collapse
|
11
|
Zhou Z, Li Y, Jiang W, Wang Z. Molecular Mechanism of Calycosin Inhibited Vascular Calcification. Nutrients 2023; 16:99. [PMID: 38201929 PMCID: PMC10781010 DOI: 10.3390/nu16010099] [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: 10/25/2023] [Revised: 12/07/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
Vascular calcification (VC) is a pathological condition frequently observed in cardiovascular diseases. Primary factors contributing to VC are osteogenic differentiation of vascular smooth muscle and hydroxyapatite deposition. Targeted autophagy (a lysosome-mediated mechanism for degradation/recycling of unnecessary cellular components) is a useful approach for inhibiting VC and promoting vascular cell health. Calycosin has been shown to alleviate atherosclerosis by enhancing macrophage autophagy, but its therapeutic effect on VC has not been demonstrated. Using an in vitro model (rat thoracic aortic smooth muscle cell line A7r5), we demonstrated effective inhibition of VC using calycosin (the primary flavonoid component of astragalus), based on the enhancement of autophagic flux. Calycosin treatment activated AMPK/mTOR signaling to induce initiation of autophagy and restored mTORC1-dependent autophagosome-lysosome fusion in late-stage autophagy by promoting soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex formation, thereby preventing stoppage of autophagy in calcified cells. Calycosin substantially reduced degrees of both osteogenic differentiation and calcium deposition in our VC cell model by enhancing autophagy. The present findings clarify the mechanism whereby calycosin mitigates autophagy stoppage in calcified smooth muscle cells and provide a basis for effective VC treatment via autophagy enhancement.
Collapse
Affiliation(s)
- Zekun Zhou
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (Z.Z.); (Y.L.)
| | - Yi Li
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (Z.Z.); (Y.L.)
| | - Wei Jiang
- College of Biological Sciences, China Agricultural University, Beijing 100193, China;
| | - Zengli Wang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (Z.Z.); (Y.L.)
| |
Collapse
|
12
|
Li X. Doxorubicin-mediated cardiac dysfunction: Revisiting molecular interactions, pharmacological compounds and (nano)theranostic platforms. ENVIRONMENTAL RESEARCH 2023; 234:116504. [PMID: 37356521 DOI: 10.1016/j.envres.2023.116504] [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: 04/22/2023] [Revised: 06/17/2023] [Accepted: 06/23/2023] [Indexed: 06/27/2023]
Abstract
Although chemotherapy drugs are extensively utilized in cancer therapy, their administration for treatment of patients has faced problems that regardless of chemoresistance, increasing evidence has shown concentration-related toxicity of drugs. Doxorubicin (DOX) is a drug used in treatment of solid and hematological tumors, and its function is based on topoisomerase suppression to impair cancer progression. However, DOX can also affect the other organs of body and after chemotherapy, life quality of cancer patients decreases due to the side effects. Heart is one of the vital organs of body that is significantly affected by DOX during cancer chemotherapy, and this can lead to cardiac dysfunction and predispose to development of cardiovascular diseases and atherosclerosis, among others. The exposure to DOX can stimulate apoptosis and sometimes, pro-survival autophagy stimulation can ameliorate this condition. Moreover, DOX-mediated ferroptosis impairs proper function of heart and by increasing oxidative stress and inflammation, DOX causes cardiac dysfunction. The function of DOX in mediating cardiac toxicity is mediated by several pathways that some of them demonstrate protective function including Nrf2. Therefore, if expression level of such protective mechanisms increases, they can alleviate DOX-mediated cardiac toxicity. For this purpose, pharmacological compounds and therapeutic drugs in preventing DOX-mediated cardiotoxicity have been utilized and they can reduce side effects of DOX to prevent development of cardiovascular diseases in patients underwent chemotherapy. Furthermore, (nano)platforms are used comprehensively in treatment of cardiovascular diseases and using them for DOX delivery can reduce side effects by decreasing concentration of drug. Moreover, when DOX is loaded on nanoparticles, it is delivered into cells in a targeted way and its accumulation in healthy organs is prevented to diminish its adverse impacts. Hence, current paper provides a comprehensive discussion of DOX-mediated toxicity and subsequent alleviation by drugs and nanotherapeutics in treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Xiaofeng Li
- Department of Emergency, Shanghai Tenth People's Hospital, School of Medicine Tongji University, Shanghai, 200072, China.
| |
Collapse
|
13
|
He Y, Xi J, Fang J, Zhang B, Cai W. Aloe-emodin alleviates doxorubicin-induced cardiotoxicity via inhibition of ferroptosis. Free Radic Biol Med 2023; 206:13-21. [PMID: 37364691 DOI: 10.1016/j.freeradbiomed.2023.06.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/22/2023] [Accepted: 06/23/2023] [Indexed: 06/28/2023]
Abstract
Aloe-emodin (AE), a novel ferroptosis inhibitor, alleviates the doxorubicin (DOX)-induced cardiotoxicity in H9c2 rat cardiomyocytes. The inhibition of ferroptosis and the protective effect against cardiotoxicity were evaluated via MTT assay in H9c2 cells. The molecular mechanism of action (MOA) of nuclear factor erythroid 2-related factor 2 (Nrf2) activation, including transactivation of multiple downstream cytoprotective genes, were further assessed by Western blot, luciferase reporter assay and qRT-PCR analyses. Fluorescent imaging was performed to detect the change of intracellular reactive oxygen species, mitochondrial membrane potential and lipid peroxidation. In addition, an infrared spectroscopy was employed to detect the AE-Fe (II) complex. AE, alleviates oxidative stress in DOX-induced H9c2 cells by activating Nrf2 and increasing the expression of Nrf2 downstream antioxidant genes, SLC7A11 and GPX4. Furthermore, AE complexes bivalent iron and regulates the intracellular iron-related genes. In conclusion, the discovery of AE as a novel ferroptosis inhibitor and its MOA provides a new perspective for further exploration of cardio-protective agents in cancer patients during chemotherapy.
Collapse
Affiliation(s)
- Ying He
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Junmin Xi
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Jianguo Fang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, 730000, China; School of Chemistry and Chemical Engineering, Nanjing University of Science & Technology, Nanjing, Jiangsu, 210094, China
| | - Baoxin Zhang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, 730000, China.
| | - Wenqing Cai
- Regor Therapeutics Inc,1206 Zhangjiang Road, Building C, Pu Dong New District, Shanghai, 201210, China.
| |
Collapse
|
14
|
Wang P, Wang Z, Zhang Z, Cao H, Kong L, Ma W, Ren W. A review of the botany, phytochemistry, traditional uses, pharmacology, toxicology, and quality control of the Astragalus memeranaceus. Front Pharmacol 2023; 14:1242318. [PMID: 37680711 PMCID: PMC10482111 DOI: 10.3389/fphar.2023.1242318] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/07/2023] [Indexed: 09/09/2023] Open
Abstract
Astragali Radix (Huangqi) is mainly distributed in the Northern Hemisphere, South America, and Africa and rarely in North America and Oceania. It has long been used as an ethnomedicine in the Russian Federation, Mongolia, Korea, Kazakhstan, and China. It was first recorded in the Shennong Ben Cao Jing and includes the effects of reinforcing healthy qi, dispelling pathogenic factors, promoting diuresis, reducing swelling, activating blood circulation, and dredging collaterals. This review systematically summarizes the botanical characteristics, phytochemistry, traditional uses, pharmacology, and toxicology of Astragalus to explore the potential of Huangqi and expand its applications. Data were obtained from databases such as PubMed, CNKI, Wan Fang Data, Baidu Scholar, and Google Scholar. The collected material also includes classic works of Chinese herbal medicine, Chinese Pharmacopoeia, Chinese Medicine Dictionary, and PhD and Master's theses. The pharmacological effects of the isoflavone fraction in Huangqi have been studied extensively; The pharmacological effects of Huangqi isoflavone are mainly reflected in its anti-inflammatory, anti-tumor, anti-oxidant, anti-allergic, and anti-diabetic properties and its ability to treat several related diseases. Additionally, the medicinal uses, chemical composition, pharmacological activity, toxicology, and quality control of Huangqi require further elucidation. Here, we provide a comprehensive review of the botany, phytochemistry, traditional uses, pharmacology, toxicology, and quality control of Astragalus to assist future innovative research and to identify and develop new drugs involving Huangqi.
Collapse
Affiliation(s)
| | | | | | | | | | - Wei Ma
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Weichao Ren
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
15
|
Ma T, Yang L, Zhang B, Lv X, Gong F, Yang W. Hydrogen inhalation enhances autophagy via the AMPK/mTOR pathway, thereby attenuating doxorubicin-induced cardiac injury. Int Immunopharmacol 2023; 119:110071. [PMID: 37080067 DOI: 10.1016/j.intimp.2023.110071] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 03/04/2023] [Accepted: 03/20/2023] [Indexed: 04/22/2023]
Abstract
AIMS Doxorubicin is a drug widely used in clinical cancer treatment, but severe cardiotoxicity limits its clinical application. Autophagy disorder is an important factor in the mechanism of doxorubicin-induced cardiac injury. As the smallest molecule in nature, hydrogen has various biological effects such as anti-oxidation, anti-apoptosis and regulation of autophagy. Hydrogen therapy is currently considered to be an emerging therapeutic method, but the effect and mechanism of hydrogen on doxorubicin-induced myocardial injury have not been determined. The purpose of this study was to investigate the protective effect of hydrogen inhalation on doxorubicin-induced chronic myocardial injury and its effect and mechanism on autophagy. METHODS In this study, we established a chronic heart injury model by intraperitoneal injection of doxorubicin in rats for 30 days, accumulating 20 mg/kg. The effect of hydrogen inhalation on the cardiac function in rats was explored by echocardiography, Elisa, and H&E staining. To clarify the influence of autophagy, we detected the expression of LC3 and related autophagy proteins in vivo and in vitro by immunofluorescence and western blot.In order to further explore the mechanism of autophagy, we added pathway inhibitors and used western blot to preliminarily investigate the protective effect of hydrogen inhalation on myocardial injury caused by doxorubicin. RESULTS Hydrogen inhalation can improve doxorubicin-induced cardiac function decline and pathological structural abnormalities in rats. It was confirmed by immunofluorescence that hydrogen treatment could restore the expression of autophagy marker protein LC3 (microtubule-associated protein 1 light chain 3) in cardiomyocytes reduced by doxorubicin, while reducing cardiomyocyte apoptosis. Mechanistically, Western blot results consistently showed that hydrogen treatment up-regulated the ratio of p-AMPK (phosphorylated AMP-dependent protein kinase) to AMPK and down-regulated p-mTOR (phosphorylated mammalian target of rapamycin) and mTOR ratio. CONCLUSIONS These results suggest that hydrogen inhalation can activate autophagy through the AMPK/mTOR pathway and protect against myocardial injury induced by doxorubicin. Hydrogen inhalation therapy may be a potential treatment for doxorubicin-induced myocardial injury.
Collapse
Affiliation(s)
- Tianjiao Ma
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150000, China
| | - Lei Yang
- Department of Urinary Surgery, The First Hospital of Harbin, Harbin 150010, China
| | - Binmei Zhang
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150000, China
| | - Xin Lv
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, 150001, China
| | - Feifei Gong
- Department of Imaging, Chest Hospital of Harbin, 150056, China
| | - Wei Yang
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150000, China.
| |
Collapse
|
16
|
Antar SA, Abd-Elsalam M, Abdo W, Abdeen A, Abdo M, Fericean L, Raslan NA, Ibrahim SF, Sharif AF, Elalfy A, Nasr HE, Zaid AB, Atia R, Atwa AM, Gebba MA, Alzokaky AA. Modulatory Role of Autophagy in Metformin Therapeutic Activity toward Doxorubicin-Induced Nephrotoxicity. TOXICS 2023; 11:273. [PMID: 36977038 PMCID: PMC10052439 DOI: 10.3390/toxics11030273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/12/2023] [Accepted: 03/15/2023] [Indexed: 06/18/2023]
Abstract
Doxorubicin (DOX) is a frequent chemotherapeutic drug used to treat various malignant tumors. One of the key factors that diminish its therapeutic importance is DOX-induced nephrotoxicity. The first-line oral antidiabetic drug is metformin (Met), which also has antioxidant properties. The purpose of our study was to investigate the underlying molecular mechanisms for the potential protective effects of Met on DOX-triggered nephrotoxicity. Four animal groups were assigned as follows; animals received vehicle (control group), 200 mg/kg Met (Met group), DOX 15 mg/kg DOX (DOX group), and a combination of DOX and Met (DOX/Met group). Our results demonstrated that DOX administration caused marked histological alterations of widespread inflammation and tubular degeneration. Notably, the DOX-induced dramatic up-regulation of the nuclear factor-kappa B/P65 (NF-κB/P65), microtubule-associated protein light chain 3B (LC3B), neutrophil gelatinase-associated lipocalin (NGAL), interleukin-1beta (IL-1β), 8-hydroxy-2' -deoxyguanosine (8-OHdG), and Beclin-1 in renal tissue. A marked increase in the malondialdehyde (MDA) tissue level and a decrease in the total antioxidant capacity (TAC) were also recorded in DOX-exposed animals. Interestingly, Met could minimize all histopathological changes as well as the disruptions caused by DOX in the aforementioned measures. Thus, Met provided a workable method for suppressing the nephrotoxicity that occurred during the DOX regimen via the deactivation of the Beclin-1/LC3B pathway.
Collapse
Affiliation(s)
- Samar A. Antar
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta 34518, Egypt
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute, Virginia Tech, Roanoke, VA 24016, USA
| | - Marwa Abd-Elsalam
- Department of Histology, Faculty of Medicine, Kafrelsheikh University, Kafr El-Sheikh 33516, Egypt
| | - Walied Abdo
- Department of Pathology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Sheikh 33516, Egypt
| | - Ahmed Abdeen
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Benha University, Toukh 13736, Egypt
| | - Mohamed Abdo
- Department of Animal Histology and Anatomy, School of Veterinary Medicine, Badr University in Cairo (BUC), Badr City 32897, Egypt
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, University of Sadat City, Sadat City 32897, Egypt
| | - Liana Fericean
- Department of Biology and Plant Protection, Faculty of Agriculture, University of Life Sciences “King Michael I” from Timișoara, Calea Aradului 119, CUI 3487181, 300645 Timisoara, Romania
| | - Nahed A. Raslan
- Clinical Pharmacy Program, College of Health Sciences and Nursing, Al-Rayan Colleges, Medina 42541, Saudi Arabia
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo 11651, Egypt
| | - Samah F. Ibrahim
- Department of Clinical Sciences, College of Medicine, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Asmaa F. Sharif
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Tanta University, Tanta 31111, Egypt
| | - Amira Elalfy
- Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Benha 13518, Egypt
| | - Hend E. Nasr
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Benha University, Benha 13518, Egypt
| | - Ahmed B. Zaid
- Department of Clinical Pathology, National Liver Institute, Menoufia University, Shibin Elkom 32511, Egypt
| | - Rania Atia
- Department of Physiology, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
- Department of Basic Medical Science, Faculty of Applied Medical Science, Al-Baha University, Al-Baha 65779, Saudi Arabia
| | - Ahmed M. Atwa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo 11829, Egypt
| | - Mohammed A. Gebba
- Department of Anatomy and Embryology, Faculty of Medicine, Benha University, Benha 13518, Egypt
| | - Amany A. Alzokaky
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta 34518, Egypt
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo 11651, Egypt
| |
Collapse
|
17
|
Wu J, Li Y, He Q, Yang X. Exploration of the Use of Natural Compounds in Combination with Chemotherapy Drugs for Tumor Treatment. Molecules 2023; 28:molecules28031022. [PMID: 36770689 PMCID: PMC9920618 DOI: 10.3390/molecules28031022] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
Currently, chemotherapy is the main treatment for tumors, but there are still problems such as unsatisfactory chemotherapy results, susceptibility to drug resistance, and serious adverse effects. Natural compounds have numerous pharmacological activities which are important sources of drug discovery for tumor treatment. The combination of chemotherapeutic drugs and natural compounds is gradually becoming an important strategy and development direction for tumor treatment. In this paper, we described the role of natural compounds in combination with chemotherapeutic drugs in synergizing, reducing drug resistance, mitigating adverse effects and related mechanisms, and providing new insights for future oncology research.
Collapse
Affiliation(s)
- Jianping Wu
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yunheng Li
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qiaojun He
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Shandong (Linyi) Institute of Modern Agriculture, Zhejiang University, Linyi 276000, China
| | - Xiaochun Yang
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Shandong (Linyi) Institute of Modern Agriculture, Zhejiang University, Linyi 276000, China
- Correspondence: ; Tel.: +86-571-8820-8076
| |
Collapse
|
18
|
Rao Y, Wang Y, Lin Z, Zhang X, Ding X, Yang Y, Liu Z, Zhang B. Comparative efficacy and pharmacological mechanism of Chinese patent medicines against anthracycline-induced cardiotoxicity: An integrated study of network meta-analysis and network pharmacology approach. Front Cardiovasc Med 2023; 10:1126110. [PMID: 37168657 PMCID: PMC10164985 DOI: 10.3389/fcvm.2023.1126110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 04/04/2023] [Indexed: 05/13/2023] Open
Abstract
Background This study aimed to evaluate the efficacy of Chinese patent medicines (CPMs) combined with dexrazoxane (DEX) against anthracycline-induced cardiotoxicity (AIC) and further explore their pharmacological mechanism by integrating the network meta-analysis (NMA) and network pharmacology approach. Methods We searched for clinical trials on the efficacy of DEX + CPMs for AIC until March 10, 2023 (Database: PubMed, Embase, Cochrane Library, Chinese National Knowledge Infrastructure, China Science and Technology Journal and China Online Journals). The evaluating outcomes were cardiac troponin I (cTnI) level, creatine kinase MB (CK-MB) level, left ventricular ejection fraction (LVEF) value, and electrocardiogram (ECG) abnormal rate. Subsequently, the results of NMA were further analyzed in combination with network pharmacology. Results We included 14 randomized controlled trials (RCTs) and 1 retrospective cohort study (n = 1,214), containing six CPMs: Wenxinkeli (WXKL), Cinobufotalin injection (CI), Shenqifuzheng injection (SQFZ), Shenmai injection (SM), Astragalus injection (AI) and AI + CI. The NMA was implemented in Stata (16.0) using the mvmeta package. Compared with using DEX only, DEX + SM displayed the best effective for lowering cTnI level (MD = -0.44, 95%CI [-0.56, -0.33], SUCRA 93.4%) and improving LVEF value (MD = 14.64, 95%CI [9.36, 19.91], SUCRA 98.4%). DEX + SQFZ showed the most effectiveness for lowering CK-MB level (MD = -11.57, 95%CI [-15.79, -7.35], SUCRA 97.3%). And DEX + AI + CI has the highest effectiveness for alleviating ECG abnormalities (MD = -2.51, 95%CI [-4.06, -0.96], SUCRA 96.8%). So that we recommended SM + DEX, SQFZ + DEX, and DEX + AI + CI as the top three effective interventions against AIC. Then, we explored their pharmacological mechanism respectively. The CPMs' active components and AIC-related targets were screened to construct the component-target network. The potential pathways related to CPMs against AIC were determined by KEGG. For SM, we identified 118 co-targeted genes of active components and AIC, which were significantly enriched in pathways of cancer pathways, EGFR tyrosine kinase inhibitor resistance and AGE-RAGE signaling pathway in diabetic complications. For SQFZ, 41 co-targeted genes involving pathways of microRNAs in cancer, Rap1 signaling pathway, MAPK signaling pathway, and lipid and atherosclerosis. As for AI + CI, 224 co-targeted genes were obtained, and KEGG analysis showed that the calcium signaling pathway plays an important role except for the consistent pathways of SM and SQFZ in anti-AIC. Conclusions DEX + CPMs might be positive efficacious interventions from which patients with AIC will derive benefits. DEX + SM, DEX + SQFZ, and DEX + AI + CI might be the preferred intervention for improving LVEF value, CK-MB level, and ECG abnormalities, respectively. And these CPMs play different advantages in alleviating AIC by targeting multiple biological processes.
Collapse
Affiliation(s)
- Yifei Rao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yu Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhijian Lin
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Center for Pharmacovigilance and Rational Use of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaomeng Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Center for Pharmacovigilance and Rational Use of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xueli Ding
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Ying Yang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zeyu Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Bing Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Center for Pharmacovigilance and Rational Use of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- Correspondence: Bing Zhang
| |
Collapse
|
19
|
Ding WJ, Chen GH, Deng SH, Zeng KF, Lin KL, Deng B, Zhang SW, Tan ZB, Xu YC, Chen S, Chen JB, Chen TF, Tan YZ, Zhou YC, Zhang JZ, Liu B. Calycosin protects against oxidative stress-induced cardiomyocyte apoptosis by activating aldehyde dehydrogenase 2. Phytother Res 2023; 37:35-49. [PMID: 36059198 DOI: 10.1002/ptr.7591] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 07/08/2022] [Accepted: 08/02/2022] [Indexed: 01/19/2023]
Abstract
Myocardial infarction (MI) is the leading cause of death worldwide, and oxidative stress is part of the process that causes MI. Calycosin, a naturally occurring substance with cardioprotective properties, is one of the major active constituents in Radix Astragali. In this study, effect of Calycosin was investigated in vivo and in vitro to determine whether it could alleviate oxidative stress and oxidative stress-induced cardiac apoptosis in neonatal cardiomyocytes (NCMs) via activation of aldehyde dehydrogenase 2 (ALDH2). Calycosin protected against oxidative stress and oxidative stress-induced apoptosis in NCMs. Molecular docking revealed that the ALDH2-Calycosin complex had a binding energy of -9.885 kcal/mol. In addition, molecular docking simulations demonstrated that the ALDH2-Calycosin complex was stable. Using BLI assays, we confirmed that Calycosin could interact with ALDH2 (KD = 1.9 × 10-4 M). Furthermore, an ALDH2 kinase activity test revealed that Calycosin increased ALDH2 activity, exhibiting an EC50 of 91.79 μM. Pre-incubation with ALDH2 inhibitor (CVT-10216 or disulfiram) reduced the cardio-protective properties Calycosin. In mice with MI, Calycosin therapy substantially reduced myocardial apoptosis, oxidative stress, and activated ALDH2. Collectively, our findings clearly suggest that Calycosin reduces oxidative stress and oxidative stress-induced apoptosis via the regulation of ALDH2 signaling, which supports potential therapeutic use in MI.
Collapse
Affiliation(s)
- Wen-Jun Ding
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Guang-Hong Chen
- School of Traditional Chinese Medicine, Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou, China
| | - Sui-Hui Deng
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ke-Feng Zeng
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Kai-Li Lin
- School of Public Health, Guangzhou Medical University, Guangzhou, China
| | - Bo Deng
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shuang-Wei Zhang
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhang-Bin Tan
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - You-Cai Xu
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Si Chen
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jun-Bang Chen
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ting-Fang Chen
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yong-Zhen Tan
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ying-Chun Zhou
- School of Traditional Chinese Medicine, Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou, China
| | - Jing-Zhi Zhang
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Bin Liu
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
20
|
Lv XF, Wen RQ, Liu K, Zhao XK, Pan CL, Gao X, Wu X, Zhi XD, Ren CZ, Chen QL, Lu WJ, Bai TY, Li YD. Role and molecular mechanism of traditional Chinese medicine in preventing cardiotoxicity associated with chemoradiotherapy. Front Cardiovasc Med 2022; 9:1047700. [PMID: 36419486 PMCID: PMC9678083 DOI: 10.3389/fcvm.2022.1047700] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 10/20/2022] [Indexed: 08/12/2023] Open
Abstract
Cardiotoxicity is a serious complication of cancer therapy. It is the second leading cause of morbidity and mortality in cancer survivors and is associated with a variety of factors, including oxidative stress, inflammation, apoptosis, autophagy, endoplasmic reticulum stress, and abnormal myocardial energy metabolism. A number of studies have shown that traditional Chinese medicine (TCM) can mitigate chemoradiotherapy-associated cardiotoxicity via these pathways. Therefore, this study reviews the effects and molecular mechanisms of TCM on chemoradiotherapy-related cardiotoxicity. In this study, we searched PubMed for basic studies on the anti-cardiotoxicity of TCM in the past 5 years and summarized their results. Angelica Sinensis, Astragalus membranaceus Bunge, Danshinone IIA sulfonate sodium (STS), Astragaloside (AS), Resveratrol, Ginsenoside, Quercetin, Danggui Buxue Decoction (DBD), Shengxian decoction (SXT), Compound Danshen Dripping Pill (CDDP), Qishen Huanwu Capsule (QSHWC), Angelica Sinensis and Astragalus membranaceus Bunge Ultrafiltration Extract (AS-AM),Shenmai injection (SMI), Xinmailong (XML), and nearly 60 other herbs, herbal monomers, herbal soups and herbal compound preparations were found to be effective as complementary or alternative treatments. These preparations reduced chemoradiotherapy-induced cardiotoxicity through various pathways such as anti-oxidative stress, anti-inflammation, alleviating endoplasmic reticulum stress, regulation of apoptosis and autophagy, and improvement of myocardial energy metabolism. However, few clinical trials have been conducted on these therapies, and these trials can provide stronger evidence-based support for TCM.
Collapse
Affiliation(s)
- Xin-Fang Lv
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine, University Hospital of Gansu Traditional Chinese Medicine, Lanzhou, China
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Ruo-Qing Wen
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine, University Hospital of Gansu Traditional Chinese Medicine, Lanzhou, China
| | - Kai Liu
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine, University Hospital of Gansu Traditional Chinese Medicine, Lanzhou, China
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Xin-Ke Zhao
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine, University Hospital of Gansu Traditional Chinese Medicine, Lanzhou, China
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Chen-Liang Pan
- The First Hospital of Lanzhou University, Lanzhou, China
| | - Xiang Gao
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine, University Hospital of Gansu Traditional Chinese Medicine, Lanzhou, China
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Xue Wu
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine, University Hospital of Gansu Traditional Chinese Medicine, Lanzhou, China
- Lanzhou University Second Hospital, Lanzhou, China
| | - Xiao-Dong Zhi
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine, University Hospital of Gansu Traditional Chinese Medicine, Lanzhou, China
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Chun-Zhen Ren
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine, University Hospital of Gansu Traditional Chinese Medicine, Lanzhou, China
| | - Qi-Lin Chen
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine, University Hospital of Gansu Traditional Chinese Medicine, Lanzhou, China
| | - Wei-Jie Lu
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine, University Hospital of Gansu Traditional Chinese Medicine, Lanzhou, China
| | - Ting-Yan Bai
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine, University Hospital of Gansu Traditional Chinese Medicine, Lanzhou, China
| | - Ying-Dong Li
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine, University Hospital of Gansu Traditional Chinese Medicine, Lanzhou, China
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| |
Collapse
|
21
|
Wu YJ, Li ZH, Li JY, Zhou Y, Wang RY, Chen XY, Qing LS, Luo P. Elucidation of the binding mechanism of astragaloside IV derivative with human serum albumin and its cardiotoxicity in zebrafish embryos. Front Pharmacol 2022; 13:987882. [PMID: 36210826 PMCID: PMC9537572 DOI: 10.3389/fphar.2022.987882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 09/01/2022] [Indexed: 11/13/2022] Open
Abstract
LS-102 is a new derivative of astragaloside IV (AGS IV) that has been shown to possess potentially significant cardioprotective effects. However, there are no reports concerning its interaction with human serum albumin (HSA) and toxicology in vertebrates. The present investigation was undertaken to characterize the interaction of AGS IV and LS-102 with HSA using equilibrium dialysis and UHPLC-MS/MS methods, along with computational methods. Notably, the effects of AGS IV and LS-102 were studied in vivo using the zebrafish embryo model. Markers related to embryonic cardiotoxicity and thrombosis were evaluated. We showed that the plasma protein binding rate of AGS IV (94.04%–97.42%) was significantly higher than that of LS-102 (66.90%–69.35%). Through site marker competitive experiments and molecular docking, we found that AGS IV and LS-102 were located at the interface of subdomains IIA and IIIA, but the site I might be the primary binding site. Molecular dynamics revealed that AGS IV showed a higher binding free energy mainly due to the stronger hydrophobic and hydrogen bonding interactions. Moreover, the secondary structure implied no obvious effect on the protein structure and conformation during the binding of LS-102. LS-102 significantly ameliorated the astramizole-induced heart rate slowing, increased SV-BA spacing, and prevented arachidonic acid-induced thrombosis in zebrafish. To our knowledge, we are the first to reveal that LS-102 binds to HSA with reversible and moderate affinity, indicating its easy diffusion from the circulatory system to the target tissue, thereby providing significant insights into its pharmacokinetic and pharmacodynamic properties when spread in the human body. Our results also provide a reference for the rational clinical application of LS-102 in the cardiovascular field.
Collapse
Affiliation(s)
- You-Jiao Wu
- State Key Laboratories for Quality Research in Chinese Medicines, Macau University of Science and Technology, Taipa, Macao, SAR, China
| | - Zhan-Hua Li
- State Key Laboratories for Quality Research in Chinese Medicines, Macau University of Science and Technology, Taipa, Macao, SAR, China
| | - Jiu-Yan Li
- State Key Laboratories for Quality Research in Chinese Medicines, Macau University of Science and Technology, Taipa, Macao, SAR, China
| | - Yan Zhou
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
| | - Run-Yue Wang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
| | - Xiao-Yi Chen
- State Key Laboratories for Quality Research in Chinese Medicines, Macau University of Science and Technology, Taipa, Macao, SAR, China
| | - Lin-Sen Qing
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
- *Correspondence: Lin-Sen Qing, ; Pei Luo,
| | - Pei Luo
- State Key Laboratories for Quality Research in Chinese Medicines, Macau University of Science and Technology, Taipa, Macao, SAR, China
- *Correspondence: Lin-Sen Qing, ; Pei Luo,
| |
Collapse
|
22
|
Chen G, Xu H, Xu T, Ding W, Zhang G, Hua Y, Wu Y, Han X, Xie L, Liu B, Zhou Y. Calycosin reduces myocardial fibrosis and improves cardiac function in post-myocardial infarction mice by suppressing TGFBR1 signaling pathways. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 104:154277. [PMID: 35752078 DOI: 10.1016/j.phymed.2022.154277] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/31/2022] [Accepted: 06/14/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Excessive myocardial fibrosis is the pathological basis of heart failure following myocardial infarction (MI). Although calycosin improves cardiac function, its effect on cardiac fibrosis and cardiac function after MI in mice and its precise mechanism remain unclear. PURPOSE Here, we firstly investigated the effects of calycosin on cardiac fibrosis and ventricular function in mice after MI and the role of transforming growth factor-beta receptor 1 (TGFBR1) signaling in the amelioration of cardiac fibrosis and ventricular function. METHODS In vivo effects of calycosin on cardiac structure and function in mice with MI induced by left anterior descending coronary artery ligation were determined by hematoxylin and eosin staining, Masson trichrome staining, and echocardiography. The molecular mechanism of the interaction between TGFBR1 and calycosin was investigated using molecular docking, molecular dynamics (MD) simulation, surface plasmon resonance imaging (SPRi), immunohistochemistry, and western blotting (WB). Subsequently, cardiac-specific Tgfbr1 knockout mice were used to verify the effects of calycosin. The effect of calycosin on primary cardiac fibroblasts (CFs) proliferation and collagen deposition was detected using cell counting (CCK-8), EdU assay, and WB in vitro. CFs infected with an adenovirus that encodes TGFBR1 were used to verify the effects of calycosin. RESULTS In vivo, calycosin attenuated myocardial fibrosis and cardiac dysfunction following MI in a dose-dependent pattern. Calycosin-TGFBR1 complex was found to have a binding energy of -9.04 kcal/mol based on molecular docking. In addition, calycosin bound steadily in the cavity of TGFBR1 during the MD simulation. Based on SPRi results, the solution equilibrium dissociation constant for calycosin and TGFBR1 was 5.11 × 10-5 M. Calycosin inhibited the expression of TGFBR1, Smad2/3, collagen I, and collagen III. The deletion of TGFBR1 partially counteracted these effects. In vitro, calycosin suppressed CFs proliferation and collagen deposition after TGF-β1 stimulation by suppressing the TGFBR1 signaling pathway. The suppressive effects of calycosin were partially rescued by overexpression of TGFBR1. CONCLUSION Calycosin attenuates myocardial fibrosis and cardiac dysfunction following MI in mice in vivo via suppressing the TGFBR1 signaling pathway. Calycosin suppresses CFs proliferation and collagen deposition induced by TGF-β1 via inhibition of the TGFBR1 signaling pathway in vitro.
Collapse
Affiliation(s)
- Guanghong Chen
- Department of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou 510515, China
| | - Honglin Xu
- Department of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou 510515, China
| | - Tong Xu
- Department of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou 510515, China
| | - Wenjun Ding
- Department of Traditional Chinese Medicine (Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, State Key Laboratory of Respiratory Disease), the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510260, China
| | - Guoyong Zhang
- Department of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou 510515, China
| | - Yue Hua
- Department of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou 510515, China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou 510515, China
| | - Yuting Wu
- Department of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou 510515, China
| | - Xin Han
- Department of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou 510515, China
| | - Lingpeng Xie
- Department of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou 510515, China
| | - Bin Liu
- Department of Traditional Chinese Medicine (Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, State Key Laboratory of Respiratory Disease), the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510260, China.
| | - Yingchun Zhou
- Department of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
23
|
Current Status and Trends of Research on Anthracycline-Induced Cardiotoxicity from 2002 to 2021: A Twenty-Year Bibliometric and Visualization Analysis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6260243. [PMID: 35993025 PMCID: PMC9388240 DOI: 10.1155/2022/6260243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/21/2022] [Accepted: 07/24/2022] [Indexed: 12/30/2022]
Abstract
Anthracyclines constitute the cornerstone of numerous chemotherapy regimens for various cancers. However, the clinical application of anthracyclines is significantly limited to their dose-dependent cardiotoxicity. A comprehensive understanding of the current status of anthracycline-induced cardiotoxicity is necessary for in-depth research and optimal clinical protocols. Bibliometric analysis is widely applied in depicting development trends and tracking frontiers of a specific field. The present study is aimed at revealing the status and trends of anthracycline-induced cardiotoxicity during the past two decades by employing bibliometric software including R-bibliometric, VOSviewer, and CiteSpace. A total of 3504 publications concerning anthracycline-induced cardiotoxicity from 2002 to 2021 were collected from the Web of Science Core Collection database. Results showed significant growth in annual yields from 90 records in 2002 to 304 papers in 2021. The United States was the most productive country with the strongest collaboration worldwide in the field. Charles University in the Czech Republic was the institution that contributed the most papers, while 7 of the top 10 productive institutions were from the United States. The United States Department of Health and Human Services and the National Institutes of Health are the two agencies that provide financial support for more than 50% of sponsored publications. The research categories of included publications mainly belong to Oncology and Cardiac Cardiovascular Systems. The Journal of Clinical Oncology had a comprehensive impact on this research field with the highest IF value and many publications. Simunek Tomas from Charles University contributed the most publications, while Lipshultz Steven E. from the State University of New York possessed the highest H-index. In addition, the future research frontiers of anthracycline-induced cardiotoxicity might include early detection, pharmacogenomics, molecular mechanism, and cardiooncology. The present bibliometric analysis may provide a valuable reference for researchers and practitioners in future research directions.
Collapse
|
24
|
Meng C, Fan L, Wang X, Wang Y, Li Y, Pang S, Lv S, Zhang J. Preparation and Evaluation of Animal Models of Cardiotoxicity in Antineoplastic Therapy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3820591. [PMID: 35847594 PMCID: PMC9277159 DOI: 10.1155/2022/3820591] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 06/03/2022] [Indexed: 11/17/2022]
Abstract
The continuous development of antineoplastic therapy has significantly reduced the mortality of patients with malignant tumors, but its induced cardiotoxicity has become the primary cause of long-term death in patients with malignant tumors. However, the pathogenesis of cardiotoxicity of antineoplastic therapy is currently unknown, and practical means of prevention and treatment are lacking in clinical practice. Therefore, how to effectively prevent and treat cardiotoxicity while treating tumors is a major challenge. Animal models are important tools for studying cardiotoxicity in antitumor therapy and are of great importance in elucidating pathophysiological mechanisms and developing and evaluating modality drugs. In this paper, we summarize the existing animal models in antitumor therapeutic cardiotoxicity studies and evaluate the models by observing the macroscopic signs, echocardiography, and pathological morphology of the animals, aiming to provide a reference for subsequent experimental development and clinical application.
Collapse
Affiliation(s)
- Chenchen Meng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China
| | - Lu Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China
| | - Xiaoming Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China
| | - Yunjiao Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China
| | - Yanyang Li
- Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Shuchao Pang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China
| | - Shichao Lv
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China
- Tianjin Key Laboratory of Traditional Research of TCM Prescription and Syndrome, Tianjin 300193, China
| | - Junping Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China
| |
Collapse
|
25
|
Xu H, Qin L, Nie L, Li L, Guo P, Chen Y, Huang C, Su M, Yang B. Biotargets for mediation of arsenic–induced coronary heart disease by calycosin. FOOD AGR IMMUNOL 2022. [DOI: 10.1080/09540105.2022.2053947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Hongyuan Xu
- Cardiology Department, Guigang City People’s Hospital The Eighth Affiliated Hospital of Guangxi Medical University, Guigang, PR People’s Republic of China
| | - Lixiu Qin
- College of Pharmacy, Guangxi Medical University, Nanning, Guangxi, PR People’s Republic of China
| | - Litao Nie
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, PR People’s Republic of China
| | - Lin Li
- Cardiology Department, Guigang City People’s Hospital The Eighth Affiliated Hospital of Guangxi Medical University, Guigang, PR People’s Republic of China
| | - Peng Guo
- Cardiology Department, Guigang City People’s Hospital The Eighth Affiliated Hospital of Guangxi Medical University, Guigang, PR People’s Republic of China
| | - Yizhao Chen
- Cardiology Department, Guigang City People’s Hospital The Eighth Affiliated Hospital of Guangxi Medical University, Guigang, PR People’s Republic of China
| | - Chuang Huang
- Cardiology Department, Guigang City People’s Hospital The Eighth Affiliated Hospital of Guangxi Medical University, Guigang, PR People’s Republic of China
| | - Min Su
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, PR People’s Republic of China
| | - Bin Yang
- College of Pharmacy, Guangxi Medical University, Nanning, Guangxi, PR People’s Republic of China
| |
Collapse
|
26
|
Calycosin Alleviates Doxorubicin-Induced Cardiotoxicity and Pyroptosis by Inhibiting NLRP3 Inflammasome Activation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1733834. [PMID: 35035656 PMCID: PMC8754606 DOI: 10.1155/2022/1733834] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/19/2021] [Accepted: 12/09/2021] [Indexed: 01/13/2023]
Abstract
Calycosin (CAL) is the main active component present in Astragalus and reportedly possesses diverse pharmacological properties. However, the cardioprotective effect and underlying mechanism of CAL against doxorubicin- (DOX-) induced cardiotoxicity need to be comprehensively examined. Herein, we aimed to investigate whether the cardioprotective effects of CAL are related to its antipyroptotic effect. A cardiatoxicity model was established by stimulating H9c2 cells and C57BL/6J mice using DOX. In vitro, CAL increased H9c2 cell viability and decreased DOX-induced pyroptosis via NLRP3, caspase-1, and gasdermin D signaling pathways in a dose-dependent manner. In vivo, CAL-DOX cotreatment effectively suppressed DOX-induced cytotoxicity as well as inflammatory and cardiomyocyte pyroptosis via the same molecular mechanism. Next, we used nigericin (Nig) and NLRP3 forced overexpression to determine whether CAL imparts antipyroptotic effects by inhibiting the NLRP3 inflammasome in vitro. Furthermore, CAL suppressed DOX-induced mitochondrial oxidative stress injury in H9c2 cells by decreasing the generation of reactive oxygen species and increasing mitochondrial membrane potential and adenosine triphosphate. Likewise, CAL attenuated the DOX-induced increase in malondialdehyde content and decreased superoxide dismutase and glutathione peroxidase activities in H9c2 cells. In vivo, CAL afforded a protective effect against DOX-induced cardiac injury by improving myocardial function, inhibiting brain natriuretic peptide, and improving the changes of the histological morphology of DOX-treated mice. Collectively, our findings confirmed that CAL alleviates DOX-induced cardiotoxicity and pyroptosis by inhibiting NLRP3 inflammasome activation in vivo and in vitro.
Collapse
|
27
|
Calycosin Alleviates Paraquat-Induced Neurodegeneration by Improving Mitochondrial Functions and Regulating Autophagy in a Drosophila Model of Parkinson's Disease. Antioxidants (Basel) 2022; 11:antiox11020222. [PMID: 35204105 PMCID: PMC8868496 DOI: 10.3390/antiox11020222] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 01/13/2022] [Accepted: 01/20/2022] [Indexed: 12/12/2022] Open
Abstract
Parkinson's disease (PD) is the second most common age-related neurodegenerative disorder with limited clinical treatments. The occurrence of PD includes both genetic and environmental toxins, such as the pesticides paraquat (PQ), as major contributors to PD pathology in both invertebrate and mammalian models. Calycosin, an isoflavone phytoestrogen, has multiple pharmacological properties, including neuroprotective activity. However, the paucity of information regarding the neuroprotective potential of calycosin on PQ-induced neurodegeneration led us to explore whether calycosin can mitigate PD-like phenotypes and the underlying molecular mechanisms. We used a PQ-induced PD model in Drosophila as a cost-effective in vivo screening platform to investigate the neuroprotective efficacy of natural compounds on PD. We reported that calycosin shows a protective role in preventing dopaminergic (DA) neuronal cell death in PQ-exposed Canton S flies. Calycosin-fed PQ-exposed flies exhibit significant resistance against PQ-induced mortality and locomotor deficits in terms of reduced oxidative stress, loss of DA neurons, the depletion of dopamine content, and phosphorylated JNK-caspase-3 levels. Additionally, mechanistic studies show that calycosin administration improves PQ-induced mitochondrial dysfunction and stimulates mitophagy and general autophagy with reduced pS6K and p4EBP1 levels, suggestive of a maintained energy balance between anabolic and catabolic processes, resulting in the inhibition of neuronal cell death. Collectively, this study substantiates the protective effect of calycosin against PQ-induced neurodegeneration by improving DA neurons' survival and reducing apoptosis, likely via autophagy induction, and it is implicated as a novel therapeutic application against toxin-induced PD pathogenesis.
Collapse
|