1
|
Chen M, He K, Wang K, Cai Y, Ying Z, Li X, Liu Y, Xiang L, Yang P, Wu H, Jiang J. Low-dose radiation ameliorates doxorubicin-induced renal injury via reducing oxidative stress and protecting mitochondrial function. PLoS One 2025; 20:e0313649. [PMID: 39932912 PMCID: PMC11813107 DOI: 10.1371/journal.pone.0313649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 10/28/2024] [Indexed: 02/13/2025] Open
Abstract
Doxorubicin (DOX) is a well-established chemotherapy drug, but its clinical application is restricted due to significant tissue toxicity, of which nephrotoxicity is a serious adverse reaction. Low-dose radiation (LDR) exerts effects through stimulating diverse cell and molecular mechanisms, which has been shown to have anti-inflammatory and alter immune adaptation effects. This study aims to investigate how LDR protects against DOX-induced nephrotoxicity and to explore the underlying mechanism involved. Sixty mice were randomly divided into control (CTR), LDR, DOX, and combination (COM) group. Nephrotoxicity was induced by injecting a single dose of DOX (7.5 mg/kg) in mice abdominal cavity, and LDR was performed 72 h before DOX treatment. Histological analysis, immunohistochemical analysis, immunofluorescence analysis and western-blotting were used to detect the related indicators. Research data was showed as mean ±SD and tested by One-way ANOVA. The results showed that compared with DOX group, the contents of serum UREA, UA, and the expression level of Bax and caspase 9 were significantly reduced in COM group (P<0.05). Western-blotting and immunohistochemical analysis showed that the expression level of MDA and Nrf2 in COM group were significantly lower than that in DOX group (P<0.05). In addition, the activity of complex Ⅰ, ATP, NDUFA1 and CYC1 were enhanced in COM group compared with DOX group (P<0.05). All the results suggested that LDR pretreatment prevented excessive accumulation of peroxides, restored antioxidants activity (SOD, GSH, CAT), activated Nrf2/HO-1/NQO1 signaling pathway, attenuated damage to the mitochondrial respiratory chain, and protected kidney cells from DOX attack. This study demonstrated that LDR could effectively and safely inhibit the progression of DOX-induced nephrotoxicity. Future studies should further investigate the mechanism of LDR protecting tissues from DOX-induced damage and find an optimal radiation dose for humans.
Collapse
Affiliation(s)
- Mengmeng Chen
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
- Department of Nursing, Zhejiang Cancer Hospital, Hangzhou, China
| | - Kang He
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| | - Kai Wang
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| | - Yibo Cai
- Department of Nursing, Zhejiang Cancer Hospital, Hangzhou, China
| | - Zhaohui Ying
- Department of Nursing, Zhejiang Cancer Hospital, Hangzhou, China
| | - Xueting Li
- Department of Nursing, Zhejiang Cancer Hospital, Hangzhou, China
| | - Yating Liu
- Department of Nursing, Zhejiang Cancer Hospital, Hangzhou, China
| | - Liting Xiang
- Department of Nursing, Zhejiang Cancer Hospital, Hangzhou, China
| | - Pingping Yang
- Department of Nursing, Zhejiang Cancer Hospital, Hangzhou, China
| | - Hongjuan Wu
- Department of Nursing, Zhejiang Cancer Hospital, Hangzhou, China
| | - Jian Jiang
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| |
Collapse
|
2
|
Tayal R, Mannan A, Singh S, Dhiman S, Singh TG. Unveiling the Complexities: Exploring Mechanisms of Anthracyclineinduced Cardiotoxicity. Curr Cardiol Rev 2025; 21:42-77. [PMID: 39484769 PMCID: PMC12060933 DOI: 10.2174/011573403x322928241021100631] [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: 04/12/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 11/03/2024] Open
Abstract
The coexistence of cancer and heart disease, both prominent causes of illness and death, is further exacerbated by the detrimental impact of chemotherapy. Anthracycline-induced cardiotoxicity is an unfortunate side effect of highly effective therapy in treating different types of cancer; it presents a significant challenge for both clinicians and patients due to the considerable risk of cardiotoxicity. Despite significant progress in understanding these mechanisms, challenges persist in identifying effective preventive and therapeutic strategies, rendering it a subject of continued research even after three decades of intensive global investigation. The molecular targets and signaling pathways explored provide insights for developing targeted therapies, emphasizing the need for continued research to bridge the gap between preclinical understanding and clinical applications. This review provides a comprehensive exploration of the intricate mechanisms underlying anthracycline-induced cardiotoxicity, elucidating the interplay of various signaling pathways leading to adverse cellular events, including cardiotoxicity and death. It highlights the extensive involvement of pathways associated with oxidative stress, inflammation, apoptosis, and cellular stress responses, offering insights into potential and unexplored targets for therapeutic intervention in mitigating anthracycline-induced cardiac complications. A comprehensive understanding of the interplay between anthracyclines and these complexes signaling pathways is crucial for developing strategies to prevent or mitigate the associated cardiotoxicity. Further research is needed to outline the specific contributions of these pathways and identify potential therapeutic targets to improve the safety and efficacy of anthracycline-based cancer treatment. Ultimately, advancements in understanding anthracycline-induced cardiotoxicity mechanisms will facilitate the development of more efficacious preventive and treatment approaches, thereby improving outcomes for cancer patients undergoing anthracycline-based chemotherapy.
Collapse
Affiliation(s)
- Rohit Tayal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Shareen Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Sonia Dhiman
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | | |
Collapse
|
3
|
Novakovic J, Muric M, Bradic J, Ramenskaya G, Jakovljevic V, Jeremic N. Diallyl Trisulfide and Cardiovascular Health: Evidence and Potential Molecular Mechanisms. Int J Mol Sci 2024; 25:9831. [PMID: 39337318 PMCID: PMC11431890 DOI: 10.3390/ijms25189831] [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: 07/23/2024] [Revised: 09/01/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024] Open
Abstract
Traditionally, garlic has a valuable role in preventing and reducing the incidence of many diseases and pathophysiological disorders. Consequently, some researchers have focused on the beneficial cardiovascular properties of diallyl trisulfide (DATS), the most potent polysulfide isolated from garlic. Therefore, in this review, we collected the available data on DATS, its biochemical synthesis, metabolism and pharmacokinetics, and gathered the current knowledge and the role of DATS in cardiovascular diseases. Overall, this review summarizes the cardioprotective effects of DATS and brings together all previous findings on its protective molecular mechanisms, which are mainly based on the potent anti-apoptotic, anti-inflammatory, and antioxidant potential of this polysulfide. Our review is an important cornerstone for further basic and clinical research on DATS as a new therapeutic agent for the treatment of numerous heart diseases.
Collapse
Affiliation(s)
- Jovana Novakovic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- Center of Excellence for the Study of Redox Balance in Cardiovascular and Metabolic Disorders, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Maja Muric
- Center of Excellence for the Study of Redox Balance in Cardiovascular and Metabolic Disorders, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Jovana Bradic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- Center of Excellence for the Study of Redox Balance in Cardiovascular and Metabolic Disorders, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Galina Ramenskaya
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| | - Vladimir Jakovljevic
- Center of Excellence for the Study of Redox Balance in Cardiovascular and Metabolic Disorders, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- Department of Human Pathology, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| | - Nevena Jeremic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- Center of Excellence for the Study of Redox Balance in Cardiovascular and Metabolic Disorders, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| |
Collapse
|
4
|
Cai K, Jiang H, Zou Y, Song C, Cao K, Chen S, Wu Y, Zhang Z, Geng D, Zhang N, Liu B, Sun G, Tang M, Li Z, Zhang Y, Sun Y, Zhang Y. Programmed death of cardiomyocytes in cardiovascular disease and new therapeutic approaches. Pharmacol Res 2024; 206:107281. [PMID: 38942341 DOI: 10.1016/j.phrs.2024.107281] [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: 04/23/2024] [Revised: 06/21/2024] [Accepted: 06/21/2024] [Indexed: 06/30/2024]
Abstract
Cardiovascular diseases (CVDs) have a complex pathogenesis and pose a major threat to human health. Cardiomyocytes have a low regenerative capacity, and their death is a key factor in the morbidity and mortality of many CVDs. Cardiomyocyte death can be regulated by specific signaling pathways known as programmed cell death (PCD), including apoptosis, necroptosis, autophagy, pyroptosis, and ferroptosis, etc. Abnormalities in PCD can lead to the development of a variety of cardiovascular diseases, and there are also molecular-level interconnections between different PCD pathways under the same cardiovascular disease model. Currently, the link between programmed cell death in cardiomyocytes and cardiovascular disease is not fully understood. This review describes the molecular mechanisms of programmed death and the impact of cardiomyocyte death on cardiovascular disease development. Emphasis is placed on a summary of drugs and potential therapeutic approaches that can be used to treat cardiovascular disease by targeting and blocking programmed cell death in cardiomyocytes.
Collapse
Affiliation(s)
- Kexin Cai
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Haoyue Jiang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Yuanming Zou
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Chunyu Song
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Kexin Cao
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Shuxian Chen
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Yanjiao Wu
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Zhaobo Zhang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Danxi Geng
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Naijin Zhang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China; Institute of health sciences, China medical university, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China; Key Laboratory of Reproductive and Genetic Medicine (China Medical University), National Health Commission, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China
| | - Bo Liu
- The first hospital of China Medical University, Department of cardiac surgery, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China.
| | - Guozhe Sun
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China.
| | - Man Tang
- Department of clinical pharmacology, College of Pharmacy, China medical university, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China.
| | - Zhao Li
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China.
| | - Yixiao Zhang
- Department of Urology Surgery, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, People's Republic of China.
| | - Yingxian Sun
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China; Institute of health sciences, China medical university, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China; Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China.
| | - Ying Zhang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China; Institute of health sciences, China medical university, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China.
| |
Collapse
|
5
|
Yoshikawa N, Hirata N, Kurone Y, Shimoeda S. Red ginseng prevents doxorubicin-induced cardiomyopathy by inhibiting cell death via activating the Nrf2 pathway. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:39. [PMID: 38909271 PMCID: PMC11193215 DOI: 10.1186/s40959-024-00242-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 06/13/2024] [Indexed: 06/24/2024]
Abstract
BACKGROUND Doxorubicin (DXR) is an effective chemotherapeutic agent. DOX-induced cardiomyopathy (DICM), a major limitation of DXR, is a complication with limited treatment options. We previously reported that Red Ginseng (steamed and dried the root of Panax Ginseng cultivated for over six years; RGin) is beneficial for the treatment of DICM. However, the mechanism underlying the action of RGin remains unclear. In this study, we investigated the mechanism of action underlying the efficacy of RGin in the treatment of DICM. METHODS Four-week-old DBA/2 mice were divided into: vehicle, DXR, RGin, and DXR + RGin (n = 10/group). Mice were treated with DXR (4 mg/kg, once a week, accumulated 20 mg/kg, i.p.) or RGin (0.5 g/kg, three times a week, i.p.). To evaluate efficacy, the survival rate and left ventricular ejection fraction (LVEF) were measured as a measure of cardiac function, and cardiomyocytes were subjected to Masson trichrome staining. To investigate the mechanism of action, western blotting was performed to evaluate the expression of nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase 1, transferrin receptor (TfR), and other related proteins. Data were analyzed using the Easy R software. Between-group comparisons were performed using one-way analysis of variance and analyzed using a post-hoc Tukey test. Survival rates were estimated using the Kaplan-Meier method and compared using the log-rank test. P < 0.05 was considered statistically significant in all analyses. RESULTS RGin treatment prolongs survival and protects against reduced LVEF. In the DXR group, Nrf2 was not activated and cell death was accelerated. Furthermore, there was an increase in the TfR levels, suggesting abnormal iron metabolism. However, the DXR + RGin group showed activation of the Nrf2 pathway and suppression of myocardial cell death. Furthermore, there was no increase in TfR expression, suggesting that there were no abnormalities in iron metabolism. Therefore, the mechanism of action of RGin in DICM involves an increase in antioxidant activity and inhibition of cell death through activation of the Nrf2 pathway. CONCLUSION RGin is a useful therapeutic candidate for DICM. Its efficacy is supported by the activation of the Nrf2 pathway, which enhances antioxidant activity and inhibits cell death.
Collapse
Affiliation(s)
- Naoki Yoshikawa
- Department of Pharmaceutical Health Care and Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| | - Naoto Hirata
- Department of Pharmaceutical Health Care and Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| | - Yuichiro Kurone
- Department of Pharmaceutical Health Care and Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| | - Sadahiko Shimoeda
- Department of Pharmaceutical Health Care and Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan.
| |
Collapse
|
6
|
Yang Y, Yang R, Deng F, Yang L, Yang G, Liu Y, Tian Q, Wang Z, Li A, Shang L, Cheng G, Zhang L. Immunoactivation by Cutaneous Blue Light Irradiation Inhibits Remote Tumor Growth and Metastasis. ACS Pharmacol Transl Sci 2024; 7:1055-1068. [PMID: 38633599 PMCID: PMC11019738 DOI: 10.1021/acsptsci.3c00355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 03/01/2024] [Accepted: 03/06/2024] [Indexed: 04/19/2024]
Abstract
An improved innate immunity will respond quickly to pathogens and initiate efficient adaptive immune responses. However, up to now, there have been limited clinical ways for effective and rapid consolidation of innate immunity. Here, we report that cutaneous irradiation with blue light of 450 nm rapidly stimulates the innate immunity through cell endogenous reactive oxygen species (ROS) regulation in a noninvasive way. The iron porphyrin-containing proteins, mitochondrial cytochrome c (Cyt-c), and cytochrome p450 (CYP450) can be mobilized by blue light, which boosts electron transport and ROS production in epidermal and dermal tissues. As a messenger of innate immune activation, the increased level of ROS activates the NF-κB signaling pathway and promotes the secretion of immunomodulatory cytokines in skin. Initiated from skin, a regulatory network composed of cytokines and immune cells is established through the circulation system for innate immune activation. The innate immunity activated by whole-body blue light irradiation inhibits tumor growth and metastasis by increasing the infiltration of antitumor neutrophils and tumor-associated macrophages. Our results elucidate the remote immune modulation mechanism of blue light and provide a clinically applicable way for innate immunity activation.
Collapse
Affiliation(s)
- Yingchun Yang
- Key
Laboratory for Space Bioscience & Biotechnology, School of Life
Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Rong Yang
- Key
Laboratory for Space Bioscience & Biotechnology, School of Life
Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Fangqing Deng
- Key
Laboratory for Space Bioscience & Biotechnology, School of Life
Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Luqiu Yang
- Key
Laboratory for Space Bioscience & Biotechnology, School of Life
Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Guanghao Yang
- Key
Laboratory for Space Bioscience & Biotechnology, School of Life
Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Yanyan Liu
- Key
Laboratory for Space Bioscience & Biotechnology, School of Life
Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Qing Tian
- Key
Laboratory for Space Bioscience & Biotechnology, School of Life
Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Zixi Wang
- Key
Laboratory for Space Bioscience & Biotechnology, School of Life
Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Aipeng Li
- School
of Chemical Engineering and Technology, Xi’an Jiaotong University, Xi’an 710049, China
| | - Li Shang
- School
of Materials Science and Engineering, Northwestern
Polytechnical University, Xi’an 710072, China
| | - Genyang Cheng
- Department
of Nephrology, the First Affiliated Hospital
of Zhengzhou University, Zhengzhou 450052, China
| | - Lianbing Zhang
- Key
Laboratory for Space Bioscience & Biotechnology, School of Life
Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| |
Collapse
|
7
|
Ma H, Mao C, Hu Y, Wang L, Guo X, Li L, Wang F, Guan R. Angiotensin-(1-9) attenuates adriamycin-induced cardiomyopathy in rats via the angiotensin type 2 receptor. Mol Cell Biochem 2024; 479:73-83. [PMID: 36995547 DOI: 10.1007/s11010-023-04718-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 03/19/2023] [Indexed: 03/31/2023]
Abstract
Adriamycin (ADR) causes irreversible damage to the heart, leading to ADR-induced cardiomyopathy (ACM). Angiotensin-(1-9) [Ang-(1-9)] is a peptide from the counter-regulatory renin-angiotensin system, but the effects on ACM is unclear. Our study was aimed to explore the effects and underlying molecular mechanisms of Ang-(1-9) against ACM in Wistar rats. Rats were injected intraperitoneally with ADR via six equal doses (each containing 2.5 mg/kg) within a period of 2 weeks to induce ACM. After 2 weeks of ADR treatment, the rats were treated with Ang-(1-9) (200 ng/kg/min) or angiotensin type 2 receptor (AT2R) antagonist PD123319 (100 ng/kg/min) for 4 weeks. Although Ang-(1-9) treatment did not influence blood pressure, it significantly improved left ventricular function and remodeling in ADR-treated rats, by inhibiting collagen deposition, the expression of TGF-β1, inflammatory response, cardiomyocyte apoptosis and oxidative stress. Moreover, Ang-(1-9) reduced ERK1/2 and P38 MAPK phosphorylation. The therapeutic effects of Ang-(1-9) were blocked by the AT2R antagonist PD123319, which also offset the down-regulation protein expression of pERK1/2 and pP38 MAPK induced by Ang-(1-9). These data suggest that Ang-(1-9) improved left ventricular function and remodeling in ADR-treated rats by an AT2R/ ERK1/2 and P38 MAPK-dependent mechanism. Thus, the Ang-(1-9)/AT2R axis may provide a novel and promising target to the prevention and treatment of ACM.
Collapse
Affiliation(s)
- Hui Ma
- Department of Pediatrics, Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Chenggang Mao
- Department of Pediatrics, Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Yang Hu
- Department of Pediatrics, Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Liqin Wang
- Department of Anesthesiology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xingqing Guo
- Department of Pediatrics, Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Lei Li
- Department of Pediatrics, Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Fang Wang
- Department of Pediatrics, Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Renzheng Guan
- Department of Pediatrics, Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
| |
Collapse
|
8
|
Ibrahim MA, Khalifa AM, Abd El-Fadeal NM, Abdel-Karim RI, Elsharawy AF, Ellawindy A, Galal HM, Nadwa EH, Abdel-Shafee MA, Galhom RA. Alleviation of doxorubicin-induced cardiotoxicity in rat by mesenchymal stem cells and olive leaf extract via MAPK/ TNF-α pathway: Preclinical, experimental and bioinformatics enrichment study. Tissue Cell 2023; 85:102239. [PMID: 37865037 DOI: 10.1016/j.tice.2023.102239] [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: 04/24/2023] [Revised: 08/31/2023] [Accepted: 10/06/2023] [Indexed: 10/23/2023]
Abstract
BACKGROUND Toxic cardiomyopathies were a potentially fatal adverse effect of anthracycline therapy. AIM This study was conducted to demonstrate the pathogenetic, morphologic, and toxicologic effects of doxorubicin on the heart and to investigate how the MAPK /TNF-α pathway can be modulated to improve doxorubicin-Induced cardiac lesions using bone marrow-derived mesenchymal stem cells (BM-MSCs) and olive leaf extract (OLE). METHODS During the study, 40 adult male rats were used. Ten were used to donate MSCs, and the other 30 were split into 5 equal groups: Group I was the negative control, Group II obtained oral OLE, Group III obtained an intraperitoneal cumulative dose of DOX (12 mg/kg) in 6 equal doses of 2 mg/kg every 48 h for 12 days, Group IV obtained intraperitoneal DOX and oral OLE at the same time, and Group V obtained intraperitoneal DOX and BM-MSCs through the tail vein at the same time for 12 days. Four weeks after their last dose of DOX, the rats were euthanized. By checking the bioinformatic databases, a molecularly targeted path was selected. Then the histological, immunohistochemistry, and gene expression of ERK, JNK, NF-κB, IL-6, and TNF-α were done. RESULTS Myocardial immunohistochemistry revealed severe fibrosis, cell degeneration, increased vimentin, and decreased CD-31 expression in the DOX-treated group, along with a marked shift in morphometric measurements, a disordered ultrastructure, and overexpression of inflammatory genes (ERK, NF-κB, IL-6, and TNF-α), oxidative stress markers, and cardiac biomarkers. Both groups IV and V displayed reduced cardiac fibrosis or inflammation, restoration of the microstructure and ultrastructure of the myocardium, downregulation of inflammatory genes, markers of oxidative stress, and cardiac biomarkers, a notable decline in vimentin, and an uptick in CD-31 expression. In contrast to group IV, group V showed a considerable beneficial effect. CONCLUSION Both OLE and BM-MSCs showed an ameliorating effect in rat models of DOX-induced cardiotoxicity, with BM-MSCs showing a greater influence than OLE.
Collapse
Affiliation(s)
- Mahrous A Ibrahim
- Department of Internal Medicine (Forensic Medicine and Clinical Toxicology division), College of Medicine, Jouf University, Aljouf 72341, Saudi Arabia.
| | - Athar M Khalifa
- Pathology Department, College of Medicine, Jouf University, Aljouf, Saudi Arabia
| | - Noha M Abd El-Fadeal
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt; Center of Excellence in Molecular and Cellular Medicine, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt; Oncology Diagnostic Unit, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Rehab I Abdel-Karim
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Ayman F Elsharawy
- Histology Department, Faculty of Medicine Al-Azhar University, Cairo, Egypt; Histology Department, College of Medicine, Shaqra University, Shaqra, Saudi Arabia
| | - Alia Ellawindy
- Medical Genetics Unit, Department of Histology and Cell Biology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Heba M Galal
- Department of Medical Physiology, College of Medicine, Jouf University, Sakaka, Saudi Arabia; Department of Medical Physiology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Eman H Nadwa
- Department of Pharmacology and Therapeutics, College of Medicine, Jouf University, Sakaka 72345, Saudi Arabia; Department of Medical Pharmacology, Faculty of Medicine, Cairo University, Giza 12613, Egypt
| | - Mohamed A Abdel-Shafee
- Department of Cardiovascular Medicine, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Rania A Galhom
- Center of Excellence in Molecular and Cellular Medicine, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt; Human Anatomy and Embryology Department, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt; Human Anatomy and Embryology Department, Faculty of Medicine, Badr University in Cairo (BUC), Cairo 11829, Egypt
| |
Collapse
|
9
|
Huang P, Zhang W, Ji J, Ma J, Cheng H, Qin M, Wei D, Ren L. LncRNA Miat knockdown protects against pirarubicin-induced cardiotoxicity by targeting miRNA-129-1-3p. ENVIRONMENTAL TOXICOLOGY 2023; 38:2751-2760. [PMID: 37471631 DOI: 10.1002/tox.23910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 05/31/2023] [Accepted: 07/09/2023] [Indexed: 07/22/2023]
Abstract
Pirarubicin (THP) is a widely used antitumor drug in clinical practice, but its cardiotoxicity limits its use. The aim of this study was to investigate the protective effect and mechanism of knockdown of lncRNA Miat in THP-induced cardiotoxicity. The extent of damage to immortalized cardiomyocytes in mice was assessed by CCK8, TUNEL, ROS, Ca2+ , RT-qPCR, and Western blot. The relative levels of Miat in THP-treated cardiomyocytes (HL-1) were measured. The protective effect of Miat on THP-treated HL-1 was assessed. The binding relationship between lncRNA Miat and mmu-miRNA-129-1-3p was verified by a dual luciferase reporter gene assay. The protective role of Miat/miRNA-129-1-3p in THP-induced HL-1 was explored by performing a rescue assay. THP reduced cell viability, induced apoptosis, triggered oxidative stress and calcium overload. Expression of Miat in HL-1 was significantly elevated after THP treatment. Miat knockdown significantly alleviated the cardiotoxicity of THP. MiR-129-1-3p is a direct target of Miat. Knockdown of miR-129-1-3p reversed the protective effect of Miat knockdown on HL-1. Miat knockdown can alleviate THP-induced cardiomyocyte injury by regulating miR-129-1-3p.
Collapse
Affiliation(s)
- Peng Huang
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Wenqing Zhang
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Jiahua Ji
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Jiulong Ma
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Hongyuan Cheng
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Meng Qin
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Dexian Wei
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Liqun Ren
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| |
Collapse
|
10
|
Hui T, Yiling J, Guangqun C, Ran L, Hui L, Lan Y, Jie H, Su Q. Diallyl disulfide downregulating RhoGDI2 induces differentiation and inhibit invasion via the Rac1/Pak1/LIMK1 pathway in human leukemia HL-60 cells. ENVIRONMENTAL TOXICOLOGY 2023; 38:1063-1077. [PMID: 36793247 DOI: 10.1002/tox.23748] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/30/2022] [Accepted: 01/16/2023] [Indexed: 06/18/2023]
Abstract
Leukemia is a type of disease in which hematopoietic stem cells proliferate clonally at the genetic level. We discovered previously by high-resolution mass spectrometry that diallyl disulfide (DADS), which is one of the effective ingredients of garlic, reduces the performance of RhoGDI2 from APL HL-60 cells. Although RhoGDI2 is oversubscribed in several cancer categories, the effect of RhoGDI2 in HL-60 cells has remained unexplained. We aimed to investigate the influence of RhoGDI2 on DADS-induced differentiation of HL-60 cells to elucidate the association among the effect of inhibition or over-expression of RhoGDI2 with HL-60 cell polarization, migration and invasion, which is important for establishing a novel generation of inducers to elicit leukemia cell polarization. Co-transfection with RhoGDI2-targeted miRNAs apparently decreases the malignant biological behavior of cells and upregulates cytopenias in DADS-treated HL-60 cell lines, which increases CD11b and decreases CD33 and mRNA levels of Rac1, PAK1 and LIMK1. Meanwhile, we generated HL-60 cell lines with high-expressing RhoGDI2. The proliferation, migration and invasion capacity of such cells were significantly increased by the treated with DADS, while the reduction capacity of the cells was decreased. There was a reduction in CD11b and an increase in CD33 production, as well as an increase in the mRNA levels of Rac1, PAK1 and LIMK1. It also confirmed that inhibition of RhoGDI2 attenuates the EMT cascade via the Rac1/Pak1/LIMK1 pathway, thereby inhibiting the malignant biological behavior of HL-60 cells. Thus, we considered that inhibition of RhoGDI2 expression might be a new therapeutic direction for the treatment of human promyelocytic leukemia. The anti-cancer property of DADS against HL-60 leukemia cells might be regulated by RhoGDI2 through the Rac1-Pak1-LIMK1 pathway, which provides new evidence for DADS as a clinical anti-cancer medicine.
Collapse
Affiliation(s)
- Tan Hui
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China
| | - Jiang Yiling
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China
- Department of Oncology, First Affiliated Hospital, University of South China, Hengyang, China
| | - Chen Guangqun
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China
- Department of Oncology, Loudi Central Hospital, Loudi, China
| | - Liu Ran
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China
- Department of Pathology, The First Hospital of Changsha, Changsha, China
| | - Ling Hui
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China
| | - Yi Lan
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China
| | - He Jie
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China
| | - Qi Su
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China
- Department of Oncology, First Affiliated Hospital, University of South China, Hengyang, China
| |
Collapse
|
11
|
Xi Y, Chi Z, Tao X, Zhai X, Zhao Z, Ren J, Yang S, Dong D. Naringin against doxorubicin-induced hepatotoxicity in mice through reducing oxidative stress, inflammation, and apoptosis via the up-regulation of SIRT1. ENVIRONMENTAL TOXICOLOGY 2023; 38:1153-1161. [PMID: 36811345 DOI: 10.1002/tox.23755] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 01/18/2023] [Accepted: 01/27/2023] [Indexed: 06/18/2023]
Abstract
Clinical application of doxorubicin is limited because of its potential side effects. The present study examined whether naringin had protective actions on doxorubicin-induced liver injury. Male BALB/c mice and alpha mouse liver 12 (AML-12) cells were used in this paper. The results showed that AML-12 cells treated with naringin significantly reduced cell injury, reactive oxygen species release and apoptosis level; Moreover, naringin notably alleviated liver injury by decreasing aspartate transaminase, alanine transaminase and malondialdehyde, and increasing superoxide dismutase, glutathione and catalase levels. Mechanism researches indicated that naringin increased the expression levels of sirtuin 1 (SIRT1), and inhibited the downstream inflammatory, apoptotic and oxidative stress signaling pathways. Further validation was obtained by knocking down SIRT1 in vitro, which proved the effects of naringin on doxorubicin-induced liver injury. Therefore, naringin is a valuable lead compound for preventing doxorubicin-induced liver damage by reducing oxidative stress, inflammation, and apoptosis via up-regulation of SIRT1.
Collapse
Affiliation(s)
- Yan Xi
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Zhongchao Chi
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
- Department of Pharmacy, Third People's Hospital of Dalian, Dalian, China
| | - Xufeng Tao
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiaohan Zhai
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Zirui Zhao
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jiaqi Ren
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shilei Yang
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Deshi Dong
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
12
|
Lu H, Yin K, Su H, Wang D, Zhang Y, Hou L, Li JB, Wang Y, Xing M. Polystyrene microplastics induce autophagy and apoptosis in birds lungs via PTEN/PI3K/AKT/mTOR. ENVIRONMENTAL TOXICOLOGY 2023; 38:78-89. [PMID: 36205374 DOI: 10.1002/tox.23663] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 06/16/2023]
Abstract
Microplastics (MPs) seriously pollute and potentially threaten human health. Birds are sentinels of environmental pollutants, which respond quickly to contamination events and reveal current environmental exposure. Therefore, birds are good bioindicators for monitoring environmental pollutants. However, the mechanism of lung injury in birds and the role of the PTEN/PI3K/AKT axis are unknown. In this study, broilers treated with different polystyrene microplastics (PS-MPs) (0, 1, 10, and 100 mg/L) were exposed to drinking water for 6 weeks to analyze the effect of PS-MPs on lung injury of broilers. The results showed that with the increase of PS-MPs concentration, malonaldehyde (MDA) content increased, and catalase (CAT) and glutathione (GSH) activity decreased, further leading to oxidative stress. PS-MPs caused the PI3K/Akt/mTOR pathway to be inhibited by phosphorylation, and autophagy accelerated formation (LC3) and degradation (p62), causing autophagy. In PS-MPs exposed lung tissues, the expression of Bax/Bcl-2 and Caspase family increased, and MAPK signaling pathways (p38, ERK, and JNK) showed an increase in phosphorylation level, thus leading to cell apoptosis. Our research showed that PS-MPs could activate the antioxidant system. The antioxidant system unbalance-regulated Caspase family, and PTEN/PI3K/AKT pathways initiated apoptosis and autophagy, which in turn led to lung tissue damage in chickens. These results are of great significance to the toxicological study of PS-MPs and the protection of the ecosystem.
Collapse
Affiliation(s)
- Hongmin Lu
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, People's Republic of China
| | - Kai Yin
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, People's Republic of China
| | - Heng Su
- College of Resources and Environment, Northeast Agricultural University, Harbin, People's Republic of China
| | - Dongxu Wang
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, People's Republic of China
| | - Yue Zhang
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, People's Republic of China
| | - Lulu Hou
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, People's Republic of China
| | - Jun Bo Li
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, People's Republic of China
| | - Yu Wang
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, People's Republic of China
| | - Mingwei Xing
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, People's Republic of China
| |
Collapse
|
13
|
Tang X, Fan X, Xu T, He Y, Chi Q, Li Z, Li S. Polystyrene nanoplastics exacerbated lipopolysaccharide-induced necroptosis and inflammation via the ROS/MAPK pathway in mice spleen. ENVIRONMENTAL TOXICOLOGY 2022; 37:2552-2565. [PMID: 35833596 DOI: 10.1002/tox.23618] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/30/2022] [Accepted: 07/03/2022] [Indexed: 06/15/2023]
Abstract
Plastics are novel environmental pollutants with potential threats to the ecosystem. At least 5.25 trillion plastic particles in the environment, of which nanoplastics are <100 nm in diameter. Polystyrene nanoplastics (PS-NPs) exposure damaged the spleen's immune function. Lipopolysaccharide (LPS) induced other toxicants to damage cells and organs, triggering inflammation. However, the mechanism of PS-NPs aggravated LPS-induced spleen injury remains unclear. In this study, the PS-NPs or/and LPS mice exposure model was replicated by intraperitoneal injection of PS-NPs or/and LPS, and PS-NPs or/and LPS were exposed to RAW264.7 cells. The histopathological and ultrastructural changes of the mice spleen were observed by H&E staining and transmission electron microscope. Western Blot, qRT-PCR, and fluorescent probes staining were used to detect reactive oxygen species (ROS), oxidative stress indicators, inflammatory factors, and necroptosis-related indicators in mice spleen and RAW264.7 cells. The results showed that PS-NPs or LPS induced oxidative stress, activated the MAPK pathway, and eventually caused necroptosis and inflammation in mice spleen and RAW264.7 cells. Compared with the single treatment group, the changes in PS-NPs + LPS group were more obvious. Furthermore, ROS inhibitor N-Acetyl-L-cysteine (NAC) significantly inhibited the activation of the mitogen-activated protein kinase (MAPK) signaling pathway caused by co-treatment of PS-NPs and LPS, reducing necroptosis and inflammation. The results demonstrated that PS-NPs promoted LPS-induced spleen necroptosis and inflammation in mice through the ROS/MAPK pathway. This study increases the data on the damage of PS-NPs to the organism and expands the research ideas and clues.
Collapse
Affiliation(s)
- Xinyu Tang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China
| | - Xue Fan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China
| | - Tong Xu
- College of Chemistry, Jilin University, Changchun, People's Republic of China
| | - Yujiao He
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China
| | - Qianru Chi
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China
| | - Zhe Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China
| | - Shu Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China
| |
Collapse
|
14
|
Ji Y, Jin D, Qi J, Wang X, Zhang C, An P, Luo Y, Luo J. Fucoidan Protects against Doxorubicin-Induced Cardiotoxicity by Reducing Oxidative Stress and Preventing Mitochondrial Function Injury. Int J Mol Sci 2022; 23:10685. [PMID: 36142635 PMCID: PMC9504360 DOI: 10.3390/ijms231810685] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/08/2022] [Accepted: 09/12/2022] [Indexed: 11/16/2022] Open
Abstract
Doxorubicin (DOXO) is a potent chemotherapeutic drug widely used to treat various cancers. However, its clinical application is limited due to serious adverse effects on dose-dependent cardiotoxicity. Although the underlying mechanism has not been fully clarified, DOXO-induced cardiotoxicity has been mainly attributed to the accumulation of reactive oxygen species (ROS) in cardiomyocytes. Fucoidan, as a kind of sulphated polysaccharide existing in numerous brown seaweed, has potent anti-oxidant, immune-regulatory, anti-tumor, anti-coagulate and anti-viral activities. Here, we explore the potential protective role and mechanism of fucoidan in DOXO-induced cardiotoxicity in mice. Our results show that oral fucoidan supplement exerts potent protective effects against DOXO-induced cardiotoxicity by reducing oxidative stress and preventing mitochondrial function injury. The improved effect of fucoidan on DOXO-induced cardiotoxicity was evaluated by echocardiography, cardiac myocytes size and cardiac fibrosis analysis, and the expression of genes related to cardiac dysfunction and remodeling. Fucoidan reduced the ROS content and the MDA levels but enhanced the activity of antioxidant enzymes GSH-PX and SOD in the mouse serum in a DOXO-induced cardiotoxicity model. In addition, fucoidan also increased the ATP production capacity and restored the levels of a mitochondrial respiratory chain complex in heart tissue. Collectively, this study highlights fucoidan as a potential polysaccharide for protecting against DOXO-induced cardiovascular diseases.
Collapse
Affiliation(s)
- Yuting Ji
- Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing 100193, China
| | - Dekui Jin
- Department of General Practice, The Third Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Jingyi Qi
- Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing 100193, China
| | - Xuan Wang
- Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing 100193, China
| | - Chengying Zhang
- Department of General Practice, The Third Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Peng An
- Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing 100193, China
| | - Yongting Luo
- Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing 100193, China
| | - Junjie Luo
- Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing 100193, China
| |
Collapse
|
15
|
Du XY, Xiang DC, Gao P, Peng H, Liu YL. Inhibition of (Pro)renin Receptor-Mediated Oxidative Stress Alleviates Doxorubicin-Induced Heart Failure. Front Oncol 2022; 12:874852. [PMID: 35574363 PMCID: PMC9106363 DOI: 10.3389/fonc.2022.874852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 03/31/2022] [Indexed: 11/22/2022] Open
Abstract
AIM Clinical utility of doxorubicin (DOX) is limited by its cardiotoxic side effect, and the underlying mechanism still needs to be fully elucidated. This research aimed to examine the role of (pro)renin receptor (PRR) in DOX-induced heart failure (HF) and its underlying mechanism. MAIN METHODS Sprague Dawley (SD) rats were injected with an accumulative dosage of DOX (15 mg/kg) to induce HF. Cardiac functions were detected by transthoracic echocardiography examination. The levels of lactate dehydrogenase (LDH) and creatine kinase (CK) in serum were detected, and oxidative stress related injuries were evaluated. Furthermore, the mRNA expression of PRR gene and its related genes were detected by real-time PCR (RT-PCR), and protein levels of PRR, RAC1, NOX4 and NOX2 were determined by Western blot. Reactive oxygen species (ROS) were determined in DOX-treated rats or cells. Additionally, PRR and RAC1 were silenced with their respective siRNAs to validate the in vitro impacts of PRR/RAC1 on DOX-induced cardiotoxicity. Moreover, inhibitors of PRR and RAC1 were used to validate their effects in vivo. KEY FINDINGS PRR and RAC1 expressions increased in DOX-induced HF. The levels of CK and LDH as well as oxidative stress indicators increased significantly after DOX treatment. Oxidative injury and apoptosis of cardiomyocytes were attenuated both in vivo and in vitro upon suppression of PRR or RAC1. Furthermore, the inhibition of PRR could significantly down-regulate the expressions of RAC1 and NOX4 but not that of NOX2, while the inhibition of RAC1 did not affect PRR. SIGNIFICANCE Our findings showed that PRR inhibition could weaken RAC1-NOX4 pathway and alleviate DOX-induced HF via decreasing ROS production, thereby suggesting a promising target for the treatment of DOX-induced HF.
Collapse
Affiliation(s)
- Xiao-yi Du
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Pediatrics, Maternal and Child Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dao-chun Xiang
- Department of Pharmacy, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Gao
- Department of Clinical Pharmacy, Wuhan Children’s Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hua Peng
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ya-li Liu
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
16
|
Zhang Q, Wu Y, Yu Y, Niu Y, Fang Q, Chen X, Qi J, Zhang C, Wu G, Su K, Chai R. Tetrandrine Prevents Neomycin-Induced Ototoxicity by Promoting Steroid Biosynthesis. Front Bioeng Biotechnol 2022; 10:876237. [PMID: 35519614 PMCID: PMC9065337 DOI: 10.3389/fbioe.2022.876237] [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: 02/15/2022] [Accepted: 03/18/2022] [Indexed: 11/23/2022] Open
Abstract
Aminoglycoside antibiotics are widely used for the treatment of serious acute infections, life-threatening sepsis, and tuberculosis, but all aminoglycosides cause side effects, especially irreversible ototoxicity. The mechanisms underlying the ototoxicity of aminoglycosides need further investigation, and there are no effective drugs in the clinic. Here we showed that tetrandrine (TET), a bioactive bisbenzylisoquinoline alkaloid derived from Stephania tetrandra, ameliorated neomycin-induced cochlear hair cell injury. In both in vitro and in vivo experiments we found that TET administration significantly improved auditory function and reduced hair cell damage after neomycin exposure. In addition, we observed that TET could significantly decrease oxidative stress and apoptosis in hair cells after neomycin exposure. Finally, RNA-seq analysis suggested that TET protected against neomycin-induced ototoxicity mainly by promoting steroid biosynthesis. Collectively, our results provide pharmacological evidence showing that TET may be a promising agent in preventing aminoglycosides-induced ototoxicity.
Collapse
Affiliation(s)
- Qilei Zhang
- The Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, China
| | - Yunhao Wu
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, China
- *Correspondence: Yunhao Wu, ; Geping Wu, ; Kaiming Su, ; Renjie Chai,
| | - Yan Yu
- The Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, China
| | - Yuguang Niu
- Department of Ambulatory Medicine, the First Medical Center of PLA General Hospital, Beijing, China
| | - Qiaojun Fang
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, China
| | - Xin Chen
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, China
| | - Jieyu Qi
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, China
| | - Chen Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Geping Wu
- The Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, China
- *Correspondence: Yunhao Wu, ; Geping Wu, ; Kaiming Su, ; Renjie Chai,
| | - Kaiming Su
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
- *Correspondence: Yunhao Wu, ; Geping Wu, ; Kaiming Su, ; Renjie Chai,
| | - Renjie Chai
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, China
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Science, Beijing, China
- Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
- *Correspondence: Yunhao Wu, ; Geping Wu, ; Kaiming Su, ; Renjie Chai,
| |
Collapse
|
17
|
Cardiac SIRT1 ameliorates doxorubicin-induced cardiotoxicity by targeting sestrin 2. Redox Biol 2022; 52:102310. [PMID: 35452917 PMCID: PMC9043985 DOI: 10.1016/j.redox.2022.102310] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/29/2022] [Accepted: 04/01/2022] [Indexed: 01/19/2023] Open
Abstract
Although it is known that the expression and activity of sirtuin 1 (SIRT1) significantly decrease in doxorubicin (DOX)-induced cardiomyopathy, the role of interaction between SIRT1 and sestrin 2 (SESN2) is largely unknown. In this study, we investigated whether SESN2 could be a crucial target of SIRT1 and the effect of their regulatory interaction and mechanism on DOX-induced cardiac injury. Here, using DOX-treated cardiomyocytes and cardiac-specific Sirt1 knockout mice models, we found SIRT1 deficiency aggravated DOX-induced cardiac structural abnormalities and dysfunction, whereas the activation of SIRT1 by resveratrol (RES) treatment or SIRT1 overexpression possessed cardiac protective effects. Further studies indicated that SIRT1 exerted these beneficial effects by markedly attenuating DOX-induced oxidative damage and apoptosis in a SESN2-dependent manner. Knockdown of Sesn2 impaired RES/SIRT1-mediated protective effects, while upregulation of SESN2 efficiently rescued DOX-induced oxidative damage and apoptosis. Most importantly, SIRT1 activation could reduce DOX-induced SESN2 ubiquitination possibly through reducing the interaction of SESN2 with mouse double minute 2 (MDM2). The recovery of SESN2 stability in DOX-impaired primary cardiomyocytes by SIRT1 was confirmed by Mdm2-siRNA transfection. Taken together, our findings indicate that disrupting the interaction between SESN2 and MDM2 by SIRT1 to reduce the ubiquitination of SESN2 is a novel regulatory mechanism for protecting hearts from DOX-induced cardiotoxicity and suggest that the activation of SIRT1-SESN2 axis has potential as a therapeutic approach to prevent DOX-induced cardiotoxicity.
Collapse
|
18
|
Kitakata H, Endo J, Ikura H, Moriyama H, Shirakawa K, Katsumata Y, Sano M. Therapeutic Targets for DOX-Induced Cardiomyopathy: Role of Apoptosis vs. Ferroptosis. Int J Mol Sci 2022; 23:1414. [PMID: 35163335 PMCID: PMC8835899 DOI: 10.3390/ijms23031414] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/20/2022] [Accepted: 01/24/2022] [Indexed: 01/04/2023] Open
Abstract
Doxorubicin (DOX) is the most widely used anthracycline anticancer agent; however, its cardiotoxicity limits its clinical efficacy. Numerous studies have elucidated the mechanisms underlying DOX-induced cardiotoxicity, wherein apoptosis has been reported as the most common final step leading to cardiomyocyte death. However, in the past two years, the involvement of ferroptosis, a novel programmed cell death, has been proposed. The purpose of this review is to summarize the historical background that led to each form of cell death, focusing on DOX-induced cardiotoxicity and the molecular mechanisms that trigger each form of cell death. Furthermore, based on this understanding, possible therapeutic strategies to prevent DOX cardiotoxicity are outlined. DNA damage, oxidative stress, intracellular signaling, transcription factors, epigenetic regulators, autophagy, and metabolic inflammation are important factors in the molecular mechanisms of DOX-induced cardiomyocyte apoptosis. Conversely, the accumulation of lipid peroxides, iron ion accumulation, and decreased expression of glutathione and glutathione peroxidase 4 are important in ferroptosis. In both cascades, the mitochondria are an important site of DOX cardiotoxicity. The last part of this review focuses on the significance of the disruption of mitochondrial homeostasis in DOX cardiotoxicity.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Motoaki Sano
- Department of Cardiology, Keio University School of Medicine, Tokyo 160-8582, Japan; (H.K.); (J.E.); (H.I.); (H.M.); (K.S.); (Y.K.)
| |
Collapse
|
19
|
Eisvand F, Imenshahidi M, Ghasemzadeh Rahbardar M, Tabatabaei Yazdi SA, Rameshrad M, Razavi BM, Hosseinzadeh H. Cardioprotective effects of alpha‐mangostin on doxorubicin‐induced cardiotoxicity in rats. Phytother Res 2021; 36:506-524. [DOI: 10.1002/ptr.7356] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 11/15/2021] [Accepted: 11/29/2021] [Indexed: 12/11/2022]
Affiliation(s)
- Farhad Eisvand
- Department of Pharmacodynamics and Toxicology, School of Pharmacy Mashhad University of Medical Sciences Mashhad Iran
| | - Mohsen Imenshahidi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy Mashhad University of Medical Sciences Mashhad Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute Mashhad University of Medical Sciences Mashhad Iran
| | | | | | - Maryam Rameshrad
- Natural Products and Medicinal Plants Research Center North Khorasan University of Medical Sciences Bojnurd Iran
| | - Bibi Marjan Razavi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy Mashhad University of Medical Sciences Mashhad Iran
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute Mashhad University of Medical Sciences Mashhad Iran
| | - Hossein Hosseinzadeh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy Mashhad University of Medical Sciences Mashhad Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute Mashhad University of Medical Sciences Mashhad Iran
| |
Collapse
|
20
|
Lin KH, Wei YM, Liu CH, Liu JS, Huang IC, Viswanadha VP, Huang CY, Kuo WW. Diallyl Trisulfide Suppresses High-Glucose-Induced Cardiomyocyte Apoptosis by Targeting Reactive Oxygen Species-Mediated Hypoxia-Inducible Factor-1α/Insulin-like Growth Factor Binding Protein 3 Activation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:11696-11708. [PMID: 34558885 DOI: 10.1021/acs.jafc.1c02384] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
It has been reported that 80% of diabetic patients die due to cardiovascular diseases. We previously demonstrated that activated hypoxia-inducible factor-1α (HIF-1 α)/insulin-like growth factor binding protein-3 (IGFBP-3) signaling by reactive oxygen species (ROS)-regulated prolyl hydroxylase domain-containing protein (PHD) is involved in high-glucose (HG)-induced cardiac apoptosis. Diallyl trisulfide (DATS), a garlic component, shows the strongest inhibitory effect on diabetic cardiomyopathy. In this study, we investigated whether HIF-1α/IGFBP-3 signaling governs the antiapoptotic effect by DATS on HG-exposed cardiomyocytes. It was observed that significantly increased levels of cell apoptosis and decreased Akt phosphorylation were reversed by DATS in HG-exposed cardiac cells. H2O2 and PHD small interfering RNA treatments increased HIF-1α and IGFBP-3 protein levels, which were decreased by DATS treatment. Overexpression of HIF-1α and IGFBP-3 increased HG-induced cell apoptosis, which was suppressed by DATS. The coimmunoprecipitation assay results showed that DATS not only increased the IGF-1 level and reduced IGFBP-3 level but also suppressed their extracellular association for cardiac cells exposed to HG. Experiments using neonatal cardiomyocytes and hearts showed similar results. These findings indicate that the effect of ROS-regulated PHD on the activation of HIF-1α/IGFBP-3 signaling governs the antiapoptotic effect by DATS on HG-exposed cardiomyocytes.
Collapse
Affiliation(s)
- Kuan-Ho Lin
- College of Medicine, China Medical University, Taichung 40402, Taiwan
- Department of Emergency Medicine, China Medical University Hospital, Taichung 40402, Taiwan
| | - Yu-Min Wei
- Department of Biological Science and Technology, College of Life Sciences, China Medical University, 100, Sec. 1, Jingmao Road, Beitun District, Taichung 404, Taiwan
| | - Chung-Hung Liu
- Department of Biological Science and Technology, College of Life Sciences, China Medical University, 100, Sec. 1, Jingmao Road, Beitun District, Taichung 404, Taiwan
| | - Jian-Sheng Liu
- Department of Biological Science and Technology, College of Life Sciences, China Medical University, 100, Sec. 1, Jingmao Road, Beitun District, Taichung 404, Taiwan
- China Medical University, Thoracic Department, Beigang Hospital, Yunlin 651, Taiwan
| | - I-Chieh Huang
- Department of Biological Science and Technology, College of Life Sciences, China Medical University, 100, Sec. 1, Jingmao Road, Beitun District, Taichung 404, Taiwan
| | | | - Chih-Yang Huang
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, 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
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung 413, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404, Taiwan
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, College of Life Sciences, China Medical University, 100, Sec. 1, Jingmao Road, Beitun District, Taichung 404, Taiwan
- Ph.D. Program for Biotechnology Industry, China Medical University, Taichung 406, Taiwan
| |
Collapse
|
21
|
Cheng D, Tu W, Chen L, Wang H, Wang Q, Liu H, Zhu N, Fang W, Yu Q. MSCs enhances the protective effects of valsartan on attenuating the doxorubicin-induced myocardial injury via AngII/NOX/ROS/MAPK signaling pathway. Aging (Albany NY) 2021; 13:22556-22570. [PMID: 34587120 PMCID: PMC8507274 DOI: 10.18632/aging.203569] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 08/17/2021] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To verify if AngII/NOX/ROS/MAPK signaling pathway is involved in Doxorubicin (DOX)-induced myocardial injury and if mesenchymal stem cells (MSCs) could enhance the protective effects of valsartan (Val) on attenuating DOX-induced injury in vitro. METHODS Reactive oxygen species (ROS) formation and the protein expression of AT1R, NOX2, NOX4, caspase-3, caspase-9 and MAPK signaling were assessed in H9c2 cardiomyocytes exposed to DOX for 24 h in the absence or presence of Val, NADPH oxidase inhibitor DPI or knockdown and overexpression of NADPH oxidase subunit: NOX2 and NOX4, co-culture with MSCs, respectively. Finally, MTT assay was used to determine the cell viability of H9c2 cells, MDA-MB-231 breast cancer cells and A549 pulmonary cancer cells under Val, DOX and Val+ DOX treatments. RESULTS DOX increased ROS formation and upregulated proteins expression of AT1R, NOX2, NOX4, caspase-3, caspase-9 and MAPK signaling including p-p38, p-JNK, p-ERK in H9c2 cells. These effects could be attenuated by Val, DPI, NOX2 siRNA and NOX4 siRNA. Meanwhile, overexpression of NOX2 and NOX4 could significantly increase DOX-induced ROS formation and further upregulate apoptotic protein expressions and protein expressions of MAPK signaling. MSCs on top of Val further enhanced the protective effects of Val on reducing the DOX-induced ROS formation and downregulating the expression of apoptotic proteins and MAPK signaling as compared with Val alone in DOX-treated H9c2 cells. Simultaneous Val and DOX treatment did not affect cell viability of DOX-treated MDA-MB-231 breast cancer cells or A549 pulmonary cancer cells but significantly improved cell viability of DOX-treated H9c2 cardiomyocytes. CONCLUSIONS AT1R/NOX/ROS/MAPK signaling pathway is involved in DOX-induced cardiotoxicity. Val treatment significantly attenuated DOX-induced cardiotoxicity, without affecting the anti-tumor effect of DOX. MSCs enhance the protective effects of Val on reducing the DOX-induced toxicity in H9c2 cells.
Collapse
Affiliation(s)
- Dong Cheng
- Department of Cardiology, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, Liaoning, China
- Medical College, Dalian University, Dalian 116622, Liaoning, China
| | - Wencheng Tu
- Medical College, Dalian University, Dalian 116622, Liaoning, China
- Department of Cardiology, Jingmen No.1 People’s Hospital, Jingmen 448000, Hubei, China
| | - Libo Chen
- Department of Cardiology, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, Liaoning, China
- Department of Cardiology, People’s Hospital of Jilin City, Jilin 132000, Jilin, China
| | - Haoren Wang
- Central Laboratory, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, Liaoning, China
| | - Qinfu Wang
- Life Engineering College, Dalian University, Dalian 116622, Liaoning, China
| | - Hainiang Liu
- Department of Cardiology, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, Liaoning, China
- Medical College, Dalian University, Dalian 116622, Liaoning, China
| | - Ning Zhu
- Department of Cardiology, The Second Affiliated Hospital of Dalian Medical University, Dalian 116023, Liaoning, China
| | - Weiyi Fang
- Department of Cardiology, Shanghai Chest Hospital, Changning 200030, Shanghai, China
| | - Qin Yu
- Department of Cardiology, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, Liaoning, China
| |
Collapse
|
22
|
Lin KH, Ng SC, Paul CR, Chen HC, Zeng RY, Liu JS, Padma VV, Huang CY, Kuo WW. MicroRNA-210 repression facilitates advanced glycation end-product (AGE)-induced cardiac mitochondrial dysfunction and apoptosis via JNK activation. J Cell Biochem 2021; 122:1873-1885. [PMID: 34545968 DOI: 10.1002/jcb.30146] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 08/31/2021] [Accepted: 09/02/2021] [Indexed: 12/12/2022]
Abstract
Hyperglycemia results in the formation of reactive oxygen species which in turn causes advanced glycation end products (AGEs) formation, leading to diabetic cardiomyopathy. Our previous study showed that AGE-induced reactive oxygen species-dependent apoptosis is mediated via protein kinase C delta (PKCδ)-enhanced mitochondrial damage in cardiomyocytes. By using microRNA (miRNA) database, miRNA-210 was predicted to target c-Jun N-terminal kinase (JNK), which were previously identified as downstream of PKCδ in regulating mitochondrial function. Therefore, we hypothesized that miR-210 mediates PKCδ-dependent upregulation of JNK to cause cardiac mitochondrial damage and apoptosis following AGE exposure. AGE-exposed cells showed activated cardiac JNK, PKCδ, and apoptosis, which were reversed by treatment with a JNK inhibitor and PKCδ-KD (deficient kinase). Cardiac miR-210 and mitochondrial function were downregulated following AGE exposure. Furthermore, JNK was upregulated and involved in AGE-induced mitochondrial damage. Interestingly, luciferase activity of the miR-210 mimic plus JNK WT-3'-untranslated region overexpressed group was significantly lower than that of miR-210 mimic plus JNK MT-3'UTR group, indicating that JNK is a target of miR-210. Moreover, JNK activation induced by AGEs was reduced by treatment with the miR-210 mimic and reversed by treatment with the miR-210 inhibitor, indicating the regulatory function of miR-210 in JNK activation following AGE exposure. Additionally, JNK-dependent mitochondrial dysfunction and apoptosis were reversed following treatment with the miR-210 mimic, while the miR-210 inhibitor showed no effect on JNK-induced mitochondrial dysfunction and apoptosis in AGE-exposed cardiac cells. Taken together, our study showed that PKCδ-enhanced JNK-dependent mitochondrial damage is mediated through the reduction of miR-210 in cardiomyocytes following AGE exposure.
Collapse
Affiliation(s)
- Kuan-Ho Lin
- College of Medicine, China Medical University, Taichung, Taiwan, ROC.,Department of Emergency Medicine, China Medical University Hospital, Taichung, Taiwan, ROC
| | - Shang-Chuan Ng
- Department of Biological Science and Technology, College of Life Sciences, China Medical University, Taichung, Taiwan, ROC.,PhD Program for Biotechnology Industry, China Medical University, Taichung, Taiwan, ROC
| | - Catherine R Paul
- Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien, Taiwan, ROC
| | - Hong-Chen Chen
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan, ROC.,Cancer Progression Research Center, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Ren-You Zeng
- Department of Biological Science and Technology, College of Life Sciences, China Medical University, Taichung, Taiwan, ROC.,PhD Program for Biotechnology Industry, China Medical University, Taichung, Taiwan, ROC
| | - Jian-Sheng Liu
- Department of Biological Science and Technology, College of Life Sciences, China Medical University, Taichung, Taiwan, ROC.,Department of Thoracic, China Medical University Beigang Hospital, Yunlin, Taiwan, ROC
| | - Viswanadha V Padma
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan, ROC
| | - Chih-Yang Huang
- Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien, Taiwan, ROC.,Department of Biotechnology, Translational Research Laboratory, School of Biotechnology and Genetic Engineering, Bharathiar University, Coimbatore, Tamil Nadu, India.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, ROC.,Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan, ROC.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan, ROC
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, College of Life Sciences, China Medical University, Taichung, Taiwan, ROC.,PhD Program for Biotechnology Industry, China Medical University, Taichung, Taiwan, ROC
| |
Collapse
|
23
|
Zheng D, Wang J, Li G, Sun Y, Deng Q, Li M, Song K, Zhao Z. Preliminary therapeutic and mechanistic evaluation of S-allylmercapto-N-acetylcysteine in the treatment of pulmonary emphysema. Int Immunopharmacol 2021; 98:107913. [PMID: 34218218 DOI: 10.1016/j.intimp.2021.107913] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/10/2021] [Accepted: 06/20/2021] [Indexed: 12/24/2022]
Abstract
The objective of this work was to study the effects and mechanisms of S-allylmercapto-N-acetylcysteine (ASSNAC) in the treatment of pulmonary emphysema based on network pharmacology analysis and other techniques. Firstly, the potential targets associated with ASSNAC and COPD were integrated using public databases. Then, a protein-protein interaction network was constructed using String database and Cytoscape software. The Gene Ontology analysis and Kyoto Encyclopedia of Genes and Genomes pathway analysis were performed on DAVID platform. The molecular docking of ASSNAC with some key disease targets was implemented on the SwissDock platform. To verify the results of the network pharmacology, a pulmonary emphysema mice model was established and treated with ASSNAC. Besides, the expressions of the predicted targets were detected by immunohistochemistry, Western blot analysis or enzyme-linked immunosorbent assay. Results showed that 33 overlapping targets are achieved, including CXCL8, ICAM1, MAP2K1, PTGS2, ACE and so on. The critical pathways of ASSNAC against COPD involved arachidonic acid metabolism, chemokine pathway, MAPK pathway, renin-angiotensin system, and others. Pharmacodynamic experiments demonstrated that ASSNAC decreased the pulmonary emphysema and inflammation in the pulmonary emphysema mice. Therefore, these results confirm the perspective of network pharmacology in the target verification, and indicate the treatment potential of ASSNAC against COPD.
Collapse
Affiliation(s)
- Dandan Zheng
- Department of Pharmaceutics, Key Laboratory of Chemical Biology of Ministry of Education, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China
| | - Jinglong Wang
- College of Food Sciences and Pharmaceutical Engineering, Zaozhuang University, Zaozhuang 277160, PR China
| | - Genju Li
- Department of Pharmaceutics, Key Laboratory of Chemical Biology of Ministry of Education, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China
| | - Yueyue Sun
- Department of Pharmaceutics, Key Laboratory of Chemical Biology of Ministry of Education, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China
| | - Qi Deng
- Department of Pharmaceutics, Key Laboratory of Chemical Biology of Ministry of Education, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China
| | - Muhan Li
- Department of Pharmaceutics, Key Laboratory of Chemical Biology of Ministry of Education, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China
| | - Kaili Song
- Department of Pharmaceutics, Key Laboratory of Chemical Biology of Ministry of Education, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China
| | - Zhongxi Zhao
- Department of Pharmaceutics, Key Laboratory of Chemical Biology of Ministry of Education, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China; Key University Laboratory of Pharmaceutics & Drug Delivery Systems of Shandong Province, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China; Pediatric Pharmaceutical Engineering Laboratory of Shandong Province, Shandong Dyne Marine Biopharmaceutical Company Limited, Rongcheng, Shandong 264300, PR China; Chemical Immunopharmaceutical Engineering Laboratory of Shandong Province, Shandong Xili Pharmaceutical Company Limited, Heze, Shandong 274300, PR China.
| |
Collapse
|
24
|
Wu YT, Xie LP, Hua Y, Xu HL, Chen GH, Han X, Tan ZB, Fan HJ, Chen HM, Li J, Liu B, Zhou YC. Tanshinone I Inhibits Oxidative Stress-Induced Cardiomyocyte Injury by Modulating Nrf2 Signaling. Front Pharmacol 2021; 12:644116. [PMID: 34084132 PMCID: PMC8167655 DOI: 10.3389/fphar.2021.644116] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 03/22/2021] [Indexed: 12/29/2022] Open
Abstract
Cardiovascular disease, a disease caused by many pathogenic factors, is one of the most common causes of death worldwide, and oxidative stress plays a major role in its pathophysiology. Tanshinone I (Tan I), a natural compound with cardiovascular protective effects, is one of the main active compounds extracted from Salvia miltiorrhiza. Here, we investigated whether Tan I could attenuate oxidative stress and oxidative stress–induced cardiomyocyte apoptosis through Nrf2/MAPK signaling in vivo and in vitro. We found that Tan I treatment protected cardiomyocytes against oxidative stress and oxidative stress–induced apoptosis, based on the detection of relevant oxidation indexes such as reactive oxygen species, superoxide dismutase, malondialdehyde, and apoptosis, including cell viability and apoptosis-related protein expression. We further examined the mechanisms underlying these effects, determining that Tan I activated nuclear factor erythroid 2 (NFE2)–related factor 2 (Nrf2) transcription into the nucleus and dose-dependently promoted the expression of Nrf2, while inhibiting MAPK signaling activation, including P38 MAPK, SAPK/JNK, and ERK1/2. Nrf2 inhibitors in H9C2 cells and Nrf2 knockout mice demonstrated aggravated oxidative stress and oxidative stress–induced cardiomyocyte injury; Tan I treatment suppressed these effects in H9C2 cells; however, its protective effect was inhibited in Nrf2 knockout mice. Additionally, the analysis of surface plasmon resonance demonstrated that Tan I could directly target Nrf2 and act as a potential Nrf2 agonist. Collectively, these data strongly indicated that Tan I might inhibit oxidative stress and oxidative stress–induced cardiomyocyte injury through modulation of Nrf2 signaling, thus supporting the potential therapeutic application of Tan I for oxidative stress–induced CVDs.
Collapse
Affiliation(s)
- Yu-Ting Wu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,Department of Traditional Chinese Medicine, Binzhou Medical University Hospital, Binzhou, China
| | - Ling-Peng Xie
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Traditional Chinese Medicine, Nanfang Hospital (Zengcheng Branch), Southern Medical University, Guangzhou, China
| | - Yue Hua
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Traditional Chinese Medicine, Nanfang Hospital (Zengcheng Branch), Southern Medical University, Guangzhou, China
| | - Hong-Lin Xu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Traditional Chinese Medicine, Nanfang Hospital (Zengcheng Branch), Southern Medical University, Guangzhou, China
| | - Guang-Hong Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Traditional Chinese Medicine, Nanfang Hospital (Zengcheng Branch), Southern Medical University, Guangzhou, China
| | - Xin Han
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Traditional Chinese Medicine, Nanfang Hospital (Zengcheng Branch), Southern Medical University, Guangzhou, China
| | - Zhang-Bin Tan
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hui-Jie Fan
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,TCM Health Construction Department of Yangjiang People's Hospital, Yangjiang, China
| | - Hong-Mei Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Traditional Chinese Medicine, Nanfang Hospital (Zengcheng Branch), Southern Medical University, Guangzhou, China
| | - Jun Li
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Bin Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ying-Chun Zhou
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Traditional Chinese Medicine, Nanfang Hospital (Zengcheng Branch), Southern Medical University, Guangzhou, China
| |
Collapse
|
25
|
Limam I, Ben Aissa-Fennira F, Essid R, Chahbi A, Kefi S, Mkadmini K, Elkahoui S, Abdelkarim M. Hydromethanolic root and aerial part extracts from Echium arenarium Guss suppress proliferation and induce apoptosis of multiple myeloma cells through mitochondrial pathway. ENVIRONMENTAL TOXICOLOGY 2021; 36:874-886. [PMID: 33393729 DOI: 10.1002/tox.23090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 12/19/2020] [Indexed: 06/12/2023]
Abstract
Echium arenarium Guss is a Mediterranean plant traditionally used in healing skin wound and it was reported exhibiting potent antioxidant, antibacterial, and antiparasitic activities. However, antitumoral activities of this plant have not yet been explored. Here we investigated for the first time, root (EARE) and aerial part (EAAPE) extracts of E. arenarium Guss to examine cytotoxicity and apoptosis activation pathway on U266 human multiple myeloma (MM) cell line. We demonstrated that EARE and EAAPE decreased U266 cell viability in a dose dependent manner. Based on 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, EARE was significantly two times more efficient (IC50 value 41 μg/ml) than EAAPE (IC50 value 82 μg/ml) considering 48 h of treatment. Furthermore, after 24 h of exposure to 100 μg/ml of EARE or EAAPE, cell cycle showed remarkable increase in sub-G1 population and a decrease of U266 cells proportion in G1 phase. In addition, EARE increased cell percentage in S phase. Moreover, analysis revealed that EAAPE or EARE induced apoptosis of U266 cells after 24 h of treatment. Interestingly, depolarization of mitochondrial membrane potential and activation of caspase 3/7 were demonstrated in treated U266 cells. Phytochemical analysis of E. arenarium extracts showed that EARE exhibited the highest content of total phenolic content. Interestingly, six phenolic compounds were identified. Myricitrin was the major compound in EARE, followed by luteolin 7-O-glucoside, resorcinol, polydatin, Trans-hydroxycinnamic acid, and hyperoside. These findings proved that an intrinsic mitochondria-mediated apoptosis pathway probably mediated the apoptotic effects of E. arenarium Guss extracts on U266 cells, and this will suggest several action plans to treat MM.
Collapse
Affiliation(s)
- Inès Limam
- Laboratory of oncohematology, Faculty of medicine of Tunis, Tunis El Manar University, Tunis, Tunisia
| | - Fatma Ben Aissa-Fennira
- Laboratory of oncohematology, Faculty of medicine of Tunis, Tunis El Manar University, Tunis, Tunisia
| | - Rim Essid
- Laboratory of Bioactive Substances, Center of Biotechnology, Ecopark of Borj Cedria, Hammam-Lif, Tunisia
| | - Ahlem Chahbi
- Laboratory of oncohematology, Faculty of medicine of Tunis, Tunis El Manar University, Tunis, Tunisia
| | - Sarra Kefi
- Laboratory of Bioactive Substances, Center of Biotechnology, Ecopark of Borj Cedria, Hammam-Lif, Tunisia
| | - Khaoula Mkadmini
- Medicinal and Aromatic Plants Laboratory, Biotechnology Center of Borj Cedria, Hammam-Lif, Tunisia
| | - Salem Elkahoui
- Laboratory of Bioactive Substances, Center of Biotechnology, Ecopark of Borj Cedria, Hammam-Lif, Tunisia
- Department of Biology, College of Science, University of Hail, Hail, Saudi Arabia
| | - Mohamed Abdelkarim
- Laboratory of oncohematology, Faculty of medicine of Tunis, Tunis El Manar University, Tunis, Tunisia
| |
Collapse
|
26
|
Geng H, Guo W, Feng L, Xie D, Bi L, Wang Y, Zhang T, Liang Z, Yu D. Diallyl trisulfide inhibited tobacco smoke-mediated bladder EMT and cancer stem cell marker expression via the NF-κB pathway in vivo. J Int Med Res 2021; 49:300060521992900. [PMID: 33730908 PMCID: PMC8166398 DOI: 10.1177/0300060521992900] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 12/04/2020] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE This study examined the effect of the NF-κB pathway on tobacco smoke-elicited bladder epithelial-mesenchymal transition (EMT) and cancer stem cell (CSC) marker expression in vivo. The effect of diallyl trisulfide (DATS) treatment was also examined. METHODS BALB/c mice were exposed to tobacco smoke and treated with an NF-κB inhibitor and DATS. Western blotting, quantitative real-time PCR, and immunohistochemical staining were used to detect the changes of relevant indices. RESULTS Phosphorylated inhibitor of kappa-B kinase alpha/beta expression and p65 and p50 nuclear transcription were increased by tobacco smoke exposure, whereas inhibitor of kappa-B expression was decreased. In addition, tobacco smoke reduced the expression of epithelial markers but increased that of mesenchymal and CSC markers. Our study further demonstrated that tobacco smoke-mediated EMT and CSC marker expression were attenuated by inhibition of the NF-κB pathway. Moreover, DATS reversed tobacco smoke-induced NF-κB pathway activation, EMT, and the acquisition of CSC properties in bladder tissues. CONCLUSIONS These data suggested that the NF-κB pathway regulated tobacco smoke-induced bladder EMT, CSC marker expression, and the protective effects of DATS.
Collapse
Affiliation(s)
- Hao Geng
- Department of Urology, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wenhao Guo
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Lei Feng
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Dongdong Xie
- Department of Urology, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Liangkuan Bi
- Department of Urology, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yi Wang
- Department of Urology, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Tao Zhang
- Department of Urology, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhaofeng Liang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Dexin Yu
- Department of Urology, Second Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
27
|
Tursunova NV, Klinnikova MG, Babenko OA, Lushnikova EL. [Molecular mechanisms of the cardiotoxic action of anthracycline antibiotics and statin-induced cytoprotective reactions of cardiomyocytes]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2021; 66:357-371. [PMID: 33140729 DOI: 10.18097/pbmc20206605357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The manifestation of the side cardiotoxic effect of anthracycline antibiotics limits their use in the treatment of malignant processes in some patients. The review analyzes the main causes of the susceptibility of cardiomyocytes to the damaging effect of anthracyclines, primarily associated with an increase in the processes of free radical oxidation. Currently, research is widely carried out to find ways to reduce anthracycline cardiotoxicity, in particular, the use of cardioprotective agents in the complex treatment of tumors. Hydroxymethylglutaryl coenzyme A reductase inhibitors (statins) have been shown to improve the function and metabolism of the cardiovascular system under various pathological impacts, therefore, it is proposed to use them to reduce cardiotoxic complications of chemotherapy. Statins exhibit direct (hypolipidemic) and pleiotropic effects due to the blockade of mevalonic acid synthesis and downward biochemical cascades that determine their cardioprotective properties. The main point of intersection of the pharmacological activity of anthracyclines and statins is the ability of both to regulate the functioning of small GTPase from the Rho family, and their effect in this regard is the opposite. The influence of statins on the modification and membrane dislocation of Rho proteins mediates the indirect antioxidant, anti-inflammatory, endothelioprotective, antiapoptotic effect. The mechanism of statin inhibition of doxorubicin blockade of the DNA-topoisomerase complex, which may be important in preventing cardiotoxic damage during chemotherapy, is discussed. At the same time, it should be noted that the use of statins can be accompanied by adverse side effects: a provocation of increased insulin resistance and glucose tolerance, which often causes them to be canceled in patients with impaired carbohydrate metabolism, so further studies are needed here. The review also analyzes data on the antitumor effect of statins, their ability to sensitize the tumor to treatment with cytostatic drug. It has been shown that the relationship between anthracycline antibiotics and statins is characterized not only by antagonism, but also in some cases by synergism. Despite some adverse effects, statins are one of the most promising cardio- and vasoprotectors for use in anthracycline cardiomyopathy.
Collapse
Affiliation(s)
- N V Tursunova
- Institute of Molecular Pathology and Pathomorphology, Federal Research Center of Fundamental and Translational Medicine, Novosibirsk, Russia
| | - M G Klinnikova
- Institute of Molecular Pathology and Pathomorphology, Federal Research Center of Fundamental and Translational Medicine, Novosibirsk, Russia
| | - O A Babenko
- Institute of Molecular Pathology and Pathomorphology, Federal Research Center of Fundamental and Translational Medicine, Novosibirsk, Russia
| | - E L Lushnikova
- Institute of Molecular Pathology and Pathomorphology, Federal Research Center of Fundamental and Translational Medicine, Novosibirsk, Russia
| |
Collapse
|
28
|
Li Y, Chandra TP, Song X, Nie L, Liu M, Yi J, Zheng X, Chu C, Yang J. H2S improves doxorubicin-induced myocardial fibrosis by inhibiting oxidative stress and apoptosis via Keap1-Nrf2. Technol Health Care 2021; 29:195-209. [PMID: 33682759 PMCID: PMC8150551 DOI: 10.3233/thc-218020] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE We waimed to investigate whether H2S can relieve the myocardial fibrosis caused by doxorubicin through Keap1-Nrf2. METHODS Sprague-Dawley (SD) rats were randomly divided into four groups: normal control group (Control); DOX model group (DOX); H2S intervention model group (DOX+H2S); H2S control group (H2S). DOX and DOX+H2S group were injected with doxorubicin (3.0 mg/kg/time) intraperitoneally. Both of the Control group and H2S groups were given normal saline in equal volume, 2 weeks later, DOX+H2S and H2S group were controlled with NaHS (56 μmol/kg/d) through the abdominal cavity, while the Control and DOX group were injected with normal saline of the same dosage intraperitoneally. RESULTS Myocardial injury and myocardial cell apoptosis were significantly increased, the H2S content in myocardial tissue was remarkably down-regulated, the expression levels of MDA, Keap1, caspase-3, caspase-9, TNF-α, IL1β, MMPs and TIMP-1 in rat myocardial tissue was significantly up-regulated (P< 0.05), and the expression levels of GSH, NQO1, Bcl-2 were down-regulated compared with those of control group. The above results can be reversed by the DOX+H2S group. There is no statistically significant difference between the Control group and the H2S control group. CONCLUSIONS These results suggest that H2S can improve DOX-induced myocardial fibrosis in rats, and the keap1/Nrf2 signaling pathway, oxidative stress, inflammation, and apoptosis may be involved in the mechanism.
Collapse
Affiliation(s)
- Yaling Li
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, China
| | - Thakur Prakash Chandra
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, China
| | - Xiong Song
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, China
| | - Liangui Nie
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, China
| | - Maojun Liu
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, China
| | - Jiali Yi
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, China
| | - Xia Zheng
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, China
| | - Chun Chu
- Department of Pharmacy, The Second Affiliated Hospital of University of South China, Hengyang, Hunan 421001, China
| | - Jun Yang
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, China
| |
Collapse
|
29
|
Xie J, Liao B, Tang RY. Functional Application of Sulfur-Containing Spice Compounds. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:12505-12526. [PMID: 33138361 DOI: 10.1021/acs.jafc.0c05002] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Sulfur-containing spice compounds possess diverse biological functions and play an important role in food, chemicals, pharmaceuticals, and agriculture. The development of functional spices has become increasingly popular, especially for medicinal functions for dietary health. Thus, this review focuses on the properties and functions of sulfur-containing spice compounds, including antioxidant, anti-inflammatory, antiobesity, anticancer, antibacterial, and insecticidal functions, among others. Developments over the last five years concerning the properties of sulfur-containing spice compounds are summarized and discussed.
Collapse
Affiliation(s)
- Jinxin Xie
- Department of Applied Chemistry, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
| | - Benjian Liao
- Department of Applied Chemistry, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
| | - Ri-Yuan Tang
- Department of Applied Chemistry, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Natural Pesticide & Chemical Biology, Ministry of Education, South China Agricultural University, Guangzhou 510642, China
- Lingnan Guangdong Laboratory of Modern Agriculture, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
30
|
Piragine E, Calderone V. Pharmacological modulation of the hydrogen sulfide (H 2 S) system by dietary H 2 S-donors: A novel promising strategy in the prevention and treatment of type 2 diabetes mellitus. Phytother Res 2020; 35:1817-1846. [PMID: 33118671 DOI: 10.1002/ptr.6923] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/15/2020] [Accepted: 10/06/2020] [Indexed: 12/25/2022]
Abstract
Type 2 diabetes mellitus (T2DM) represents the most common age-related metabolic disorder, and its management is becoming both a health and economic issue worldwide. Moreover, chronic hyperglycemia represents one of the main risk factors for cardiovascular complications. In the last years, the emerging evidence about the role of the endogenous gasotransmitter hydrogen sulfide (H2 S) in the pathogenesis and progression of T2DM led to increasing interest in the pharmacological modulation of endogenous "H2 S-system". Indeed, H2 S directly contributes to the homeostatic maintenance of blood glucose levels; moreover, it improves impaired angiogenesis and endothelial dysfunction under hyperglycemic conditions. Moreover, H2 S promotes significant antioxidant, anti-inflammatory, and antiapoptotic effects, thus preventing hyperglycemia-induced vascular damage, diabetic nephropathy, and cardiomyopathy. Therefore, H2 S-releasing molecules represent a promising strategy in both clinical management of T2DM and prevention of macro- and micro-vascular complications associated to hyperglycemia. Recently, growing attention has been focused on dietary organosulfur compounds. Among them, garlic polysulfides and isothiocyanates deriving from Brassicaceae have been recognized as H2 S-donors of great pharmacological and nutraceutical interest. Therefore, a better understanding of the therapeutic potential of naturally occurring H2 S-donors may pave the way to a more rational use of these nutraceuticals in the modulation of H2 S homeostasis in T2DM.
Collapse
Affiliation(s)
| | - Vincenzo Calderone
- Department of Pharmacy, University of Pisa, Pisa, Italy.,Interdepartmental Research Center Nutrafood "Nutraceuticals and Food for Health", University of Pisa, Pisa, Italy.,Interdepartmental Research Centre of Ageing Biology and Pathology, University of Pisa, Pisa, Italy
| |
Collapse
|
31
|
Hao Y, Liu HM, Wei X, Gong X, Lu ZY, Huang ZH. Diallyl trisulfide attenuates hyperglycemia-induced endothelial apoptosis by inhibition of Drp1-mediated mitochondrial fission. Acta Diabetol 2019; 56:1177-1189. [PMID: 31115753 PMCID: PMC6768919 DOI: 10.1007/s00592-019-01366-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 05/07/2019] [Indexed: 02/07/2023]
Abstract
AIMS Hyperglycemia induces endothelial cell apoptosis and blood vessel damage, while diallyl trisulfide (DATS) has shown cardiovascular protection in animal models and humans. The aim of this study was to investigate the effects of DATS on inhibition of high glucose-induced endothelial cell apoptosis and the underlying molecular events. METHODS Human umbilical vein endothelial cells (HUVECs) were incubated with DATS (100 μM) for 30 min and then cultured in high-glucose medium (HG, 33 mM) for 24 h for assessment of apoptosis, glutathione (GSH), reactive oxygen species (ROS), superoxide dismutase (SOD), and gene expression using the terminal deoxyuridine triphosphate nick end labeling (TUNEL), flow cytometry, caspase-3 activity, ROS, SOD, and western blot assays as well as JC-1 and MitoTracker Red staining, respectively. RESULTS DATS treatment significantly inhibited high glucose-induced HUVEC apoptosis by blockage of intracellular and mitochondrial ROS generation, maintenance of the mitochondrial membrane potential, and suppression of high glucose-induced dynamin-related protein 1 (Drp1) expression. Furthermore, DATS blockage of high glucose-induced mitochondrial fission and apoptosis was through adenosine monophosphate-activated protein kinase (AMPK) activation-inhibited Drp1 expression in HUVECs. CONCLUSIONS DATS demonstrated the ability to inhibit high glucose-induced HUVEC apoptosis via suppression of Drp1-mediated mitochondrial fission in an AMPK-dependent fashion.
Collapse
Affiliation(s)
- Ying Hao
- Department of Cardiology, Shanghai East Hospital, Tongji University, 1800 Yuntai Road, Shanghai, 200126, China
| | - Hui-Min Liu
- Department of Hematology, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, China
| | - Xin Wei
- Department of Pharmacy, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Xue Gong
- Department of Cardiology, Delta Health Hospital, 109 Xule Road, Shanghai, 201702, China
| | - Zhao-Yang Lu
- Department of Cardiology, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, China.
| | - Zhen-Hao Huang
- Department of Cardiology, Shanghai East Hospital, Tongji University, 1800 Yuntai Road, Shanghai, 200126, China.
| |
Collapse
|
32
|
Xiao Q, Che X, Cai B, Tao Z, Zhang H, Shao Q, Pu J. Macrophage autophagy regulates mitochondria-mediated apoptosis and inhibits necrotic core formation in vulnerable plaques. J Cell Mol Med 2019; 24:260-275. [PMID: 31660692 PMCID: PMC6933382 DOI: 10.1111/jcmm.14715] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 08/27/2019] [Accepted: 08/31/2019] [Indexed: 12/19/2022] Open
Abstract
The vulnerable plaque is a key distinguishing feature of atherosclerotic lesions that can cause acute atherothrombotic vascular disease. This study was designed to explore the effect of autophagy on mitochondria-mediated macrophage apoptosis and vulnerable plaques. Here, we generated the mouse model of vulnerable carotid plaque in ApoE-/- mice. Application of ApoE-/- mice with rapamycin (an autophagy inducer) inhibited necrotic core formation in vulnerable plaques by decreasing macrophage apoptosis. However, 3-methyladenine (an autophagy inhibitor) promoted plaque vulnerability through deteriorating these indexes. To further explore the mechanism of autophagy on macrophage apoptosis, we used macrophage apoptosis model in vitro and found that 7-ketocholesterol (7-KC, one of the primary oxysterols in oxLDL) caused macrophage apoptosis with concomitant impairment of mitochondria, characterized by the impairment of mitochondrial ultrastructure, cytochrome c release, mitochondrial potential dissipation, mitochondrial fragmentation, excessive ROS generation and both caspase-9 and caspase-3 activation. Interestingly, such mitochondrial apoptotic responses were ameliorated by autophagy activator, but exacerbated by autophagy inhibitor. Finally, we found that MAPK-NF-κB signalling pathway was involved in autophagy modulation of 7-KC-induced macrophage apoptosis. So, we provide strong evidence for the potential therapeutic benefit of macrophage autophagy in regulating mitochondria-mediated apoptosis and inhibiting necrotic core formation in vulnerable plaques.
Collapse
Affiliation(s)
- Qingqing Xiao
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinyu Che
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Cai
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenyu Tao
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hengyuan Zhang
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qin Shao
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Pu
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
33
|
Sun J, Zhang L, Zhang Y, Yue CW, Lin J, Wang H, Fang ZJ, Wu J. Smart albumin-loaded Rose Bengal and doxorubicin nanoparticles for breast cancer therapy. J Microencapsul 2019; 36:728-737. [PMID: 31544561 DOI: 10.1080/02652048.2019.1671908] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Jing Sun
- Department of Pharmacy, The Second Hospital of Shandong University, Jinan, China
| | - Li Zhang
- Department of Pharmacy, The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Ying Zhang
- Department of Pharmacy, The Second Hospital of Shandong University, Jinan, China
| | - Chun-Wen Yue
- Department of Pharmacy, The Second Hospital of Shandong University, Jinan, China
| | - Jia Lin
- Department of Pharmacy, Shandong Provincial Hospital Group, Jinan, China
| | - Haisheng Wang
- Department of Pharmacy, The Second Hospital of Shandong University, Jinan, China
| | - Zeng-Jun Fang
- Department of Pharmacy, The Second Hospital of Shandong University, Jinan, China
| | - Jing Wu
- Department of Pharmacy, The Second Hospital of Shandong University, Jinan, China
| |
Collapse
|
34
|
Abstract
BACKGROUND AND OBJECTIVE Allitridin [diallyl trisulfide (DATS)] is an extract from garlic (Allium sativum) that putatively improves endothelial function and is protective against cardiovascular diseases. Endothelial dysfunction after tissue ischemia in diabetic patients is partially due to poor angiogenic response. This study investigated whether DATS may improve angiogenesis in a diabetic mouse model with hind limb ischemia. METHODS Streptozotocin was administered by intraperitoneal injection to establish the model of diabetes in male C57BL/6 mice. After 14 days, nondiabetic and diabetic mice (n = 24, each) underwent unilateral hind limb ischemia by femoral artery ligation. The mice were apportioned to 4 groups: nondiabetic treated (or not) with DATS and diabetic treated (or not) with DATS. DATS treatment consisted of a single daily intraperitoneal injection of 500 μg·kg·d for 14 days, beginning on the day of induced ischemia. Ischemia was scored by standard criteria. Blood perfusion was determined using thermal infrared imaging. Tissue capillary density and oxidative stress levels were measured by immunohistochemistry and immunofluorescence, respectively. Serum lipids were measured by enzymatic colorimetric assay. Fasting serum insulin was detected using an insulin enzyme-linked immunosorbent assay kit. Nitric oxide (NO) metabolites and protein carbonyls in tissues were determined by enzyme-linked immunosorbent assay. Targeted protein concentrations were measured by western blotting. RESULTS At 14 days after ligation, the ischemic skeletal muscle of the streptozotocin-induced diabetic mice had lower levels of endothelial NO synthase, phosphorylated endothelial NO synthase, and vascular endothelial growth factor compared with nondiabetic group. In addition, the hind limb blood perfusion, capillary density, and NO bioactivity were lower in the diabetic group, whereas oxidative stress and protein carbonyl levels were higher. These changes were ameliorated by DATS treatment. CONCLUSIONS DATS treatment of diabetic mice promoted revascularization in ischemic tissue.
Collapse
|
35
|
Liu Y, Jiang X, Gu Y, Chen Y. Preventive effect of Diallyl Trisulfide on cutaneous toxicities induced by EGFR inhibitor. Int Immunopharmacol 2019; 69:79-87. [PMID: 30682720 DOI: 10.1016/j.intimp.2019.01.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 01/09/2019] [Accepted: 01/15/2019] [Indexed: 01/29/2023]
Abstract
Cutaneous toxicities are the commonest side effects in patients with cancer treated using epidermal growth factor receptor inhibitors such as erlotinib. For patients with such toxicities, there is a lack of safe, effective pharmacological agents. Here we established a skin toxicity model and investigated the preventive and therapeutic effect of Diallyl Trisulfide (DATS) in vivo. The mouse skin toxicities model was established through continuous administration of erlotinib for 49 days. Meanwhile, the mice in the experimental group underwent DATS treatment for 49 days. Hematoxylin and eosin (HE) staining and oil red O staining of back and limb skin was performed to determine whether DATS aqueous extract can reverse the skin toxicities caused by erlotinib. Compared with the erlotinib group, the incidence of rash in the DATS group was lower. In addition, in the DATS group, the degree of skin redness and herpes was mild, the body weight was stable, and the activity was favorable. By comparing the HE and oil red O staining results for the mouse skin, the degree of keratin hyperplasia was determined to be lower in the experimental group than in the erlotinib group, and the number of purulent neutrophils decreased. The number of follicles was relatively less. The release of TNF-α, IL-6 and other inflammatory factors was reduced by DATS. Erlotinib hydrochloride can cause severe skin toxicities, and DATS prevents skin toxicities, its mechanism may be related to DATS reduced erlotinib-induced inflammatory injury.
Collapse
Affiliation(s)
- Yuping Liu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Xiangliang Jiang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Yue Gu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Yan Chen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China.
| |
Collapse
|
36
|
Yao H, Luo S, Liu J, Xie S, Liu Y, Xu J, Zhu Z, Xu S. Controllable thioester-based hydrogen sulfide slow-releasing donors as cardioprotective agents. Chem Commun (Camb) 2019; 55:6193-6196. [DOI: 10.1039/c9cc02829c] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Hydrogen sulfide (H2S) is an important signaling molecule with promising protective effects in many physiological and pathological processes.
Collapse
Affiliation(s)
- Hong Yao
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University
- Nanjing 210009
- P. R. China
| | - Shanshan Luo
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University
- Nanjing 211166
- P. R. China
| | - Junkai Liu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University
- Nanjing 210009
- P. R. China
| | - Shaowen Xie
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University
- Nanjing 210009
- P. R. China
| | - Yanpeng Liu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University
- Nanjing 210009
- P. R. China
| | - Jinyi Xu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University
- Nanjing 210009
- P. R. China
| | - Zheying Zhu
- Division of Molecular Therapeutics & Formulation, School of Pharmacy, the University of Nottingham, University Park Campus
- Nottingham NG7 2RD
- UK
| | - Shengtao Xu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University
- Nanjing 210009
- P. R. China
| |
Collapse
|
37
|
Hsueh KC, Lin CL, Tung JN, Yang SF, Hsieh YH. Nimbolide induced apoptosis by activating ERK-mediated inhibition of c-IAP1 expression in human hepatocellular carcinoma cells. ENVIRONMENTAL TOXICOLOGY 2018; 33:913-922. [PMID: 29962003 DOI: 10.1002/tox.22576] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 05/04/2018] [Accepted: 05/06/2018] [Indexed: 06/08/2023]
Abstract
Nimbolide is one of the major compounds from the leaves and flowers of the neem tree and exhibits antitumor properties on various cancer cells. However, no report has shown that nimbolide induces apoptosis in vitro and in vivo in human hepatocellular carcinoma cells. Our results indicated that it inhibited cell growth in Huh-7 and PLC/PRF/5 cells. We also found that nimbolide induced cell death through the induction of G2/M phase arrest and mitochondrial dysfunction, accompanied by the increased expression of cleaved caspase-7, caspase-9, caspase-3, caspase-PARP, and Bax and decreased expression of Mcl-1 and Bcl-2. A human apoptosis antibody array analysis demonstrated that inhibition of the apoptosis family proteins (XIAP, c-IAP1, and c-IAP2) was one of the major targets of nimbolide. Additionally, nimbolide sustained activation of ERK expression. Moreover, pretreatment with U0126 (MEK inhibitor) markedly abolished nimbolide-inhibited cell viability, induced cell apoptosis, ERK phosphorylation, cleaved caspase-9, caspase-3, cleaved-PARP activation, and increased c-IAP1 expression in Huh-7 cells. An in vivo study showed that nimbolide significantly reduced Huh-7 tumor growth and weight in a xenograft mouse model. This study indicated the antitumor potential of nimbolide in human hepatocellular carcinoma cells.
Collapse
Affiliation(s)
- Kuan-Chun Hsueh
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Division of General Surgery, Department of Surgery, Tungs' Taichung MetroHarbour Hospital, Taichung, Taiwan
| | - Chia-Liang Lin
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
| | - Jai-Nien Tung
- Department of Neurosurgery, Tungs'Taichung MetroHarbor Hospital, Taichung, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yi-Hsien Hsieh
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
- Department of Biochemistry, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
38
|
Jeremic J, Srejovic I, Zivkovic V, Turnic TN, Milosavljevic I, Jeremic N, Suresh T, Jakovljevic V. CARDIOPROTECTIVE EFFECTS OF DIALLYL TRISULFIDE IN A RAT MODEL OF TYPE-1 DIABETES MELLITUS. PATHOPHYSIOLOGY 2018. [DOI: 10.1016/j.pathophys.2018.07.126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
|
39
|
Yu W, Xiang Y, Luo G, Zhao X, Xiao B, Cheng Y, Feng C, Duan C, Xia X, Wong VKW, Dai R. Salubrinal Enhances Doxorubicin Sensitivity in Human Cholangiocarcinoma Cells Through Promoting DNA Damage. Cancer Biother Radiopharm 2018; 33:258-265. [PMID: 29957018 DOI: 10.1089/cbr.2018.2447] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Cholangiocarcinoma (CCA) is a highly malignant and aggressive tumor of the bile duct that arises from epithelial cells. Chemotherapy is an important treatment strategy for CCA patients, but its efficacy remains limited due to drug resistance. Salubrinal, an inhibitor of eukaryotic translation initiation factor 2 alpha (eIF2α), has been reported to affect antitumor activities in cancer chemotherapy. In this study, the authors investigated the effect of salubrinal on the chemosensitivity of doxorubicin in CCA cells. They showed that doxorubicin induces CCA cell death in a dose- and time-dependent manner. Doxorubicin triggers reactive oxygen species (ROS) generation and induces DNA damage in CCA cells. In addition, ROS inhibitor N-acetylcysteine (NAC) pretreatment inhibits doxorubicin-induced CCA cell death. Importantly, these data demonstrate a synergistic death induction effect contributed by the combination of salubrinal and doxorubicin in CCA cells. It is notable that salubrinal promotes doxorubicin-induced ROS production and DNA damage in CCA cells. Taken together, these data suggest that salubrinal enhances the sensitivity of doxorubicin in CCA cells through promoting ROS-mediated DNA damage.
Collapse
Affiliation(s)
- Wenjing Yu
- 1 State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology , Taipei, China .,2 Department of Biochemistry and Molecular Biology, Southwest Medical University , Luzhou, China
| | - Yuancai Xiang
- 2 Department of Biochemistry and Molecular Biology, Southwest Medical University , Luzhou, China
| | - Guosong Luo
- 3 Department of Hepatobiliary Surgery of the Affiliated Hospital, Southwest Medical University , Luzhou, China
| | - Xiaofang Zhao
- 2 Department of Biochemistry and Molecular Biology, Southwest Medical University , Luzhou, China
| | - Bin Xiao
- 2 Department of Biochemistry and Molecular Biology, Southwest Medical University , Luzhou, China
| | - Ying Cheng
- 2 Department of Biochemistry and Molecular Biology, Southwest Medical University , Luzhou, China
| | - Chunhong Feng
- 3 Department of Hepatobiliary Surgery of the Affiliated Hospital, Southwest Medical University , Luzhou, China
| | - Chunyan Duan
- 2 Department of Biochemistry and Molecular Biology, Southwest Medical University , Luzhou, China
| | - Xianming Xia
- 3 Department of Hepatobiliary Surgery of the Affiliated Hospital, Southwest Medical University , Luzhou, China
| | - Vincent Kam Wai Wong
- 1 State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology , Taipei, China
| | - Rongyang Dai
- 1 State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology , Taipei, China .,2 Department of Biochemistry and Molecular Biology, Southwest Medical University , Luzhou, China
| |
Collapse
|
40
|
Li A, Liu Y, Zhu X, Sun X, Feng X, Li D, Zhang J, Zhu M, Zhao Z. Methylallyl sulfone attenuates inflammation, oxidative stress and lung injury induced by cigarette smoke extract in mice and RAW264.7 cells. Int Immunopharmacol 2018; 59:369-374. [PMID: 29689496 DOI: 10.1016/j.intimp.2018.04.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 03/28/2018] [Accepted: 04/13/2018] [Indexed: 12/28/2022]
Abstract
In this study, we revealed that methylallyl sulfone (AMSO2), the metabolite of active organosulfur compounds, had anti-inflammatory and antioxidant effect in a cigarette smoke extract (CSE)-induced lung injury model. Firstly, histological analysis showed that the CSE group exhibited lung injury compared with the control, which was alleviated by AMSO2. Secondly, we estimated its anti-inflammatory capacity. The results indicated that pretreatment with AMSO2 significantly decreased CSE-elevated tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) in serum. Thirdly, AMSO2 also showed antioxidant properties through enhancing activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) as well as reducing the level of malondialdehyde (MDA) and myeloperoxidase (MPO). Finally, we elucidated that AMSO2 alleviated inflammation and oxidative stress probably via suppressing ERK/p38 MAPK and inhibiting NF-κB expressions. In conclusion, we proposed that AMSO2 protected against the development of CSE-induced lung injury by reducing inflammatory cytokine levels and augmenting antioxidant activity via ERK/p38 MAPK and NF-κB pathways.
Collapse
Affiliation(s)
- Ang Li
- Institute of Pharmaceutics, Medicine School, Shandong University, 44 West Wenhua Road, Jinan 250012, Shandong, China
| | - Yan Liu
- Institute of Pharmaceutics, Medicine School, Shandong University, 44 West Wenhua Road, Jinan 250012, Shandong, China
| | - Xiaosong Zhu
- Institute of Pharmaceutics, Medicine School, Shandong University, 44 West Wenhua Road, Jinan 250012, Shandong, China
| | - Xiao Sun
- Institute of Pharmaceutics, Medicine School, Shandong University, 44 West Wenhua Road, Jinan 250012, Shandong, China
| | - Xiuli Feng
- Institute of Pharmaceutics, Medicine School, Shandong University, 44 West Wenhua Road, Jinan 250012, Shandong, China
| | - Dawei Li
- Shandong Provincial Key Laboratory of Mucosal and Transdermal Drug Delivery Technologies, Shandong Freda Pharmaceutical Group Co., Ltd., 888 Xinluo Street, Jinan 250101, Shandong, China
| | - Jiangqiang Zhang
- Shandong Provincial Key Laboratory of Mucosal and Transdermal Drug Delivery Technologies, Shandong Freda Pharmaceutical Group Co., Ltd., 888 Xinluo Street, Jinan 250101, Shandong, China
| | - Meihua Zhu
- Shandong Provincial Key Laboratory of Mucosal and Transdermal Drug Delivery Technologies, Shandong Freda Pharmaceutical Group Co., Ltd., 888 Xinluo Street, Jinan 250101, Shandong, China
| | - Zhongxi Zhao
- Institute of Pharmaceutics, Medicine School, Shandong University, 44 West Wenhua Road, Jinan 250012, Shandong, China; Shandong Engineering & Technology Research Center for Jujube Food and Drug, 44 West Wenhua Road, Jinan 250012, Shandong, China; Shandong Provincial Key Laboratory of Mucosal and Transdermal Drug Delivery Technologies, Shandong Freda Pharmaceutical Group Co., Ltd., 888 Xinluo Street, Jinan 250101, Shandong, China.
| |
Collapse
|
41
|
Zhao L, Tao X, Qi Y, Xu L, Yin L, Peng J. Protective effect of dioscin against doxorubicin-induced cardiotoxicity via adjusting microRNA-140-5p-mediated myocardial oxidative stress. Redox Biol 2018; 16. [PMID: 29524841 PMCID: PMC5953242 DOI: 10.1016/j.redox.2018.02.026 10.1016/j.redox.2019.101303] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023] Open
Abstract
Clinical application of doxorubicin (DOX) is limited because of its cardiotoxicity. Thus, exploration of effective lead compounds against DOX-induced cardiotoxicity is necessary. The aim of the present study was to investigate the effects and possible mechanisms of dioscin against DOX-induced cardiotoxicity. The in vitro model of DOX- treated H9C2 cells and the in vivo models of DOX-treated rats and mice were used in this study. The results showed that discoin markedly increased H9C2 cell viability, decreased the levels of CK, LDH, and improved histopathological and electrocardio- gram changes in rats and mice to protect DOX-induced cardiotoxicity. Furthermore, dioscin significantly inhibited myocardial oxidative insult through adjusting the levels of intracellular ROS, MDA, SOD, GSH and GSH-Px in vitro and in vivo. Our data also indicated that dioscin activated Nrf2 and Sirt2 signaling pathways, and thereby affected the expression levels of HO-1, NQO1, Gst, GCLM, Keap1 and FOXO3a through decreasing miR-140-5p expression level. In addition, the level of intracellular ROS was significantly increased in H9C2 cells treated by DOX after miR-140-5p mimic transfection, as well as the down-regulated expression levels of Nrf2 and Sirt2, which were markedly reversed by dioscin. In conclusion, our data suggested that dioscin alleviated DOX-induced cardiotoxicity through modulating miR-140-5p-mediated myocardial oxidative stress. This natural product should be developed as a new candidate to alleviate cardiotoxicity caused by DOX in the future.
Collapse
Affiliation(s)
- Lisha Zhao
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Xufeng Tao
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Yan Qi
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Lina Xu
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Lianhong Yin
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Jinyong Peng
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China.
| |
Collapse
|
42
|
Zhao L, Tao X, Qi Y, Xu L, Yin L, Peng J. Protective effect of dioscin against doxorubicin-induced cardiotoxicity via adjusting microRNA-140-5p-mediated myocardial oxidative stress. Redox Biol 2018; 16:189-198. [PMID: 29524841 PMCID: PMC5953242 DOI: 10.1016/j.redox.2018.02.026] [Citation(s) in RCA: 171] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 02/24/2018] [Accepted: 02/26/2018] [Indexed: 12/21/2022] Open
Abstract
Clinical application of doxorubicin (DOX) is limited because of its cardiotoxicity. Thus, exploration of effective lead compounds against DOX-induced cardiotoxicity is necessary. The aim of the present study was to investigate the effects and possible mechanisms of dioscin against DOX-induced cardiotoxicity. The in vitro model of DOX- treated H9C2 cells and the in vivo models of DOX-treated rats and mice were used in this study. The results showed that discoin markedly increased H9C2 cell viability, decreased the levels of CK, LDH, and improved histopathological and electrocardio- gram changes in rats and mice to protect DOX-induced cardiotoxicity. Furthermore, dioscin significantly inhibited myocardial oxidative insult through adjusting the levels of intracellular ROS, MDA, SOD, GSH and GSH-Px in vitro and in vivo. Our data also indicated that dioscin activated Nrf2 and Sirt2 signaling pathways, and thereby affected the expression levels of HO-1, NQO1, Gst, GCLM, Keap1 and FOXO3a through decreasing miR-140-5p expression level. In addition, the level of intracellular ROS was significantly increased in H9C2 cells treated by DOX after miR-140-5p mimic transfection, as well as the down-regulated expression levels of Nrf2 and Sirt2, which were markedly reversed by dioscin. In conclusion, our data suggested that dioscin alleviated DOX-induced cardiotoxicity through modulating miR-140-5p-mediated myocardial oxidative stress. This natural product should be developed as a new candidate to alleviate cardiotoxicity caused by DOX in the future.
Collapse
Affiliation(s)
- Lisha Zhao
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Xufeng Tao
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Yan Qi
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Lina Xu
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Lianhong Yin
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Jinyong Peng
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China.
| |
Collapse
|