1
|
Huang M, Mo Y, Lei H, Chen M. Edaravone: A Possible Treatment for Acute Lung Injury. Int J Gen Med 2024; 17:3975-3986. [PMID: 39286534 PMCID: PMC11403130 DOI: 10.2147/ijgm.s467891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 09/06/2024] [Indexed: 09/19/2024] Open
Abstract
Despite technological advances in science and medicine, acute lung injury (ALI) is still associated with high mortality rates in the ICU. Therefore, finding novel drugs and treatment approaches is crucial to preventing ALI. Drug repurposing is a common practice in clinical research, primarily for drugs that have previously received approval for use in patients, to investigate novel uses of drugs and therapies. One such medication is edaravone, which is a highly effective free-radical scavenger that also has anti-inflammatory, anti-apoptotic, antioxidant, and anti-fibrotic effects. Both basic and clinical studies have shown that edaravone can treat different types of lung injury through its distinct properties. Edaravone exhibits significant protective benefits and holds promising clinical treatment potential for ALI caused by diverse factors, thereby offering a novel approach to treating ALI. This study aims to provide new insights and treatment options for ALI by reviewing both basic and clinical research on the use of edaravone. The focus is on evaluating the effectiveness of edaravone in treating ALI caused by various factors.
Collapse
Affiliation(s)
- Ma Huang
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People's Republic of China
| | - Yalan Mo
- Centre for Infectious Diseases, General Hospital of Hunan Medical College, Huaihua, Hunan, People's Republic of China
| | - Haiyun Lei
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People's Republic of China
| | - Miao Chen
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People's Republic of China
| |
Collapse
|
2
|
Long X, Liu M, Nan Y, Chen Q, Xiao Z, Xiang Y, Ying X, Sun J, Huang Q, Ai K. Revitalizing Ancient Mitochondria with Nano-Strategies: Mitochondria-Remedying Nanodrugs Concentrate on Disease Control. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2308239. [PMID: 38224339 DOI: 10.1002/adma.202308239] [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: 08/14/2023] [Revised: 01/04/2024] [Indexed: 01/16/2024]
Abstract
Mitochondria, widely known as the energy factories of eukaryotic cells, have a myriad of vital functions across diverse cellular processes. Dysfunctions within mitochondria serve as catalysts for various diseases, prompting widespread cellular demise. Mounting research on remedying damaged mitochondria indicates that mitochondria constitute a valuable target for therapeutic intervention against diseases. But the less clinical practice and lower recovery rate imply the limitation of traditional drugs, which need a further breakthrough. Nanotechnology has approached favorable regiospecific biodistribution and high efficacy by capitalizing on excellent nanomaterials and targeting drug delivery. Mitochondria-remedying nanodrugs have achieved ideal therapeutic effects. This review elucidates the significance of mitochondria in various cells and organs, while also compiling mortality data for related diseases. Correspondingly, nanodrug-mediate therapeutic strategies and applicable mitochondria-remedying nanodrugs in disease are detailed, with a full understanding of the roles of mitochondria dysfunction and the advantages of nanodrugs. In addition, the future challenges and directions are widely discussed. In conclusion, this review provides comprehensive insights into the design and development of mitochondria-remedying nanodrugs, aiming to help scientists who desire to extend their research fields and engage in this interdisciplinary subject.
Collapse
Affiliation(s)
- Xingyu Long
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P. R. China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, P. R. China
| | - Min Liu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, P. R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P. R. China
| | - Yayun Nan
- Geriatric Medical Center, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia, 750002, P. R. China
| | - Qiaohui Chen
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, P. R. China
| | - Zuoxiu Xiao
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, P. R. China
| | - Yuting Xiang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, P. R. China
| | - Xiaohong Ying
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, P. R. China
| | - Jian Sun
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830017, P. R. China
| | - Qiong Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P. R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P. R. China
| | - Kelong Ai
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, P. R. China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, 410078, P. R. China
| |
Collapse
|
3
|
Salehzadeh J, Nasiri F. A facile one-pot synthesis of new functionalized pyrazolone-1,4-dithiafulvene hybrids. Mol Divers 2024; 28:19-28. [PMID: 35761142 DOI: 10.1007/s11030-022-10473-x] [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: 04/09/2022] [Accepted: 05/30/2022] [Indexed: 11/25/2022]
Abstract
In this study, a one-pot reaction between β-keto esters or dialkyl acetylenedicarboxylates with hydrazines, carbon disulfide, and dialkyl acetylenedicarboxylates in the presence of triethylamine is reported. This reaction proceeded at room temperature and was completed within 6 h to produce functionalized pyrazolone-1,4-dithiafulvene hybrids in good yields.
Collapse
Affiliation(s)
- Jaber Salehzadeh
- Department of Applied Chemistry, University of Mohaghegh Ardabili, P.O. Box 56199, Ardabil, 11367, Iran
| | - Farough Nasiri
- Department of Applied Chemistry, University of Mohaghegh Ardabili, P.O. Box 56199, Ardabil, 11367, Iran.
| |
Collapse
|
4
|
Wang XB, Zhou LY, Chen XQ, Li R, Yu BB, Pan MX, Fang L, Li J, Cui XJ, Yao M, Lu X. Neuroprotective effect and possible mechanism of edaravone in rat models of spinal cord injury: a protocol for a systematic review and meta-analysis. Syst Rev 2023; 12:177. [PMID: 37752580 PMCID: PMC10521558 DOI: 10.1186/s13643-023-02306-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 08/04/2023] [Indexed: 09/28/2023] Open
Abstract
BACKGROUND Spinal cord injury (SCI) is one of the most disabling neurological conditions, afflicting thousands of human beings. Edaravone, a well-known reactive oxygen species scavenger, is expanding its new scope in field of SCI. The objective of this systematic review is to determine the neuroprotective effects and discuss the underlying mechanism of edaravone in management of SCI. METHODS The systematic review will include the controlled studies evaluating the neurological roles of edaravone on experiment rat models following SCI. The primary outcome will be the 21-point Basso, Beattie, and Bresnahan locomotor rating scale. The secondary outcomes will include the preservation of white matter areas and malondialdehyde levels. Two researchers will independently search PubMed, Embase, Web of Science, Scopus and Cochrane Library from their inception date. Following study selection, data extraction, and assessment of methodological quality in included studies using the SYRCLE's RoB tool, data from eligible studies will be pooled and analyzed using random-effects models with RevMan 5.3 software. In case of sufficient data, subgroup analyses with respect to species, age, gender, injury characteristics, or administration details will be carried out to explore the factors modifying efficacy of edaravone. For exploring the appropriate dose of edaravone, a network meta-analysis approach will be conducted based on the Bayesian method. Importantly, the proposed mechanisms and changes of related molecules will be also extracted from included studies for comprehensively investigating the mechanisms underlying the neuroprotective effects of edaravone. DISCUSSION In this study, we aim to quantitatively analyze the role of edaravone in locomotor recovery and tissue damage in SCI rat model. The efficacy of edaravone in distinct scenarios will be investigated by subgroup analyses, and we expect to predict the candidate dose that offers a superior treatment effect using network meta-analyses. Moreover, a comprehensive framework regarding the neuroprotective mechanisms behind edaravone will be constructed via a combination of systematic and traditional review. This study will bring implications for future preclinical studies and clinical applications of SCI. Nonetheless, in light of the anticipated limitations in animal experimental design and methodological quality, the results in this review should be interpreted with caution.
Collapse
Affiliation(s)
- Xiao-Bo Wang
- Spine Disease Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Long-Yun Zhou
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Xu-Qing Chen
- Department of Otolaryngology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Ran Li
- Traditional Chinese Medicine Hospital of LuAn, Luan, 237006, Anhui, China
| | - Bin-Bin Yu
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Meng-Xiao Pan
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Lu Fang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Jian Li
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Xue-Jun Cui
- Spine Disease Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Min Yao
- Spine Disease Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Xiao Lu
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
5
|
Yıldızhan K, Huyut Z, Altındağ F. Involvement of TRPM2 Channel on Doxorubicin-Induced Experimental Cardiotoxicity Model: Protective Role of Selenium. Biol Trace Elem Res 2023; 201:2458-2469. [PMID: 35922740 DOI: 10.1007/s12011-022-03377-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 07/30/2022] [Indexed: 12/31/2022]
Abstract
Doxorubicin (DOXR) is an important chemotherapeutic drug used in cancer treatment for many years. Several studies reported that the use of DOXR increased toxicity by causing an increase in oxidative stress (OS), especially in the heart. In this study, we investigated the protective effect of selenium (Se) and the role of transient receptor potential melastatin-2 (TRPM2) channel activation by using N-(p-amylcinnamoyl) anthranilic acid (ACA) in a model of DOXR-induced cardiotoxicity. Sixty female rats were equally divided into the control, dimethyl sulfoxide (DMSO), DOXR, DOXR + Se, DOXR + ACA, and DOXR + Se + ACA groups. Glutathione (GSH), glutathione peroxidase (GSH-Px), caspases (Cas) 3 and 9, interleukin 1β (IL-1β), tumor necrosis factor-α (TNF-α), reactive oxygen species (ROS), poly [ADP-ribose] polymerase 1 (PARP-1), and TRPM2 channel levels were measured by ELISA. In addition, histopathological examination was performed in cardiac tissues and TNF-α, caspase 3, and TRPM2 channel expression levels were determined immunohistochemically. The levels of GSH, GSH-Px, caspases 3 and 9, IL-1β, TNF-α, ROS, PARP-1, and TRPM2 channel in serum, and cardiac tissue in the DOXR group were higher than in the control and DMSO groups (p < 0.05). However, these parameters in Se and/or ACA treatment groups were lower than in the DOXR group (p < 0.05). Also, we determined that Se and/or ACA treatment together with DOXR application decreased the TNF-α, Cas-3, and TRPM2 channel expression levels in the cardiac tissue. The data showed that administration of Se and/or ACA treatment together with DOXR may be used as a therapeutic agent in preventing DOXR-induced cardiotoxicity.
Collapse
Affiliation(s)
- Kenan Yıldızhan
- Department of Biophysics, Faculty of Medicine, Van Yuzuncu Yil University, TR-65090, Van, Turkey.
| | - Zübeyir Huyut
- Department of Biochemistry, Faculty of Medicine, Van Yuzuncu Yil University, Van, Turkey
| | - Fikret Altındağ
- Department of Histology and Embryology, Faculty of Medicine, Van Yuzuncu Yil University, Van, Turkey
| |
Collapse
|
6
|
Shao Z, Li R, Shao D, Tang H, Han Y. Albumin-Based Zn (II)-Quercetin Enzyme Mimic Scavenging ROS for Protection against Cardiotoxicity Induced by Doxorubicin. Pharmaceuticals (Basel) 2022; 15:ph15121524. [PMID: 36558975 PMCID: PMC9781925 DOI: 10.3390/ph15121524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/03/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
Doxorubicin (DOX) is a chemotherapeutic agent that can cause cardiotoxicity leading to progressive, chronic, life-threatening cardiomyopathy, called DOX-induced cardiomyopathy (DIC). DIC is a fatal cardiomyopathy with a worse prognosis compared to other cardiomyopathies and limits the use of DOX in malignancies due to its cardiotoxicity. DIC has been proven to be associated with reactive oxygen species (ROS)-induced side effect damage in cardiac myocytes. Currently, scavenging of reactive oxygen species is a practical strategy to reduce chemotherapy-associated DIC. Although quercetin has already been reported to have superior antioxidant activity, its clinical application is severely limited due to its rapid degradation and poor tissue absorption. Herein, we reported the preparation of a novel enzyme mimic via coordinated albumin, Zinc Ion (Zn2+) and quercetin. The enzyme mimics were capable of simultaneously increasing the biocompatibility and efficiently overcame the drawbacks of free quercetin, and were achieved by long circulation in vivo. Most importantly, these quercetin-based enzyme mimics had no effect on the antioxidant activity of quercetin. These beneficial therapeutic properties, together with high drug-carrying capacity and redox stimuli, will significantly improve quercetin's alleviation of chemotherapeutic cardiotoxicity without causing significant side effects. Therefore, nanoparticles of albumin-based Zn (II)-Quercetin have a promising clinical application as an effective agent for mitigating the cardiotoxicity of chemotherapy.
Collapse
Affiliation(s)
- Zehua Shao
- Heart Center of Zhengzhou University People’s Hospital, Fuwai Central China Cardiovascular Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou 450003, China
| | - Ran Li
- Heart Center of Zhengzhou University People’s Hospital, Fuwai Central China Cardiovascular Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou 450003, China
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Fuwai Central China Cardiovascular Hospital & Central China Branch of National Center for Cardiovascular Diseases, Zhengzhou 451464, China
| | - Dongxing Shao
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Fuwai Central China Cardiovascular Hospital & Central China Branch of National Center for Cardiovascular Diseases, Zhengzhou 451464, China
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Beijing 100191, China
| | - Hao Tang
- Heart Center of Zhengzhou University People’s Hospital, Fuwai Central China Cardiovascular Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou 450003, China
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Fuwai Central China Cardiovascular Hospital & Central China Branch of National Center for Cardiovascular Diseases, Zhengzhou 451464, China
- Correspondence: (H.T.); (Y.H.)
| | - Yu Han
- Heart Center of Zhengzhou University People’s Hospital, Fuwai Central China Cardiovascular Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou 450003, China
- Correspondence: (H.T.); (Y.H.)
| |
Collapse
|
7
|
Chen Y, Shi S, Dai Y. Research progress of therapeutic drugs for doxorubicin-induced cardiomyopathy. Biomed Pharmacother 2022; 156:113903. [PMID: 36279722 DOI: 10.1016/j.biopha.2022.113903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/16/2022] [Accepted: 10/19/2022] [Indexed: 12/06/2022] Open
|
8
|
Jiao Y, Yuan C, Wu T, Zhang H, Wei Y, Ma Y, Zhang X, Han L. Incidence of pressure injuries in fracture patients: A systematic review and meta-analysis. J Tissue Viability 2022; 31:726-734. [PMID: 36109261 DOI: 10.1016/j.jtv.2022.08.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 08/17/2022] [Accepted: 08/28/2022] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To systematically evaluate the incidence of pressure injuries (PIs) in hospitalized fracture patients and to provide evidence for the prevention and treatment of PIs. METHODS A systematic review and meta-analysis was conducted according to the Preferred Reporting Items for Systematic Reviews and Meta-analysis (PRISMA) guidelines. Electronic databases including PubMed, Embase, the Cochrane Library, Web of Science, CINAHL, China Knowledge Resource Integrated Database (CNKI), WanFang Database, Weipu Database (VIP), and Chinese Biomedical Database (CBM) were searched to collect cross-sectional studies and cohort studies related to PIs among hospitalized fracture patients. All electronic literature sources were searched from inception to March 2022, and a hand-search through references was also conducted to find relevant articles. Studies were evaluated independently by two researchers and audited by a third researcher. The data were extracted and presented in tables. The risk of bias was assessed using the Joanna Briggs Institute (JBI) Critical Appraisal Checklist. All data analysis used Stata14.0. The I2 statistic and random-effects model were used to determine the heterogeneity. RESULTS A total of 7906 articles were screened, and 18 studies with 8956 patients were ultimately involved in this review. The pooled incidence of PIs in the fracture patients was 20.4% (95%CI: 14.9 to 25.8), and the incidence of PIs only in spinal and hip fracture patients was 23.9% (95%CI: 19.6 to 28.2). The incidence of PIs in 65 years old or over was significantly high (23.3% [95%CI: 15.3 to 31.2]). The most affected body sites were sacrococcygeal regions (56.7%) and heels (19.9%). The most common stages were stage 2 (62.2%) and stage 1 (17.4%). CONCLUSION The overall incidence of PIs in fracture patients was as high as 20.4%, significantly higher than the average incidence of adults. We found that the potential for PIs in fracture patients increases with age. Hence, our discoveries recommended that healthcare givers should consider reducing the occurrence of PIs. Additionally, more research may be conducted to improve the understanding of characteristics of PIs among fracture patients and to identify PIs risk factors to prevent and treat them effectively.
Collapse
Affiliation(s)
- Yanxia Jiao
- Evidence-based Nursing Center, School of Nursing, Lanzhou University, Lanzhou City, Gansu Province, 730000, #28 Yanxi Road, Chenguan District, China.
| | - Chenlu Yuan
- Evidence-based Nursing Center, School of Nursing, Lanzhou University, Lanzhou City, Gansu Province, 730000, #28 Yanxi Road, Chenguan District, China.
| | - Tong Wu
- Evidence-based Nursing Center, School of Nursing, Lanzhou University, Lanzhou City, Gansu Province, 730000, #28 Yanxi Road, Chenguan District, China.
| | - Hongyan Zhang
- Department of Nursing, Gansu Provincial Hospital, Lanzhou City, Gansu Province, #204 Donggang Road, Chenguan District, 730000, China.
| | - Yuting Wei
- Evidence-based Nursing Center, School of Nursing, Lanzhou University, Lanzhou City, Gansu Province, 730000, #28 Yanxi Road, Chenguan District, China.
| | - Yuxia Ma
- Evidence-based Nursing Center, School of Nursing, Lanzhou University, Lanzhou City, Gansu Province, 730000, #28 Yanxi Road, Chenguan District, China.
| | - Xiujuan Zhang
- Department of Respiratory, Gansu Provincial Hospital, Lanzhou City, Gansu Province, #204 Donggang Road, Chenguan District, 730000, China.
| | - Lin Han
- Department of Nursing, Gansu Provincial Hospital, Lanzhou City, Gansu Province, #204 Donggang Road, Chenguan District, 730000, China.
| |
Collapse
|
9
|
LCZ696 ameliorates doxorubicin-induced cardiomyocyte toxicity in rats. Sci Rep 2022; 12:4930. [PMID: 35322164 PMCID: PMC8943022 DOI: 10.1038/s41598-022-09094-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 03/17/2022] [Indexed: 12/11/2022] Open
Abstract
Doxorubicin (DOX)-based chemotherapy induces cardiotoxicity, which is considered the main bottleneck for its clinical application. In this study, we investigated the potential benefit of LCZ696, an angiotensin receptor-neprilysin inhibitor against DOX-induced cardiotoxicity in rats and H9c2 cells and determined whether the mechanism underlying any such effects involves its antioxidant activity. Male Sprague-Dawley rats were randomly separated into four groups, each consisting of 15 rats (DOX (1.5 mg/kg/day intraperitoneally for 10 days followed by non-treatment for 8 days); DOX + valsartan (31 mg/kg/day by gavage from day 1 to day 18); DOX + LCZ696 (68 mg/kg/day by gavage from day 1 to day 18); and control (saline intraperitoneally for 10 days). DOX-induced elevation of cardiac troponin T levels on day 18 was significantly reduced by LCZ696, but not valsartan. The DOX-induced increase in myocardial reactive oxygen species (ROS) levels determined using dihydroethidium was significantly ameliorated by LCZ696, but not valsartan, and was accompanied by the suppression of DOX-induced increase in p47phox. LCZ696 recovered the DOX-induced decrease in phosphorylation of adenosine monophosphate-activated protein kinase and increased the ratio of Bax and Bcl-2. In H9c2 cardiomyocytes, LCZ696 reduced DOX-induced mitochondrial ROS generation and improved cell viability more than valsartan. Our findings indicated that LCZ696 ameliorated DOX-induced cardiotoxicity in rat hearts in vivo and in vitro, possibly by mediating a decrease in oxidative stress.
Collapse
|
10
|
Cha SJ, Kim K. Effects of the Edaravone, a Drug Approved for the Treatment of Amyotrophic Lateral Sclerosis, on Mitochondrial Function and Neuroprotection. Antioxidants (Basel) 2022; 11:antiox11020195. [PMID: 35204078 PMCID: PMC8868074 DOI: 10.3390/antiox11020195] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 01/13/2022] [Accepted: 01/19/2022] [Indexed: 01/27/2023] Open
Abstract
Edaravone, the first known free radical scavenger, has demonstrated cellular protective properties in animals and humans. Owing to its antioxidant activity, edaravone modulates oxidative damage in various diseases, especially neurodegenerative diseases. In 2015, edaravone was approved in Japan to treat amyotrophic lateral sclerosis. The distinguishing pathogenic features of neurodegenerative diseases include high reactive oxygen species levels and mitochondrial dysfunction. However, the correlation between mitochondria and edaravone has not been elucidated. This review highlights recent studies on novel therapeutic perspectives of edaravone in terms of its effect on oxidative stress and mitochondrial function.
Collapse
Affiliation(s)
- Sun Joo Cha
- Department of Medical Sciences, Soonchunhyang University, Asan 31538, Korea;
| | - Kiyoung Kim
- Department of Medical Sciences, Soonchunhyang University, Asan 31538, Korea;
- Department of Medical Biotechnology, Soonchunhyang University, Asan 31538, Korea
- Correspondence: ; Tel.: +82-41-413-5024; Fax: +82-41-413-5006
| |
Collapse
|
11
|
Alqudah MA, Al-Nosairy A, Alzoubi KH, Kahbour OF, Alazzam SI. Edaravone prevents memory impairment in diabetic rats: Role of oxidative stress. INFORMATICS IN MEDICINE UNLOCKED 2022. [DOI: 10.1016/j.imu.2022.101096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
|
12
|
Ommati MM, Attari H, Siavashpour A, Shafaghat M, Azarpira N, Ghaffari H, Moezi L, Heidari R. Mitigation of cholestasis-associated hepatic and renal injury by edaravone treatment: Evaluation of its effects on oxidative stress and mitochondrial function. LIVER RESEARCH 2021; 5:181-193. [PMID: 39957848 PMCID: PMC11791843 DOI: 10.1016/j.livres.2020.10.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/20/2020] [Accepted: 10/25/2020] [Indexed: 02/07/2023]
Abstract
Background and aim The liver is the primary organ affected by cholestasis, and complications such as renal injury, renal failure, and the need for renal transplantation are associated with cholestatic liver disease. There is substantial evidence indicating that reactive oxygen species (ROS) and mitochondrial impairment are fundamental mechanisms underlying cholestasis-induced hepatic and renal injury. Edaravone (EDV) is a potent radical scavenger and antioxidant that may prevent oxidative stress and improve impaired mitochondrial function in various diseases. This study was performed to evaluate the effects and mechanisms of action of EDV on hepatic and renal injury in an animal model of cholestasis. Methods Rats subjected to bile duct ligation (BDL) were treated with EDV 1 or 10 mg/kg/day intraperitoneally for 14 consecutive days. Biomarkers of oxidative stress and mitochondrial impairment in the liver and kidney were assessed in EDV-treated and untreated rats with cholestasis. Results Significant increases in tissue ROS level, lipid peroxidation, protein carbonylation, and oxidized glutathione level were detected in rats subjected to BDL. Additionally, significant decreases in tissue glutathione level and antioxidant capacity were observed in the hepatic and renal tissues of rats with cholestasis. Markers of mitochondrial impairment, including mitochondrial depolarization, lipid peroxidation, mitochondrial permeabilization, depleted adenosine triphosphate content, and decreased dehydrogenase activity, were also detected in rats subjected to BDL. Furthermore, portal inflammation, necrosis, and tissue fibrosis were detected in the liver and significant tubular atrophy and interstitial inflammation, as well as fibrotic lesions, were detected in the kidneys of rats with cholestasis. EDV treatment significantly mitigated cholestasis-associated hepatic and renal injury. Conclusions The antioxidative properties of EDV and its positive effects on the indices of mitochondrial function may be critical factors contributing to protection against cholestasis-associated hepatic and renal injury.
Collapse
Affiliation(s)
| | - Hanie Attari
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Asma Siavashpour
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Marzieh Shafaghat
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hasti Ghaffari
- Department of Veterinary Sciences, Islamic Azad University, Urmia Branch, Urmia, Iran
| | - Leila Moezi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmacology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
13
|
Guo H, Yang R, He J, Chen K, Yang W, Liu J, Xiao K, Li H. Edaravone combined with dexamethasone exhibits synergic effects on attenuating smoke-induced inhalation lung injury in rats. Biomed Pharmacother 2021; 141:111894. [PMID: 34225014 DOI: 10.1016/j.biopha.2021.111894] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 06/06/2021] [Accepted: 06/28/2021] [Indexed: 02/05/2023] Open
Abstract
Inhalational lung injury often leads to morbidity and mortality during fire disasters. In this study, we aimed to evaluate the protective effects of edaravone combined with dexamethasone on smoke-induced inhalational lung injury. Sprague-Dawley rats were divided into five groups, namely, the control, model (inhalation), and three treatment groups (edaravone, dexamethasone, and edaravone combined with dexamethasone). After drug intervention in the acute lung injury model, arterial blood gas, wet:dry weight ratio of the lung tissue, bronchoalveolar lavage fluid, and pulmonary histopathology were determined. The production of reactive oxygen species (ROS), mitochondrial membrane potential (MMP), inflammatory cytokines, peroxidase and apoptosis were further analyzed to explore the underlying mechanisms. The results of blood gas and inflammatory cytokine analysis and the histopathological data demonstrated that edaravone combined with dexamethasone had obvious protective effects on smoke infiltration and tissue injury. Moreover, after the co-administration of edaravone and dexamethasone, malondialdehyde and myeloperoxidase levels in the lung tissue decreased, whereas those of glutathione peroxidase and superoxide dismutase were elevated. In addition, this drug combination could inhibit smoke-induced apoptosis in lung tissues by reducing the cleavage of caspase-3, caspase-9, and poly ADP-ribose polymerase (PARP), and also reverse smoke-mediated mitochondrial dysfunction, including ROS generation, loss of MMP, early release of cytochrome C, second mitochondrial activator of caspases, and apoptosis-inducing factor. In conclusion, edaravone combined with dexamethasone had a protective effect on smoke-induced inhalational lung injury in rats and can be further explored as an attractive therapeutic option for the treatment of smoke inhalation-induced pulmonary dysfunction.
Collapse
Affiliation(s)
- Haidong Guo
- National Chengdu Center for Safety Evaluation of Drugs and National Clinical Research Center for Geriatrics, West China hospital, Sichuan University, Chengdu 610041, PR China; Sichuan Fire Research Institute of Ministry of Emergency Management, Chengdu 610036, PR China
| | - Runfang Yang
- National Chengdu Center for Safety Evaluation of Drugs and National Clinical Research Center for Geriatrics, West China hospital, Sichuan University, Chengdu 610041, PR China
| | - Jin He
- Sichuan Fire Research Institute of Ministry of Emergency Management, Chengdu 610036, PR China
| | - Ke Chen
- National Chengdu Center for Safety Evaluation of Drugs and National Clinical Research Center for Geriatrics, West China hospital, Sichuan University, Chengdu 610041, PR China
| | - Wen Yang
- National Chengdu Center for Safety Evaluation of Drugs and National Clinical Research Center for Geriatrics, West China hospital, Sichuan University, Chengdu 610041, PR China
| | - Junjun Liu
- Sichuan Fire Research Institute of Ministry of Emergency Management, Chengdu 610036, PR China
| | - Kai Xiao
- National Chengdu Center for Safety Evaluation of Drugs and National Clinical Research Center for Geriatrics, West China hospital, Sichuan University, Chengdu 610041, PR China; Precision Medicine Research Center, Sichuan Provincial Key Laboratory of Precision Medicine, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Hongxia Li
- National Chengdu Center for Safety Evaluation of Drugs and National Clinical Research Center for Geriatrics, West China hospital, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
14
|
Qian XK, Zhang J, Song PF, Zhao YS, Ma HY, Jin Q, Wang DD, Guan XQ, Li SY, Bao X, Zou LW. Discovery of pyrazolones as novel carboxylesterase 2 inhibitors that potently inhibit the adipogenesis in cells. Bioorg Med Chem 2021; 40:116187. [PMID: 33965840 DOI: 10.1016/j.bmc.2021.116187] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/09/2021] [Accepted: 04/26/2021] [Indexed: 10/21/2022]
Abstract
Carboxylesterase 2 (CES2) is one of the most important Phase I drug metabolizing enzymes in the carboxylesterase family. It plays crucial roles in the bioavailability of oral ester prodrugs and the therapeutic effect of some anticancer drugs such as irinotecan (CPT11) and capecitabine. In addition to the well-known roles of CES2 in xenobiotic metabolism, the enzyme also participates in endogenous metabolism and the production of lipids. In this study, we synthesized a series of pyrazolones and assayed their inhibitory effects against CES2 in vitro. Structure-activity relationship analysis of these pyrazolones reveals that the introduction of 4-methylphenyl unit (R1), 4-methylbenzyl (R2) and cyclohexyl (R3) moieties are beneficial for CES2 inhibition. Guided by these SARs results, 1-cyclohexyl-4-(4-methylbenzyl)-3-p-tolyl-1H- pyrazol-5(4H)-one (27) was designed and synthesized. Further investigations demonstrated that the compound 27 exhibited stronger CES2 inhibition activity with a lower IC50 value (0.13 μM). The inhibition kinetic study demonstrated that compound 27 inhibited the hydrolysis of CES2-fluorescein diacetate (FD) through non-competitive inhibition. In addition, the molecular docking showed that the core of pyrazolone, the cyclohexane moiety, 4-methylbenzyl and 4-methylphenyl groups in compound 27 all played important roles with the amino acid residues of CSE2. Also, compound 27 could inhibit adipocyte adipogenesis induced by mouse preadipocytes. In brief, we designed and synthesized a novel pyrazolone compound with a strong inhibitory ability on CES2 and could inhibit the adipogenesis induced by mouse preadipocytes, which can be served as a promising lead compound for the development of more potent pyrazolone-type CES2 inhibitors, and also used as a potential tool for exploring the biological functions of CES2 in human being.
Collapse
Affiliation(s)
- Xing-Kai Qian
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jing Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Pei-Fang Song
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yi-Su Zhao
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hong-Ying Ma
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qiang Jin
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Dan-Dan Wang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiao-Qing Guan
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Shi-Yang Li
- Analytical Central Laboratory, Shengyang Harmony Health Medical Laboratory Co Ltd, 19 Wen Hui Road Shenyang 210112, China
| | - XiaoZe Bao
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China.
| | - Li-Wei Zou
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
15
|
Vairetti M, Di Pasqua LG, Cagna M, Richelmi P, Ferrigno A, Berardo C. Changes in Glutathione Content in Liver Diseases: An Update. Antioxidants (Basel) 2021; 10:364. [PMID: 33670839 PMCID: PMC7997318 DOI: 10.3390/antiox10030364] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 02/22/2021] [Accepted: 02/24/2021] [Indexed: 02/07/2023] Open
Abstract
Glutathione (GSH), a tripeptide particularly concentrated in the liver, is the most important thiol reducing agent involved in the modulation of redox processes. It has also been demonstrated that GSH cannot be considered only as a mere free radical scavenger but that it takes part in the network governing the choice between survival, necrosis and apoptosis as well as in altering the function of signal transduction and transcription factor molecules. The purpose of the present review is to provide an overview on the molecular biology of the GSH system; therefore, GSH synthesis, metabolism and regulation will be reviewed. The multiple GSH functions will be described, as well as the importance of GSH compartmentalization into distinct subcellular pools and inter-organ transfer. Furthermore, we will highlight the close relationship existing between GSH content and the pathogenesis of liver disease, such as non-alcoholic fatty liver disease (NAFLD), alcoholic liver disease (ALD), chronic cholestatic injury, ischemia/reperfusion damage, hepatitis C virus (HCV), hepatitis B virus (HBV) and hepatocellular carcinoma. Finally, the potential therapeutic benefits of GSH and GSH-related medications, will be described for each liver disorder taken into account.
Collapse
Affiliation(s)
| | - Laura Giuseppina Di Pasqua
- Unit of Cellular and Molecular Pharmacology and Toxicology, Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy; (M.V.); (M.C.); (P.R.); (C.B.)
| | | | | | - Andrea Ferrigno
- Unit of Cellular and Molecular Pharmacology and Toxicology, Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy; (M.V.); (M.C.); (P.R.); (C.B.)
| | | |
Collapse
|
16
|
Li Z, Chinnathambi A, Ali Alharbi S, Yin F. Plumbagin protects the myocardial damage by modulating the cardiac biomarkers, antioxidants, and apoptosis signaling in the doxorubicin-induced cardiotoxicity in rats. ENVIRONMENTAL TOXICOLOGY 2020; 35:1374-1385. [PMID: 32691977 DOI: 10.1002/tox.23002] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/18/2020] [Accepted: 06/28/2020] [Indexed: 06/11/2023]
Abstract
Cardiovascular disease created enormous health and economic burdens worldwide, which is responsible for the highest mobility and mortality that results in nearly 6.2% of in-hospital deaths every year. Plumbagin is a major bioactive compound that occurs in the Plumbago indica and P. zeylanica with numerous therapeutic benefits. The current research exploration was planned to investigate the therapeutic role of plumbagin against doxorubicin stimulated cardiotoxicity in rats. The cardiotoxicity was stimulated to the rats by administering the 2.5 mg/kg of doxorubicin for 14 days with concurrent supplementation with plumbagin. The hemodynamic parameters were studied by using the tail-cuff plethysmography. The lipid peroxidation and antioxidant status was examined by the standard procedures. The myocardial function and damage markers were assessed with the help of commercial kits. The expression status of inflammatory markers and PI3K/Akt signaling markers were investigated by reverse transcription polymerase chain reaction (RT-PCR) and western blotting analysis, respectively. The plumbagin supplementation appreciably regained the body weight and heart weight of the investigational animals. Hemodynamic parameters and antioxidants statuses were escalated by the plumbagin treatment. The severe elevation in the cardiac damage markers and inflammatory markers were noticeably ameliorated by the plumbagin treatment. The plumbagin treatment also assuaged the overexpression of inflammatory and apoptotic proteins in the heart tissues of doxorubicin-challenged rats. The histopathological analysis revealed that the plumbagin appreciably protected the heart tissues from the doxorubicin-induced damages. The findings of this exploration evidenced that plumbagin treatment attenuated the doxorubicin-stimulated cardiotoxicity in rats.
Collapse
Affiliation(s)
- Zhe Li
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi Province, China
| | - Arunachalam Chinnathambi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Sulaiman Ali Alharbi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Fuyu Yin
- Department of Cardiology, Xidian Group Hospital, Xi'an, Shaanxi Province, China
| |
Collapse
|
17
|
Hassanein EHM, Abd El-Ghafar OAM, Ahmed MA, Sayed AM, Gad-Elrab WM, Ajarem JS, Allam AA, Mahmoud AM. Edaravone and Acetovanillone Upregulate Nrf2 and PI3K/Akt/mTOR Signaling and Prevent Cyclophosphamide Cardiotoxicity in Rats. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:5275-5288. [PMID: 33299300 PMCID: PMC7721127 DOI: 10.2147/dddt.s281854] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 11/11/2020] [Indexed: 01/17/2023]
Abstract
Introduction Cyclophosphamide (CP) causes redox imbalance and its use is associated with marked cardiotoxicity that limits its clinical applications. The present study investigated the protective effects of acetovanillone (AV) and edaravone (ED) against CP-induced oxidative stress and cardiac damage, emphasizing the role of PI3K/Akt/mTOR and Nrf2 signaling. Materials and Methods Rats received either AV (100 mg/kg) or ED (20 mg/kg) orally for 10 days and CP (200 mg/kg) on day 7. At day 11, the rats were sacrificed, and samples were collected for analysis. Results AV and ED ameliorated serum troponin I, CK-MB, LDH, AST and ALP, and prevented cardiac histological alterations in CP-intoxicated rats. Both treatments decreased cardiac lipid peroxidation and enhanced GSH, SOD and cytoglobin in CP-induced rats. AV and ED downregulated Keap1, whereas increased the expression of PI3K, Akt, mTOR and Nrf2 in the heart of rats received CP. Additionally, the binding modes of AV and ED to Keap1 were pinpointed in silico using molecular docking simulations. Conclusion AV and ED prevent CP cardiotoxicity by attenuating oxidative stress and tissue injury, and modulating cytoglobin, and PI3K/Akt/mTOR and Keap1/Nrf2 signaling. Therefore, AV and ED may represent promising agents that can prevent cardiac injury in patients receiving CP.
Collapse
Affiliation(s)
- Emad H M Hassanein
- Department of Pharmacology and Toxicology Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt
| | - Omnia A M Abd El-Ghafar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Nahda University, Beni-Suef, Egypt
| | - Marwa A Ahmed
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Ahmed M Sayed
- Biochemistry Laboratory, Chemistry Department, Faculty of Science, Assiut University, Assiut, Egypt
| | - Wail M Gad-Elrab
- Human Anatomy & Embryology Department Faculty of Medicine, Al-Azhar University, Assiut, Egypt
| | - Jamaan S Ajarem
- Zoology Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed A Allam
- Zoology Department Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| | - Ayman M Mahmoud
- Zoology Department Faculty of Science, Beni-Suef University, Beni-Suef, Egypt.,Biotechnology Department, Research Institute of Medicinal and Aromatic Plants, Beni-Suef University, Beni-Suef, Egypt
| |
Collapse
|
18
|
Li H, Xia B, Chen W, Zhang Y, Gao X, Chinnathambi A, Alharbi SA, Zhao Y. Nimbolide prevents myocardial damage by regulating cardiac biomarkers, antioxidant level, and apoptosis signaling against doxorubicin-induced cardiotoxicity in rats. J Biochem Mol Toxicol 2020; 34:e22543. [PMID: 32627270 DOI: 10.1002/jbt.22543] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 05/08/2020] [Accepted: 05/29/2020] [Indexed: 12/31/2022]
Abstract
The current work planned to assess the protecting properties of nimbolide against doxorubicin (DOX)-treated myocardial damage. Myocardial damage was produced with 2.5 mg/kg of DOX given on alternative days (14 days). Thiobarbituric acid reactive substances (TBARS) levels of a lipid peroxidative marker were elevated, whereas reduced body weight, heart weight, blood pressure indices and reduced levels of antioxidants like glutathione-S-transferase, superoxide dismutase, catalase, glutathione peroxidase, glutathione, and glutathione reductase were observed in the heart tissue of DOX-treated animals. DOX-treated animals showed augmented levels of cardiac markers likes monocyte chemotactic protein-1, interferon-gamma, aspartate transferase, creatine kinase, lactate dehydrogenase, creatine kinase-muscle/brain, heart-type fatty acid-binding protein, glycogen phosphorylase isoenzyme BB, transforming growth factor-β, brain natriuretic peptide, myoglobin, and cTnI in serum. Histopathological assessment confirmed the DOX-induced cardiotoxicity. Furthermore, DOX-induced rats showed augmented inflammatory mediators (nuclear factor-κB [NF-kB], tumor necrosis factor-α [TNF-α], and interleukin-1β [IL-1β]) and increased PI3K/Akt signaling proteins (PI3K, p-Bad/Bad, caspase-3, and p-Akt), whereas decreased oxidative markers (HO-1 and NQO-1) and p-PTEN were observed. Nimbolide-supplemented rats showed reduced activity/levels of cardiac markers and TBARS levels in serum and heart tissue. Levels of enzymatic and nonenzymatic antioxidants were augmented in the heart tissue of nimbolide-supplemented rats. Nimbolide influence decreased apoptosis, inflammation, and enhanced antioxidant markers through the modulation of p-Bad/Bad, caspase-3, PI3K, p-Akt, TNF-α, NF-kB, IL-1β, HO-1, NQO-1, and p-PTEN markers. The histopathological explanations were observed to be in line with biochemical analysis. Therefore, the finding of current work was that nimbolide has a defensive effect on the myocardium against DOX-induced cardiac tissue damage.
Collapse
Affiliation(s)
- Haining Li
- Department of Critical Care Medicine, Shenyang Tenth People's Hospital, Shenyang, Liaoning, China
| | - Bihua Xia
- Internal Medicine-Cardiovascular Department, The Second Affiliated Hospital of GuiZhou Medical University, Kaili, GuiZhou, China
| | - Wei Chen
- Department of Critical Care Medicine, Shenyang Tenth People's Hospital, Shenyang, Liaoning, China
| | - Yumeng Zhang
- Department of Critical Care Medicine, Shenyang Tenth People's Hospital, Shenyang, Liaoning, China
| | - Xia Gao
- Ultrasonic Room, Shenyang Tenth People's Hospital, Shenyang, Liaoning, China
| | - Arunachalam Chinnathambi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Sulaiman A Alharbi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Yujie Zhao
- ICU, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| |
Collapse
|