1
|
Intharuksa A, Kuljarusnont S, Sasaki Y, Tungmunnithum D. Flavonoids and Other Polyphenols: Bioactive Molecules from Traditional Medicine Recipes/Medicinal Plants and Their Potential for Phytopharmaceutical and Medical Application. Molecules 2024; 29:5760. [PMID: 39683916 DOI: 10.3390/molecules29235760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
Currently, natural bioactive ingredients and/or raw materials are of significant interest to scientists around the world. Flavonoids and other polyphenols are a major group of phytochemicals that have been researched and noted as bioactive molecules. They offer several pharmacological and medical benefits. This current review aims to (1) illustrate their benefits for human health, such as antioxidant, anti-aging, anti-cancer, anti-inflammatory, anti-microbial, cardioprotective, neuroprotective, and UV-protective effects, and also (2) to perform a quality evaluation of traditional medicines for future application. Consequently, keywords were searched on Scopus, Google Scholar, and PubMed so as to search for related publications. Then, those publications were carefully checked in order to find current and non-redundant studies that matched the objective of this review. According to this review, researchers worldwide are very interested in discovering the potential of flavonoids and other polyphenols, used in traditional medicines and taken from medicinal plants, in relation to medical and pharmaceutical applications. Many studies focus on the health benefits of flavonoids and other polyphenols have been tested using in silico, in vitro, and in vivo models. However, few studies have been carried out using clinical trials that have trustworthy subject sizes and are in accordance with clinical practice guidelines. Additionally, interesting research directions and perspectives for future studies are highlighted in this work.
Collapse
Affiliation(s)
- Aekkhaluck Intharuksa
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sompop Kuljarusnont
- Department of Obstetrics and Gynecology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Yohei Sasaki
- Division of Pharmaceutical Sciences, Graduate School of Medical Plant Sciences, Kanazawa University, Kanazawa 920-1192, Japan
| | - Duangjai Tungmunnithum
- Department of Pharmaceutical Botany, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand
- Le Studium Institute for Advanced Studies, 1 Rue Dupanloup, 45000 Orléans, France
| |
Collapse
|
2
|
Ononiwu CP, Joshua PE, Amah CC, Asomadu RO, Okorigwe EM, Nnemolisa CS, Ezeorba TPC, Nwanelo VO, Iyidiegwu FC, Duru JO, Okeke PN, Adiele OB. Cleistopholis patens root bark extract exerts cardioprotective effect against doxorubicin-induced myocardial toxicity in rats. Lab Anim Res 2024; 40:39. [PMID: 39551811 PMCID: PMC11572060 DOI: 10.1186/s42826-024-00225-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 10/17/2024] [Accepted: 10/26/2024] [Indexed: 11/19/2024] Open
Abstract
BACKGROUND Myocardial Infarction still persists as the most prevalent cardiovascular disease and is a top cause of morbidity and mortality in doxorubicin treated cancer patients. This study evaluated the prophylactic effect of the ethanol root bark extract of Cleistopholis patens (ERBECP) against doxorubicin-induced myocardial infarction in wistar rats. Extraction, preliminary phytochemical analysis, acute toxicity study and body weight (b.w.) of ERBECP were achieved using standard methods. Phyto-constituents in ERBECP were indentified using Gas Chromatography-Mass Spectrometry (GC-MS) technique. Thirty (30) male albino Wistar rats of average b.w. ranging between 100 and 130 g were divided into six groups of five rats each. Groups I, II and III served as normal, doxorubicin (DOX) and standard (Vasoprin 150 mg/kg b.w) controls respectively, while groups IV, V and VI were orally pre-treated with the extract (200, 400 and 600 mg/kgb.w) for two weeks prior to intraperitoneal induction of cardiotoxicity with DOX (20 mg/kg bw) on day 14. RESULTS Disturbances in serum cardiac function bio-markers such as; Cardiac Troponin-I (CTnI), Creatine Kinase (CK), Lactate Dehydrogenase (LDH), Aspartate aminotransferase (AST), Alanine aminotransferase (ALT). Lipid profile markers such as; Total cholesterol (TC), Triacylglycerol (TAG), Low Density Lipoprotein (LDL), High Density Lipoprotein (HDL). Oxidative stress markers such as; Malondialdehyde (MDA), Superoxide Dismutase (SOD), Catalase (CAT), Glutathione (GSH) confirmed the induction of myocardial infarction. Histological assessment of heart tissues was performed to validate biochemical results. The GC-MS analysis of ERBECP identified a total of 69 compounds. Safety profile of the aqueous extract was safe for the animals up to the highest dose of 5000 mg/kg b.w. Pre-treatment of DOX group with ERBECP could significantly increase the b.w. compared to the DOX-treated group during the experimental period of 2 weeks. There were significant (p < 0.05) alterations in the levels of CTnI, CK, LDH, AST, ALT and lipid profile indices in the DOX control rats. Also, significant (p < 0.05) increase was observed in MDA and decrease in SOD, CAT and GSH in the DOX control rats. However, administration of the extract significantly (p < 0.05) normalized these alterations and reversed the architectural changes in the heart. The 69 compounds were screened against the target protein (CBR1); we identified seven hits based on the docking score and interactions with the active site residues. All the C. patens constituents had MW (g/mol) less than 500, HBA < 10 and HBD not more than 5. Apart, 9-Octadecenoic acid (Z)-, 2,3-dihydroxy propyl ester and Estra-1,3,5(10)-trien-17. beta. -ol, all the constituents had LD50 lower than 2000 mg/kg. CONCLUSIONS The findings reveals ERBECP demonstrated promising potential and can be exploited in the development novel cardiac therapeutic agents.
Collapse
Affiliation(s)
- Chidinma Pamela Ononiwu
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Nsukka, 410001, Enugu State, Nigeria
| | - Parker Elijah Joshua
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Nsukka, 410001, Enugu State, Nigeria
| | - Christian Chijioke Amah
- Department of Biochemistry, Faculty of Natural and Applied Sciences, State University of Medical and Applied Sciences Igbo-Eno, Nsukka, Enugu State, Nigeria.
| | - Rita Onyekachukwu Asomadu
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Nsukka, 410001, Enugu State, Nigeria
| | - Ekezie Matthew Okorigwe
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Nsukka, 410001, Enugu State, Nigeria
| | | | - Timothy Prince Chidike Ezeorba
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Nsukka, 410001, Enugu State, Nigeria
- Department of Molecular Biotechnology, School of Biosciences, University of Birmingham, Edgbaston, B15 2TT, UK
| | | | - Favour Chinagorom Iyidiegwu
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Nsukka, 410001, Enugu State, Nigeria
| | - Justin Onuawuchi Duru
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Nsukka, 410001, Enugu State, Nigeria
| | - Peace Nkiruka Okeke
- Department of Biotechnology, School of Medicine, 3900 University Blvd., Tyler, TX, 75799, USA
| | - Onyinyechi Becky Adiele
- Department of Science Laboratory Technology, Faculty of Physical Sciences, University of Nigeria, Nsukka, 410001, Enugu State, Nigeria
| |
Collapse
|
3
|
Kuşi M, Becer E, Vatansever HS. Basic approach on the protective effects of hesperidin and naringin in Alzheimer's disease. Nutr Neurosci 2024:1-13. [PMID: 39225173 DOI: 10.1080/1028415x.2024.2397136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
OBJECTIVES Alzheimer's disease (AD) is a neurodegenerative disease characterized by cognitive impairment. This situation imposes a great burden on individuals, both economically and socially. Today, an effective method for treating the disease and protective approach to tau accumulation has not been developed yet. Studies have been conducted on the effects of hesperidin and naringin flavonoids found in citrus fruits on many diseases. METHODS In this review, the pathophysiology of AD is defined, and the effects of hesperidin and naringin on these factors are summarized. RESULTS Studies have shown that both components may potentially affect AD due to their antioxidative and anti-inflammatory properties. Based on these effects of the components, it has been shown that they may have ameliorative effects on Aβ, α-synuclein aggregation, tau pathology, and cognitive functions in the pathophysiology of AD. DISCUSSION There are studies suggesting that hesperidin and naringin may be effective in the prevention/treatment of AD. When these studies are examined, it is seen that more studies should be conducted on the subject.
Collapse
Affiliation(s)
- Müjgan Kuşi
- Faculty of Health Sciences, Department of Nutrition and Dietetics, Near East University, Nicosia, North Cyprus via Mersin 10, Turkey
- Research Center for Science, Technology and Engineering (BILTEM), Near East University, Nicosia, North Cyprus via Mersin 10, Turkey
| | - Eda Becer
- Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, North Cyprus via Mersin 10, Turkey
| | - Hafize Seda Vatansever
- DESAM Institute, Near East University, Nicosia, North Cyprus via Mersin 10, Turkey
- Faculty of Medicine, Department of Histology and Embryology, Manisa Celal Bayar University, Manisa, Turkey
| |
Collapse
|
4
|
Chen Y, Yang W, Cui X, Zhang H, Li L, Fu J, Guo H. Research Progress on the Mechanism, Monitoring, and Prevention of Cardiac Injury Caused by Antineoplastic Drugs-Anthracyclines. BIOLOGY 2024; 13:689. [PMID: 39336116 PMCID: PMC11429024 DOI: 10.3390/biology13090689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 08/28/2024] [Accepted: 09/02/2024] [Indexed: 09/30/2024]
Abstract
Anthracyclines represent a highly efficacious class of chemotherapeutic agents employed extensively in antitumor therapy. They are universally recognized for their potency in treating diverse malignancies, encompassing breast cancer, gastrointestinal tumors, and lymphomas. Nevertheless, the accumulation of anthracyclines within the body can lead to significant cardiac toxicity, adversely impacting both the survival rates and quality of life for tumor patients. This limitation somewhat restricts their clinical utilization. Determining how to monitor and mitigate their cardiotoxicity at an early stage has become an urgent clinical problem to be solved. Therefore, this paper reviews the mechanism of action, early monitoring, and strategies for the prevention of anthracycline-induced cardiotoxicity for clinical reference.
Collapse
Affiliation(s)
- Yuanyuan Chen
- Graduate School, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Wenwen Yang
- Graduate School, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Department of Cardiology, Shaanxi Academy of Traditional Chinese Medicine, Xi'an 710005, China
| | - Xiaoshan Cui
- Graduate School, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Huiyu Zhang
- Graduate School, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Liang Li
- Graduate School, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Jianhua Fu
- Graduate School, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Hao Guo
- Graduate School, China Academy of Chinese Medical Sciences, Beijing 100700, China
| |
Collapse
|
5
|
Yaseen HS, Zubair HM, Jamal A, Farrukh M, Mikrani R, Shaukat B, Hill JW, Rana R, Nazir A, Naveed M, Malik S. Naringin: Cardioprotective properties and safety profile in diabetes treatment. Fitoterapia 2024; 176:106011. [PMID: 38740344 DOI: 10.1016/j.fitote.2024.106011] [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: 11/04/2023] [Revised: 04/19/2024] [Accepted: 05/11/2024] [Indexed: 05/16/2024]
Abstract
Flavonoids derived from plants offer a broad spectrum of therapeutic potential for addressing metabolic syndrome, particularly diabetes mellitus (DM), a prevalent non-communicable disease. Hyperglycemia in DM is a known risk factor for cardiovascular diseases (CVDs), which substantially impact global mortality rates. This review examines the potential effects of naringin, a citrus flavonoid, on both DM and its associated cardiovascular complications, including conditions like diabetic cardiomyopathy. The safety profile of naringin is summarized based on various pre-clinical studies. The data for this review was gathered from diverse electronic databases, including Medline, PubMed, ScienceDirect, SpringerLink, Google Scholar, and Emerald Insight. Multiple pre-clinical studies have demonstrated that naringin exerts hypoglycemic and cardioprotective effects by targeting various vascular mechanisms. Specifically, research indicates that naringin down-regulates the renin-angiotensin and oxidative stress systems while concurrently upregulating β-cell and immune system functions. Clinical trial outcomes also support the therapeutic potential of naringin in managing hyperglycemic states and associated cardiovascular issues. Moreover, toxicity studies have confirmed the safety of naringin in animal models, suggesting its potential for safe administration in humans. In conclusion, naringin emerges as a promising natural candidate for both antidiabetic and cardioprotective purposes, offering potential improvements in health outcomes. While naringin presents a new avenue for therapies targeting DM and CVDs, additional controlled and long-term clinical trials are necessary to validate its efficacy and safety for human use.
Collapse
Affiliation(s)
| | - Hafiz Muhammad Zubair
- Post-Graduate Medical College, Faculty of Medicine and Allied Health Sciences, The Islamia University of Bahawalpur, Bahawalpur, Pakistan.
| | - Adil Jamal
- Sciences and Research, College of Nursing, Umm Al Qura University, Makkah 715, Saudi Arabia
| | - Maryam Farrukh
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Pakistan
| | - Reyaj Mikrani
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Bushra Shaukat
- Sciences and Research, College of Nursing, Umm Al Qura University, Makkah 715, Saudi Arabia
| | - Jennifer W Hill
- Department of Pharmacology and Physiology, School of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Reemal Rana
- Sciences and Research, College of Nursing, Umm Al Qura University, Makkah 715, Saudi Arabia
| | - Ansa Nazir
- Faculty of Pharmacy, The University of Lahore, Lahore 54000, Pakistan
| | - Muhammad Naveed
- Department of Pharmacology and Physiology, School of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Samiullah Malik
- Post-Graduate Medical College, Faculty of Medicine and Allied Health Sciences, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| |
Collapse
|
6
|
Liu S, Zhong M, Wu H, Su W, Wang Y, Li P. Potential Beneficial Effects of Naringin and Naringenin on Long COVID-A Review of the Literature. Microorganisms 2024; 12:332. [PMID: 38399736 PMCID: PMC10892048 DOI: 10.3390/microorganisms12020332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) caused a severe epidemic due to severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). Recent studies have found that patients do not completely recover from acute infections, but instead, suffer from a variety of post-acute sequelae of SARS-CoV-2 infection, known as long COVID. The effects of long COVID can be far-reaching, with a duration of up to six months and a range of symptoms such as cognitive dysfunction, immune dysregulation, microbiota dysbiosis, myalgic encephalomyelitis/chronic fatigue syndrome, myocarditis, pulmonary fibrosis, cough, diabetes, pain, reproductive dysfunction, and thrombus formation. However, recent studies have shown that naringenin and naringin have palliative effects on various COVID-19 sequelae. Flavonoids such as naringin and naringenin, commonly found in fruits and vegetables, have various positive effects, including reducing inflammation, preventing viral infections, and providing antioxidants. This article discusses the molecular mechanisms and clinical effects of naringin and naringenin on treating the above diseases. It proposes them as potential drugs for the treatment of long COVID, and it can be inferred that naringin and naringenin exhibit potential as extended long COVID medications, in the future likely serving as nutraceuticals or clinical supplements for the comprehensive alleviation of the various manifestations of COVID-19 complications.
Collapse
Affiliation(s)
- Siqi Liu
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-Evaluation of Post-Market Traditional Chinese Medicine, State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China; (S.L.); (M.Z.); (H.W.); (W.S.); (Y.W.)
| | - Mengli Zhong
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-Evaluation of Post-Market Traditional Chinese Medicine, State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China; (S.L.); (M.Z.); (H.W.); (W.S.); (Y.W.)
| | - Hao Wu
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-Evaluation of Post-Market Traditional Chinese Medicine, State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China; (S.L.); (M.Z.); (H.W.); (W.S.); (Y.W.)
| | - Weiwei Su
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-Evaluation of Post-Market Traditional Chinese Medicine, State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China; (S.L.); (M.Z.); (H.W.); (W.S.); (Y.W.)
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming 525000, China
| | - Yonggang Wang
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-Evaluation of Post-Market Traditional Chinese Medicine, State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China; (S.L.); (M.Z.); (H.W.); (W.S.); (Y.W.)
| | - Peibo Li
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Re-Evaluation of Post-Market Traditional Chinese Medicine, State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China; (S.L.); (M.Z.); (H.W.); (W.S.); (Y.W.)
| |
Collapse
|
7
|
Tang X, Liu H, Rao R, Huang Y, Dong M, Xu M, Feng S, Shi X, Wang L, Wang Z, Zhou B. Modeling drug-induced mitochondrial toxicity with human primary cardiomyocytes. SCIENCE CHINA. LIFE SCIENCES 2024; 67:301-319. [PMID: 37864082 DOI: 10.1007/s11427-023-2369-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 05/16/2023] [Indexed: 10/22/2023]
Abstract
Mitochondrial toxicity induced by therapeutic drugs is a major contributor for cardiotoxicity, posing a serious threat to pharmaceutical industries and patients' lives. However, mitochondrial toxicity testing is not incorporated into routine cardiac safety screening procedures. To accurately model native human cardiomyocytes, we comprehensively evaluated mitochondrial responses of adult human primary cardiomyocytes (hPCMs) to a nucleoside analog, remdesivir (RDV). Comparison of their response to human pluripotent stem cell-derived cardiomyocytes revealed that the latter utilized a mitophagy-based mitochondrial recovery response that was absent in hPCMs. Accordingly, action potential duration was elongated in hPCMs, reflecting clinical incidences of RDV-induced QT prolongation. In a screen for mitochondrial protectants, we identified mitochondrial ROS as a primary mediator of RDV-induced cardiotoxicity. Our study demonstrates the utility of hPCMs in the detection of clinically relevant cardiac toxicities, and offers a framework for hPCM-based high-throughput screening of cardioprotective agents.
Collapse
Affiliation(s)
- Xiaoli Tang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China
| | - Hong Liu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China
| | - Rongjia Rao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China
| | - Yafei Huang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China
| | - Mengqi Dong
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China
| | - Miaomiao Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China
| | - Shanshan Feng
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China
| | - Xun Shi
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China
| | - Li Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, Shenzhen, 518020, China
| | - Zengwu Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China
- Department of Epidemiology, Cardiovascular Institute and Fuwai Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100037, China
| | - Bingying Zhou
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China.
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, Shenzhen, 518020, China.
| |
Collapse
|
8
|
Adeyemi DH, Obembe OO, Hamed MA, Akhigbe RE. Sodium acetate ameliorates doxorubicin-induced cardiac injury via upregulation of Nrf2/HO-1 signaling and downregulation of NFkB-mediated apoptotic signaling in Wistar rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:423-435. [PMID: 37458777 DOI: 10.1007/s00210-023-02620-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 07/10/2023] [Indexed: 01/07/2024]
Abstract
Despite the effectiveness of doxorubicin (DOX) in the management of a wide range of cancers, a major challenge is its cardio-toxic effect. Oxidative stress, inflammation, and apoptosis are major pathways for the cardiotoxic effect of DOX. On the other hand, acetate reportedly exerts antioxidant, anti-inflammatory, and anti-apoptotic activities. This particular research assessed the impact of acetate on cardiotoxicity induced by DOX. Mechanistically, acetate dramatically inhibited DOX-induced upregulation of xanthine oxidase and uric acid pathway as well as downregulation of Nrf2/HO-1 signaling and its upstream proteins (reduced glutathione peroxidase, superoxide dismutase, glutathione-S-transferase, glutathione, and catalase, glutathione reductase). In addition, acetate markedly attenuated DOX-driven rise inTNF-α, NFkB IL-6 and IL-1β expression, and myeloperoxidase activity. Furthermore, acetate significantly ameliorated DOX-led suppression of Bcl-2 and Ca2+-ATPase activity and upregulation of Bax, caspase 3, and caspase 9 actions. Improved body weight, heart structural integrity, and cardiac function as depicted by cardiac injury markers convoyed these cascades of events. Summarily, the present study demonstrated that acetate protects against DOX-induced cardiotoxicity by upregulating Nrf2/HO-1 signaling and downregulating NFkB-mediated activation of Bax/Bcl-2 and caspase signaling.
Collapse
Affiliation(s)
- D H Adeyemi
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Osun State University, Osogbo, Osun State, Nigeria
| | - O O Obembe
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Osun State University, Osogbo, Osun State, Nigeria
| | - M A Hamed
- Department of Medical Laboratory Sciences, Afe Babalola University, Ado Ekiti, Ekiti State, Nigeria
- The Brainwill Laboratories, Osogbo, Osun State, Nigeria
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Osun State, Nigeria
| | - R E Akhigbe
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Osun State, Nigeria.
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria.
| |
Collapse
|
9
|
Zaafar D, Khalil HMA, Elnaggar R, Saad DZ, Rasheed RA. Protective role of hesperetin in sorafenib-induced hepato- and neurotoxicity in mice via modulating apoptotic pathways and mitochondrial reprogramming. Life Sci 2024; 336:122295. [PMID: 38007145 DOI: 10.1016/j.lfs.2023.122295] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/20/2023] [Accepted: 11/22/2023] [Indexed: 11/27/2023]
Abstract
INTRODUCTION Sorafenib, an FDA-approved standard chemotherapy for advanced hepatocellular carcinoma, is associated with numerous adverse effects that significantly impact patients' physiological well-being. Consequently, identifying agents that mitigate these side effects while enhancing efficacy is crucial. Hesperetin, a flavone present in fruits and vegetables, possesses antioxidant, anti-inflammatory, and anti-cancer properties. This study aimed to investigate the hepatotoxic and neurotoxic effects of sorafenib and the potential protective role of hesperetin. MATERIALS AND METHODS Swiss albino mice were orally administered sorafenib (100 mg/kg) alone or in combination with hesperetin (50 mg/kg) over 21 days. Behavioral assessments for anxiety and depressive-like behaviors were conducted. Additionally, evaluations encompassed apoptotic activity, mitochondrial integrity, liver enzyme levels, proliferation rates, and histopathological changes. RESULTS Combining hesperetin with sorafenib showed improvements in behavioral alterations, liver damage, brain mitochondrial dysfunction, and liver apoptosis compared to the sorafenib-only group in mice. CONCLUSION Hesperetin exhibits potential as an adjunct to sorafenib, mitigating its side effects by attenuating its toxicity, enhancing efficacy, and potentially reducing the occurrence of sorafenib-induced resistance through the downregulation of hepatocyte growth factor levels.
Collapse
Affiliation(s)
- Dalia Zaafar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Modern University, Cairo, Egypt.
| | - Heba M A Khalil
- Department of Veterinary Hygiene and Management, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt.
| | - Reham Elnaggar
- Department of Pharmacology and Toxicology, College of Pharmacy and Drug Manufacturing, Misr University of Science and Technology (MUST), 6th October, Giza 12566, Egypt.
| | - Diana Z Saad
- Department of Pathology, Faculty of Medicine, Ain Shams University, Cairo, Egypt; Department of Basic Medical Sciences, Faculty of Medicine, King Salman International University, South Sinai, Egypt.
| | - Rabab Ahmed Rasheed
- Department of Medical Histology and Cell Biology, Faculty of Medicine, King Salman International University, South Sinai, Egypt.
| |
Collapse
|
10
|
Liao Y, Meng Q. Protection against cancer therapy-induced cardiovascular injury by planed-derived polyphenols and nanomaterials. ENVIRONMENTAL RESEARCH 2023; 238:116896. [PMID: 37586453 DOI: 10.1016/j.envres.2023.116896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/18/2023] [Accepted: 08/13/2023] [Indexed: 08/18/2023]
Abstract
Cancer therapy-induced heart injury is a significant concern for cancer patients undergoing chemotherapy, radiotherapy, immunotherapy, and also targeted molecular therapy. The use of these treatments can lead to oxidative stress and cardiomyocyte damage in the heart, which can result in heart failure and other cardiac complications. Experimental studies have revealed that chemotherapy drugs such as doxorubicin and cyclophosphamide can cause severe side effects such as cardiac fibrosis, electrophysiological remodeling, chronic oxidative stress and inflammation, etc., which may increase risk of cardiac disorders and attacks for patients that underwent chemotherapy. Similar consequences may also be observed for patients that undergo radiotherapy for left breast or lung malignancies. Polyphenols, a group of natural compounds with antioxidant and anti-inflammatory properties, have shown the potential in protecting against cancer therapy-induced heart injury. These compounds have been found to reduce oxidative stress, necrosis and apoptosis in the heart, thereby preserving cardiac function. In recent years, nanoparticles loaded with polyphenols have also provided for the delivery of these compounds and increasing their efficacy in different organs. These nanoparticles can improve the bioavailability and efficacy of polyphenols while minimizing their toxicity. This review article summarizes the current understanding of the protective effects of polyphenols and nanoparticles loaded with polyphenols against cancer therapy-induced heart injury. The article discusses the mechanisms by which polyphenols protect the heart, including antioxidant and anti-inflammation abilities. The article also highlights the potential benefits of using nanoparticles for the delivery of polyphenols.
Collapse
Affiliation(s)
- Yunshu Liao
- Department of Cardiac Surgery, The First Hospital Affiliated to the Army Medical University, Chongqing, 400038, China
| | - Qinghua Meng
- Department of Cardiac Surgery, The First Hospital Affiliated to the Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
11
|
Gupta T, Najumuddin, Rajendran D, Gujral A, Jangra A. Metabolism configures immune response across multi-systems: Lessons from COVID-19. Adv Biol Regul 2023; 90:100977. [PMID: 37690286 DOI: 10.1016/j.jbior.2023.100977] [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: 03/10/2023] [Revised: 07/19/2023] [Accepted: 08/16/2023] [Indexed: 09/12/2023]
Abstract
Several studies over the last decade demonstrate the recruitment of immune cells, increased inflammatory cytokines, and chemokine in patients with metabolic diseases, including heart failure, parenchymal inflammation, obesity, tuberculosis, and diabetes mellitus. Metabolic rewiring of immune cells is associated with the severity and prevalence of these diseases. The risk of developing COVID-19/SARS-CoV-2 infection increases in patients with metabolic dysfunction (heart failure, diabetes mellitus, and obesity). Several etiologies, including fatigue, dyspnea, and dizziness, persist even months after COVID-19 infection, commonly known as Post-Acute Sequelae of CoV-2 (PASC) or long COVID. A chronic inflammatory state and metabolic dysfunction are the factors that contribute to long COVID. Here, this study explores the potential link between pathogenic metabolic and immune alterations across different organ systems that could underlie COVID-19 and PASC. These interactions could be utilized for targeted future therapeutic approaches.
Collapse
Affiliation(s)
- Tinku Gupta
- Department of Pharmacognosy & Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard (Deemed University), M. B. Road, New Delhi 110062, India
| | - Najumuddin
- Program of Biotechnology, Department of Applied Sciences, Faculty of Engineering, Science and Technology, Hamdard University, Karachi, Pakistan
| | - Dhanya Rajendran
- Rajiv Gandhi Centre for Biotechnology, Trivandrum, Kerala, 695014, India
| | - Akash Gujral
- Department of Medicine, Nyu Grossman School of Medicine, NY, USA
| | - Ashok Jangra
- Department of Pharmaceutical Sciences, Central University of Haryana, Mahendergarh, Haryana, India.
| |
Collapse
|
12
|
Cicek B, Hacimuftuoglu A, Yeni Y, Danisman B, Ozkaraca M, Mokhtare B, Kantarci M, Spanakis M, Nikitovic D, Lazopoulos G, Tsarouhas K, Tsatsakis A, Taghizadehghalehjoughi A. Chlorogenic Acid Attenuates Doxorubicin-Induced Oxidative Stress and Markers of Apoptosis in Cardiomyocytes via Nrf2/HO-1 and Dityrosine Signaling. J Pers Med 2023; 13:jpm13040649. [PMID: 37109035 PMCID: PMC10140899 DOI: 10.3390/jpm13040649] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/26/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
(1) Background: Doxorubicin (DOX) is extensively used for cancer treatments; however, its clinical application is limited because of its cardiotoxic adverse effects. A combination of DOX and agents with cardioprotective properties is an effective strategy to ameliorate DOX-related cardiotoxicity. Polyphenolic compounds are ideal for the investigation of novel cardioprotective agents. Chlorogenic acid (CGA), an essential dietary polyphenol found in plants, has been previously reported to exert antioxidant, cardioprotective, and antiapoptotic properties. The current research evaluated CGA's in vivo cardioprotective properties in DOX-induced cardiotoxicity and the probable mechanisms underlying this protection. (2) Methods: CGA's cardioprotective properties were investigated in rats that were treated with CGA (100 mg/kg, p.o.) for fourteen days. The experimental model of cardiotoxicity was induced with a single intraperitoneal (15 mg/kg i.p.) injection of DOX on the 10th day. (3) Results: Treatment with CGA significantly improved the DOX-caused altered cardiac damage markers (LDH, CK-MB, and cTn-T), and a marked improvement in cardiac histopathological features accompanied this. DOX downregulated the expression of Nrf2/HO-1 signaling pathways, and the CGA reversed this effect. Consistently, caspase-3, an apoptotic-related marker, and dityrosine expression were suppressed, while Nrf2 and HO-1 expressions were elevated in the cardiac tissues of DOX-treated rats after treatment with the CGA. Furthermore, the recovery was confirmed by the downregulation of 8-OHdG and dityrosine (DT) expressions in immunohistochemical findings. (4) Conclusions: CGA demonstrated a considerable cardioprotective effect against DOX-induced cardiotoxicity. One of the possible mechanisms for these protective properties was the upregulation of the Nrf2/HO-1-dependent pathway and the downregulation of DT, which may ameliorate oxidative stress and cardiomyocyte apoptosis. These findings suggest that CGA may be cardioprotective, particularly in patients receiving DOX-based chemotherapy.
Collapse
Affiliation(s)
- Betul Cicek
- Department of Physiology, Faculty of Medicine, Erzincan Binali Yildirim University, 24100 Erzincan, Turkey
| | - Ahmet Hacimuftuoglu
- Department of Medical Pharmacology, Faculty of Medicine, Ataturk University, 25240 Erzurum, Turkey
| | - Yesim Yeni
- Department of Medical Pharmacology, Faculty of Medicine, Malatya Turgut Ozal University, 44210 Malatya, Turkey
| | - Betul Danisman
- Department of Biophysics, Faculty of Medicine, Ataturk University, 25240 Erzurum, Turkey
| | - Mustafa Ozkaraca
- Department of Pathology, Faculty of Veterinary, Sivas Cumhuriyet University, 58140 Sivas, Turkey
| | - Behzad Mokhtare
- Department of Pathology, Faculty of Veterinary, Sivas Cumhuriyet University, 58140 Sivas, Turkey
| | - Mecit Kantarci
- Department of Radiology, Faculty of Medicine, Ataturk University, 25240 Erzurum, Turkey
| | - Marios Spanakis
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Dragana Nikitovic
- Dragana Nikitovic, Laboratory of Histology-Embryology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Georgios Lazopoulos
- Department of Cardiac Surgery, University General Hospital of Heraklion, Medical School, University of Crete, 71003 Heraklion, Greece
| | | | - Aristidis Tsatsakis
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Ali Taghizadehghalehjoughi
- Department of Medical Pharmacology, Faculty of Medicine, Bilecik Seyh Edebali University, 11230 Bilecik, Turkey
| |
Collapse
|
13
|
Mishra A, Singla R, Kumar R, Sharma A, Joshi R, Sarma P, Kaur G, Prajapat M, Bhatia A, Medhi B. Granulocyte Colony-Stimulating Factor Improved Core Symptoms of Autism Spectrum Disorder via Modulating Glutamatergic Receptors in the Prefrontal Cortex and Hippocampus of Rat Brains. ACS Chem Neurosci 2022; 13:2942-2961. [PMID: 36166499 DOI: 10.1021/acschemneuro.2c00270] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Chronic neuroinflammation-induced anomalous glutamate receptor activation has been identified as one of the important factors in the pathogenesis of autism spectrum disorder (ASD). Thus, the current study was designed to elucidate the neuroprotective effect of the granulocyte colony-stimulating factor (G-CSF), a haemopoietic growth factor, an anti-inflammatory, and a neuroprotectant to decipher the underlying mechanism(s) in the valproic acid (VPA)-induced experimental model of ASD. Experimentally, the ASD rat model was induced by a single dose of VPA (600 mg/kg; i.p.) on gestation day 12.5 to the pregnant female rats. After birth, pups were treated with vehicle, normal saline 0.9% i.p., risperidone (2.5 mg/kg; i.p.), and G-CSF (10, 35, and 70 μg/kg; i.p.) from postnatal day (PND) 23 to 43. All the groups were subjected to various developmental and behavior tests from birth. The rats were sacrificed on PND 55, and their brain was excised and processed for biochemical parameters (oxidative stress, inflammatory markers, BDNF), histological examination (H&E, Nissl staining), NMDA, and AMPA receptor expression by immunohistochemistry, western blot, and real-time polymerase chain reaction evaluation. Also, the possible interaction of the G-CSF with NMDA and AMPA receptors was evaluated using the in-silico method. The results of the study showed that in VPA-exposed rats, postnatal treatment of G-CSF rescued all the behavioral abnormalities, oxidative stress, and inflammatory parameters in a dose-dependent manner while risperidone did not show any significant results. The in-silico analysis showed the direct interaction of G-CSF with NMDA and AMPA receptors. The upregulated expression of NMDA and AMPA both in the prefrontal cortex as well as hippocampus was alleviated by G-CSF thereby validating its anti-inflammatory and excitoprotective properties. Thus, G-CSF demonstrated neuroprotection against the core symptoms of autism in the VPA-induced rodent model, making it a potential candidate for the treatment of ASD.
Collapse
Affiliation(s)
- Abhishek Mishra
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh160012, India
| | - Rubal Singla
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh160012, India
| | - Rohit Kumar
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh160012, India
| | - AmitRaj Sharma
- Department of Neurology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh160012, India
| | - Rupa Joshi
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh160012, India
| | - Phulen Sarma
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh160012, India
| | - Gurjeet Kaur
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh160012, India
| | - Manisha Prajapat
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh160012, India
| | - Alka Bhatia
- Department of Experimental Medicine and Biotechnology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh160012, India
| | - Bikash Medhi
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh160012, India
| |
Collapse
|
14
|
The Preventive Effects of Naringin and Naringenin against Paclitaxel-Induced Nephrotoxicity and Cardiotoxicity in Male Wistar Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:8739815. [PMID: 36212979 PMCID: PMC9546692 DOI: 10.1155/2022/8739815] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/14/2022] [Accepted: 08/28/2022] [Indexed: 11/17/2022]
Abstract
This study assessed the preventive properties of naringin and naringenin on paclitaxel-induced nephrotoxicity and cardiotoxicity in adult male Wistar rats. Intraperitoneal injection of paclitaxel 2 mg/kg body weight, two days/week on the 2nd and 5th days of each week, with or without oral administration of naringin and/or naringenin 10 mg/kg body weight every other day, was continued for six weeks. Treatment of rats with naringin and/or naringenin significantly reversed elevated serum creatinine, urea, and uric acid levels caused by paclitaxel, reflecting improved kidney function. Similarly, heart dysfunction induced by paclitaxel was alleviated after treatment with naringin and/or naringenin, as evidenced by significant decreases in elevated CK-MB and LDH activities. After drug administration, histopathological findings and lesion scores in the kidneys and heart were markedly decreased by naringin and/or naringenin. Moreover, the treatments reversed renal and cardiac lipid peroxidation and the negative impacts on antioxidant defenses via raising GSH, SOD, and GPx. The preventive effects of naringin and naringenin were associated with suppressing oxidative stress and reestablishing antioxidant defenses. A combination of naringin and naringenin was the most efficacious in rescuing organ function and structure.
Collapse
|
15
|
Azmy AM, Abd Elbaki BT, Ali MA, Mahmoud AA. Effect of ozone versus naringin on testicular injury in experimentally induced ulcerative colitis in adult male albino rats. Ultrastruct Pathol 2022; 46:439-461. [DOI: 10.1080/01913123.2022.2132337] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Abeer M. Azmy
- Medical Histology and Cell Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Bassant T. Abd Elbaki
- Medical Histology and Cell Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Mohammed A. Ali
- Medical Histology and Cell Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Abeer A Mahmoud
- Medical Histology and Cell Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
16
|
Islam J, Shree A, Khan HA, Sultana S. Chemopreventive potential of Diosmin against benzo[a]pyrene induced lung carcinogenesis in Swiss Albino mice. J Biochem Mol Toxicol 2022; 36:e23187. [PMID: 35920545 DOI: 10.1002/jbt.23187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 04/28/2022] [Accepted: 07/25/2022] [Indexed: 12/24/2022]
Abstract
Lung cancer, one of the most common cancer is a cause of concern associated with cancer-related mortality. Benzo[a]pyrene [B(a)P], a potent carcinogen as well as an environmental contaminant is reported to be found in cigarette smoke among various sources. The present study focuses on the chemopreventive potential of Diosmin against B[a]P-induced lung carcinogenesis and its possible mechanism in male Swiss Albino mice (SAM). SAM were treated orally with Diosmin (200 mg/kg b.w.) for 16 weeks and/or B[a]P (50 mg/kg b.w) for a period of 4 weeks. B[a]P treated cancerous mice showed increased peroxidation of membrane lipid as well as a decrease in the level/activity of antioxidant proteins. Cancerous mice also showed an increased level of carcinoembryonic antigen (CEA) and neuron-specific enolase (NSE). Diosmin treatment, however, leads to decreased peroxidation of lipids, increased antioxidant proteins as well decrease in the level of CEA and NSE. B[a]P-induced cancerous animals also exhibited increased expression of cyclic AMP response element-binding protein (CREB), COX2 as well as prostaglandin-E2 (PGE2) while Diosmin-treated mice were found to have an ameliorative effect. Histopathological results further confirm the protective effect of Diosmin in averting B[a]P-induced pathological alterations of lung tissue. Overall, our results suggest Diosmin exerts its chemopreventive potential possibly via targeting the CREB/cyclooxygenase-2 (COX-2)/PGE2 pathway thereby repressing inflammation.
Collapse
Affiliation(s)
- Johirul Islam
- Department of Toxicology, Jamia Hamdard, New Delhi, Delhi, India
| | - Alpa Shree
- Department of Toxicology, Jamia Hamdard, New Delhi, Delhi, India
| | - Haider Ali Khan
- Department of Toxicology, Jamia Hamdard, New Delhi, Delhi, India
| | - Sarwat Sultana
- Department of Toxicology, Jamia Hamdard, New Delhi, Delhi, India
| |
Collapse
|
17
|
Syahputra RA, Harahap U, Dalimunthe A, Nasution MP, Satria D. The Role of Flavonoids as a Cardioprotective Strategy against Doxorubicin-Induced Cardiotoxicity: A Review. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27041320. [PMID: 35209107 PMCID: PMC8878416 DOI: 10.3390/molecules27041320] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 01/26/2022] [Accepted: 02/02/2022] [Indexed: 12/14/2022]
Abstract
Doxorubicin is a widely used and promising anticancer drug; however, a severe dose-dependent cardiotoxicity hampers its therapeutic value. Doxorubicin may cause acute and chronic issues, depending on the duration of toxicity. In clinical practice, the accumulative toxic dose is up to 400 mg/m2 and increasing the dose will increase the probability of cardiac toxicity. Several molecular mechanisms underlying the pathogenesis of doxorubicin cardiotoxicity have been proposed, including oxidative stress, topoisomerase beta II inhibition, mitochondrial dysfunction, Ca2+ homeostasis dysregulation, intracellular iron accumulation, ensuing cell death (apoptosis and necrosis), autophagy, and myofibrillar disarray and loss. Natural products including flavonoids have been widely studied both in cell, animal, and human models which proves that flavonoids alleviate cardiac toxicity caused by doxorubicin. This review comprehensively summarizes cardioprotective activity flavonoids including quercetin, luteolin, rutin, apigenin, naringenin, and hesperidin against doxorubicin, both in in vitro and in vivo models.
Collapse
Affiliation(s)
- Rony Abdi Syahputra
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan 20155, Indonesia;
- Correspondence: (R.A.S.); (U.H.)
| | - Urip Harahap
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan 20155, Indonesia;
- Correspondence: (R.A.S.); (U.H.)
| | - Aminah Dalimunthe
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan 20155, Indonesia;
| | - M. Pandapotan Nasution
- Department of Pharmaceutical Biology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan 20155, Indonesia; (M.P.N.); (D.S.)
| | - Denny Satria
- Department of Pharmaceutical Biology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan 20155, Indonesia; (M.P.N.); (D.S.)
| |
Collapse
|
18
|
Asiedu-Gyekye IJ, Arhin E, Arthur SA, N'guessan BB, Amponsah SK. Genotoxicity, nitric oxide level modulation and cardio-protective potential of Kalanchoe Integra Var. Crenata (Andr.) Cuf Leaves in murine models. JOURNAL OF ETHNOPHARMACOLOGY 2022; 283:114640. [PMID: 34606947 DOI: 10.1016/j.jep.2021.114640] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/10/2021] [Accepted: 09/11/2021] [Indexed: 02/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Advancement in cancer therapy has improved survival among patients. However, use of anticancer drugs like anthracyclines (e.g., doxorubicin) is not without adverse effects. Notable among adverse effects of doxorubicin (DOX) is cardiotoxicity, which ranges from mild transient blood pressure changes to potentially serious heart failure. Anecdotal reports suggest that Kalanchoe integra (KI) may have cardio-protective potential. AIMS OF THE STUDY This study sought to determine the cardio-protective potential of KI against doxorubicin-induced cardiotoxicity and also examined any possible genotoxic potential of KI in selected organs. Additionally, the nitric oxide modulatory potential of KI was assessed. MATERIALS AND METHODS The leaves of KI were collected, air-dried, pulverised and extracted using 70% ethanol. High-performance liquid chromatography (HPLC) fingerprinting was done for KI. Also, the single-cell gel electrophoresis assay (Comet assay) was employed to ascertain the genotoxic potential of KI. In assessment of cardio-protective potential of KI against doxorubicin-induced cardiotoxicity, a total of 42 female Sprague-Dawley rats were put into 7 groups (n = 6). Group I: vehicle control, received normal saline (1 mL/kg p.o) for 30 days. Group II: toxic control, received DOX (20 mg/kg i.p.) once on the 29th day. Group III: KI control, received KI (300 mg/kg p.o) for 30 days. Group IV: vitamin E control, received vitamin E (100 mg/kg p.o) for 30 days. Group V: KI treated-1, received KI (300 mg/kg p.o) for 30 days and DOX (20 mg/kg i.p) on the 29th day. Group VI: KI treated-2, received KI (600 mg/kg p.o) for 30 days and DOX (20 mg/kg i.p) on the 29th day. Group VII: vitamin E treated, received vitamin E (100 mg/kg p.o) for 30 days and DOX (20 mg/kg i.p) on the 29th day. Thirty-six (36) hours after last administration, rats were sacrificed. Blood samples were taken via cardiac puncture to determine levels of aspartate aminotransferase (AST), alanine transaminase (ALT), alkaline phosphatase (ALP), creatine kinase (CK), lactate dehydrogenase (LDH), enzymatic antioxidants such as glutathione (GSH), superoxide dismutase (SOD), and catalase (CAT). Nitric oxide level was also determined. Hearts of rats in each group were excised and taken through histopathological examination. RESULTS In the HPLC fingerprint analysis, 13 peaks were identified, and peak with retention time of 24.0 min had the highest peak area (3.223 x104 mAU). Comet assay showed that the KI extract was non-genotoxic. Pretreatment with KI protected rats against doxorubicin-induced cardiotoxicity as evidenced by the low levels of AST, ALT, ALP, CK and LDH compared with the controls (p < 0.05). SOD, CAT and GPX levels were also high for rats administered KI extracts, further showing that KI protected rats against doxorubicin-induced cardiotoxicity. KI also inhibited nitric oxide levels at 300 mg/kg and 600 mg/kg effective doses. Histological examination revealed that rats pretreated with KI showed no signs of abnormal myocardial fibres (shape, size and configuration). CONCLUSION Ethanolic (70%) leaf extract of KI showed no genotoxic potential and possessed cardioprotective effects against doxorubicin-induced cardiotoxicity in Sprague-Dawley rats. KI also inhibited nitric oxide production, thus, a potential nitric oxide scavenger.
Collapse
Affiliation(s)
- Isaac Julius Asiedu-Gyekye
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, University of Ghana, PO Box LG 43 Legon, Accra, Ghana.
| | - Emmanuel Arhin
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, University of Ghana, PO Box LG 43 Legon, Accra, Ghana.
| | - Stella Amaaba Arthur
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, University of Ghana, PO Box LG 43 Legon, Accra, Ghana.
| | - Benoit Banga N'guessan
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, University of Ghana, PO Box LG 43 Legon, Accra, Ghana.
| | - Seth Kwabena Amponsah
- Department of Medical Pharmacology, University of Ghana Medical School, College of Health Sciences, Korle-Bu, Accra, Ghana.
| |
Collapse
|
19
|
Viswanatha GL, Shylaja H, Keni R, Nandakumar K, Rajesh S. A systematic review and meta‐analysis on the cardio‐protective activity of naringin based on pre‐clinical evidences. Phytother Res 2022; 36:1064-1092. [DOI: 10.1002/ptr.7368] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 10/15/2021] [Accepted: 12/10/2021] [Indexed: 12/12/2022]
Affiliation(s)
| | | | - Raghuvir Keni
- Department of Pharmacology Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education Manipal India
| | - Krishnadas Nandakumar
- Department of Pharmacology Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education Manipal India
| | - Subbanna Rajesh
- Department of Pharmacology Government College of Pharmacy Bangalore India
| |
Collapse
|
20
|
Waz S, Matouk AI. Cardioprotective effect of allyl isothiocyanate in a rat model of doxorubicin acute toxicity. Toxicol Mech Methods 2021; 32:194-203. [PMID: 34635025 DOI: 10.1080/15376516.2021.1992064] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Doxorubicin (DOX) is an effective anthracycline chemotherapeutic drug. Nevertheless, the cardiotoxicity adverse effect restricts its clinical benefit. Allyl isothiocyanate (AITC) is a natural antioxidant and anti-inflammatory agent. In the present study, we investigated the effect of AITC on cardiotoxicity of DOX. Thirty-two adult male albino rats were divided into four groups; control, AITC, DOX, and AITC + DOX. AITC was administrated orally (25 mg/kg/day) for 7 days, and DOX was given as a single i.p. injection (15 mg/kg) on the third day. Mortality rate was observed during the experiment. Cardiac toxicity markers (lactate dehydrogenase (LDH), creatine kinase (CK-MB), and cardiac Troponin I (cTn-I)) were evaluated in serum samples obtained from all groups after 48 hours of DOX injection. DOX-treated group showed 40% mortality and a significant increase in cardiac enzymes. This increase was accompanied by degenerated cardiomyocytes, and inflammatory cells infiltrates. Interestingly, AITC administration alleviated myocardial oxidative stress induced by DOX as attenuated the increase in malondialdehyde (MDA), and nitric oxide (NO) while resulted in elevations of the antioxidant reduced glutathione (GSH) level as well as superoxide dismutase (SOD) activity. Furthermore, the inflammatory cytokine, TNF-α, was reduced upon administration of AITC with DOX. The cardio-protection of AITC is attributed to increase the expression of cytoprotective nuclear factor erythroid 2-related factor 2 (Nrf2). Subsequently, heme oxygenase 1 (HO-1) level was elevated by AITC to correct the oxidative stress induced by DOX in the heart. Accordingly, AITC ameliorated acute cardiotoxicity associated with DOX treatment via attenuation of oxidative stress and the induced-tissue inflammatory injury. Abbreviations: DOX: doxrubicin; Nrf2: nuclear factor erythroid 2-related factor 2; HO-1: heme oxygenase 1; AITC: ally isothiocyanate; MDA: malondialdehyde; SOD: superoxide dismutase; GSH: reduced glutathione; TNF-α: tumor necrosis factor alpha.
Collapse
Affiliation(s)
- Shaimaa Waz
- Department of Biochemistry, Faculty of Pharmacy, Minia University, El-Minia, Egypt
| | - Asmaa I Matouk
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, El-Minia, Egypt
| |
Collapse
|
21
|
Kalyankumarraju M, Puppala ER, Ahmed S, Jagadeesh Kumar G, Tene K, N P S, Sahu BD, Barua CC, Naidu VGM. Zanthoxylum alatum Roxb. seed extract ameliorates stress aggravated DSS-induced ulcerative colitis in mice: Plausible role on NF-κB signaling axis. JOURNAL OF ETHNOPHARMACOLOGY 2021; 279:114385. [PMID: 34217795 DOI: 10.1016/j.jep.2021.114385] [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] [Received: 03/31/2021] [Revised: 06/25/2021] [Accepted: 06/30/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Zanthoxylum alatum (ZA) Roxb (family: Rutaceae) plant has been traditionally used for multiple indications by local healers among different communities of South Asian countries mainly in India and Bangladesh. The extracts of ZA have reported strong anti-inflammatory and anti-oxidant activities, but no scientific report is available on its efficacy in intestinal inflammatory disorders like ulcerative colitis. AIM OF THE STUDY The overall objective of our study was to evaluate the anti-inflammatory potency of hydro-ethanolic extract of Zanthoxylum alatum seed (ZAHA) using both in-vitro NF-κB-luciferase translocation assay and in-vivo stress aggravated dextran sodium sulfate (DSS)-induced ulcerative colitis model. MATERIALS AND METHODS The in-vitro anti-inflammatory effect of ZAHA extract was evaluated by luciferase assay in HEK293 cells. Parameters such as body weights, behavioural, colonoscopy, colon lengths and spleen weights were measured and recorded in stress aggravated DSS-induced colitis model in C57BL/6 mice. Biochemical, histological and immunoblot analysis in the colon tissues were determined to prove its anti-inflammatory and anti-oxidant activities. Characterization of the extract was done by LC-MS/MS study. RESULTS Initial in vitro NF-κB-luciferase translocation assay showed that the hydroalcoholic extract of ZA (ZAHA) showed potent inhibitory activity for NF-κB translocation by TNF-α stimulation and hence this particular extract was further evaluated in stress aggravated DSS-induced ulcerative colitis model in C57BL/6 mice. Treatment of ZAHA for two weeks at a dose of 200 mg/kg significantly ameliorated the stress aggravated DSS-induced colitis in mice. Histological alterations, infiltration of inflammatory cells, and the levels of IL-1β, IL-6, TNF-α in colon tissue and serum samples were significantly decreased in ZAHA treatment groups compared to the stress aggravated DSS induced colitis animals. Moreover, the protein expressions of p-NF-κB, p-IκBα, p-STAT3, COX-2, and TNF-α were significantly reduced in colon tissues of ZAHA treated groups and also increased anti-oxidant markers like SOD-1, Nrf2 significantly when compared with disease control group. Characterization of the extract further by LC-MS/MS revealed the presence of several active compounds which could be responsible for its anti-inflammatory activity. CONCLUSIONS Thus from the above findings it can be concluded that ZAHA ameliorates stress aggravated DSS-induced ulcerative colitis due to its anti-inflammatory and anti-oxidant activity.
Collapse
Affiliation(s)
- Malayamarutham Kalyankumarraju
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Assam, 781101, India
| | - Eswara Rao Puppala
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Assam, 781101, India
| | - Sahabuddin Ahmed
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Assam, 781101, India
| | - Gangasani Jagadeesh Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Assam, 781101, India
| | - Kalyani Tene
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Assam, 781101, India
| | - Syamprasad N P
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Assam, 781101, India
| | - Bidya Dhar Sahu
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Assam, 781101, India
| | - Chandana Choudhury Barua
- Department of Pharmacology and Toxicology, College of Veterinary Science, Assam Agricultural University, Guwahati, Assam, 781022, India
| | - V G M Naidu
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Assam, 781101, India.
| |
Collapse
|
22
|
Uryash A, Mijares A, Flores V, Adams JA, Lopez JR. Effects of Naringin on Cardiomyocytes From a Rodent Model of Type 2 Diabetes. Front Pharmacol 2021; 12:719268. [PMID: 34497520 PMCID: PMC8419284 DOI: 10.3389/fphar.2021.719268] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 08/10/2021] [Indexed: 12/15/2022] Open
Abstract
Diabetic cardiomyopathy (DCM) is a primary disease in diabetic patients characterized by diastolic dysfunction leading to heart failure and death. Unfortunately, even tight glycemic control has not been effective in its prevention. We have found aberrant diastolic Ca2+ concentrations ([Ca2+]d), decreased glucose transport, elevated production of reactive oxygen species (ROS), and increased calpain activity in cardiomyocytes from a murine model (db/db) of type 2 diabetes (T2D). Cardiomyocytes from these mice demonstrate significant cell injury, increased levels of tumor necrosis factor-alpha and interleukin-6 and expression of the transcription nuclear factor-κB (NF-κB). Furthermore, decreased cell viability, and reduced expression of Kir6.2, SUR1, and SUR2 subunits of the ATP-sensitive potassium (KATP) channels. Treatment of T2D mice with the citrus fruit flavonoid naringin for 4 weeks protected cardiomyocytes by reducing diastolic Ca2+ overload, improving glucose transport, lowering reactive oxygen species production, and suppressed myocardial inflammation. In addition, naringin reduced calpain activity, decreased cardiac injury, increased cell viability, and restored the protein expression of Kir6.2, SUR1, and SUR2 subunits of the KATP channels. Administration of the KATP channel inhibitor glibenclamide caused a further increase in [Ca2+]d in T2D cardiomyocytes and abolished the naringin effect on [Ca2+]d. Nicorandil, a KATP channel opener, and nitric oxide donor drug mimic the naringin effect on [Ca2+]d in T2D cardiomyocyte; however, it aggravated the hyperglycemia in T2D mice. These data add new insights into the mechanisms underlying the beneficial effects of naringin in T2D cardiomyopathy, thus suggesting a novel approach to treating this cardiovascular complication.
Collapse
Affiliation(s)
- A. Uryash
- Department of Neonatology, Mount Sinai Medical Center, Miami, FL, United States
| | - A. Mijares
- Centro de Biofísica y Bioquímica, Instituto Venezolano de Investigaciones Científicas, Caracas, Venezuela
| | - V. Flores
- Department of Research, Mount Sinai Medical Center, Miami, FL, United States
| | - J. A. Adams
- Department of Neonatology, Mount Sinai Medical Center, Miami, FL, United States
| | - J. R. Lopez
- Department of Research, Mount Sinai Medical Center, Miami, FL, United States
| |
Collapse
|
23
|
Sandamali JAN, Hewawasam RP, Jayatilaka KAPW, Mudduwa LKB. Cinnamomum zeylanicum Blume (Ceylon cinnamon) bark extract attenuates doxorubicin induced cardiotoxicity in Wistar rats. Saudi Pharm J 2021; 29:820-832. [PMID: 34408544 PMCID: PMC8363100 DOI: 10.1016/j.jsps.2021.06.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 06/13/2021] [Indexed: 01/02/2023] Open
Abstract
Anti-tumour efficacy of doxorubicin is hindered by the cumulative dose-dependent cardiotoxicity induced by reactive oxygen species during its metabolism. As Cinnamomum zeylanicum has proven antioxidant potential, objective of this study was to investigate the cardioprotective activity of Cinnamomum bark extract against doxorubicin induced cardiotoxicity in Wistar rats. Physicochemical and phytochemical analysis was carried out and dose response effect and the cardioprotective activity of Cinnamomum were determined in vivo. 180 mg/kg dexrazoxane was used as the positive control. Plant extracts were free of heavy metals and toxic phytoconstituents. In vivo study carried out in Wistar rats revealed a significant increase (p < 0.05) in cardiac troponin I, NT-pro brain natriuretic peptide, AST and LDH concentrations in the doxorubicin control group (18 mg/kg) compared to the normal control. Rats pre-treated with the optimum dosage of Cinnmamomum (2.0 g/kg) showed a significant reduction (p < 0.05) in all above parameters compared to the doxorubicin control. A significant reduction was observed in the total antioxidant capacity, reduced glutathione, glutathione peroxidase, glutathione reductase, superoxide dismutase and catalase activity while the lipid peroxidation and myeloperoxidase activity were significantly increased in the doxorubicin control group compared to the normal control (p < 0.05). Pre-treatment with Cinnamomum bark showed a significant decrease in lipid peroxidation, myeloperoxidase activity and significant increase in rest of the parameters compared to the doxorubicin control (p < 0.05). Histopathological analysis revealed a preserved appearance of the myocardium and lesser degree of cellular changes of necrosis in rats pre-treated with Cinnamomum extract. In conclusion, Cinnamomum bark extract has the potential to significantly reduce doxorubicin induced oxidative stress and inflammation in Wistar rats.
Collapse
Key Words
- ABEC, Aqueous bark extract of Cinnamomum zeylanicum
- ABTS, 2,2′-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid)
- AST, Aspartate aminotransferase
- Antioxidant effect
- Cardiotoxicity
- Cinnamomum zeylanicum bark extract
- DNA, Deoxyribonucleic acid
- DPPH, 2,2-diphenyl-1-picrylhydrazyl
- Doxorubicin
- ELISA, Enzyme-linked immunosorbent assay
- FRAP, Ferric reducing antioxidant power
- GPx, Glutathione peroxidase
- GR, Glutathione reductase
- GSH, Reduced glutathione
- H & E, Haematoxylin and eosin
- IP, Intraperitoneal
- LDH, Lactate dehydrogenase
- MDA, Malondialdehyde
- MPO, Myeloperoxidase
- Myeloperoxidase
- NADPH, Nicotinamide adenine dinucleotide phosphate hydrogen
- NO, Nitric oxide
- NT-pro BNP, N terminal- pro brain natriuretic peptide
- Oxidative-stress
- PBS, Phosphate buffered saline
- ROS, Reactive oxygen species
- SOD, Superoxide dismutase
- USA, United States of America
- WHO, World Health Organization
- cTnI, Cardiac troponin I
Collapse
|
24
|
Ikewuchi JC, Ikewuchi CC, Ifeanacho MO, Jaja VS, Okezue EC, Jamabo CN, Adeku KA. Attenuation of doxorubicin-induced cardiotoxicity in Wistar rats by aqueous leaf-extracts of Chromolaena odorata and Tridax procumbens. JOURNAL OF ETHNOPHARMACOLOGY 2021; 274:114004. [PMID: 33727109 DOI: 10.1016/j.jep.2021.114004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 01/02/2021] [Accepted: 03/04/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Chromolaena odorata (L) King and Robinson and Tridax procumbens Linn are used in traditional medicine in the treatment of diabetes mellitus and hypertension. AIM OF THE STUDY This study investigated the potential protective role of aqueous leaf-extracts of Chromolaena odorata and Tridax procumbens against cardiotoxicity induced by doxorubicin. MATERIALS AND METHODS To this end, their impact on plasma markers of cardiac integrity, cardiac markers of oxidative stress, cardiac lipids and electrolyte profiles, and activities of cardiac ATPases, lactate dehydrogenase and creatine kinase, were monitored in doxorubicin treated rats. Metformin (250 mg/kg body weight, orally) and both extracts (50, 75 and 100 mg/kg, orally) were daily administered for 14 days; while cardiotoxicity was induced with doxorubicin (15 mg/kg, intra-peritioneally, once on the 12th day of study). RESULTS The plasma activities of creatine kinase, lactate dehydrogenase and AST of Test control were significantly (p < 0.05) higher than those of the other groups. Also, the cardiac malondialdehyde, calcium, chloride, sodium, cholesterol and triglyceride concentrations of Test control were significantly (p < 0.05) higher than those of the others. However, the cardiac concentrations of ascorbic acid, reduced glutathione, magnesium and potassium, and cardiac activities of catalase, glutathione peroxidase, superoxide dismutase, Ca2+-ATPase, Mg2+-ATPase, Na+,K+-ATPase, creatine kinase and lactate dehydrogenase of Test control were significantly (p < 0.05) lower than those of the others. CONCLUSIONS Pre-treatment with the extracts and metformin elicited a cardioprotective effect, as indicated by the prevention of doxorubicin-induced cardiac oxidative stress and prevention of adverse alterations in plasma cardiac markers, cardiac lipids and electrolyte profiles, as well as improvement of the activities of cardiac ATPases, creatine kinase and lactate dehydrogenase.
Collapse
Affiliation(s)
- Jude C Ikewuchi
- Department of Biochemistry, Faculty of Science, University of Port Harcourt, P.M.B, 5323, Port Harcourt, Nigeria
| | - Catherine C Ikewuchi
- Department of Biochemistry, Faculty of Science, University of Port Harcourt, P.M.B, 5323, Port Harcourt, Nigeria
| | - Mercy O Ifeanacho
- Department of Food Science, Faculty of Agriculture, University of Port Harcourt, Nigeria.
| | - Victoria S Jaja
- Department of Biochemistry, Faculty of Science, University of Port Harcourt, P.M.B, 5323, Port Harcourt, Nigeria
| | - Esther C Okezue
- Department of Biochemistry, Faculty of Science, University of Port Harcourt, P.M.B, 5323, Port Harcourt, Nigeria
| | - Caius N Jamabo
- Department of Biochemistry, Faculty of Science, University of Port Harcourt, P.M.B, 5323, Port Harcourt, Nigeria
| | - Kehinde A Adeku
- Department of Biochemistry, Faculty of Science, University of Port Harcourt, P.M.B, 5323, Port Harcourt, Nigeria
| |
Collapse
|
25
|
Furihata T, Maekawa S, Takada S, Kakutani N, Nambu H, Shirakawa R, Yokota T, Kinugawa S. Premedication with pioglitazone prevents doxorubicin-induced left ventricular dysfunction in mice. BMC Pharmacol Toxicol 2021; 22:27. [PMID: 33962676 PMCID: PMC8103594 DOI: 10.1186/s40360-021-00495-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 04/26/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Doxorubicin (DOX) is widely used as an effective chemotherapeutic agent for cancers; however, DOX induces cardiac toxicity, called DOX-induced cardiomyopathy. Although DOX-induced cardiomyopathy is known to be associated with a high cumulative dose of DOX, the mechanisms of its long-term effects have not been completely elucidated. Pioglitazone (Pio) is presently contraindicated in patients with symptomatic heart failure owing to the side effects. The concept of drug repositioning led us to hypothesize the potential effects of Pio as a premedication before DOX treatment, and to analyze this hypothesis in mice. METHODS First, for the hyperacute (day 1) and acute (day 7) DOX-induced dysfunction models, mice were fed a standard diet with or without 0.02% (wt/wt) Pio for 5 days before DOX treatment (15 mg/kg body weight [BW] via intraperitoneal [i.p.] administration). The following 3 treatment groups were analyzed: standard diet + vehicle (Vehicle), standard diet + DOX (DOX), and Pio + DOX. Next, for the chronic model (day 35), the mice were administrated DOX once a week for 5 weeks (5 mg/kg BW/week, i.p.). RESULTS In the acute phase after DOX treatment, the percent fractional shortening of the left ventricle (LV) was significantly decreased in DOX mice. This cardiac malfunction was improved in Pio + DOX mice. In the chronic phase, we observed that LV function was preserved in Pio + DOX mice. CONCLUSIONS Our findings may provide a new pathophysiological explanation by which Pio plays a role in the treatment of DOX-induced cardiomyopathy, but the molecular links between Pio and DOX-induced LV dysfunction remain largely elusive.
Collapse
Affiliation(s)
- Takaaki Furihata
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo, 060-8638, Japan.
| | - Satoshi Maekawa
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Shingo Takada
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo, 060-8638, Japan
- Faculty of Lifelong Sport, Department of Sports Education, Hokusho University, Ebetsu, 069-8511, Japan
| | - Naoya Kakutani
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo, 060-8638, Japan
- Research Fellow of the Japan Society for the Promotion of Science, Tokyo, Japan
| | - Hideo Nambu
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Ryosuke Shirakawa
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Takashi Yokota
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Shintaro Kinugawa
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo, 060-8638, Japan
| |
Collapse
|
26
|
Ahmed OM, AbouZid SF, Ahmed NA, Zaky MY, Liu H. An Up-to-Date Review on Citrus Flavonoids: Chemistry and Benefits in Health and Diseases. Curr Pharm Des 2021; 27:513-530. [PMID: 33245267 DOI: 10.2174/1381612826666201127122313] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 08/09/2020] [Indexed: 11/22/2022]
Abstract
Flavonoids, the main class of polyphenols, are characterized by the presence of 2-phenyl-benzo-pyrane nucleus. They are found in rich quantities in citrus fruits. Citrus flavonoids are classified into flavanones, flavones, flavonols, polymethoxyflavones and anthocyanins (found only in blood oranges). Flavanones are the most abundant flavonoids in citrus fruits. In many situations, there are structure-function relationships. Due to their especial structures and presence of many hydroxyls, polymethoxies and glycoside moiety, the flavonoids have an array of multiple biological and pharmacological activities. This article provides an updated overview of the differences in chemical structures of the classes and members of citrus flavonoids and their benefits in health and diseases. The review article also sheds light on the mechanisms of actions of citrus flavonoids in the treatment of different diseases, including arthritis, diabetes mellitus, cancer and neurodegenerative disorders as well as liver, kidney and heart diseases. The accumulated and updated knowledge in this review may provide useful information and ideas in the discovery of new strategies for the use of citrus flavonoids in the protection, prevention and therapy of diseases.
Collapse
Affiliation(s)
- Osama M Ahmed
- Physiology Division, Department of Zoology, Faculty of Science, Beni-Suef University, Egypt
| | - Sameh F AbouZid
- Pharmacognosy Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Noha A Ahmed
- Physiology Division, Department of Zoology, Faculty of Science, Beni-Suef University, Egypt
| | - Mohamed Y Zaky
- Physiology Division, Department of Zoology, Faculty of Science, Beni-Suef University, Egypt
| | - Han Liu
- Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| |
Collapse
|
27
|
Ifeanacho MO, Ikewuchi JC, Ikewuchi CC, Nweke PC, Okere R, Nwate TL. Prevention of doxorubicin-induced dyslipidaemia, plasma oxidative stress and electrolytes imbalance in Wistar rats by aqueous leaf-extracts of Chromolaena odorata and Tridax procumbens. SCIENTIFIC AFRICAN 2021. [DOI: 10.1016/j.sciaf.2020.e00636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
28
|
Navarro-Hortal MD, Varela-López A, Romero-Márquez JM, Rivas-García L, Speranza L, Battino M, Quiles JL. Role of flavonoids against adriamycin toxicity. Food Chem Toxicol 2020; 146:111820. [PMID: 33080329 DOI: 10.1016/j.fct.2020.111820] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 10/13/2020] [Accepted: 10/15/2020] [Indexed: 12/13/2022]
Abstract
Doxorubicin (DOX), or adriamycin, is an anthracycline antineoplastic drug widely used in the chemotherapy of a large variety of cancers due to its potency and action spectrum. However, its use is limited by the toxicity on healthy cells and its acute and chronic side effects. One of the developed strategies to attenuate DOX toxicity is the combined therapy with bioactive compounds such as flavonoids. This review embraces the role of flavonoids on DOX treatment side effects. Protective properties of some flavonoidss against DOX toxicity have been investigated and observed mainly in heart but also in liver, kidney, brain, testis or bone marrow. Protective mechanisms involve reduction of oxidative stress by decrease of ROS levels and/or increase antioxidant defenses and interferences with autophagy, apoptosis and inflammation. Studies in cancer cells have reported that the anticancer activity of DOX was not compromised by the flavonoids. Moreover, some of them increased DOX efficiency as anti-cancer drug even in multidrug resistant cells.
Collapse
Affiliation(s)
- María D Navarro-Hortal
- Biomedical Research Centre, Institute of Nutrition and Food Technology "José Mataix Verdú", Department of Physiology, University of Granada, Avda. del Conocimiento s/n, 18100, Armilla, Granada, Spain.
| | - Alfonso Varela-López
- Biomedical Research Centre, Institute of Nutrition and Food Technology "José Mataix Verdú", Department of Physiology, University of Granada, Avda. del Conocimiento s/n, 18100, Armilla, Granada, Spain.
| | - José M Romero-Márquez
- Biomedical Research Centre, Institute of Nutrition and Food Technology "José Mataix Verdú", Department of Physiology, University of Granada, Avda. del Conocimiento s/n, 18100, Armilla, Granada, Spain.
| | - Lorenzo Rivas-García
- Biomedical Research Centre, Institute of Nutrition and Food Technology "José Mataix Verdú", Department of Physiology, University of Granada, Avda. del Conocimiento s/n, 18100, Armilla, Granada, Spain; Sport and Health Research Centre, University of Granada, C/. Menéndez Pelayo 32, 18016, Armilla, Granada, Spain.
| | - Lorenza Speranza
- Department of Medicine and Aging Sciences, University "G. d'Annunzio" Chieti-Pescara, Via dei Vestini 31, 66100, CH, Italy.
| | - Maurizio Battino
- Department of Clinical Sicences, Università Politecnica delle Marche, 60131, Ancona, Italy; International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang, 212013, China.
| | - José L Quiles
- Biomedical Research Centre, Institute of Nutrition and Food Technology "José Mataix Verdú", Department of Physiology, University of Granada, Avda. del Conocimiento s/n, 18100, Armilla, Granada, Spain.
| |
Collapse
|
29
|
Liu C, Ma X, Zhuang J, Liu L, Sun C. Cardiotoxicity of doxorubicin-based cancer treatment: What is the protective cognition that phytochemicals provide us? Pharmacol Res 2020; 160:105062. [DOI: 10.1016/j.phrs.2020.105062] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/29/2020] [Accepted: 06/30/2020] [Indexed: 12/11/2022]
|
30
|
Clerodendrum volubile Ethanol Leaf Extract: A Potential Antidote to Doxorubicin-Induced Cardiotoxicity in Rats. J Toxicol 2020; 2020:8859716. [PMID: 32714390 PMCID: PMC7355376 DOI: 10.1155/2020/8859716] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 06/11/2020] [Indexed: 01/09/2023] Open
Abstract
Doxorubicin is widely applied in hematological and solid tumor treatment but limited by its off-target cardiotoxicity. Thus, cardioprotective potential and mechanism(s) of CVE in DOX-induced cardiotoxicity were investigated using cardiac and oxidative stress markers and histopathological endpoints. 50–400 mg/kg/day CVE in 5% DMSO in distilled water were investigated in Wistar rats intraperitoneally injected with 2.5 mg/kg DOX on alternate days for 14 days, using serum troponin I and LDH, complete lipid profile, cardiac tissue oxidative stress marker assays, and histopathological examination of DOX-treated cardiac tissue. Preliminary qualitative and quantitative assays of CVE's secondary metabolites were also conducted. Phytochemical analyses revealed the presence of flavonoids (34.79 ± 0.37 mg/100 mg dry extract), alkaloids (36.73 ± 0.27 mg/100 mg dry extract), reducing sugars (07.78 ± 0.09 mg/100 mg dry extract), and cardiac glycosides (24.55 ± 0.12 mg/100 mg dry extract). 50–400 mg/kg/day CVE significantly attenuated increases in the serum LDH and troponin I levels. Similarly, the CVE dose unrelatedly decreased serum TG and VLDL-c levels without significant alterations in the serum TC, HDL-c, and LDL-c levels. Also, CVE profoundly attenuated alterations in the cardiac tissue oxidative stress markers' activities while improving DOX-associated cardiac histological lesions that were possibly mediated via free radical scavenging and/or antioxidant mechanisms. Overall, CVE may play a significant therapeutic role in the management of DOX-induced cardiotoxicity in humans.
Collapse
|
31
|
Wali AF, Rashid S, Rashid SM, Ansari MA, Khan MR, Haq N, Alhareth DY, Ahmad A, Rehman MU. Naringenin Regulates Doxorubicin-Induced Liver Dysfunction: Impact on Oxidative Stress and Inflammation. PLANTS 2020; 9:plants9040550. [PMID: 32344607 PMCID: PMC7238146 DOI: 10.3390/plants9040550] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 03/29/2020] [Accepted: 04/08/2020] [Indexed: 12/18/2022]
Abstract
Doxorubicin (Dox) is an operational and largely used anticancer drug, used to treat an array of malignancies. Nonetheless, its beneficial use is constrained due to its renal and hepatotoxicity dose dependently. Numerous research findings favor the use of antioxidants may impact Dox-induced liver injury/damage. In the current study, Wistar rats were given naringenin (50 and 100 mg/kg b.wt.) orally for 20 days as prophylactic dose, against the hepatotoxicity induced by single intraperitoneal injection of Dox (20 mg/kg b.wt.). Potency of naringenin against the liver damage caused by Dox was assessed by measuring malonyl aldehyde (MDA) as a by-product of lipid peroxidation, biochemical estimation of antioxidant enzyme system, reactive oxygen species (ROS) level, and inflammatory mediators. Naringenin-attenuated ROS production, ROS-induced lipid peroxidation, and replenished reduced antioxidant armory, namely, catalase (CAT), glutathione reductase (GR), superoxide dismutase (SOD), glutathione peroxidase (GPx), and glutathione (GSH). Naringenin similarly diminished expression of Cox-2 and levels of NF-κB and other inflammatory molecules induced by the Dox treatment. Histology added further evidence to the defensive effects of naringenin on Dox-induced liver damage. The outcomes of the current study reveal that oxidative stress and inflammation are meticulously linked with Dox-triggered damage, and naringenin illustrates the potential effect on Dox-induced hepatotoxicity probably through diminishing the oxidative stress and inflammation.
Collapse
Affiliation(s)
- Adil Farooq Wali
- RAK College of Pharmaceutical Sciences, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, UAE;
| | - Summya Rashid
- Department of Pharmacology & Toxicology, College of Pharmacy Girls Section, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia;
| | - Shahzada Mudasir Rashid
- Division of Veterinary Biochemistry, Faculty of Veterinary Science and Animal Husbandry, SKUAST-Kashmir, Shuhama, J&K 190006, India;
| | - Mushtaq Ahmad Ansari
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; (M.A.A.); (M.R.K.); (D.Y.A.)
| | - Mohammad Rashid Khan
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; (M.A.A.); (M.R.K.); (D.Y.A.)
| | - Nazrul Haq
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia;
| | - Dhafer Yahya Alhareth
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; (M.A.A.); (M.R.K.); (D.Y.A.)
| | - Ajaz Ahmad
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
- Correspondence: (A.A.); (M.U.R.); Tel.: +96-6114670765 (A.A. & M.U.R.)
| | - Muneeb U. Rehman
- Division of Veterinary Biochemistry, Faculty of Veterinary Science and Animal Husbandry, SKUAST-Kashmir, Shuhama, J&K 190006, India;
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
- Correspondence: (A.A.); (M.U.R.); Tel.: +96-6114670765 (A.A. & M.U.R.)
| |
Collapse
|
32
|
Salehcheh M, Alboghobeish S, Dehghani MA, Zeidooni L. Multi-walled carbon nanotubes induce oxidative stress, apoptosis, and dysfunction in isolated rat heart mitochondria: protective effect of naringin. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:13447-13456. [PMID: 32026367 DOI: 10.1007/s11356-020-07943-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 01/29/2020] [Indexed: 06/10/2023]
Abstract
Multi-walled carbon nanotubes (MWCNTs) are material with exclusive features that can be applied in different fields including industrial and medicine. It has been determined that the accumulation of MWCNTs in the organs is along with genotoxic and cytotoxic injuries. Previous studies have shown mitochondrial dysfunction in MWCNTs exposure with cell lines, but their exact mechanisms with isolated mitochondria have remained unclear. The present study evaluated toxicity induced by MWCNTs in isolated rat heart mitochondria and protective effect of naringin. Our results showed that MWCNTs toxicity caused the prevention of heart mitochondrial complex II activity. Treatment of isolated heart mitochondria with MWCNTs led to an increase in mitochondrial reactive oxygen species (ROS) generation, mitochondrial membrane potential (MMP) collapse, and mitochondrial malondialdehyde (MDA) and a decrease in mitochondrial glutathione (GSH) level and mitochondrial catalase (CAT) activity. Pretreatment of isolated heart mitochondria with naringin decreased mitochondrial oxidative damage through decreasing lipid peroxidation, returned mitochondrial complex II changes, decreasing MMP collapse and ROS production, and restoration of GSH level and CAT activity. Our findings indicated that MWCNTs had toxic effects on isolated heart mitochondria by inducing oxidative stress and possibly apoptosis pathway. The protection effects of naringin may be accompanied by mitochondrial conservation by its antioxidant property or due to its free radical scavenging. Our findings indicated that naringin had a possible role in preventing the mitochondria complaints in the heart.
Collapse
Affiliation(s)
- Maryam Salehcheh
- Toxicology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Soheila Alboghobeish
- Student Research Committee, Department of Pharmacology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Amin Dehghani
- Student Research Committee, Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Leila Zeidooni
- Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
33
|
George MY, Menze ET, Esmat A, Tadros MG, El-Demerdash E. Potential therapeutic antipsychotic effects of Naringin against ketamine-induced deficits in rats: Involvement of Akt/GSK-3β and Wnt/β-catenin signaling pathways. Life Sci 2020; 249:117535. [PMID: 32151688 DOI: 10.1016/j.lfs.2020.117535] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/13/2020] [Accepted: 03/05/2020] [Indexed: 12/11/2022]
Abstract
AIM Schizophrenia is a chronic, disabling and one of the major neurological illnesses affecting nearly 1% of the global population. Currently available antipsychotic medications possess limited effects. The current research aimed at investigating potential therapeutic add-on benefit to enhance the effects of clozapine anti-schizophrenic. MAIN METHODS To induce schizophrenia, ketamine was administered at a dose of 25 mg/kg i.p. for 14 consecutive days. Naringin was administered to Wistar rats at a dose of 100 mg/kg orally, alone or in combination with clozapine 5 mg/kg i.p from day 8 to day 14. Furthermore, behavioral tests were conducted to evaluate positive, negative and cognitive symptoms of schizophrenia. In addition, neurotransmitters' levels were detected using HPLC. Moreover, oxidative stress markers were assessed using spectrophotometry. Furthermore, apoptotic and wnt/β-catenin pathway markers were determined using western blotting (Akt, GSK-3β and β-catenin), colorimetric methods (Caspase-3) and immunohistochemistry (Bax, Bcl2 and cytochrome c). KEY FINDINGS Ketamine induced positive, negative and cognitive schizophrenia symptoms together with neurotransmitters' imbalance. In addition, ketamine treatment caused significant glutathione depletion, lipid peroxidation and reduction in catalase activity. Naringin and/or clozapine treatment significantly attenuated ketamine-induced schizophrenic symptoms and oxidative injury. Additionally, ketamine provoked apoptosis via increasing Bax/Bcl2 expression, caspase-3 activity, and Cytochrome C and Akt protein expression while naringin/clozapine treatment significantly inhibited this apoptotic effect. Moreover, naringin activated the neurodevelopmental wnt/β-catenin signaling pathway evidenced by increasing pGSK-3β and reducing pβ-catenin protein expression. SIGNIFICANCE These findings may suggest that naringin possesses a potential therapeutic add-on effect against ketamine-induced schizophrenia.
Collapse
Affiliation(s)
- Mina Y George
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Esther T Menze
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Ahmed Esmat
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mariane G Tadros
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - E El-Demerdash
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
34
|
El-Agamy DS, Ibrahim SRM, Ahmed N, Khoshhal S, Abo-Haded HM, Elkablawy MA, Aljuhani N, Mohamed GA. Aspernolide F, as a new cardioprotective butyrolactone against doxorubicin-induced cardiotoxicity. Int Immunopharmacol 2020; 72:429-436. [PMID: 31030099 DOI: 10.1016/j.intimp.2019.04.045] [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: 01/29/2019] [Revised: 04/03/2019] [Accepted: 04/21/2019] [Indexed: 12/15/2022]
Abstract
Endophytic fungi have known as a promising source of secondary metabolites. γ-Butyrolactones are a class of metabolites reported from Aspergillus genus, which attracted much attention for their bioactivities. This study aimed to assess the potential cardioprotective effects of aspernolide F (AF) separated from the endophytic fungus A. terreus against doxorubicin (DOX)-induced cardiotoxic effects in rats. Animals were treated with two different doses of AF for 10 days prior to DOX injection. Electrocardiographic (ECG), biochemical, histopathological and immunohistochemical analyses were performed. Results have shown that AF effectively protected against DOX-induced cardiac damage as AF counteracted DOX-induced ECG abnormalities and attenuated serum markers of cardiotoxicity (creatine kinase-MB, lactate dehydrogenase, troponin I, and troponin T). Histopathological examination of cardiac tissue revealed a remarkable improvement in DOX-induced lesions. In addition, AF ameliorated DOX-induced oxidative damage and increased the levels of antioxidants in cardiac tissues. AF treatment inhibited the activation of nuclear factor-κB (NF-κB) and decreased the immuno-expression of NF-κB in cardiac tissue. Furthermore, AF caused a marked lowering in the level of inflammatory cytokines (nitric oxide, tumor necrosis factor-α, and interleukin-6) in the cardiac tissue. Collectively, this study demonstrates the cardioprotective activity of AF against DOX-induced cardiac damage which may be due to its antioxidant and anti-inflammatory activities.
Collapse
Affiliation(s)
- Dina S El-Agamy
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Al Madinah Al-Munawwarah 30001, Saudi Arabia; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Sabrin R M Ibrahim
- Department of Pharmacognosy and Pharmaceutical Chemistry, College of Pharmacy, Taibah University, Al Madinah Al-Munawwarah 30001, Saudi Arabia; Department of Pharmacognosy, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt.
| | - Nishat Ahmed
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Al Madinah Al-Munawwarah 30001, Saudi Arabia
| | - Saad Khoshhal
- Cardiogenetic Team, Pediatric Dept., College of Medicine, Taibah University, Al Madinah Al-Munawwarah 30078, Saudi Arabia
| | - Hany M Abo-Haded
- Cardiogenetic Team, Pediatric Dept., College of Medicine, Taibah University, Al Madinah Al-Munawwarah 30078, Saudi Arabia; Cardiology Unit, Department of Pediatrics, Mansoura University, 35516, Egypt
| | - Mohamed A Elkablawy
- Department of Pathology, College of Medicine, Taibah University, Al Madinah Al-Munawwarah 30078, Saudi Arabia; Department of Pathology, Faculty of Medicine, Menoufia University, Menoufia 32511, Egypt
| | - Naif Aljuhani
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Al Madinah Al-Munawwarah 30001, Saudi Arabia
| | - Gamal A Mohamed
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Pharmacognosy Department, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut 71524, Egypt
| |
Collapse
|
35
|
Al-Malky HS, Osman AMM, Damanhouri ZA, Alkreathy HM, Al Aama JY, Ramadan WS, Al Qahtani AA, Al Mahdi HB. Modulation of doxorubicin-induced expression of the multidrug resistance gene in breast cancer cells by diltiazem and protection against cardiotoxicity in experimental animals. Cancer Cell Int 2019; 19:191. [PMID: 31367189 PMCID: PMC6657176 DOI: 10.1186/s12935-019-0912-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 07/16/2019] [Indexed: 12/11/2022] Open
Abstract
Background Doxorubicin (DOX) is one of the most important anticancer agents used in treating breast cancer. However, chronic cardiotoxicity and multidrug resistance limit the chemotherapeutic use of DOX. Methods This study aimed to evaluate the capability of calcium channel blocker diltiazem (DIL) to reverse DOX resistance in breast cancer MCF-7 cells and to confer protection against DOX-induced cardiotoxicity in Wistar rats. For this purpose, we explored the effects of DOX on cell cycle phase distribution and expression of ABCB1, FOXO3a, and p53 genes in the presence and absence of DIL (20 μg/ml) and studied the ability of DIL to prevent DOX-induced cardiotoxicity after a single injection of DOX (15 mg/kg) in male Wister rats. Results We found that compared with DOX alone treatment, DIL + DOX treatment down regulated the ABCB1 gene expression by > fourfold but up regulated the FOXO3a and p53 genes expression by 1.5 fold. DIL treatment conferred protection against DOX-induced cardiotoxicity, as indicated by a decrease in the levels of the cardiac enzyme creatine kinase MB and malondialdehyde and an increase in the total antioxidant capacity and glutathione peroxidase levels. These biochemical results were further confirmed by the histopathological investigation of cardiac cells, which showed normal cardiac cells with central vesicular nuclei and prevention of DOX-induced disruption of normal cardiac architecture in the DIL to DOX group. Conclusions Taken together, our results indicate that DIL treatment can reverse the resistance of breast cancer cells to the therapeutic effects of DOX and can protect against DOX-induced cardiotoxicity in rats.
Collapse
Affiliation(s)
- Hamdan S Al-Malky
- Pharmacology Department, Faculty of Medicine, KAU, Jeddah, Saudi Arabia
| | - Abdel-Moneim M Osman
- Pharmacology Department, Faculty of Medicine, KAU, Jeddah, Saudi Arabia.,2Pharmacology Unit, National Cancer Institute, Cairo University, Cairo, Egypt
| | | | - Huda M Alkreathy
- Pharmacology Department, Faculty of Medicine, KAU, Jeddah, Saudi Arabia
| | - Jumana Y Al Aama
- Department of Genetic Medicine, Faculty of Medicine, KAU, Jeddah, Saudi Arabia.,Princess Aljawhara Center of Excellence in Research of Hereditary Disorders, KAU, Jeddah, Saudi Arabia
| | - Wafaa S Ramadan
- Anatomy Department, Faculty of Medicine, KAU, Jeddah, Saudi Arabia.,6Anatomy Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Ali A Al Qahtani
- Pharmacology Department, Faculty of Medicine, KAU, Jeddah, Saudi Arabia
| | - Hadiah B Al Mahdi
- Department of Genetic Medicine, Faculty of Medicine, KAU, Jeddah, Saudi Arabia.,Princess Aljawhara Center of Excellence in Research of Hereditary Disorders, KAU, Jeddah, Saudi Arabia
| |
Collapse
|
36
|
McCluskey SP, Haslop A, Coello C, Gunn RN, Tate EW, Southworth R, Plisson C, Long NJ, Wells LA. Imaging of Chemotherapy-Induced Acute Cardiotoxicity with 18F-Labeled Lipophilic Cations. J Nucl Med 2019; 60:1750-1756. [PMID: 31147403 DOI: 10.2967/jnumed.119.226787] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 05/29/2019] [Indexed: 12/25/2022] Open
Abstract
Many chemotherapy agents are toxic to the heart, such that increasing numbers of cancer survivors are now living with the potentially lethal cardiovascular consequences of their treatment. Earlier and more sensitive detection of chemotherapy-induced cardiotoxicity may allow improved treatment strategies and increase long-term survival. Lipophilic cation PET tracers may be suitable for early detection of cardiotoxicity. This study aimed to evaluate an 18F-labeled lipophilic phosphonium cation, [1-(2-18F-fluoroethyl),1H[1,2,3]triazole-4-ethylene]triphenylphosphonium bromide (18F-MitoPhos), as a cardiac imaging agent, comparing it with leading PET and SPECT lipophilic cationic tracers before further assessing its potential for imaging cardiotoxicity in an acute doxorubicin model. Methods: Cardiac uptake and response to decreased mitochondrial membrane potential of 18F-MitoPhos and 99mTc-sestamibi were tested in isolated perfused rat hearts. Baseline pharmacokinetic profiles of 18F-MitoPhos and 18F-fluorobenzyltriphenylphosphonium and their response to acute doxorubicin-induced cardiotoxicity were assessed in rats in vivo (10, 15, or 20 mg of doxorubicin per kilogram, intravenously, 48 h beforehand). Results: Cardiac retention of 18F-MitoPhos was more than double that of 99mTc-sestamibi in isolated perfused rat hearts. A favorable biodistribution of 18F-MitoPhos in vivo was observed, with heart-to-tissue ratios of 304 ± 186, 11.2 ± 1.2, and 3.8 ± 0.6 for plasma, liver, and lung, respectively (60 min). A significant dose-dependent loss of cardiac retention of 18F-MitoPhos was observed on doxorubicin treatment, with average cardiac SUV from 30 to 60 min (mean ± SD) decreasing from 3.5 ± 0.5 (control) to 1.8 ± 0.1 (doxorubicin, 20 mg/kg). Other assessed biomarkers showed no alterations. Conclusion: 18F-MitoPhos showed pharmacokinetic parameters suitable for cardiac imaging. A significant dose response of cardiac uptake to doxorubicin treatment was observed before detectable biomarker alterations. 18F-MitoPhos is therefore a promising tracer for imaging chemotherapy-induced cardiotoxicity. To our knowledge, this is the first demonstration of radiolabeled lipophilic cations being used for the PET imaging of chemotherapy-induced cardiotoxicity and indicates the potential application of these compounds in this area.
Collapse
Affiliation(s)
- Stuart P McCluskey
- Department of Chemistry, Imperial College London, London, United Kingdom.,Invicro LLC, London, United Kingdom
| | - Anna Haslop
- Department of Chemistry, Imperial College London, London, United Kingdom
| | | | - Roger N Gunn
- Invicro LLC, London, United Kingdom.,Division of Brain Sciences, Imperial College London, Imperial College Centre for Drug Discovery Science, London, United Kingdom; and
| | - Edward W Tate
- Department of Chemistry, Imperial College London, London, United Kingdom
| | - Richard Southworth
- Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | | | - Nicholas J Long
- Department of Chemistry, Imperial College London, London, United Kingdom
| | | |
Collapse
|
37
|
Zakaria N, Khalil SR, Awad A, Khairy GM. Quercetin Reverses Altered Energy Metabolism in the Heart of Rats Receiving Adriamycin Chemotherapy. Cardiovasc Toxicol 2019; 18:109-119. [PMID: 28702745 DOI: 10.1007/s12012-017-9420-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The primary aim of this study was to find the potential modulatory roles of quercetin (QUE) against Adriamycin (ADR)-induced cardiotoxicity. A total of 50 rats were assigned to five groups: a control group, an ADR-treated group, a QUE-treated group, a prophylaxis-cotreated group, and a therapeutic-cotreated group, respectively. QUE exhibited a significant cardioprotective effect, particularly, when it was administered prior to and concurrently with ADR treatment (prophylaxis-cotreated group). This effect was biochemically evident by the significant decreases in the serum levels of myocardial injury biomarkers such as troponin, creatine kinase-myocardium bound, and creatine phosphokinase. In addition, significant elevations in myocardial antioxidant indices coupled with significant reductions in myocardial malondialdehyde contents and DNA damage, elicited by ADR injection, were observed. All these biochemical improvements were accompanied by a significant histopathological recovery and obvious modulation of the AMP-activated protein kinase (AMPK) signaling pathway by promoting the expression of the AMPKα2, PPARα, and PCG-1α genes. Taken together, these findings conclusively showed that QUE administration through its antioxidant capacity and myocardial energy metabolism restoration provides a prophylactic effect in response to ADR-induced deleterious effects, in the rat heart.
Collapse
Affiliation(s)
- Naglaa Zakaria
- Pharmacology Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Samah R Khalil
- Forensic Medicine and Toxicology Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44511, Egypt.
| | - Ashraf Awad
- Animal Wealth Development Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Ghada M Khairy
- Pharmacology Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
38
|
Lin SR, Weng CF. PG-Priming Enhances Doxorubicin Influx to Trigger Necrotic and Autophagic Cell Death in Oral Squamous Cell Carcinoma. J Clin Med 2018; 7:jcm7100375. [PMID: 30347872 PMCID: PMC6210351 DOI: 10.3390/jcm7100375] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 10/15/2018] [Accepted: 10/18/2018] [Indexed: 12/13/2022] Open
Abstract
Synergistic effects between natural compounds and chemotherapy drugs are believed to have fewer side effects with equivalent efficacy. However, the synergistic potential of prodigiosin (PG) with doxorubicin (Dox) chemotherapy is still unknown. This study explores the synergistic mechanism of PG and Dox against oral squamous cell carcinoma (OSCC) cells. Three OSCC cell lines were treated with different PG/Dox combinatory schemes for cytotoxicity tests and were further investigated for cell death characteristics by cell cycle flow cytometry and autophagy/apoptosis marker labelling. When OSCC cells were pretreated with PG, the cytotoxicity of the subsequent Dox-treatment was 30% higher than Dox alone. The cytotoxic efficacy of PG-pretreated was found better than those of PG plus Dox co-treatment and Dox-pretreatment. Increase of Sub-G1 phase and caspase-3/LC-3 levels without poly (ADP-ribose) polymeras (PARP) elevation indicated both autophagy and necrosis occurred in OSCC cells. Dox flux after PG-priming was further evaluated by rhodamine-123 accumulation and Dox transporters analysis to elucidate the PG-priming effect. PG-priming autophagy enhanced Dox accumulation according to the increase of rhodamine-123 accumulation without the alterations of Dox transporters. Additionally, the cause of PG-triggered autophagy was determined by co-treatment with endoplasmic reticulum (ER) stress or AMP-activated protein kinase (AMPK) inhibitor. PG-induced autophagy was not related to nutrient deprivation and ER stress was proved by co-treatment with specific inhibitor. Taken together, PG-priming autophagy could sensitize OSCC cells by promoting Dox influx without regulation of Dox transporter. The PG-priming might be a promising adjuvant approach for the chemotherapy of OSCC.
Collapse
Affiliation(s)
- Shian-Ren Lin
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Hualien 97401, Taiwan.
| | - Ching-Feng Weng
- Department of Life Science and Institute of Biotechnology, National Dong Hwa University, Hualien 97401, Taiwan.
| |
Collapse
|
39
|
Khodayar MJ, Kalantari H, Mahdavinia M, Khorsandi L, Alboghobeish S, Samimi A, Alizadeh S, Zeidooni L. Protective effect of naringin against BPA-induced cardiotoxicity through prevention of oxidative stress in male Wistar rats. Drug Chem Toxicol 2018; 43:85-95. [PMID: 30264589 DOI: 10.1080/01480545.2018.1504958] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Mohammad Javad Khodayar
- Toxicology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Heibatollah Kalantari
- Medicinal Plants Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Masoud Mahdavinia
- Toxicology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Layasadat Khorsandi
- Cellular and Molecular Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Soheila Alboghobeish
- Department of Pharmacology, School of Pharmacy, Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Azin Samimi
- Department of Toxicology, School of Pharmacy, Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Saeid Alizadeh
- Department of Toxicology, School of Pharmacy, Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Leila Zeidooni
- Department of Toxicology, School of Pharmacy, Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
40
|
Aygun H, Gul SS. Effects of melatonin and agomelatine on doxorubicin induced anxiety and depression-like behaviors in rats. ACTA ACUST UNITED AC 2018. [DOI: 10.17546/msd.433289] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
41
|
Shi W, Deng H, Zhang J, Zhang Y, Zhang X, Cui G. Mitochondria-Targeting Small Molecules Effectively Prevent Cardiotoxicity Induced by Doxorubicin. Molecules 2018; 23:E1486. [PMID: 29921817 PMCID: PMC6099719 DOI: 10.3390/molecules23061486] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 06/13/2018] [Accepted: 06/13/2018] [Indexed: 02/07/2023] Open
Abstract
Doxorubicin (Dox) is a chemotherapeutic agent widely used for the treatment of numerous cancers. However, the clinical use of Dox is limited by its unwanted cardiotoxicity. Mitochondrial dysfunction has been associated with Dox-induced cardiotoxicity. To mitigate Dox-related cardiotoxicity, considerable successful examples of a variety of small molecules that target mitochondria to modulate Dox-induced cardiotoxicity have appeared in recent years. Here, we review the related literatures and discuss the evidence showing that mitochondria-targeting small molecules are promising cardioprotective agents against Dox-induced cardiac events.
Collapse
Affiliation(s)
- Wei Shi
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai 519041, China.
| | - Hongkuan Deng
- School of Life Sciences, Shandong University of Technology, Zibo 255000, China.
| | - Jianyong Zhang
- Pharmacy School, Zunyi Medical University, Zunyi 563003, China.
| | - Ying Zhang
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai 519041, China.
| | - Xiufang Zhang
- School of Life Sciences, Shandong University of Technology, Zibo 255000, China.
| | - Guozhen Cui
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai 519041, China.
| |
Collapse
|
42
|
Bose C, Awasthi S, Sharma R, Beneš H, Hauer-Jensen M, Boerma M, Singh SP. Sulforaphane potentiates anticancer effects of doxorubicin and attenuates its cardiotoxicity in a breast cancer model. PLoS One 2018; 13:e0193918. [PMID: 29518137 PMCID: PMC5843244 DOI: 10.1371/journal.pone.0193918] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 02/20/2018] [Indexed: 11/19/2022] Open
Abstract
Breast cancer is the most common malignancy in women of the Western world. Doxorubicin (DOX) continues to be used extensively to treat early-stage or node-positive breast cancer, human epidermal growth factor receptor-2 (HER2)-positive breast cancer, and metastatic disease. We have previously demonstrated in a mouse model that sulforaphane (SFN), an isothiocyanate isolated from cruciferous vegetables, protects the heart from DOX-induced toxicity and damage. However, the effects of SFN on the chemotherapeutic efficacy of DOX in breast cancer are not known. Present studies were designed to investigate whether SFN alters the effects of DOX on breast cancer regression while also acting as a cardioprotective agent. Studies on rat neonatal cardiomyocytes and multiple rat and human breast cancer cell lines revealed that SFN protects cardiac cells but not cancer cells from DOX toxicity. Results of studies in a rat orthotopic breast cancer model indicated that SFN enhanced the efficacy of DOX in regression of tumor growth, and that the DOX dosage required to treat the tumor could be reduced when SFN was administered concomitantly. Additionally, SFN enhanced mitochondrial respiration in the hearts of DOX-treated rats and reduced cardiac oxidative stress caused by DOX, as evidenced by the inhibition of lipid peroxidation, the activation of NF-E2-related factor 2 (Nrf2) and associated antioxidant enzymes. These studies indicate that SFN not only acts synergistically with DOX in cancer regression, but also protects the heart from DOX toxicity through Nrf2 activation and protection of mitochondrial integrity and functions.
Collapse
Affiliation(s)
- Chhanda Bose
- University of Arkansas for Medical Sciences, Department of Geriatrics, Little Rock, Arkansas, United States of America
| | - Sanjay Awasthi
- Texas Tech Health Sciences Center, Division of Hematology & Oncology, Department of Internal Medicine, Lubbock, Texas, United States of America
| | - Rajendra Sharma
- University of Arkansas for Medical Sciences, Department of Pharmacology and Toxicology, Little Rock, Arkansas, United States of America
| | - Helen Beneš
- University of Arkansas for Medical Sciences, Department of Neurobiology and Developmental Sciences, Little Rock, Arkansas, United States of America
| | - Martin Hauer-Jensen
- University of Arkansas for Medical Sciences, Division of Radiation Health, Little Rock, Arkansas, United States of America
| | - Marjan Boerma
- University of Arkansas for Medical Sciences, Division of Radiation Health, Little Rock, Arkansas, United States of America
| | - Sharda P. Singh
- Texas Tech Health Sciences Center, Division of Hematology & Oncology, Department of Internal Medicine, Lubbock, Texas, United States of America
- University of Arkansas for Medical Sciences, Department of Pharmacology and Toxicology, Little Rock, Arkansas, United States of America
- Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, United States of America
| |
Collapse
|
43
|
Prevention of doxorubicin (DOX)-induced genotoxicity and cardiotoxicity: Effect of plant derived small molecule indole-3-carbinol (I3C) on oxidative stress and inflammation. Biomed Pharmacother 2018; 101:228-243. [PMID: 29494960 DOI: 10.1016/j.biopha.2018.02.088] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 02/02/2018] [Accepted: 02/20/2018] [Indexed: 01/22/2023] Open
Abstract
Doxorubicin (DOX) is an anthracycline group of antibiotic available for the treatment of broad spectrum of human cancers. However, patient receiving DOX-therapy, myelosuppression and genotoxicity which may lead to secondary malignancy and dose dependent cardiotoxicity is an imperative adverse effect. Mechanisms behind the DOX-induced toxicities are increased level of oxidative damage, inflammation and apoptosis. Therefore, in search of a potential chemoprotectant, naturally occurring glucosinolate breakdown product Indole-3-Carbinol (I3C) was evaluated against DOX-induced toxicities in Swiss albino mice. DOX was administered (5 mg/kg b.w., i.p.) and I3C was administered (20 mg/kg b.w., p.o.) in concomitant and 15 days pretreatment schedule. Results of the present study showed that I3C appreciably mitigated DOX-induced chromosomal aberrations, micronuclei formation, DNA damage and apoptosis in bone marrow niche. Histopathological observations revealed that DOX-intoxication resulted in massive structural and functional impairment of heart and bone marrow niche. However, oral administration of I3C significantly attenuated DOX-induced oxidative stress in the cardiac tissues as evident from decreased levels of ROS/RNS and lipid peroxidation, and by increased level of glutathione (reduced) and the activity of phase-II antioxidant enzymes. Additionally, administration of I3C significantly (P < 0.05) stimulated Nrf2-mediated activation of antioxidant response element (ARE) pathway and promoted expression of cytoprotective proteins heme oxygenase 1 (HO-1), NAD(P)H:quinine oxidoreductase 1 (NQO1) and GSTπ in bone marrow and cardiac tissues. In connection with that, I3C significantly attenuated DOX-induced inflammation by downregulation of pro-inflammatory mediators, viz., NF-kβ(p50), iNOS, COX-2 and IL-6 expression. Moreover, I3C attenuate DOX-induced apoptosis by up-regulation of Bcl2 and down-regulation of Bax and caspase-3 expression in bone marrow cells. Thus, this study suggests that I3C has promising chemoprotective efficacy against DOX-induced toxicities and indicates its future use as an adjuvant in chemotherapy.
Collapse
|
44
|
Omóbòwálé TO, Oyagbemi AA, Folasire AM, Ajibade TO, Asenuga ER, Adejumobi OA, Ola-Davies OE, Oyetola O, James G, Adedapo AA, Yakubu MA. Ameliorative effect of gallic acid on doxorubicin-induced cardiac dysfunction in rats. J Basic Clin Physiol Pharmacol 2018; 29:19-27. [PMID: 29016351 DOI: 10.1515/jbcpp-2016-0194] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 07/23/2017] [Indexed: 06/07/2023]
Abstract
BACKGROUND The use of doxorubicin (DOX) as an antineoplastic agent has been greatly limited because of the myriad of toxic sequelae associated with it. The aim of this study was to assess the protective effects of gallic acid (GA) on DOX-induced cardiac toxicity in rats. METHODS Sixty male rats (Wistar strain) were used in this study. They were divided into six groups (A-F) each containing 10 animals. Group A was the control. Rats in Groups B, C, and D were treated with DOX at the dosage of 15 mg/kg body weight i.p. Prior to this treatment, rats in Groups C and D had been treated orally with GA for 7 days at the dosage of 60 and 120 mg/kg, respectively. Animals from Groups E and F received only 60 and 120 mg/kg GA, respectively, which were administered orally for 7 days. RESULTS The exposure of rats to DOX led to a significant (p<0.05) decrease in the cardiac antioxidant defence system and elevation of creatine kinase myocardial band and lactate dehydrogenase. The electrocardiography results showed a significant decrease in heart rate, QRS, and QT-segment prolongation. GA alone improved the antioxidant defence system. CONCLUSIONS The GA pretreatment significantly alleviated GA-associated ECG abnormalities, restored the antioxidant status and prevented cardiac damage.
Collapse
Affiliation(s)
| | - Ademola A Oyagbemi
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Ayorinde M Folasire
- Department of Radiation Oncology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Temitayo O Ajibade
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Ebunoluwa R Asenuga
- Department of Veterinary Physiology, Pharmacology and Biochemistry, University of Benin, Benin City, Nigeria
| | | | - Olufunke E Ola-Davies
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Orotusin Oyetola
- Department of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Gana James
- Department of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Adeolu A Adedapo
- Department of Veterinary Pharmacology and Toxicology, University of Ibadan, Ibadan, Nigeria
| | - Momoh A Yakubu
- Department of Environmental and Interdisciplinary Sciences, College of Science, Engineering and Technology, Vascular Biology Unit, Center for Cardiovascular Diseases, COPHS, Texas Southern University, Houston, TX, USA
| |
Collapse
|
45
|
Heated naringin mitigate the genotoxicity effect of Mitomycin C in BALB/c mice through enhancing the antioxidant status. Biomed Pharmacother 2017; 97:1417-1423. [PMID: 29156531 DOI: 10.1016/j.biopha.2017.11.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 10/26/2017] [Accepted: 11/03/2017] [Indexed: 11/23/2022] Open
Abstract
A major problem with cancer chemotherapy is its severe toxic effects on non-target tissues. Assessment of natural products for their protective effect against anticancer drugs induced toxicity is gaining importance in cancer biology. The aim of the present study was to evaluate the effect of native and thermal treated naringin on the protective effect against mitomycin C (MMC) induced genotoxicity. The genotoxicity in liver kidney and brain cells isolated from Balb/C mice were evaluated by performing the comet assay. Antioxidant and lipid peroxidation assays were carried out to understand the protective effects of these compounds. The comet assay showed that heated and native naringin were not genotoxic at the tested dose (40 mg/kg b.w) on liver, kidney and brain cells. A significant decrease in DNA damages was observed, at the tested doses (20 mg/kg b.w and 40 mg/kg b.w) suggesting a protective role of these molecules against the genotoxicity induced by mitomycin C on liver, kidney and brain cells. Moreover, administration of MMC (6 mg/kg b.w.) altered the activities of glutathione peroxidase and superoxide dismutase accompanied by a significant increase of lipid peroxidation. Pretreatment of mouse with heated and native naringin before MMC administration significantly raised the glutathione peroxidase and superoxide dismutase activities followed by a reduced MMC-induced lipid peroxidation. Our study demonstrated that heat treatment of naringin preserve activities of native naringin. The genoprotective properties of heated and native naringin against MMC could be attributed to its antioxidant activities and its inhibitory effect on lipid peroxidation.
Collapse
|
46
|
Tian XQ, Ni XW, Xu HL, Zheng L, ZhuGe DL, Chen B, Lu CT, Yuan JJ, Zhao YZ. Prevention of doxorubicin-induced cardiomyopathy using targeted MaFGF mediated by nanoparticles combined with ultrasound-targeted MB destruction. Int J Nanomedicine 2017; 12:7103-7119. [PMID: 29026304 PMCID: PMC5627735 DOI: 10.2147/ijn.s145799] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The present study seeks to observe the preventive effects of doxorubicin-induced cardiomyopathy (DOX-CM) in rats using targeted non-mitogenic acidic fibroblast growth factor (MaFGF) mediated by nanoparticles (NP) combined with ultrasound-targeted MB destruction (UTMD). DOX-CM rats were induced by intraperitoneally injected doxorubicin. Six weeks after intervention, the indices from the transthoracic echocardiography and velocity vector imaging showed that the left ventricular function in the MaFGF-loaded NP (MaFGF-NP) + UTMD group was significantly improved compared with the DOX-CM group. The increased malondialdehyde and decreased superoxide dismutase were observed in the DOX-CM group, while a significant increase in superoxide dismutase and a decrease in malondialdehyde were detected in the groups treated with MaFGF-NP + UTMD. From the Masson staining, the MaFGF-NP + UTMD group showed a significant difference from the DOX-CM group. The cardiac collagen volume fraction and the ratio of the perivascular collagen area to the luminal area number of terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end labelling positive cells in the MaFGF-NP + UTMD group decreased to 8.9%, 0.55-fold, compared with the DOX-CM group (26.5%, 1.7-fold). From terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end labelling staining, the results showed the strongest inhibition of apoptosis progress in MaFGF-NP + UTMD group. The immunohistochemical staining of the TGF-β1 in MaFGF-NP + UTMD group reached 3.6%, which was much lower than that of the DOX-CM group (12.6%). These results confirmed that the abnormalities, including left ventricular dysfunction, myocardial fibrosis, cardiomyocytes apoptosis and oxidative stress, could be suppressed by twice weekly MaFGF treatments for 6 consecutive weeks (free MaFGF or MaFGF-NP+/UTMD), with the strongest improvements observed in the MaFGF-NP + UTMD group. Western blot analyses of the heart tissue further revealed the highest pAkt levels, highest anti-apoptosis protein (Bcl-2) levels and strongest reduction in proapoptosis protein (Bax) levels in the MaFGF-NP + UTMD group. This study confirmed the preventive effects of DOX-CM in the rats with MaFGF-NP and UTMD by retarding myocardial fibrosis, inhibiting oxidative stress, and decreasing cardiomyocyte apoptosis.
Collapse
Affiliation(s)
- Xin-Qiao Tian
- Department of Ultrasonography, Henan Provincial People's Hospital (People's Hospital of Zhengzhou University), Zhengzhou.,Department of Pharmacology, College of Pharmaceutical Sciences, Wenzhou Medical University
| | - Xian-Wei Ni
- Department of Ultrasonography, The Second Affiliated Hospital of Wenzhou Medical University
| | - He-Lin Xu
- Department of Pharmacology, College of Pharmaceutical Sciences, Wenzhou Medical University
| | - Lei Zheng
- Department of Ultrasonography, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - De-Li ZhuGe
- Department of Pharmacology, College of Pharmaceutical Sciences, Wenzhou Medical University
| | - Bin Chen
- Department of Ultrasonography, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Cui-Tao Lu
- Department of Pharmacology, College of Pharmaceutical Sciences, Wenzhou Medical University
| | - Jian-Jun Yuan
- Department of Ultrasonography, Henan Provincial People's Hospital (People's Hospital of Zhengzhou University), Zhengzhou
| | - Ying-Zheng Zhao
- Department of Pharmacology, College of Pharmaceutical Sciences, Wenzhou Medical University
| |
Collapse
|
47
|
Viswanatha GL, Shylaja H, Moolemath Y. The beneficial role of Naringin- a citrus bioflavonoid, against oxidative stress-induced neurobehavioral disorders and cognitive dysfunction in rodents: A systematic review and meta-analysis. Biomed Pharmacother 2017; 94:909-929. [PMID: 28810519 DOI: 10.1016/j.biopha.2017.07.072] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 07/04/2017] [Accepted: 07/17/2017] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVES Naringin is a bioflavonoid, very abundantly found in citrus species. In literature, naringin has been scientifically well documented for its beneficial effects in various neurological disorders. In this systematic review and meta-analysis, we have made an attempt to correlate the protective role of naringin against oxidative stress-induced neurological disorders in rodents. METHODS The systematic search was performed using electronic databases; the search was mainly focused on the role of naringin in oxidative stress-induced neuropathological conditions in rodents. While, the meta-analysis was performed on the effect of naringin on oxidative stress markers [superoxide dismutase (SOD), catalase (CAT), glutathione-S-transferase (GST), reduced glutathione (GSH), lipid peroxidation (LPO)], nitrite, mitochondrial complexes (I to IV) and enzymes (acetylcholinesterase, Na+-K+-ATPase, Ca2+-ATPase, and Mg2+-ATPase) in the rodent brain. The data was analyzed using Review Manager Software. THE RESULTS Based on the inclusion and exclusion criteria, twenty studies were selected. The meta-analysis revealed that, naringin could significantly inhibit various physical and chemical stimuli- induced neurological perturbances in the rodent brain, mediated through oxidative stress. Further, naringin also significantly restored the levels of all the oxidative stress markers (oxidative, nitrosative, enzymes, and mitochondrial complexes) in different parts of the rodent brain. SUMMARY This systematic review and meta-analysis supports the available scientific evidence on the beneficial role of naringin in the management of various neurological ailments. However, further studies involving human subjects is recommended to establish the safety and therapeutic efficacy in humans.
Collapse
Affiliation(s)
| | - H Shylaja
- Independent Researcher, Kengeri, Bangalore, 560060, Karnataka, India
| | - Yogananda Moolemath
- Vittarthaa Life Sciences, Bommasandra Industrial Area, Bangalore, 560099, Karnataka, India
| |
Collapse
|
48
|
Xianchu L, Lan PZ, Qiufang L, Yi L, Xiangcheng R, Wenqi H, Yang D. Naringin protects against lipopolysaccharide-induced cardiac injury in mice. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2016; 48:1-6. [PMID: 27716530 DOI: 10.1016/j.etap.2016.09.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 09/06/2016] [Accepted: 09/11/2016] [Indexed: 06/06/2023]
Abstract
Previous research has demonstrated that lipopolysaccharide (LPS) can induce sepsis and lead to myocardial dysfunction. Naringin has various biological activities in LPS-induced sepsis. In this study, our aim was to investigate the effects of Naringin on LPS-induced cardiac injury and clarify its potential mechanism. We found that in vivo treatment with Naringin significantly ameliorated body weight loss, and attenuated cardiac histopathological changes after LPS challenge. Furthermore, Naringin inhibited LPS-induced increase of TNF-α, IL-1β and IL-6 activities to alleviate inflammatory response in heart. Moreover, Naringin supplement dramatically increased SOD levels, and prevented MDA levels to ameliorate oxidative stress compared with the LPS group in heart. Lastly, treatment with Naringin also significantly decreased the ratio of BAX to BCL-2 to resist apoptosis in heart. It is concluded that Naringin may be a promising therapeutic agent on LPS-induced cardiac injury by anti-inflammatory, anti-oxidant and anti-apoptotic effects.
Collapse
Affiliation(s)
- Liu Xianchu
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, Hunan Normal University, Changsha, China
| | - Professor Zheng Lan
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, Hunan Normal University, Changsha, China.
| | - Li Qiufang
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, Hunan Normal University, Changsha, China
| | - Liu Yi
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, Hunan Normal University, Changsha, China
| | - Ruan Xiangcheng
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, Hunan Normal University, Changsha, China
| | - Hou Wenqi
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, Hunan Normal University, Changsha, China
| | - Ding Yang
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, Hunan Normal University, Changsha, China
| |
Collapse
|
49
|
Jangra A, Kwatra M, Singh T, Pant R, Kushwah P, Ahmed S, Dwivedi D, Saroha B, Lahkar M. Edaravone alleviates cisplatin-induced neurobehavioral deficits via modulation of oxidative stress and inflammatory mediators in the rat hippocampus. Eur J Pharmacol 2016; 791:51-61. [PMID: 27492363 DOI: 10.1016/j.ejphar.2016.08.003] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 07/29/2016] [Accepted: 08/01/2016] [Indexed: 12/19/2022]
Abstract
Cisplatin is a chemotherapeutic agent used in the treatment of malignant tumors. A major clinical limitation of cisplatin is its potential toxic effects, including neurotoxicity. Edaravone, a potent free radical scavenger, has been reported to have the neuroprotective effect against neurological deficits. The aim of the present study was to determine the neuroprotective effect of edaravone against cisplatin-induced behavioral and biochemical anomalies in male Wistar rats. Our results showed that cisplatin (5mg/kg/week, i.p.) administration for seven weeks caused marked cognitive deficits and motor incoordination in rats. This was accompanied by oxido-nitrosative stress, neuroinflammation, NF-κB activation and down-regulation of Nrf2/HO-1 gene expression level in the hippocampus. Edaravone (10mg/kg/week, i.p.) treatment for seven weeks inhibited the aforementioned neurobehavioral and neurochemical deficits. Furthermore, edaravone was found to up-regulate the gene expression level of Nrf2/HO-1 and prevented the cisplatin-induced NF-κB activation. These findings demonstrated that oxido-nitrosative stress and inflammatory signaling mediators play a key role in the development of cisplatin-induced neurobehavioral deficits which were prevented by edaravone treatment.
Collapse
Affiliation(s)
- Ashok Jangra
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Guwahati, Assam, India
| | - Mohit Kwatra
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Guwahati, Assam, India
| | - Tavleen Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Guwahati, Assam, India
| | - Rajat Pant
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Guwahati, Assam, India
| | - Pawan Kushwah
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Guwahati, Assam, India
| | - Sahabuddin Ahmed
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Guwahati, Assam, India
| | - Durgesh Dwivedi
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Guwahati, Assam, India
| | - Babita Saroha
- Department of Biotechnology, University Institute of Engineering & Technology (UIET), Maharshi Dayanand University, Rohtak, Haryana, India
| | - Mangala Lahkar
- Department of Pharmacology, Gauhati Medical College, Guwahati, India.
| |
Collapse
|
50
|
Naringin and Sertraline Ameliorate Doxorubicin-Induced Behavioral Deficits Through Modulation of Serotonin Level and Mitochondrial Complexes Protection Pathway in Rat Hippocampus. Neurochem Res 2016; 41:2352-66. [DOI: 10.1007/s11064-016-1949-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 04/02/2016] [Accepted: 05/06/2016] [Indexed: 01/16/2023]
|