1
|
Yavuz Türel G, Aslan Koşar P. Protective efficacy of ramelteon on methotrexate-induced DNA damage. Drug Chem Toxicol 2024:1-7. [PMID: 38984403 DOI: 10.1080/01480545.2024.2375300] [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: 04/06/2024] [Accepted: 06/26/2024] [Indexed: 07/11/2024]
Abstract
Ramelteon (RMLT) is a melatonin receptor agonist that it has antioxidative and anti-inflammatory effects associated with DNA damage through different mechanisms of action. In this regard, we investigated the potential usefulness of RMLT as a protective agent against methotrexate (MTX)-induced DNA damage. Four groups were constituted from 32 Wistar albino rats: Negative control, RMLT, MTX, and MTX + RMLT. Twenty mg/kg MTX (i.p., single dose) and RMLT 10 mg/kg (oral, 7 days) was administered. Comet assay was used and the parameter %TailDNA was used to detect DNA damage. %TailDNA was 4.90 ± 0.19 in the control group, 7.85 ± 0.33 in the MTX group, 5.49 ± 0.24 in the RMLT group, and 5.86 ± 0.23 in the MTX + RMLT group. While there was a significant increase in DNA damage in the MTX-treated group compared to the control group, there was a significant reduction in DNA damage in the MTX + RMLT group, compared to the MTX group (p < 0.001). In conclusion, it was observed that combined treatment with RMLT significantly reduced MTX-induced DNA damage.
Collapse
Affiliation(s)
- Gülçin Yavuz Türel
- Faculty of Medicine, Department of Medical Biology, Süleyman Demirel University, Isparta, Turkey
| | - Pınar Aslan Koşar
- Faculty of Medicine, Department of Medical Biology, Süleyman Demirel University, Isparta, Turkey
| |
Collapse
|
2
|
da Cunha de Medeiros P, Nunes EA, Barcelos GRM, Perobelli JE. Genotoxicity and cytotoxicity of antineoplastic drugs at environmentally relevant concentrations after long-term exposure. Toxicol Res (Camb) 2024; 13:tfae049. [PMID: 38533178 PMCID: PMC10962016 DOI: 10.1093/toxres/tfae049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 02/20/2024] [Accepted: 03/12/2024] [Indexed: 03/28/2024] Open
Abstract
Introduction 5-fluorouracil (5-FU) and methotrexate (MTX) are the antineoplastic drugs most commonly used worldwide. Considered cytotoxic, these pharmaceuticals exhibit low specificity, causing damage not only to cancer cells but also to healthy cells in organisms. After being consumed and metabolized, these drugs are excreted through urine and feces, followed by wastewater treatment. However, conventional treatments do not have the capacity to completely remove these substances, risking their introduction into freshwater systems. This could pose a risk to human health even at low concentrations. Aims Thus, the present study aimed to investigate the genotoxicity, cytotoxicity, and mutagenicity of 5-FU and MTX at environmentally relevant concentrations after a long-term exposure, using adult male rats as an experimental model. Methods Male Wistar rats (70 days old) were distributed into 4 groups (n = 10/group): control, received only vehicle; MTX, received methotrexate at 10ngL-1; 5-FU received 5-fluorouracil at 10ngL-1; and MTX + 5-FU, received a combination of MTX and 5-FU at 10ngL-1 each. The period of exposure was from postnatal day (PND) 70 to PND 160, through drinking water. After that, the animals were euthanized and the samples (liver, testis, femoral bone marrow, and peripheral blood) were obtained. Results Increased DNA fragmentation was observed in the peripheral blood, liver, and testis, altering the parameters of the tail moment and tail intensity in the Comet assay. Besides, the change in the ratio between PCE and NCE indicates bone marrow suppression. Conclusion These findings warn the adverse effects for the general population worldwide chronically exposed to these drugs at trace concentration unintentionally.
Collapse
Affiliation(s)
- P da Cunha de Medeiros
- Laboratory of Experimental Toxicology – LATOEX, Universidade Federal de São Paulo, Instituto do Mar, Carvalho de Mendonça, 144, Santos 11070-100, SP, Brazil
| | - E A Nunes
- Department of Biosciences, Laboratory of Gene-Environmental Interactions in Toxicology – GENINTOX, Universidade Federal de São Paulo, XV de novembro 195, sala 614, Santos 11.010-151, SP, Brazil
| | - G R M Barcelos
- Department of Biosciences, Laboratory of Gene-Environmental Interactions in Toxicology – GENINTOX, Universidade Federal de São Paulo, XV de novembro 195, sala 614, Santos 11.010-151, SP, Brazil
| | - J E Perobelli
- Laboratory of Experimental Toxicology – LATOEX, Universidade Federal de São Paulo, Instituto do Mar, Carvalho de Mendonça, 144, Santos 11070-100, SP, Brazil
| |
Collapse
|
3
|
Zhang Y, Sheng Z, Xiao J, Li Y, Huang J, Jia J, Zeng X, Li L. Advances in the roles of glycyrrhizic acid in cancer therapy. Front Pharmacol 2023; 14:1265172. [PMID: 37649893 PMCID: PMC10463042 DOI: 10.3389/fphar.2023.1265172] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 08/07/2023] [Indexed: 09/01/2023] Open
Abstract
Since the first 70 years of reporting cancer chemotherapy, malignant tumors have been the second most common cause of death in children and adults. Currently, the commonly used anti-cancer methods include surgery, chemotherapy, radiotherapy, and immunotherapy. Although these treatment methods could alleviate cancer, they lead to different forms of side effects and have no particularly significant effect on prolonging the patients' life span. Glycyrrhizic acid (GL), a native Chinese herbal extract, has a wide range of pharmacological effects, such as anti-cancer, anti-inflammatory, antioxidant, and immune regulation. In this review, the anti-cancer effects and mechanisms of GL are summarized in various cancers. The inhibition of GL on chemotherapy-induced side effects, including hepatotoxicity, nephrotoxicity, genotoxicity, neurotoxicity and pulmonary toxicity, is highlighted. Therefore, GL may be a promising and ideal drug for cancer therapy.
Collapse
Affiliation(s)
- Yuqian Zhang
- Research Center of Neuroscience, Jiaxing University Medical College, Jiaxing, China
| | - Zixuan Sheng
- Research Center of Neuroscience, Jiaxing University Medical College, Jiaxing, China
| | - Jing Xiao
- Research Center of Neuroscience, Jiaxing University Medical College, Jiaxing, China
| | - Yang Li
- Research Center of Neuroscience, Jiaxing University Medical College, Jiaxing, China
| | - Jie Huang
- Research Center of Neuroscience, Jiaxing University Medical College, Jiaxing, China
| | - Jinjing Jia
- Research Center of Neuroscience, Jiaxing University Medical College, Jiaxing, China
- Department of Physiology, Jiaxing University Medical College, Jiaxing, China
| | - Xiansi Zeng
- Research Center of Neuroscience, Jiaxing University Medical College, Jiaxing, China
- Department of Biochemistry and Molecular Biology, Jiaxing University Medical College, Jiaxing, China
| | - Li Li
- Research Center of Neuroscience, Jiaxing University Medical College, Jiaxing, China
- Department of Physiology, Jiaxing University Medical College, Jiaxing, China
| |
Collapse
|
4
|
Ibrahim MA, Khalifa AM, Mohamed AA, Galhom RA, Korayem HE, Abd El-Fadeal NM, Abd-Eltawab Tammam A, Khalifa MM, Elserafy OS, Abdel-Karim RI. Bone-Marrow-Derived Mesenchymal Stem Cells, Their Conditioned Media, and Olive Leaf Extract Protect against Cisplatin-Induced Toxicity by Alleviating Oxidative Stress, Inflammation, and Apoptosis in Rats. TOXICS 2022; 10:toxics10090526. [PMID: 36136492 PMCID: PMC9504158 DOI: 10.3390/toxics10090526] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/26/2022] [Accepted: 09/01/2022] [Indexed: 06/01/2023]
Abstract
BACKGROUND Hepatic and renal damage is a cisplatin (Cis)-induced deleterious effect that is a major limiting factor in clinical chemotherapy. OBJECTIVES The current study was designed to investigate the influence of pretreatment with olive leaf extract (OLE), bone-marrow-derived mesenchymal stem cells (BM-MSC), and their conditioned media (CM-MSC) against genotoxicity, nephrotoxicity, hepatotoxicity, and immunotoxicity induced by cisplatin in rats. METHODS The rats were randomly divided into six groups (six rats each) as follows: Control; OLE group, treated with OLE; Cis group, treated with a single intraperitoneal dose of Cis (7 mg/kg bw); Cis + OLE group, treated with OLE and cisplatin; Cis + CM-MSC group, treated with BM-MSC conditioned media and Cis; and Cis + MSC group, treated with BM-MSC in addition to Cis. RESULTS Cis resulted in a significant deterioration in hepatic and renal functions and histological structures. Furthermore, it increased inflammatory markers (TNF-α, IL-6, and IL-1β) and malondialdehyde (MDA) levels and decreased glutathione (GSH) content, total antioxidant capacity (TAC), catalase (CAT), and superoxide dismutase (SOD) activity in hepatic and renal tissues. Furthermore, apoptosis was evident in rat tissues. A significant increase in serum 8-hydroxy-2-deoxyguanosine (8-OH-dG), nitric oxide (NO) and lactate dehydrogenase (LDH), and a decrease in lysozyme activity were detected in Cis-treated rats. OLE, CM-MSC, and BM-MSC have significantly ameliorated Cis-induced deterioration in hepatic and renal structure and function and improved oxidative stress and inflammatory markers, with preference to BM-MSC. Moreover, apoptosis was significantly inhibited, evident from the decreased expression of Bax and caspase-3 genes and upregulation of Bcl-2 proteins in protective groups as compared to Cis group. CONCLUSIONS These findings indicate that BM-MSC, CM-MSC, and OLE have beneficial effects in ameliorating cisplatin-induced oxidative stress, inflammation, and apoptosis in the hepatotoxicity, nephrotoxicity, immunotoxicity, and genotoxicity in a rat model.
Collapse
Affiliation(s)
- Mahrous A. Ibrahim
- Forensic Medicine and Clinical Toxicology, College of Medicine, Jouf University, Sakaka 41412, Saudi Arabia
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Suez Canal University (SCU), Ismailia 41522, Egypt or
| | - Athar M. Khalifa
- Pathology Department, College of Medicine, Jouf University, Sakaka 41412, Saudi Arabia
| | - Alaa A. Mohamed
- Medical Biochemistry Division, Pathology Department, College of Medicine, Jouf University, Sakaka 41412, Saudi Arabia
- Medical Biochemistry Department, Faculty of Medicine, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Rania A. Galhom
- Human Anatomy and Embryology Department, Faculty of Medicine, Suez Canal University (SCU), Ismailia 41522, Egypt
- Center of Excellence in Molecular and Cellular Medicine (CEMCM), Faculty of Medicine, Suez Canal University (SCU), Ismailia 41522, Egypt
- Human Anatomy and Embryology Department, Faculty of Medicine, Badr University in Cairo (BUC), Cairo 11829, Egypt
| | - Horeya E. Korayem
- Histology and Cell Biology Department, Faculty of Medicine, Suez Canal University (SCU), Ismailia 41522, Egypt
| | - Noha M. Abd El-Fadeal
- Center of Excellence in Molecular and Cellular Medicine (CEMCM), Faculty of Medicine, Suez Canal University (SCU), Ismailia 41522, Egypt
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Suez Canal University (SCU), Ismailia 41522, Egypt
- Oncology Diagnostic Unit, Faculty of Medicine, Suez Canal University (SCU), Ismailia 41522, Egypt
| | - Ahmed Abd-Eltawab Tammam
- Physiology Department, College of Medicine, Jouf University, Sakaka 41412, Saudi Arabia
- Physiology Department, Faculty of Medicine, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Mohamed Mansour Khalifa
- Human Physiology Department, Faculty of Medicine, Cairo University, Cairo 11562, Egypt
- Human Physiology Department, College of Medicine, King Saud University, Riyadh 11472, Saudi Arabia
| | - Osama S. Elserafy
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Cairo University, Cairo 11562, Egypt
- Criminal Justice and Forensic Sciences Department, King Fahd Security College, Riyadh 11451, Saudi Arabia
| | - Rehab I. Abdel-Karim
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Suez Canal University (SCU), Ismailia 41522, Egypt or
| |
Collapse
|
5
|
Abadi AJ, Mirzaei S, Mahabady MK, Hashemi F, Zabolian A, Hashemi F, Raee P, Aghamiri S, Ashrafizadeh M, Aref AR, Hamblin MR, Hushmandi K, Zarrabi A, Sethi G. Curcumin and its derivatives in cancer therapy: Potentiating antitumor activity of cisplatin and reducing side effects. Phytother Res 2021; 36:189-213. [PMID: 34697839 DOI: 10.1002/ptr.7305] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/03/2021] [Accepted: 09/25/2021] [Indexed: 12/12/2022]
Abstract
Curcumin is a phytochemical isolated from Curcuma longa with potent tumor-suppressor activity, which has shown significant efficacy in pre-clinical and clinical studies. Curcumin stimulates cell death, triggers cycle arrest, and suppresses oncogenic pathways, thereby suppressing cancer progression. Cisplatin (CP) stimulates DNA damage and apoptosis in cancer chemotherapy. However, CP has adverse effects on several organs of the body, and drug resistance is frequently observed. The purpose of the present review is to show the function of curcumin in decreasing CP's adverse impacts and improving its antitumor activity. Curcumin administration reduces ROS levels to prevent apoptosis in normal cells. Furthermore, curcumin can inhibit inflammation via down-regulation of NF-κB to maintain the normal function of organs. Curcumin and its nanoformulations can reduce the hepatoxicity, neurotoxicity, renal toxicity, ototoxicity, and cardiotoxicity caused by CP. Notably, curcumin potentiates CP cytotoxicity via mediating cell death and cycle arrest. Besides, curcumin suppresses the STAT3 and NF-ĸB as tumor-promoting pathways, to enhance CP sensitivity and prevent drug resistance. The targeted delivery of curcumin and CP to tumor cells can be mediated nanostructures. In addition, curcumin derivatives are also able to reduce CP-mediated side effects, and increase CP cytotoxicity against various cancer types.
Collapse
Affiliation(s)
- Asal Jalal Abadi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Mahmood Khaksary Mahabady
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Farid Hashemi
- Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Amirhossein Zabolian
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Fardin Hashemi
- School of Rehabilitation, Department of Physical Therapy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Pourya Raee
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shahin Aghamiri
- Student Research Committee, Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Tuzla, Turkey.,Sabanci University Nanotechnology Research and Application Center (SUNUM), Istanbul, Turkey
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA.,Vice President at Translational Sciences, Xsphera Biosciences Inc, Boston, Massachusetts, USA
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, South Africa.,Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Istanbul, Turkey.,Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Sariyer, Istanbul, Turkey
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
6
|
Rababa'H AM, Alzoubi KH, Khabour OF, Ababneh M. Ameliorative effect of metformin on methotrexate-induced genotoxicity: An in vitro study in human cultured lymphocytes. Biomed Rep 2021; 15:59. [PMID: 34094535 PMCID: PMC8165753 DOI: 10.3892/br.2021.1435] [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: 02/16/2021] [Accepted: 04/22/2021] [Indexed: 11/05/2022] Open
Abstract
Methotrexate is a folic acid antagonist that has been shown to be genotoxic to normal healthy cells. Metformin is an insulin-sensitizing agent, with multiple potential pharmacodynamic profiles. The aim of the present study was to evaluate the genotoxic effect of methotrexate on DNA and the potential ameliorative effect of metformin on chromosomal damage induced by methotrexate. The present study was performed in vitro, and the frequency of chromosomal aberrations (CAs) and sister chromatid exchanges (SCEs) in human cultured lymphocytes were measured. Blood samples from five non-smoking healthy men aged 20-35 years were donated and used in the present study. Treatment of cultured blood cells with methotrexate significantly increased the number of cells with CAs (P<0.0001) and the frequency of SCEs (P<0.0001). The chromosomal injury induced by methotrexate was significantly reduced by pretreatment of the samples with metformin (P<0.0001). Importantly, the treatment of the cells with metformin alone did not affect the frequency of SCEs compared with the control group (P>0.05). Additionally, methotrexate and metformin alone, and combined, induced significant decreases in the proliferative index compared with the control group (P<0.05). In conclusion, metformin ameliorated the genotoxicity induced by methotrexate in cultured human lymphocytes.
Collapse
Affiliation(s)
- Abeer M Rababa'H
- Department of Clinical Pharmacy, College of Pharmacy, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Karem H Alzoubi
- Department of Clinical Pharmacy, College of Pharmacy, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Omar F Khabour
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Mera Ababneh
- Department of Clinical Pharmacy, College of Pharmacy, Jordan University of Science and Technology, Irbid 22110, Jordan
| |
Collapse
|
7
|
Rababa'h AM, Hussein SA, Khabour OF, Alzoubi KH. The Protective Effect of Cilostazol in Genotoxicity Induced by Methotrexate in Human Cultured Lymphocytes. Curr Mol Pharmacol 2021; 13:137-143. [PMID: 31702497 DOI: 10.2174/1874467212666191023120118] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 10/13/2019] [Accepted: 10/15/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Methotrexate is an antagonist of folic acid that has been shown to be genotoxic to healthy body cells via induction of oxidative stress. Cilostazol is a phosphodiesterase III inhibitor and a potent antioxidant drug. OBJECTIVE To evaluate the potential protective effect of cilostazol on methotrexate genotoxicity. METHODS The genotoxic effect of methotrexate by measuring the frequency of chromosomal aberrations (CAs) and sister chromatid exchanges (SCEs) in human cultured lymphocytes was studied. RESULTS Methotrexate significantly increased the frequency of CAs and SCEs (p < 0.0001) as compared to control cultures. This chromosomal damage induced by methotrexate was considerably decreased by pretreatment of the cells with cilostazol (P < 0.01). Moreover, the results showed that methotrexate resulted in a notable reduction (P < 0.01) in cells kinetic parameters, the mitotic index (MI) and the proliferative index (PI). Similarly, cilostazol significantly reduced the mitotic index, which could be related to the anti-proliferative effect (P < 0.01). CONCLUSION Methotrexate is genotoxic, and cilostazol could prevent the methotrexate-induced chromosomal damage with no modulation of methotrexate-induced cytotoxicity.
Collapse
Affiliation(s)
- Abeer M Rababa'h
- Department of Clinical Pharmacy, College of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Samah A Hussein
- Department of Clinical Pharmacy, College of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Omar F Khabour
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, Jordan University of Science and Technology, Irbid, Jordan
| | - Karem H Alzoubi
- Department of Clinical Pharmacy, College of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
8
|
Jensen NB, Justesen SD, Larsen A, Ernst E, Pedersen LH. A systematic overview of the spermatotoxic and genotoxic effects of methotrexate, ganciclovir and mycophenolate mofetil. Acta Obstet Gynecol Scand 2021; 100:1557-1580. [PMID: 33755191 DOI: 10.1111/aogs.14151] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 02/23/2021] [Accepted: 03/14/2021] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Immunosuppressant drugs are increasingly being used in the reproductive years. Theoretically, such medications could affect fetal health either through changes in the sperm DNA or through fetal exposure caused by a presence in the seminal fluid. This systematic overview summarizes existing literature on the spermatotoxic and genotoxic potentials of methotrexate (MTX), a drug widely used to treat rheumatic and dermatologic diseases, and mycophenolate mofetil (MMF), which alone or supplemented with ganciclovir (GCV) may be crucial for the survival of organ transplants. MATERIAL AND METHODS The systematic overview was performed in accordance with the PRISMA guidelines: A systematic literature search of the MEDLINE and Embase databases was done using a combination of relevant terms to search for studies on spermatotoxic or genotoxic changes related to treatment with MTX, GCV or MMF. The search was restricted to English language literature, and to in vivo animal studies (mammalian species) and clinical human studies. RESULTS A total of 102 studies were identified, hereof 25 human and 77 animal studies. For MTX, human studies of immunosuppressive dosages show transient effect on sperm quality parameters, which return to reference values within 3 months. No human studies have investigated the sperm DNA damaging effect of MTX, but in other organs the genotoxic effects of immunosuppressive doses of MTX are fluctuating. In animals, immunosuppressive and cytotoxic doses of MTX adversely affect sperm quality parameters and show widespread genotoxic damages in various organs. Cytotoxic doses transiently change the DNA material in all cell stages of spermatogenesis in rodents. For GCV and MMF, data are limited and the results are indeterminate, for which reason spermatotoxic and genotoxic potentials cannot be excluded. CONCLUSIONS Data from human and animal studies indicate transient spermatotoxic and genotoxic potentials of immunosuppressive and cytotoxic doses of MTX. There are a limited number of studies investigating GCV and MMF.
Collapse
Affiliation(s)
| | | | - Agnete Larsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Erik Ernst
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Laboratory for Reproduction, Institute of Anatomy, Aarhus University, Aarhus, Denmark
| | - Lars H Pedersen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Obstetrics and Gynecology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
9
|
Martinovich GG, Martinovich IV, Vcherashniaya AV, Zenkov NK, Menshchikova EB, Cherenkevich SN. Chemosensitization of Tumor Cells by Phenolic Antioxidants: The Role of the Nrf2 Transcription Factor. Biophysics (Nagoya-shi) 2020. [DOI: 10.1134/s000635092006010x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
10
|
Curcumin as a preventive or therapeutic measure for chemotherapy and radiotherapy induced adverse reaction: A comprehensive review. Food Chem Toxicol 2020; 145:111699. [PMID: 32858134 DOI: 10.1016/j.fct.2020.111699] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/27/2020] [Accepted: 08/19/2020] [Indexed: 12/12/2022]
Abstract
Curcumin has attracted much attention for medicinal purposes in wide range of illnesses including cancer. In some studies, its efficacy is evaluated against chemotherapy and radiotherapy induced adverse reaction and also as adjuvant to cancer treatment. Here we have tried to present a comprehensive review on protective and therapeutic effect of curcumin against these side effects. METHOD: The data were collected by searching Scopus, PubMed, Medline, and Cochrane database systematic reviews, using key words "nephrotoxicity", "cardiotoxicity", "genotoxicity", "ototoxicity", "hepatotoxicity", "reproductive toxicity", "myelosuppression", "pulmonary toxicity", "radiotherapy induced side effect" with "turmeric" and "curcumin". Although curcumin has low bioavailability, it has shown brilliant profile on prevention and management of chemotherapy and radiotherapy induced adverse reactions, particularly based on in vitro and in vivo studies and limited number of human studies on radiotherapy adverse reactions. Antioxidant and anti-inflammatory properties of the curcumin are the main proposed mechanism of action for management and prevention of adverse reactions. One of the major points regarding the protective effect of curcumin is its wide tolerable therapeutic range of dose with minimal side effects. Furthermore, new nano-formulations help to improve the bioavailability, increase in efficacy and lower the adverse effects. In conclusion, based on the present knowledge, curcumin has significant supportive potential in patients receiving chemotherapy or radiotherapy and may be suggested as adjutant with cancer treatments. Further well-designed human studies are recommended.
Collapse
|
11
|
Zhang K, Weng H, Yang J, Wu C. Protective effect of Liuwei Dihuang Pill on cisplatin-induced reproductive toxicity and genotoxicity in male mice. JOURNAL OF ETHNOPHARMACOLOGY 2020; 247:112269. [PMID: 31610261 DOI: 10.1016/j.jep.2019.112269] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 09/24/2019] [Accepted: 10/01/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cisplatin (CP) is the classical chemotherapeutic drug for various cancer, but it also accompanies reproductive toxicity and genotoxicity. Liuwei Dihuang Pill (LW) is the traditional Chinese medicine prescription for treating Kidney-Yin deficiency syndrome, which has been reported to prevent and treat various diseases. However, the protective effect of LW on CP-induced reproductive toxicity and genotoxicity has not been reported. AIM OF THE STUDY To investigate the potential protective effect and mechanism of LW on CP-induced reproductive toxicity and genotoxicity in male mice. MATERIALS AND METHODS Mice were given LW (0.4, 1.2 and 3.6 g/kg) or Vitamin C (0.1 g/kg) once daily by oral gavage for thirteen consecutive days. Then, CP (3.00 mg/kg) was given intraperitoneal injection once daily for five consecutive days starting on the ninth day. The protective effects of LW against CP-induced reproductive toxicity and genotoxicity were evaluated by body weight, testis ratio, sperm count, sperm viability, sperm abnormal morphology type, micronuclei test, testicular histopathology, serum malondialdehyde (MDA), total superoxide dismutase (T-SOD), glutathione peroxidase (GSH-Px) and catalase (CAT) level. RESULTS The results demonstrated that LW could significantly increase CP-induced the reduction of sperm count and sperm viability, then decrease abnormal sperm type rate and micronucleus rate. Moreover, LW also could improve testicular abnormal histopathologic morphology induced by CP exposure. Meanwhile, LW decreased serum MDA level and increased T-SOD, GSH-Px and CAT level compared to CP group. CONCLUSION our findings show that LW has protective effects on CP-induced reproductive toxicity and genotoxicity. LW decreases serum MDA level and increases T-SOD, GSH-Px and CAT level, which indicates that antioxidant activity may be the potential mechanism of LW to resist reproductive toxicity and genotoxicity.
Collapse
Affiliation(s)
- Kuo Zhang
- Department of Pharmacology, Shenyang Pharmaceutical University, 110016, Shenyang, PR China
| | - Huili Weng
- Department of Pharmacology, Shenyang Pharmaceutical University, 110016, Shenyang, PR China; Department of Pharmacy, Chengde Maternal and Child Health-Care Hospital, Chengde, China
| | - Jingyu Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, 110016, Shenyang, PR China
| | - Chunfu Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, 110016, Shenyang, PR China.
| |
Collapse
|
12
|
Lu Y, Wu S, Xiang B, Li L, Lin Y. Curcumin Attenuates Oxaliplatin-Induced Liver Injury and Oxidative Stress by Activating the Nrf2 Pathway. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:73-85. [PMID: 32021093 PMCID: PMC6956999 DOI: 10.2147/dddt.s224318] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 12/13/2019] [Indexed: 12/21/2022]
Abstract
Purpose Oxaliplatin (OXA)-induced liver injury is one of the main limiting factors affecting the efficacy of OXA-based chemotherapy in patients with colorectal liver metastases. In addition, oxidative stress is an important pathophysiological mechanism of OXA-induced liver injury. Therefore, dietary antioxidants may decrease or prevent hepatic toxicity in vivo and be beneficial to OXA-based chemotherapy. Methods An experimental OXA-induced liver injury animal model was established, and the protective effects of curcumin (CUR) against OXA-induced liver injury were investigated. ELISA was used to determine the levels of MDA, SOD, CAT, and GSH in liver tissue. The effect of CUR treatment on the expression of cytokines and the Nrf2 pathway was determined by real-time PCR and Western blotting. Results CUR treatment alleviated OXA-induced hepatic pathological damage and splenomegaly. The protective effect of CUR was demonstrated to be correlated with inhibition of oxidative stress, inflammation, and the coagulation system. Furthermore, Western blotting revealed that CUR treatment reverses the suppression of Nrf2 nuclear translocation and increases the expression of HO-1 and NOQ1 in mice with OXA-induced liver injury. Moreover, the Nrf2 activation and hepatoprotective effect of CUR were abolished by brusatol. Conclusion Curcumin attenuates oxaliplatin-induced liver injury and oxidative stress by activating the Nrf2 pathway, which suggests that CUR may be potentially used in the prevention and treatment of OXA-induced liver injury.
Collapse
Affiliation(s)
- Yulei Lu
- Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, People's Republic of China
| | - Shengming Wu
- Departments of Pathology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, People's Republic of China
| | - Bangde Xiang
- Departments of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, People's Republic of China
| | - Lequn Li
- Departments of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, People's Republic of China
| | - Youzhi Lin
- Departments of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, People's Republic of China
| |
Collapse
|
13
|
The mechanisms of pharmacokinetic food-drug interactions - A perspective from the UNGAP group. Eur J Pharm Sci 2019; 134:31-59. [PMID: 30974173 DOI: 10.1016/j.ejps.2019.04.003] [Citation(s) in RCA: 191] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 03/12/2019] [Accepted: 04/02/2019] [Indexed: 02/06/2023]
Abstract
The simultaneous intake of food and drugs can have a strong impact on drug release, absorption, distribution, metabolism and/or elimination and consequently, on the efficacy and safety of pharmacotherapy. As such, food-drug interactions are one of the main challenges in oral drug administration. Whereas pharmacokinetic (PK) food-drug interactions can have a variety of causes, pharmacodynamic (PD) food-drug interactions occur due to specific pharmacological interactions between a drug and particular drinks or food. In recent years, extensive efforts were made to elucidate the mechanisms that drive pharmacokinetic food-drug interactions. Their occurrence depends mainly on the properties of the drug substance, the formulation and a multitude of physiological factors. Every intake of food or drink changes the physiological conditions in the human gastrointestinal tract. Therefore, a precise understanding of how different foods and drinks affect the processes of drug absorption, distribution, metabolism and/or elimination as well as formulation performance is important in order to be able to predict and avoid such interactions. Furthermore, it must be considered that beverages such as milk, grapefruit juice and alcohol can also lead to specific food-drug interactions. In this regard, the growing use of food supplements and functional food requires urgent attention in oral pharmacotherapy. Recently, a new consortium in Understanding Gastrointestinal Absorption-related Processes (UNGAP) was established through COST, a funding organisation of the European Union supporting translational research across Europe. In this review of the UNGAP Working group "Food-Drug Interface", the different mechanisms that can lead to pharmacokinetic food-drug interactions are discussed and summarised from different expert perspectives.
Collapse
|
14
|
Mortezaee K, Salehi E, Mirtavoos-Mahyari H, Motevaseli E, Najafi M, Farhood B, Rosengren RJ, Sahebkar A. Mechanisms of apoptosis modulation by curcumin: Implications for cancer therapy. J Cell Physiol 2019; 234:12537-12550. [PMID: 30623450 DOI: 10.1002/jcp.28122] [Citation(s) in RCA: 202] [Impact Index Per Article: 40.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 12/21/2018] [Indexed: 12/12/2022]
Abstract
Cancer incidences are growing and cause millions of deaths worldwide. Cancer therapy is one of the most important challenges in medicine. Improving therapeutic outcomes from cancer therapy is necessary for increasing patients' survival and quality of life. Adjuvant therapy using various types of antibodies or immunomodulatory agents has suggested modulating tumor response. Resistance to apoptosis is the main reason for radioresistance and chemoresistance of most of the cancers, and also one of the pivotal targets for improving cancer therapy is the modulation of apoptosis signaling pathways. Apoptosis can be induced by intrinsic or extrinsic pathways via stimulation of several targets, such as membrane receptors of tumor necrosis factor-α and transforming growth factor-β, and also mitochondria. Curcumin is a naturally derived agent that induces apoptosis in a variety of different tumor cell lines. Curcumin also activates redox reactions within cells inducing reactive oxygen species (ROS) production that leads to the upregulation of apoptosis receptors on the tumor cell membrane. Curcumin can also upregulate the expression and activity of p53 that inhibits tumor cell proliferation and increases apoptosis. Furthermore, curcumin has a potent inhibitory effect on the activity of NF-κB and COX-2, which are involved in the overexpression of antiapoptosis genes such as Bcl-2. It can also attenuate the regulation of antiapoptosis PI3K signaling and increase the expression of MAPKs to induce endogenous production of ROS. In this paper, we aimed to review the molecular mechanisms of curcumin-induced apoptosis in cancer cells. This action of curcumin could be applicable for use as an adjuvant in combination with other modalities of cancer therapy including radiotherapy and chemotherapy.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Ensieh Salehi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hanifeh Mirtavoos-Mahyari
- Department of Medical Genetics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Elahe Motevaseli
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Science, Kermanshah, Iran
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Rhonda J Rosengren
- Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
15
|
Farkhondeh T, Samarghandian S, Azimi-Nezhad M, Shahri AMP. Protective Effects of Curcumin Against Nephrotoxic Agents. Cardiovasc Hematol Disord Drug Targets 2019; 19:176-182. [PMID: 30205807 DOI: 10.2174/1871529x18666180905160830] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 08/08/2018] [Accepted: 08/28/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND Curcumin is the one of the main phenolic ingredients in curcuma species rhizome. Curcuma species have traditionally been used for the treatment of diabetes, cardiovascular, and renal diseases. METHODS The present study was designed to review the scientific literature on the protective effects of curcumin against nephrotoxic agents. RESULTS Studies have shown the protective effects of curcumin against nephrotoxic agents such as gallic acid, glucose, tartrazine, streptozotocin, lead, cadmium, fluoride, maleate, malathion, nicotine, cisplatin, gentamicin, and methotrexate. However, further investigations are needed to determine the efficacy of curcumin as an antidote agent due to the lack of clinical trial studies. Therefore, it is recommended to conduct clinical trials in humans to confirm these effects. CONCLUSION The current review indicated that curcumin may be effective against nephrotoxicity by modulating oxidative stress and inflammatory responses.
Collapse
Affiliation(s)
- Tahereh Farkhondeh
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Saeed Samarghandian
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Mohsen Azimi-Nezhad
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran
- Department of Medical Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali M P Shahri
- Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| |
Collapse
|
16
|
Liu Z, Huang P, Law S, Tian H, Leung W, Xu C. Preventive Effect of Curcumin Against Chemotherapy-Induced Side-Effects. Front Pharmacol 2018; 9:1374. [PMID: 30538634 PMCID: PMC6277549 DOI: 10.3389/fphar.2018.01374] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Accepted: 11/08/2018] [Indexed: 12/29/2022] Open
Abstract
Cancer is still a severe threat to the health of people worldwide. Chemotherapy is one of main therapeutic approaches to combat cancer. However, chemotherapy only has a limited success with severe side effects, especially causing damage to normal tissues such as bone marrow, gastrointestine, heart, liver, renal, neuron, and auditory tissues, etc. The side-effects limit clinical outcome of chemotherapy and lower patients’ quality of life, and even make many patients discontinue the chemotherapy. Thus, there is a need to explore effective adjuvant strategies to prevent and reduce the chemotherapy-induced side effects. Naturally occurring products provide a rich source for exploring effective adjuvant agents to prevent and reduce the side effects in anticancer chemotherapy. Curcumin is an active compound from natural plant Curcuma longa L., which is widely used as a coloring and flavoring agent in food industry and a herbal medicine in Asian countries for thousands of years to treat vomiting, headache, diarrhea, etc. Modern pharmacological studies have revealed that curcumin has strong antioxidative, anti-microbial, anti-inflammatory and anticancer activities. Growing evidence shows that curcumin is able to prevent carcinogenesis, sensitize cancer cells to chemotherapy, and protect normal cells from chemotherapy-induced damages. In the present article, we review the preventive effect of curcumin against chemotherapy-induced myelosuppression, gastrointestinal toxicity, cardiotoxicity, hepatotoxicity, nephrotoxicity, neurotoxicity, ototoxicity, and genotoxicity, and discuss its action mechanisms.
Collapse
Affiliation(s)
- Zhijun Liu
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Pengyun Huang
- Faculty of Medicine, School of Chinese Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, Shenzhen, China
| | - Siukan Law
- Faculty of Medicine, School of Chinese Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, Shenzhen, China
| | - Haiyan Tian
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou, China
| | - Wingnang Leung
- Division of Chinese Medicine, School of Professional and Continuing Education, The University of Hong Kong, Pokfulam, Hong Kong
| | - Chuanshan Xu
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,Faculty of Medicine, School of Chinese Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, Shenzhen, China
| |
Collapse
|
17
|
Yilmaz E, Melekoglu R, Ciftci O, Eraslan S, Cetin A, Basak N. The therapeutic effects of curcumin and capsaicin against cyclophosphamide side effects on the uterus in rats. Acta Cir Bras 2018; 33:499-507. [DOI: 10.1590/s0102-865020180060000004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 05/23/2018] [Indexed: 02/06/2023] Open
|