1
|
Satyam SM, Bairy LK, Rehman A, Farook M, Khan S, Nair AA, Binu NN, Yehya M, Khan MM. Dapagliflozin: A Promising Strategy to Combat Cisplatin-Induced Hepatotoxicity in Wistar Rats. BIOLOGY 2024; 13:672. [PMID: 39336099 PMCID: PMC11428795 DOI: 10.3390/biology13090672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/23/2024] [Accepted: 08/28/2024] [Indexed: 09/30/2024]
Abstract
Recognizing the challenges posed by chemotherapy, specifically the hepatotoxic effects of drugs like cisplatin, this study aimed to examine the hepatoprotective potential of dapagliflozin to mitigate cisplatin-induced hepatotoxicity in a rat model. This study focused on repurposing drugs such as dapagliflozin and natural agents like silymarin as potential interventions to address cisplatin-induced hepatotoxicity. Thirty adult female Wistar rats were distributed into five groups and treated with cisplatin alone, silymarin, dapagliflozin, or a combination of dapagliflozin and silymarin accordingly for 45 days. Body weight, fasting blood glucose levels, liver function tests, and histopathological analysis were conducted to evaluate the hepatoprotective effects. Cisplatin-induced hepatotoxicity significantly (p < 0.05) increased the serum levels of ALT, AST, TB, and reduced the TP and albumin levels. Dapagliflozin administration led to significant reductions in ALT, AST, TB, and increased albumin levels. Silymarin demonstrated comparable effects. Combining dapagliflozin and silymarin showed synergistic effects, further reducing the liver enzymes and improving albumin levels. Histopathological examination supported these findings, revealing the restoration of liver structure with dapagliflozin and silymarin treatment. Dapagliflozin and silymarin exhibited substantial hepatoprotective benefits against cisplatin-induced hepatotoxicity in rats. The combination therapy demonstrated synergistic effects, highlighting a potential therapeutic approach for mitigating chemotherapy-induced liver damage. Further research into molecular mechanisms and clinical translation is warranted, offering hope for improved clinical outcomes in cancer patients undergoing cisplatin-based chemotherapy.
Collapse
Affiliation(s)
- Shakta Mani Satyam
- Faculty of Pharmacology, RAK College of Medical Sciences, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
| | - Laxminarayana Kurady Bairy
- Faculty of Pharmacology, RAK College of Medical Sciences, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
| | - Abdul Rehman
- Faculty of Pathology, RAK College of Medical Sciences, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
| | - Mohamed Farook
- RAK College of Medical Sciences, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
| | - Sofiya Khan
- RAK College of Medical Sciences, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
| | - Anuradha Asokan Nair
- RAK College of Medical Sciences, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
| | - Nirmal Nachiketh Binu
- RAK College of Medical Sciences, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
| | - Mohamed Yehya
- RAK College of Medical Sciences, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
| | - Mohammed Moin Khan
- RAK College of Medical Sciences, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
| |
Collapse
|
2
|
Yadav V, Fuentes JL, Krishnan A, Singh N, Vohora D. Guidance for the use and interpretation of assays for monitoring anti-genotoxicity. Life Sci 2024; 337:122341. [PMID: 38101613 DOI: 10.1016/j.lfs.2023.122341] [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: 08/03/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023]
Abstract
Since DNA damage can occur spontaneously or be produced by the environmental genotoxins in living cells, it is important to investigate compounds that can reverse or protect DNA damage. An appropriate methodology is essential for the responsive identification of protection offered against DNA damage. This review includes information on the current state of knowledge on prokaryotic cell-based assays (SOS chromotest, umu test, vitotox assay) and cytogenetic techniques (micronucleus assay, chromosome aberration test and sister chromatid exchange assay) with an emphasis on the possibility to explore genoprotective compounds. Throughout the last decade, studies have extrapolated the scientific methodologies utilized for genotoxicity to assess genoprotective compounds. Therefore, shortcomings of genotoxicity studies are also mirrored in antigenotoxicity studies. While regulatory authorities around the world (OECD, US-EPA and ICH) continue to update diverse genotoxic assay strategies, there are still no clear guidelines/approaches for efficient experimental design to screen genoprotective compounds. As a consequence, non-synergetic and inconsistent implementation of the test method by the researchers to execute such simulations has been adopted, which inevitably results in unreliable findings. The review has made the first attempt to collect various facets of experimentally verified approaches for evaluating genoprotective compounds, as well as to acknowledge potential significance and constraints, and further focus on the assessment of end points which are required to validate such action. Henceforth, the review makes an incredible commitment by permitting readers to equate several components of their test arrangement with the provided simplified information, allowing the selection of convenient technique for the predefined compound from a central repository.
Collapse
Affiliation(s)
- Vaishali Yadav
- Neurobehavioral Pharmacology Laboratory, Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Jorge L Fuentes
- School of Biology, Science Faculty, Industrial University of Santander, Bucaramanga 680002, Santander, Colombia
| | - Anuja Krishnan
- Department of Molecular Medicine, School of Interdisciplinary Science and Technology, Jamia Hamdard, New Delhi 110062, India
| | - Neenu Singh
- Leicester School of Allied Health Sciences, Faculty of Health & Life Sciences, De Montfort University, The Gateway, Leicester LE1 9BH, UK
| | - Divya Vohora
- Neurobehavioral Pharmacology Laboratory, Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
3
|
Al-Bataineh WM, Alzoubi KH, Khabour OF, Mahasneh A, Al Momany EM. Vitamin B12 Protects against Genotoxicity Induced by Cisplatin. Curr Cancer Drug Targets 2024; 24:1169-1176. [PMID: 38299397 DOI: 10.2174/0115680096284684240110044954] [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: 09/28/2023] [Revised: 11/18/2023] [Accepted: 11/29/2023] [Indexed: 02/02/2024]
Abstract
BACKGROUND Cisplatin is an effective synthetic chemotherapeutic drug used for cancer treatment. Vitamin B12 has been shown to possess anti-genotoxic activity. This study aimed to investigate the effect of vitamin B12 on chromosomal damage induced by cisplatin. METHODS The level of sister chromatid exchanges (SCEs) and chromosomal aberrations (CAs) were measured in cultured human blood lymphocytes treated with cisplatin and/or vitamin B12. RESULTS The results showed a significantly elevated frequency of CAs and SCEs of cisplatin-treated cultures compared to the control (P < 0.05). The CAs and SCEs induced by cisplatin were significantly lowered by pretreatment of cell cultures with vitamin B12. In addition, cisplatin caused a slight reduction in the mitotic index (MI), while vitamin B12 did not modulate the effect of cisplatin on MI. CONCLUSION Vitamin B12 can protect human lymphocytes against genotoxicity associated with cisplatin.
Collapse
Affiliation(s)
- Wejdan M Al-Bataineh
- Department of Applied Biological Sciences, Faculty of Science and Art, Jordan University of Science and Technology, Irbid, Jordan
| | - Karem H Alzoubi
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, UAE
- Department of Clinical Pharmacy, Faculty 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, Jordan
| | - Amjad Mahasneh
- Department of Applied Biological Sciences, Faculty of Science and Art, Jordan University of Science and Technology, Irbid, Jordan
| | - Enaam M Al Momany
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmaceutical Sciences, The Hashemite University, P.O. box 330127, Zarqa 13133, Jordan
| |
Collapse
|
4
|
Alzoubi KH, Halboup AM, Khabour OF, Alomari MA. The Protective Effects of the Combination of Vitamin E and Swimming Exercise on Memory Impairment Induced by Exposure to Waterpipe Smoke. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2023; 22:304-312. [PMID: 35306997 DOI: 10.2174/1871527321666220318113635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/12/2021] [Accepted: 12/29/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Waterpipe smoking (WP) exposure involves a negative health impact, including memory deficit, which is attributed to the elevation of oxidative stress. Vitamin E (VitE) in combination with swimming exercise exerts protective effects that prevent memory impairment. In the current study, the modulation of WP-induced memory impairment by the combined effect of VitE and swimming exercise (SE) was investigated. METHODS Animals were exposed to WP one hour/day, five days per week for four weeks. Simultaneously, VitE (100 mg/kg, six days/week for four weeks) was administered via oral gavage, and the rats were made to swim one hour/day, five days/week for four weeks. Changes in memory were evaluated using radial arm water maze (RAWM), and oxidative stress biomarkers were examined in the hippocampus. RESULTS WP exposure induced short-term/long-term memory impairment (p<0.05). This impairment was prevented by a combination of VitE with SE (p<0.05). Additionally, this combination normalized the hippocampal catalase, GPx, and GSH/GSSG ratios that were modulated by WP (p<0.05). The combination further reduced TBARs levels below those of the control group (p<0.05). CONCLUSION WP-induced memory impairments were prevented by the combination of VitE with SE. This could be attributed to preserving the hippocampal oxidative mechanism by combining VitE and SE during WP exposure.
Collapse
Affiliation(s)
- Karem H Alzoubi
- Department of Pharmacy Practice and Pharmacotherapeutics, University of Sharjah, Sharjah, UAE
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Abdulsalam M Halboup
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, University of Science and Technology, Sana\'a, Yemen
| | - Omar F Khabour
- Department of Medical Laboratory Sciences, Jordan University of Science and Technology, Jordan
| | - Mahmoud A Alomari
- Department of Rehabilitation Sciences, Division of Physical Therapy, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
5
|
Al-Khdour MS, Khabour OF, Al-Eitan LN, Alzoubi KH. Genotoxicity of nedaplatin in cultured lymphocytes: modulation by vitamin E. Drug Chem Toxicol 2023; 46:176-180. [PMID: 34965829 DOI: 10.1080/01480545.2021.2015369] [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: 02/02/2023]
Abstract
Nedaplatin is a chemotherapeutic agent used widely in cancer therapy. Nedaplatin has been shown to cause DNA damage to cells via the induction of oxidative stress. Vitamin E (Vit E) has an anti-mutagenic activity that can protect cells from DNA damaging agents. The objective of this study is to examine the genotoxic and cytotoxic effects of nedaplatin in human cultured lymphocytes. In addition, modulation of such effects by Vit E was also examined. The frequencies of sister chromatid exchange (SCE) and chromosomal aberrations (CAs) were used as an indicator for genotoxicity. The mitotic and proliferative indices were used to examine the cytotoxic effects of nedaplatin. The results showed that nedaplatin significantly elevated SCE and CA frequencies in human lymphocytes (p ˂ 0.01). The increases in the frequencies of SCE and CA caused by nedaplatin were lowered by pretreatment treatment with Vit E (p < 0.05). Nedaplatin significantly lowered mitotic index but Vit E pretreatment did not modulate this effect. These results suggest that Vit E has the potential to ameliorate the genotoxicity of nedaplatin in cultured lymphocytes.
Collapse
Affiliation(s)
- Muntaha S Al-Khdour
- Department of Applied Biological Sciences, Faculty of Science, Jordan University of Science and Technology, Irbid, Jordan
| | - Omar F Khabour
- Department of Medical Laboratory Sciences, Faculty of Medical Sciences, Jordan University of Science and Technology, Irbid, Jordan
| | - Laith N Al-Eitan
- Department of Applied Biological Sciences, Faculty of Science, Jordan University of Science and Technology, Irbid, Jordan
| | - Karem H Alzoubi
- Department of Pharmacy Practice and Pharmacotherapeutics, University of Sharjah, Sharjah, United Arab Emirates.,Department of Clinical Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
6
|
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
|
7
|
Gilani SJ, Bin-Jumah MN, Al-Abbasi FA, Nadeem MS, Alzarea SI, Ahmed MM, Sayyed N, Kazmi I. Rosinidin Protects against Cisplatin-Induced Nephrotoxicity via Subsiding Proinflammatory and Oxidative Stress Biomarkers in Rats. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:9719. [PMID: 35955076 PMCID: PMC9368304 DOI: 10.3390/ijerph19159719] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/30/2022] [Accepted: 07/31/2022] [Indexed: 05/28/2023]
Abstract
BACKGROUND Rosinidin is a flavonoid anthocyanin pigmentation found in shrub flowers such as Catharanthus roseus and Primula rosea. The molecular docking studies predicted that rosinidin has adequate structural competency, making it a viable medicinal candidate for the treatment of a wide range of disorders. The current study intends to assess rosinidin nephroprotective efficacy against nephrotoxicity induced by cisplatin in rats. MATERIALS AND METHODS Oral acute toxicity tests of rosinidin were conducted to assess potential toxicity in animals, and it was shown to be safe. The nephroprotective effect of rosinidin 10, and 20 mg/kg were tested in rats for 25 days with concurrent administration of cisplatin. Several biochemical parameters were measured to support enzymatic and non-enzymatic oxidative stress such as superoxide dismutase (SOD), malondialdehyde (MDA), and glutathione peroxidase (GSH). Likewise, changes in several non-protein-nitrogenous components and blood chemistry parameters were made to support the theory linked with the pathogenesis of chemical-induced nephrotoxicity. RESULTS Cisplatin caused significant changes in biochemical, enzymatic, and blood chemistry, which rosinidin efficiently controlled. CONCLUSIONS The present investigation linked rosinidin with nephroprotective efficacy in experimental models.
Collapse
Affiliation(s)
- Sadaf Jamal Gilani
- Department of Basic Health Sciences, Preparatory Year, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia
| | - May Nasser Bin-Jumah
- Biology Department, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia
- Environment and Biomaterial Unit, Health Sciences Research Center, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia
- Saudi Society for Applied Science, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia
| | - Fahad A. Al-Abbasi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Muhammad Shahid Nadeem
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Sami I. Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka 72341, Saudi Arabia
| | - Mohammed Muqtader Ahmed
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Nadeem Sayyed
- Glocal School of Pharmacy, Glocal University, Saharanpur 247121, India
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
8
|
Fathy M, Darwish MA, Abdelhamid ASM, Alrashedy GM, Othman OA, Naseem M, Dandekar T, Othman EM. Kinetin Ameliorates Cisplatin-Induced Hepatotoxicity and Lymphotoxicity via Attenuating Oxidative Damage, Cell Apoptosis and Inflammation in Rats. Biomedicines 2022; 10:biomedicines10071620. [PMID: 35884925 PMCID: PMC9312964 DOI: 10.3390/biomedicines10071620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 11/16/2022] Open
Abstract
Though several previous studies reported the in vitro and in vivo antioxidant effect of kinetin (Kn), details on its action in cisplatin-induced toxicity are still scarce. In this study we evaluated, for the first time, the effects of kinetin in cisplatin (cp)- induced liver and lymphocyte toxicity in rats. Wistar male albino rats were divided into nine groups: (i) the control (C), (ii) groups 2,3 and 4, which received 0.25, 0.5 and 1 mg/kg kinetin for 10 days; (iii) the cisplatin (cp) group, which received a single intraperitoneal injection of CP (7.0 mg/kg); and (iv) groups 6, 7, 8 and 9, which received, for 10 days, 0.25, 0.5 and 1 mg/kg kinetin or 200 mg/kg vitamin C, respectively, and Cp on the fourth day. CP-injected rats showed a significant impairment in biochemical, oxidative stress and inflammatory parameters in hepatic tissue and lymphocytes. PCR showed a profound increase in caspase-3, and a significant decline in AKT gene expression. Intriguingly, Kn treatment restored the biochemical, redox status and inflammatory parameters. Hepatic AKT and caspase-3 expression as well as CD95 levels in lymphocytes were also restored. In conclusion, Kn mitigated oxidative imbalance, inflammation and apoptosis in CP-induced liver and lymphocyte toxicity; therefore, it can be considered as a promising therapy.
Collapse
Affiliation(s)
- Moustafa Fathy
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia 61519, Egypt;
| | - Mostafa A. Darwish
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Nahda University, Beni-Suef 62521, Egypt;
| | - Al-Shaimaa M. Abdelhamid
- Department of Chemistry, Biochemistry Division, Faculty of Science, Minia University, Minia 61519, Egypt; (A.-S.M.A.); (G.M.A.); (O.A.O.)
| | - Gehad M. Alrashedy
- Department of Chemistry, Biochemistry Division, Faculty of Science, Minia University, Minia 61519, Egypt; (A.-S.M.A.); (G.M.A.); (O.A.O.)
| | - Othman Ali Othman
- Department of Chemistry, Biochemistry Division, Faculty of Science, Minia University, Minia 61519, Egypt; (A.-S.M.A.); (G.M.A.); (O.A.O.)
| | - Muhammad Naseem
- Department of Life and Environmental Sciences, College of Natural and Health Sciences, Zayed University, Abu Dhabi 144534, United Arab Emirates;
- Department of Bioinformatics, Biocenter, University of Wuerzburg, Am Hubland, 97074 Wuerzburg, Germany
| | - Thomas Dandekar
- Department of Bioinformatics, Biocenter, University of Wuerzburg, Am Hubland, 97074 Wuerzburg, Germany
- Correspondence: (T.D.); (E.M.O.)
| | - Eman M. Othman
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia 61519, Egypt;
- Department of Bioinformatics, Biocenter, University of Wuerzburg, Am Hubland, 97074 Wuerzburg, Germany
- Correspondence: (T.D.); (E.M.O.)
| |
Collapse
|
9
|
Circ-PKD2 promotes Atg13-mediated autophagy by inhibiting miR-646 to increase the sensitivity of cisplatin in oral squamous cell carcinomas. Cell Death Dis 2022; 13:192. [PMID: 35220397 PMCID: PMC8882170 DOI: 10.1038/s41419-021-04497-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 11/03/2021] [Accepted: 12/20/2021] [Indexed: 12/16/2022]
Abstract
Autophagy is an evolutionally conserved catabolic process that degrades cells to maintain homeostasis. Cisplatin-activated autophagy promotes the expression of circ-PKD2, which plays a role as a tumor suppressor gene in the proliferation, migration, and invasion in oral squamous cell carcinoma (OSCC). However, the role of circ-PKD2 in regulating the sensitivity of OSCC patients to cisplatin remains to be elucidated. Overexpression of circ-PKD2 increased the formation of autophagosomes in OSCC cells and activation of proteins, such as LC3 II/I. Its activation effect on autophagy was, however, alleviated by 3-MA. Bioinformatics analyses and double luciferases reporter assays conducted in this study confirmed the existence of targeted relationships between circ-PKD2 and miR-646 and miR-646 and Atg13. Functional experiments further revealed that miR-646 reversed the autophagy and apoptosis effects of circ-PKD2 in OSCC cells treated with cisplatin. In addition, circ-PKD2 promoted the expression of ATG13 by adsorption of miR-646. Its interference with Atg13 alleviated the activation effects of circ-PKD2 on autophagy and apoptosis of miR-646. Notably, the in vivo animal experiments also confirmed that circ-PKD2 inhibited tumor proliferation and activated autophagy in OSCC cells. This study provides a theoretical basis for using circ-PKD2 as a target to regulate the sensitivity of OSCC patients to cisplatin, thus increasing its chemotherapeutic effects.
Collapse
|
10
|
Eslamifar Z, Moridnia A, Sabbagh S, Ghaffaripour R, Jafaripour L, Behzadifard M. Ameliorative Effects of Gallic Acid on Cisplatin-Induced Nephrotoxicity in Rat Variations of Biochemistry, Histopathology, and Gene Expression. BIOMED RESEARCH INTERNATIONAL 2021; 2021:2195238. [PMID: 34746299 PMCID: PMC8564201 DOI: 10.1155/2021/2195238] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/18/2021] [Accepted: 09/21/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND Cisplatin is a powerful chemotherapeutic drug mainly used in the treatment of solid tumors. Aggregation of the drug in renal proximal tubule cells causes nephrotoxicity and renal failure. Investigations showed nephrotoxicity as Cisplatin's dose-limiting side effect. One of the Cisplatin toxicity mechanisms is generation of reactive oxygen species, which leads to oxidative stress and renal damage. The purpose of this study was evaluation of the modulating effects of Gallic acid on Cisplatin-induced variations including Caspase-3 and Clusterin expression and histopathological and biochemical parameters in adult male Wistar rats. METHOD Rats were kept under standard condition of temperature, light, and humidity. The animals were divided into 4 groups: GpI: control group (received distilled water for 10 days); GpII: Gallic acid (alone) (50 mg/kg bw, once a day for 10 days); GpIII: Cisplatin (alone), single dose (6 mg/kg bw, I.P. on 5th day of study); GpIV: Gallic acid (50 mg/kg bw, once a day for 10 days) and also injected with single dose of Cisplatin (6 mg/kg bw, I.P., on 5th day of study). After 10 days, all rats were anaesthetized and plasma collected to estimate urea, creatinine, and uric acid. The right kidneys were removed for the study of gene expression and biochemical parameters. The left kidneys were used for histopathological studies. RESULTS The Cisplatin-induced nephrotoxicity was evident from the elevated levels of creatinine, urea, uric acid, and renal tissue MDA and also decreased levels of SOD, CAT, GPX, and GSH in renal tissue. Administration of Gallic acid significantly modulated nephrotoxicity markers, gene expression variations, and histopathological damage. CONCLUSION Outcomes of the present investigation suggest that Gallic acid provides protection against CP-induced nephrotoxicity, but for application in people, further studies are needed.
Collapse
Affiliation(s)
- Zahra Eslamifar
- Department of Medical Laboratory Sciences, School of Paramedical Sciences, Dezful University of Medical Sciences, Dezful, Iran
| | - Abbas Moridnia
- Department of Immunology, School of Medicine, Dezful University of Medical Sciences, Dezful, Iran
| | - Susan Sabbagh
- Department of Anatomy, Faculty of Medicine, Dezful University of Medical Sciences, Dezful, Iran
| | - Reza Ghaffaripour
- Department of Medical Laboratory Sciences, School of Paramedical Sciences, Dezful University of Medical Sciences, Dezful, Iran
| | - Leila Jafaripour
- Department of Anatomy, Faculty of Medicine, Dezful University of Medical Sciences, Dezful, Iran
| | - Mahin Behzadifard
- Department of Medical Laboratory Sciences, School of Paramedical Sciences, Dezful University of Medical Sciences, Dezful, Iran
| |
Collapse
|
11
|
Kocaman AY, Asfuroğlu K. The genotoxic effects of perchloroethylene in human peripheral blood lymphocytes and the possible ameliorative role of α-tocopherol. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:39576-39586. [PMID: 33763835 DOI: 10.1007/s11356-021-13523-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/15/2021] [Indexed: 06/12/2023]
Abstract
Perchloroethylene (PCE), also known as tetrachloroethylene, is a commercially important chlorinated solvent commonly used in dry cleaning, textile processing, and metal degreasing industries. According to the available studies, the potential genotoxic impacts of this chlorinated solvent on human beings are still controversial. The present work was carried out to determine the genotoxic effects of PCE on human peripheral blood lymphocytes (PBLs) using chromosome aberrations (CAs) and cytokinesis-block micronucleus (CBMN) tests. Additionally, the antigenotoxic potential of α-tocopherol (α-Toc), a well-known antioxidant agent, on human lymphocytes treated with PCE in vitro was assessed. The cells were exposed for 48 h to PCE (25, 50, 100, and 150 μg/mL) alone as well as in combination with α-Toc (100 μg/mL). The findings of the study suggested that, relative to solvent control, PCE significantly increased the structural CA and MN formation for all concentrations. However, simultaneous treatment of PCE and α-Toc caused a significant reduction of CAs and MNi as compared to cultures treated with PCE alone. Besides, the results showed that PCE has cytotoxic effects on human PBLs as indicated by the significant decrease in mitotic index (MI) and nuclear division index (NDI). Nevertheless, the co-treatment of α-Toc with PCE did not reduce the cytotoxicity of PCE at a significant level. In conclusion, it can be suggested that PCE is genotoxic and cytotoxic in human PBLs, and α-Toc has an antigenotoxic effect on PCE-induced genotoxicity but has no significant effect on the cytotoxicity triggered by PCE.
Collapse
Affiliation(s)
- Ayşe Yavuz Kocaman
- Department of Biology, Faculty of Sciences and Letters, Hatay Mustafa Kemal University, 31000, Antakya, Hatay, Turkey.
| | - Kübra Asfuroğlu
- Basic and Applied Sciences Institute, Hatay Mustafa Kemal University, Antakya, Hatay, Turkey
| |
Collapse
|
12
|
Yarmohammadi F, Hayes AW, Karimi G. Protective effects of curcumin on chemical and drug-induced cardiotoxicity: a review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2021; 394:1341-1353. [PMID: 33666716 DOI: 10.1007/s00210-021-02072-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 02/18/2021] [Indexed: 12/15/2022]
Abstract
Cardiotoxicity is a major adverse effect that can be induced by both therapeutic agents and industrial chemicals. The pathogenesis of such cardiac damage is multifactorial, often injuring the cardiac tissue by generating free radicals, oxidative stress, and/or inflammation. Curcumin (CUR) is a bright yellow chemical produced by Curcuma longa plants. It is the principal curcuminoid of turmeric (Curcuma longa), a member of the ginger family, Zingiberaceae. Administration of CUR has been reported to ameliorate the chemical and drug-induced cardiac injury in several studies. CUR has been suggested to act as an effective candidate against oxidative stress and inflammation in heart tissue via regulation of Nrf2 and suppression of p38 MAPK/NF-κB and NLRP3 inflammasomes. The anti-apoptotic properties of CUR have also been reported to modulate the AMPK, Akt, JNK, and ERK signaling pathways. This review explores the potential protective effects of CUR regarding the detrimental effects often observed in cardiac tissue following exposure to several chemicals including drugs.
Collapse
Affiliation(s)
- Fatemeh Yarmohammadi
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - A Wallace Hayes
- Center for Environmental Occupational Risk Analysis and Management, College of Public Health, University of South Florida, Tampa, FL, 33617, USA
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
13
|
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
|
14
|
Sanz-Serrano J, Vettorazzi A, Muruzabal D, López de Cerain A, Azqueta A. In vitro genotoxicity assessment of functional ingredients: DHA, rutin and α-tocopherol. Food Chem Toxicol 2021; 153:112237. [PMID: 33894296 DOI: 10.1016/j.fct.2021.112237] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/31/2021] [Accepted: 04/16/2021] [Indexed: 11/26/2022]
Abstract
The in vitro genotoxicity of three compounds widely used as functional ingredients, docosahexaenoic acid (DHA), rutin and α-tocopherol, was assessed. A miniaturized version of the Ames test in Salmonella typhimurium TA97a, TA98, TA100, TA102, and TA1535 strains (following the principles of OECD 471), and the in vitro micronucleus test in TK6 cells (OECD 487) were performed. This strategy is recommended by the European Food Safety Authority for the in vitro genotoxicity assessment of food and feed. In addition, this approach was complemented with the in vitro standard and enzyme-modified comet assay (S9-/S9+) using hOGG1, EndoIII and hAAG in order to assess potential premutagenic lesions in TK6 cells. Rutin showed an equivocal response in the in vitro micronucleus test and also was a potent Salmonella typhimurium revertant inductor in the Ames test. DHA showed equivocal results in the in vitro micronucleus test. In this regard, DHA and rutin seemed to interact with the DNA at a chromosomal level, but rutin is also capable of producing frameshift mutations. No genotoxicity was observed in cells treated with α-tocopherol. This article complements the evidence already available about the genotoxicity of these compounds. However, more studies are needed in order to elucidate the consequences of their use as functional ingredients in human health.
Collapse
Affiliation(s)
- Julen Sanz-Serrano
- Universidad de Navarra, School of Pharmacy and Nutrition, Department of Pharmacology and Toxicology, Irunlarrea 1, 31008, Pamplona, Spain
| | - Ariane Vettorazzi
- Universidad de Navarra, School of Pharmacy and Nutrition, Department of Pharmacology and Toxicology, Irunlarrea 1, 31008, Pamplona, Spain; IdiSNA, Navarra Institute for Health Research, Irunlarrea 3, 31008, Pamplona, Spain
| | - Damian Muruzabal
- Universidad de Navarra, School of Pharmacy and Nutrition, Department of Pharmacology and Toxicology, Irunlarrea 1, 31008, Pamplona, Spain
| | - Adela López de Cerain
- Universidad de Navarra, School of Pharmacy and Nutrition, Department of Pharmacology and Toxicology, Irunlarrea 1, 31008, Pamplona, Spain; IdiSNA, Navarra Institute for Health Research, Irunlarrea 3, 31008, Pamplona, Spain.
| | - Amaya Azqueta
- Universidad de Navarra, School of Pharmacy and Nutrition, Department of Pharmacology and Toxicology, Irunlarrea 1, 31008, Pamplona, Spain; IdiSNA, Navarra Institute for Health Research, Irunlarrea 3, 31008, Pamplona, Spain
| |
Collapse
|
15
|
Halevas E, Mitrakas A, Mavroidi B, Athanasiou D, Gkika P, Antoniou K, Samaras G, Lialiaris E, Hatzidimitriou A, Pantazaki A, Koukourakis M, Sagnou M, Pelecanou M, Lialiaris T. Structurally characterized copper-chrysin complexes display genotoxic and cytotoxic activity in human cells. Inorganica Chim Acta 2021. [DOI: 10.1016/j.ica.2020.120062] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
16
|
Simultaneous Determination of Vitamin E and Vitamin K in Food Supplements Using Adsorptive Stripping Square-Wave Voltammetry at Glassy Carbon Electrode. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10144759] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
A new voltammetric method for the simultaneous determination of vitamin E and vitamin K present in different types of commercially available food supplements has been developed. This electroanalytical method is based on the ex situ adsorptive accumulation of these biologically active compounds onto the surface of a solid glassy carbon electrode (GCE) with subsequent electrochemical detection by square-wave adsorptive stripping voltammetry in 0.01-mol L−1 HNO3 containing 0.1-mol L−1 KCl at pH 2.08. Due to reversible electrochemical reactions of phylloquinone, a subsequent voltammetric detection of both vitamins in anodic mode can be performed. Since individual forms of vitamins E and K usually exhibit nearly identical electrochemical behavior, it is therefore impossible to distinguish individual forms (quinones and tocopherols) and determine their molar concentrations in this way. Thus, the values of vitamin content were expressed as mass equivalent of phylloquinone and α-tocopherol as they are the most biologically active forms. Despite the high sensitivity, relatively short linear ranges were obtained due to the interaction (competition) of both vitamins during adsorption onto the freshly polished surface of the GCE from a 50% aqueous–acetonitrile mixture. The obtained results showed that the voltammetric approach is a very simple and low-cost analytical method that can be used in analyses of food supplements.
Collapse
|
17
|
Scagliarini A, Mathey A, Aires V, Delmas D. Xanthohumol, a Prenylated Flavonoid from Hops, Induces DNA Damages in Colorectal Cancer Cells and Sensitizes SW480 Cells to the SN38 Chemotherapeutic Agent. Cells 2020; 9:E932. [PMID: 32290112 PMCID: PMC7226974 DOI: 10.3390/cells9040932] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/26/2020] [Accepted: 04/07/2020] [Indexed: 12/21/2022] Open
Abstract
In spite of chemotherapy and systematic screening for people at risk, the mortality rate of colorectal cancer (CRC) remains consistently high, with 600,000 deaths per year. This low success rate in the treatment of CRC results from many failures associated with high resistance and the risk of metastasis. Therefore, in response to these therapeutic failures, new strategies have been under development for several years aimed at increasing the effect of anticancer compounds and/or at reducing their secondary effects on normal cells, thus enabling the host to better withstand chemotherapy. This study highlights that xanthohumol (Xn) concentrations under the IC50 values were able to induce apoptosis and to enhance the DNA-damage response (DDR). We demonstrate for the first time that Xn exerts its anticancer activity in models of colon cancer through activation of the ataxia telangiectasia mutated (ATM) pathway. Subsequently, the ability of Xn to restore DNA damage in CRC cells can sensitize them to anticancer agents such as SN38 (7-ethyl-10-hydroxycamptothecin) used in chemotherapy.
Collapse
Affiliation(s)
- Alessandra Scagliarini
- Université de Bourgogne Franche-Comté, F-21000 Dijon, France; (A.S.); (A.M.); (V.A.)
- INSERM Research Center U1231—Cancer and Adaptive Immune Response Team, Bioactive Molecules and Health Research Group, F-21000 Dijon, France
| | - Aline Mathey
- Université de Bourgogne Franche-Comté, F-21000 Dijon, France; (A.S.); (A.M.); (V.A.)
- INSERM Research Center U1231—Cancer and Adaptive Immune Response Team, Bioactive Molecules and Health Research Group, F-21000 Dijon, France
| | - Virginie Aires
- Université de Bourgogne Franche-Comté, F-21000 Dijon, France; (A.S.); (A.M.); (V.A.)
- INSERM Research Center U1231—Cancer and Adaptive Immune Response Team, Bioactive Molecules and Health Research Group, F-21000 Dijon, France
| | - Dominique Delmas
- Université de Bourgogne Franche-Comté, F-21000 Dijon, France; (A.S.); (A.M.); (V.A.)
- INSERM Research Center U1231—Cancer and Adaptive Immune Response Team, Bioactive Molecules and Health Research Group, F-21000 Dijon, France
- Centre Anticancéreux Georges François Leclerc, F-21000 Dijon, France
| |
Collapse
|
18
|
Hashem MA, Shoeeb SB, Abd-Elhakim YM, Mohamed WA. The antitumor activity of Arthrospira platensis and/or cisplatin in a murine model of Ehrlich ascites carcinoma with hematinic and hepato-renal protective action. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.103831] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
19
|
Sioud F, Amor S, Toumia IB, Lahmar A, Aires V, Chekir-Ghedira L, Delmas D. A New Highlight of Ephedra alata Decne Properties as Potential Adjuvant in Combination with Cisplatin to Induce Cell Death of 4T1 Breast Cancer Cells In Vitro and In Vivo. Cells 2020; 9:E362. [PMID: 32033130 PMCID: PMC7072491 DOI: 10.3390/cells9020362] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 01/24/2020] [Accepted: 01/28/2020] [Indexed: 12/29/2022] Open
Abstract
Despite major advances in the last 10 years, whether in terms of prevention or treatment, the 5 year survival rate remains relatively low for a large number of cancers. These therapeutic failures can be the consequence of several factors associated with the cellular modifications or with the host by itself, especially for some anticancer drugs such as cisplatin, which induces a nephrotoxicity. In the strategy of research for active molecules capable both of exerting a protective action against the deleterious effects of cisplatin and exerting a chemosensitizing action with regard to cancer cells, we tested the potential effects of Ephedra alata Decne extract (E.A.) rich in polyphenolic compounds towards a 4T1 breast cancer model in vitro and in vivo. We showed that E.A. extract inhibited cell viability of 4T1 breast cancer cells and induced apoptosis in a caspase-dependent manner, which involved intrinsic pathways. Very interestingly, we observed a synergic antiproliferative and pro-apoptotic action with cisplatin. These events were associated with a strong decrease of breast tumor growth in mice treated with an E.A./cisplatin combination and simultaneously with a decrease of hepato- and nephrotoxicities of cisplatin.
Collapse
Affiliation(s)
- Fairouz Sioud
- Research Unit Bioactive Natural Products and Biotechnology UR17ES49, Faculty of Dental Medicine of Monastir, University of Monastir, Avicenne street, Monastir 5000, Tunisia; (F.S.); (I.b.T.); (A.L.); (L.C.-G.)
- Université de Bourgogne Franche-Comté, F-21000 Dijon, France; (S.A.); (V.A.)
- INSERM Research Center U1231—Cancer and Adaptive Immune Response Team, F-21000 Dijon, France
| | - Souheila Amor
- Université de Bourgogne Franche-Comté, F-21000 Dijon, France; (S.A.); (V.A.)
- INSERM Research Center U1231—Cancer and Adaptive Immune Response Team, F-21000 Dijon, France
| | - Imène ben Toumia
- Research Unit Bioactive Natural Products and Biotechnology UR17ES49, Faculty of Dental Medicine of Monastir, University of Monastir, Avicenne street, Monastir 5000, Tunisia; (F.S.); (I.b.T.); (A.L.); (L.C.-G.)
| | - Aida Lahmar
- Research Unit Bioactive Natural Products and Biotechnology UR17ES49, Faculty of Dental Medicine of Monastir, University of Monastir, Avicenne street, Monastir 5000, Tunisia; (F.S.); (I.b.T.); (A.L.); (L.C.-G.)
| | - Virginie Aires
- Université de Bourgogne Franche-Comté, F-21000 Dijon, France; (S.A.); (V.A.)
- INSERM Research Center U1231—Cancer and Adaptive Immune Response Team, F-21000 Dijon, France
| | - Leila Chekir-Ghedira
- Research Unit Bioactive Natural Products and Biotechnology UR17ES49, Faculty of Dental Medicine of Monastir, University of Monastir, Avicenne street, Monastir 5000, Tunisia; (F.S.); (I.b.T.); (A.L.); (L.C.-G.)
| | - Dominique Delmas
- Université de Bourgogne Franche-Comté, F-21000 Dijon, France; (S.A.); (V.A.)
- INSERM Research Center U1231—Cancer and Adaptive Immune Response Team, F-21000 Dijon, France
- Centre anticancéreux Georges François Leclerc Center, F-21000 Dijon, France
| |
Collapse
|