1
|
Saif-Elnasr M, Elmalawany AM, Abdel-Khalek AF. The antioxidant and anti-inflammatory effects of gallic acid and/or cerium oxide nanoparticles synthesized by gamma-irradiation ameliorate cisplatin-induced hepatic injury. Arch Physiol Biochem 2025:1-13. [PMID: 40402839 DOI: 10.1080/13813455.2025.2507760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/30/2025] [Accepted: 05/12/2025] [Indexed: 05/24/2025]
Abstract
CONTEXT Cisplatin is a widely utilised chemotherapeutic agent for cancer therapy, but various systemic toxicities limit its effectiveness. OBJECTIVE This study aimed to assess the hepatoprotective properties expressed by gallic acid (GA) and gamma-irradiation synthesized cerium oxide nanoparticles (CONPs) in response to hepatic damage produced by cisplatin in albino rats. MATERIALS AND METHODS Serum AST and ALT activities and levels of MDA, TAC, NF‑kB, TNF‑α, and TGF‑β were determined in hepatic tissue, along with histopathological examination. RESULTS The executive impact of cisplatin led to a notable rise in the serum activity of hepatic enzymes AST and ALT. Conversely, in the groups receiving treatment with either or both GA and CONPs, the liver function enzymes exhibited a decline in their activity. In addition, in the hepatotoxicity models, the levels of hepatic MDA were significantly increased, accompanied by a reduction of hepatic TAC. While administration of GA, CONPs or their combination to cisplatin-injected rats resulted in a noteworthy reduction in MDA level. Conversely, the hepatic TAC level increased compared to the group that received cisplatin. The hepatic tissue architecture in rats exposed to cisplatin was found to undergo significant alterations. Furthermore, the cisplatin induced overexpression of NF-kB, TNF-α, and TGF-β. The hepatic histopathological changes observed in rats induced with cisplatin were significantly attenuated after pre-treatment with GA, CONPs, and their combination. GA, CONPs, and their co-administration resulted in reducing the levels of NF-κB, TNF-α and TGF-β compared to the group received cisplatin. DISCUSSION AND CONCLUSION In summary, GA and CONPs synthesized by gamma-irradiation resulted in a noteworthy reduction of liver damage caused by cisplatin exposure. Their potent antioxidant and immunoprotective properties were cited as the cause of this phenomenon.
Collapse
Affiliation(s)
- Mostafa Saif-Elnasr
- Health Radiation Research Department, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Alshimaa M Elmalawany
- Clinical Pathology Department, National Liver Institute, Menoufia University, Menoufia, Egypt
| | - Assmaa Fathi Abdel-Khalek
- Internal Medicine Unit, Health Radiation Research Department, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| |
Collapse
|
2
|
Saad KM, Elmasry K, Baban B, Livingston MJ, Dong Z, Abdelmageed ME, Abdelaziz RR, Suddek GM, Elmarakby AA. Protocatechuic Acid Ameliorates Cisplatin-Induced Inflammation and Apoptosis in Mouse Proximal Tubular Cells. Int J Mol Sci 2025; 26:4115. [PMID: 40362355 PMCID: PMC12071929 DOI: 10.3390/ijms26094115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 04/16/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Cisplatin is a highly cytotoxic drug used for the treatment of head, neck, and soft tissue cancers; however, it has nephrotoxic effects that can lead to acute kidney injury. Protocatechuic acid (PCA) is a natural widely available antioxidant found in many fruits such as kiwi, mango, and berries. We have recently shown that PCA reduced renal injury in a mouse model of unilateral ureteral obstruction. The current study aims to investigate the protective effects of PCA in Cisplatin-induced inflammation in vitro in Boston University Mouse Proximal Tubular (BUMPT) cells. BUMPT cells were cultured in complete DMEM. Confluent BUMPT cells were then treated with 20 μM Cisplatin ± PCA 50 or 100 μM for 24 h. PCA treatment showed a dose-depending increase in % cell viability in Cisplatin-treated BUMPT cells. PCA treatment also dose-dependently decreased Cisplatin-induced increases in oxidative stress (ROS and TBARS), inflammation (p-NF-κB and IL-6), and apoptosis (cleaved caspase-3 and % of TUNEL+ cells) compared to Cisplatin-only treatment. The reduction in oxidative stress, inflammation, and apoptosis with PCA treatment in Cisplatin-treated BUMPT cells was associated with decreases in tubular physical barrier resistance and the expression of the tight junction protein zonula occludens-1 (ZO-1) when compared to BUMPT cells treated with Cisplatin alone. The current findings suggest that PCA treatment improves tubular barrier function in Cisplatin-treated BUMPT cells via reductions in oxidative stress, inflammation, and apoptosis.
Collapse
Affiliation(s)
- Karim M. Saad
- Department of Oral Biology and Diagnostic Sciences, The Dental College of Georgia, Augusta University, 1450 Laney Walker Blvd, Augusta, GA 30912, USA; (K.M.S.); (K.E.); (B.B.)
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; (M.E.A.); (R.R.A.); (G.M.S.)
| | - Khaled Elmasry
- Department of Oral Biology and Diagnostic Sciences, The Dental College of Georgia, Augusta University, 1450 Laney Walker Blvd, Augusta, GA 30912, USA; (K.M.S.); (K.E.); (B.B.)
- Department of Cellular Biology and Anatomy, The Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (M.J.L.); (Z.D.)
- Department of Human Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Babak Baban
- Department of Oral Biology and Diagnostic Sciences, The Dental College of Georgia, Augusta University, 1450 Laney Walker Blvd, Augusta, GA 30912, USA; (K.M.S.); (K.E.); (B.B.)
| | - Man J. Livingston
- Department of Cellular Biology and Anatomy, The Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (M.J.L.); (Z.D.)
| | - Zheng Dong
- Department of Cellular Biology and Anatomy, The Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (M.J.L.); (Z.D.)
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
| | - Marwa E. Abdelmageed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; (M.E.A.); (R.R.A.); (G.M.S.)
| | - Rania R. Abdelaziz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; (M.E.A.); (R.R.A.); (G.M.S.)
| | - Ghada M. Suddek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; (M.E.A.); (R.R.A.); (G.M.S.)
| | - Ahmed A. Elmarakby
- Department of Oral Biology and Diagnostic Sciences, The Dental College of Georgia, Augusta University, 1450 Laney Walker Blvd, Augusta, GA 30912, USA; (K.M.S.); (K.E.); (B.B.)
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; (M.E.A.); (R.R.A.); (G.M.S.)
| |
Collapse
|
3
|
Latif S, Sohaib M, Iqbal S, Mushtaq MH, Sultan MT. Nephroprotective Potential of Lyophilized Grewia asiatica Powder Against Renal Biomarkers and Inflammation In Vivo. J Nutr Metab 2025; 2025:3726752. [PMID: 40291032 PMCID: PMC12033067 DOI: 10.1155/jnme/3726752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 03/27/2025] [Indexed: 04/30/2025] Open
Abstract
Introduction: Phalsa (Grewia asiatica) fruit is known for its rich nutritional profile and diverse pharmacological properties such as antioxidants, anti-inflammatory, and anti-cancer, making it a promising contender for preventive measures against cisplatin-induced acute kidney injury (AKI) in living organisms. Material and Methods: In the present study, rats were provided with different levels of lyophilized Grewia asiatica, i.e., 200, 300, and 400 mg/kg body weight along with control, fed on the basal diet. After trial completion, blood serum samples of rats subjected to renal biomarkers, hematology, and liver function tests, interleukin-6 (IL-6), whereas enzymes (alanine aminotransferase (ALT); sodium oxide dismutase, and glutathione) for kidney tissues along with photomicrographs for kidney tissue damage were measured. Results: The findings revealed that lyophilized Grewia asiatica provision effectively reduced renal biomarkers, blood urea nitrogen, and creatinine with AKI in the rats as well as treatments demonstrated significant improvements in antioxidant activity by reducing malonaldehyde levels and increasing the activity of glutathione, catalase, and superoxide dismutase in groups treated with dosages of 300 and 400 mg/kg powder. Conclusion: Grewia asiatica exhibited remarkable hepatoprotective properties by decreasing ALT and displayed anti-inflammatory properties, as evidenced by a substantial decrease in interleukin-6 serum levels. The study findings also added valuable insight into the multiform nephroprotective reverberation of lyophilized phalsa powder, emphasizing its plausible protective use in reducing cisplatin-induced nephrotoxicity.
Collapse
Affiliation(s)
- Saima Latif
- Department of Food Science & Human Nutrition, University of Veterinary & Animal Sciences, Lahore 54000, Punjab, Pakistan
| | - Muhammad Sohaib
- Department of Food Science & Human Nutrition, University of Veterinary & Animal Sciences, Lahore 54000, Punjab, Pakistan
| | - Sanaullah Iqbal
- Department of Food Science & Human Nutrition, University of Veterinary & Animal Sciences, Lahore 54000, Punjab, Pakistan
| | - Muhammad Hassan Mushtaq
- Department of Epidemiology & Public Health, University of Veterinary and Animal Sciences, Lahore 54000, Punjab, Pakistan
| | | |
Collapse
|
4
|
Arzani S, Farzipour S, Talebpour Amiri F, Hosseinimehr SJ. Gliclazide protects ionizing radiation-induced intestinal injury in mice by inhibiting oxidative stress and caspase-3. BIOTECHNOLOGIA 2024; 105:367-376. [PMID: 39844874 PMCID: PMC11748219 DOI: 10.5114/bta.2024.145257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 05/26/2024] [Accepted: 09/11/2024] [Indexed: 01/24/2025] Open
Abstract
Gliclazide (GLZ), an oral antihyperglycemic medication, has additional beneficial effects, such as anti-inflammatory and antioxidant properties, besides lowering blood glucose levels. In this study, the radio-protective effect of GLZ was evaluated against ionizing radiation (IR)-induced intestinal injury in mice. Eight groups of mice were randomized as follows: control, GLZ (5, 10, and 25 mg/kg), IR (6 Gy), and IR + GLZ (at 5, 10, and 25 mg/kg). GLZ was administered to the mice for eight consecutive days, after which they were exposed to X-rays at a single dose of 6 Gy. After irradiation, biochemical parameters, immunohistochemical, and histological examinations were conducted on the ileum of the mice. IR exposure increased the levels of malondialdehyde and protein carbonyl, while glutathione levels, as oxidative stress biomarkers, decreased. Apoptosis in ileum tissues was also assessed. Furthermore, histopathological changes were observed in the irradiated mice. GLZ treatment significantly mitigated these changes. The administration of GLZ resulted in a marked decrease in caspase-3 immunoreactivity in the ileum of irradiated mice. This preclinical study exhibited that GLZ has a radioprotective effect against intestinal injury by inhibiting oxidative stress and apoptosis.
Collapse
Affiliation(s)
- Soroush Arzani
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Soghra Farzipour
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | | | - Seyed Jalal Hosseinimehr
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
5
|
Hafezizadeh M, Salehcheh M, Mohtadi S, Mansouri E, Khodayar MJ. Zingerone effects on arsenic-induced glucose intolerance and hepatotoxicity in mice via suppression of oxidative stress-mediated hepatic inflammation and apoptosis. J Trace Elem Med Biol 2024; 86:127562. [PMID: 39531827 DOI: 10.1016/j.jtemb.2024.127562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 10/04/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Arsenic (As), a poisonous metalloid, is widely distributed in air, water, and soil and has been associated with the occurrence of diabetes and liver toxicity. Zingerone (ZNG), one of the active compounds in ginger, has several pharmacological benefits such as antioxidant and anti-inflammatory characteristics. The objective of this research was to assess the protective role of ZNG against arsenic (As)-induced glucose intolerance (GI) and hepatotoxicity in mice. METHODS Male NMRI mice were treated with ZNG (25, 50, and 100 mg/kg, oral gavage for 29 days) before As administration (10 mg/kg, oral gavage for 29 days). On the 29th day, fasting blood glucose (FBG) and glucose tolerance test were measured. The animals were euthanized (day 30), and samples from blood and tissue (liver and pancreas) were gathered for further evaluations. RESULTS Administration of ZNG inhibited As-induced elevation of FBG and GI. Moreover, hepatic tissue damage and decreased Langerhans islets' diameter caused by As administration were improved by ZNG treatment. Pretreatment with ZNG attenuated the elevation of serum liver enzymes induced by As (alanine aminotransferase, aspartate aminotransferase, and alkaline phosphatase). Also, the reduction in total thiol content, as well as the decline in antioxidant enzyme activities (catalase, superoxide dismutase, and glutathione peroxidase) and the increase in lipid peroxidation marker (thiobarbituric acid reactive substances) in the liver tissue of As-exposed mice were reversed in ZNG-treated mice. Furthermore, ZNG prevented the increase of hepatic inflammatory markers (nitric oxide and tumor necrosis factor-alpha levels, and protein expression of nuclear factor-kappa B) and apoptosis-related marker (caspase-3 protein expression) in As-treated mice. CONCLUSIONS This study has provided evidence indicating that ZNG can act as a beneficial agent in preventing As-induced hepatotoxicity and diabetes.
Collapse
Affiliation(s)
- Mobina Hafezizadeh
- Toxicology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Department of Toxicology, Faculty of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maryam Salehcheh
- Toxicology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Department of Toxicology, Faculty of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Shokooh Mohtadi
- Department of Toxicology, Faculty of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Esrafil Mansouri
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Javad Khodayar
- Toxicology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Department of Toxicology, Faculty of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
6
|
Bingul E, Bulut S, Mammadov R, Cicek B, Dogru TE, Suleyman H, Mendil AS. Effect of ethyl acetate extract from Usnea longissima on chemotherapy-associated multiple organ dysfunction in rats. Biomed Pharmacother 2024; 181:117636. [PMID: 39489120 DOI: 10.1016/j.biopha.2024.117636] [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: 08/03/2024] [Revised: 10/10/2024] [Accepted: 10/28/2024] [Indexed: 11/05/2024] Open
Abstract
BACKGROUND The toxic effects of doxorubicin and cisplatin in various organs have been associated with oxidative stress. Studies have shown that Usnea longissima has strong antioxidant effects. This study aimed to investigate the protective effect of ethyl acetate extract from Usnea longissima (ULE), which is known to have strong antioxidant effects, on chemotherapeutic-induced heart, kidney, liver, and ovarian toxicity. METHODS Albino Wistar female rats were divided into five groups (12 rats per group): healthy (HG), doxorubicin (DOX), Cisplatin (CIS), Doxorubicin+ ULE (DULE), Cisplatin+ ULE (CULE). In this experiment, ULE was given 100 mg/kg orally. After 1 hour, 2.5 mg/kg doxorubicin and 2.5 mg/kg cisplatin were administered intraperitoneally. Drug treatments continued once a day for seven days. At the end of seven days, six rats from each group were euthanized and heart, kidney, liver, and ovary tissues were analyzed biochemically. The remaining rats were left in the laboratory with male rats for 45 days for reproduction. RESULTS ULE inhibited chemotherapeutic-induced increase in malondialdehyde, tumor necrosis factor-alpha, and interleukin 6 and a decrease in total glutathione in liver, kidney, and ovarian tissues. ULE also inhibited the increase of blood urea nitrogen, creatinine, alanine aminotransferase, and aspartate aminotransferase in serum. ULE treatment had no protective effect against doxorubicin and cisplatin cardiac toxicity. On the other hand, ULE also decreased the delay in pregnancy induced by chemotherapy. CONCLUSION ULE may be considered an adjuvant therapy in patients receiving chemotherapy to reduce liver, kidney, and ovarian toxicity.
Collapse
Affiliation(s)
- Eda Bingul
- Gynecology and Obstetrics Clinic, Erzurum City Hospital, Erzurum 25240, Turkey.
| | - Seval Bulut
- Department of Pharmacology, Faculty of Medicine, Erzincan Binali Yildirim University, Erzincan 24100, Turkey.
| | - Renad Mammadov
- Department of Pharmacology, Faculty of Medicine, Erzincan Binali Yildirim University, Erzincan 24100, Turkey.
| | - Betul Cicek
- Department of Physiology, Faculty of Medicine, Erzincan Binali Yildirim University, Erzincan 24100, Turkey.
| | - Tugba Erkmen Dogru
- Department of Biochemistry, Faculty of Medicine, Erzincan Binali Yildirim University, Erzincan 24100, Turkey.
| | - Halis Suleyman
- Department of Pharmacology, Faculty of Medicine, Erzincan Binali Yildirim University, Erzincan 24100, Turkey.
| | - Ali Sefa Mendil
- Department of Pathology, Faculty of Veterinary, Erciyes University, Kayseri 38280, Turkey.
| |
Collapse
|
7
|
Kounatidis D, Vallianou NG, Karampela I, Rebelos E, Kouveletsou M, Dalopoulos V, Koufopoulos P, Diakoumopoulou E, Tentolouris N, Dalamaga M. Anti-Diabetic Therapies and Cancer: From Bench to Bedside. Biomolecules 2024; 14:1479. [PMID: 39595655 PMCID: PMC11591849 DOI: 10.3390/biom14111479] [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: 10/24/2024] [Revised: 11/17/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024] Open
Abstract
Diabetes mellitus (DM) is a significant risk factor for various cancers, with the impact of anti-diabetic therapies on cancer progression differing across malignancies. Among these therapies, metformin has gained attention for its potential anti-cancer effects, primarily through modulation of the AMP-activated protein kinase/mammalian target of rapamycin (AMPK/mTOR) pathway and the induction of autophagy. Beyond metformin, other conventional anti-diabetic treatments, such as insulin, sulfonylureas (SUs), pioglitazone, and dipeptidyl peptidase-4 (DPP-4) inhibitors, have also been examined for their roles in cancer biology, though findings are often inconclusive. More recently, novel medications, like glucagon-like peptide-1 (GLP-1) receptor agonists, dual GLP-1/glucose-dependent insulinotropic polypeptide (GIP) agonists, and sodium-glucose co-transporter-2 (SGLT-2) inhibitors, have revolutionized DM management by not only improving glycemic control but also delivering substantial cardiovascular and renal benefits. Given their diverse metabolic effects, including anti-obesogenic properties, these novel agents are now under meticulous investigation for their potential influence on tumorigenesis and cancer advancement. This review aims to offer a comprehensive exploration of the evolving landscape of glucose-lowering treatments and their implications in cancer biology. It critically evaluates experimental evidence surrounding the molecular mechanisms by which these medications may modulate oncogenic signaling pathways and reshape the tumor microenvironment (TME). Furthermore, it assesses translational research and clinical trials to gauge the practical relevance of these findings in real-world settings. Finally, it explores the potential of anti-diabetic medications as adjuncts in cancer treatment, particularly in enhancing the efficacy of chemotherapy, minimizing toxicity, and addressing resistance within the framework of immunotherapy.
Collapse
Affiliation(s)
- Dimitris Kounatidis
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Medical School, Laiko General Hospital, National and Kapustina University of Athens, 11527 Athens, Greece; (D.K.); (E.R.); (M.K.); (E.D.); (N.T.)
| | - Natalia G. Vallianou
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126 Athens, Greece; (N.G.V.); (V.D.); (P.K.)
| | - Irene Karampela
- 2nd Department of Critical Care, Medical School, Attikon General University Hospital, University of Athens, 1 Rimini str., 12461 Athens, Greece;
| | - Eleni Rebelos
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Medical School, Laiko General Hospital, National and Kapustina University of Athens, 11527 Athens, Greece; (D.K.); (E.R.); (M.K.); (E.D.); (N.T.)
| | - Marina Kouveletsou
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Medical School, Laiko General Hospital, National and Kapustina University of Athens, 11527 Athens, Greece; (D.K.); (E.R.); (M.K.); (E.D.); (N.T.)
| | - Vasileios Dalopoulos
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126 Athens, Greece; (N.G.V.); (V.D.); (P.K.)
| | - Petros Koufopoulos
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126 Athens, Greece; (N.G.V.); (V.D.); (P.K.)
| | - Evanthia Diakoumopoulou
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Medical School, Laiko General Hospital, National and Kapustina University of Athens, 11527 Athens, Greece; (D.K.); (E.R.); (M.K.); (E.D.); (N.T.)
| | - Nikolaos Tentolouris
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Medical School, Laiko General Hospital, National and Kapustina University of Athens, 11527 Athens, Greece; (D.K.); (E.R.); (M.K.); (E.D.); (N.T.)
| | - Maria Dalamaga
- Department of Biological Chemistry, National and Kapodistrian University of Athens, 75 Mikras Asias str., 11527 Athens, Greece
| |
Collapse
|
8
|
Sohail N, Farhat H, Qureshi SA, Ullah I, Ali MS. The brown algae: Sargassum binderi sonder shows a potential nephroprotective activity in in-vivo experimental model. ANNALES PHARMACEUTIQUES FRANÇAISES 2024; 82:1046-1061. [PMID: 38871117 DOI: 10.1016/j.pharma.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 06/02/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024]
Abstract
OBJECTIVES This study aimed to investigate the protective activity of brown seaweed, the ethanolic and water extracts of Sargassum binderi (S. binderi) were examined. Anticancer drug, cisplatin is normally used for the treatment of solid tumors that cause acute kidney damage after assemblage in the renal tubules. MATERIAL AND METHODS It was an acute nephrotoxicity study, animals were divided into several groups randomly, cisplatin (7mg/kg i.p.) and normal saline were used as positive and negative control respectively. The S. bindari ethanolic and water extract were given orally in a dose of 200mg/kg for 5days. Various biomarkers were assessed to observe the nephroprotective potential, while antioxidant activities were investigated using reduced glutathione, catalase and malondialdehyde as oxidative stress. GCMS was performed to validate the presence of important therapeutic moieties. RESULTS The current result justified that pretreatment with S. binderi inhibited the elevation of antioxidant parameters and also showed protection against lipid peroxidation, induced by cisplatin challenge. The overall impact was the nephroprotection, which has been revealed from the results. GCMS evaluation of hexanes fraction revealed the presence of therapeutically important compounds including heptasiloxane, 3,7,11,15-tetramethyl-2-hexadecen-1-ol, hexadecamethyl, cyclooctasiloxane, and hexadecamethyl. These compounds have been reported for their antioxidant, antibacterial, anticancer, and antifungal activities. CONCLUSION S. binderi showed reno-protective effect by checking their well-known biochemical parameters probably due to the antioxidant activity as confirmed by the presence of compounds.
Collapse
Affiliation(s)
- Nida Sohail
- Department of Biochemistry, University of Karachi, Main University Road, Karachi City, Sindh 75270, Pakistan.
| | - Hafiza Farhat
- Institute of Biological Sciences, Gomal University, D.I Khan, D.I Khan 29050, Pakistan
| | - Shamim Akhtar Qureshi
- Department of Biochemistry, University of Karachi, Main University Road, Karachi City, Sindh 75270, Pakistan
| | - Irfan Ullah
- Department of Neuroscience, The University of Minnesota, Minneapolis, MN 55455, United States
| | - Muhammad Shaiq Ali
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Main University Road, Karachi City, Sindh 75270, Pakistan
| |
Collapse
|
9
|
Yilmaz A, Dizman F, Akyildiz K, Mataraci Karakas S, Mercantepe T, Uydu HA, Tumkaya L, Ozturk K. The Hepatoprotective Effects of Camellia sinensis on Cisplatin-Induced Acute Liver Injury. Life (Basel) 2024; 14:1077. [PMID: 39337861 PMCID: PMC11432752 DOI: 10.3390/life14091077] [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: 08/01/2024] [Revised: 08/20/2024] [Accepted: 08/26/2024] [Indexed: 09/30/2024] Open
Abstract
Acute liver injury is an increasing global health problem. It is a widespread side effect of cisplatin treatment in the clinic and can lead to liver failure if not treated promptly. Previous studies have revealed that green tea can protect some organs from treatments. However, the potential of white tea to prevent the negative effects of acute liver injury has not been addressed so far. The purpose of this study was to investigate the reduction in cisplatin-induced liver injury in rats receiving white tea. Female Sprague Dawley rats with similar weight were selected in this study. Twenty-four rats were divided into three groups of eight animals each and ad libitum nutrition was provided. The control and cisplatin groups were given tap water only, while the white tea + cisplatin group received white tea at a 0.5% weight/volume concentration for four weeks. At the end of the fourth week, the white tea + cisplatin group and the cisplatin group received a single dose of cisplatin (7 mg/kg) via the intraperitoneal route. Five days after that procedure, the rats were anesthetized. Liver tissues and blood samples were collected, which were used for biochemical and histopathological analyses. According to biochemical results, liver tissue MDA and GSH, serum ALT, and AST levels significantly increased in the cisplatin group compared to the control group. Compared with the cisplatin group, although MDA, AST, ALT, and GSH levels were lower in the white tea + cisplatin group, only GSH levels were statistically different. The examination of histopathological and immunohistochemical findings revealed apoptotic cells, vascular congestion, and sinusoidal dilatation in the cisplatin group compared to the control group. This adverse event decreased in the white tea + cisplatin group compared to the cisplatin group. In conclusion, white tea exhibits an ameliorating effect on cisplatin-induced liver injury.
Collapse
Affiliation(s)
- Adnan Yilmaz
- Department of Biochemistry, Faculty of Medicine, Recep Tayyip Erdogan University, 53010 Rize, Turkey
| | - Fatih Dizman
- Recep Tayyip Erdogan University Research and Training Hospital, 53010 Rize, Turkey
| | - Kerimali Akyildiz
- Department of Biochemistry, Faculty of Medicine, Recep Tayyip Erdogan University, 53010 Rize, Turkey
| | - Sibel Mataraci Karakas
- Department of Biochemistry, Faculty of Medicine, Recep Tayyip Erdogan University, 53010 Rize, Turkey
| | - Tolga Mercantepe
- Department of Histology and Embryology, Faculty of Medicine, Recep Tayyip Erdogan University, 53010 Rize, Turkey
| | - Huseyin Avni Uydu
- Department of Biochemistry, Faculty of Medicine, Samsun University, 55000 Samsun, Turkey
| | - Levent Tumkaya
- Department of Histology and Embryology, Faculty of Medicine, Ondokuz Mayis University, 55000 Samsun, Turkey
| | - Koksal Ozturk
- Recep Tayyip Erdogan University Research and Training Hospital, 53010 Rize, Turkey
| |
Collapse
|
10
|
Oliveira CA, Mercês ÉAB, Portela FS, Malheiro LFL, Silva HBL, De Benedictis LM, De Benedictis JM, Silva CCDE, Santos ACL, Rosa DP, Velozo HS, de Jesus Soares T, de Brito Amaral LS. An integrated view of cisplatin-induced nephrotoxicity, hepatotoxicity, and cardiotoxicity: characteristics, common molecular mechanisms, and current clinical management. Clin Exp Nephrol 2024; 28:711-727. [PMID: 38678166 DOI: 10.1007/s10157-024-02490-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 03/18/2024] [Indexed: 04/29/2024]
Abstract
Cisplatin (CP) is a chemotherapy drug widely prescribed to treat various neoplasms. Although fundamental for the therapeutic action of the drug, its cytotoxic mechanisms trigger adverse effects in several tissues, such as the kidney, liver, and heart, which limit its clinical use. In this sense, studies point to an essential role of damage to nuclear and mitochondrial DNA associated with oxidative stress, inflammation, and apoptosis in the pathophysiology of tissue injuries. Due to the limitation of effective preventive and therapeutic measures against CP-induced toxicity, new strategies with potential cytoprotective effects have been studied. Therefore, this article is timely in reviewing the characteristics and main molecular mechanisms common to renal, hepatic, and cardiac toxicity previously described, in addition to addressing the main validated strategies for the current management of these adverse events in clinical practice. We also handle the main promising antioxidant substances recently presented in the literature to encourage the development of new research that consolidates their potential preventive and therapeutic effects against CP-induced cytotoxicity.
Collapse
Affiliation(s)
- Caroline Assunção Oliveira
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
| | - Érika Azenathe Barros Mercês
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
- Programa de Pós-Graduação em Biociências, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
| | - Fernanda Santos Portela
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
- Programa de Pós-Graduação em Biociências, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
| | - Lara Fabiana Luz Malheiro
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
- Programa de Pós-Graduação em Biociências, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
| | | | | | | | | | | | | | - Helloisa Souza Velozo
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
| | - Telma de Jesus Soares
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
- Programa de Pós-Graduação em Biociências, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil
| | - Liliany Souza de Brito Amaral
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil.
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil.
- Programa de Pós-Graduação em Biociências, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Bahia, 45029-094, Brazil.
| |
Collapse
|
11
|
Aksoy S, Kuloğlu N, Karabulut D, Yakan B. Investigation of the effect of Myricetin on Cisplatin-induced liver hepatotoxicity. REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2024; 70:e20240136. [PMID: 39045937 PMCID: PMC11262330 DOI: 10.1590/1806-9282.20240136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 04/25/2024] [Indexed: 07/25/2024]
Abstract
OBJECTIVE Cisplatin, a widely used anticancer agent, induces hepatotoxicity alongside organ damage. Understanding Cisplatin's toxicity mechanism and developing preventive measures are crucial. Our study explores Myricetin, a flavonoid, for its protective effects against Cisplatin-induced hepatotoxicity. METHODS In our study, a total of 32 Wistar albino male rats were utilized, which were categorized into four distinct groups: Control, Myricetin, Cisplatin, and Myricetin+Cisplatin. For the histological assessment of hepatic tissues, hematoxylin-eosin and periodic acid Schiff staining were employed, alongside immunohistochemical measurements of TNF-α, interleukin-17, and interleukin-6 immunoreactivity. Additionally, aspartate transaminase and alanine transaminase values were examined by biochemical analysis. RESULTS In the histological evaluation of the tissues, a normal healthy cell structure and a strong periodic acid Schiff (+) reaction were observed in the hepatocyte cells in the tissues of the Control and Myricetin groups, while intense eosinophilia, minimal vacuolization, congestion, and sinusoidal expansions were observed in the hematoxylin-eosin stainings, and a decrease in the positive reaction in the periodic acid Schiff staining was observed in the Cisplatin group. Consistent with these histological findings, an increase in TNF-α, interleukin-17, and interleukin-6 expressions (p<0.0001) and a concomitant increase in aspartate transaminase and alanine transaminase values were observed in the Cisplatin group. In the group protected by Myricetin, a significant improvement was observed in all these histological and biochemical values. CONCLUSION Cisplatin induces notable histopathological alterations in the liver. In this context, Myricetin exhibits the potential to alleviate Cisplatin-induced damage by modulating histological parameters and biochemical processes.
Collapse
Affiliation(s)
- Sümeyye Aksoy
- Erciyes University, Institute of Health Sciences – Kayseri, Turkey
| | - Nurhan Kuloğlu
- Niğde Ömer Halisdemir University, Department of Healthcare Services – Niğde, Turkey
| | - Derya Karabulut
- Erciyes University, Faculty of Medicine, Department of Histology and Embryology – Kayseri, Turkey
| | - Birkan Yakan
- Erciyes University, Faculty of Medicine, Department of Histology and Embryology – Kayseri, Turkey
| |
Collapse
|
12
|
Wu DP, Yi W, Zhao YD, Wei YS, Liu LL, Yan QQ, Yu C, Liu JY, Zhu XX, Zhong ZG, Huang JL. Gliclazide Ameliorates Neuronal Injury by Attenuating Oxidative Stress in D-gal-Induced Senescent Cells and Aging Mice. Mol Neurobiol 2024; 61:4391-4401. [PMID: 38087171 DOI: 10.1007/s12035-023-03850-6] [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: 04/11/2023] [Accepted: 11/17/2023] [Indexed: 07/11/2024]
Abstract
Enhancement of oxidative stress and resultant neuronal injury play important roles in initiating cognitive impairment during the aging process. Thus, attenuating oxidative injury is regarded as a profitable therapeutic strategy for age-associated cognitive impairment. Previous studies showed that gliclazide (Gli) had a protective role in neuronal injury from cerebral ischemia/reperfusion (I/R) injury. However, whether Gli has a profitable effect on age-associated cognitive impairment remains largely unclear. The present study showed that Gli held the potential to attenuate neuronal apoptosis in D-gal-induced senescent cells and aging mice. Additionally, Gli could alleviate synaptic injury and cognitive function in D-gal-induced aging mice. Further study showed that Gli could attenuate oxidative stress in D-gal-induced senescent cells and aging mice. The p38 MAPK pathway was predicted as the downstream target of Gli retarding oxidative stress using in silico analysis. Further studies revealed that Gli attenuated D-gal-induced phosphorylation of p38 and facilitated Nrf2 nuclear expression, indicating that the anti-oxidative property of Gli may be associated with the p38 MAPK pathway. The study demonstrates that Gli has a beneficial effect on ameliorating D-gal-induced neuronal injury and cognitive impairment, making this compound a promising agent for the prevention and treatment of age-associated cognitive impairment.
Collapse
Affiliation(s)
- Deng-Pan Wu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Pharmacy School of Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- Xuzhou Ruihu Health Management Consulting Co., Ltd, Xuzhou, 221002, Jiangsu, China
| | - Wen Yi
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Pharmacy School of Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Yuan-Dan Zhao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Pharmacy School of Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Yan-Su Wei
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Pharmacy School of Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Ling-Ling Liu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Pharmacy School of Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Qiu-Qing Yan
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Pharmacy School of Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Chao Yu
- School of Basic Medicine, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Jin-Yuan Liu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Pharmacy School of Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Xiao-Xiao Zhu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Pharmacy School of Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Zhen-Guo Zhong
- Scientific Research Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, 530200, Guangxi, China
| | - Jin-Lan Huang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Pharmacy School of Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- Xuzhou Ruihu Health Management Consulting Co., Ltd, Xuzhou, 221002, Jiangsu, China.
| |
Collapse
|
13
|
Gad F, Abdelghaffar Emam M, Eldeeb AA, Abdelhameed AA, Soliman MM, Alotaibi KS, Albattal SB, Abughrien B. Mitigative Effects of l-Arginine and N-Acetyl Cysteine against Cisplatin-Induced Testicular Dysfunction and Toxicity through the Regulation of Antioxidant, Anti-inflammatory, and Antiapoptotic Markers: Role of miR-155 and miR-34c Expression. ACS OMEGA 2024; 9:27680-27691. [PMID: 38947789 PMCID: PMC11209920 DOI: 10.1021/acsomega.4c03742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/28/2024] [Accepted: 06/05/2024] [Indexed: 07/02/2024]
Abstract
Testicular dysfunction is a common adverse effect of cisplatin (CIS) administration as a chemotherapeutic drug. The current study has outlined the role of micro-RNAs (miR-155 and 34c) in CIS-induced testicular dysfunction and evaluated the protective effect of N-acetyl cysteine (NAC) and/or l-arginine (LA). Seven groups of Albino rats were used for this study. The control (C) group received physiological saline; the CIS group was injected CIS (7 mg/kg IP, once) on day 21 of the experiment; the NAC group was administered NAC (150 mg/kg intragastric, for 28 days); and the LA group was injected LA (50 mg/kg IP, for 28 days). NAC+CIS, LA+CIS, and NAC+LA+CIS groups received the above regime. CIS significantly reduced serum testosterone, LH, and FSH concentrations with decline of testicular enzyme activities. CIS caused significant elevation in testicular oxidative-stress biomarkers, inflammation-associated cytokines, and apoptosis markers, along with overexpression of miR-155 and low miR-34c expression. Additionally, marked testicular degenerative changes were observed in the examined histological section; a significant decrease in the expression of PCNA with significant increase in expressions of F4/80 and BAX was confirmed. The administration of NAC or LA upregulated testicular functions and improved histopathological and immunohistochemical changes as well as miRNA expression compared with the CIS-administered group. Rats receiving both NAC and LA showed a more significant ameliorative effect compared with groups receiving NAC or LA alone. In conclusion, NAC or LA showed an ameliorative effect against CIS-induced testicular toxicity and dysfunction through the regulation of antioxidant, anti-inflammatory, and antiapoptotic markers and via modulating miR-155 and miR-34c expression.
Collapse
Affiliation(s)
- Fatma
A. Gad
- Clinical
Pathology Department, Faculty of Veterinary Medicine, Benha University, P.O. Box13736 Benha, Egypt
| | - Mahmoud Abdelghaffar Emam
- Histology
Department., Faculty of Veterinary Medicine, Benha University, P.O. Box 13736 Benha, Egypt
| | - Abeer A. Eldeeb
- Clinical
Pharmacology Department, Faculty of Medicine, Benha University, 13511 Benha, Egypt
| | - Abeer A. Abdelhameed
- Clinical
Pharmacology Department, Faculty of Medicine, Benha University, 13511 Benha, Egypt
| | - Mohamed Mohamed Soliman
- Department
of Clinical Laboratory Sciences, Turabah University College, Taif University, P.O.
Box 11099, Taif 21944, Saudi Arabia
| | - Khalid S. Alotaibi
- General
Science and English Language Department, College of Applied Sciences, AlMaarefa University, Riyadh 71666, Saudi Arabia
| | - Shatha B. Albattal
- General
Science and English Language Department, College of Applied Sciences, AlMaarefa University, Riyadh 71666, Saudi Arabia
| | - Badia Abughrien
- Anatomy and
Histology Department, Faculty of Veterinary Medicine, Tripoli University, 15673 Tripoli, Libya
| |
Collapse
|
14
|
Okkay IF, Famurewa A, Bayram C, Okkay U, Mendil AS, Sezen S, Ayaz T, Gecili I, Ozkaraca M, Senyayla S, Hacimuftuoglu A. Arbutin abrogates cisplatin-induced hepatotoxicity via upregulating Nrf2/HO-1 and suppressing genotoxicity, NF-κB/iNOS/TNF-α and caspase-3/Bax/Bcl2 signaling pathways in rats. Toxicol Res (Camb) 2024; 13:tfae075. [PMID: 38770183 PMCID: PMC11102346 DOI: 10.1093/toxres/tfae075] [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: 11/24/2023] [Revised: 05/08/2024] [Accepted: 05/10/2024] [Indexed: 05/22/2024] Open
Abstract
BACKGROUND Cisplatin is a potent anticancer agent widely employed in chemotherapy. However, cisplatin leads to toxicity on non-targeted healthy organs, including the liver. We investigated the hepatoprotective mechanism of arbutin (ARB), a glycosylated hydroquinone, against cisplatin-induced hepatotoxicity. METHODS Rats were orally administered with ARB (ARB1 = 50 mg/kg; ARB2 = 100 mg/kg) for 14 consecutive days against hepatotoxicity induced by a single dose of cisplatin (10 mg/kg) on day 15. Three days after the intraperitoneal cisplatin injection, serum and liver tissue were collected for subsequent analyses. RESULTS Cisplatin triggered marked increases in serum AST, ALT, and ALP activities, hepatic malondialdehyde (MDA) and reactive oxygen species (ROS) coupled with a considerable diminution in hepatic activities of superoxide dismutase (SOD), catalase (CAT) and the concentration of reduced glutathione (GSH). The gene expressions of interleukin-1β (IL-1β), tumor necrosis factor (TNF-α), and IL-6 were notably increased. The pre-administration of ARB1 and ARB2 reduced AST, ALT and ALP in serum and restored SOD, CAT, GSH, ROS, MDA and cytokine levels which was also evidenced by alleviated hepatic lesions. Further, cisplatin-induced prominent alterations in the gene expressions of nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), iNOS, NF-κB, Bax, Bcl-2, caspase-3 and 8-OHdG in the liver. Interestingly, ARB protected the liver and mitigated the cisplatin-induced alterations in serum AST, ALT, ALP, and reduced hepatic redox markers, 8-OdG, inflammatory markers and gene expressions. CONCLUSION The findings demonstrate that ARB is a potential protective adjuvant against cisplatin-induced hepatotoxicity via inhibition of hepatic oxidative stress, inflammation, and apoptosis.
Collapse
Affiliation(s)
- Irmak Ferah Okkay
- Pharmacology Department, Faculty of Pharmacy, Ataturk University, Ataturk Street, 25240, Yakutiye, Erzurum, Turkey
| | - Ademola Famurewa
- Medical Biochemistry Department, Faculty of Basic Medical Sciences, College of Medical Sciences, Alex-Ekwueme Federal University, Ndufu-Alike Ikwo, Abakaliki, Ebonyi State, Nigeria
| | - Cemil Bayram
- Medical Pharmacology Department, Faculty of Medicine, Ataturk University, Ataturk Street, 25240, Yakutiye, Erzurum, Turkey
| | - Ufuk Okkay
- Medical Pharmacology Department, Faculty of Medicine, Ataturk University, Ataturk Street, 25240, Yakutiye, Erzurum, Turkey
- Vaccine Development Application and Research Center, Ataturk University, Ataturk Street, 25240, Yakutiye, Erzurum, Turkey
| | - Ali Sefa Mendil
- Department of Pathology, Faculty of Veterinary Medicine, Erciyes University, Yenidogan, Kume Evleri, 38280 Talas, Kayseri, Turkey
| | - Selma Sezen
- Pharmacology Department, Faculty of Medicine, Agri Ibrahim Cecen University, New University Street, No 2, 04100, Agri, Turkey
| | - Teslime Ayaz
- Internal Medicine Department, Faculty of Medicine, Recep Tayyip Erdogan University, 53200, Islampasa, Rize, Turkey
| | - Ibrahim Gecili
- Medical Pharmacology Department, Faculty of Medicine, Ataturk University, Ataturk Street, 25240, Yakutiye, Erzurum, Turkey
| | - Mustafa Ozkaraca
- Pathology Department, Faculty of Veterinary Medicine, Sivas Cumhuriyet University, 58070, Imaret, Sivas, Turkey
| | - Selcuk Senyayla
- Medical Pharmacology Department, Faculty of Medicine, Ataturk University, Ataturk Street, 25240, Yakutiye, Erzurum, Turkey
| | - Ahmet Hacimuftuoglu
- Medical Pharmacology Department, Faculty of Medicine, Ataturk University, Ataturk Street, 25240, Yakutiye, Erzurum, Turkey
- Vaccine Development Application and Research Center, Ataturk University, Ataturk Street, 25240, Yakutiye, Erzurum, Turkey
| |
Collapse
|
15
|
ERTUNÇ O, ERZURUMLU Y, SAVRAN M, ÇATAKLI D, DOĞAN KIRAN E, PEKGÖZ Ş. Potential Hepatoprotective Effects of Irbesartan, an Accessible Angiotensin II Receptor Blocker, Against Cisplatin-Induced Liver Injury in a Rat Model. Turk J Pharm Sci 2024; 21:88-94. [PMID: 38742755 PMCID: PMC11096784 DOI: 10.4274/tjps.galenos.2023.90846] [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: 11/06/2022] [Accepted: 05/06/2023] [Indexed: 05/16/2024]
Abstract
Objectives Drug-induced liver injury is a common adverse reaction that frequently occurs with chemotherapeutic agents, such as cisplatin (CIS). This study seeks to enhance our understanding of drug actions and their associated adverse effects by examining the toxicity of CIS on rat liver tissue. We aimed to investigate the potential hepatoprotective effects of irbesartan (IRB), an easily accessible angiotensin II receptor blocker, in mitigating CIS-induced hepatotoxicity. Materials and Methods Wistar albino rats were divided into four groups. These groups included a control group [saline, per oral (p.o.)] for seven days, and 1 mL saline intraperitoneal [(i.p.) on the fourth day]; a CIS group (1 mL saline for seven days and 7.5 mg/kg CIS i.p. on the fourth day); a CIS + IRB group (IRB: 50 mg/kg p.o. for seven days and 7.5 mg/kg CIS i.p. on the fourth day), and an IRB group (50 mg/kg IRB p.o. for seven days). The effect of IRB on interleukin-1 beta (IL-1β) and caspase 3 levels was evaluated by immunohistochemical analysis, and its effects on mRNA expression levels of CCAAT/enhancer-binding protein homologous protein (CHOP) and immunoglobulin-heavy-chain-binding protein (BiP) were tested by quantitative real-time polymerase chain reaction. Results IRB administration mitigated CIS-induced liver toxicity by inhibiting endoplasmic reticulum (ER) stress. Specifically, this drug reduced the mRNA expression of ER stress markers, including CHOP and BiP. In addition, IRB treatment decreased oxidative stress, inflammatory responses, and apoptotic markers. Conclusion These findings suggest that IRB is a promising therapeutic option for preventing CIS-induced liver injury, potentially by modulating ER stress-related pathways.
Collapse
Affiliation(s)
- Onur ERTUNÇ
- Süleyman Demirel University Faculty of Medicine, Department of Pathology, Isparta, Türkiye
| | - Yalçın ERZURUMLU
- Süleyman Demirel University Faculty of Pharmacy, Department of Biochemistry, Isparta, Türkiye
| | - Mehtap SAVRAN
- Süleyman Demirel University Faculty of Medicine, Department of Pharmacology, Isparta, Türkiye
| | - Deniz ÇATAKLI
- Süleyman Demirel University Faculty of Medicine, Department of Pharmacology, Isparta, Türkiye
| | - Eltaf DOĞAN KIRAN
- Süleyman Demirel University Faculty of Medicine, Department of Biochemistry, Isparta, Türkiye
| | - Şakir PEKGÖZ
- Süleyman Demirel University Faculty of Medicine, Department of Pathology, Isparta, Türkiye
| |
Collapse
|
16
|
Moynihan E, Galiana-Cameo M, Sandri M, Ruffini A, Panseri S, Velasco-Torrijos T, Montesi M, Montagner D. 2D and 3D anticancer properties of C2-functionalised glucosamine-Pt (IV) prodrugs based on cisplatin scaffold. Front Chem 2024; 12:1388332. [PMID: 38770272 PMCID: PMC11102980 DOI: 10.3389/fchem.2024.1388332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 04/22/2024] [Indexed: 05/22/2024] Open
Abstract
A series of C2-functionalied Pt (IV) glycoconjugates based on glucosamine have been synthesised, characterised and tested as anticancer agents on a series of different 2D and 3D cancer cell lines. The carbohydrate will act as a targeted delivery system to improve the selectivity, exploiting the Warburg Effect and the GLUTs receptors that are overexpressed in most of the cancer cells. The hydroxyl at C2 of the carbohydrates does not participate in hydrogen bonding with the GLUTs receptors, making C2 an attractive position for drug conjugation as seen in literature. In this study, we use the amino functionality at the C2 position in glucosamine and Copper-catalysed Azide-Alkyne Cycloaddition "click" (CuAAC) reaction to connect the prodrug Pt (IV) scaffold to the carbohydrate. We have investigated complexes with different linker lengths, as well as acetyl protected and free derivatives. To the best of our knowledge, this study represents the first series of Pt (IV) glucosamine-conjugates functionalised at C2.
Collapse
Affiliation(s)
- Eoin Moynihan
- Department of Chemistry, Maynooth University, Maynooth, Ireland
| | | | - Monica Sandri
- Institute of Science, Technology and Sustainability for Ceramics (ISSMC)– National Research Council (CNR), Faenza, Italy
| | - Andrea Ruffini
- Institute of Science, Technology and Sustainability for Ceramics (ISSMC)– National Research Council (CNR), Faenza, Italy
| | - Silvia Panseri
- Institute of Science, Technology and Sustainability for Ceramics (ISSMC)– National Research Council (CNR), Faenza, Italy
| | - Trinidad Velasco-Torrijos
- Department of Chemistry, Maynooth University, Maynooth, Ireland
- Kathleen Londsdale for Human Health Research, Maynooth University, Maynooth, Ireland
| | - Monica Montesi
- Institute of Science, Technology and Sustainability for Ceramics (ISSMC)– National Research Council (CNR), Faenza, Italy
| | - Diego Montagner
- Department of Chemistry, Maynooth University, Maynooth, Ireland
- Kathleen Londsdale for Human Health Research, Maynooth University, Maynooth, Ireland
| |
Collapse
|
17
|
Ingersoll MA, Lutze RD, Pushpan CK, Kelmann RG, Liu H, May MT, Hunter WJ, He DZ, Teitz T. Dabrafenib protects from cisplatin-induced hearing loss in a clinically relevant mouse model. JCI Insight 2023; 8:e171140. [PMID: 37934596 PMCID: PMC10807719 DOI: 10.1172/jci.insight.171140] [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: 04/03/2023] [Accepted: 11/02/2023] [Indexed: 11/09/2023] Open
Abstract
The widely used chemotherapy cisplatin causes permanent hearing loss in 40%-60% of patients with cancer. One drug, sodium thiosulfate, is approved by the FDA for use in pediatric patients with localized solid tumors for preventing cisplatin-induced hearing loss, but more drugs are desperately needed. Here, we tested dabrafenib, an FDA-approved BRAF kinase inhibitor and anticancer drug, in a clinically relevant multidose cisplatin mouse model. The protective effects of dabrafenib, given orally twice daily with cisplatin, were determined by functional hearing tests and cochlear outer hair cell counts. Toxicity of the drug cotreatment was evaluated, and levels of phosphorylated ERK were measured. A dabrafenib dose of 3 mg/kg BW, twice daily, in mice, was determined to be the minimum effective dose, and it is equivalent to one-tenth of the daily FDA-approved dose for human cancer treatment. The levels of hearing protection acquired, 20-25 dB at the 3 frequencies tested, in both female and male mice, persisted for 4 months after completion of treatments. Moreover, dabrafenib exhibited a good in vivo therapeutic index (> 25), protected hearing in 2 mouse strains, and diminished cisplatin-induced weight loss. This study demonstrates that dabrafenib is a promising candidate drug for protection from cisplatin-induced hearing loss.
Collapse
Affiliation(s)
| | | | | | | | | | | | - William J. Hunter
- Department of Pathology, School of Medicine, Creighton University, Omaha, Nebraska, USA
| | | | - Tal Teitz
- Department of Pharmacology and Neuroscience
| |
Collapse
|
18
|
Bekhit AA, Beshay ON, Fawzy MA, Abdel-Hafez SMN, Batiha GES, Ataya FS, Fathy M. Curative Effect of AD-MSCs against Cisplatin-Induced Hepatotoxicity in Rats is Potentiated by Azilsartan: Targeting Oxidative Stress, MAPK, and Apoptosis Signaling Pathways. Stem Cells Int 2023; 2023:6767735. [PMID: 37908315 PMCID: PMC10615573 DOI: 10.1155/2023/6767735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 09/02/2023] [Accepted: 10/03/2023] [Indexed: 11/02/2023] Open
Abstract
Despite its clinical value, cisplatin (CISP) is complicated by marked hepatotoxicity via inducing oxidative stress, inflammatory, and apoptotic pathways. This study aims to explore the protective impact of azilsartan (AZIL), an antihypertensive drug, in addition to adipose tissue-derived mesenchymal stem cells (AD-MSCs) on CISP-induced hepatotoxicity. After characterization and labeling of AD-MSCs by PKH26 dye, 54 Wistar male albino rats were randomly divided into nine groups: I (CONT), II (AZIL.H), III (CISP), IV (CISP + AZIL.L), V (CISP + AZIL.H), VI (CISP + AD-MSCs), VII (CISP + AZIL.L + AD-MSCs), VIII (CISP + AZIL.H + AD-MSCs), and IX (CISP + VITA C). Serum alanine aminotransferase (ALT), alanine aminotransferase (AST), and albumin levels were determined. Assessment of reactive oxygen species, malondialdehyde, and glutathione contents, and superoxide dismutase activity and histopathological evaluations were done on hepatic tissue. Quantitative real-time PCR was utilized to estimate the expression of TNF-α and IL-6 genes. Cell homing of labeled AD-MSCs to the liver tissues was investigated. Hepatic expression of JNK1/2, ERK1/2, p38, Bax, Bcl-2, and cleaved caspase-3 proteins was investigated by western blot analysis. CISP elevated serum ALT and AST activities, reduced albumin level, and remarkably changed the hepatic architecture. It increased the expression TNF-α and IL-6 genes, raised the expression of JNK1/2, ERK1/2, p38, Bax, and cleaved caspase-3 proteins, and diminished the Bcl-2 protein. By contrast, treatment of animals with either AZIL or AD-MSCs dramatically reduced the effects of CISP injection. Moreover, treatment with combination therapy (AZIL.L or H + AD-MSCs) considerably mitigated all previously mentioned alterations superior to AZIL or AD-MSCs alone, which might be attributed to the AZIL-enhanced homing ability of AD-MSCs into the injured liver tissue. In conclusion, the present findings demonstrated that AZIL improves the hepatoprotective potential of AD-MSCs against CISP-induced hepatotoxicity by modulating oxidative stress, mitogen-activated protein kinase, and apoptotic pathways.
Collapse
Affiliation(s)
| | - Olivia N. Beshay
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | - Michael A. Fawzy
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | | | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, AlBeheira 22511, Egypt
| | - Farid S. Ataya
- Department of Biochemistry, College of Science, King Saud University, P.O. Box, 2455, Riyadh 11451, Saudi Arabia
| | - Moustafa Fathy
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
- Department of Regenerative Medicine, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| |
Collapse
|
19
|
Hwang DB, Seo Y, Lee E, Won DH, Kim C, Kang M, Jeon Y, Kim HS, Park JW, Yun JW. Diagnostic potential of serum miR-532-3p as a circulating biomarker for experimental intrinsic drug-induced liver injury by acetaminophen and cisplatin in rats. Food Chem Toxicol 2023:113890. [PMID: 37308052 DOI: 10.1016/j.fct.2023.113890] [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: 04/30/2023] [Revised: 06/07/2023] [Accepted: 06/09/2023] [Indexed: 06/14/2023]
Abstract
Evaluating tissue injury largely depends on serum biochemical analysis despite insufficient tissue specificity and low sensitivity. Therefore, attention has been paid to the potential of microRNAs (miRNAs) to overcome the limitations of the current diagnostic tools, as tissue-enriched miRNAs are detected in the blood upon tissue injury. First, using a cisplatin-injected rats, we screened a specific pattern of altered hepatic miRNAs and their target mRNAs. Subsequently, we identified novel liver-specific circulating miRNAs for drug-induced liver injury by comparing miRNA expression changes in organs and serum. RNA sequencing revealed that 32 hepatic miRNAs were differentially expressed (DE) in the cisplatin-treated group. Furthermore, among the 1217 targets predicted using miRDB on these DE-miRNAs, 153 hepatic genes involved in different liver function-related pathways and processes were found to be dysregulated by cisplatin. Next, comparative analyses of the liver, kidneys, and serum DE-miRNAs were conducted to select circulating miRNA biomarker candidates reflecting drug-induced liver injury. Finally, among the four liver-specific circulating miRNAs selected based on their expression patterns in tissue and serum, miR-532-3p was increased in the serum after cisplatin or acetaminophen administration. Our findings suggest that miR-532-3p is potential as a serum biomarker for identifying drug-induced liver injury, leading to the accurate diagnosis.
Collapse
Affiliation(s)
- Da-Bin Hwang
- Department of Biotechnology, The Catholic University of Korea, Bucheon, 14662, Republic of Korea
| | - Yoojin Seo
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Eunji Lee
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Dong-Hoon Won
- Department of Biotechnology, The Catholic University of Korea, Bucheon, 14662, Republic of Korea
| | - Changuk Kim
- Department of Biotechnology, The Catholic University of Korea, Bucheon, 14662, Republic of Korea
| | - MinHwa Kang
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Young Jeon
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hyung-Sik Kim
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Jun Won Park
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Jun-Won Yun
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
20
|
Park JE, Ahn CH, Lee HJ, Sim DY, Park SY, Kim B, Shim BS, Lee DY, Kim SH. Antioxidant-Based Preventive Effect of Phytochemicals on Anticancer Drug-Induced Hepatotoxicity. Antioxid Redox Signal 2023; 38:1101-1121. [PMID: 36242510 DOI: 10.1089/ars.2022.0144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Significance: Drug-induced liver injury (DILI) or hepatotoxicity has been a hot issue to overcome on the safety and physiological function of the liver, since it is known to have biochemical, cellular, immunological, and molecular alterations in the liver mainly induced by alcohol, chemicals, drugs, heavy metals, and genetic factors. Recently efficient therapeutic and preventive strategies by some phytochemicals are of interest, targeting oxidative stress-mediated hepatotoxicity alone or in combination with anticancer drugs. Recent Advances: To assess DILI, the variety of in vitro and in vivo animal models has been developed mainly by using carbon tetrachloride, d-galactosamine, acetaminophen, and lipopolysaccharide. Also, the mechanisms on hepatotoxicity by several drugs and herbs have been explored in detail. Recent studies reveal that antioxidants including vitamins and some phytochemicals were reported to prevent against DILI. Critical Issues: Antioxidant therapy with some phytochemicals is noteworthy, since oxidative stress is critically involved in DILI via production of chemically reactive oxygen species or metabolites, impairment of mitochondrial respiratory chain, and induction of redox cycling. Future Directions: For efficient antioxidant therapy, DILI susceptibility, Human Leukocyte Antigen genetic factors, biomarkers, and pathogenesis implicated in hepatotoxicity should be further explored in association with oxidative stress-mediated signaling, while more randomized preclinical and clinical trials are required with optimal safe doses of drugs and/or phytochemicals alone or in combination for efficient clinical practice along with the development of advanced DILI diagnostic tools.
Collapse
Affiliation(s)
- Ji Eon Park
- Cancer Molecular Target Herbal Research Laboratory, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Chi-Hoon Ahn
- Cancer Molecular Target Herbal Research Laboratory, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Hyo-Jung Lee
- Cancer Molecular Target Herbal Research Laboratory, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Deok Yong Sim
- Cancer Molecular Target Herbal Research Laboratory, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Su Yeon Park
- Cancer Molecular Target Herbal Research Laboratory, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Bonglee Kim
- Cancer Molecular Target Herbal Research Laboratory, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Bum Sang Shim
- Cancer Molecular Target Herbal Research Laboratory, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Dae Young Lee
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, Rural Development Administration (RDA), Eumseong, Republic of Korea
| | - Sung-Hoon Kim
- Cancer Molecular Target Herbal Research Laboratory, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
21
|
Mahmoud NA, Hassanein EHM, Bakhite EA, Shaltout ES, Sayed AM. Apocynin and its chitosan nanoparticles attenuated cisplatin-induced multiorgan failure: Synthesis, characterization, and biological evaluation. Life Sci 2023; 314:121313. [PMID: 36565813 DOI: 10.1016/j.lfs.2022.121313] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 12/08/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022]
Abstract
Cisplatin (CDDP) is an effective chemotherapeutic drug that has been used successfully in treating various tumors. Although its higher antineoplastic agent activity, CDDP exhibited severe side effects that limit its use. CDDP-induced toxicity is attributed to oxidative stress and inflammation. Apocynin (APO) is a bioactive phytochemical with potent antioxidant and anti-inflammatory properties. However, pharmaceutical experts face significant hurdles due to the limited bioavailability and quick elimination of APO. Therefore, we synthesized a chitosan (CTS)-based nano delivery system using the ionic gelation method to enhance APO bioactivity. CTS-APO-NPs were characterized using different physical and chemical approaches, including FTIR, XRD, TGA, Zeta-sizer, SEM, and TEM. In addition, the protective effect of CTS-APO-NPs against CDDP-induced nephrotoxicity, hepatotoxicity, and cardiotoxicity in rats was evaluated. CTS-APO-NPs restored serum biomarkers and antioxidants to their normal levels. Also, histopathological examination was used to assess the recovery of heart, kidney, and liver tissues. CTS-APO-NPs attenuated the oxidative stress mediated by Nrf2 activation while it dampened inflammation mediated by NF-κB suppression. CTS-APO-NPs is a potentially attractive target for more therapeutic trials.
Collapse
Affiliation(s)
- Nahed A Mahmoud
- Biochemistry Laboratory, Chemistry Department, Faculty of Science, Assiut University, Egypt
| | - Emad H M Hassanein
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Al-Azhar University, Egypt
| | - Etify A Bakhite
- Chemistry Department, Faculty of Science, Assiut University, Assiut 71516, Egypt
| | - Eman S Shaltout
- Department of Forensic Medicine & Clinical Toxicology, Faculty of Medicine, Assiut University, Assiut 71515, Egypt
| | - Ahmed M Sayed
- Biochemistry Laboratory, Chemistry Department, Faculty of Science, Assiut University, Egypt.
| |
Collapse
|
22
|
Mobasher MA, Ahmed EI, Hakami NY, Germoush MO, Awad NS, Khodeer DM. The Combined Effect of Licorice Extract and Bone Marrow Mesenchymal Stem Cells on Cisplatin-Induced Hepatocellular Damage in Rats. Metabolites 2023; 13:94. [PMID: 36677019 PMCID: PMC9861302 DOI: 10.3390/metabo13010094] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/12/2022] [Accepted: 12/20/2022] [Indexed: 01/11/2023] Open
Abstract
Drug-induced liver damage is a life-threatening disorder, and one major form of it is the hepatotoxicity induced by the drug cisplatin. In folk medicine, Licorice (Glycyrrhiza glabra (is used for detoxification and is believed to be a potent antioxidant. Currently, the magically self-renewable potential of bone marrow mesenchymal stem cells (BM-MSCs) has prompted us to explore their hepatoregenerative capability. The impact of G. glabra extract (GGE) and BM-MSCs alone and, in combination, on protecting against hepatotoxicity was tested on cisplatin-induced liver injury in rats. Hepatic damage, as revealed by liver histopathology and increased levels of serum aspartate aminotransferase (AST), alanine aminotransferase (ALT), alkaline phosphatase (ALP), and malondialdehyde (MDA), was elevated in rats by received 7 mg/kg of cisplatin intraperitoneally. The combination of GGE and BM-MSCs returned the enzyme levels to near the normal range. It also improved levels of liver superoxide dismutase (SOD) and glutathione (GSH) and reduced MDA levels. Additionally, it was found that when GGE and BM-MSCs were used together, they significantly downregulated caspase9 (Casp9), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB), and interleukin-1β (IL-1β), which are involved in severe proinflammatory and apoptotic signaling cascades in the liver. Moreover, combining GGE and BM-MSCs led to the normal result of hepatocytes in several examined liver histological sections. Therefore, our findings suggest that GGE may have protective effects against oxidative liver damage and the promising regenerative potential of BM-MSCs.
Collapse
Affiliation(s)
- Maysa A. Mobasher
- Department of Pathology, Biochemistry Division, College of Medicine, Jouf University, Sakaka 72388, Saudi Arabia
| | - Eman Ibrahim Ahmed
- Pharmacology and Therapeutics Department, College of Medicine, Jouf University, Sakaka 72346, Saudi Arabia
- Pharmacology Department, Faculty of Medicine, Fayoum University, Fayoum 63511, Egypt
| | - Nora Y. Hakami
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21423, Saudi Arabia
| | - Mousa O. Germoush
- Biology Department, College of Science, Jouf University, Sakaka 72388, Saudi Arabia
| | - Nabil S Awad
- Department of Genetics, Faculty of Agriculture and Natural Resources, Aswan University, Aswan 81528, Egypt
- College of Biotechnology, Misr University for Science and Technology, Giza 12563, Egypt
| | - Dina M. Khodeer
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
23
|
Ahmed RF, Nasr M, Abd Elbaset M, Hussein AF, Ahmed-Farid OAH, Shafee N, Shabana ME. Combating hematopoietic and hepatocellular abnormalities resulting from administration of cisplatin: Role of liver targeted glycyrrhetinic acid nanoliposomes loaded with amino acids. Pharm Dev Technol 2022; 27:925-941. [PMID: 36168910 DOI: 10.1080/10837450.2022.2129687] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
The effectiveness of cisplatin in cancer treatment renders its use vital to clinicians. However, the accompanying side effects as cachexia, emesis and liver damage necessitate the use of a dietary supplement which is capable of hindering such undesirable complications. The branched chain amino acids as well as glutamine and arginine have been proven to be effective nutritional co-adjuvant therapeutic agents. Furthermore, new pharmaceutical approaches encompass designing organ-targeted nanoformulations to increase the medicinal efficacy. Therefore, the aim of the present study was to investigate the beneficial effects of liver-targeted amino acids-loaded nanoliposomes in counteracting the adverse hematopoietic and hepatic complications associated with cisplatin. Results revealed the use of the combination of two nanoliposomal formulations (one loading leucine + isolecuine + valine, and the other loading glutamine and arginine) given orally at a dose of 200 mg/kg for twelve days was effective against cisplatin-induced toxicities represented by improvement in the complete blood picture parameters, decrease in the serum hepatic enzymes levels, amelioration of the hepatic oxidative stress and cellular energy imbalance along with reduction in the histopathological abnormalities. It can be concluded that amino acids loaded nanoliposomes could be considered a new strategy in preventing cisplatin's adverse effects.
Collapse
Affiliation(s)
- Rania F Ahmed
- Department of Pharmacology, Medical Research and Clinical studies Institute, National Research Centre, (ID: 60014618), Dokki, 12622, Giza, Egypt
| | - Maha Nasr
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Egypt
| | - Marawan Abd Elbaset
- Department of Pharmacology, Medical Research and Clinical studies Institute, National Research Centre, (ID: 60014618), Dokki, 12622, Giza, Egypt
| | - Alyaa F Hussein
- Department of Pharmacology, Medical Research and Clinical studies Institute, National Research Centre, (ID: 60014618), Dokki, 12622, Giza, Egypt
| | - Omar A H Ahmed-Farid
- Department of Physiology, National Organization for Drug Control and Research, 12553, Giza, Egypt
| | - Nermin Shafee
- Department of Pathology, Medical Research and Clinical studies Institute, National Research Centre, (ID: 60014618), Dokki, 12622, Giza, Egypt
| | - Marwa E Shabana
- Department of Pathology, Medical Research and Clinical studies Institute, National Research Centre, (ID: 60014618), Dokki, 12622, Giza, Egypt
| |
Collapse
|
24
|
Wang X, Wang R, Qiao Y, Li Y. Progress on the efficacy and mechanism of action of panax ginseng monomer saponins treat toxicity. Front Pharmacol 2022; 13:1022266. [PMID: 36199681 PMCID: PMC9527293 DOI: 10.3389/fphar.2022.1022266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 09/05/2022] [Indexed: 12/06/2022] Open
Abstract
As a traditional Chinese herbal medicine, Panax ginseng C. A. Meyer (PG) has preventive and therapeutic effects on various diseases. Ginsenosides are main active ingredients of PG and have good pharmacological effects. Due to the diversity of chemical structures and physicochemical properties of ginsenosides, Currently, related studies on PG monomer saponins are mainly focused on the cardiovascular system, nervous system, antidiabetic, and antitumor. There are few types of research on the toxin treatment, predominantly exogenous toxicity. PG and its monomer ginsenosides are undoubtedly a practical option for treating exogenous toxicity for drug-induced or metal-induced side effects such as nephrotoxicity, hepatotoxicity, cardiotoxicity, metal toxicity and other exogenous toxicity caused by drugs or metals. The mechanism focuses on antioxidant, anti-inflammatory, and anti-apoptotic, as well as modulation of signaling pathways. It summarized the therapeutic effects of ginseng monomer saponins on exogenous toxicity and demonstrated that ginsenosides could be used as potential drugs to treat exogenous toxicity and reduce drug toxicities.
Collapse
Affiliation(s)
- Xinyi Wang
- Institute of Special Animal and Plant Sciences of Chinese Academy of Agricultural Sciences, Changchun, China
| | - Rongcan Wang
- Institute of Special Animal and Plant Sciences of Chinese Academy of Agricultural Sciences, Changchun, China
| | - Yongfei Qiao
- Institute of Special Animal and Plant Sciences of Chinese Academy of Agricultural Sciences, Changchun, China
| | - Yali Li
- Institute of Special Animal and Plant Sciences of Chinese Academy of Agricultural Sciences, Changchun, China
- Jilin Provincial Key Laboratory of Traditional Chinese Medicinal Materials Cultivation and Propagation, Changchun, China
- *Correspondence: Yali Li,
| |
Collapse
|
25
|
Famurewa AC, Mukherjee AG, Wanjari UR, Sukumar A, Murali R, Renu K, Vellingiri B, Dey A, Valsala Gopalakrishnan A. Repurposing FDA-approved drugs against the toxicity of platinum-based anticancer drugs. Life Sci 2022; 305:120789. [PMID: 35817170 DOI: 10.1016/j.lfs.2022.120789] [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: 05/24/2022] [Revised: 07/02/2022] [Accepted: 07/05/2022] [Indexed: 11/17/2022]
Abstract
Platinum-based anticancer drugs (PADs), mainly cisplatin, carboplatin, and oxaliplatin, are widely used efficacious long-standing anticancer agents for treating several cancer types. However, clinicians worry about PAD chemotherapy and its induction of severe non-targeted organ toxicity. Compelling evidence has shown that toxicity of PAD on delicate body organs is associated with free radical generation, DNA impairment, endocrine and mitochondrial dysfunctions, oxidative inflammation, apoptosis, endoplasmic reticulum stress, and activation of regulator signaling proteins, cell cycle arrest, apoptosis, and pathways. The emerging trend is the repurposing of FDA-approved non-anticancer drugs (FNDs) for combating the side effects toxicity of PADs. Thus, this review chronicled the mechanistic preventive and therapeutic effects of FNDs against PAD organ toxicity in preclinical studies. FNDs are potential clinical drugs for the modulation of toxicity complications associated with PAD chemotherapy. Therefore, FNDs may be suggested as non-natural agent inhibitors of unpalatable side effects of PADs.
Collapse
Affiliation(s)
- Ademola C Famurewa
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, Alex Ekwueme Federal University, Ndufu-Alike lkwo, Nigeria.
| | - Anirban Goutam Mukherjee
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India
| | - Uddesh Ramesh Wanjari
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India
| | - Aarthi Sukumar
- Department of Integrative Biology, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India
| | - Reshma Murali
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India
| | - Kaviyarasi Renu
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600077, Tamil Nadu, India
| | - Balachandar Vellingiri
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641 046, Tamil Nadu, India
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata, West Bengal 700073, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India.
| |
Collapse
|
26
|
Talebpour Amiri F, Arzani S, Farzipour S, Hosseinimehr SJ. Radioprotective effects of gliclazide against irradiation-induced cardiotoxicity and lung injury through inhibiting oxidative stress. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:199. [PMID: 36071308 DOI: 10.1007/s12032-022-01803-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 07/18/2022] [Indexed: 10/14/2022]
Abstract
Radiotherapy is one of the main treatments for localized primary cancer in patients. Cardiotoxicity and lung injury are two of the main side effects of oxidative stress following radiotherapy in patients with thoracic region cancer. Gliclazide (GLZ) as an antihyperglycemic drug has antioxidant, anti-inflammatory, and anti-apoptotic activities. This study aimed to evaluate the effect of GLZ in cardiotoxicity and lung injury induced by irradiation (IR). In this experimental study, 64 mice were divided into eight groups: control, GLZ (5, 10, and 25 mg/kg), IR (6 Gy), and IR + GLZ (in three doses). GLZ was administrated for 8 consecutive successive days and mice were exposed with IR on the 9th day of study. On the 10th day of study, tissue biochemical assay and at 14th day of study, histopathological assay were performed to evaluate for cardiotoxicity and lung injury. The findings revealed that IR induces atypical features in heart and lung histostructure, and oxidative stress (an increase of MDA, PC levels, and decrease of GSH content) in these tissues. GLZ administration preserved heart and lung damages and improves oxidative stress markers in mice. Data have authenticated that GLZ could protect heart and lung histostructure against oxidative stress-induced injury through inhibiting oxidative stress.
Collapse
Affiliation(s)
- Fereshteh Talebpour Amiri
- Department of Anatomy, Faculty of Medicine, Molecular and Cell Biology Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Soroush Arzani
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran.,Student Research Committee, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Soghra Farzipour
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Guilan University of Medical Sciences, Rasht, Iran
| | - Seyed Jalal Hosseinimehr
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
27
|
Hassan SMA, Saeed AK, Rahim OO, Mahmood SAF. Alleviation of cisplatin-induced hepatotoxicity and nephrotoxicity by L-carnitine. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:897-903. [PMID: 36033956 PMCID: PMC9392569 DOI: 10.22038/ijbms.2022.65427.14395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 06/21/2022] [Indexed: 11/06/2022]
Abstract
Objectives To assess the protective effect of L-carnitine in reducing cisplatin toxicity via estimating biochemical tests, histomorphometric, and immunohistochemistry (IHC) of β-catenin and cyclin D. Materials and Methods Fifteen adult male rabbits were used in this study and allocated into 3 groups; Group 1 (Control negative), rabbits of this group were not given any treatment. In group 2, the animals were injected with cisplatin single-dose/per week. Group 3 rabbits were treated with Cisplatin+L-carnitine orally by gavage tube for 29 days. At the end of the experiments, blood samples from all rabbits were taken from the earlobe, and then the biochemical test was done, the kidney and tissue sections were prepared for both H& E and IHC for both β-catenin and cyclin D genes. Results Treatment with L-carnitine reduced the injury effect of cisplatin via a decline in serum creatinine, urea, bilirubin, GPT, GOP, and ALP significantly (P<0.05). Also, administration of LC attenuates the histopathologic abnormality in the kidney (15.71% vs 85.18%) and liver (score 3 vs 15 ) induced by cisplatin. L-carnitine elevates the expression of β-catenin and cyclin D in renal and hepatic parenchyma by diffuse, moderate-strong positivity vs cisplatin that showed local-weak staining. Conclusion These findings imply that L-carnitine, by its pleiotropic actions in activating Wnt signaling, alleviates cisplatin-induced renal and hepatic destruction. It might be a method of preventing cisplatin-related nephrotoxicity and hepatotoxicity.
Collapse
Affiliation(s)
- Snur Mohammad Amen Hassan
- Department of Anatomy and Histopathology, College of Veterinary Medicine, Sulaimani University, Sulaimani, 4601, KRG, Iraq,Corresponding author: Snur Mohammad Amen Hassan. Department of Anatomy and Histopathology, College of Veterinary Medicine, Sulaimani University, Sulaimani, 4601, KRG, Iraq. Tel/ Fax: +964-07701953823;
| | - Azad K Saeed
- Department of Anatomy and Histopathology, College of Veterinary Medicine, Sulaimani University, Sulaimani, 4601, KRG, Iraq
| | - Omed Omer Rahim
- Department of Food Science and Quality Control, Bakrajo Technical Institute, Sulaimani Polytechnic University, Sulaimani, 4601, KRG, Iraq
| | - Shler A F Mahmood
- Department of Microbiology, School of Medicine, Sulaimani University, Sulaimani, 4601, KRG, Iraq
| |
Collapse
|
28
|
Sadasivam N, Kim YJ, Radhakrishnan K, Kim DK. Oxidative Stress, Genomic Integrity, and Liver Diseases. Molecules 2022; 27:3159. [PMID: 35630636 PMCID: PMC9147071 DOI: 10.3390/molecules27103159] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 02/04/2023] Open
Abstract
Excess reactive oxygen species production and free radical formation can lead to oxidative stress that can damage cells, tissues, and organs. Cellular oxidative stress is defined as the imbalance between ROS production and antioxidants. This imbalance can lead to malfunction or structure modification of major cellular molecules such as lipids, proteins, and DNAs. During oxidative stress conditions, DNA and protein structure modifications can lead to various diseases. Various antioxidant-specific gene expression and signal transduction pathways are activated during oxidative stress to maintain homeostasis and to protect organs from oxidative injury and damage. The liver is more vulnerable to oxidative conditions than other organs. Antioxidants, antioxidant-specific enzymes, and the regulation of the antioxidant responsive element (ARE) genes can act against chronic oxidative stress in the liver. ARE-mediated genes can act as the target site for averting/preventing liver diseases caused by oxidative stress. Identification of these ARE genes as markers will enable the early detection of liver diseases caused by oxidative conditions and help develop new therapeutic interventions. This literature review is focused on antioxidant-specific gene expression upon oxidative stress, the factors responsible for hepatic oxidative stress, liver response to redox signaling, oxidative stress and redox signaling in various liver diseases, and future aspects.
Collapse
Affiliation(s)
- Nanthini Sadasivam
- Department of Integrative Food, Bioscience and Biotechnology, Chonnam National University, Gwangju 61186, Korea; (N.S.); (Y.-J.K.)
| | - Yu-Ji Kim
- Department of Integrative Food, Bioscience and Biotechnology, Chonnam National University, Gwangju 61186, Korea; (N.S.); (Y.-J.K.)
| | - Kamalakannan Radhakrishnan
- Clinical Vaccine R&D Center, Department of Microbiology, Combinatorial Tumor Immunotherapy MRC, Medical School, Chonnam National University, Gwangju 58128, Korea
| | - Don-Kyu Kim
- Department of Integrative Food, Bioscience and Biotechnology, Chonnam National University, Gwangju 61186, Korea; (N.S.); (Y.-J.K.)
| |
Collapse
|
29
|
Ferah Okkay I, Okkay U, Aydin IC, Bayram C, Ertugrul MS, Mendil AS, Hacimuftuoglu A. Centella asiatica extract protects against cisplatin-induced hepatotoxicity via targeting oxidative stress, inflammation, and apoptosis. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:33774-33784. [PMID: 35029831 DOI: 10.1007/s11356-022-18626-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 01/07/2022] [Indexed: 06/14/2023]
Abstract
This study was designed to investigate the protective effects of Centella asiatica (CA) on cisplatin-induced hepatotoxicity and to clarify the underlying mechanism by biochemical, molecular, immunohistochemical, and histopathological analyses. Rats were pre-treated with two doses of CA (100 and 200 mg/kg, p.o.) for 14 consecutive days. Then, on the 15th day, hepatotoxicity was induced by a single cisplatin injection (10 mg/kg i.p.). On the 18th day, the rats were euthanized. CA effectively alleviated cisplatin-induced hepatic injury via reduction in AST, ALT, and ALP enzymes and a decrease in oxidative stress (decreased MDA and ROS, and increased SOD, CAT, and GSH). CA also mitigated the inflammatory damage by the inhibition of TNF-α, IL-1β, and NF-κB. The liver expression of caspase-3 and Bax was downregulated, while Bcl-2 was upregulated. Moreover, immunohistochemical results confirmed the recovery with CA by downregulation of iNOS and 8-OHdG expression. These results showed that with its antioxidant, anti-inflammatory, and anti-apoptotic activities, CA could help alleviate the hepatotoxic effects of cisplatin chemotherapy.
Collapse
Affiliation(s)
- Irmak Ferah Okkay
- Department of Pharmacology, Faculty of Pharmacy, Ataturk University, 25100, Erzurum, Turkey.
| | - Ufuk Okkay
- Department of Medical Pharmacology, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Ismail Cagri Aydin
- Department of Pharmacology, Faculty of Pharmacy, Erzincan Binali Yildirim University, Erzincan, Turkey
| | - Cemil Bayram
- Department of Medical Pharmacology, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Muhammed Sait Ertugrul
- Department of Medical Pharmacology, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Ali Sefa Mendil
- Department of Pathology, Faculty of Veterinary Medicine, Erciyes University, Kayseri, Turkey
| | - Ahmet Hacimuftuoglu
- Department of Medical Pharmacology, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| |
Collapse
|
30
|
Gad El-Hak HN, Mahmoud HS, Ahmed EA, Elnegris HM, Aldayel TS, Abdelrazek HMA, Soliman MTA, El-Menyawy MAI. Methanolic Phoenix dactylifera L. Extract Ameliorates Cisplatin-Induced Hepatic Injury in Male Rats. Nutrients 2022; 14:1025. [PMID: 35268000 PMCID: PMC8912432 DOI: 10.3390/nu14051025] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 02/20/2022] [Accepted: 02/21/2022] [Indexed: 12/20/2022] Open
Abstract
This study investigated the ameliorative potential of methanolic date flesh extract (MDFE) against cisplatin-induced hepatic injury. Twenty male rats (weighing 180-200 g) were allocated into four groups: control; date flesh (DF) group (oral 600 mg/kg MDFE for 21 days); Cis group (7.5 mg/kg i.p. at day 16); and date flesh/cisplatin (DF/Cis) group (oral 600 mg/kg MDFE for 21 days and 7.5 mg/kg i.p. at day 16). Hepatic biochemical parameters in sera, and inflammatory and oxidant/antioxidant hepatic biomarkers were estimated. Hepatic histological changes and the immunohistochemistry of cyclooxygenase-2 (COX-2), nuclear factor kappa B (NF-κB), and alpha smooth muscle actin (α-SMA) were assessed. Pretreatment with MDFE decreased Cis-triggered liver biochemical parameters, oxidative stress, inflammatory biomarkers, and histological damage. Moreover, MDFE treatment reduced Cis-induced hepatic NF-κB, COX-2, and α-SMA protein expression. MDFE exerted a hepatoprotective effect when used concomitantly with Cis. Its effect was mediated via its antioxidant and anti-inflammatory ingredients.
Collapse
Affiliation(s)
- Heba Nageh Gad El-Hak
- Zoology Department, Faculty of Sciences, Suez Canal University, Ismailia 41522, Egypt;
| | - Hany Salah Mahmoud
- Center of Scientific Foundation for Experimental Studies and Research, Ismailia 41511, Egypt;
| | - Eman A. Ahmed
- Department of Pharmacology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt;
| | - Heba M. Elnegris
- Department of Histology and Cell Biology, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt;
- Department of Histology and Cell Biology, Faculty of Medicine, Badr University in Cairo, Cairo 11829, Egypt
| | - Tahany Saleh Aldayel
- Department of Physical Sport Sciences, College of Education, Princess Nourah Bint Abdulrahman University, Riyadh 11671, Saudi Arabia
| | - Heba M. A. Abdelrazek
- Department of Physiology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt;
| | - Mohamed T. A. Soliman
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 67614, Saudi Arabia;
| | | |
Collapse
|