1
|
Alzoubi KH, Rababa’h AM, Khabour OF, Nuseir F. Cilostazol protective effect on nedaplatin-induced genotoxicity in cultured human lymphocytes. Toxicol Rep 2025; 14:101928. [PMID: 39917038 PMCID: PMC11800108 DOI: 10.1016/j.toxrep.2025.101928] [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: 12/06/2024] [Revised: 01/18/2025] [Accepted: 01/24/2025] [Indexed: 02/09/2025] Open
Abstract
Background Nedaplatin has demonstrated remarkable efficacy in combating various malignancies. However, the administration of nedaplatin has been associated with the induction of DNA damage within normal cells. Cilostazol is a phosphodiesterase III inhibitor with an antioxidant mechanism that could protect cells from genotoxicity. We aimed to evaluate the genotoxic effect of nedaplatin on cultured human lymphocytes and the potential protective effect of cilostazol on chromosomal damage induced by nedaplatin. Methods The proposed nedaplatin's genotoxic effect was studied in vitro by evaluating the frequencies of sister chromatid exchanges (SCEs) in human cultured lymphocytes. Both the mitotic and proliferative indices (MI and PI, respectively) were used to assess the cytotoxic effects of nedaplatin. Results Nedaplatin significantly increased the frequency of SCEs compared to control and cilostazol-treated cells. The chromosomal injury induced by nedaplatin was significantly reduced by pretreatment of cells with cilostazol (P < 0.0001). Treating with cilostazol alone also reduced the frequency of SCEs, MI, and PI compared to the control group. Nedaplatin induced significant decreases in the MI and PI compared to the control group. Pretreatment with cilostazol partially debilitated the nedaplatin-induced changes in MI but not PI. Conclusion Cilostazol ameliorated the genotoxicity of nedaplatin in cultured human lymphocytes.
Collapse
Affiliation(s)
- Karem H. Alzoubi
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, The University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Abeer M. Rababa’h
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
- Division of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First St SW, Rochester, MN 55902, USA
| | - Omar F. Khabour
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, Jordan University of Science and Technology, Irbid, Jordan
| | - Fian Nuseir
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
2
|
Zaaba NE, Al-Salam S, Beegam S, Elzaki O, Aldhaheri F, Nemmar A, Ali BH, Nemmar A. Attenuation of cisplatin-induced acute kidney injury by sanguinarine: modulation of oxidative stress, inflammation, and cellular damage. Front Pharmacol 2025; 16:1567888. [PMID: 40242453 PMCID: PMC11999955 DOI: 10.3389/fphar.2025.1567888] [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: 01/28/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
Introduction Cisplatin (CP)-induced acute kidney injury (AKI) is a significant side effect of CP chemotherapy, driven by oxidative stress and inflammation. Sanguinarine (SANG), an alkaloid from the rhizomes of Sanguinaria canadensis and poppy-fumaria species, exhibits antioxidant and anti-inflammatory properties. This study examined SANG's effect on CP-induced AKI in mice and its underlying mechanisms. Methods Mice were orally administered 5 mg/kg SANG for 10 days. On the seventh day, they received a single intraperitoneal CP injection (20 mg/kg) and were sacrificed on the 11th day. Results SANG significantly improved CP-induced decreases in body weight, water intake, urine volume, relative kidney weight, creatinine clearance, albumin-to-creatinine ratio, and plasma urea and creatinine levels. It also reduced elevated plasma neutrophil gelatinase-associated lipocalin, kidney injury molecule-1, cystatin C, and adiponectin levels, as well as renal markers of inflammation and oxidative stress induced by CP administration. SANG normalized kidney mitochondrial dysfunction, DNA damage, and apoptosis caused by CP. It also inhibited the CP-induced increase in the expression of phosphorylated nuclear factor-κB and autophagy markers in the kidney. Histological analysis showed that SANG reduced acute tubular necrosis and intraluminal protein accumulation due to CP. Discussion In conclusion, SANG mitigated CP-induced AKI by reducing inflammation, oxidative stress, DNA damage, apoptosis, and autophagy. Pending more comprehensive pharmacological and toxicological assessments, SANG may be regarded as a potential therapeutic agent for mitigating CP-induced AKI.
Collapse
Affiliation(s)
- Nur Elena Zaaba
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Suhail Al-Salam
- Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Sumaya Beegam
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Ozaz Elzaki
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Fatima Aldhaheri
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Anas Nemmar
- College of Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - Badreldin H. Ali
- Emeritus Professor, Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Science, Sultan Qaboos University, Muscat, Oman
| | - Abderrahim Nemmar
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
3
|
Mossa FB, Bakry N, El-Sawi MR. Potential ameliorative effects of bilberry (Vaccinium myrtillus L.) fruit extract on cisplatin-induced reproductive damage in adult male albino rats. Clin Exp Reprod Med 2024; 51:192-204. [PMID: 39210717 PMCID: PMC11372312 DOI: 10.5653/cerm.2023.06380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 12/19/2023] [Indexed: 09/04/2024] Open
Abstract
OBJECTIVE Cisplatin (CP) is a widely used chemotherapeutic agent, but its severe side effects impact testicular function. We investigated the potential protective effects of bilberry extract against CP-induced testicular toxicity. METHODS Forty adult male albino rats were divided into four groups. Control animals received a single oral dose of 0.9% saline. Bilberry-treated rats received oral bilberry extract (200 mg/kg body weight [BW] dissolved in 1 mL of saline) daily for 10 consecutive days. CP-treated animals were administered a single intraperitoneal dose (7.5 mg/kg BW). Finally, a bilberry+CP group received oral bilberry extract (200 mg/kg BW) daily for 10 consecutive days, with one intraperitoneal dose of CP (7.5 mg/kg BW) on day 2. We assessed sperm count, motility, viability, and abnormalities, along with testis weight, testis weight-to-BW ratio, antioxidant activity, levels of oxidative stress markers (malondialdehyde [MDA] and hydrogen peroxide [H2O2]), sex hormones (follicle-stimulating hormone [FSH], luteinizing hormone [LH], and testosterone), and apoptotic and anti-apoptotic markers, and DNA damage. Testicular tissue underwent histopathological examination. RESULTS Among CP-treated rats, significantly lower values were observed for testis weight; testis weight-to-BW ratio; levels of FSH, LH, testosterone, superoxide dismutase, catalase, glutathione S-transferase, glutathione, and B-cell lymphoma 2; and sperm count, motility, and proportion of normal sperm. CP administration was associated with higher MDA, H2O2, p53, Bax, cytochrome c, caspase 9, and caspase 3 levels, along with elevated tail moment. However, bilberry extract administration significantly improved all altered parameters. CONCLUSION Bilberry treatment demonstrated protective effects and reduced CP-induced testicular toxicity via antioxidant activity and cytoprotection.
Collapse
Affiliation(s)
- Fatma B Mossa
- Physiology Division, Zoology Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Nadia Bakry
- Bone Marrow Transplantation and Cord Blood Unit, Mansoura University Children's Hospital, Mansoura, Egypt
| | - Mamdouh Rashad El-Sawi
- Physiology Division, Zoology Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| |
Collapse
|
4
|
Queiroz GCDAD, Dias FCR, Torres SMD, Pereira MDF, Morais DB, Silva WED, Silva Junior VAD. Bioconjugate based on cisplatin and bacterial exopolysaccharide with reduced side effects: A novel proposal for cancer treatment. J Trace Elem Med Biol 2024; 83:127374. [PMID: 38266419 DOI: 10.1016/j.jtemb.2023.127374] [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: 08/17/2023] [Revised: 12/12/2023] [Accepted: 12/20/2023] [Indexed: 01/26/2024]
Abstract
BACKGROUND In the search for alternatives that attenuate the toxicity associated to oncologic treatment with cisplatin (CDDP) and considering the potential health-beneficial properties of exopolysaccharides (EPS) produced by lactic acid bacteria, it was aimed on this study to evaluate the cytotoxic, toxicologic and antitumoral efficacy of a bioconjugate based on CDDP and EPS, on the experimental tumor of sarcoma 180. METHODS After the synthesis of the cis-[Pt(NH3)2(Cl)2] complex and of the conjugate containing Lactobacillus fermentum exopolysaccharide was tested both in vitro and in vivo for evaluating the acute toxicity. RESULTS The antitumoral study was performed using mice transplanted with sarcoma 180. The bioconjugate showed low to medium cytotoxicity for the cell lines tested, as well moderated acute toxicity. After determining the LD50, the following experimental groups were established for the antitumor assay: Control (NaCl 0,9%), CDDP (1 mg/kg), EPS and bioconjugate composition (200 mg/kg). The bioconjugate promoted a 38% regression in tumor mass when compared to the control, and a regression of 41% when compared to CDDP. Liver histopathological analysis revealed discrete alterations in animals treated with (CDDP + EPS) when compared to control. The bioconjugate also minimized changes in the renal parenchyma resulting from the tumor. CONCLUSION Our results indicate that when CDDP is associated with EPS, this composition was more biocompatible, showing itself as a potent chemotherapeutic agent and lower tissue toxicity.
Collapse
Affiliation(s)
- Gian Carlo D Angelo de Queiroz
- Programa de Pós-Graduação em Desenvolvimento e Inovação Tecnológica em Medicamentos, Universidade Federal Rural de Pernambuco, UFRPE, Recife, PE, Brazil
| | - Fernanda Carolina Ribeiro Dias
- Departamento de Medicina Veterinária, Universidade Federal Rural de Pernambuco, UFRPE, Recife, PE, Brazil; Department of Structural Biology, Federal University of Triangulo Mineiro, UFTM, Uberaba, MG, Brazil.
| | - Sandra Maria de Torres
- Departamento de Medicina Veterinária, Universidade Federal Rural de Pernambuco, UFRPE, Recife, PE, Brazil
| | | | - Danielle Barbosa Morais
- Departamento de Morfologia, Universidade Federal do Rio Grande do Norte, UFRN, Natal, RN, Brazil
| | - Wagner Eduardo da Silva
- Departamento de Química, Universidade Federal Rural de Pernambuco, UFRPE, Recife, PE, Brazil
| | | |
Collapse
|
5
|
Singh S. Antioxidant nanozymes as next-generation therapeutics to free radical-mediated inflammatory diseases: A comprehensive review. Int J Biol Macromol 2024; 260:129374. [PMID: 38242389 DOI: 10.1016/j.ijbiomac.2024.129374] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 12/30/2023] [Accepted: 01/08/2024] [Indexed: 01/21/2024]
Abstract
Recent developments in exploring the biological enzyme mimicking properties in nanozymes have opened a separate avenue, which provides a suitable alternative to the natural antioxidants and enzymes. Due to high and tunable catalytic activity, low cost of synthesis, easy surface modification, and good biocompatibility, nanozymes have garnered significant research interest globally. Several inorganic nanomaterials have been investigated to exhibit catalytic activities of some of the key natural enzymes, including superoxide dismutase (SOD), catalase, glutathione peroxidase, peroxidase, and oxidase, etc. These nanozymes are used for diverse biomedical applications including therapeutics, imaging, and biosensing in various cells/tissues and animal models. In particular, inflammation-related diseases are closely associated with reactive oxygen and reactive nitrogen species, and therefore effective antioxidants could be excellent therapeutics due to their free radical scavenging ability. Although biological enzymes and other artificial antioxidants could perform well in scavenging the reactive oxygen and nitrogen species, however, suffer from several drawbacks such as the requirement of strict physiological conditions for enzymatic activity, limited stability in the environment beyond their optimum pH and temperature, and high cost of synthesis, purification, and storage make then unattractive for broad-spectrum applications. Therefore, this review systematically and comprehensively presents the free radical-mediated evolution of various inflammatory diseases (inflammatory bowel disease, mammary gland fibrosis, and inflammation, acute injury of the liver and kidney, mammary fibrosis, and cerebral ischemic stroke reperfusion) and their mitigation by various antioxidant nanozymes in the biological system. The mechanism of free radical scavenging by antioxidant nanozymes under in vitro and in vivo experimental models and catalytic efficiency comparison with corresponding natural enzymes has also been presented.
Collapse
Affiliation(s)
- Sanjay Singh
- National Institute of Animal Biotechnology (NIAB), Opposite Journalist Colony, Near Gowlidoddy, Extended Q-City Road, Gachibowli, Hyderabad 500032, Telangana, India.
| |
Collapse
|
6
|
Abdel-Wahab WM, Daifalla NS, Essawy AE. L-methionine protects against nephrotoxicity induced by methotrexate through modulation of redox status and inflammation. Redox Rep 2023; 28:2270886. [PMID: 37931136 PMCID: PMC10629423 DOI: 10.1080/13510002.2023.2270886] [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] [Indexed: 11/08/2023] Open
Abstract
Objective: Methotrexate (MTX) is a drug used in the treatment of cancer and autoimmune disorders; however, its clinical use is limited because of serious side effects including renal toxicity. This study aimed to investigate the protective effect of Lmethionine (L-Met) on MTX toxicity in the kidneys of rats.Methods: Thirty male rats were divided equally into five groups: control (saline), Met400 (400 mg/kg L-Met), MTX (20 mg/kg MTX), MTX-Met300 (300 mg/kg L-Met and 20 mg/kg MTX), and MTX-Met400 (400 mg/kg L-Met and 20 mg/kg MTX). Rats were euthanized one day after the last dose administration (day 16) and serum and renal tissue samples were collected. Renal function and injury indices, oxidative stress/antioxidant indices and proinflammatory cytokines were evaluated.Results: The results showed that L-Met could effectively counteract the nephrotoxic effects of MTX, in a dose-related manner, by improving most of the tested parameters. Furthermore, the higher dose of L-Met was able to restore several parameters to normal levels. In addition, investigation of MTX-induced hematological changes revealed a corrective potential of L-Met.Conclusion: L-Met can be an effective adjuvant therapy to modulate renal toxicity associated with MTX because of its antioxidant and antiinflammatory effects.
Collapse
Affiliation(s)
- Wessam M. Abdel-Wahab
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria, Egypt
- Department of Basic Sciences, Deanship of Preparatory Year and Supporting Studies, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Nada S. Daifalla
- Department of Basic Sciences, Deanship of Preparatory Year and Supporting Studies, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Amina E. Essawy
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria, Egypt
| |
Collapse
|
7
|
Bershawy R, Hafez HS, El-Sakka SS, Hammad A, Soliman MH. The anticancer and anti-inflammatory activity screening of pyridazinone-based analogs against human epidermoid skin cancer with detailed mechanistic analyses. J Biomol Struct Dyn 2023; 42:12885-12899. [PMID: 37916672 DOI: 10.1080/07391102.2023.2273985] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 09/07/2023] [Indexed: 11/03/2023]
Abstract
3(2H)-Pyridazinone derivatives based on 4-biphenyl, naphtha-2-yl, pyridine, or piperidine moiety were synthesized and characterized using I-R and 1HNMR spectra. The activity and cytotoxicity of some synthesized compounds on the skin epidermoid cancer cell proliferation and progression were investigated. The pyridazine isomer with pyridine revealed a significant decrease in the level of nitric oxide p < 0.01 than the activity of caffeine phenecyl ester. The activity of the three active isomers recorded significant activity for their total antioxidant content that triggers their ability for the scavenging the oxygen free radicals significantly p < 0.01. Moreover, revealing the pharmaceutical activity of the isomers as anti-inflammatory agents, IL-6, IL10, and IL12 have been decreased by variable significant values. Additionally, the active isomers revealed variable actions on the skin cancer cell to induce apoptosis using annexin V-FITC/PI. Pyridine was the highest isomer to induce late apoptosis and necrosis for the skin cancer cells against the use of cisplatin. Importantly, Molecular modeling experiments including docking and dynamic simulations were done for the most active 3 analogs to explore the ligand binding and stability leading to exploring the structure-activity relationship with biological target PARP1 which showed a good binding propensity to pyridazine binding site which supports the in vitro data. In conclusion, the pyridazine moieties with piperdine, naphthayl, and pyridine have pharmacological activities against skin cancer epidermoid by triggering action in inhibition of the proliferation and progression with an up-regulated apoptotic mechanism that evades the emergence of cisplatin resistance among different cancer cells.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Rana Bershawy
- Department of Chemistry, Faculty of Science, Suez University, Suez, Egypt
| | - Hani S Hafez
- Department of Zoology, Faculty of Science, Suez University, Suez, Egypt
| | - Sahar S El-Sakka
- Department of Chemistry, Faculty of Science, Suez University, Suez, Egypt
| | - Ali Hammad
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Mohammed H Soliman
- Department of Chemistry, Faculty of Science, Suez University, Suez, Egypt
| |
Collapse
|
8
|
Yoshioka T, Goda M, Kanda M, Itobayashi S, Sugimoto Y, Izawa‐Ishizawa Y, Yagi K, Aizawa F, Miyata K, Niimura T, Hamano H, Sakurada T, Zamami Y, Ishizawa K. Valproic acid treatment attenuates cisplatin-induced kidney injury by suppressing proximal tubular cell damage. Clin Transl Sci 2023; 16:2369-2381. [PMID: 37700528 PMCID: PMC10651653 DOI: 10.1111/cts.13638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 09/14/2023] Open
Abstract
Cisplatin treatment is effective against several types of carcinomas. However, it frequently leads to kidney injury, which warrants effective prevention methods. Sodium valproic acid is a prophylactic drug candidate with a high potential for clinical application against cisplatin-induced kidney injury. Therefore, in this study, we aimed to elucidate the mechanism underlying the prophylactic effect of valproic acid on cisplatin-induced kidney injury in a mouse model and HK2 and PODO cells with cisplatin-induced toxicity. In the mouse model of cisplatin-induced kidney injury, various renal function parameters and tubular damage scores were worsened by cisplatin, but they were significantly improved upon combination with valproic acid. No difference was observed in cisplatin accumulation between the cisplatin-treated and valproic acid-treated groups in whole blood and the kidneys. The mRNA expression levels of proximal tubular damage markers, apoptosis markers, and inflammatory cytokines significantly increased in the cisplatin group 72 h after cisplatin administration but significantly decreased upon combination with valproic acid. In HK2 cells, a human proximal tubular cell line, the cisplatin-induced decrease in cell viability was significantly suppressed by co-treatment with valproic acid. Valproic acid may inhibit cisplatin-induced kidney injury by suppressing apoptosis, inflammatory responses, and glomerular damage throughout the kidneys by suppressing proximal tubular cell damage. However, prospective controlled trials need to evaluate these findings before their practical application.
Collapse
Affiliation(s)
- Toshihiko Yoshioka
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical SciencesTokushima UniversityTokushimaJapan
- Department of PharmacyTokushima University HospitalTokushimaJapan
| | - Mitsuhiro Goda
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical SciencesTokushima UniversityTokushimaJapan
- Department of PharmacyTokushima University HospitalTokushimaJapan
| | - Masaya Kanda
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical SciencesTokushima UniversityTokushimaJapan
- Department of PharmacyTokushima University HospitalTokushimaJapan
| | - Sayuri Itobayashi
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical SciencesTokushima UniversityTokushimaJapan
| | - Yugo Sugimoto
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical SciencesTokushima UniversityTokushimaJapan
| | - Yuki Izawa‐Ishizawa
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical SciencesTokushima UniversityTokushimaJapan
- Department of General MedicineTaoka HospitalTokushimaJapan
| | - Kenta Yagi
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical SciencesTokushima UniversityTokushimaJapan
- Clinical Research Center for Developmental TherapeuticsTokushima University HospitalTokushimaJapan
| | - Fuka Aizawa
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical SciencesTokushima UniversityTokushimaJapan
- Department of PharmacyTokushima University HospitalTokushimaJapan
| | - Koji Miyata
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical SciencesTokushima UniversityTokushimaJapan
| | - Takahiro Niimura
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical SciencesTokushima UniversityTokushimaJapan
- Clinical Research Center for Developmental TherapeuticsTokushima University HospitalTokushimaJapan
| | - Hirofumi Hamano
- Department of PharmacyOkayama University HospitalOkayamaJapan
| | - Takumi Sakurada
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical SciencesTokushima UniversityTokushimaJapan
- Department of PharmacyTokushima University HospitalTokushimaJapan
| | - Yoshito Zamami
- Department of PharmacyOkayama University HospitalOkayamaJapan
| | - Keisuke Ishizawa
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical SciencesTokushima UniversityTokushimaJapan
- Department of PharmacyTokushima University HospitalTokushimaJapan
- Clinical Research Center for Developmental TherapeuticsTokushima University HospitalTokushimaJapan
| |
Collapse
|
9
|
Samoylova NA, Gureev AP, Popov VN. Methylene Blue Induces Antioxidant Defense and Reparation of Mitochondrial DNA in a Nrf2-Dependent Manner during Cisplatin-Induced Renal Toxicity. Int J Mol Sci 2023; 24:ijms24076118. [PMID: 37047089 PMCID: PMC10094522 DOI: 10.3390/ijms24076118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
Cisplatin is a platinum-based cytostatic drug that is widely used for cancer treatment. Mitochondria and mtDNA are important targets for platinum-based cytostatics, which mediates its nephrotoxicity. It is important to develop therapeutic approaches to protect the kidneys from cisplatin during chemotherapy. We showed that the exposure of mitochondria to cisplatin increased the level of lipid peroxidation products in the in vitro experiment. Cisplatin caused strong damage to renal mtDNA, both in the in vivo and in vitro experiments. Cisplatin injections induced oxidative stress by depleting renal antioxidants at the transcriptome level but did not increase the rate of H2O2 production in isolated mitochondria. Methylene blue, on the contrary, induced mitochondrial H2O2 production. We supposed that methylene blue-induced H2O2 production led to activation of the Nrf2/ARE signaling pathway. The consequences of activation of this signaling pathway were manifested in an increase in the expression of some antioxidant genes, which likely caused a decrease in the amount of mtDNA damage. Methylene blue treatment induced an increase in the expression of genes that were involved in the base excision repair (BER) pathway: the main pathway for mtDNA reparation. It is known that the expression of these genes can also be regulated by the Nrf2/ARE signaling pathway. We can assume that the protective effect of methylene blue is related to the activation of Nrf2/ARE signaling pathways, which can activate the expression of genes related to antioxidant defense and mtDNA reparation. Thus, the protection of kidney mitochondria from cisplatin-induced damage using methylene blue can significantly expand its application in medicine.
Collapse
Affiliation(s)
- Natalia A Samoylova
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, 394018 Voronezh, Russia
| | - Artem P Gureev
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, 394018 Voronezh, Russia
- Laboratory of Metagenomics and Food Biotechnology, Voronezh State University of Engineering Technology, 394036 Voronezh, Russia
| | - Vasily N Popov
- Department of Genetics, Cytology and Bioengineering, Voronezh State University, 394018 Voronezh, Russia
- Laboratory of Metagenomics and Food Biotechnology, Voronezh State University of Engineering Technology, 394036 Voronezh, Russia
| |
Collapse
|
10
|
He H, Ge J, Yi S, Wang Y, Liu Y, Liu Y, Liu X. Ginkgolide A downregulates transient receptor potential (melastatin) 2 to protect cisplatin-induced acute kidney injury in rats through the TWEAK/Fn14 pathway: Ginkgolide A improve acute renal injury. Hum Exp Toxicol 2023; 42:9603271231200868. [PMID: 37715308 DOI: 10.1177/09603271231200868] [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] [Indexed: 09/17/2023]
Abstract
PURPOSE In order to seek effective drugs for treating cisplatin-induced acute renal injury and explore the corresponding potential mechanism. METHODS Mouse kidney injury model was established by intraperitoneal injection of 20 mg/kg cisplatin. The temporal expression of TRPM2 and the regulation of Ginkgolide A on its expression were analyzed by western blot. In order to perform the mechanical analysis, we used TRPM2-KO knockout mice. In this study, we evaluated the repair effect of GA on acute kidney injury through renal function factors, inflammatory factors and calcium and potassium content. Pathological injury and cell apoptosis were detected by H&E and TUNEL, respectively. RESULT Ginkgolide A inhibited inflammatory reaction and excessive oxidative stress, reduced renal function parameters, and improved pathological injury. Meanwhile, we also found that the repair effect of Ginkgolide A on renal injury is related to TRPM2, and Ginkgolide A downregulated TRPM2 expression and inactivated TWEAK/Fn14 pathway in cisplatin-induced renal injury model. We also found that inhibition of TWEAK/Fn14 pathway was more effective in TRPM2-KO mice than TRPM2-WT mice. CONCLUSION Ginkgolide A was the effective therapeutic drug for cisplatin-induced renal injury through acting on TRPM2, and TWEAK/Fn14 pathway was the downstream pathway of Ginkgolide A in acute renal injury, and Ginkgolide A inhibited TWEAK/Fn14 pathway in cisplatin-induced renal injury model.
Collapse
Affiliation(s)
- Haiyan He
- Department of Nephrology, Yantaishan Hospital, Yantai, China
| | - Jun Ge
- Department of Nephrology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Shaona Yi
- Department of Nephrology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Yuhong Wang
- Department of Nephrology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Ye Liu
- Department of Nephrology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Ying Liu
- Department of Pathology, Yantaishan Hospital, Yantai, China
| | - Xiaoming Liu
- Department of Nephrology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| |
Collapse
|
11
|
Al-Khdour MS, Khabour OF, Al-Eitan LN, Alzoubi KH. Genotoxicity of nedaplatin in cultured lymphocytes: modulation by vitamin E. Drug Chem Toxicol 2023; 46:176-180. [PMID: 34965829 DOI: 10.1080/01480545.2021.2015369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Nedaplatin is a chemotherapeutic agent used widely in cancer therapy. Nedaplatin has been shown to cause DNA damage to cells via the induction of oxidative stress. Vitamin E (Vit E) has an anti-mutagenic activity that can protect cells from DNA damaging agents. The objective of this study is to examine the genotoxic and cytotoxic effects of nedaplatin in human cultured lymphocytes. In addition, modulation of such effects by Vit E was also examined. The frequencies of sister chromatid exchange (SCE) and chromosomal aberrations (CAs) were used as an indicator for genotoxicity. The mitotic and proliferative indices were used to examine the cytotoxic effects of nedaplatin. The results showed that nedaplatin significantly elevated SCE and CA frequencies in human lymphocytes (p ˂ 0.01). The increases in the frequencies of SCE and CA caused by nedaplatin were lowered by pretreatment treatment with Vit E (p < 0.05). Nedaplatin significantly lowered mitotic index but Vit E pretreatment did not modulate this effect. These results suggest that Vit E has the potential to ameliorate the genotoxicity of nedaplatin in cultured lymphocytes.
Collapse
Affiliation(s)
- Muntaha S Al-Khdour
- Department of Applied Biological Sciences, Faculty of Science, Jordan University of Science and Technology, Irbid, Jordan
| | - Omar F Khabour
- Department of Medical Laboratory Sciences, Faculty of Medical Sciences, Jordan University of Science and Technology, Irbid, Jordan
| | - Laith N Al-Eitan
- Department of Applied Biological Sciences, Faculty of Science, Jordan University of Science and Technology, Irbid, Jordan
| | - Karem H Alzoubi
- Department of Pharmacy Practice and Pharmacotherapeutics, University of Sharjah, Sharjah, United Arab Emirates.,Department of Clinical Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
12
|
Amorim JM, Ribeiro de Souza LC, Lemos de Souza RA, da Silva Filha R, de Oliveira Silva J, de Almeida Araújo S, Tagliti CA, Simões E Silva AC, Castilho RO. Costus spiralis extract restores kidney function in cisplatin-induced nephrotoxicity model: Ethnopharmacological use, chemical and toxicological investigation. JOURNAL OF ETHNOPHARMACOLOGY 2022; 299:115510. [PMID: 35772602 DOI: 10.1016/j.jep.2022.115510] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 06/21/2022] [Accepted: 06/22/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Costus spiralis (Jacq.). Roscoe (Costaceae) is traditionally used in Brazil for the treatment of kidney diseases such as pyelonephritis, urethra inflammation, kidney stones, and inflammatory conditions. There are reports of its use by Brazilian Indians since the 17th century when it was known as "pacocatinga." Currently, the use of the Costus species in Brazil is widespread, which was evidenced by the inclusion of the genus in the Brazilian National List of Medicinal Plants of Interest to the Unified Health System (RENISUS). AIM OF THE STUDY This study aimed to confirm the ethnopharmacological use of Costus spiralis in the treatment of kidney diseases, toxicity study using animal models, and the phytochemistry of the species. MATERIALS AND METHODS The chemical profile of Costus spiralis leaves extract (CSLE) was obtained for the hydroethanolic extract by ultra-performance liquid chromatography coupled to a mass spectrometer and ultraviolet detector with diode array (UPLC-UV/DAD-ESI-MS). The acute oral toxicity of the extract was predicted using the neutral red uptake cytotoxicity assay. Wistar rats were used in a model in vivo for confirmation of acute oral toxicity (2000 mg/kg p.o. for 14 days.) and determination of the effect on a cisplatin-induced nephrotoxicity model. RESULTS The analysis by UPLC-UV/DAD-ESI-MS showed that the chemical composition of the extract is mostly di-glycosylated flavones of apigenin. In the extract were identified the flavones vicenin II and schaftoside. The quantification of total flavonoids by spectrometry showed 0.880%. CSLE proved to be safe for acute oral administration (2000 mg/kg) with an IC50 value of 222.9 μg/mL and predicted oral toxic dose of 523.82 μg/mL in a neutral red uptake cytotoxicity assay. The absence of death allows the classification of the extract in class 5 according to OECD 423 guidelines and therefore it can be considered as a high acute safety product, which is highly relevant, considering the wide popular use of the species. In the cisplatin-induced nephrotoxicity model, C. spiralis extract (5, 15, and 30 mg/kg) significantly improved renal function, reversing almost completely the effects on plasma creatinine levels and creatinine clearance (p < 0.001). CONCLUSIONS This study demonstrates that oral administration of Costus spiralis extract leaves is safe and effective in restoring the renal function in rats in a cisplatin-induced nephrotoxicity. It is suggested that the observed activity is related to the flavonoids present. This hypothesis should be confirmed, and the participation of other secondary metabolites should be investigated in the future.
Collapse
Affiliation(s)
- Juliana Mendes Amorim
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Faculdade de Farmácia, Avenida Antônio Carlos 6627, Campus Pampulha, 31.270-901, Belo Horizonte, MG, Brazil.
| | - Larissa Camila Ribeiro de Souza
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Faculdade de Farmácia, Avenida Antônio Carlos 6627, Campus Pampulha, 31.270-901, Belo Horizonte, MG, Brazil.
| | - Rebecca Almeida Lemos de Souza
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Faculdade de Farmácia, Avenida Antônio Carlos 6627, Campus Pampulha, 31.270-901, Belo Horizonte, MG, Brazil.
| | - Roberta da Silva Filha
- Universidade Federal de Minas Gerais, Faculdade de Medicina- Avenida Professor Alfredo Balena, 190, Campus Sáude, 30.130-190, Belo Horizonte, MG, Brazil.
| | - Juliana de Oliveira Silva
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Faculdade de Farmácia, Avenida Antônio Carlos 6627, Campus Pampulha, 31.270-901, Belo Horizonte, MG, Brazil.
| | - Stanley de Almeida Araújo
- Instituto de Nefrologia - Av. Bernardo Monteiro, 971, Funcionários, 30150-281, Belo Horizonte, MG, Brazil.
| | - Carlos Alberto Tagliti
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Faculdade de Farmácia, Avenida Antônio Carlos 6627, Campus Pampulha, 31.270-901, Belo Horizonte, MG, Brazil.
| | - Ana Cristina Simões E Silva
- Universidade Federal de Minas Gerais, Faculdade de Medicina- Avenida Professor Alfredo Balena, 190, Campus Sáude, 30.130-190, Belo Horizonte, MG, Brazil.
| | - Rachel Oliveira Castilho
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Faculdade de Farmácia, Avenida Antônio Carlos 6627, Campus Pampulha, 31.270-901, Belo Horizonte, MG, Brazil; Consórcio Acadêmico Brasileiro de Saúde Integrativa, CABSIN, Brazil.
| |
Collapse
|
13
|
Layos L, Martínez-Balibrea E, Ruiz de Porras V. Curcumin: A Novel Way to Improve Quality of Life for Colorectal Cancer Patients? Int J Mol Sci 2022; 23:ijms232214058. [PMID: 36430537 PMCID: PMC9695864 DOI: 10.3390/ijms232214058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/07/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer in men and the second most common in women. Treatment of metastatic CRC consists of highly toxic chemotherapeutic drug combinations that often negatively affect patient quality of life (QoL). Moreover, chemotherapy-induced toxicity and chemotherapy resistance are among the most important factors limiting cancer treatment and can lead to the interruption or discontinuation of potentially effective therapy. Several preclinical studies have demonstrated that curcumin acts through multiple cellular pathways and possesses both anti-cancer properties against CRC and the capacity to mitigate chemotherapy-related side effects and overcome drug resistance. In this review article, we suggest that the addition of curcumin to the standard chemotherapeutic treatment for metastatic CRC could reduce associated side-effects and overcome chemotherapy resistance, thereby improving patient QoL.
Collapse
Affiliation(s)
- Laura Layos
- Medical Oncology Department, Catalan Institute of Oncology, Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain
- Catalan Institute of Oncology, Badalona Applied Research Group in Oncology (B·ARGO), Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain
- Germans Trias i Pujol Research Institute (IGTP), Ctra. Can Ruti-Camí de les escoles s/n, 08916 Badalona, Spain
| | - Eva Martínez-Balibrea
- Germans Trias i Pujol Research Institute (IGTP), Ctra. Can Ruti-Camí de les escoles s/n, 08916 Badalona, Spain
- ProCURE Program, Catalan Institute of Oncology, Ctra. Can Ruti-Camí de les escoles s/n, 08916 Badalona, Spain
| | - Vicenç Ruiz de Porras
- Catalan Institute of Oncology, Badalona Applied Research Group in Oncology (B·ARGO), Ctra. Can Ruti-Camí de les Escoles s/n, 08916 Badalona, Spain
- Germans Trias i Pujol Research Institute (IGTP), Ctra. Can Ruti-Camí de les escoles s/n, 08916 Badalona, Spain
- Correspondence: ; Tel.: +34-(93)-5546301
| |
Collapse
|
14
|
ElBakary NM, Hagag SA, Ismail MA, El-Sayed WM. New thiophene derivative augments the antitumor activity of γ-irradiation against colorectal cancer in mice via anti-inflammatory and pro-apoptotic pathways. Discov Oncol 2022; 13:119. [PMID: 36326938 PMCID: PMC9633918 DOI: 10.1007/s12672-022-00583-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 10/17/2022] [Indexed: 04/17/2023] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is one of the most common types of cancer worldwide and the second cause of cancer-related deaths. It usually starts as an inflammation that progresses to adenocarcinoma. The goal of the present study was to investigate the antitumor efficacy of a new thiophene derivative against CRC in mice and explore the possible associated molecular pathways. The potential of this thiophene derivative to sensitize the CRC tumor tissue to a low dose of gamma irradiation was also investigated. METHODS Adult male mice were divided into seven groups; control, group treated with dimethylhydrazine (DMH) for the induction of CRC. The DMH-group was further divided into six groups and treated with either cisplatin, thiophene derivative, γ-irradiation, cisplatin + γ-irradiation, thiophene derivative + γ-irradiation, or left untreated. RESULTS DMH induced CRC as evidenced by the macroscopic examination of colon tissues and histopathology, and elevated the activities of cyclooxygenase2 (COX2) and nitric oxide synthase (iNOS). DMH also elevated kirsten rat sarcoma (KRAS) and downregulated the peroxisome proliferator activated receptor (PPARγ) as shown by RT-PCR and Western blotting. DMH exerted anti-apoptotic activity by reducing the expression of phosphorylated p53 and cleaved caspase3 at the gene and protein levels. The flow cytometry analysis showed that DMH elevated the necrosis and reduced the apoptosis compared to the other groups. The colon tissue from DMH-treated mice showed hyperplasia, aberrant crypt foci, loss of cell polarity, typical CRC of grade 4 with lymphocytes and macrophages infiltrating mucosa, muscularis mucosa, and submucosa score 3. Treatment with thiophene derivative or γ-irradiation ameliorated most of these deleterious effects of DMH. The concomitant action of thiophene derivative + γ-irradiation was typified by the better amelioration of tumor incidence and multiplicity, iNOS, PPARγ, p53, caspase 3, and histopathology of colon. CONCLUSION Taken together, the new thiophene derivative is a promising therapeutic candidate for treatment of colorectal cancer in mice. It also sensitizes the CRC tumor to the ionizing radiation through anti-inflammatory and pro-apoptotic pathways.
Collapse
Affiliation(s)
- Nermeen M ElBakary
- Radiation Biology Department, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Nasr City, Cairo, Egypt
| | - Sanaa A Hagag
- Radiation Biology Department, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Nasr City, Cairo, Egypt
| | - Mohamed A Ismail
- Department of Chemistry, Faculty of Science, Mansoura University, Mansoura, 35516, Egypt
| | - Wael M El-Sayed
- Department of Zoology, Faculty of Science, University of Ain Shams, Abbassia, Cairo, 11566, Egypt.
| |
Collapse
|
15
|
Gilani SJ, Bin-Jumah MN, Al-Abbasi FA, Nadeem MS, Alzarea SI, Ahmed MM, Sayyed N, Kazmi I. Rosinidin Protects against Cisplatin-Induced Nephrotoxicity via Subsiding Proinflammatory and Oxidative Stress Biomarkers in Rats. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:9719. [PMID: 35955076 PMCID: PMC9368304 DOI: 10.3390/ijerph19159719] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/30/2022] [Accepted: 07/31/2022] [Indexed: 05/28/2023]
Abstract
BACKGROUND Rosinidin is a flavonoid anthocyanin pigmentation found in shrub flowers such as Catharanthus roseus and Primula rosea. The molecular docking studies predicted that rosinidin has adequate structural competency, making it a viable medicinal candidate for the treatment of a wide range of disorders. The current study intends to assess rosinidin nephroprotective efficacy against nephrotoxicity induced by cisplatin in rats. MATERIALS AND METHODS Oral acute toxicity tests of rosinidin were conducted to assess potential toxicity in animals, and it was shown to be safe. The nephroprotective effect of rosinidin 10, and 20 mg/kg were tested in rats for 25 days with concurrent administration of cisplatin. Several biochemical parameters were measured to support enzymatic and non-enzymatic oxidative stress such as superoxide dismutase (SOD), malondialdehyde (MDA), and glutathione peroxidase (GSH). Likewise, changes in several non-protein-nitrogenous components and blood chemistry parameters were made to support the theory linked with the pathogenesis of chemical-induced nephrotoxicity. RESULTS Cisplatin caused significant changes in biochemical, enzymatic, and blood chemistry, which rosinidin efficiently controlled. CONCLUSIONS The present investigation linked rosinidin with nephroprotective efficacy in experimental models.
Collapse
Affiliation(s)
- Sadaf Jamal Gilani
- Department of Basic Health Sciences, Preparatory Year, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia
| | - May Nasser Bin-Jumah
- Biology Department, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia
- Environment and Biomaterial Unit, Health Sciences Research Center, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia
- Saudi Society for Applied Science, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia
| | - Fahad A. Al-Abbasi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Muhammad Shahid Nadeem
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Sami I. Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka 72341, Saudi Arabia
| | - Mohammed Muqtader Ahmed
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Nadeem Sayyed
- Glocal School of Pharmacy, Glocal University, Saharanpur 247121, India
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
16
|
Elbeltagy A, Mohamed G, Akeel M, Abdelaziz K, Elbakry K, Elsayed A. Modulatory role of garlic ( Allium sativum) extract against cisplatin- induced nephrotoxicity in female albino rats and their offspring. F1000Res 2022; 11:504. [PMID: 37547786 PMCID: PMC10403747 DOI: 10.12688/f1000research.111293.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/19/2022] [Indexed: 08/08/2023] Open
Abstract
Background: Cisplatin (CP) is one of the chemotherapeutic drugs widely utilized in the treatment of several malignancies. However, recently; its use has been limited because of its hazardous health drawbacks. Previous researches confirmed that CP has severe deleterious side effects on pregnant mothers and their fetuses. Garlic ( Allium sativum) extract has been claimed to exhibit potent antioxidative and free radical scavenging abilities. Aim: This work is mainly designed to evaluate the potential therapeutic role of garlic extract against CP-induced nephrotoxicity in pregnant rats and their offspring. Methods: 24 pregnant rats were used in the current study. They were randomly allocated into four groups (n=6): control, garlic, CP, and CP + garlic group. At the end of the weaning period, the mothers and the offsprings of all groups were sacrificed, the kidneys were immediately excised, and processed for histological and biochemical investigations. Also, blood samples were withdrawn and processed for estimation of the assigned biochemical parameters. Results: The renal histological sections from CP-treated mother rats displayed pronounced histopathological lesions however, their offspring showed mild renal histopathological lesions if compared with those of their mothers. The levels of renal tissue Superoxide dismutase, catalase, and glutathione peroxidase enzymes were significantly decreased. On the contrary, the levels of malondialdehyde, serum urea, and creatinine were significantly increased in CP-treated mother rats and their offspring as compared with control. The percentage value of caspase 3 activity was markedly elevated in the renal tissues of CP-treated mother rats and their offspring compared to the control group. Supplementation of garlic extract to the CP treated rats; the overall histological lesions, as well as biochemical parameters, were restored nearly to the control ones. It is concluded that garlic ( Allium sativum) extract has a powerful ameliorative role against CP-induced nephrotoxicity in pregnant rats and their offspring.
Collapse
Affiliation(s)
| | - Gamal Mohamed
- Department of Human Anatomy, , Faculty of Medicine, Jazan University, Jazan, KSA, Jazan, 45142, Saudi Arabia
| | - Mohammed Akeel
- Department of Human Anatomy, , Faculty of Medicine, Jazan University, Jazan, KSA, Jazan, 45142, Saudi Arabia
| | - Karoline Abdelaziz
- Zoology, Damanhour University Faculty of Science, Damanhour, 22511, Egypt
| | - Kadry Elbakry
- Zoology, Faculty of Science, Damietta, University, Damietta, Egypt, Damietta, 34611, Egypt
| | - Ahmed Elsayed
- Zoology, Damanhour University Faculty of Science, Damanhour, 22511, Egypt
| |
Collapse
|
17
|
Abdel-Rahman Mohamed A, Khater SI, Metwally MMM, Bin Emran T, Nassan MA, Abd El-Emam MM, Mostafa-Hedeab G, El-Shetry ES. TGF-β1, NAG-1, and antioxidant enzymes expression alterations in Cisplatin-induced nephrotoxicity in a rat model: Comparative modulating role of Melatonin, Vit. E and Ozone. Gene 2022; 820:146293. [PMID: 35143943 DOI: 10.1016/j.gene.2022.146293] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 01/23/2022] [Accepted: 02/03/2022] [Indexed: 12/24/2022]
Abstract
Cisplatin (CP) is an anticancer medication that is commonly used to treat solid tumors. Its use is, however, dose-restricted due to nephrotoxicity. We planned to compare the nephroprotective effects of three major compounds, including melatonin (MN), Ozone, or vitamin E, against the CP-induced renal damage in rats. CP was given once intraperitoneally (10 mg/kg,) eliciting acute kidney injury as assured by several adverse histological changes; glomerulopathy, tubulopathy, and vasculopathy, an inflammatory response including elevated TNF-α, IL-6, and IL-1β. Furthermore, biochemical alterations including, elevated plasma levels of urea, uric acid, creatinine, phosphorous, decreased plasma calcium levels, and gene expression abnormalities; upregulation of N-acetyl-β-d-glucosaminidase (NAG) and Transforming growth factor-β1 (TGF-β1), downregulation of CAT and SOD. Concurrent supplementation with either MN (10 mg/kg per os) or Ozone (1.1 mg/kg ip) and Vit E given by oral gavage (1 g/kg) for five consecutive days prior to CP injection and five days afterward displayed variable significant nephroprotective effects by mitigating the pro-inflammatory secretion, augmenting antioxidant competence, and modulating the gene expression in the renal tissue. The obtained biochemical, histological, and gene expression data suggested that MN had foremost rescue effects followed by Ozone then Vit E. MN's ameliorative effect was augmented in many indices including TNF-α, IL-6 , IL1-β, uric acid, creatinine, sNGAL and GGT, more than observed in Ozone, and Vit E therapy. A combination of these medications is expected to be more useful in relieving the damaging renal effects of CP given to cancer patients, pending further toxicological and pharmacological research.
Collapse
Affiliation(s)
| | - Safaa I Khater
- Department of Biochemistry, Faculty of Veterinary Medicine, Zagazig University, 4511, Egypt
| | - Mohamed M M Metwally
- Department of Pathology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh
| | - Mohamed A Nassan
- Department of Clinical Laboratory Sciences, Turabah University College, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Mahran M Abd El-Emam
- Department of Biochemistry, Faculty of Veterinary Medicine, Zagazig University, 4511, Egypt
| | - Gomaa Mostafa-Hedeab
- Pharmacology department & Health Research Unit, Medical College, Jouf University, Saudi Arabia; Pharmacology department, Faculty of Medicine, Beni-Suef University, Egypt
| | - Eman S El-Shetry
- Department of Human Anatomy and Embryology, Faculty of Medicine, Zagazig University, Zagazig 44511, Egypt
| |
Collapse
|
18
|
Kanda M, Goda M, Maegawa A, Yoshioka T, Yoshida A, Miyata K, Aizawa F, Niimura T, Hamano H, Okada N, Sakurada T, Chuma M, Yagi K, Izawa-Ishizawa Y, Yanagawa H, Zamami Y, Ishizawa K. Discovery of preventive drugs for cisplatin-induced acute kidney injury using big data analysis. Clin Transl Sci 2022; 15:1664-1675. [PMID: 35445533 PMCID: PMC9283743 DOI: 10.1111/cts.13282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/21/2022] [Accepted: 03/30/2022] [Indexed: 11/30/2022] Open
Abstract
Cisplatin is effective against many types of carcinoma. However, a high rate of renal damage is a clinical problem. Thus, there is a need to establish a method to prevent it. Although various compounds have been reported to be effective against cisplatin-induced renal injury, there are no examples of their clinical application. Therefore, we attempted to search for prophylactic agents with a high potential for clinical application. We used Cascade Eye to identify genes that are altered during cisplatin-induced renal injury, Library of Integrated Network-based Cellular Signatures (LINCS) to identify drugs that inhibit changes in gene expression, and a large database of spontaneous adverse drug reaction reports to identify drugs that could prevent cisplatin-induced kidney injury in clinical practice. In total, 10 candidate drugs were identified. Using the US FDA Adverse Event Reporting System (FAERS), we identified drugs that reduce cisplatin-induced kidney injury. Fenofibrate was selected as a candidate drug to prevent cisplatin-induced kidney injury based on the FAERS analysis. A model was used to evaluate the efficacy of fenofibrate against cisplatin-induced renal injury. Studies using HK2 cells and mouse models showed that fenofibrate significantly inhibited cisplatin-induced renal injury but did not inhibit the antitumor effect of cisplatin. Fenofibrate is a candidate prophylactic drug with high clinical applicability for cisplatin-induced renal injury. Analysis of data from multiple big databases will improve the search for novel prophylactic drugs with high clinical applicability. For the practical application of these findings, evaluation in prospective controlled trials is necessary.
Collapse
Affiliation(s)
- Masaya Kanda
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan.,Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
| | - Mitsuhiro Goda
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan.,Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan.,Clinical Research Center for Developmental Therapeutics, Tokushima University Hospital, Tokushima, Japan
| | - Akiko Maegawa
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Toshihiko Yoshioka
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan.,Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
| | - Ami Yoshida
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Koji Miyata
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Fuka Aizawa
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan.,Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
| | - Takahiro Niimura
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan.,Clinical Research Center for Developmental Therapeutics, Tokushima University Hospital, Tokushima, Japan
| | - Hirofumi Hamano
- Department of Pharmacy, Okayama University Hospital, Okayama, Japan
| | - Naoto Okada
- Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
| | - Takumi Sakurada
- Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
| | - Masayuki Chuma
- Department of Pharmacy, Asahikawa Medical University Hospital, Hokkaido, Japan
| | - Kenta Yagi
- Clinical Research Center for Developmental Therapeutics, Tokushima University Hospital, Tokushima, Japan
| | - Yuki Izawa-Ishizawa
- Department of Pharmacology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Hiroaki Yanagawa
- Clinical Research Center for Developmental Therapeutics, Tokushima University Hospital, Tokushima, Japan
| | - Yoshito Zamami
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan.,Department of Pharmacy, Okayama University Hospital, Okayama, Japan
| | - Keisuke Ishizawa
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan.,Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan.,Clinical Research Center for Developmental Therapeutics, Tokushima University Hospital, Tokushima, Japan
| |
Collapse
|
19
|
Abdel-Rasol MA, El-Beih NM, Yahya SS, El-Sayed WM. The Antitumor Activity of Ginger against Colorectal Cancer Induced by Dimethylhydrazine in Rats. Anticancer Agents Med Chem 2022; 22:1601-1610. [PMID: 34477526 DOI: 10.2174/1871520621666210903112813] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 07/07/2021] [Accepted: 07/12/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND Bowl or colorectal cancer (CRC) is the third most common type of cancer with about two million new cases every year. CRC is the second leading cause of cancer related mortalities. OBJECTIVE The study aims to evaluate the anticancer activity of ethanolic Ginger Extract (GE) in HCT-116 colon cells and colorectal tumors induced by dimethylhydrazine (DMH). METHODS The antiproliferative activity was measured by MTT assay and the gene expression was assessed by q-RTPCR. For the antitumor study, rats were divided into five groups in random; control, group two was orally treated with 300 mg/kg of GE for 21 weeks, group three was s.c. injected with DMH (20 mg/kg) for 9 weeks, and groups four and five were treated with DMH and then treated with cisplatin (2.5 mg/kg, i.p) or GE, respectively, for 21 weeks. RESULTS GE had a significant antiproliferative activity with IC50~ 12.5 µg/ml. GE induced both extrinsic and intrinsic apoptotic pathways. GE induced the expression of FasL, TRAIL, p53, and caspase-8 and downregulated Bcl-2 and survivin genes. Treatment of rats with DMH resulted in 100% tumor incidence and 2.3 tumors/rat. DMH significantly elevated the serum ALT, urea, and creatinine and significantly decreased the body weight gain. DMH also caused significant reductions in the hepatic GSH level, and the activities of catalase, SOD, GST, and GR in the liver as well as the renal GSH content and γ-GT activity. The colon from rats insulted with DMH showed adenomatous polyps with polymorphism and mitosis. The mucosa and submucosa were infested with inflammatory cells while serosa and muscularis were devoid from these cells. However, the muscularis was infiltrated with cystic formation, anaplastic changes, and hemorrhage. GE was able to alleviate all the previous deleterious effects of DMH and it was superior to cisplatin in its ameliorative effects. It did so without eliciting hepatotoxicity or nephrotoxicity which were shown in the group treated with DMH and cisplatin. CONCLUSION This study proved that the antitumor activity of GE against the DMH induced-CRC is superior to cisplatin. GE was also safer than cisplatin and did not elicit hepatotoxicity or nephrotoxicity. GE induced apoptosis and has carcinostatic activity.
Collapse
Affiliation(s)
- Mohammed A Abdel-Rasol
- Department of Zoology, Faculty of Science, University of Ain Shams, Abbassia 11566, Cairo, Egypt
| | - Nadia M El-Beih
- Department of Zoology, Faculty of Science, University of Ain Shams, Abbassia 11566, Cairo, Egypt
| | - Shaymaa S Yahya
- Department of Hormones, National Research Centre, Dokki 12622, Giza, Egypt
| | - Wael M El-Sayed
- Department of Zoology, Faculty of Science, University of Ain Shams, Abbassia 11566, Cairo, Egypt
| |
Collapse
|
20
|
Eslamifar Z, Moridnia A, Sabbagh S, Ghaffaripour R, Jafaripour L, Behzadifard M. Ameliorative Effects of Gallic Acid on Cisplatin-Induced Nephrotoxicity in Rat Variations of Biochemistry, Histopathology, and Gene Expression. BIOMED RESEARCH INTERNATIONAL 2021; 2021:2195238. [PMID: 34746299 PMCID: PMC8564201 DOI: 10.1155/2021/2195238] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/18/2021] [Accepted: 09/21/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND Cisplatin is a powerful chemotherapeutic drug mainly used in the treatment of solid tumors. Aggregation of the drug in renal proximal tubule cells causes nephrotoxicity and renal failure. Investigations showed nephrotoxicity as Cisplatin's dose-limiting side effect. One of the Cisplatin toxicity mechanisms is generation of reactive oxygen species, which leads to oxidative stress and renal damage. The purpose of this study was evaluation of the modulating effects of Gallic acid on Cisplatin-induced variations including Caspase-3 and Clusterin expression and histopathological and biochemical parameters in adult male Wistar rats. METHOD Rats were kept under standard condition of temperature, light, and humidity. The animals were divided into 4 groups: GpI: control group (received distilled water for 10 days); GpII: Gallic acid (alone) (50 mg/kg bw, once a day for 10 days); GpIII: Cisplatin (alone), single dose (6 mg/kg bw, I.P. on 5th day of study); GpIV: Gallic acid (50 mg/kg bw, once a day for 10 days) and also injected with single dose of Cisplatin (6 mg/kg bw, I.P., on 5th day of study). After 10 days, all rats were anaesthetized and plasma collected to estimate urea, creatinine, and uric acid. The right kidneys were removed for the study of gene expression and biochemical parameters. The left kidneys were used for histopathological studies. RESULTS The Cisplatin-induced nephrotoxicity was evident from the elevated levels of creatinine, urea, uric acid, and renal tissue MDA and also decreased levels of SOD, CAT, GPX, and GSH in renal tissue. Administration of Gallic acid significantly modulated nephrotoxicity markers, gene expression variations, and histopathological damage. CONCLUSION Outcomes of the present investigation suggest that Gallic acid provides protection against CP-induced nephrotoxicity, but for application in people, further studies are needed.
Collapse
Affiliation(s)
- Zahra Eslamifar
- Department of Medical Laboratory Sciences, School of Paramedical Sciences, Dezful University of Medical Sciences, Dezful, Iran
| | - Abbas Moridnia
- Department of Immunology, School of Medicine, Dezful University of Medical Sciences, Dezful, Iran
| | - Susan Sabbagh
- Department of Anatomy, Faculty of Medicine, Dezful University of Medical Sciences, Dezful, Iran
| | - Reza Ghaffaripour
- Department of Medical Laboratory Sciences, School of Paramedical Sciences, Dezful University of Medical Sciences, Dezful, Iran
| | - Leila Jafaripour
- Department of Anatomy, Faculty of Medicine, Dezful University of Medical Sciences, Dezful, Iran
| | - Mahin Behzadifard
- Department of Medical Laboratory Sciences, School of Paramedical Sciences, Dezful University of Medical Sciences, Dezful, Iran
| |
Collapse
|
21
|
Perše M. Cisplatin Mouse Models: Treatment, Toxicity and Translatability. Biomedicines 2021; 9:biomedicines9101406. [PMID: 34680523 PMCID: PMC8533586 DOI: 10.3390/biomedicines9101406] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/26/2021] [Accepted: 10/05/2021] [Indexed: 02/06/2023] Open
Abstract
Cisplatin is one of the most widely used chemotherapeutic drugs in the treatment of a wide range of pediatric and adult malignances. However, it has various side effects which limit its use. Cisplatin mouse models are widely used in studies investigating cisplatin therapeutic and toxic effects. However, despite numerous promising results, no significant improvement in treatment outcome has been achieved in humans. There are many drawbacks in the currently used cisplatin protocols in mice. In the paper, the most characterized cisplatin protocols are summarized together with weaknesses that need to be improved in future studies, including hydration and supportive care. As demonstrated, mice respond to cisplatin treatment in similar ways to humans. The paper thus aims to illustrate the complexity of cisplatin side effects (nephrotoxicity, gastrointestinal toxicity, neurotoxicity, ototoxicity and myelotoxicity) and the interconnectedness and interdependence of pathomechanisms among tissues and organs in a dose- and time-dependent manner. The paper offers knowledge that can help design future studies more efficiently and interpret study outcomes more critically. If we want to understand molecular mechanisms and find therapeutic agents that would have a potential benefit in clinics, we need to change our approach and start to treat animals as patients and not as tools.
Collapse
Affiliation(s)
- Martina Perše
- Medical Experimental Centre, Institute of Pathology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| |
Collapse
|
22
|
Crook A, De Lima Leite A, Payne T, Bhinderwala F, Woods J, Singh VK, Powers R. Radiation exposure induces cross-species temporal metabolic changes that are mitigated in mice by amifostine. Sci Rep 2021; 11:14004. [PMID: 34234212 PMCID: PMC8263605 DOI: 10.1038/s41598-021-93401-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 06/24/2021] [Indexed: 11/09/2022] Open
Abstract
Exposure to acute, damaging radiation may occur through a variety of events from cancer therapy and industrial accidents to terrorist attacks and military actions. Our understanding of how to protect individuals and mitigate the effects of radiation injury or Acute Radiation Syndrome (ARS) is still limited. There are only a few Food and Drug Administration-approved therapies for ARS; whereas, amifostine is limited to treating low dose (0.7-6 Gy) radiation poisoning arising from cancer radiotherapy. An early intervention is critical to treat ARS, which necessitates identifying diagnostic biomarkers to quickly characterize radiation exposure. Towards this end, a multiplatform metabolomics study was performed to comprehensively characterize the temporal changes in metabolite levels from mice and non-human primate serum samples following γ-irradiation. The metabolomic signature of amifostine was also evaluated in mice as a model for radioprotection. The NMR and mass spectrometry metabolomics analysis identified 23 dysregulated pathways resulting from the radiation exposure. These metabolomic alterations exhibited distinct trajectories within glucose metabolism, phospholipid biosynthesis, and nucleotide metabolism. A return to baseline levels with amifostine treatment occurred for these pathways within a week of radiation exposure. Together, our data suggests a unique physiological change that is independent of radiation dose or species. Furthermore, a metabolic signature of radioprotection was observed through the use of amifostine prophylaxis of ARS.
Collapse
Affiliation(s)
- Alexandra Crook
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588-0304, USA
| | - Aline De Lima Leite
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE, 68588-0304, USA
| | - Thomas Payne
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE, 68588-0304, USA
| | - Fatema Bhinderwala
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588-0304, USA
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE, 68588-0304, USA
| | - Jade Woods
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588-0304, USA
| | - Vijay K Singh
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, USUHS, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
- Armed Forces Radiobiology Research Institute, USUHS, Bethesda, MD, 20814, USA.
| | - Robert Powers
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588-0304, USA.
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE, 68588-0304, USA.
| |
Collapse
|
23
|
Singh VK, Seed TM, Cheema AK. Metabolomics-based predictive biomarkers of radiation injury and countermeasure efficacy: current status and future perspectives. Expert Rev Mol Diagn 2021; 21:641-654. [PMID: 34024238 DOI: 10.1080/14737159.2021.1933448] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION There is an urgent need for specific and sensitive bioassays to augment biodosimetric assessments of unwanted and excessive radiation exposures that originate from unexpected nuclear/radiological events, including nuclear accidents, acts of terrorism, or the use of a radiological dispersal device. If sufficiently intense, such ionizing radiation exposures are likely to impact normal metabolic processes within the cells and organs of the body, thus inducing multifaceted biological responses. AREAS COVERED This review covers the application of metabolomics, an emerging and promising technology based on quantitative and qualitative determinations of small molecules in biological samples for the rapid assessment of an individual's exposure to ionizing radiation. Recent advancements in the analytics of high-resolution chromatography, mass spectrometry, and bioinformatics have led to untargeted (global) and targeted (quantitative phase) approaches to identify biomarkers of radiation injury and countermeasure efficacy. Biomarkers are deemed essential for both assessing the radiation exposure levels and for extrapolative processes involved in determining scaling factors of a given radiation countering medicinal between experimental animals and humans. EXPERT OPINION The discipline of metabolomics appears to be highly informative in assessing radiation exposure levels and for identifying biomarkers of radiation injury and countermeasure efficacy.
Collapse
Affiliation(s)
- Vijay K Singh
- Division of Radioprotectants,Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Serices University of the Health Sciences, Bethesda, MD, USA.,Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | | | - Amrita K Cheema
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA.,Department of Biochemistry, Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
24
|
Al Za'abi M, Ali H, Al Sabahi M, Ali BH. The salutary action of melatonin and betaine, given singly or concomitantly, on cisplatin-induced nephrotoxicity in mice. Naunyn Schmiedebergs Arch Pharmacol 2021; 394:1693-1701. [PMID: 34003327 DOI: 10.1007/s00210-021-02097-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/03/2021] [Indexed: 12/13/2022]
Abstract
Cisplatin (CP) is commonly used in the treatment of various solid tumors. Its use, however, is hampered by nephrotoxicity. In this study, we compared the effect of betaine and melatonin given singly, with that of a combination of these two agents on CP-induced nephrotoxicity in mice. CP (20 mg/kg, given intraperitoneally on the 8th day of 12 days of the experiment) showed the typical physiological, biochemical, and histologic features of nephrotoxicity. CP-treated mice showed a significant reduction in food intake, body weight, and urine and fecal output. It also induced significant increases in the plasma concentrations of urea, creatinine, uric acid, phosphorous, adiponectin, interleukin-1β, interleukin-6, transforming growth factor -β1, tumor necrosis factor-α, and cystatin C. All these effects were significantly reduced by daily administration of betaine or melatonin at oral doses of 200 mg/kg and 10 mg/kg, respectively. Furthermore, using the two agents in combination caused further significant reductions in the above parameters. These findings suggest that betaine and melatonin concomitant use is likely to provide greater protection against CP-induced nephrotoxicity than when they are given singly, rendering them potentially suitable and safe agents to use in clinical trials to assess their possible beneficial actions in cancer patients receiving CP.
Collapse
Affiliation(s)
- Mohammed Al Za'abi
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, Al Khoud, P. O. Box 35, Muscat, Postal code 123, Oman.
| | - Haytham Ali
- Department of Animal and Veterinary Sciences, College of Agricultural and Marine Sciences, Sultan Qaboos University, Muscat, Postal code 123, Oman
| | - Mohammed Al Sabahi
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, Al Khoud, P. O. Box 35, Muscat, Postal code 123, Oman
| | - Badreldin H Ali
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, Al Khoud, P. O. Box 35, Muscat, Postal code 123, Oman
| |
Collapse
|
25
|
Saeidy S, Petera B, Pierre G, Fenoradosoa TA, Djomdi D, Michaud P, Delattre C. Plants arabinogalactans: From structures to physico-chemical and biological properties. Biotechnol Adv 2021; 53:107771. [PMID: 33992708 DOI: 10.1016/j.biotechadv.2021.107771] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 04/10/2021] [Accepted: 05/08/2021] [Indexed: 01/02/2023]
Abstract
Arabinogalactans (AGs) are plant heteropolysaccharides with complex structures occasionally attached to proteins (AGPs). AGs in cell matrix of different parts of plant are freely available or chemically bound to pectin rhamnogalactan. Type I with predominantly β-d-(1 → 4)-galactan and type II with β-d-(1 → 3) and/or (1 → 6)-galactan structural backbones construct the two main groups of AGs. In the current review, the chemical structure of AGs is firstly discussed focusing on non-traditional plant sources and not including well known industrial gums. After that, processes for their extraction and purification are considered and finally their techno-functional and biological properties are highlighted. The role of AG structure and function on health advantages such as anti-tumor, antioxidant, anti-ulcer- anti-diabetic and other activites and also the immunomodulatory effects on in-vivo model systems are overviewed.
Collapse
Affiliation(s)
- S Saeidy
- Department of Food Science and Technology, College of Agriculture, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - B Petera
- Faculté des Sciences de l'Université d'Antsiranana, BP O 201 Antsiranana, Madagascar; Université Clermont Auvergne, CNRS, SIGMA Clermont, Institut Pascal, F-63000 Clermont-Ferrand, France
| | - G Pierre
- Université Clermont Auvergne, CNRS, SIGMA Clermont, Institut Pascal, F-63000 Clermont-Ferrand, France
| | - T A Fenoradosoa
- Faculté des Sciences de l'Université d'Antsiranana, BP O 201 Antsiranana, Madagascar
| | - Djomdi Djomdi
- Department of Renewable Energy, National Advanced School of Engineering of Maroua, University of Maroua, Cameroon
| | - P Michaud
- Université Clermont Auvergne, CNRS, SIGMA Clermont, Institut Pascal, F-63000 Clermont-Ferrand, France.
| | - C Delattre
- Université Clermont Auvergne, CNRS, SIGMA Clermont, Institut Pascal, F-63000 Clermont-Ferrand, France; Institut Universitaire de France (IUF), 1 rue Descartes, 75005 Paris, France
| |
Collapse
|
26
|
Abdel-Rasol M, El-Beih NM, Yahya SMM, Ismail MA, El-Sayed WM. The Antitumor Activity of a Novel Fluorobenzamidine against Dimethylhydrazine- Induced Colorectal Cancer in Rats. Anticancer Agents Med Chem 2021; 20:450-463. [PMID: 31736450 DOI: 10.2174/1871520619666191021162411] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 09/25/2019] [Accepted: 10/01/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Colorectal cancer is among the leading causes of death worldwide. The incidence of deaths is expected to be 11.4 million in 2030. OBJECTIVE We aimed to evaluate the in vitro and in vivo antioxidant and antitumor activities of a novel Bithiophene- Fluorobenzamidine (BFB) against DMH-induced colorectal cancer in rats. METHODS The antiproliferative activity of BFB against HCT-116 colon cancer cells and apoptotic genes was assessed. In vivo study was also conducted in which 80 adult male rats were divided into 5 groups; control, BFB, and the other 3 groups were injected with DMH (20mg/kg, s.c., for 9 weeks). Group 4 was injected with 5 doses of cisplatin (2.5mg/kg, i.p over 21 weeks) and group 5 was injected with 3 doses/week of BFB (2.5mg/kg, i.p, for 21 weeks). RESULTS BFB exhibited weak to moderate in vitro antioxidant activity. It had a strong antiproliferative activity with IC50 ~0.3µg/ml. BFB induced extrinsic apoptosis through the upregulation of FasL, TRAL, p53 and caspase-8, and intrinsic apoptosis through the downregulation of Bcl-2 and survivin. BFB decreased the tumor incidence, multiplicity and size and improved the decreased body weight. BFB also ameliorated the functions of kidney and liver and antioxidants deteriorated by DMH. BFB significantly improved the pathological changes caused by DMH in colon tissues. CONCLUSION BFB showed a very promising antitumor activity against colorectal cancer induced by DMH in rats without causing hepato- or nephrotoxicity.
Collapse
Affiliation(s)
- Mohammed Abdel-Rasol
- Department of Zoology, Faculty of Science, University of Ain Shams, Abbassia 11566, Cairo, Egypt
| | - Nadia M El-Beih
- Department of Zoology, Faculty of Science, University of Ain Shams, Abbassia 11566, Cairo, Egypt
| | - Shaymaa M M Yahya
- Department of Hormones, Medical Research Division, National Research Center, Dokki 12622, Giza, Egypt
| | - Mohamed A Ismail
- Department of Chemistry, Faculty of Science, University of Mansoura, Mansoura, Egypt
| | - Wael M El-Sayed
- Department of Zoology, Faculty of Science, University of Ain Shams, Abbassia 11566, Cairo, Egypt
| |
Collapse
|
27
|
Nephroprotective Effects of Zataria multiflora Boiss. Hydroalcoholic Extract, Carvacrol, and Thymol on Kidney Toxicity Induced by Cisplatin in Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:8847212. [PMID: 33574884 PMCID: PMC7857888 DOI: 10.1155/2021/8847212] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 01/05/2021] [Accepted: 01/17/2021] [Indexed: 11/17/2022]
Abstract
Background Cisplatin (Cis) is an anticancer drug; however, it has dose-dependent renal toxicity. The current study aims to investigate the protective effects of Zataria multiflora Boiss. hydroalcoholic extract (Z.M.B), carvacrol, and thymol on cisplatin-induced nephrotoxicity in rats. Materials and Methods Forty-two Wistar male rats were randomly allocated into six groups (n = 7). Group I received normal saline; group II received Cis (7 mg/kg. ip); group III received the Z.M.B extract only (500 mg/kg/d, po); group IV received Z.M.B extract (500 mg/kg/d, po) + Cis; group V received carvacrol (50 mg/kg/d, po) + Cis; and group VI received thymol (50 mg/kg/d, po) + Cis. The levels of biochemical markers, oxidative stress parameters, and histopathological staining were determined in serum and renal tissues. Also, the chemical compositions (carvacrol and thymol) of the Z.M.B extract were assayed by HPLC analysis. Result The results revealed that Z.M.B extract, carvacrol, and thymol markedly decreased the renal index as compared with the Cis-only group. Also, carvacrol and thymol significantly reduced the blood urea nitrogen level as compared with the Cis-only group. Furthermore, Z.M.B extract, carvacrol, and thymol significantly attenuated the Cis-induced increase in malondialdehyde and nitric oxide metabolite. Additionally, histopathological examination showed that Z.M.B extract, carvacrol, and thymol markedly ameliorated Cis-induced renal tubular necrosis. Conclusion The results showed renoprotective effects of Z.M.B extract, carvacrol, and thymol in Cis-induced nephrotoxicity in rats. Therefore, Z.M.B extract can be considered as a potential candidate for the protection of nephrotoxicity induced by Cis.
Collapse
|
28
|
Abdelhamid AM, Mahmoud SS, Abdelrahman AE, Said NM, Toam M, Samy W, Amer MAM. Protective effect of cerium oxide nanoparticles on cisplatin and oxaliplatin primary toxicities in male albino rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2020; 393:2411-2425. [PMID: 32710137 DOI: 10.1007/s00210-020-01946-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 07/09/2020] [Indexed: 01/09/2023]
Abstract
Cisplatin and oxaliplatin are widely used anticancer drugs. Their use is restricted by their dose-limiting side effects: nephrotoxicity and neurotoxicity, respectively. Cerium oxide nanoparticles (CONPs) are promising antioxidant and anti-inflammatory agent. To test the possible ameliorative impact of CONPs on the toxic effect of cisplatin and oxaliplatin in male albino rats. Forty eight rats were divided into 6 groups: control group, CONPs group, cisplatin group, cisplatin and CONPs group, oxaliplatin group, and oxaliplatin and CONPs group. After 4 weeks, serum urea and creatinine, renal tissue level of interleukin 10 (IL10), and total antioxidant (TAO) were measured in control, CONPs, and cisplatin groups. The other kidney was used for histopathological and immunohistochemical studies. The right sciatic nerves and the lumbar spinal cord of rats from control, CONPs, and oxaliplatin groups were used for immunohistochemical evaluations of nitrotyrosine, myelin basic protein (MBP), and glial fibrillary acidic protein (GFAP). Cisplatin significantly increased serum urea and creatinine levels, significantly decreased the kidney level of IL10 and TAO with marked tubular necrosis, hemorrhage and renal damage. Also, it decreased IL10 immunohistochemical expression. CONPs significantly decreased the serum urea and creatinine level and increased IL10 and TAO with lower renal damage and strong IL10 expression compared with cisplatin group. Oxaliplatin significantly decreased MBP immunoreactivity and increased nitrotyrosine immunoreactivity. In the lumbar spinal cord, GFAP immunoreactivity was significantly increased. CONPs significantly increased MBP and decreased nitrotyrosine immunoreactivity. GFAP immunoreactivity was significantly decreased. CONPs ameliorated cisplatin and oxaliplatin primary toxicities through anti-inflammatory and antioxidant characteristics.
Collapse
Affiliation(s)
- Amira Mohamed Abdelhamid
- Clinical pharmacology department, faculty of medicine, Zagazig University, Zagazig, Sharqia, Egypt.
| | - Shireen Sami Mahmoud
- Clinical pharmacology department, faculty of medicine, Zagazig University, Zagazig, Sharqia, Egypt
| | - Aziza E Abdelrahman
- Pathology department, faculty of medicine, Zagazig University, Zagazig, Egypt
| | - Nelly Mohamed Said
- Pathology department, faculty of medicine, Zagazig University, Zagazig, Egypt
| | - Mostafa Toam
- Clinical Oncology department, faculty of medicine, Zagazig University, Zagazig, Egypt
| | - Walaa Samy
- Biochemistry department, faculty of medicine, Zagazig University, Zagazig, Egypt
| | - Marwa AbdEl-Moniem Amer
- Forensic Medicine and Clinical toxicology department, faculty of medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
29
|
Zhang X, Dong Y, Gao M, Hao M, Ren H, Guo L, Guo H. Knockdown of TRAP1 promotes cisplatin-induced apoptosis by promoting the ROS-dependent mitochondrial dysfunction in lung cancer cells. Mol Cell Biochem 2020; 476:1075-1082. [PMID: 33196942 DOI: 10.1007/s11010-020-03973-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 11/06/2020] [Indexed: 10/23/2022]
Abstract
The tumor necrosis factor receptor-associated protein 1 (TRAP1) is associated with the occurrence and development of various diseases, including inflammation and cancer. However, the role and mechanism of TRAP1 in the development of lung cancer need to be further explored. Therefore, the purpose of this study is to investigate the role of TRAP1 in the regulation of apoptosis by cisplatin and its special mechanism. The RT-qPCR and Western blot were used to detect the mRNA and protein expression of ANGPTL4 in A549 and H1299 cells, respectively. And the cell apoptosis and cell cycle were measured by flow cytometry (FCM). The expression of genes related to apoptosis and drug resistance as well as the cell cycle regulators, including MDM2, CyclinB1, and CDK1, were detected by Western blot. Finally, the reactive oxygen species (ROS) indicator DCFH-DA was performed to detect the generation of ROS, and the mitochondrial membrane potential (ΔΨm) was detected by JC-1 staining. The results showed that the expression of TRAP1 was significantly increased in A549/DDP and H1299/DDP than A549 and H1299 cells. Further research found that knockdown of TRAP1 induced apoptosis and caused G2/M cell cycle arrest in A549/DDP and H1299/DDP cells. What is more, siTRAP1 reduced the relative JC-1 polymer monomer fluorescence ratio and decreased the ΔΨm, up-regulated the expression of Cytochrome C. Importantly, siTRAP1 induces ROS-dependent mitochondrial dysfunction. It is suggested that that TRAP1 suppresses cisplatin-induced apoptosis by promoting ROS-dependent mitochondrial dysfunction.
Collapse
Affiliation(s)
- Xiaowei Zhang
- Department of Oncology, Xi'an Central Hospital, The Affiliated Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, 710003, Shaanxi, China
| | - Yu Dong
- Department of Respiratory, Xi'an Central Hospital, The Affiliated Hospital of Xian Jiaotong University College of Medicine, 185 Houzai Men, Xi'an, 710003, Shaanxi, China
| | - Miao Gao
- Department of Obstetrics and Gynecology, Xi'an No.5 Hospital, Xi'an, 710082, Shaanxi, China
| | - Minfeng Hao
- Department of Neurology, Xi'an Central Hospital, The Affiliated Hospital of Xian Jiaotong University College of Medicine, Xi'an, 710003, Shaanxi, China
| | - Hui Ren
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Ling Guo
- Yan'an University College of Medicine, Yan'an, 716000, Shaanxi, China
| | - Hua Guo
- Department of Respiratory, Xi'an Central Hospital, The Affiliated Hospital of Xian Jiaotong University College of Medicine, 185 Houzai Men, Xi'an, 710003, Shaanxi, China.
| |
Collapse
|
30
|
Casanova AG, Hernández-Sánchez MT, Martínez-Salgado C, Morales AI, Vicente-Vicente L, López-Hernández FJ. A meta-analysis of preclinical studies using antioxidants for the prevention of cisplatin nephrotoxicity: implications for clinical application. Crit Rev Toxicol 2020; 50:780-800. [PMID: 33170047 DOI: 10.1080/10408444.2020.1837070] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Cisplatin is an effective chemotherapeutic drug whose clinical use and efficacy are limited by its nephrotoxicity, which affects mainly the renal tubules and vasculature. It accumulates in proximal and distal epithelial tubule cells and causes oxidative stress-mediated cell death and malfunction. Consequently, many antioxidants have been tested for their capacity to prevent cisplatin nephrotoxicity. In this study, we made a systematic review of the literature and meta-analyzed 152 articles, which tested the nephroprotective effect of isolated compounds or mixtures of natural origin on cisplatin nephrotoxicity in preclinical models. This meta-analysis identified the most effective candidates and examined the efficacy obtained by antioxidants administered by the oral and intraperitoneal routes. By comparing with a recent, similar meta-analysis performed on clinical studies, this article identifies a disconnection between preclinical and clinical research, and contextualizes, discusses, and integrates the existing preclinical information toward the optimized selection of candidates to be further explored (clinical level). Despite proved efficacy, this article discusses the barriers limiting the clinical development of natural mixtures, such as those in extracts from Calendula officinalis flowers and Heliotropium eichwaldii roots. On the contrary, isolated compounds are more straightforward candidates, among which arjunolic acid and quercetin stand out in this meta-analysis.
Collapse
Affiliation(s)
- Alfredo G Casanova
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Department of Physiology and Pharmacology, University of Salamanca (USAL), Salamanca, Spain.,Fundación Instituto de Estudios de Ciencias de la Salud de Castilla y León (IECSCYL), Soria, Spain.,Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Salamanca, Spain.,National Network for Kidney Research REDINREN, Instituto de Salud Carlos III, Madrid, Spain
| | - M Teresa Hernández-Sánchez
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Department of Physiology and Pharmacology, University of Salamanca (USAL), Salamanca, Spain.,Fundación Instituto de Estudios de Ciencias de la Salud de Castilla y León (IECSCYL), Soria, Spain.,Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Salamanca, Spain.,National Network for Kidney Research REDINREN, Instituto de Salud Carlos III, Madrid, Spain
| | - Carlos Martínez-Salgado
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Department of Physiology and Pharmacology, University of Salamanca (USAL), Salamanca, Spain.,Fundación Instituto de Estudios de Ciencias de la Salud de Castilla y León (IECSCYL), Soria, Spain.,Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Salamanca, Spain.,National Network for Kidney Research REDINREN, Instituto de Salud Carlos III, Madrid, Spain
| | - Ana I Morales
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Department of Physiology and Pharmacology, University of Salamanca (USAL), Salamanca, Spain.,Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Salamanca, Spain.,National Network for Kidney Research REDINREN, Instituto de Salud Carlos III, Madrid, Spain.,Group of Biomedical Research on Critical Care (BioCritic), Valladolid, Spain
| | - Laura Vicente-Vicente
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Department of Physiology and Pharmacology, University of Salamanca (USAL), Salamanca, Spain.,Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Salamanca, Spain.,National Network for Kidney Research REDINREN, Instituto de Salud Carlos III, Madrid, Spain
| | - Francisco J López-Hernández
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Department of Physiology and Pharmacology, University of Salamanca (USAL), Salamanca, Spain.,Fundación Instituto de Estudios de Ciencias de la Salud de Castilla y León (IECSCYL), Soria, Spain.,Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Salamanca, Spain.,National Network for Kidney Research REDINREN, Instituto de Salud Carlos III, Madrid, Spain.,Group of Biomedical Research on Critical Care (BioCritic), Valladolid, Spain
| |
Collapse
|
31
|
Ruggiero A, Ariano A, Triarico S, Capozza MA, Romano A, Maurizi P, Mastrangelo S, Attinà G. Cisplatin-induced nephrotoxicity in children: what is the best protective strategy? J Oncol Pharm Pract 2020; 27:180-186. [PMID: 32990190 DOI: 10.1177/1078155220961550] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
INTRODUCTION Platinum compounds, which are considerably effective for the treatment of childhood malignancies, have significantly contributed to the increase in long-term survival of children with cancer. Unfortunately, children receiving cisplatin-based chemotherapy have been known to be at risk for severe disabling adverse effects, such as nephrotoxicity. METHODS A literature research of the MEDLINE PubMed database was conducted to identify articles published between 1980 and 2019 reviewing "Cisplatin AND mannitol." RESULTS The primary pharmacodynamics and clinical characteristics of cisplatin were described, focusing on its renal toxic effects and potential preventive strategies, in order to improve clinical outcomes among children with cancer aged 1 to 14 years. Currently, selecting either hydration alone or hydration plus mannitol for preventing nephrotoxicity has been controversial considering the lack of guidelines to provide treatment recommendations both among adults and children. CONCLUSIONS Appropriate knowledge regarding the pharmacokinetics and toxicological profile of cisplatin may help physicians prevent renal toxicity. Unfortunately, published data regarding the nephroprotective utility of adding mannitol appear to be inconclusive. As such, appropriate hydration remains the main fundamental strategy for reducing the risk of cisplatin-induced nephrotoxicity. Considering the increasing number of children safely cured of their tumours, it is imperative that those treated with cisplatin receive the most appropriate nephroprotective strategy for reducing the negative impact of platinum compounds on quality of life.
Collapse
Affiliation(s)
- Antonio Ruggiero
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Università Cattolica Sacro Cuore, Rome, Italy
| | - Anna Ariano
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Università Cattolica Sacro Cuore, Rome, Italy
| | - Silvia Triarico
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Università Cattolica Sacro Cuore, Rome, Italy
| | - Michele Antonio Capozza
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Università Cattolica Sacro Cuore, Rome, Italy
| | - Alberto Romano
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Università Cattolica Sacro Cuore, Rome, Italy
| | - Palma Maurizi
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Università Cattolica Sacro Cuore, Rome, Italy
| | - Stefano Mastrangelo
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Università Cattolica Sacro Cuore, Rome, Italy
| | - Giorgio Attinà
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Università Cattolica Sacro Cuore, Rome, Italy
| |
Collapse
|
32
|
Ali BH, Abdelrahman A, Al Suleimani Y, Manoj P, Ali H, Nemmar A, Al Za'abi M. Effect of concomitant treatment of curcumin and melatonin on cisplatin-induced nephrotoxicity in rats. Biomed Pharmacother 2020; 131:110761. [PMID: 33152924 DOI: 10.1016/j.biopha.2020.110761] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/14/2020] [Accepted: 09/16/2020] [Indexed: 12/15/2022] Open
Abstract
Cisplatin (CP) is a potent anticancer drug used to treat solid tumors. Its use, however, is dose-limited by its nephrotoxicity. We aimed to compare the effect of melatonin and curcumin given singly, with that of a combination of these two agents on CP-induced nephrotoxicity in rats. CP (6 mg/kg, given once intraperitoneally) induced nephrotoxicity as evidenced by several significant adverse physiological, biochemical and histopathological actions that included a reduction in body weight, increased urine production, and significant alterations in some conventional and novel renal damage indices in plasma, urine and kidneys. CP also elevated several pro-inflammatory cytokines and caused renal oxidative/nitrosative stress. Supplementation with either curcumin (200 mg/kg) or melatonin (10 mg /kg) given singly by oral gavage for eight consecutive days prior to CP injection and four days thereafter, significantly mitigated the adverse renal effects of CP, by attenuating the pro-inflammatory and apoptotic mediators and improving antioxidant competence in renal tissues of CP- treated rats. When curcumin and melatonin were given together, the ameliorative effect was augmented in some of the measured indices e.g. tumor necrosis factor alpha, cystatin C, uric acid, phosphorus in plasma and, urine creatinine and creatinine clearance. Renal platinum concertation was reduced more with curcumin than that with melatonin, while the reduction was maximized when both melatonin and curcumin were given. Pending further pharmacological and toxicological studies, a combination of these two agents is likely to be mor effective in mitigating the adverse renal effects of CP administered to cancer patients.
Collapse
Affiliation(s)
- Badreldin H Ali
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, P. O. Box 35 Al Khoud, 123, Oman
| | - Aly Abdelrahman
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, P. O. Box 35 Al Khoud, 123, Oman
| | - Yousuf Al Suleimani
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, P. O. Box 35 Al Khoud, 123, Oman
| | - Priyadarsini Manoj
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, P. O. Box 35 Al Khoud, 123, Oman
| | - Haytham Ali
- Department of Animal and Veterinary Sciences, College of Agricultural and Marine Sciences, Sultan Qaboos University, Muscat, 123, Oman
| | - Abderrahim Nemmar
- Department of Physiology, College of Medicine and Health Sciences, UAE University, Al Ain, United Arab Emirates
| | - Mohammed Al Za'abi
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, P. O. Box 35 Al Khoud, 123, Oman.
| |
Collapse
|
33
|
Park HR, Jo SK, Cho HH, Jung U. Synergistic Anti-cancer Activity of MH-30 in a Murine Melanoma Model Treated With Cisplatin and its Alleviated Effects Against Cisplatin-induced Toxicity in Mice. In Vivo 2020; 34:1845-1856. [PMID: 32606154 DOI: 10.21873/invivo.11979] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/10/2020] [Accepted: 04/12/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND/AIM Although cisplatin is an effective anticancer drug, its toxic effects on normal tissues limit its use. We developed a herbal formula, MH-30, with increased fat-soluble polyphenols by improving the manufacturing method of HemoHIM. In this study, we examined whether the combination of MH-30 with cisplatin exerts synergistic antitumor effect while it reduces cisplatin-induced toxicities. MATERIALS AND METHODS MH-30 was produced by adding the ethanol-insoluble fraction to its extract after decocting herbs in 30% ethanol and water. We used a melanoma-bearing mice model to investigate synergistic anticancer effects. The NK cell activity and cytokine levels were measured by Cr51-release assay and ELISA. The AST, ALT, BUN, and creatinine levels were estimated in the serum. RESULTS MH-30 effectively inhibited melanoma growth in vitro. Furthermore, MH-30 had a synergistic effect in combination with cisplatin on melanoma growth inhibition in vitro and in vivo. In melanoma-bearing mice, cisplatin alone decreased the activity of NK cells and the levels of IL-2 and IFN-γ, which were effectively restored by the combination of MH-30 with cisplatin. Combined treatment with MH-30 and cisplatin significantly inhibited the cisplatin-induced increase in the levels of AST, ALT, BUN, and creatinine. CONCLUSION Combination of MH-30 with cisplatin may be a beneficial anticancer treatment with reduced adverse effects.
Collapse
Affiliation(s)
- Hae-Ran Park
- Radiation Research Division, Korea Atomic Energy Research Institute (KAERI), Jeongeup, Republic of Korea
| | - Sung-Kee Jo
- Radiation Research Division, Korea Atomic Energy Research Institute (KAERI), Jeongeup, Republic of Korea
| | - Hyang-Hee Cho
- Radiation Research Division, Korea Atomic Energy Research Institute (KAERI), Jeongeup, Republic of Korea
| | - Uhee Jung
- Environmental Safety Evaluation Research Division, Korea Atomic Energy Research Institute (KAERI), Jeongeup, Republic of Korea
| |
Collapse
|
34
|
Alterations in Tissue Metabolite Profiles with Amifostine-Prophylaxed Mice Exposed to Gamma Radiation. Metabolites 2020; 10:metabo10050211. [PMID: 32455594 PMCID: PMC7281564 DOI: 10.3390/metabo10050211] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 05/18/2020] [Accepted: 05/19/2020] [Indexed: 12/02/2022] Open
Abstract
Acute exposure to high-dose ionizing irradiation has the potential to severely injure the hematopoietic system and its capacity to produce vital blood cells that innately serve to ward off infections and excessive bleeding. Developing a medical radiation countermeasure that can protect individuals from the damaging effects of irradiation remains a significant, unmet need and an area of great public health interest and concern. Despite significant advancements in the field of radiation countermeasure development to find a nontoxic and effective prophylactic agent for acute radiation syndrome, no such drug has yet been approved by the Food and Drug Administration. This study focuses on examining the metabolic corrections elicited by amifostine, a potent radioprotector, on tissues of vital body organs, such as the heart, spleen, and kidney. Our findings indicate that prophylaxis with this drug offers significant protection against potentially lethal radiation injury, in part, by correction of radiation-induced metabolic pathway perturbations.
Collapse
|
35
|
Yin M, Li N, Makinde EA, Olatunji OJ, Ni Z. N6-2-hydroxyethyl-adenosine ameliorate cisplatin induced acute kidney injury in mice. ALL LIFE 2020. [DOI: 10.1080/26895293.2020.1760149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Affiliation(s)
- Min Yin
- Department of Nephrology, China–Japan Union Hospital of Jilin University, Changchun, People’s Republic of China
| | - Na Li
- Department of Nephrology, China–Japan Union Hospital of Jilin University, Changchun, People’s Republic of China
| | | | | | - Ziyuan Ni
- Department of Nephrology, China–Japan Union Hospital of Jilin University, Changchun, People’s Republic of China
| |
Collapse
|
36
|
Renoprotective Effects of a New Free Radical Scavenger, XH-003, against Cisplatin-Induced Nephrotoxicity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:9820168. [PMID: 32377314 PMCID: PMC7189338 DOI: 10.1155/2020/9820168] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 03/31/2020] [Indexed: 12/18/2022]
Abstract
Acute renal injury has an incidence of 25%–30% in patients with tumors who are treated with cisplatin and in patients for whom no specific drugs are available for treatment. Amifostine is the only FDA-approved chemoprotective drug; however, its clinical application is limited because of side effects. The small-molecule antioxidant XH-003, an acute radiation syndrome- (ARS-) protective drug independently developed in our laboratory, with 100% intellectual property rights, overcomes the side effects of amifostine but retains its high efficacy. In this study, XH-003 showed a chemoprotective effect similar to that of amifostine. A mechanistic study showed that XH-003 could significantly reduce cisplatin-induced increases in serum creatinine and urea nitrogen, increase the activity of antioxidant enzymes (SOD, CAT, and GSH-Px), reduce oxidative stress and tissue inflammation, and alleviate renal tissue damage by blocking the activity of the mitochondrial apoptosis pathway. Most importantly, XH-003 could reduce the accumulation of cisplatin in renal tissue by regulating the expression of proteins involved in cisplatin uptake and excretion, such as organic cation transporter 2 and MRP2. Moreover, in an in vivo xenotransplantation model, XH-003 did not interfere with the antitumor effect of cisplatin. These data provide strong evidence that the ARS-protective agent has a great potential for protecting against chemotherapy-induced toxicity. Thus, XH-003 can be considered in antitumor therapy.
Collapse
|
37
|
Liu DX, Chen G, Guo HL, Chen DM, Li SH, Zhang WX. Synthesis, Crystal Structure, and Anticancer Activity of a In(III) Coordination Polymer Based on a Flexible 5-(4-Carboxybenzyl)Isophthalic Acid Ligand. J STRUCT CHEM+ 2020. [DOI: 10.1134/s0022476620040113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
38
|
Sadeghi H, Mansourian M, Panahi Kokhdan E, Salehpour Z, Sadati I, Abbaszadeh-Goudarzi K, Asfaram A, Doustimotlagh AH. Antioxidant and protective effect of Stachys pilifera Benth against nephrotoxicity induced by cisplatin in rats. J Food Biochem 2020; 44:e13190. [PMID: 32155675 DOI: 10.1111/jfbc.13190] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 01/04/2020] [Accepted: 01/30/2020] [Indexed: 11/26/2022]
Abstract
The aim of current study was to assess the antioxidant and renoprotective effects of Stachys pilifera Benth (S.P.B.) hydroalcoholic extract on nephrotoxicity induced with cisplatin (CP). Adult rats with bodyweight of 180-220 g were divided into five groups (n = 7) treated as follows: group 1, control; group 2, CP; group 3, pretreatment with S.P.B. before CP; group 4, posttreatment with S.P.B. after CP; and, group 5, S.P.B. extract. A single dose of CP (7 mg/kg) was intraperitoneally injected on the fifth day and hydroalcoholic extract of S.P.B. (500 mg kg-1 day-1 ) was orally administered. The levels of oxidative stress markers, biochemical tests, and histopathological staining were assayed in serum and renal tissue. Also, the chemical composition of S.P.B. extract was determined by GC-MS analysis. The main compositions of S.P.B. extract identified by GC-MS analysis, were hexadeca-2,6,10,14-tetraen-1-ol, 3,7,11,16-tetramethyl (24.77%), thymol (14.1%), and linolenic acid (13.4%). In groups treated and pretreated with S.P.B., blood urea nitrogen, creatinine, malondialdehyde, and nitric oxide metabolite in serum as well as malondialdehyde and protein carbonyl content of kidney tissues were significantly decreased in comparison to CP group; in contrast, total thiol group showed a significant increase in treated group as compared to CP group. Furthermore, histological investigation indicated that treatment with S.P.B. improved renal damages induced by CP. The current study showed that S.P.B. hydroalcoholic extract improved the biochemical parameters and kidney function as well as restored antioxidant activity in CP-induced nephrotoxicity. However, it needs more investigations to define the mechanism of S.P.B. action. PRACTICAL APPLICATIONS: In different regions of Iran, Stachys is demonstrated by 34 species, out of which 13 are endemic, one of these endemic species is Stachys pilifera Benth (S.P.B.). The oil of S.P.B. is mainly consisted of cis-chrysanthenyl acetate, cis-chrysanthenol, spathulenol, β-caryophyllene, linalool, and terpinen-4-ol. Moreover, phytochemical studies have shown the presence of compounds such as diterpenes, phenylethanoid glycosides, saponins, terpenoides, and flavonoids in Stachys species. The aerial parts of S.P.B. are consumed as herbal tea to treat several disorders, for example, infections, asthma, and rheumatoid arthritis in Iranian folk medicine. The aim of current study was to evaluate the antioxidant and protective effects of S.P.B. hydroalcoholic extract on nephrotoxicity induced with cisplatin (CP). The current study showed that S.P.B. hydroalcoholic extract improved the biochemical parameters and kidney function as well as restored antioxidant activity in CP-induced nephrotoxicity. However, it needs more researches to define the mechanism of S.P.B. action.
Collapse
Affiliation(s)
- Hossein Sadeghi
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Mahboubeh Mansourian
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | | | - Zeinab Salehpour
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Iman Sadati
- Student Research Committee, Yasuj University of Medical Sciences, Yasuj, Iran
| | | | - Arash Asfaram
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | | |
Collapse
|
39
|
Singla P, Salunke DB. Recent advances in steroid amino acid conjugates: Old scaffolds with new dimensions. Eur J Med Chem 2020; 187:111909. [PMID: 31830636 DOI: 10.1016/j.ejmech.2019.111909] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 11/20/2019] [Accepted: 11/22/2019] [Indexed: 12/13/2022]
|
40
|
Ruggiero A, Trombatore G, Triarico S, Capozza MA, Coccia P, Attina G, Mastrangelo S, Maurizi P. Cisplatin Toxicity in Children with Malignancy. BIOMEDICAL AND PHARMACOLOGY JOURNAL 2019; 12:1603-1611. [DOI: 10.13005/bpj/1791] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Platinum’ derivates are antineoplastic agents widely adopted for their efficacy for the treatment of many pediatric cancers. The use of cisplatin has positively influenced the results of the cure of different childhood malignancies. However, cisplatin-based treatments are limited by the risk of severe and progressive toxicities, such as oto- or nephrotoxicity, that can be more serious in very young children expecially when high cumulative doses and/or radiotherapy is administered. A correct knowledge of the cisplatin’ pharmacological features might be of interest for clinicians in order to manage its potential toxicities. Based on the positive trend in the cure of children with cancer, it is crucial that all children receiving cisplatin-based chemotherapy have and appropriate and long-term follow-up to improve their quality of life.
Collapse
Affiliation(s)
- Antonio Ruggiero
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A .Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Roma, Italy
| | - Giovanna Trombatore
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A .Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Roma, Italy
| | - Silvia Triarico
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A .Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Roma, Italy
| | - Michele Antonio Capozza
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A .Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Roma, Italy
| | - Paola Coccia
- Pediatric Hemato-oncology Unit, Ospedale Salesi, Azienda Ospedali Riuniti Ancona, Ancona, Italy
| | - Giorgio Attina
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A .Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Roma, Italy
| | - Stefano Mastrangelo
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A .Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Roma, Italy
| | - Palma Maurizi
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A .Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Roma, Italy
| |
Collapse
|
41
|
Ali BH, Al Salam S, Al Suleimani Y, Al Za'abi M, Ashique M, Manoj P, Sudhadevi M, Al Tobi M, Nemmar A. Ameliorative effect of sesamin in cisplatin-induced nephrotoxicity in rats by suppressing inflammation, oxidative/nitrosative stress, and cellular damage. Physiol Res 2019; 69:61-72. [PMID: 31852200 DOI: 10.33549/physiolres.934142] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Nephrotoxicity of cisplatin (CP) involves renal oxidative stress and inflammation, and sesamin (a major liganin in many plants) has strong antioxidant and antiinflammatory actions. Therefore, we investigated here the possible mitigative action of sesamin on CP nephrotoxicity in rats. Sesamin was given orally (5 mg/kg/day, 10 days), and on the 7th day, some of the treated rats were injected intraperitoneally with either saline or CP (5 mg/kg). On the 11th day, rats were sacrificed, and blood and urine samples and kidneys were collected for biochemical estimation of several traditional and novel indices of renal damage in plasma and urine, several oxidative and nitrosative indices in kidneys, and assessment of histopathological renal damage. CP significantly and adversely altered all the physiological, biochemical and histopathological indices of renal function measured. Kidneys of CP-treated rats had a moderate degree of necrosis. This was markedly lessened when CP was given simultaneously with sesamin. Sesamin treatment did not significantly alter the renal CP concentration. The results suggested that sesamin had ameliorated CP nephrotoxicity in rats by reversing the CP-induced oxidative stress and inflammation. Pending further pharmacological and toxicological studies sesamin may be considered a potentially useful nephroprotective agent.
Collapse
Affiliation(s)
- B H Ali
- Department of Pharmacology, College of Medicine, Sultan Qaboos University, Al Khod, Oman.
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Cheema AK, Li Y, Girgis M, Jayatilake M, Simas M, Wise SY, Olabisi AO, Seed TM, Singh VK. Metabolomic studies in tissues of mice treated with amifostine and exposed to gamma-radiation. Sci Rep 2019; 9:15701. [PMID: 31666611 PMCID: PMC6821891 DOI: 10.1038/s41598-019-52120-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 10/14/2019] [Indexed: 02/07/2023] Open
Abstract
Although multiple radioprotectors are currently being investigated preclinically for efficacy and safety, few studies have investigated concomitant metabolic changes. This study examines the effects of amifostine on the metabolic profiles in tissues of mice exposed to cobalt-60 total-body gamma-radiation. Global metabolomic and lipidomic changes were analyzed using ultra-performance liquid chromatography (UPLC) quadrupole time-of-flight mass spectrometry (QTOF-MS) in bone marrow, jejunum, and lung samples of amifostine-treated and saline-treated control mice. Results demonstrate that radiation exposure leads to tissue specific metabolic responses that were corrected in part by treatment with amifostine in a drug-dose dependent manner. Bone marrow exhibited robust responses to radiation and was also highly responsive to protective effects of amifostine, while jejunum and lung showed only modest changes. Treatment with amifostine at 200 mg/kg prior to irradiation seemed to impart maximum survival benefit, while the lower dose of 50 mg/kg offered only limited survival benefit. These findings show that the administration of amifostine causes metabolic shifts that would provide an overall benefit to radiation injury and underscore the utility of metabolomics and lipidomics to determine the underlying physiological mechanisms involved in the radioprotective efficacy of amifostine. This approach may be helpful in identifying biomarkers for radioprotective efficacy of amifostine and other countermeasures under development.
Collapse
Affiliation(s)
- Amrita K Cheema
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
- Department of Biochemistry, Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC, USA
| | - Yaoxiang Li
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Michael Girgis
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Meth Jayatilake
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Madison Simas
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Serices University of the Health Sciences, Bethesda, MD, USA
- Armed Forces Radiobiology Research Institute, Uniformed Serices University of the Health Sciences, Bethesda, MD, USA
| | - Stephen Y Wise
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Serices University of the Health Sciences, Bethesda, MD, USA
- Armed Forces Radiobiology Research Institute, Uniformed Serices University of the Health Sciences, Bethesda, MD, USA
| | - Ayodele O Olabisi
- Armed Forces Radiobiology Research Institute, Uniformed Serices University of the Health Sciences, Bethesda, MD, USA
| | | | - Vijay K Singh
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Serices University of the Health Sciences, Bethesda, MD, USA.
- Armed Forces Radiobiology Research Institute, Uniformed Serices University of the Health Sciences, Bethesda, MD, USA.
| |
Collapse
|
43
|
Tamadon MR, Tirom S, Ghahremanfard F, Baradaran A, Ghorbani R. Evaluation of the Protective Effect of Cystone Against Cisplatin-induced Nephrotoxicity in Patients with Cancer: A Pilot Study. Int J Prev Med 2019; 10:180. [PMID: 32133098 PMCID: PMC6826770 DOI: 10.4103/ijpvm.ijpvm_66_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 07/16/2018] [Indexed: 11/04/2022] Open
Abstract
Introduction Cisplatin is a widely used anti-cancer drug that is commonly administered for the treatment of various cancers. However, nephrotoxicity is the most important side effect of this drug which limits its use. This study aimed to investigate the protective effect of Cystone against nephrotoxicity induced by Cisplatin in patients with cancer. Methods This pilot clinical trial study was conducted on 43 cancer patients treated with Cisplatin (75 mg/m2 for a period of six months). The subjects were divided into treatment group (receiving Cystone, two per 8 hours; n = 21) and control group (n = 22). The two groups were compared with each other in terms of demographic and laboratory variables. Results In the intervention group receiving Cystone, serum creatinine-based GFR level (P = 0.453) and 24-hour urine creatinine-based GFR level (P = 0.397) did not change significantly during the studied period, but in the control group, serum creatinine-based GFR level (P = 0.013) and 24-hour urine creatinine-based GFR level (P = 0.016) significantly changed. Serum creatinine-based GFR level increased by 2.3 units in the intervention group and 10.5 units in the control group (P = 0.005) in the six months of the study. At the end of the sixth month, 24-hour urine creatinine-based GFR level increased by 2.2 units in the intervention group and 0.8 unit in the control group (P = 0.008). Conclusions The use of Cystone resulted in more stable kidney function indices in the intervention group, as compared with the control group. Therefore, Cystone seems to have a protective effect against nephrotoxicity induced by Cisplatin in cancer patients.
Collapse
Affiliation(s)
- Mohammad Reza Tamadon
- Department of Internal Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Samaneh Tirom
- Department of Internal Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | | | - Azar Baradaran
- Department of Pathology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Raheb Ghorbani
- Social Determinants of Health Research Center, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
44
|
Singh VK, Seed TM. The efficacy and safety of amifostine for the acute radiation syndrome. Expert Opin Drug Saf 2019; 18:1077-1090. [PMID: 31526195 DOI: 10.1080/14740338.2019.1666104] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction: A radiation countermeasure that can be used prior to radiation exposure to protect the population from the harmful effects of radiation exposure remains a major unmet medical need and is recognized as an important area for research. Despite substantial advances in the research and development for finding nontoxic, safe, and effective prophylactic countermeasures for the acute radiation syndrome (ARS), no such agent has been approved by the United States Food and Drug Administration (FDA). Area covered: Despite the progress made to improve the effectiveness of amifostine as a radioprotector for ARS, none of the strategies have resolved the issue of its toxicity/side effects. Thus, the FDA has approved amifostine for limited clinical indications, but not for non-clinical uses. This article reviews recent strategies and progress that have been made to move forward this potentially useful countermeasure for ARS. Expert opinion: Although the recent investigations have been promising for fielding safe and effective radiation countermeasures, additional work is needed to improve and advance drug design and delivery strategies to get FDA approval for broadened, non-clinical use of amifostine during a radiological/nuclear scenario.
Collapse
Affiliation(s)
- Vijay K Singh
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences , Bethesda , MD , USA.,Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences , Bethesda , MD , USA
| | | |
Collapse
|
45
|
Zakaria MM, El-Tantawy FMM, Khater SM, Derbala SA, Farag VMEM, Abdel-Aziz AAF. Protective and curative role of Spirulina platensis extracts on cisplatin induce acute kidney injury in rats. ACTA ACUST UNITED AC 2019. [DOI: 10.1080/2314808x.2019.1653570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
| | | | | | - Safaa A. Derbala
- Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | | | | |
Collapse
|
46
|
Sohail N, Hira K, Tariq A, Sultana V, Ehteshamul-Haque S. Marine macro-algae attenuates nephrotoxicity and hepatotoxicity induced by cisplatin and acetaminophen in rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:25301-25311. [PMID: 31256398 DOI: 10.1007/s11356-019-05704-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 06/06/2019] [Indexed: 06/09/2023]
Abstract
Cisplatin is considered one of the best anticancer medications often used for the treatment of various cancers even with its adverse effects. Acetaminophen (paracetamol) is a widely used analgesic-antipyretic drug that causes hepatotoxicity at higher than the effective doses. The present study assesses the nephroprotective and hepatoprotective effects of two seaweeds against cisplatin and acetaminophen toxicity in rats. Damage to the liver and kidney was induced by administering a single intraperitoneal dose of acetaminophen (600 mg/kg) or cisplatin (7 mg/kg) to groups of rats. The damage to the liver and kidney was assessed by the elevated liver (ALT, AST, ALP, LDH, electrolytes) and kidney (urea, creatinine) biomarkers. The ethanol extract of brown seaweed reversed the elevated levels of kidney and liver biomarkers along with triglycerides, cholesterol, and glucose. Among the two seaweeds, Sargassum ilicifolium showed better nephroprotective and hepatoprotective effects than the standard drug N-Acetyl-cysteine, Halymenia porphyroides showed only limited protection. Findings of this study provide evidence of nephroprotective and hepatoprotective effects of S. ilicifolium. Seaweed could be a beneficial dietary supplement to attenuate nephrotoxicity and hepatotoxicity.
Collapse
Affiliation(s)
- Nida Sohail
- Biotechnology and Drug Development Laboratory, Department of Biochemistry, University of Karachi, Karachi, 75270, Pakistan
| | - Khan Hira
- Institute of Biomedical Sciences, Dow University of Health Sciences, Karachi, Pakistan
| | - Amna Tariq
- M.A.H. Qadri Biological Research Centre, University of Karachi, Karachi, 75270, Pakistan
| | - Viqar Sultana
- Biotechnology and Drug Development Laboratory, Department of Biochemistry, University of Karachi, Karachi, 75270, Pakistan
| | | |
Collapse
|
47
|
Afsar T, Razak S, Almajwal A, Shabbir M, Khan MR. Evaluating the protective potency of Acacia hydaspica R. Parker on histological and biochemical changes induced by Cisplatin in the cardiac tissue of rats. Altern Ther Health Med 2019; 19:182. [PMID: 31337380 PMCID: PMC6651963 DOI: 10.1186/s12906-019-2575-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 06/24/2019] [Indexed: 12/30/2022]
Abstract
Background Increase oxidative trauma is the main cause behind Cisplatin (CP) induced cardiotoxicity which restricts its clinical application as anti-neoplastic prescription. Acacia hydaspica is a natural shrub with diverse bioactivities. Acacia hydaspica ethyl acetate extract (AHE) ameliorated drug-induced cardiotoxicity in animals with anti-oxidative mechanisms. Current study aimed to evaluate the protective potential of A. hydaspica against cisplatin-induced myocardial injury. Methods Rats were indiscriminately distributed into six groups (n = 6). Group 1: control; Groups 2: Injected with CP (7.5 mg/kg bw, i.p, single dose) on day 16; Group 3: Treated for 21 days with AHE (400 mg/kg b.w, oral); Group 4: Received CP injection on day 16 and treated with AHE for 5 days post injection; Group 5: Received AHE (400 mg/kg b.w/day, p.o.) for 21 days and CP (7.5 mg/kg b.w., i.p.) on day 16; Group 6: Treated with silymarin (100 mg/kg b.w., p.o.) after 1 day interval for 21 days and CP injection (7.5 mg/kg b.w., i.p.) on day 16. On 22nd day, the animals were sacrificed and their heart tissues were removed. Cisplatin induced cardiac toxicity and the influence of AHE were evaluated by examination of serum cardiac function markers, cardiac tissue antioxidant enzymes, oxidative stress markers and histology. Results CP inoculation considerably altered cardiac function biomarkers in serum and diminished the antioxidant enzymes levels, while increased oxidative stress biomarkers in cardiac tissues AHE treatment attenuated CP-induced deteriorations in creatine kinase (CK), Creatine kinase isoenzymes MB (CK-MB), cardiac Troponin I (cTNI) and lactate dehydrogenase (LDH) levels and ameliorated cardiac oxidative stress markers as evidenced by decreasing lipid peroxidation, H2O2 and NO content along with augmentation in phase I and phase II antioxidant enzymes. Additionally, CP inoculation also induced morphological alterations which were ameliorated by AHE. In pretreatment group more significant protection was observed compared to post-treatment group indicating preventive potential of AHE. The protective potency of AHE was comparable to silymarin. Conclusion Results demonstrate that AHE attenuated CP induce cardiotoxicity. The polyphenolic metabolites and antioxidant properties of AHE might be responsible for its protective influence.
Collapse
|
48
|
Kheira HS, El-Sayed SAES, Elsayed GR, Rizk MA. Dietary flaxseed oil inhibits kidney NF-kappa B activation and pro-inflammatory cytokine expression in cisplatin-treated rats. COMPARATIVE CLINICAL PATHOLOGY 2019; 28:349-357. [DOI: 10.1007/s00580-018-2871-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/20/2018] [Indexed: 09/02/2023]
|
49
|
|
50
|
Zangui M, Atkin SL, Majeed M, Sahebkar A. Current evidence and future perspectives for curcumin and its analogues as promising adjuncts to oxaliplatin: state-of-the-art. Pharmacol Res 2019; 141:343-356. [DOI: 10.1016/j.phrs.2019.01.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 01/11/2019] [Accepted: 01/11/2019] [Indexed: 02/06/2023]
|