1
|
Shaban DA, El-Shahawy AAG, Zanaty MI, Eldin ZE, Abd-Elbaset M, Shams A, Tamur S, Ahmed OM. Utilizing Nanoparticles of Hesperidin Loaded on Layered Double Hydroxide to Reduce Hepatotoxicity Caused by Paracetamol in Rats: Controlling of Biotransformation, Oxidative Stress, Inflammation, and Apoptosis. Pharmaceutics 2025; 17:429. [PMID: 40284423 PMCID: PMC12030007 DOI: 10.3390/pharmaceutics17040429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/10/2025] [Accepted: 03/18/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: The most used antipyretic and pain relief treatment is paracetamol (acetaminophen), also known as N-acetyl-para-aminophenol (APAP). However, it is considered potentially hazardous if consumed repeatedly in large doses or over prolonged periods. This investigation explores the effectiveness of hesperidin (Hesp) and Hesp loaded on layered double hydroxide nanoparticles (Hesp-NPs) in inhibiting the progression of acute hepatotoxicity in rats induced by APAP. Methods: LDH-Hesp-NPs were prepared and characterized. Male Wistar rats were orally treated with Hesp and Hesp-NPs at the same adjusted dose (100 mg/kg) every other day for six weeks. After 2 h of the first doses of Hesp and Hesp-NPs, the rats received one oral dose of APAP (750 mg/kg). Results: Administering of Hesp and Hesp-NPs to APAP-treated rats significantly reduced oxidant parameter (malondialdehyde) and serum enzymes (ALT, AST, LDH, and ALP) associated with liver function. Antioxidant markers in the liver, such as catalase and glutathione, also increased notably. Moreover, Hesp and Hesp-NPs enhanced the mRNA expression of liver UGT1A6, IL-10, and HO-1. Conversely, the mRNA expressions of liver CYP1A1, KEAP1, TGF-β, P53, and BAX decreased. These improvements in biochemical and molecular markers were corroborated by liver histopathology. Conclusions: Hesp and Hesp-NPs protect significantly against APAP-induced hepatotoxicity in male Wistar rats. Hesp-NPs treatment was more potent. The protective effects may be mediated via modulation of APAP biotransformation, oxidative stress, inflammation and apoptosis.
Collapse
Affiliation(s)
- Deyaa A. Shaban
- Department of Biotechnology and Life Sciences, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef 62511, Egypt; (D.A.S.); (M.I.Z.)
| | - Ahmed A. G. El-Shahawy
- Materials Science and Nanotechnology Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef 62511, Egypt; (A.A.G.E.-S.); (Z.E.E.)
| | - Mohamed I. Zanaty
- Department of Biotechnology and Life Sciences, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef 62511, Egypt; (D.A.S.); (M.I.Z.)
| | - Zienab E. Eldin
- Materials Science and Nanotechnology Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef 62511, Egypt; (A.A.G.E.-S.); (Z.E.E.)
| | - Mohamed Abd-Elbaset
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, El Salehya El Gadida University (SGU), El Salheya 44813, Egypt
| | - Anwar Shams
- Department of Pharmacology, College of Medicine, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
- Research Center for Health Sciences, Deanship of Graduate Studies and Scientific Research, Taif University, Taif 26432, Saudi Arabia
- High Altitude Research Center, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Shadi Tamur
- Department of Pediatric, College of Medicine, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia;
| | - Osama M. Ahmed
- Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef 62521, Egypt;
| |
Collapse
|
2
|
Elsayed MA, Radwan DA, Rabah HM, El-Horany HES, Nasef NA, Abo El Gheit RE, Emam MN, Elesawy RO, Elseady W, Mahmoud A. Protective Effects of Galangin Against Cyclophosphamide-Induced Cardiotoxicity via Suppressing NF-κB and Improving Mitochondrial Biogenesis. J Biochem Mol Toxicol 2025; 39:e70193. [PMID: 39999301 DOI: 10.1002/jbt.70193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 02/06/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025]
Abstract
Cyclophosphamide (CYP) is an effective chemotherapeutic and immunosuppressive agent; however, its clinical application is limited by a variety of toxic side effects. Mitochondrial dysfunction has been associated with the pathogenesis of chemotherapy-induced cardiotoxicity. This work aimed to evaluate the possible protective effect of galangin (Gal) on CYP-induced cardiotoxicity, pointing to its ability to promote mitochondrial biogenesis. Thirty two male rats were allocated equally into four groups: control; Gal-treated; CYP-treated; and Gal + CYP-treated groups. Markers of cardiac injury, oxidative/antioxidant status, inflammation, apoptosis, and mitochondrial function were assessed in addition to histopathological and electrocardiographic (ECG) evaluation. The current results revealed that Gal treatment significantly attenuated the cardiac injury and retrieved the alterations in cardiac histopathology and ECG changes. Also, it restored redox balance, as evidenced by the alleviation of malondialdehyde (MDA) levels and increased glutathione peroxidase (GPx) activity. Gal activated the sirtuin (SIRT) 1/nuclear factor erythroid 2-related factor 2 (Nrf2)-mediated signaling pathway, as indicated by upregulation of SIRT1, Nrf2, SIRT3, and mitochondrial transcription factor (TFAM), in addition to increased levels of superoxide dismutase 2 (SOD)2 and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), together with increased activity of citrate synthase (CS), pointing to improved mitochondrial function. It ameliorated the inflammation and apoptosis-associated markers supported by biochemical and immunostaining data. Our study provided novel insights elucidating the mitigative potential of against CYP-induced cardiac oxidative damage, inflammation, apoptosis, and mitochondrial dysfunction by upregulating the SIRT1/Nrf2/SIRT3/PGC-1α/TFAM survival pathway.
Collapse
Affiliation(s)
- Manar Ali Elsayed
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Doaa A Radwan
- Department of Anatomy and Embryology, Faculty of Medicinen, Tanta University, Tanta, Egypt
| | - Hanem Mohamed Rabah
- Medical Biochemistry Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Hemat El-Sayed El-Horany
- Medical Biochemistry Department, Faculty of Medicine, Tanta University, Tanta, Egypt
- Biochemistry Department, College of Medicine, Hail University, Hail, Saudi Arabia
| | - Nahla Anas Nasef
- Medical Biochemistry Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Rehab E Abo El Gheit
- Department of Physiology, Faculty of Medicine, Tanta University, Tanta, Egypt
- Department of Physiology, Faculty of Physical Therapy, AlSalam University, Tanta, Egypt
| | - Marwa N Emam
- Department of Physiology, Faculty of Medicine, Tanta University, Tanta, Egypt
- Bio-Physiology Department, Ibn Sina National College for Medical Studies, Jeddah, Saudi Arabia
| | - Rasha Osama Elesawy
- Department of Pharmacology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Walaa Elseady
- Department of Anatomy and Embryology, Faculty of Medicinen, Tanta University, Tanta, Egypt
| | - Alia Mahmoud
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
3
|
Ali M, Din Wani SU, Manjula S, Mruthunjaya K, Shakeel F, DR B, Sridhar SB, Mohiuddin I, Mir RH, Dey T. Divine noni's protective impact on Swiss albino mice's short-term memory impairment caused by cyclophosphamide: A behavioral and biochemical approach. Heliyon 2024; 10:e37557. [PMID: 39309823 PMCID: PMC11415706 DOI: 10.1016/j.heliyon.2024.e37557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 08/26/2024] [Accepted: 09/05/2024] [Indexed: 09/25/2024] Open
Abstract
Cyclophosphamide (CYL) is a first-line cancer chemotherapeutic agent widely used for the treatment of cancer that has severe toxic effects. The primary mechanism by which CYL induces toxicity through free radical generation. Morinda citrifolia (Noni) fruit juice is an herbal remedy documented to have antioxidant properties. The aim of the current study was to investigate the protective effect of noni against CYL-induced memory impairment in Swiss albino mice. Treatment schedule: Group 1: Normal: Received vehicle; Group 2: CYL treatment: Received CYL (40.0 mg/kg b.w. i.p.) on day one; Group 3: NJ treatment: Received NJ (360 mg/b.w. p.o.) once daily for 14 days. Group 4: DNG treatment: DNG (360 mg/b.w. p.o.) once daily for 14 days, Group 5: NJ + CYL treatment: Received CYL (40.0 mg/kg b.w. i.p.) on day one and after half an hour of received NJ (360 mg/b.w. p.o.) once daily for 14 days. Group 6: DNG + CYL treatment: Received CYL (40.0 mg/kg b.w. i.p.) on day one and after half an hour received DNG (360 mg/b.w. p.o.) once daily for 14 days. Mice were subjected to the Morris water maze (MWM) challenge for two weeks as part of a behavioral study. Short-term memory impairment was observed in the behavioral activity of CYL-treated mice in the MWM test in the 1st week trial, and this effect was reversed in the 2nd week trial in the combination treatment group. The behavioral analysis proved that noni supplementation reduced the risk of memory impairment caused by CYL. Biochemical analysis revealed that CYL markedly increased the levels of AChE and MDA in brain tissue. Similarly, decreases in the levels of antioxidants, i.e., GSH, CAT, SOD and GST, were detected in the brain tissue of the mice exposed to CYL. Qualitative and quantitative examinations of histopathological examination of the mouse hippocampus supported the above findings. The results demonstrated that noni supplement therapy reversed the changes in the MDA, AChE, and antioxidant enzyme levels while improving the behavioral and histological alterations caused by CYL. Long-term hippocampal growth and memory are unaffected, suggesting that CYL is less harmful. According to our research, supplementing with noni in conjunction with CYL may be a helpful treatment strategy for treating memory impairment caused by CYL.
Collapse
Affiliation(s)
- Mohammad Ali
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, 570015, India
- Department of Pharmacology, Sri Adichunchanagiri College of Pharmacy, Adichunchanagiri University, B. G Nagar, Bellur, Karnataka, 571418, India
| | - Shahid Ud Din Wani
- Department of Pharmaceutical Sciences, School of Applied Science and Technology, University of Kashmir, Srinagar, 190006, India
| | - S.N. Manjula
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, 570015, India
| | - K. Mruthunjaya
- Department of Pharmacognosy, JSS College of Pharmacy, JSS Academy of Higher, Education and Research, Mysuru, 570015, India
| | - Faiyaz Shakeel
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Bharathi DR
- Department of Pharmacology, Sri Adichunchanagiri College of Pharmacy, Adichunchanagiri University, B. G Nagar, Bellur, Karnataka, 571418, India
| | - Sathvik B. Sridhar
- Department of Clinical Pharmacy and Pharmacology, RAK College of Pharmacy, RAK Medical and Health Sciences University, Ras Al Khaimah, PO Box, 11172, United Arab Emirates
| | - Ishfaq Mohiuddin
- Department of Zoology, Annamalai University, Annamalainagar, 608002, India
| | - Reyaz Hassan Mir
- Department of Pharmaceutical Sciences, School of Applied Science and Technology, University of Kashmir, Srinagar, 190006, India
| | - Tathagata Dey
- Department of Pharmaceutical Chemistry, East Point College of Pharmacy, Bangalore, 560049, India
| |
Collapse
|
4
|
Gent DG, Saif M, Dobson R, Wright DJ. Cardiovascular Disease After Hematopoietic Stem Cell Transplantation in Adults: JACC: CardioOncology State-of-the-Art Review. JACC CardioOncol 2024; 6:475-495. [PMID: 39239331 PMCID: PMC11372032 DOI: 10.1016/j.jaccao.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 06/11/2024] [Accepted: 06/28/2024] [Indexed: 09/07/2024] Open
Abstract
The use of hematopoietic cell transplantation (HCT) has expanded in the last 4 decades to include an older and more comorbid population. These patients face an increased risk of cardiovascular disease after HCT. The risk varies depending on several factors, including the type of transplant (autologous or allogeneic). Many therapies used in HCT have the potential to be cardiotoxic. Cardiovascular complications after HCT include atrial arrhythmias, heart failure, myocardial infarction, and pericardial effusions. Before HCT, patients should undergo a comprehensive cardiovascular assessment, with ongoing surveillance tailored to their individual level of cardiovascular risk. In this review, we provide an overview of cardiotoxicity after HCT and outline our approach to risk assessment and ongoing care.
Collapse
Affiliation(s)
- David G Gent
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart and Chest Hospital, Liverpool, United Kingdom
| | - Muhammad Saif
- The Clatterbridge Cancer Centre, Liverpool, United Kingdom
| | - Rebecca Dobson
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart and Chest Hospital, Liverpool, United Kingdom
| | - David J Wright
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart and Chest Hospital, Liverpool, United Kingdom
| |
Collapse
|
5
|
Cetik Yildiz S, Demir C, Cengiz M, Irmak H, Cengiz BP, Ayhanci A. In Vitro Antitumor and Antioxidant Capacity as well as Ameliorative Effects of Fermented Kefir on Cyclophosphamide-Induced Toxicity on Cardiac and Hepatic Tissues in Rats. Biomedicines 2024; 12:1199. [PMID: 38927407 PMCID: PMC11200811 DOI: 10.3390/biomedicines12061199] [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: 03/28/2024] [Revised: 05/20/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024] Open
Abstract
Fermented prebiotic and probiotic products with kefir are very important to slow down and prevent the growth of tumors and to treat cancer by stimulating the immune response against tumor cells. Cyclophosphamide (CPx) is widely preferred in cancer treatment but its effectiveness in high doses is restricted because of its side effects. The aim of this study was to investigate the protective effects of kefir against CPx-induced heart and liver toxicity. In an experiment, 42 Wistar albino rats were divided into six treatment groups: the control (Group 1), the group receiving 150 mg/kg CPx (Group 2), the groups receiving 5 and 10 mg/kg kefir (Groups 3 and 4) and the groups receiving 5 and 10 mg/kg kefir + CPx (Group 5 and 6). Fermented kefirs obtained on different days by traditional methods were mixed and given by gavage for 12 days, while a single dose of CPx was administered intraperitoneally (i.p.) on the 12th day of the experiment. It was observed that alanine transaminase (ALT), aspartate transaminase (AST), alkaline phosphatase (ALP), lactate dehydrogenase (LDH), creatinine kinase-MB (CK-MB), ischemia modified albumin (IMA) and Troponin I values, which indicate oxidative stress, increased in the CPx-administered group, and this level approached that of the control in the CPx + kefir groups. Likewise, as a result of the kefir, the rats' CPx-induced histopathological symptoms were reduced, and their heart and liver tissue were significantly improved. In conclusion, it was observed that kefir had a cytoprotective effect against CPx-induced oxidative stress, hepatotoxicity and cardiotoxicity, bringing their biochemical parameters closer to those of the control by suppressing oxidative stress and reducing tissue damage.
Collapse
Affiliation(s)
- Songul Cetik Yildiz
- Department of Medical Services and Techniques, Health Services Vocational School, Mardin Artuklu University, 47200 Mardin, Türkiye;
| | - Cemil Demir
- Department of Medical Services and Techniques, Health Services Vocational School, Mardin Artuklu University, 47200 Mardin, Türkiye;
| | - Mustafa Cengiz
- Department of Elementary Education, Faculty of Education, Siirt University, 56100 Siirt, Türkiye;
| | - Halit Irmak
- Department of Computer Sciences, Mardin Artuklu University, 47200 Mardin, Türkiye;
| | | | - Adnan Ayhanci
- Department of Biology, Science Faculty, Eskisehir Osmangazi University, 26040 Eskisehir, Türkiye;
| |
Collapse
|
6
|
Nishikawa T, Miyahara E, Yamazaki I, Ikawa K, Nakagawa S, Kodama Y, Kawano Y, Okamoto Y. Effects of High-Dose Cyclophosphamide on Ultrastructural Changes and Gene Expression Profiles in the Cardiomyocytes of C57BL/6J Mice. Diseases 2024; 12:85. [PMID: 38785740 PMCID: PMC11120609 DOI: 10.3390/diseases12050085] [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: 04/02/2024] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024] Open
Abstract
The pathogenesis of cyclophosphamide (CY)-induced cardiotoxicity remains unknown, and methods for its prevention have not been established. To elucidate the acute structural changes that take place in myocardial cells and the pathways leading to myocardial damage under high-dose CY treatments, we performed detailed pathological analyses of myocardial tissue obtained from C57BL/6J mice subjected to a high-dose CY treatment. Additionally, we analysed the genome-wide cardiomyocyte expression profiles of mice subjected to the high-dose CY treatment. Treatment with CY (400 mg/kg/day intraperitoneally for two days) caused marked ultrastructural aberrations, as observed using electron microscopy, although these aberrations could not be observed using optical microscopy. The expansion of the transverse tubule and sarcoplasmic reticulum, turbulence in myocardial fibre travel, and a low contractile protein density were observed in cardiomyocytes. The high-dose CY treatment altered the cardiomyocyte expression of 1210 genes (with 675 genes upregulated and 535 genes downregulated) associated with cell-cell junctions, inflammatory responses, cardiomyopathy, and cardiac muscle function, as determined using microarray analysis (|Z-score| > 2.0). The expression of functionally important genes related to myocardial contraction and the regulation of calcium ion levels was validated using real-time polymerase chain reaction analysis. The results of the gene expression profiling, functional annotation clustering, and Kyoto Encyclopedia of Genes and Genomes pathway functional-classification analysis suggest that CY-induced cardiotoxicity is associated with the disruption of the Ca2+ signalling pathway.
Collapse
Affiliation(s)
- Takuro Nishikawa
- Department of Pediatrics, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8520, Japan; (E.M.); (S.N.); (Y.K.); (Y.K.); (Y.O.)
| | - Emiko Miyahara
- Department of Pediatrics, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8520, Japan; (E.M.); (S.N.); (Y.K.); (Y.K.); (Y.O.)
| | | | - Kazuro Ikawa
- Department of Clinical Pharmacotherapy, Hiroshima University, Hiroshima 734-8553, Japan;
| | - Shunsuke Nakagawa
- Department of Pediatrics, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8520, Japan; (E.M.); (S.N.); (Y.K.); (Y.K.); (Y.O.)
| | - Yuichi Kodama
- Department of Pediatrics, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8520, Japan; (E.M.); (S.N.); (Y.K.); (Y.K.); (Y.O.)
| | - Yoshifumi Kawano
- Department of Pediatrics, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8520, Japan; (E.M.); (S.N.); (Y.K.); (Y.K.); (Y.O.)
| | - Yasuhiro Okamoto
- Department of Pediatrics, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8520, Japan; (E.M.); (S.N.); (Y.K.); (Y.K.); (Y.O.)
| |
Collapse
|
7
|
Hayek SS, Zaha VG, Bogle C, Deswal A, Langston A, Rotz S, Vasbinder A, Yang E, Okwuosa T. Cardiovascular Management of Patients Undergoing Hematopoietic Stem Cell Transplantation: From Pretransplantation to Survivorship: A Scientific Statement From the American Heart Association. Circulation 2024; 149:e1113-e1127. [PMID: 38465648 DOI: 10.1161/cir.0000000000001220] [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] [Indexed: 03/12/2024]
Abstract
Hematopoietic stem cell transplantation can cure various disorders but poses cardiovascular risks, especially for elderly patients and those with cardiovascular diseases. Cardiovascular evaluations are crucial in pretransplantation assessments, but guidelines are lacking. This American Heart Association scientific statement summarizes the data on transplantation-related complications and provides guidance for the cardiovascular management throughout transplantation. Hematopoietic stem cell transplantation consists of 4 phases: pretransplantation workup, conditioning therapy and infusion, immediate posttransplantation period, and long-term survivorship. Complications can occur during each phase, with long-term survivors facing increased risks for late effects such as cardiovascular disease, secondary malignancies, and endocrinopathies. In adults, arrhythmias such as atrial fibrillation and flutter are the most frequent acute cardiovascular complication. Acute heart failure has an incidence ranging from 0.4% to 2.2%. In pediatric patients, left ventricular systolic dysfunction and pericardial effusion are the most common cardiovascular complications. Factors influencing the incidence and risk of complications include pretransplantation therapies, transplantation type (autologous versus allogeneic), conditioning regimen, comorbid conditions, and patient age. The pretransplantation cardiovascular evaluation consists of 4 steps: (1) initial risk stratification, (2) exclusion of high-risk cardiovascular disease, (3) assessment of cardiac reserve, and (4) optimization of cardiovascular reserve. Clinical risk scores could be useful tools for the risk stratification of adult patients. Long-term cardiovascular management of hematopoietic stem cell transplantation survivors includes optimizing risk factors, monitoring, and maintaining a low threshold for evaluating cardiovascular causes of symptoms. Future research should prioritize refining risk stratification and creating evidence-based guidelines and strategies to optimize outcomes in this growing patient population.
Collapse
|
8
|
Yahyazadeh A, Başak F, Demirel MA. Efficacy of coenzyme Q10 and curcumin on antioxidant enzyme activity and hippocampal alteration following exposure to cyclophosphamide in male rat. Tissue Cell 2024; 86:102296. [PMID: 38184921 DOI: 10.1016/j.tice.2023.102296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 12/03/2023] [Accepted: 12/27/2023] [Indexed: 01/09/2024]
Abstract
Coenzyme Q10 (KQ10) and curcumin (KUR) supplements are extensively used for their potential antioxidant, anticancer, and antiapoptotic properties. The present study investigated the neuroprotective potential of KQ10 and KUR against the side effect of cyclophosphamide (SF) (150 mg/kg) on the hippocampus of male Wistar albino rats. Forty-nine 10-12 weeks old rats were randomly divided into seven groups: control, olive oil (OL), SF, KQ10, KUR, SF+KQ10, and SF+KUR. Our biochemical finding showed a significant decrease in superoxide dismutase (SOD) level in the SF group compared to the control group (p < 0.05). There was also a significant reduction in the total number of the hippocampal pyramidal neurons in the CA1, CA2, and CA1-3 regions in the SF group compared to the control group (p < 0.05). In the SF+KQ10 group, we found a significant increase in serum SOD level and the total number of the hippocampal pyramidal neurons in the CA1, CA2, and CA1-3 regions compared to the SF group (p < 0.05). Immunohistochemical and histopathological examination exhibited noteworthy findings in the hippocampus tissues. Our findings showed that KQ10 administration significantly mitigated the hippocampal alteration caused by SF through enhancing antioxidant enzyme activity and reducing apoptosis. However, we found no protective activity of KUR on the hippocampus tissue, which may be due to its weak antioxidative activity.
Collapse
Affiliation(s)
- Ahmad Yahyazadeh
- Department of Histology and Embryology, Faculty of Medicine, Karabuk University, Karabuk, Turkey.
| | - Feyza Başak
- Department of Histology and Embryology, Faculty of Medicine, Karabuk University, Karabuk, Turkey
| | - Mürşide Ayşe Demirel
- Laboratory Animals Breeding and Experimental Research Centre, Department of Basic Pharmaceutical Sciences, Faculty of Pharmacy, Gazi University, Turkey
| |
Collapse
|
9
|
Adeyemi DH, Hamed MA, Oluwole DT, Omole AI, Akhigbe RE. Acetate attenuates cyclophosphamide-induced cardiac injury via inhibition of NF-kB signaling and suppression of caspase 3-dependent apoptosis in Wistar rats. Biomed Pharmacother 2024; 170:116019. [PMID: 38128178 DOI: 10.1016/j.biopha.2023.116019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/08/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023] Open
Abstract
AIM The goal of the current study was to examine the potential therapeutic effects of sodium acetate on cardiac toxicities caused by cyclophosphamide in Wistar rats. The possible involvement of NF-kB/caspase 3 signaling was also explored. MAIN METHODS Thirty-two male Wistar rats were divided into four groups at random. (n = 8). The control animals received 0.5 mL of distilled water orally for 14 days, the acetate-treated group received 200 mg/kg/day of sodium acetate orally for 14 consecutive days, and cyclophosphamide-treated rats received 150 mg/kg /day of cyclophosphamide i.p. on day 8, while cyclophosphamide + acetate group received sodium acetate and cyclophosphamide as earlier stated. KEY FINDINGS Results showed that cyclophosphamide-induced cardiotoxicity, which manifested as a marked drop in body and cardiac weights as well as cardiac weight/tibial length, increased levels of troponin, C-reactive protein, lactate, and creatinine kinase, and lactate dehydrogenase activities in the plasma and cardiac tissue. Histopathological examination also revealed toxic cardiac histopathological changes. These alterations were associated with a significant increase in xanthine oxidase and myeloperoxidase activities, uric acid, malondialdehyde, TNF-α, IL-1β, NFkB, DNA fragmentation, and caspase 3 and caspase 9 activities in addition to a marked decline in Nrf2 and GSH levels, and SOD and catalase activities in the cardiac tissue. Acetate co-administration significantly attenuated cyclophosphamide cardiotoxicity by its antioxidant effect, preventing NFkB activation and caspase 9/caspase 3 signalings. SIGNIFICANCE This study shows that acetate co-administration may have cardio-protective effects against cyclophosphamide-induced cardiotoxicity by inhibiting NF-kB signaling and suppressing caspase-3-dependent apoptosis.
Collapse
Affiliation(s)
- D H Adeyemi
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Osun State University, Osun State, Nigeria
| | - M A Hamed
- Department of Medical Laboratory Sciences, Afe Babalola University, Ado Ekiti, Ekiti State, Nigeria; The Brainwill Laboratories, Osogbo, Osun State, Nigeria; Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Osun State, Nigeria
| | - D T Oluwole
- Department of Physiology, Crescent University, Abeokuta, Ogun State, Nigeria
| | - A I Omole
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Osun State, Nigeria; Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - R E Akhigbe
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Osun State, Nigeria; Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria.
| |
Collapse
|
10
|
Jali AM, Alam MF, Hanbashi A, Mawkili W, Abdlasaed BM, Alshahrani S, Qahl AM, Alrashah ASS, Shahi HA. Sesamin's Therapeutic Actions on Cyclophosphamide-Induced Hepatotoxicity, Molecular Mechanisms, and Histopathological Characteristics. Biomedicines 2023; 11:3238. [PMID: 38137459 PMCID: PMC10741447 DOI: 10.3390/biomedicines11123238] [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: 10/12/2023] [Revised: 11/22/2023] [Accepted: 11/28/2023] [Indexed: 12/24/2023] Open
Abstract
Cyclophosphamide, an alkylating agent integral to specific cancer chemotherapy protocols, is often curtailed in application owing to its significant hepatotoxic side effects. Therefore, this study was conducted to assess the hepatoprotective potential of sesamin, a plant-originated antioxidant, using rat models. The rats were divided into five groups: a control group received only the vehicle for six days; a cyclophosphamide group received an intraperitoneal (i.p.) single injection of cyclophosphamide (150 mg/kg) on day four; a sesamin group received a daily high oral dose (20 mg/kg) of sesamin for six days; and two groups were pretreated with oral sesamin (10 and 20 mg/kg daily from day one to day six) followed by an i.p. injection of cyclophosphamide on day four. The final and last sesamin dose was administered 24 h before euthanasia. At the end of the experiment, blood and liver tissue were collected for biochemical and histopathological assessments. The results indicated significantly increased liver markers (AST, ALT, ALP, and BIL), cytokines (TNFα and IL-1β), caspase-3, and malondialdehyde (MDA) in the cyclophosphamide group as compared to the normal control. Additionally, there was a significant decline in antioxidants (GSH) and antioxidant enzymes (CAT and SOD), but the sesamin treatment reduced liver marker enzymes, cytokines, and caspase-3 and improved antioxidants and antioxidant enzymes. Thus, sesamin effectively countered these alterations and helped to normalize the histopathological alterations. In conclusion, sesamin demonstrated the potential for attenuating cyclophosphamide-induced hepatotoxicity by modulating cytokine networks, apoptotic pathways, and oxidative stress, suggesting its potential role as an adjunct in chemotherapy to reduce hepatotoxicity.
Collapse
Affiliation(s)
- Abdulmajeed M. Jali
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia; (A.M.J.); (A.H.); (W.M.); (S.A.); (A.M.Q.); (A.S.S.A.); (H.A.S.)
| | - Mohammad Firoz Alam
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia; (A.M.J.); (A.H.); (W.M.); (S.A.); (A.M.Q.); (A.S.S.A.); (H.A.S.)
| | - Ali Hanbashi
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia; (A.M.J.); (A.H.); (W.M.); (S.A.); (A.M.Q.); (A.S.S.A.); (H.A.S.)
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Wedad Mawkili
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia; (A.M.J.); (A.H.); (W.M.); (S.A.); (A.M.Q.); (A.S.S.A.); (H.A.S.)
| | - Basher M. Abdlasaed
- Department of Biology, Faculty of Education, Alasmaray Islamic University, Zliten 218521, Libya;
| | - Saeed Alshahrani
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia; (A.M.J.); (A.H.); (W.M.); (S.A.); (A.M.Q.); (A.S.S.A.); (H.A.S.)
| | - Abdullah M. Qahl
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia; (A.M.J.); (A.H.); (W.M.); (S.A.); (A.M.Q.); (A.S.S.A.); (H.A.S.)
- Pharmacy Department, Jazan University Hospital, Jazan University, Jazan 45142, Saudi Arabia
| | - Ahmad S. S. Alrashah
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia; (A.M.J.); (A.H.); (W.M.); (S.A.); (A.M.Q.); (A.S.S.A.); (H.A.S.)
- Pharmacy Administration, Ministry of Health, Health Affairs General Directorate, Najran 66251, Saudi Arabia
| | - Hamad Al Shahi
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia; (A.M.J.); (A.H.); (W.M.); (S.A.); (A.M.Q.); (A.S.S.A.); (H.A.S.)
| |
Collapse
|
11
|
İpek E, Hesapçıoğlu M, Karaboğa M, Avcı H. Selenium protection from DNA damage and regulation of apoptosis signaling following cyclophosphamide induced cardiotoxicity in rats. Biotech Histochem 2023; 98:534-542. [PMID: 37695070 DOI: 10.1080/10520295.2023.2253424] [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: 09/12/2023] Open
Abstract
We investigated the mechanism of the cardioprotective effect of selenium (Se) against cyclophosphamide (CPA) induced cardiotoxicity in rats. We divided 24 female Wistar albino rats into four groups. The control group was injected intraperitoneally (i.p.) with normal saline. The CPA group was injected i.p. with 200 mg/kg CPA. The Se group was injected i.p. with 1 mg/kg Se. The CPA + Se group was injected i.p. with 200 mg/kg CPA and 1 mg/kg Se. Rats were euthanized 24 h after injection and heart tissues were harvested. Histopathological examination revealed reduced severity of myocardial lesions in the CPA + Se group compared to CPA induced cardiotoxicity of the CPA group; this finding was confirmed by increased immunoreactivity of cardiac troponin-I (cTn-I) in the CPA + Se group compared to decreased cTn-I immunoreactivity in the CPA group. Administration of CPA increased the immunoreactivity of phosphorylated histone-2AX (γH2AX). Se reduced the CPA induced increase in γH2AX immunoreactivity. Se administration reversed the CPA induced increase of Bax and decrease of Bcl2 gene expressions. Our findings suggest that Se is cardioprotective by reducing DNA damage and regulating the genes responsible for apoptosis caused by CPA in rats.
Collapse
Affiliation(s)
- Emrah İpek
- Department of Pathology, Faculty of Veterinary Medicine, Aydın Adnan Menderes University, Aydın, Turkey
| | - Mehmet Hesapçıoğlu
- Department of Pathology, Faculty of Veterinary Medicine, Aydın Adnan Menderes University, Aydın, Turkey
| | - Mehmet Karaboğa
- Department of Pathology, Faculty of Veterinary Medicine, Aydın Adnan Menderes University, Aydın, Turkey
| | - Hamdi Avcı
- Department of Pathology, Faculty of Veterinary Medicine, Aydın Adnan Menderes University, Aydın, Turkey
| |
Collapse
|
12
|
Xie Y, Li Y, Chen J, Ding H, Zhang X. Early growth response-1: Key mediators of cell death and novel targets for cardiovascular disease therapy. Front Cardiovasc Med 2023; 10:1162662. [PMID: 37057102 PMCID: PMC10086247 DOI: 10.3389/fcvm.2023.1162662] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
SignificanceCardiovascular diseases are seen to be a primary cause of death, and their prevalence has significantly increased across the globe in the past few years. Several studies have shown that cell death is closely linked to the pathogenesis of cardiovascular diseases. Furthermore, many molecular and cellular mechanisms are involved in the pathogenesis of the cardiac cell death mechanism. One of the factors that played a vital role in the pathogenesis of cardiac cell death mechanisms included the early growth response-1 (Egr-1) factor.Recent AdvancesStudies have shown that abnormal Egr-1 expression is linked to different animal and human disorders like heart failure and myocardial infarction. The biosynthesis of Egr-1 regulates its activity. Egr-1 can be triggered by many factors such as serum, cytokines, hormones, growth factors, endotoxins, mechanical injury, hypoxia, and shear stress. It also displays a pro-apoptotic effect on cardiac cells, under varying stress conditions. EGR1 mediates a broad range of biological responses to oxidative stress and cell death by combining the acute changes occurring in the cellular environment with sustained changes in gene expression.Future DirectionsThe primary regulatory role played by the Egr-1-targeting DNAzymes, microRNAs, and oligonucleotide decoy strategies in cardiovascular diseases were identified to provide a reference to identify novel therapeutic targets for cardiovascular diseases.
Collapse
Affiliation(s)
- Yixin Xie
- Department of Cardiology, Lanzhou University Second Hospital, Lanzhou, China
| | - Yongnan Li
- Department of Cardiac Surgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Jianshu Chen
- Department of Cardiology, Lanzhou University Second Hospital, Lanzhou, China
| | - Hong Ding
- Department of Cardiology, Lanzhou University Second Hospital, Lanzhou, China
| | - Xiaowei Zhang
- Department of Cardiology, Lanzhou University Second Hospital, Lanzhou, China
- Correspondence: Xiaowei Zhang
| |
Collapse
|
13
|
Glutathione system enhancement for cardiac protection: pharmacological options against oxidative stress and ferroptosis. Cell Death Dis 2023; 14:131. [PMID: 36792890 PMCID: PMC9932120 DOI: 10.1038/s41419-023-05645-y] [Citation(s) in RCA: 72] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/31/2023] [Accepted: 02/02/2023] [Indexed: 02/17/2023]
Abstract
The glutathione (GSH) system is considered to be one of the most powerful endogenous antioxidant systems in the cardiovascular system due to its key contribution to detoxifying xenobiotics and scavenging overreactive oxygen species (ROS). Numerous investigations have suggested that disruption of the GSH system is a critical element in the pathogenesis of myocardial injury. Meanwhile, a newly proposed type of cell death, ferroptosis, has been demonstrated to be closely related to the GSH system, which affects the process and outcome of myocardial injury. Moreover, in facing various pathological challenges, the mammalian heart, which possesses high levels of mitochondria and weak antioxidant capacity, is susceptible to oxidant production and oxidative damage. Therefore, targeted enhancement of the GSH system along with prevention of ferroptosis in the myocardium is a promising therapeutic strategy. In this review, we first systematically describe the physiological functions and anabolism of the GSH system, as well as its effects on cardiac injury. Then, we discuss the relationship between the GSH system and ferroptosis in myocardial injury. Moreover, a comprehensive summary of the activation strategies of the GSH system is presented, where we mainly identify several promising herbal monomers, which may provide valuable guidelines for the exploration of new therapeutic approaches.
Collapse
|
14
|
Therapeutic Potential of Capsaicin against Cyclophosphamide-Induced Liver Damage. J Clin Med 2023; 12:jcm12030911. [PMID: 36769559 PMCID: PMC9917381 DOI: 10.3390/jcm12030911] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/06/2023] [Accepted: 01/19/2023] [Indexed: 01/25/2023] Open
Abstract
Cyclophosphamide (CPM) is a classical alkylating agent used in different cancer chemotherapy regimens and is restricted due to severe adverse effects, including hepatotoxicity. Natural or plant-derived antioxidants such as capsaicin were utilized in this study to examine the hepatoprotective benefits against cyclophosphamide-induced hepatotoxicity. The rats were divided into five groups: a normal control group, a toxic group (CPM), an intraperitoneal injection of a single dose of 200 mg/kg b.w. on the fourth day, a pretreated group with two doses of CPS (10 mg and 20 mg/kg b.w.) orally for six consecutive days, and an intraperitoneal administration of 200 mg/kg b.w. on the fourth day of treatment. The fifth group was administered with the highest dose of CPS (20 mg/kg b.w.) orally for six consecutive days. After 24 h of administration of CPS, the rats were anesthetized, blood was collected, and the serum enzyme toxicity was evaluated. After the blood sampling and euthanasia of all the animals, the liver was isolated for further toxicity and histopathological examination. The results revealed that serum liver markers (AST, ALT, ALP, BLI) significantly increased after CPM administration, but were subsequently restored after CPS treatment with both doses. In addition, lipid peroxidation (MDA), inflammatory cytokines (IL-1β, TNF-α), and apoptotic markers (Caspase-3) increased, and antioxidant enzymes (GSH, CAT, SOD) were significantly decreased after CPM administration, and it was re-established by CPS treatment. However, CPS effectively protected against the CPM-induced histopathological architects of liver tissues. In conclusion, CPS attenuates CPM-induced hepatotoxicity via modulating oxidative stress, apoptotic signals, and cytokine pathway. Therefore, CPS could play a significant role as a supplement during the chemotherapy of patients.
Collapse
|
15
|
Assessing Drug-Induced Mitochondrial Toxicity in Cardiomyocytes: Implications for Preclinical Cardiac Safety Evaluation. Pharmaceutics 2022; 14:pharmaceutics14071313. [PMID: 35890211 PMCID: PMC9319223 DOI: 10.3390/pharmaceutics14071313] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/09/2022] [Accepted: 06/14/2022] [Indexed: 02/07/2023] Open
Abstract
Drug-induced cardiotoxicity not only leads to the attrition of drugs during development, but also contributes to the high morbidity and mortality rates of cardiovascular diseases. Comprehensive testing for proarrhythmic risks of drugs has been applied in preclinical cardiac safety assessment for over 15 years. However, other mechanisms of cardiac toxicity have not received such attention. Of them, mitochondrial impairment is a common form of cardiotoxicity and is known to account for over half of cardiovascular adverse-event-related black box warnings imposed by the U.S. Food and Drug Administration. Although it has been studied in great depth, mitochondrial toxicity assessment has not yet been incorporated into routine safety tests for cardiotoxicity at the preclinical stage. This review discusses the main characteristics of mitochondria in cardiomyocytes, drug-induced mitochondrial toxicities, and high-throughput screening strategies for cardiomyocytes, as well as their proposed integration into preclinical safety pharmacology. We emphasize the advantages of using adult human primary cardiomyocytes for the evaluation of mitochondrial morphology and function, and the need for a novel cardiac safety testing platform integrating mitochondrial toxicity and proarrhythmic risk assessments in cardiac safety evaluation.
Collapse
|
16
|
Dionísio F, Araújo AM, Duarte-Araújo M, Bastos MDL, Guedes de Pinho P, Carvalho F, Costa VM. Cardiotoxicity of cyclophosphamide's metabolites: an in vitro metabolomics approach in AC16 human cardiomyocytes. Arch Toxicol 2022; 96:653-671. [PMID: 35088106 DOI: 10.1007/s00204-021-03204-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 12/09/2021] [Indexed: 12/11/2022]
Abstract
Cyclophosphamide is a widely used anticancer and immunosuppressive prodrug that unfortunately causes severe adverse effects, including cardiotoxicity. Although the exact cardiotoxic mechanisms are not completely understood, a link between cyclophosphamide's pharmacologically active metabolites, namely 4-hydroxycyclophosphamide and acrolein, and the toxicity observed after the administration of high doses of the prodrug is likely. Therefore, the objective of this study is to shed light on the cardiotoxic mechanisms of cyclophosphamide and its main biotransformation products, through classic and metabolomics studies. Human cardiac proliferative and differentiated AC16 cells were exposed to several concentrations of the three compounds, determining their basic cytotoxic profile and preparing the next study, using subtoxic and toxic concentrations for morphological and biochemical studies. Finally, metabolomics studies were applied to cardiac cells exposed to subtoxic concentrations of the aforementioned compounds to determine early markers of damage. The cytotoxicity, morphological and biochemical assays showed that 4-hydroxycyclophosphamide and acrolein induced marked cardiotoxicity at µM concentrations (lower than 5 µM), being significantly lower than the ones observed for cyclophosphamide (higher than 2500 μM). Acrolein led to increased levels of ATP and total glutathione on proliferative cells at 25 µM, while no meaningful changes were observed in differentiated cells. Higher levels of carbohydrates and decreased levels of fatty acids and monoacylglycerols indicated a metabolic cardiac shift after exposure to cyclophosphamide's metabolites, as well as a compromise of precursor amino acids used in the synthesis of glutathione, seen in proliferative cells' metabolome. Overall, differences in cytotoxic mechanisms were observed for the two different cellular states used and for the three molecules, which should be taken into consideration in the study of cyclophosphamide cardiotoxic mechanisms.
Collapse
Affiliation(s)
- Flávio Dionísio
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira nº 228, 4050-313, Porto, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
| | - Ana Margarida Araújo
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira nº 228, 4050-313, Porto, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
| | - Margarida Duarte-Araújo
- LAQV/REQUIMTE, Department of Imuno-Physiology and Pharmacology, Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Maria de Lourdes Bastos
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira nº 228, 4050-313, Porto, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
| | - Paula Guedes de Pinho
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira nº 228, 4050-313, Porto, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
| | - Félix Carvalho
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira nº 228, 4050-313, Porto, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
| | - Vera Marisa Costa
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira nº 228, 4050-313, Porto, Portugal.
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.
| |
Collapse
|
17
|
Ahmed OM, Ahmed AA, Fahim HI, Zaky MY. Quercetin and naringenin abate diethylnitrosamine/acetylaminofluorene-induced hepatocarcinogenesis in Wistar rats: the roles of oxidative stress, inflammation and cell apoptosis. Drug Chem Toxicol 2022; 45:262-273. [PMID: 31665932 DOI: 10.1080/01480545.2019.1683187] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 10/10/2019] [Accepted: 10/15/2019] [Indexed: 01/10/2023]
Abstract
This study was designed to assess the preventive effects and to suggest the probable mechanisms of action of quercetin and naringein in diethylnitrosamine (DEN)/2-acetylaminofluorene (2AAF)-induced hepatocarcinogenesis in Wistar male rats. The chemical-induction of hepatocarcinogenesis was performed by injection of DEN intraperitoneally at 150 mg/kg body weight (b.w.) twice/week for two weeks, followed by oral administration of 2AAF at 20 mg/kg body weight (b.w.) 4 times/week for 3 weeks. The DEN/2AAF-administered rats were co-treated with quercetin and naringenin at dose level of 10 mg/kg b. w. by oral gavage for 20 weeks. The treatment of DEN/2AAF-administered rats with quercetin and naringenin significantly prevented the elevations in serum levels of liver function indicators (ALT, AST, ALP, γ-GT, total bilirubin and albumin) and liver tumor biomarkers including AFP, CEA and CA19.9. The cancerous histological lesions and inflammatory cells infiltration in liver of DEN/2AAF-administered rats were remarkably suppressed by treatments with quercetin and naringenin. The hepatic oxidative stress markers including NO level and lipid peroxidation significantly decreased while the SOD, GPx and CAT activities and GSH content significantly increased in DEN/2AAF-administered rats treated with quercetin and naringenin when compared to DEN/2AFF-administered control rats. Furthermore, the lowered mRNA expression of liver IL-4, P53 and Bcl-2 in of DEN/2AAF-administered rats were significantly counteracted by treatment with quercetin and naringenin. Taken together, our results demonstrate that quercetin and naringenin may abate hepatocarcinogenesis via enhancement of anti-inflammatory, anti-oxidant and apoptotic actions.
Collapse
Affiliation(s)
- Osama M Ahmed
- Molecular Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| | - Adel A Ahmed
- Molecular Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| | - Hanaa I Fahim
- Molecular Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| | - Mohamed Y Zaky
- Molecular Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|
18
|
Poojary KK, Nayak G, Vasani A, Kumari S, Dcunha R, Kunhiraman JP, Gopalan D, Rao RR, Mutalik S, Kalthur SG, Murari MS, Raghu SV, Adiga SK, Kalthur G. Curcumin nanocrystals attenuate cyclophosphamide-induced testicular toxicity in mice. Toxicol Appl Pharmacol 2021; 433:115772. [PMID: 34715073 DOI: 10.1016/j.taap.2021.115772] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 10/09/2021] [Accepted: 10/18/2021] [Indexed: 12/11/2022]
Abstract
The cancer therapy using cyclophosphamide (CP) has been associated with adverse effects on the testicular function that raises concerns about the future fertility potential among cancer survivors. Curcumin, a polyphenol, has shown to possess a plethora of biological functions including tissue protective effects. In the present study, we investigated the protective effects of curcumin nanocrystals (NC) in mitigation of CP-induced testicular toxicity. Healthy adult (8-10 week) and prepubertal (2 week) male Swiss albino mice were injected with a single dose of CP (200 mg/kg) intraperitoneally (i.p). NC (4 mg/kg, i.p.) was administered every alternate day, for 35 days in adult mice while, a single dose of NC was injected intraperitoneally to prepubertal mice 1 h prior to CP. Administration of multiple doses of NC ameliorated CP-induced testicular toxicity in adult mice, which was evident from the improved sperm functional competence, sperm chromatin condensation, seminiferous tubule architecture and decreased apoptosis in testicular cells. Further, administration of NC 1 h prior to CP in prepubertal mice modulated the expression of genes pertaining to proliferation, pluripotency, DNA damage and DNA repair in spermatogonial cells at 24 h after the treatment. Overall, these results suggest that NC could be a promising chemoprotective agent, which can have potential application in male fertility preservation.
Collapse
Affiliation(s)
- Keerthana Karunakar Poojary
- Division of Clinical Embryology, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, India
| | - Guruprasad Nayak
- Division of Reproductive Biology, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, India
| | - Ashna Vasani
- Division of Reproductive Biology, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, India
| | - Sandhya Kumari
- Division of Reproductive Biology, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, India
| | - Reyon Dcunha
- Division of Reproductive Biology, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, India
| | - Jyolsna Ponnaratta Kunhiraman
- Division of Reproductive Biology, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, India
| | - Divya Gopalan
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Rajat Radhakrishna Rao
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Srinivas Mutalik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Sneha Guruprasad Kalthur
- Department of Anatomy, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, India
| | - M S Murari
- DST PURSE Program, Mangalore University, Mangalagangotri 574199, Karnataka, India
| | - Shamprasad Varija Raghu
- Neurogenetics Lab, Department of Applied Zoology, Mangalore University, Mangalagangotri 574199, Karnataka, India
| | - Satish Kumar Adiga
- Division of Clinical Embryology, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, India
| | - Guruprasad Kalthur
- Division of Reproductive Biology, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, India.
| |
Collapse
|
19
|
Oztopuz O, Coskun O, Buyuk B. Alterations in aquaporin gene expression level on cyclophosphamide-induced cardiac injury and possible protective role of Ganoderma lucidum. Biologia (Bratisl) 2021. [DOI: 10.1007/s11756-021-00817-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
20
|
Khordad E, Alipour F, Pourabbas M, Mansouri S, Salimnejad R. Hepatoprotective Impact of Ghrelin against Cyclophosphamide-Induced Toxicity in the Male Mice. Drug Res (Stuttg) 2021; 71:407-412. [PMID: 34282601 DOI: 10.1055/a-1508-5368] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Despite its vast spectrum of clinical usage, cyclophosphamide (CP) exerts many adverse impacts, including hepatotoxicity. Antioxidant properties of ghrelin might protect the liver from CP-induced toxicity. The current study aimed to assess the protective impacts of ghrelin on CP-induced liver toxicity. METHODS Forty male mice were randomly divided into four groups (n=10) Group 1 as control received no intervention,group 2 received cyclophosphamide (CP) (100 mg/kg, i.p.) for five weeks and once a week. Group 3 received CP+ghrelin (CP+G), (80 µg/kg daily, i.p.) for five weeks. Group 4 received ghrelin with above-mentioned dose. At the end of the experiment, the mice were sacrificed to remove liver tissuesfor histological and biochemical examination. RESULTS Malondialdehyde (MDA) level increased after CP treatment but ghrelin administration significantly decreased the level of MDA (P<0.05). Measurement of the total antioxidant capacity (TAC) noted a significant decrease in the CP group against the control group (P<0.05). Ghrelin treatment in the CP+G group considerably increased the TAC activity when compared to the CP group (P<0.05). Histological examinations also confirmed the hepatocyte necrosis, local bleeding and inflammation, vacuolation, and sinusoidal dilation in the CP group, ghrelin administration reduced the destructive effects of CP on the liver significantly (P<0.05). CONCLUSION Our results reveal the hepatoprotective effect of ghrelin against CP. Therefore, ghrelin might be useful in protecting the body against the adverse impacts of injuries induced by chemotherapeutic drugs.
Collapse
Affiliation(s)
- Elnaz Khordad
- Department of Physiology, School of Paramedical Sciences, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
- Neuroscience Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Fatemeh Alipour
- Department of Anatomy and Cellular Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahdieh Pourabbas
- Student Research Committee, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Somaieh Mansouri
- Department of Anatomy, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Ramin Salimnejad
- Department of Anatomical Sciences and Pathology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
21
|
Hassan RA, Hozayen WG, Abo Sree HT, Al-Muzafar HM, Amin KA, Ahmed OM. Naringin and Hesperidin Counteract Diclofenac-Induced Hepatotoxicity in Male Wistar Rats via Their Antioxidant, Anti-Inflammatory, and Antiapoptotic Activities. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9990091. [PMID: 34422219 PMCID: PMC8376442 DOI: 10.1155/2021/9990091] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/27/2021] [Accepted: 07/14/2021] [Indexed: 01/18/2023]
Abstract
This study is aimed at evaluating the preventive effect and at suggesting the mode of actions of naringin and hesperidin and their combination in diclofenac-induced hepatotoxicity. Male Wistar rats, intraperitoneally injected with diclofenac sodium (3 mg/kg b.wt/day), were orally treated with naringin (20 mg/kg b.wt/day) and hesperidin (20 mg/kg b.wt/day) and their combination for 4 weeks. The administrations of naringin and hesperidin to diclofenac-injected rats led to a significant decrease in the elevated serum ALT, AST, LDH, ALP, GGT, total bilirubin, TNF-α, and IL-17 levels as well as liver lipid peroxidation and liver p53 and caspase-3 mRNA expressions. In contrast, serum IL-4 level, liver GSH content, and liver GPx and SOD activities increased. In association, diclofenac-induced deleterious histological alterations including hydropic degeneration, cytoplasmic vacuolization, apoptosis, and focal hepatic necrosis of hepatocytes associated with inflammatory cells' infiltration were remarkably improved by treatments with naringin and hesperidin. In conclusion, naringin, hesperidin, and their combination, which was the most potent, counteract diclofenac-induced liver injury via antioxidant, anti-inflammatory, and antiapoptotic actions. Thus, this study recommends the use of naringin and hesperidin or their combination to resolve the side effects of drugs like diclofenac on the liver.
Collapse
Affiliation(s)
- Rasha A. Hassan
- Department of Biochemistry, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| | - Walaa G. Hozayen
- Department of Biochemistry, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| | - Haidy T. Abo Sree
- Faculty of Oral and Dental Medicine, Nahda University, Beni-Suef, Egypt
| | - Hessah M. Al-Muzafar
- Chemistry Department, College of Science, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | - Kamal A. Amin
- Chemistry Department, College of Science, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
- Basic & Applied Scientific Research Center, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | - Osama M. Ahmed
- Physiology Division, Department of Zoology, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| |
Collapse
|
22
|
Afkhami-Ardakani M, Hasanzadeh S, Shahrooz R, Delirezh N, Malekinejad H. Spirulina platensis (Arthrospira platensis) attenuates Cyclophosphamide-induced reproductive toxicity in male Wistar rats: Evidence for sperm apoptosis and p53/Bcl-2 expression. J Food Biochem 2021; 45:e13854. [PMID: 34245022 DOI: 10.1111/jfbc.13854] [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: 04/12/2021] [Revised: 06/14/2021] [Accepted: 06/23/2021] [Indexed: 12/01/2022]
Abstract
Cyclophosphamide is an antitumor agent that causes disorders in fertility. This study aimed to evaluate the protective effects of Spirulina platensis against Cyclophosphamide-induced testicular toxicity. 42 male Wistar rats were randomly divided into six groups. Experimental groups included three groups. The first experimental group received Cyclophosphamide at a dose of 5 mg/kg body weight (BW) orally. The second and third experimental groups received 5 mg/kg BW Cyclophosphamide and 500 and 1,000 mg/kg BW S. platensis orally, respectively. The control groups included a control group, and two S. platensis control groups. Following 28 days, two flow cytometry techniques were used to determine sperm apoptosis and testicular protein expression of tumor protein (p53) and B-cell lymphoma 2 (Bcl-2). p53 is a tumor suppressor protein that causes the cell to enter the apoptosis cycle after DNA damage and Bcl-2 is an anti-apoptotic protein that acts through the mitochondrial pathway of apoptosis. FITC-Annexin V assay was used for sperm apoptosis evaluation. For protein expression assay, primary and secondary antibodies staining were performed. The Cyclophosphamide group showed a significant increase in sperm apoptosis compared to the control group. Cyclophosphamide significantly increased p53 and decreased Bcl-2 expression compared to the control group. S. platensis co-treated groups exhibited a significant decrease in sperm apoptosis compared to the Cyclophosphamide group. Moreover, S. platensis co-treated groups displayed a significant decreasing in p53 and increasing in Bcl-2 expression compared to the Cyclophosphamide group. The results of this study indicated that S. platensis protected rats against Cyclophosphamide-induced reproductive toxicity. PRACTICAL APPLICATIONS: Cyclophosphamide is the chemotherapy agent used to treat different cancers. Cyclophosphamide has side effects on the male reproductive system. Spirulina plantesis has a protective effect because of its antioxidant and anti-apoptotic properties. Co-administration of Spirulina plantesis with Cyclophosphamide reduces sperm apoptosis also decreases P53 protein expression and increases Bcl-2 protein expression. This study validated the anti-apoptotic potential of Spirulina plantesis against Cyclophosphamide-induced male reproductive toxicity.
Collapse
Affiliation(s)
- Mohammad Afkhami-Ardakani
- Department of Comparative Histology and Embryology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Shapour Hasanzadeh
- Department of Comparative Histology and Embryology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Rasoul Shahrooz
- Department of Comparative Histology and Embryology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Norouz Delirezh
- Department of Microbiology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Hasan Malekinejad
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
23
|
A Review on Recent Advancement on Age-Related Hearing Loss: The Applications of Nanotechnology, Drug Pharmacology, and Biotechnology. Pharmaceutics 2021; 13:pharmaceutics13071041. [PMID: 34371732 PMCID: PMC8309044 DOI: 10.3390/pharmaceutics13071041] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/01/2021] [Accepted: 07/01/2021] [Indexed: 11/30/2022] Open
Abstract
Aging is considered a contributing factor to many diseases such as cardiovascular disease, Alzheimer’s disease, and hearing loss. Age-related hearing loss, also termed presbycusis, is one of the most common sensory impairments worldwide, affecting one in five people over 50 years of age, and this prevalence is growing annually. Associations have emerged between presbycusis and detrimental health outcomes, including social isolation and mental health. It remains largely untreatable apart from hearing aids, and with no globally established prevention strategies in the clinical setting. Hence, this review aims to explore the pathophysiology of presbycusis and potential therapies, based on a recent advancement in bile acid-based bio-nanotechnologies. A comprehensive online search was carried out using the following keywords: presbycusis, drugs, hearing loss, bile acids, nanotechnology, and more than 150 publications were considered directly relevant. Evidence of the multifaceted oxidative stress and chronic inflammation involvement in cellular damage and apoptosis that is associated with a loss of hair cells, damaged and inflamed stria vascularis, and neuronal signalling loss and apoptosis continues to emerge. New robust and effective therapies require drug delivery deeper into the various layers of the cochlea. Bile acid-based nanotechnology has gained wide interest in its permeation-enhancing ability and potential for numerous applications in treating presbycusis.
Collapse
|
24
|
Akamo AJ, Rotimi SO, Akinloye DI, Ugbaja RN, Adeleye OO, Dosumu OA, Eteng OE, Amah G, Obijeku A, Cole OE. Naringin prevents cyclophosphamide-induced hepatotoxicity in rats by attenuating oxidative stress, fibrosis, and inflammation. Food Chem Toxicol 2021; 153:112266. [PMID: 33992719 DOI: 10.1016/j.fct.2021.112266] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 05/09/2021] [Accepted: 05/11/2021] [Indexed: 02/06/2023]
Abstract
Cyclophosphamide (CYCP), a synthetic alkylating antineoplastic, disrupts both cancerous and non-cancerous cells to cause cancer regression and multi organotoxicity respectively. CYCP-induced hepatotoxicity is rare but possible. Evidence has shown that naringin has several beneficial potentials against oxidative stress, inflammation, and fibrosis. This study examined the chemoprotective potentials of naringin on exited radical scavenging, hepatic integrity, oxidative stress, fibrosis, and inflammation in CYCP-mediated hepatotoxicity. Rats were pre-treated orally by gavage for fourteen consecutive days with three doses (50, 100, and 200 mg/kg) of naringin before single CYCP (200 mg/kg, i.p.) administration. Subsequently, the rats were euthanized; blood and liver were removed, and assessed for serum and hepatic enzymes, oxidative stress, inflammation, and gene expression dynamics. Naringin concentrations required for 50% scavenging hydroxyl radical and 2,2'-azinobis-(3-ethylbenzothiazoline-6-sulphonic acid) radical cation were 0.32 mg/mL and 0.39 mg/mL, respectively. Pretreatment with naringin significantly (p < 0.05) abolish CYCP-induced changes in the activities of serum and hepatic ALT, AST, GGT, ALP, and LDH. Pretreatment with naringin remarkably (p < 0.05) reversed CYCP-mediated increases in hepatic levels of malondialdehyde, hydroperoxide, and nitric oxide; reverse CYCP-induced decreases in the hepatic glutathione levels, activities of catalase, glutathione peroxidase, and glutathione reductase; and also attenuated CYCP-induced upregulation of expression of hepatic chemokine (C-C motif) ligand 2 (CCL2), interferon alpha1 (IFN-α1), interleukine-1β, interleukine-1 receptor, and transforming growth factor beta 1 (TGF-β1). Taken together, different doses of naringin can prevent CYCP-induced oxidants generation, hepatocytes dysfunctions, oxidative stress as well as inflammatory perturbations in rats when pre-administered for as few as 14 days.
Collapse
Affiliation(s)
- Adio J Akamo
- Department of Biochemistry, Federal University of Agriculture, Abeokuta, Ogun State, Nigeria.
| | - Solomon O Rotimi
- Biochemistry Unit and Molecular Biology Research Laboratory, Department of Biological Sciences, Covenant University, Ota, Ogun State, Nigeria
| | - Dorcas I Akinloye
- Department of Biochemistry, Federal University of Agriculture, Abeokuta, Ogun State, Nigeria
| | - Regina N Ugbaja
- Department of Biochemistry, Federal University of Agriculture, Abeokuta, Ogun State, Nigeria
| | - Oluwagbemiga O Adeleye
- Department of Animal Production and Health, Federal University of Agriculture, Abeokuta, Ogun State, Nigeria
| | - Oluwatosin A Dosumu
- Department of Biochemistry, Federal University of Agriculture, Abeokuta, Ogun State, Nigeria
| | - Ofem E Eteng
- Department of Biochemistry, Federal University of Agriculture, Abeokuta, Ogun State, Nigeria
| | - Gogonte Amah
- Department of Biochemistry, Benjamin Carson (SRN) School of Medicine, Babcock University, Ilisan, Ogun State, Nigeria
| | - Augustine Obijeku
- Department of Biochemistry, Federal University of Agriculture, Abeokuta, Ogun State, Nigeria
| | - Oluwatosin E Cole
- Department of Biochemistry, Federal University of Agriculture, Abeokuta, Ogun State, Nigeria
| |
Collapse
|
25
|
Abd-ElRaouf A, Nada AS, Mohammed NEDA, Amer HA, Abd-ElRahman SS, Abdelsalam RM, Salem HA. Low dose gamma irradiation attenuates cyclophosphamide-induced cardiotoxicity in rats: role of NF-κB signaling pathway. Int J Radiat Biol 2021; 97:632-641. [PMID: 33635746 DOI: 10.1080/09553002.2021.1893856] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/24/2021] [Accepted: 02/14/2021] [Indexed: 01/03/2023]
Abstract
PURPOSE Cyclophosphamide (Cyp) is one of the most commonly used, wide spectrum chemotherapeutic agents. Cyp has multi-organ toxicities that are dose limiting, thus it's mostly used in chemotherapeutic combinations. Radiation is well known as a hazardous sort of energy, recent studies are interested in studying the beneficial therapeutic effects of low-dose gamma radiation. This study examined the protective effect of two different doses/dose-rates of irradiation either alone or combined with telmisartan against Cyp-induced cardiotoxicity. MATERIALS AND METHODS Rats were divided into seven groups; (1): Control, (2): Cyp, (3-4): 0.05 Gy low dose rate (LDR) irradiation, 0.25 Gy high dose rate (HDR) irradiation, respectively, prior to Cyp dose, (5-7): telmisartan either alone or with 0.05 Gy LDR-irradiation or 0.25 Gy HDR-irradiation, respectively, prior to Cyp dose. The current investigation studied the effect of Cyp alone or combined with different treatment regimens on serum cTn-I and LDH, nuclear factor-κB (NF-κB) pathway (p65/IκB/IKK-α/IKK-ß) in the myocardium. Pro-inflammatory cytokines IL-1ß, IL-6 and TNF-α were assessed in addition to histopathological examination of the heart. RESULTS Low-dose irradiation attenuated cardiac enzymes, pro-inflammatory cytokines, NF-κB content, and histology, in both low and HDRs. Furthermore, the combination of low-dose irradiation with telmisartan (an angiotensin-II receptor type-1 blocker and a known cardio-protective drug) offered the best histological results. CONCLUSIONS Low-dose irradiation-induced amelioration is partially but not completely through canonical activation of NF-κB, and may have another atypical pathway. While telmisartan probably ameliorates NF-κB totally through canonical pathway.
Collapse
Affiliation(s)
- Amira Abd-ElRaouf
- National Centre of Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Ahmed S Nada
- National Centre of Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Nour El-Din A Mohammed
- National Centre of Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Hany A Amer
- National Centre for Nuclear Safety and Radiation Control, Atomic Energy Authority, Cairo, Egypt
| | - Sahar S Abd-ElRahman
- Department of Pathology, Faculty of Veterinary, Cairo University, Giza Square, Egypt
| | - Rania M Abdelsalam
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Hesham A Salem
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
26
|
Tesfaye BA, Berhe AH, Wondafrash DZ, Berhe DF. Cardioprotective Effect of Crude Extract and Solvent Fractions of Urtica simensis Leaves on Cyclophosphamide-Induced Myocardial Injury in Rats. J Exp Pharmacol 2021; 13:147-160. [PMID: 33628065 PMCID: PMC7897978 DOI: 10.2147/jep.s270038] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 02/03/2021] [Indexed: 01/26/2023] Open
Abstract
Background Globally, cardiovascular diseases (CVDs) are becoming the major cause of death. Urtica simensis is one of endogenous plant which treats a wide range of disease conditions including heart diseases. However, there is limited information on safety and efficacy of the plant. Objective To evaluate the in vitro antioxidant, the in vivo cardioprotective activity of crude extract and solvent fractions of Urtica simensis leaves on cyclophosphamide-induced myocardial injury. Methods The cardioprotective activity of the crude extract, aqueous and hexane fraction of Urtica simensis leaves was evaluated based on anatomical, biochemical and histopathological methods. The in vitro antioxidant activity of the plant was also assayed in terms of free radical scavenging activity (RSA). Results Crude extract and solvent fractions of Urtica simensis significantly prevented the deleterious effect of cyclophosphamide on body weight (P<0.001), heart weight to body ratio (P<0.01), cardiac biomarkers including troponin I (P<0.01), alanine transaminase (ALT) (P<0.001), aspartate aminotransferase (AST) (P<0.01) and lipid profiles including triglycerides (P<0.001) and total cholesterol (P<0.01). The histopathological study confirmed presence of necrosis, oedema and haemorrhage on cyclophosphamide alone-treated groups while the 200mg/kg and 400mg/kg of the crude extract and aqueous fraction showed normal cardiocytes. The antioxidant assay of Urtica simensis plant exhibited free radical scavenging activity of inhibitory concentration of 50% (IC50) for the crude extract with the values of 63.27µg/mL, aqueous fraction with the values of 136.38µg/mL and hexane fraction with the values of 258.70µg/mL. Conclusion Crude extract and solvent fractions of Urtica simensis leaves have cardioprotective activities. The cardioprotective effect could be attributed to the antioxidant activity of the plant extracts. However, this requires further in-depth understanding.
Collapse
Affiliation(s)
- Bekalu Amare Tesfaye
- Department of Pharmacology and Toxicology, School of Pharmacy, Mekelle University, Mekelle, Ethiopia
| | - Abera Hadgu Berhe
- Department of Pharmacology and Toxicology, School of Pharmacy, Mekelle University, Mekelle, Ethiopia
| | - Dawit Zewdu Wondafrash
- Department of Pharmacology and Toxicology, School of Pharmacy, Mekelle University, Mekelle, Ethiopia
| | - Derbew Fikadu Berhe
- Department of Pharmacology and Toxicology, School of Pharmacy, Mekelle University, Mekelle, Ethiopia
| |
Collapse
|
27
|
Kalinin RA, Suchkov IA, Klimentova ÉA, Shchul'kin AV, Egorov AA. [Effect of an antioxidant on vascular wall cell apoptosis markers after reconstructive operations]. ANGIOLOGIIA I SOSUDISTAIA KHIRURGIIA = ANGIOLOGY AND VASCULAR SURGERY 2021; 27:8-15. [PMID: 34528583 DOI: 10.33529/angio2021301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
AIM This study was aimed at determining Bcl-2 and Bax proteins expression before and after reconstructive-repairing operations in patients with atherosclerosis obliterans of lower extremities and at assessing the effect of an antioxidant (vitamin E at a dose of 100 mg once daily for 1 month after surgery) on the dynamics of changes of Bcl-2 and Bax proteins in the postoperative period. PATIENTS AND METHODS The study included a total of 60 patients with stage III-IV lower limb atherosclerosis obliterans. All patients underwent reconstructive-repairing operations on the arteries of the aortofemoral segment. After surgery the patients were divided into two groups. Group A included 30 patients who during 1 month received additionally to basic therapy vitamin E at a daily dose of 100 mg. Group B was composed of 30 patients receiving basic therapy alone according to the National guidelines of managing patients with peripheral artery disease. All patients before, on POD 1, and 1 month after surgery were subjected to venous blood test aimed at determining Bcl-2 and Bax apoptosis proteins expression by means of enzyme-linked immunosorbent assay. RESULTS In patients of groups A and B, the baseline level of Bcl-2 protein (4.75 and 4.2 ng/ml, respectively) was comparable with that in apparently healthy volunteers (5.3 ng/ml). The baseline levels of Bax protein in patients of the operated groups (26.9 and 26.0 ng/ml, respectively) were increased compared with the values in healthy volunteers (16.5 ng/ml). On POD 1 there was increased expression of Bax protein in Group A and B patients to 39.4 and 30.2 ng/ml, respectively. One month after surgery, Group B patients demonstrated a decrease in the Bcl-2 values below the baseline level - 1.1 ng/ml (p=0.003), with the Bax level continuing to increase - 36 ng/ml (p=0.004). In turn, Group A patients after 1 month were found to have increased levels of the Bcl-2 protein - 5.75 ng/ml, with the Bax level returning to the baseline values - 27.4 ng/ml. CONCLUSION In stage III and IV lower limb obliterating atherosclerosis, the level of the Bax proapoptoric protein was higher than that in healthy volunteers. On POD 1, there occurred increased expression of the pro-apoptotic protein Bax and activation of apoptosis markers. On the background of using vitamin E at a dose of 100 mg once daily for 1 month, there was a decrease in level of the Bax propapoptotic protein (p=0.003) and an increase in level of the anti-apoptotic Bcl-2 protein level (p=0.0007).
Collapse
Affiliation(s)
- R A Kalinin
- I.P. Pavlov Ryazan State Medical University of the RF Ministry of Public Health, Ryazan, Russia
| | - I A Suchkov
- I.P. Pavlov Ryazan State Medical University of the RF Ministry of Public Health, Ryazan, Russia
| | - É A Klimentova
- I.P. Pavlov Ryazan State Medical University of the RF Ministry of Public Health, Ryazan, Russia
| | - A V Shchul'kin
- I.P. Pavlov Ryazan State Medical University of the RF Ministry of Public Health, Ryazan, Russia
| | - A A Egorov
- I.P. Pavlov Ryazan State Medical University of the RF Ministry of Public Health, Ryazan, Russia
| |
Collapse
|
28
|
Hassan RA, Hozayen WG, Abo Sree HT, Al-Muzafar HM, Amin KA, Ahmed OM. Naringin and Hesperidin Counteract Diclofenac‐Induced Hepatotoxicity in Male Wistar Rats via Their Antioxidant, Anti‐Inflammatory, and Antiapoptotic Activities. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021. [DOI: https:/doi.org/10.1155/2021/9990091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 07/14/2021] [Indexed: 08/29/2023]
Abstract
This study is aimed at evaluating the preventive effect and at suggesting the mode of actions of naringin and hesperidin and their combination in diclofenac‐induced hepatotoxicity. Male Wistar rats, intraperitoneally injected with diclofenac sodium (3 mg/kg b.wt/day), were orally treated with naringin (20 mg/kg b.wt/day) and hesperidin (20 mg/kg b.wt/day) and their combination for 4 weeks. The administrations of naringin and hesperidin to diclofenac‐injected rats led to a significant decrease in the elevated serum ALT, AST, LDH, ALP, GGT, total bilirubin, TNF‐α, and IL‐17 levels as well as liver lipid peroxidation and liver p53 and caspase‐3 mRNA expressions. In contrast, serum IL‐4 level, liver GSH content, and liver GPx and SOD activities increased. In association, diclofenac‐induced deleterious histological alterations including hydropic degeneration, cytoplasmic vacuolization, apoptosis, and focal hepatic necrosis of hepatocytes associated with inflammatory cells’ infiltration were remarkably improved by treatments with naringin and hesperidin. In conclusion, naringin, hesperidin, and their combination, which was the most potent, counteract diclofenac‐induced liver injury via antioxidant, anti‐inflammatory, and antiapoptotic actions. Thus, this study recommends the use of naringin and hesperidin or their combination to resolve the side effects of drugs like diclofenac on the liver.
Collapse
|
29
|
Elrashidy RA, Hasan RA. Cilostazol preconditioning alleviates cyclophosphamide-induced cardiotoxicity in male rats: Mechanistic insights into SIRT1 signaling pathway. Life Sci 2020; 266:118822. [PMID: 33275987 DOI: 10.1016/j.lfs.2020.118822] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 11/18/2020] [Accepted: 11/20/2020] [Indexed: 01/08/2023]
Abstract
AIMS Cyclophosphamide (CYP) is a potent anticancer agent with well-known cardiotoxicity that limits its clinical applications. Cilostazol is a vosodilating drug, showing a cardioprotective effect in some cardiac disorders; however its effect in CYP-induced cardiotoxicity is still uncertain. We investigated the effect of cilostazol against CYP-induced cardiotoxicity and the contribution of SIRT1 signaling. MATERIALS AND METHODS 7 week-old male Wistar albino rats were treated with cilostazol (30 mg/kg/day, orally) in the absence or presence of SIRT1 inhibitor, EX-527 (5 mg/kg/day, IP) for 10 days and injected with CYP (200 mg/kg, IP) on the 7th day of the study. Age-matched rats were used as control group. On the 11th day, hearts were harvested for biochemical, immunoblotting and histological analyses. Markers of cardiac injury were assessed in plasma samples. KEY FINDINGS CYP injection contributed to cardiac injury manifested as significant increases in plasma activities of heart enzymes and cardiac troponin I levels. Cilostazol attenuated cardiac injury and minimized the histological lesions in hearts of CYP-treated rats. Cilostazol induced 3 fold up-regulation of SIRT1 and promoted the antioxidant defense response through FoxO1-related mechanism in hearts of CYP-treated rats. Cilostazol suppressed the CYP-induced up-regulation of PARP1 and p53, and blocked the NF-kB p65-mediated inflammatory response in hearts of CYP-treated rats. All the beneficial effects of cilostazol were almost abolished by EX-527. SIGNIFICANCE These data provided insights into the mechanism underlying the cardioprotective effect of cilostazol in CYP-treated rats through upregulation of SIRT1 signaling, suggesting that cilostazol might be a candidate modality for CYP-induced cardiotoxicity.
Collapse
Affiliation(s)
- Rania A Elrashidy
- Biochemistry Department, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt.
| | - Rehab A Hasan
- Histology Department, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
30
|
Mohamed MT, Zaitone SA, Ahmed A, Mehanna ET, El-Sayed NM. Raspberry Ketones Attenuate Cyclophosphamide-Induced Pulmonary Toxicity in Mice through Inhibition of Oxidative Stress and NF-ΚB Pathway. Antioxidants (Basel) 2020; 9:antiox9111168. [PMID: 33238601 PMCID: PMC7700149 DOI: 10.3390/antiox9111168] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/09/2020] [Accepted: 11/15/2020] [Indexed: 02/07/2023] Open
Abstract
Cyclophosphamide (CP) was found to have a potential toxic effect on lung tissues. Raspberry ketones (RKs) are natural antioxidant chemicals isolated from red raspberries (Rubus ideaus). They are commonly used for weight loss and obesity. The current study aimed to evaluate the possible protective effects of RKs against lung toxicity induced by CP. Mice were allocated into six groups: (1) control group; (2) CP group: received a single intraperitoneal dose of CP (150 mg/kg, i.p.); and (3–6) mice were pre-treated orally with different doses of RKs (25, 50, 100, and 200 mg/kg) for 14 consecutive days, respectively, before the administration of an intraperitoneal dose of CP (150 mg/kg, i.p.). Mice were then sacrificed under anesthesia, then lungs were removed for histopathological and biochemical investigations. A single dose of CP markedly altered the levels of some oxidative stress biomarkers and resulted in the fragmentation of DNA in lung homogenates. Histological examination of CP-treated mice demonstrated diffuse alveolar damage that involved apparent hyalinization of membranes, thickening of inter alveolar septa, and proliferation of type II pneumocytes. The immunohistochemical results of CP-treated mice revealed strongly positive Bax and weakly positive proliferating cell nuclear antigen (PCNA) staining reactivity of the nuclei of the lining epithelium of the bronchioles and alveoli. CP activated the cyclooxygenase-2/nuclear factor-kappa B pathway. However, pre-treatment with RKs significantly attenuated CP-evoked alterations in the previously mentioned parameters, highlighting their antioxidant, anti-inflammatory, and anti-apoptotic potential. RKs may be suggested to be a potential candidate to ameliorate CP-induced pulmonary toxicity.
Collapse
Affiliation(s)
| | - Sawsan A. Zaitone
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt;
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk 741, Saudi Arabia
| | - Amal Ahmed
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt;
| | - Eman T. Mehanna
- Department of Biochemistry, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt;
| | - Norhan M. El-Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt;
- Correspondence: ; Tel.: +20-12-2722-2915; Fax: +20-64-3230741
| |
Collapse
|
31
|
Santos DB, Colle D, Moreira ELG, Santos AA, Hort MA, Santos K, Oses JP, Razzera G, Farina M. Probucol Protects Neuronal Cells Against Peroxide-Induced Damage and Directly Activates Glutathione Peroxidase-1. Mol Neurobiol 2020; 57:3245-3257. [PMID: 32506382 DOI: 10.1007/s12035-020-01963-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 05/28/2020] [Indexed: 10/24/2022]
Abstract
Experimental evidence has shown that probucol, a hypocholesterolemic agent, is also able to increase glutathione peroxidase (GPx) activity. However, there is a lack of knowledge about the mechanism(s) involved in this event. In this study, in vitro experiments with purified GPx1 from bovine erythrocytes and cultured SH-SY5Y neuroblastoma cells, as well as in silico studies with GPx1, were performed in order to elucidate mechanisms mediating the stimulatory effect of probucol on GPx activity and to investigate the relevance of this event in terms of susceptibility against peroxide-induced cytotoxicity. In vitro experiments with purified GPx1 showed a direct stimulatory effect of probucol on the activity of GPx1, which was related to an increase in Vmax with no changes in KM. Probucol also increased GPx activity in cultured SH-SY5Y neuroblastoma cells, while the levels of GPx1 expression were not changed, corroborating the results found with the purified enzyme. In addition, probucol rendered SH-SY5Y cells more resistant to hydroperoxide-induced cytotoxicity, and this event was abolished in GPx1 knocked-down cells. In silico studies with GPx1 pointed to a potential binding site for probucol at the close vicinity of the GSH pocket. Collectively, the results presented herein indicate that GPx1 plays a central role in the probucol-induced protective effects against peroxide toxicity. This highlights a novel target (GPx1) and a new mechanism of action (direct activation) for an "old drug." The relevance of such results for in vivo conditions deserves further investigation.
Collapse
Affiliation(s)
- Danúbia B Santos
- Department of Biochemistry, Federal University of Santa Catarina, Florianopolis, Santa Catarina, 88040-900, Brazil.
| | - Dirleise Colle
- Department of Clinical Analyses, Federal University of Santa Catarina, Florianopolis, Santa Catarina, Brazil
| | - Eduardo L G Moreira
- Department of Physiological Sciences, Federal University of Santa Catarina, Florianopolis, Santa Catarina, Brazil
| | - Alessandra A Santos
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Mariana A Hort
- Institute of Biological Sciences, Federal University of Rio Grande, Rio Grande, Rio Grande do Sul, Brazil
| | - Karin Santos
- Department of Biochemistry, Federal University of Santa Catarina, Florianopolis, Santa Catarina, 88040-900, Brazil
| | - Jean P Oses
- Institute of Bioscience, Sector of Biochemistry, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Guilherme Razzera
- Department of Biochemistry, Federal University of Santa Catarina, Florianopolis, Santa Catarina, 88040-900, Brazil
| | - Marcelo Farina
- Department of Biochemistry, Federal University of Santa Catarina, Florianopolis, Santa Catarina, 88040-900, Brazil.
| |
Collapse
|
32
|
El kiki SM, Omran MM, Mansour HH, Hasan HF. Metformin and/or low dose radiation reduces cardiotoxicity and apoptosis induced by cyclophosphamide through SIRT-1/SOD and BAX/Bcl-2 pathways in rats. Mol Biol Rep 2020; 47:5115-5126. [DOI: 10.1007/s11033-020-05582-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 06/10/2020] [Indexed: 12/27/2022]
|
33
|
Alhoshani A, Alanazi FE, Alotaibi MR, Attwa MW, Kadi AA, Aldhfyan A, Akhtar S, Hourani S, Agouni A, Zeidan A, Korashy HM. EGFR Inhibitor Gefitinib Induces Cardiotoxicity through the Modulation of Cardiac PTEN/Akt/FoxO3a Pathway and Reactive Metabolites Formation: In Vivo and in Vitro Rat Studies. Chem Res Toxicol 2020; 33:1719-1728. [PMID: 32370496 DOI: 10.1021/acs.chemrestox.0c00005] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Gefitinib (GEF) is a selective inhibitor of the epidermal growth factor receptor (EGFR) used to treat non-small cell lung cancer. Yet, few cases of cardiotoxicity have been reported. However, the role of the PTEN/Akt/FoxO3a pathway, which mediates GEF anticancer activity, in GEF cardiotoxicity remains unclear. For this purpose, in vitro H9c2 cells and in vivo rat cardiomyocytes were utilized as study models. Treatment of H9c2 cells and Sprague-Dawley rats with GEF significantly induced the expression of hypertrophic and apoptotic markers at mRNA and protein levels with an increased plasma level of troponin. This was accompanied by induction of autophagy and mitochondrial dysfunction in H9c2 cells. Inhibition of cardiac EGFR activity and Akt cellular content of in vitro and in vivo rat cardiomyocytes by GEF increased PTEN and FoxO3a gene expression and cellular content. Importantly, treatment of H9c2 cells with PI3K/Akt inhibitor increased PTEN and FoxO3a mRNA expression associated with potentiation of GEF cardiotoxicity. In addition, by using LC-MS/MS, we showed that GEF is metabolized in the rat heart microsomes into one cyanide- and two methoxylamine-adduct reactive metabolites, where their formation was entirely blocked by CYP1A1 inhibitor, α-naphthoflavone. The current study concludes that GEF induces cardiotoxicity through modulating the expression and function of the cardiac PTEN/AKT/FoxO3a pathway and the formation of CYP1A1-mediated reactive metabolites.
Collapse
Affiliation(s)
- Ali Alhoshani
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Fawaz E Alanazi
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia.,Security Forces Hospital Program, P.O. Box 3643, Riyadh 11481, Saudi Arabia
| | - Moureq R Alotaibi
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Mohamed W Attwa
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia.,Students' University Hospital, Mansoura University, Mansoura 35516, Egypt
| | - Adnan A Kadi
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Abdullah Aldhfyan
- Stem Cell & Tissue Re-Engineering, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia
| | - Sabah Akhtar
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Shireen Hourani
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Abdelali Agouni
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Asad Zeidan
- College of Medicine, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Hesham M Korashy
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| |
Collapse
|
34
|
The Role of Antioxidants in Ameliorating Cyclophosphamide-Induced Cardiotoxicity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:4965171. [PMID: 32454939 PMCID: PMC7238386 DOI: 10.1155/2020/4965171] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 04/09/2020] [Accepted: 04/21/2020] [Indexed: 12/11/2022]
Abstract
The chemotherapeutic and immunosuppressive agent cyclophosphamide has previously been shown to induce complications within the setting of bone marrow transplantation. More recently, cardiotoxicity has been shown to be a dose-limiting factor during cyclophosphamide therapy, and cardiooncology is getting wider attention. Though mechanism of cyclophosphamide-induced cardiotoxicity is not completely understood, it is thought to encompass oxidative and nitrative stress. As such, this review focuses on antioxidants and their role in preventing or ameliorating cyclophosphamide-induced cardiotoxicity. It will give special emphasis to the cardioprotective effects of natural, plant-derived antioxidants that have garnered significant interest in recent times.
Collapse
|
35
|
Singh S, Kumar A. Protective Effect of Edaravone on Cyclophosphamide Induced Oxidative Stress and Neurotoxicity in Rats. Curr Drug Saf 2020; 14:209-216. [PMID: 31057112 PMCID: PMC6864589 DOI: 10.2174/1574886314666190506100717] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 04/11/2019] [Accepted: 04/24/2019] [Indexed: 01/06/2023]
Abstract
BACKGROUND Cyclophosphamide (CPA) is the most widely prescribed cancer chemotherapeutic agent which shows serious neurotoxic side effect. Generation of reactive oxygen species at the cellular level is the basic mechanism of cyclophosphamide induced neurotoxicity. Edaravone is the synthetic drug used for brain stroke and has potent antioxidant property. OBJECTIVE This study aimed to investigate the effect of edaravone on neurobehavioral and neuropathological alteration induced by cyclophosphamide in male rats. METHODS Twenty eight Sprague-Dawley rats were equally divided into four groups of seven rats in each. The control group received saline, and other groups were given CPA intraperitoneally (100 mg/kg), CPA (100 mg/kg) intraperitoneally + Edaravone (10 mg/kg) orally, or Edaravone (10 mg/kg) orally for one month. RESULTS Our data showed that CPA significantly elevated brain AChE activity in the hippocampal region. A decrease in the total antioxidant capacity and a reduction in the CAT, SOD, and GPX activity occurred in the brains of the rats exposed to CPA. CPA-treated rats showed a significant impairment in long-termmemory and motor coordination. These results were supported by histopathological observations of the brain. Results revealed that administration of edaravone reversed AChE activity alternation and ameliorated behavioral and histopathological changes induced by CPA. CONCLUSION This study suggests that co-administration of edaravone with cyclophosphamide may be a useful intriguing therapeutic approach to overcome cyclophosphamide induced neurotoxicity.
Collapse
Affiliation(s)
- Sanjiv Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Bihar, India.,Truba Institute of Pharmacy, Bhopal, MP, India
| | | |
Collapse
|
36
|
Rohilla S, Dureja H, Chawla V. Cytoprotective Agents to Avoid Chemotherapy Induced Sideeffects on Normal Cells: A Review. Curr Cancer Drug Targets 2019; 19:765-781. [PMID: 30914026 DOI: 10.2174/1568009619666190326120457] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 03/01/2019] [Accepted: 03/10/2019] [Indexed: 01/16/2023]
Abstract
Anticancer agents play a vital role in the cure of patients suffering from malignancy. Though, the chemotherapeutic agents are associated with various adverse effects which produce significant toxic symptoms in the patients. But this therapy affects both the malignant and normal cells and leads to constricted therapeutic index of antimalignant drugs which adversely impacts the quality of patients’ life. Due to these adversities, sufficient dose of drug is not delivered to patients leading to delay in treatment or improper treatment. Chemoprotective agents have been developed either to minimize or to mitigate the toxicity allied with chemotherapeutic agents. Without any concession in the therapeutic efficacy of anticancer drugs, they provide organ specific guard to normal tissues.
Collapse
Affiliation(s)
- Seema Rohilla
- Department of Pharmaceutics, Hindu College of Pharmacy, Sonepat- 131001, India
| | - Harish Dureja
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak-124001, India
| | - Vinay Chawla
- Institute of Pharmaceutical Sciences, Baba Farid University of Health Sciences, Faridkot-151203, India
| |
Collapse
|
37
|
El-Sheikh AA, Abdelzaher WY, Gad AA, Abdel-Gaber SA. Purine versus non-purine xanthine oxidase inhibitors against cyclophosphamide-induced cardiac and bone marrow toxicity in rats. Hum Exp Toxicol 2019; 39:249-261. [PMID: 31640406 DOI: 10.1177/0960327119883412] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND AIM Cancer is a fatal and serious disease. Cyclophosphamide (CYC) is a commonly used anticancer drug. Cardiotoxicity and myelotoxicity are life-threatening side effects of CYC treatment. We aimed to evaluate the effect of the xanthine oxidase (XO) inhibitors, allopurinol (ALL) and febuxostat (FEB), on CYC-induced cardio- and hematopoietic toxicity in rats. METHODS ALL (100 mg/kg/day) or FEB (10 mg/kg/day) were administered orally to rats in the presence and absence of CYC (200 mg/kg kg i.p. single dose) treatment. Serum creatine kinase-MB creatine kinase myocardial band (CK-MB) and lactate dehydrogenase (LDH) activities were estimated. Complete blood counting (CBC), cardiac and bone marrow XO activity, malondialdehyde level, and superoxide dismutase activity were determined. Cardiac and bone marrow histopathological changes were also evaluated. RESULTS ALL and FEB significantly decreased CK-MB and LDH induced by CYC. Disturbed levels of XO, oxidative stress parameters, and CBC were also corrected by both XO inhibitors tested, with amelioration of cardiac histopathological changes caused by CYC. Treatment with FEB, but not ALL, prior to CYC challenges normalized bone marrow histopathological changes. CONCLUSION These results suggest that both XO inhibitors tested; ALL and FEB can ameliorate CYC-induced cardiotoxicity. However, only FEB can protect against CYC-induced myelotoxicity, whereas ALL, to the contrary, might aggravate it.
Collapse
Affiliation(s)
- A A El-Sheikh
- Basic Health Sciences Department, College of Medicine, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia.,Pharmacology Department, Faculty of Medicine, Minia University, Minia, Egypt
| | - W Y Abdelzaher
- Pharmacology Department, Faculty of Medicine, Minia University, Minia, Egypt
| | - A A Gad
- Pharmacology Department, Faculty of Medicine, Minia University, Minia, Egypt
| | - S A Abdel-Gaber
- Pharmacology Department, Faculty of Medicine, Minia University, Minia, Egypt
| |
Collapse
|
38
|
Iqubal A, Sharma S, Ansari MA, Najmi AK, Syed MA, Ali J, Alam MM, Ahmad S, Haque SE. Nerolidol attenuates cyclophosphamide-induced cardiac inflammation, apoptosis and fibrosis in Swiss Albino mice. Eur J Pharmacol 2019; 863:172666. [PMID: 31541628 DOI: 10.1016/j.ejphar.2019.172666] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 09/06/2019] [Accepted: 09/17/2019] [Indexed: 02/07/2023]
Abstract
Incidence and prevalence of cancer is an alarming situation globally. For the treatment of cancer many anticancer drugs have been developed but, unfortunately, their potential cardiotoxic side effects raised serious concerns about their use among clinicians. Cyclophosphamide is a potent anticancer and immunosuppressant drug but its use is limited due to cardiotoxic side effect. Thus, there is a need for the development of certain drug which can reduce cardiotoxicity and can be used as an adjuvant therapy in cancer patients. In this direction we, therefore planned to evaluate nerolidol (NER) for its cardioprotective potential against cyclophosphamide-induced cardiotoxicity in Swiss Albino mice. Animals were divided into 6 groups. Vehicle control; Cyclophosphamide (CP 200); NER 400 per se; NER 200 + CP 200; NER 400 + CP 200; and fenofibrate (FF 80) + CP 200. Dosing was done for 14 days along with a single dose of CP 200 on the 7th day. On 15th day animals were sacrificed and various biochemical parameters pertaining to oxidative stress, nitrative stress, inflammation, apoptosis and fibrosis were estimated in the blood and heart tissues. Histopathological analysis (H & E and Masson's trichrome staining); ultrastructural analysis (transmission electron microscopy) and immunohistochemical analysis were also performed along with mRNA expression and molecular docking to establish the cardioprotective potential of nerolidol. Nerolidol acted as a potent cardioprotective molecule and attenuated CP-induced cardiotoxicity.
Collapse
Affiliation(s)
- Ashif Iqubal
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Sumit Sharma
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Mohd Asif Ansari
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Abul Kalam Najmi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Mansoor Ali Syed
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, 110025, India
| | - Javed Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - M Mumtaz Alam
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Shaniya Ahmad
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, 110025, India
| | - Syed Ehtaishamul Haque
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
39
|
Salimi A, Pirhadi R, Jamali Z, Ramazani M, Yousefsani BS, Pourahmad J. Mitochondrial and lysosomal protective agents ameliorate cytotoxicity and oxidative stress induced by cyclophosphamide and methotrexate in human blood lymphocytes. Hum Exp Toxicol 2019; 38:1266-1274. [PMID: 31446784 DOI: 10.1177/0960327119871096] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Cyclophosphamide (CYP) and methotrexate (MTX) have been evaluated for their ability to induce toxicity in human peripheral blood lymphocytes (PBLs) and the protective role of mitochondrial and lysosomal stabilizing agents. The potential toxicity effects of CYP and MTX were measured in vitro by cellular parameters assays such as cellular viability, reactive oxygen species (ROS) formation, mitochondrial membrane permeability transition (mitochondrial membrane potential (MMP)) collapse, lysosomal membrane damage, intracellular reduced glutathione (GSH), extracellular oxidized glutathione (GSSG), and lipid peroxidation. Separately, human lymphocytes were treated with concentrations of 0.1, 0.2, 0.4, 0.8, and 1.6 ng/mL for CYP and 1, 2, 5, and 10 µg/mL for MTX for 6 h. Statistical evaluations showed that CYP and MTX significantly decreased the cell viability at the three highest concentrations when compared with both the negative and solvent controls. In addition, CYP and MTX were significantly induced ROS formation, MMP collapse, lysosomal membrane damage, lipid peroxidation, and GSH depletion compared with the controls. Mitochondrial and lysosomal protective agents like cyclosporine A and chloroquine, respectively, decreased cytotoxicity and oxidative stress induced by CYP and MTX. The present results indicate that CYP and MTX are toxic to human PBLs and their toxicity could be ameliorated by mitochondrial and lysosomal protective agents.
Collapse
Affiliation(s)
- A Salimi
- Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - R Pirhadi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Z Jamali
- Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - M Ramazani
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - B S Yousefsani
- Research Institute for Islamic and Complementary Medicine, Iran University of Medical Sciences, Tehran, Iran.,School of Persian Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - J Pourahmad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
40
|
Diethelm-Varela B, Ai Y, Liang D, Xue F. Nitrogen Mustards as Anticancer Chemotherapies: Historic Perspective, Current Developments and Future Trends. Curr Top Med Chem 2019; 19:691-712. [PMID: 30931858 DOI: 10.2174/1568026619666190401100519] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 02/11/2019] [Accepted: 02/25/2019] [Indexed: 12/30/2022]
Abstract
Nitrogen mustards, a family of DNA alkylating agents, marked the start of cancer pharmacotherapy. While traditionally characterized by their dose-limiting toxic effects, nitrogen mustards have been the subject of intense research efforts, which have led to safer and more effective agents. Even though the alkylating prodrug mustards were first developed decades ago, active research on ways to improve their selectivity and cytotoxic efficacy is a currently active topic of research. This review addresses the historical development of the nitrogen mustards, outlining their mechanism of action, and discussing the improvements on their therapeutic profile made through rational structure modifications. A special emphasis is made on discussing the nitrogen mustard prodrug category, with Cyclophosphamide (CPA) serving as the main highlight. Selected insights on the latest developments on nitrogen mustards are then provided, limiting such information to agents that preserve the original nitrogen mustard mechanism as their primary mode of action. Additionally, future trends that might follow in the quest to optimize these invaluable chemotherapeutic medications are succinctly suggested.
Collapse
Affiliation(s)
- Benjamin Diethelm-Varela
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| | - Yong Ai
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| | - Dongdong Liang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| | - Fengtian Xue
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| |
Collapse
|
41
|
Refaie MMM, Shehata S, El-Hussieny M, Abdelraheem WM, Bayoumi AMA. Role of ATP-Sensitive Potassium Channel (KATP) and eNOS in Mediating the Protective Effect of Nicorandil in Cyclophosphamide-Induced Cardiotoxicity. Cardiovasc Toxicol 2019; 20:71-81. [DOI: 10.1007/s12012-019-09535-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
42
|
Abdallah HMI, Abdel-Rahman RF, El Awdan SA, Allam RM, El-Mosallamy AEMK, Selim MS, Mohamed SS, Arbid MS, Farrag ARH. Protective effect of some natural products against chemotherapy-induced toxicity in rats. Heliyon 2019; 5:e01590. [PMID: 31080906 PMCID: PMC6507045 DOI: 10.1016/j.heliyon.2019.e01590] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 03/01/2019] [Accepted: 04/24/2019] [Indexed: 02/07/2023] Open
Abstract
Aim There is a great interest in combining anticancer drugs with natural products aiming at maximizing their efficacy while minimizing systemic toxicity. Hence, the present study was constructed aiming to investigate the protective potential of three natural products, 1,8-cineole an essential oil from Artemisia herba alba, exopolysaccharide (EPS) from locally identified marine streptomycete, and ellagic acid (EA), against chemotherapy-induced organ toxicity. Methods Isolation, production and characterization of EPS from marine streptomycete was done. Animals were allocated into five groups, GP1: normal control, GP2: cyclophosphamide (CYC), GP3: 1,8-cineole + CYC, GP4: EPS + CYC, GP4: EA + CYC. All drugs were administered orally 1 week before and concomitantly with CYC. Electrocardiography (ECG) analysis, liver enzymes (ALT and AST), cardiac serum markers (LDH and CK), oxidative stress biomarkers in hepatic and cardiac tissues (GSH and MDA), TGF-β1 and histopathological examination of hepatic and cardiac tissues were executed. Results The isolated stain produced EPS was identified as Streptomyces xiamenensis. EPS contains uronic, sulphate groups and different monosugars with Mw 4.65 × 104 g/mol and showed antioxidant activity against DPPH. Pretreatment of rats with 1,8-cineole, EPS and EA improved ECG abnormalities, decrease serum markers of hepato- and cardiotoxicity, prevent oxidative stress and decrease TGF-β1 in liver and heart tissues. Conclusion The present results demonstrate the hepatoprotective and cardioprotective effects of the above-mentioned natural products against CYC organ toxicity.
Collapse
Affiliation(s)
- Heba M I Abdallah
- Department of Pharmacology, Medical Division, National Research Centre, Giza, Egypt
| | - Rehab F Abdel-Rahman
- Department of Pharmacology, Medical Division, National Research Centre, Giza, Egypt
| | - Sally A El Awdan
- Department of Pharmacology, Medical Division, National Research Centre, Giza, Egypt
| | - Rasha M Allam
- Department of Pharmacology, Medical Division, National Research Centre, Giza, Egypt
| | | | - Manal S Selim
- Department of Microbial Biotechnology, Genetic Engineering and Biotechnology Research Division, National Research Centre, Giza, Egypt
| | - Sahar S Mohamed
- Department of Microbial Biotechnology, Genetic Engineering and Biotechnology Research Division, National Research Centre, Giza, Egypt
| | - Mahmoud S Arbid
- Department of Pharmacology, Medical Division, National Research Centre, Giza, Egypt
| | - Abdel Razik H Farrag
- Department of Pathology, Medical Division, National Research Centre, Giza, Egypt
| |
Collapse
|
43
|
Omole JG, Ayoka OA, Alabi QK, Adefisayo MA, Asafa MA, Olubunmi BO, Fadeyi BA. Protective Effect of Kolaviron on Cyclophosphamide-Induced Cardiac Toxicity in Rats. J Evid Based Integr Med 2019; 23:2156587218757649. [PMID: 29468886 PMCID: PMC5871040 DOI: 10.1177/2156587218757649] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Cyclophosphamide (CP) is a nitrogen mustard alkylating drug used for the treatment of chronic and acute malignant lymphomas, myeloma, leukemia, neuroblastoma, adenocarcinoma, retinoblastoma, breast carcinoma, and immunosuppressive therapy. Despite its vast therapeutic uses, it is known to cause severe cardiac toxicity. Kolaviron (KV), a Garcinia kola seed extract containing a mixture of flavonoids, is reputed for its antioxidant and membrane stabilizing properties. OBJECTIVE This study investigated the protective effect of KV on CP-induced cardiotoxicity in rats. METHODS Thirty rats were used, and they were divided into 6 groups of 5 rats each. Group I received 2 mL/kg propylene glycol orally for 14 days; group II received CP (50 mg/kg/d, intraperitoneally [i.p.]) for 3 days; groups III and IV received 200 and 400 mg/kg/d KV, respectively, orally for 14 days and groups V and VI were pretreated with 200 and 400 mg/kg/d KV, respectively, orally for 14 days followed by CP (50 mg/kg/d, i.p.) for 3 days. RESULTS CP treatment resulted in a significantly lower food consumption and body weight in rats. The lactate dehydrogenase and creatine kinase enzymes in cardiac tissues of rats treated with CP were significantly higher. In cardiac tissues, 3-day doses of CP resulted in significantly higher heart weight, cardiac troponin I, myeloperoxidase, malondialdehyde, hydrogen peroxide and lower superoxide dismutase, catalase, glutathione peroxidase activities, and reduced glutathione levels. Histological examination of cardiac tissues showed sign of necrosis of myocardium after CP treatment. However, administration of KV at 200 and 400 mg/kg for 14 days prior to CP treatment, increase food consumption, body weight, and attenuates the biochemical and histological changes induced by CP. CONCLUSIONS These results revealed that KV attenuates CP-induced cardiotoxicity by inhibiting oxidative stress and preserving the activity of antioxidant enzymes.
Collapse
Affiliation(s)
| | | | - Quadri Kunle Alabi
- 1 Obafemi Awolowo University, Ile-Ife, Osun State, Nigeria.,2 Afe-Babalola University, Ado-Ekiti, Ekiti State, Nigeria
| | - Modinat Adebukola Adefisayo
- 1 Obafemi Awolowo University, Ile-Ife, Osun State, Nigeria.,3 University of Medical Sciences, Ondo City, Ondo State, Nigeria
| | | | | | - Benson Akinloye Fadeyi
- 1 Obafemi Awolowo University, Ile-Ife, Osun State, Nigeria.,4 Federal Teaching Hospital, Ido-Ekiti, Ekiti State, Nigeria
| |
Collapse
|
44
|
Eltantawy FM, Sobh MAA, EL-Waseef AM, Ibrahim RAA, Saad MA. Protective effect of Spirulina against cyclophosphamide-induced urotoxicity in mice. ACTA ACUST UNITED AC 2019. [DOI: 10.1016/j.ejbas.2018.06.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Affiliation(s)
- Fatma M. Eltantawy
- Urology and Nephrology Center, Mansoura University, Mansoura 35511, Egypt
| | - Mohamed A. Ali Sobh
- Urology and Nephrology Center, Mansoura University, Mansoura 35511, Egypt
- Medical and Experimental Research Center, Faculty of Medicine, Mansoura University, Mansoura 35511, Egypt
| | - Ahmed M. EL-Waseef
- Department of Chemistry/Biochemistry, Faculty of Science, Mansoura University, Mansoura 35511, Egypt
| | - Rehab-Allah A. Ibrahim
- Department of Pathology, Faculty of Medicine, Mansoura University, Mansoura 35511, Egypt
| | - Mohamed A.A. Saad
- Medical and Experimental Research Center, Faculty of Medicine, Mansoura University, Mansoura 35511, Egypt
- Department of Pharmacology, Faculty of Medicine, Mansoura University, Mansoura 35511, Egypt
| |
Collapse
|
45
|
Ma X, Jiao Z, Liu Y, Chen J, Li G, Liu T, Tse G, Yuan R. Probucol Protects Against Contrast-Induced Acute Kidney Injury via the Extracellular Signal-Regulated Kinases 1 and 2 (ERK1/2)/JNK-Caspase 3 Pathway in Diabetic Rats. Med Sci Monit 2019; 25:1038-1045. [PMID: 30728344 PMCID: PMC6375284 DOI: 10.12659/msm.913106] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 11/01/2018] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Contrast-induced acute kidney injury is an important clinical problem, yet its pathogenic mechanisms are incompletely understood. In this study we explored the potential beneficial effects of probucol as treatment of contrast-induced acute kidney injury in diabetic rats. MATERIAL AND METHODS Rats were divided into 3 groups: i) diabetic control, ii) diabetic with contrast, and iii) probucol treatment groups. Probucol was administered by gavage and the contrast diatrizoate (60%) was injected via femoral vein. After 24 h, the rats were sacrificed and samples were taken to measure biochemical indicators. Pathological damage of renal tubules was evaluated by HE staining. Expression of Bcl-2, Bax, p-ERKs, and p-JNK proteins in the kidneys was examined by Western blotting, whereas expression level of caspase-3 in kidneys was detected by immunohistochemistry. RESULTS Compared to the probucol treatment group, the diabetes with contrast group showed higher serum creatinine and lower creatinine clearance. The pathological changes of kidneys in the probucol treatment group were improved compared with the contrast group. Moreover, Western blot analyses revealed that use of contrast agent led to lower p-ERK1/2, higher p-JNK, lower Bcl-2, and higher Bax levels, which were reversed by probucol. Finally, immunohistochemical findings revealed higher caspase-3 after contrast use, which was partially reversed by probucol. CONCLUSIONS Probucol exerts protective effects on contrast-induced acute kidney injury in diabetic rats by inhibition of renal cell apoptosis. This is achieved by reducing mitochondrial caspase-3 expression through increasing and decreasing the expression of the upstream mediators p-ERK1/2 and p-JNK, respectively.
Collapse
Affiliation(s)
- Xingxing Ma
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, P.R China
| | - Zhanquan Jiao
- Department of Cardiology, Tianjin Haihe Hospital, Tianjin, P.R. China
| | - Yanhong Liu
- Department of Cardiology, Tianjin Third Central Hospital, Tianjin, P.R. China
| | - Jun Chen
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, P.R China
| | - Guangping Li
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, P.R China
| | - Tong Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, P.R China
| | - Gary Tse
- Department of Medicine and Therapeutics, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, P.R. China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, P.R. China
| | - Ruyu Yuan
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, P.R China
| |
Collapse
|
46
|
Novel Mesenchymal Stem Cell Strategy in Alleviating Toll-Like Receptor-4, p53 and Nrf2 Signaling in Isoproterenol-Induced Myocardial Infarction in Rat Model. Cardiovasc Toxicol 2019; 18:232-241. [PMID: 29110132 DOI: 10.1007/s12012-017-9432-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Mesenchymal stem cells (MSCs) are multipotent stromal cells that merit the differentiation into various cell types. The present study was designed to test the hypothesis that the cardioprotective effect of MSCs transplantation and digoxin treatment is mediated via the regulation of messenger RNA gene expression of pro-inflammatory cytokines and apoptotic markers. Myocardial infarction was induced in Wistar rats via isoproterenol injection in a dose of (85 mg/kg, subcutaneously, twice at an interval of 24 h). Four weeks post-MSCs transplantation and digoxin treatment a significant reduction in serum cardiac markers, aspartate aminotransferase, creatine kinase-MB and troponine II was observed. Meanwhile, isoproterenol significantly reduced the gene and protein expression of the oxidative stress marker nuclear-related factor-2 (Nrf2) with a concomitant elevation in (MDA) level and inflammatory markers toll-like receptor-4 (TLR-4), intercellular adhesion molecules (ICAMs) and (VCAM-1). Moreover, apoptotic marker (P53) was significantly down-regulated. This was confirmed by histopathological investigations. It was hypothesized that MSCs transplantation was superior over digoxin treatment regimen in improving heart function.
Collapse
|
47
|
Iqubal A, Iqubal MK, Sharma S, Ansari MA, Najmi AK, Ali SM, Ali J, Haque SE. Molecular mechanism involved in cyclophosphamide-induced cardiotoxicity: Old drug with a new vision. Life Sci 2018; 218:112-131. [PMID: 30552952 DOI: 10.1016/j.lfs.2018.12.018] [Citation(s) in RCA: 185] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 11/08/2018] [Accepted: 12/10/2018] [Indexed: 12/20/2022]
Abstract
Cyclophosphamide (CP) is an important anticancer drug which belongs to the class of alkylating agent. Cyclophosphamide is mostly used in bone marrow transplantation, rheumatoid arthritis, lupus erythematosus, multiple sclerosis, neuroblastoma and other types of cancer. Dose-related cardiotoxicity is a limiting factor for its use. CP-induced cardiotoxicity ranges from 7 to 28% and mortality ranges from 11 to 43% at the therapeutic dose of 170-180 mg/kg, i.v. CP undergoes hepatic metabolism that results in the production of aldophosphamide. Aldophosphamide decomposes into phosphoramide mustard & acrolein. Phosphoramide is an active neoplastic agent, and acrolein is a toxic metabolite which acts on the myocardium and endothelial cells. This is the first review article that talks about cyclophosphamide-induced cardiotoxicity and the different signaling pathways involved in its pathogenicity. Based on the available literature, CP is accountable for cardiomyocytes energy pool alteration by affecting the heart fatty acid binding proteins (H-FABP). CP has been found associated with cardiomyocytes apoptosis, inflammation, endothelial dysfunction, calcium dysregulation, endoplasmic reticulum damage, and mitochondrial damage. Molecular mechanism of cardiotoxicity has been discussed in detail through crosstalk of Nrf2/ARE, Akt/GSK-3β/NFAT/calcineurin, p53/p38MAPK, NF-kB/TLR-4, and Phospholamban/SERCA-2a signaling pathway. Based on the available literature we support the fact that metabolites of CP are responsible for cardiotoxicity due to depletion of antioxidants/ATP level, altered contractility, damaged endothelium and enhanced pro-inflammatory/pro-apoptotic activities resulting into cardiomyopathy, myocardial infarction, and heart failure. Dose adjustment, elimination/excretion of acrolein and maintenance of endogenous antioxidant pool could be the therapeutic approach to mitigate the toxicities.
Collapse
Affiliation(s)
- Ashif Iqubal
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Mohammad Kashif Iqubal
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Sumit Sharma
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Mohd Asif Ansari
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Abul Kalam Najmi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Syed Mansoor Ali
- Department of Biosciences, Jamia Millia Islamia,110025 New Delhi, India
| | - Javed Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Syed Ehtaishamul Haque
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
48
|
Zhao K, Gao Q, Zong C, Ge L, Liu J. Cordyceps sinensis prevents contrast-induced nephropathy in diabetic rats: its underlying mechanism. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:5571-5580. [PMID: 31949644 PMCID: PMC6963074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 10/25/2018] [Indexed: 06/10/2023]
Abstract
Apoptosis is recognized as an important mechanism in contrast-induced nephropathy (CIN). This study investigated the renal protective effect of cordyceps sinensis (CS) in a diabetic rat model of CIN and the mechanism of its effect. Sixty SD rats were randomly divided into 4 groups, the control group, model group, probucol group, and CS group. We used a diabetic rat model of Iodixanol-induced CIN. Serum creatinine (Scr), blood urea nitrogen (BUN), urinary kidney injury molecule-1 (KIM-1), neutrophil gelatinase associated lipocalin (NGAL) levels were measured to evaluate renal function. Total antioxidative ability (T-AOC), superoxide dismutase (SOD), and malonaldehyde (MDA) levels were assessed to discuss the effect of probucol and CS on oxidative stress. The pathologic changes in the kidney were observed by hematoxylin and eosin (HE) staining and periodic acid-Schiff (PAS) staining. Apoptosis was assessed by transmission electron microscopy and TUNEL staining. Caspase-3, Bax, Bcl2 and phospho-p38 mitogen-activated protein kinase (MAPK) protein expressions were assessed by Western blotting. The model group of rats showed significantly elevated levels of BUN, Scr, urinary KIM-1, NGAL, and parameters of oxidative stress (P<0.05). Both the probucol and CS groups demonstrated significantly lower Scr, BUN, and urinary KIM-1, NGAL levels compared to the model group (P<0.05), with no significant difference between these two groups. The probucol group and the CS group had significantly lower MDA and higher T-AOC, SOD than the model group after modeling (P<0.05). Caspase-3, Bax activation were effectively repressed while Bcl-2 expression was increased by probucol and CS pretreatment. Mechanistically, probucol and CS decreased the expression of JNK protein and increased the expression of ERK protein. CS can effectively reduce kidney damage caused by contrast medium. The underlying mechanism may be that CS accelerates the recovery of renal function and renal pathology by reducing local renal oxidative stress and influencing MAPK signal pathways.
Collapse
Affiliation(s)
- Kai Zhao
- Department of Cardiology, Tianjin Nankai HospitalTianjin, China
| | - Qiaoying Gao
- Department of Pharmacology, Institute of Acute Abdominal Diseases, Tianjin Nankai HospitalTianjin, China
| | - Chunhui Zong
- Department of Pharmacology, Institute of Acute Abdominal Diseases, Tianjin Nankai HospitalTianjin, China
| | - Lixiu Ge
- Department of Clinical Laboratory, Tianjin Nankai HospitalTianjin, China
| | - Jinjin Liu
- Department of Pharmacology, Institute of Acute Abdominal Diseases, Tianjin Nankai HospitalTianjin, China
| |
Collapse
|
49
|
Di Paolo C, Müller Y, Thalmann B, Hollert H, Seiler TB. p53 induction and cell viability modulation by genotoxic individual chemicals and mixtures. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2018; 25:4012-4022. [PMID: 28303539 DOI: 10.1007/s11356-017-8790-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 03/08/2017] [Indexed: 06/06/2023]
Abstract
The binding of the p53 tumor suppression protein to DNA response elements after genotoxic stress can be quantified by cell-based reporter gene assays as a DNA damage endpoint. Currently, bioassay evaluation of environmental samples requires further knowledge on p53 induction by chemical mixtures and on cytotoxicity interference with p53 induction analysis for proper interpretation of results. We investigated the effects of genotoxic pharmaceuticals (actinomycin D, cyclophosphamide) and nitroaromatic compounds (4-nitroquinoline 1-oxide, 3-nitrobenzanthrone) on p53 induction and cell viability using a reporter gene and a colorimetric assay, respectively. Individual exposures were conducted in the absence or presence of metabolic activation system, while binary and tertiary mixtures were tested in its absence only. Cell viability reduction tended to present direct correlation with p53 induction, and induction peaks occurred mainly at chemical concentrations causing cell viability below 80%. Mixtures presented in general good agreement between predicted and measured p53 induction factors at lower concentrations, while higher chemical concentrations gave lower values than expected. Cytotoxicity evaluation supported the selection of concentration ranges for the p53 assay and the interpretation of its results. The often used 80% viability threshold as a basis to select the maximum test concentration for cell-based assays was not adequate for p53 induction assessment. Instead, concentrations causing up to 50% cell viability reduction should be evaluated in order to identify the lowest observed effect concentration and peak values following meaningful p53 induction.
Collapse
Affiliation(s)
- Carolina Di Paolo
- Department of Ecosystem Analysis, Institute for Environmental Research, Aachen Biology and Biotechnology (ABBt), RWTH Aachen University, Aachen, Germany.
| | - Yvonne Müller
- Department of Ecosystem Analysis, Institute for Environmental Research, Aachen Biology and Biotechnology (ABBt), RWTH Aachen University, Aachen, Germany
| | - Beat Thalmann
- Department of Ecosystem Analysis, Institute for Environmental Research, Aachen Biology and Biotechnology (ABBt), RWTH Aachen University, Aachen, Germany
| | - Henner Hollert
- Department of Ecosystem Analysis, Institute for Environmental Research, Aachen Biology and Biotechnology (ABBt), RWTH Aachen University, Aachen, Germany
- College of Resources and Environmental Science, Chongqing University, 1 Tiansheng Road, Beibei, Chongqing, 400715, China
- College of Environmental Science and Engineering and State Key Laboratory of Pollution Control and Resource Reuse, Tongji University, 1239 Siping Road, Shanghai, China
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, China
| | - Thomas-Benjamin Seiler
- Department of Ecosystem Analysis, Institute for Environmental Research, Aachen Biology and Biotechnology (ABBt), RWTH Aachen University, Aachen, Germany
| |
Collapse
|
50
|
Afkhami-Ardakani M, Hasanzadeh S, Shahrooz R, Delirezh N, Malekinejad H. Antioxidant effects of Spirulina platensis ( Arthrospira platensis) on cyclophosphamide-induced testicular injury in rats. VETERINARY RESEARCH FORUM : AN INTERNATIONAL QUARTERLY JOURNAL 2018; 9:35-41. [PMID: 29719662 PMCID: PMC5913559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 09/12/2017] [Indexed: 10/24/2022]
Abstract
Cyclophosphamide (CP) is known to reduce fertility. The protective effects of Spirulina plantesis (SP) against CP-induced testicular toxicity were investigated. Male Wistar rats were categorized into eight groups (n = 7). Four groups of rats were administered CP at a dose of 5 mg in 5 mL distilled water kg-1 per day orally. Two of these groups were received SP (500 and 1000 mg kg-1 per day) orally after CP administration. One of these groups was also received vitamin E (100 mg kg-1 per day) intraperitoneally. A vehicle treated control group, two SP control groups (500 and 1000 mg kg-1 per day) and a vitamin E control group were also included. Body and testes weights, sperm count, serum levels of glutathione peroxidase (GPx), malondialdehyde (MDA), histological and histomorphometric alternations in testes were investigated after four weeks. The CP-treated group exhibited significant decreases in the body and testes weights and spermatogenic activities. Several histological alterations were observed in this group. The CP treatment caused a significant reduction in sperm count, in serum level of GPx, as well increased serum concentration of MDA. The SP co-administration caused an increase in GPx serum level, a decrease in MDA serum level and improvements in histological and histomorphometric alternations. Vitamin E co-treatment showed partial recovery in above-mentioned parameters. These results suggest that SP due to a reduction in oxidative stress has more effective protection against CP-induced reproductive damages in rat than vitamin E.
Collapse
Affiliation(s)
| | - Shapour Hasanzadeh
- Department of Basic Sciences, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran;,Correspondence:: Shapour Hasanzadeh. DVM, PhD, Department of Basic Sciences, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran. E-mail:
| | - Rasoul Shahrooz
- Department of Basic Sciences, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran;
| | - Norouz Delirezh
- Department of Microbiology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran;
| | - Hassan Malekinejad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|