1
|
Alkandahri MY, Sadino A, Pamungkas BT, Oktoba Z, Arfania M, Yuniarsih N, Wahyuningsih ES, Dewi Y, Winarti SA, Dinita ST. Potential Nephroprotective Effect of Kaempferol: Biosynthesis, Mechanisms of Action, and Clinical Prospects. Adv Pharmacol Pharm Sci 2024; 2024:8907717. [PMID: 39377015 PMCID: PMC11458287 DOI: 10.1155/2024/8907717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 08/16/2024] [Accepted: 09/10/2024] [Indexed: 10/09/2024] Open
Abstract
Kidney is an essential organ that is highly susceptible to cellular injury caused by various toxic substances in the blood. Several studies have shown that untreated injuries to this organ can cause glomerulosclerosis, tubulointerstitial fibrosis, and tubular cell apoptosis, leading to kidney failure. Despite significant advancements in modern treatment, there is no fully effective drug for repairing its function, providing complete protection, and assisting in cell regeneration. Furthermore, some available medications have been reported to exacerbate injuries, showing the need to explore alternative treatments. Natural drugs are currently being explored as a new therapeutic strategy for managing kidney diseases. Kaempferol, a polyphenol found in plants, including vegetables, legumes, and fruits, has been extensively studied in various nephrotoxicity protocols. The compound has been reported to have potential as a nephroprotective agent with beneficial effects on various physiological pathways, such as CPL-induced kidney injury, DOX, LPO, ROS, RCC, and diabetic nephropathy. Therefore, this study aims to provide a brief overview of the current nephroprotective effects of kaempferol, as well as its molecular mechanisms of action, biosynthesis pathways, and clinical prospects.
Collapse
Affiliation(s)
- Maulana Yusuf Alkandahri
- Department of Pharmacology and Clinical PharmacyFaculty of PharmacyUniversitas Buana Perjuangan Karawang, Karawang, West Java, Indonesia
| | - Asman Sadino
- Department of PharmacyFaculty of Mathematics and Natural ScienceUniversitas Garut, Garut, West Java, Indonesia
| | - Barolym Tri Pamungkas
- Department of Pharmaceutical BiologyFaculty of PharmacyUniversitas Mulawarman, Samarinda, East Kalimantan, Indonesia
| | - Zulpakor Oktoba
- Department of PharmacyFaculty of MedicineUniversitas Lampung, Bandar Lampung, Indonesia
| | - Maya Arfania
- Department of Pharmacology and Clinical PharmacyFaculty of PharmacyUniversitas Buana Perjuangan Karawang, Karawang, West Java, Indonesia
| | - Nia Yuniarsih
- Department of Pharmaceutical TechnologyFaculty of PharmacyUniversitas Buana Perjuangan Karawang, Karawang, West Java, Indonesia
| | - Eko Sri Wahyuningsih
- Department of Pharmaceutical BiologyFaculty of PharmacyUniversitas Buana Perjuangan Karawang, Karawang, West Java, Indonesia
| | - Yuliani Dewi
- Faculty of PharmacyUniversitas Buana Perjuangan Karawang, Karawang, West Java, Indonesia
| | - Sri Ayu Winarti
- Faculty of PharmacyUniversitas Buana Perjuangan Karawang, Karawang, West Java, Indonesia
| | - Sri Tantia Dinita
- Faculty of PharmacyUniversitas Buana Perjuangan Karawang, Karawang, West Java, Indonesia
| |
Collapse
|
2
|
Manavi MA, Fathian Nasab MH, Mohammad Jafari R, Dehpour AR. Mechanisms underlying dose-limiting toxicities of conventional chemotherapeutic agents. J Chemother 2024:1-31. [PMID: 38179685 DOI: 10.1080/1120009x.2023.2300217] [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: 09/20/2023] [Accepted: 12/21/2023] [Indexed: 01/06/2024]
Abstract
Dose-limiting toxicities (DLTs) are severe adverse effects that define the maximum tolerated dose of a cancer drug. In addition to the specific mechanisms of each drug, common contributing factors include inflammation, apoptosis, ion imbalances, and tissue-specific enzyme deficiencies. Among various DLTs are bleomycin-induced pulmonary fibrosis, doxorubicin-induced cardiomyopathy, cisplatin-induced nephrotoxicity, methotrexate-induced hepatotoxicity, vincristine-induced neurotoxicity, paclitaxel-induced peripheral neuropathy, and irinotecan, which elicits severe diarrhea. Currently, specific treatments beyond dose reduction are lacking for most toxicities. Further research on cellular and molecular pathways is imperative to improve their management. This review synthesizes preclinical and clinical data on the pharmacological mechanisms underlying DLTs and explores possible treatment approaches. A comprehensive perspective reveals knowledge gaps and emphasizes the need for future studies to develop more targeted strategies for mitigating these dose-dependent adverse effects. This could allow the safer administration of fully efficacious doses to maximize patient survival.
Collapse
Affiliation(s)
- Mohammad Amin Manavi
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Razieh Mohammad Jafari
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Dehpour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Zeng B, Liu Y, Xu J, Niu L, Wu Y, Zhang D, Tang X, Zhu Z, Chen Y, Hu L, Yu S, Yu P, Zhang J, Wang W. Future Directions in Optimizing Anesthesia to Reduce Perioperative Acute Kidney Injury. Am J Nephrol 2023; 54:434-450. [PMID: 37742618 DOI: 10.1159/000533534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 08/01/2023] [Indexed: 09/26/2023]
Abstract
BACKGROUND Perioperative acute kidney injury (AKI) is common in surgical patients and is associated with high morbidity and mortality. There are currently few options for AKI prevention and treatment. Due to its complex pathophysiology, there is no efficient medication therapy to stop the onset of the injury or repair the damage already done. Certain anesthetics, however, have been demonstrated to affect the risk of perioperative AKI in some studies. The impact of anesthetics on renal function is particularly important as it is closely related to the prognosis of patients. Some anesthetics can induce anti-inflammatory, anti-necrotic, and anti-apoptotic effects. Propofol, sevoflurane, and dexmedetomidine are a few examples of anesthetics that have protective association with AKI in the perioperative period. SUMMARY In this study, we reviewed the clinical characteristics, risk factors, and pathogenesis of AKI. Subsequently, the protective effects of various anesthetic agents against perioperative AKI and the latest research are introduced. KEY MESSAGE This work demonstrates that a thorough understanding of the reciprocal effects of anesthetic drugs and AKI is crucial for safe perioperative care and prognosis of patients. However, more complete mechanisms and pathophysiological processes still need to be further studied.
Collapse
Affiliation(s)
- Bin Zeng
- Department of Gastroenterology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Yinuo Liu
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Nanchang, China,
- The Second Clinical Medical College of Nanchang University, Nanchang, China,
| | - Jiawei Xu
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Liyan Niu
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- The Second Clinical Medical College of Nanchang University, Nanchang, China
- Huan Kui College, Nanchang University, Nanchang, China
| | - Yuting Wu
- Huan Kui College, Nanchang University, Nanchang, China
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Deju Zhang
- Huan Kui College, Nanchang University, Nanchang, China
- Food and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Hong Kong, Hong Kong, China
| | - Xiaoyi Tang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zicheng Zhu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yixuan Chen
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Leilei Hu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shuchun Yu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Peng Yu
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wenting Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
- Department of Cardiopulmonary Bypass, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
4
|
Mansour M, Nasr M, Ahmed-Farid OAH, Ahmed RF. Intranasal ondansetron microemulsion counteracting the adverse effects of cisplatin: animal study. Pharmacol Rep 2023; 75:199-210. [PMID: 36517694 PMCID: PMC9889460 DOI: 10.1007/s43440-022-00435-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/25/2022] [Accepted: 11/09/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Cisplatin is considered one of the most effective and commonly used chemotherapeutic drugs, but despite its high therapeutic effectiveness, most patients treated with cisplatin suffer from nausea and vomiting, neurotoxic side effects, and cerebral psychiatric disorders such as depression. Therefore, the aim of the current work was to explore whether a selective 5-HT3 receptor antagonist (Ondansetron) administered via the oral route or intranasally in microemulsion form would alleviate cisplatin's adverse effects. METHODS The selected ondansetron microemulsion was characterized in vitro for particle size, polydispersity, zeta potential, morphology, and nasal permeation, and in vivo in terms of anti-emetic and antidepressant activity, with the assessment of biochemical markers in brain homogenates. RESULTS Results revealed that both orally administered ondansetron and intranasally administered microemulsion were able to counteract the pica effect by increasing food consumption, water intake, and decreasing kaolin intake. They were also able to increase BDNF, normalize IL-6, increase serotonin, and normalize NOx, MDA, GSSH/GSH as well as 8OHdG levels in rats' brain homogenates. The intranasal ondansetron microemulsion displayed superiority compared to oral conventional ondansetron in terms of increasing food intake, reduction of stomach content, and normalization of serotonin turnover. CONCLUSION Ondansetron microemulsion can be administered by an alternative route of administration (intranasal) rather than oral, for patients on cisplatin chemotherapy.
Collapse
Affiliation(s)
- Mai Mansour
- grid.7269.a0000 0004 0621 1570Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, African Organization Unity Street, Cairo, 11566 Egypt
| | - Maha Nasr
- grid.7269.a0000 0004 0621 1570Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, African Organization Unity Street, Cairo, 11566 Egypt
| | - Omar A. H. Ahmed-Farid
- grid.419698.bDepartment of Physiology, National Organization for Drug Control and Research, Giza, 12553 Egypt
| | - Rania F. Ahmed
- grid.419725.c0000 0001 2151 8157Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Centre (ID: 60014618), Dokki, Giza, 12622 Egypt
| |
Collapse
|
5
|
Xiong D, Xiong C. Early Postoperative Ondansetron Exposure is Associated with Reduced 90-Day Mortality in Patients Undergoing Cardiac Surgery. Front Surg 2022; 9:885137. [PMID: 35784927 PMCID: PMC9243460 DOI: 10.3389/fsurg.2022.885137] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 05/27/2022] [Indexed: 11/13/2022] Open
Abstract
Background Ondansetron is a widely used anti-emetic for the prevention and treatment of nausea and vomiting for patients in critical care. Recent retrospective cohort studies suggest the potential beneficial effects of ondansetron in critically ill patients. In this study, we investigate the impact of ondansetron use on patient outcomes after cardiac surgery. Material and Methods The MIMIC-III database was used to identify two types of cardiac surgical patients: those who were administered early ondansetron and those who were not given this early medication in the first 48 h in the postoperative period. Multivariable logistic regression was used to investigate the effect of ondansetron exposure on 90-day mortality, acute kidney injury, and malignant ventricular arrhythmias. Sensitivity analyses utilizing the inverse probability of treatment weighting and covariate balancing propensity score models were conducted to test the robustness of our findings. Results A total of 12.4% of patients received ondansetron. Ondansetron use was associated with a lower risk of 90-day mortality in the multivariable logistic regression model (OR: 0.31, 95% CI: 0.13 to 0.72; P = 0.006) and sensitivity analyses. Additionally, ondansetron exposure was associated with less postoperative acute kidney injury (OR: 0.82, 95%CI: 0.69 to 0.96; P = 0.017) but did not increase the risk of postoperative malignant ventricular arrhythmias (OR: 0.38, 95%CI: 0.09 to 1.16; P = 0.191). Conclusions In a population of cardiac surgical patients, early postoperative use of ondansetron appears to be associated with decreased 90-day mortality and acute kidney injury.
Collapse
Affiliation(s)
- Dexin Xiong
- Department of Thoracic Surgery, Wuhan Red Cross Hospital, Wuhan, China
| | - Chao Xiong
- Department of Anesthesiology, Fuwai Hospital, National Center of Cardiovascular Diseases, Chinese Academy of Medical Sciences, and Peking Union Medical College, Beijing, China
- Correspondence: Chao Xiong
| |
Collapse
|
6
|
Saad AAA, Zhang F, Mohammed EAH, Wu X. Clinical Aspects of Drug–Drug Interaction and Drug Nephrotoxicity at Renal Organic Cation Transporters 2 (OCT2) and Multidrug and Toxin Exclusion 1, and 2-K (MATE1/MATE2-K). Biol Pharm Bull 2022; 45:382-393. [DOI: 10.1248/bpb.b21-00916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
| | - Fan Zhang
- Department of Pharmacy, the First Hospital of Lanzhou University
| | | | - Xin’an Wu
- Department of Pharmacy, the First Hospital of Lanzhou University
| |
Collapse
|
7
|
Domnik NJ, Vincent SG, Fisher JT. Mechanosensitivity of Murine Lung Slowly Adapting Receptors: Minimal Impact of Chemosensory, Serotonergic, and Purinergic Signaling. Front Physiol 2022; 13:833665. [PMID: 35250636 PMCID: PMC8889033 DOI: 10.3389/fphys.2022.833665] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 01/19/2022] [Indexed: 11/13/2022] Open
Abstract
Murine slowly adapting receptors (SARs) within airway smooth muscle provide volume-related feedback; however, their mechanosensitivity and morphology are incompletely characterized. We explored two aspects of SAR physiology: their inherent static mechanosensitivity and a potential link to pulmonary neuroepithelial bodies (NEBs). SAR mechanosensitivity displays a rate sensitivity linked to speed of inflation; however, to what extent static SAR mechanosensitivity is tuned for the very rapid breathing frequency (B f ) of small mammals (e.g., mouse) is unclear. NEB-associated, morphologically described smooth muscle-associated receptors (SMARs) may be a structural analog for functionally characterized SARs, suggesting functional linkages between SARs and NEBs. We addressed the hypotheses that: (1) rapid murine B f is associated with enhanced in vivo SAR static sensitivity; (2) if SARs and NEBs are functionally linked, stimuli reported to impact NEB function would alter SAR mechanosensitivity. We measured SAR action potential discharge frequency (AP f, action potentials/s) during quasi-static inflation [0-20 cmH2O trans-respiratory pressure (PTR)] in NEB-relevant conditions of hypoxia (FIO2 = 0.1), hypercarbia (FICO2 = 0.1), and pharmacologic intervention (serotonergic 5-HT3 receptor antagonist, Tropisetron, 4.5 mg/kg; P2 purinergic receptor antagonist, Suramin, 50 mg/kg). In all protocols, we obtained: (1) AP f vs. PTR; (2) PTR threshold; and (3) AP f onset at PTR threshold. The murine AP f vs. PTR response comprises high AP f (average maximum AP f: 236.1 ± 11.1 AP/s at 20 cmH2O), a low PTR threshold (mean 2.0 ± 0.1 cmH2O), and a plateau in AP f between 15 and 20 cmH2O. Murine SAR mechanosensitivity (AP f vs. PTR) is up to 60% greater than that reported for larger mammals. Even the maximum difference between intervention and control conditions was minimally impacted by NEB-related alterations: Tropisetron -7.6 ± 1.8% (p = 0.005); Suramin -10.6 ± 1.5% (p = 0.01); hypoxia +9.3 ± 1.9% (p < 0.001); and hypercarbia -6.2 ± 0.9% (p < 0.001). We conclude that the high sensitivity of murine SARs to inflation provides enhanced resolution of operating lung volume, which is aligned with the rapid B f of the mouse. We found minimal evidence supporting a functional link between SARs and NEBs and speculate that the <10% change in SAR mechanosensitivity during altered NEB-related stimuli is not consistent with a meaningful physiologic role.
Collapse
Affiliation(s)
- Nicolle J. Domnik
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON, Canada
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Sandra G. Vincent
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON, Canada
| | - John T. Fisher
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON, Canada
| |
Collapse
|
8
|
Yuan J, Liang X, Zhou W, Feng J, Wang Z, Shen S, Guan X, Zhao L, Deng F. TRPA1 promotes cisplatin-induced nephrotoxicity through inflammation mediated by the MAPK/NF-κB signaling pathway. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1578. [PMID: 34790784 PMCID: PMC8576655 DOI: 10.21037/atm-21-5125] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 10/21/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND The nephrotoxicity induced by cisplatin (DDP) has been a severe obstacle for its clinical use in anticancer treatment. The apoptosis and inflammation induced by DDP are the main causes of the nephrotoxicity. Transient receptor potential ankyrin 1 (TRPA1) is a non-selective cation ligand-gated channel that is involved in the inflammation progress. METHODS The apoptosis, inflammation, MAPK/NF-κB signaling pathway, and TRPA1 expression were assessed after HEK293 cells had been induced by DDP, and the role of TRPA1 in apoptosis and inflammation of DDP-induced HEK293 cells treated with TRPA1 antagonist HC-030031 was also evaluated by quantitative reverse transcription-polymerase chain reaction (qRT-PCR), flow cytometry, and western blot assays. RESULTS The cell viability was reduced by DDP in both a time-dependent and dose-dependent manner with a minimal cytotoxic concentration of 10 μM. Moreover, DDP induced an enhancement of the apoptosis and inflammation in a dose-dependent manner, as indicated by the increase of the relative protein level of cleaved-caspase3 (cleaved-cas3), the cleavage product of caspase-3 substrate poly-ADP-ribose polymerase (cleaved-PARP) and inducible nitric oxide synthase (iNOS), and the messenger RNA (mRNA) expression level of interleukin (IL)-1β, IL-6, tumor necrosis factor-α (TNF-α), and interferon-γ (INF-γ). Additionally, DDP treatment increased the protein phosphorylation expression of IKKβ, JNK, ERK, and p38 in a dose-dependent manner, which was antagonized by the treatment of NF-κB-specific inhibitor BAY 11-7082 and pan-MAPK inhibitor U0126. It was also found that DDP upregulated the expression of TRPA1 at both the mRNA and protein levels in a dose-dependent manner. Besides, block of TRPA1 with HC-030031 relieved the apoptosis, diminished the level of IL-1β, IL-6, TNF-α, and INF-γ, reduced the level of cleaved-cas3, cleaved-PARP, and iNOS, decreased the p-IKKβ, p-JNK, p-ERK, and p-p38 expression, and enhanced the expression of IκBα. CONCLUSIONS Taken together, these results indicate that TRPA1 regulates DDP-induced nephrotoxicity via inflammation mediated by the MAPK/NF-κB signaling pathway in HEK293 cells.
Collapse
Affiliation(s)
- Jinyan Yuan
- Department of Nephrology, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiao Liang
- Department of Internal Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Wei Zhou
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jing Feng
- Department of Traditional Chinese Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Zhenyang Wang
- Department of Nephrology, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Shaoxian Shen
- Department of Nephrology, Jinniu Hospital of Sichuan Provincial People’s Hospital and Chengdu Jinniu District People’s Hospital, Chengdu, China
| | - Xin Guan
- Department of Nephrology, Jinniu Hospital of Sichuan Provincial People’s Hospital and Chengdu Jinniu District People’s Hospital, Chengdu, China
| | - Liangbin Zhao
- Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fei Deng
- Department of Nephrology, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Department of Nephrology, Jinniu Hospital of Sichuan Provincial People’s Hospital and Chengdu Jinniu District People’s Hospital, Chengdu, China
| |
Collapse
|
9
|
Effects of ondansetron use on outcomes of acute kidney injury in critically ill patients: An analysis based on the MIMIC-IV database. J Crit Care 2021; 66:117-122. [PMID: 34509800 DOI: 10.1016/j.jcrc.2021.07.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 07/07/2021] [Accepted: 07/15/2021] [Indexed: 12/19/2022]
Abstract
PURPOSE Acute kidney injury (AKI) in intensive care units (ICUs) is a health priority with limited treatment options. This study aimed to estimate the effects of ondansetron use on AKI patient outcomes. MATERIALS AND METHODS We used the MIMIC-IV database to compare AKI patient mortality in the ICU with and without ondansetron and calculated hazard ratios (HRs) with 95% confidence intervals (95%CI) by multivariable Cox regression. Propensity score matching (PSM) and inverse probability of treatment weighting (IPTW) were applied to adjust for confounding factors. RESULTS In total, 26,004 AKI patients were included. Ondansetron use reduced in-hospital mortality risk by 16% among AKI patients (HR: 0.84, 95%CI: 0.77-0.90, p < 0.001). In-hospital mortality was significantly reduced among patients administered ondansetron at AKI stage 1 (11.4% vs. 16.5%. p < 0.001) and stage 2 (16.1% vs. 19.6%. p < 0.001) but not stage 3 (24.0% vs. 23.9%. p = 0.890). Patients younger than 60 years or receiving surgery received greater benefits from ondansetron use. (HR: 0.62, 95%CI:0.53-0.72 and HR: 0.59, 95%CI:0.50-0.69, respectively). CONCLUSIONS This cohort study showed that ondansetron use is significantly associated with reduced risk-adjusted in-hospital mortality in stages 1 and 2 AKI patients in the ICU. Further randomized controlled trials are needed.
Collapse
|
10
|
Gholizadeh-Ghaleh Aziz S, Naderi R, Mahmodian N. Ameliorative effects of tropisetron on liver injury in streptozotocin-induced diabetic rats. Arch Physiol Biochem 2021; 127:367-372. [PMID: 31306054 DOI: 10.1080/13813455.2019.1640743] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
This study aimed to evaluate the effect of tropisetron on liver injury induced by diabetes. Thirty-five male Wistar rats were assigned to five groups (n = 7): control (C), tropisetron (T), diabetic (D), diabetic + tropisetron (D + T) and diabetic + glibenclamide (D + G). Diabetic rats were treated with tropisetron (3 mg/kg body weight/day) or glibenclamide (1 mg/kg/day) for two weeks. Liver from diabetic rats exhibited a significant increase in alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP), cholesterol (Chol), triglycerides (TG), low-density lipoprotein (LDL), and atherogenic index, and a significant decrease in liver glycogen, serum albumin and high-density lipoprotein. Treatment with tropisetron significantly abrogated diabetes-induced perturbation in these parameters. These effects were equipotent with glibenclamide, suggesting that tropisetron treatment is associated with a hepatoprotective effect against diabetic injury. Therefore, the results of this study manifested the significance of using tropisetron as a promising remedial agent to improve diabetic complications.
Collapse
Affiliation(s)
| | - Roya Naderi
- Nephrology and Kidney Transplant Research Center, Urmia University of Medical Sciences, Urmia, Iran
- Department of Physiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Nima Mahmodian
- Department of Physiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
11
|
Kamil M, Fatima A, Ullah S, Ali G, Khan R, Ismail N, Qayum M, Irimie M, Dinu CG, Ahmedah HT, Cocuz ME. Toxicological Evaluation of Novel Cyclohexenone Derivative in an Animal Model through Histopathological and Biochemical Techniques. TOXICS 2021; 9:119. [PMID: 34070633 PMCID: PMC8227666 DOI: 10.3390/toxics9060119] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 04/04/2021] [Accepted: 05/19/2021] [Indexed: 12/13/2022]
Abstract
Toxicity studies were conducted to provide safety data of potential drug candidates by determining lethal and toxic doses. This study was designed for pre-clinical evaluation of novel cyclohexenone derivative with respect to the acute and sub-acute toxicity along with the diabetogenic potential. Acute and sub-acute toxicity were assessed after intraperitoneal (i.p) injection of the investigational compound through selected doses for 21 days. This was followed by assessment of isolated body organs (liver, kidney, heart and pancreas) via biochemical indicators and histopathological techniques. No signs of toxicity were revealed in the study of acute toxicity. Similarly, a sub-acute toxicity study showed no significant difference in biochemical indicators on 11th and 21st days between treated and control groups. However, in blood urea nitrogen (BUN) and random blood glucose/sugar (RBS) values, significant differences were recorded. Histopathological evaluation of liver, kidney, pancreas and heart tissues revealed mild to severe changes in the form of steatosis, inflammation, fibrosis, necrosis and myofibrillary damages on 11th and 21st days of treatment. In conclusion, the median lethal dose of the tested compound was expected to be greater than 500 mg/kg. No significant change occurred in selected biomarkers, except BUN and RBS levels, but a histopathological study showed moderate toxic effect on liver, kidney, pancreas and heart tissues by the cyclohexenone derivative.
Collapse
Affiliation(s)
- Muhammad Kamil
- Department of Pharmacy, University of Peshawar, Peshawar 25120, Pakistan; (M.K.); (A.F.); (G.A.)
| | - Arifa Fatima
- Department of Pharmacy, University of Peshawar, Peshawar 25120, Pakistan; (M.K.); (A.F.); (G.A.)
| | - Sami Ullah
- Department of Pharmacy, University of Peshawar, Peshawar 25120, Pakistan; (M.K.); (A.F.); (G.A.)
| | - Gowhar Ali
- Department of Pharmacy, University of Peshawar, Peshawar 25120, Pakistan; (M.K.); (A.F.); (G.A.)
| | - Rasool Khan
- Department of Organic Chemistry, Institute of Chemical Sciences, University of Peshawar, Peshawar 25120, Pakistan;
| | - Naila Ismail
- Department of Pathology, Kabir Medical College, Gandhara University, Peshawar 25000, Pakistan;
| | - Mughal Qayum
- Department of Pharmacy, Kohat University of Science and Technology, Kohat 26000, Pakistan;
| | - Marius Irimie
- Faculty of Medicine, Transilvania University of Brasov, 500019 Brasov, Romania;
| | | | - Hanadi Talal Ahmedah
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Rabigh 25732, Saudi Arabia
| | - Maria Elena Cocuz
- Faculty of Medicine, Transilvania University of Brasov, 500019 Brasov, Romania;
| |
Collapse
|
12
|
Karimollah A, Hemmatpur A, Hosseini N, Manshadi MD. Tropisetron balances immune responses via TLR2, TLR4 and JAK2/STAT3 signalling pathway in LPS-stimulated PBMCs. Basic Clin Pharmacol Toxicol 2021; 128:669-676. [PMID: 33523585 DOI: 10.1111/bcpt.13565] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/18/2021] [Accepted: 01/27/2021] [Indexed: 11/28/2022]
Abstract
Numerous documents have been stated that tropisetron, an antagonist of the 5-HT3 receptor and α7nAChR agonist, modulates immune responses. However, the mechanistic basis for this aspect of tropisetron action is largely unknown. Here, the immuno-modulatory effects of tropisetron are investigated, focusing on the possible molecular targets and the mechanisms. Aside from the well-characterized role in immune signalling, JAK2/STAT3, TLR2 and TLR4 are signal transducers linked to both immuno-modulatory actions of acetylcholine and serotonin. Therefore, we evaluated their involvement in the immunoregulatory effects of tropisetron. To test the hypothesis, we assessed the expression of pro-/anti-inflammatory cytokines including TNF-α, IL-1β, IL-17 and IL-10 following tropisetron treatment in lipopolysaccharide (LPS)-stimulated peripheral blood mononuclear cells (PBMCs) derived from healthy subjects. Tropisetron up-regulates the transcription of TLR2, TLR4, JAK2 and STAT3 genes. Tropisetron also increases the expression of target pro-inflammatory cytokines, although considerably suppresses the pro-inflammatory cytokines (IL-1β, IL-17 and TNF-α) levels in media. Tropisetron notably promotes both IL-10 gene expression and secretion. These findings confirm the antiphlogistic properties of tropisetron. The present data also shed light on a new aspect of tropisetron immune-modulatory action that engaged TLR2, TLR4 and JAK2/STAT3 signalling cascades.
Collapse
Affiliation(s)
- Alireza Karimollah
- Department of Pharmacology, School of Pharmacy, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Anahid Hemmatpur
- Department of Biochemistry, School of medicine, Shahid Sadoughi University of medical sciences and Health Services, Yazd, Iran
| | - Nafise Hosseini
- Department of Pharmacology, School of Pharmacy, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Mahdi Dehghan Manshadi
- Reproductive Immunology Research Center, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| |
Collapse
|
13
|
Okur ME, Ayla Ş, Karadağ AE, Çiçek Polat D, Demirci S, Seçkin İ. Opuntia ficus indica Fruits Ameliorate Cisplatin-Induced Nephrotoxicity in Mice. Biol Pharm Bull 2021; 43:831-838. [PMID: 32378560 DOI: 10.1248/bpb.b19-01044] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This study aims to determine the potential renal protective effects of Opuntia ficus-indica (L.) Miller (OFI) fruits against cisplatin-induced nephrotoxicity in mice. The antioxidant activity of OFI methanol extract was calculated by 2,2-diphenyl-1-picrylhydrazyl (DPPH) and 2,2'-azino-bis-3-ethylbenzthiazoline-6-sulfonic acid (ABTS) free radical scavenging assays. Furthermore, the LC-mass spectroscopy (MS) analysis of the OFI fruit extract was carried out. Mice were treated with OFI extract (250 mg/kg) for 10 d and injected with a single dose of cisplatin (20 mg/kg) on the 7th day. The blood samples were collected to measure blood urea nitrogen (BUN) and serum creatinine level on the 10th day. Their kidneys were removed for histopathological examination. The renal morphological alterations were assessed through the mesangial matrix index and transmission electron microscopy (TEM). The OFI fruit extract showed significant in vitro antioxidant activity. In further, it was revealed that the cisplatin-induced nephrotoxicity in mice was ameliorated; this outcome was supported by both histological examination results and the depicted reduced levels of BUN and serum creatinine. The potent antioxidant compounds which were detected in the extract of OFI fruits such as myricetin, quercetin, luteolin might be responsible for the observed renoprotective effect. The results clarified that the OFI fruit extract could ameliorate cisplatin-induced renal toxicity in mice via including antioxidant and renoprotective compounds.
Collapse
Affiliation(s)
- Mehmet Evren Okur
- University of Health Sciences, Faculty of Pharmacy, Department of Pharmacology
| | - Şule Ayla
- Istanbul Medipol University, School of Medicine, Department of Histology and Embryology
| | - Ayşe Esra Karadağ
- Istanbul Medipol University, School of Pharmacy, Department of Pharmacognosy.,Anadolu University, Graduate School of Health Sciences
| | - Derya Çiçek Polat
- Ankara University, Faculty of Pharmacy, Department of Pharmaceutical Botany
| | - Sibel Demirci
- Cerrahpaşa University, School of Medicine, Department of Histology and Embryology
| | - İsmail Seçkin
- Cerrahpaşa University, School of Medicine, Department of Histology and Embryology
| |
Collapse
|
14
|
An Integrated In Silico and In Vivo Approach to Identify Protective Effects of Palonosetron in Cisplatin-Induced Nephrotoxicity. Pharmaceuticals (Basel) 2020; 13:ph13120480. [PMID: 33419241 PMCID: PMC7766590 DOI: 10.3390/ph13120480] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/17/2020] [Accepted: 12/17/2020] [Indexed: 11/17/2022] Open
Abstract
Cisplatin is widely used to treat various types of cancers, but it is often limited by nephrotoxicity. Here, we employed an integrated in silico and in vivo approach to identify potential treatments for cisplatin-induced nephrotoxicity (CIN). Using publicly available mouse kidney and human kidney organoid transcriptome datasets, we first identified a 208-gene expression signature for CIN and then used the bioinformatics database Cmap and Lincs Unified Environment (CLUE) to identify drugs expected to counter the expression signature for CIN. We also searched the adverse event database, Food and Drug Administration. Adverse Event Reporting System (FAERS), to identify drugs that reduce the reporting odds ratio of developing cisplatin-induced acute kidney injury. Palonosetron, a serotonin type 3 receptor (5-hydroxytryptamine receptor 3 (5-HT3R)) antagonist, was identified by both CLUE and FAERS analyses. Notably, clinical data from 103 patients treated with cisplatin for head and neck cancer revealed that palonosetron was superior to ramosetron in suppressing cisplatin-induced increases in serum creatinine and blood urea nitrogen levels. Moreover, palonosetron significantly increased the survival rate of zebrafish exposed to cisplatin but not to other 5-HT3R antagonists. These results not only suggest that palonosetron can suppress CIN but also support the use of in silico and in vivo approaches in drug repositioning studies.
Collapse
|
15
|
Un H, Ugan RA, Gurbuz MA, Bayir Y, Kahramanlar A, Kaya G, Cadirci E, Halici Z. Phloretin and phloridzin guard against cisplatin-induced nephrotoxicity in mice through inhibiting oxidative stress and inflammation. Life Sci 2020; 266:118869. [PMID: 33309722 DOI: 10.1016/j.lfs.2020.118869] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/26/2020] [Accepted: 12/01/2020] [Indexed: 12/13/2022]
Abstract
AIM Cisplatin (Cis) is widely used chemotherapeutic and has some serious side effects as nephrotoxicity. Phloretin (PH) and Phloridzin (PZ) are known their anti-oxidant anti-inflammatory effects. We aimed to examine the protective effects of PH and PZ on cisplatin-induced nephrotoxicity. MAIN METHODS Totally, 48 Balb/C female mice were separated into eight groups (n = 6). First day, single dose of cisplatin (20 mg/kg intraperitoneal) was administered to induce toxicity. PH and PZ were given (50 and 100 mg/kg orally) to treatment groups during 3 days. After the experimental procedures serum renal function enzymes (BUN and Creatinine), oxidative parameters (SOD, GSH and MDA), nuclear agent NFKβ, inflammatory cytokines (Tnf-α and IL1β) and HSP70 expressions and histopathological assessments were analyzed. KEY FINDINGS Serum enzymes, tissue cytokines and oxidative stress were increased after the Cis treatment. PH and PZ treatments normalized all parameters compared to Cis administrated group. After the treatments, SOD activities and GSH levels were increased while MDA levels were decreased. PH and PZ treatments decreased Tnf-α, IL1β and NFKβ mRNA expressions. Cis significantly increased the HSP70 expression while PH and PZ administrations significantly decreased. Similar the biochemical and molecular results, PH and PZ showed positive effects on tissue pathological parameters. Cisplatin cause a lot of abnormal structures as tubular and glomeruli damages on the kidney. SIGNIFICANCE PH and PZ play important physiological roles in the prevention of nephrotoxicity. Antioxidant and anti-inflammatory effects of PH and PZ demonstrated visible protective effects in the cisplatin-induced nephrotoxicity model.
Collapse
Affiliation(s)
- Harun Un
- Agri Ibrahim Cecen University, Faculty of Pharmacy, Department of Biochemistry, Agri, Turkey.
| | - Rustem Anil Ugan
- Ataturk University, Faculty of Pharmacy, Department of Pharmacology, Erzurum, Turkey
| | - Muhammet Ali Gurbuz
- Ataturk University, Faculty of Medicine, Department of Histology and Embryology, Erzurum, Turkey
| | - Yasin Bayir
- Ataturk University, Faculty of Pharmacy, Department of Biochemistry, Erzurum, Turkey
| | - Aysenur Kahramanlar
- Ataturk University, Faculty of Pharmacy, Department of Biochemistry, Erzurum, Turkey
| | - Gokce Kaya
- Ataturk University, Faculty of Medicine, Department of Pharmacology, Erzurum, Turkey
| | - Elif Cadirci
- Ataturk University, Faculty of Medicine, Department of Pharmacology, Erzurum, Turkey; Clinical Research, Development and Design Application and Research Center, Ataturk University, Erzurum, Turkey
| | - Zekai Halici
- Ataturk University, Faculty of Medicine, Department of Pharmacology, Erzurum, Turkey; Clinical Research, Development and Design Application and Research Center, Ataturk University, Erzurum, Turkey
| |
Collapse
|
16
|
Casanova AG, Hernández-Sánchez MT, Martínez-Salgado C, Morales AI, Vicente-Vicente L, López-Hernández FJ. A meta-analysis of preclinical studies using antioxidants for the prevention of cisplatin nephrotoxicity: implications for clinical application. Crit Rev Toxicol 2020; 50:780-800. [PMID: 33170047 DOI: 10.1080/10408444.2020.1837070] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Cisplatin is an effective chemotherapeutic drug whose clinical use and efficacy are limited by its nephrotoxicity, which affects mainly the renal tubules and vasculature. It accumulates in proximal and distal epithelial tubule cells and causes oxidative stress-mediated cell death and malfunction. Consequently, many antioxidants have been tested for their capacity to prevent cisplatin nephrotoxicity. In this study, we made a systematic review of the literature and meta-analyzed 152 articles, which tested the nephroprotective effect of isolated compounds or mixtures of natural origin on cisplatin nephrotoxicity in preclinical models. This meta-analysis identified the most effective candidates and examined the efficacy obtained by antioxidants administered by the oral and intraperitoneal routes. By comparing with a recent, similar meta-analysis performed on clinical studies, this article identifies a disconnection between preclinical and clinical research, and contextualizes, discusses, and integrates the existing preclinical information toward the optimized selection of candidates to be further explored (clinical level). Despite proved efficacy, this article discusses the barriers limiting the clinical development of natural mixtures, such as those in extracts from Calendula officinalis flowers and Heliotropium eichwaldii roots. On the contrary, isolated compounds are more straightforward candidates, among which arjunolic acid and quercetin stand out in this meta-analysis.
Collapse
Affiliation(s)
- Alfredo G Casanova
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Department of Physiology and Pharmacology, University of Salamanca (USAL), Salamanca, Spain.,Fundación Instituto de Estudios de Ciencias de la Salud de Castilla y León (IECSCYL), Soria, Spain.,Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Salamanca, Spain.,National Network for Kidney Research REDINREN, Instituto de Salud Carlos III, Madrid, Spain
| | - M Teresa Hernández-Sánchez
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Department of Physiology and Pharmacology, University of Salamanca (USAL), Salamanca, Spain.,Fundación Instituto de Estudios de Ciencias de la Salud de Castilla y León (IECSCYL), Soria, Spain.,Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Salamanca, Spain.,National Network for Kidney Research REDINREN, Instituto de Salud Carlos III, Madrid, Spain
| | - Carlos Martínez-Salgado
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Department of Physiology and Pharmacology, University of Salamanca (USAL), Salamanca, Spain.,Fundación Instituto de Estudios de Ciencias de la Salud de Castilla y León (IECSCYL), Soria, Spain.,Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Salamanca, Spain.,National Network for Kidney Research REDINREN, Instituto de Salud Carlos III, Madrid, Spain
| | - Ana I Morales
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Department of Physiology and Pharmacology, University of Salamanca (USAL), Salamanca, Spain.,Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Salamanca, Spain.,National Network for Kidney Research REDINREN, Instituto de Salud Carlos III, Madrid, Spain.,Group of Biomedical Research on Critical Care (BioCritic), Valladolid, Spain
| | - Laura Vicente-Vicente
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Department of Physiology and Pharmacology, University of Salamanca (USAL), Salamanca, Spain.,Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Salamanca, Spain.,National Network for Kidney Research REDINREN, Instituto de Salud Carlos III, Madrid, Spain
| | - Francisco J López-Hernández
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Department of Physiology and Pharmacology, University of Salamanca (USAL), Salamanca, Spain.,Fundación Instituto de Estudios de Ciencias de la Salud de Castilla y León (IECSCYL), Soria, Spain.,Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), Salamanca, Spain.,National Network for Kidney Research REDINREN, Instituto de Salud Carlos III, Madrid, Spain.,Group of Biomedical Research on Critical Care (BioCritic), Valladolid, Spain
| |
Collapse
|
17
|
Zirak MR, Karimi G, Rahimian R, Jafarian AH, Hayes AW, Mehri S. Tropisetron ameliorates cyclophosphamide-induced hemorrhagic cystitis in rats. Eur J Pharmacol 2020; 883:173310. [PMID: 32619674 DOI: 10.1016/j.ejphar.2020.173310] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 06/20/2020] [Accepted: 06/22/2020] [Indexed: 01/05/2023]
Abstract
Hemorrhagic cystitis is one of the most important complications of cyclophosphamide, a drug widely used in cancer chemotherapy and bone marrow transplantation. 5-HT3 antagonists are anti-emetic agents and have been shown to have notable anti-inflammatory and antioxidant properties. This study was designed to investigate the possible protective effects of tropisetron against cyclophosphamide-induced hemorrhagic cystitis in rats. Hemorrhagic cystitis was induced in female rats by cyclophosphamide (270 mg/kg). Tropisetron (2.5, 5 and 7.5 mg/kg), granisetron (2.5 and 5 mg/kg), and ondansetron (5 mg/kg) were injected 15 min before, 4 and 8 h after cyclophosphamide. To evaluate the role of alpha7 nicotinic acetylcholine receptor (α7nAChR), its antagonist, methyllycaconitine (5 mg/kg) was administered 30 min before tropisetron. After 24 h, animals were killed under anesthesia. Macroscopic and histological changes were evaluated. Malondialdehyde (MDA), glutathione (GSH) and Evans blue were measured spectrophotometrically. Furthermore, the protein levels of p38 mitogen-activated protein kinases (P38 MAPK), p-P38, signal transducer and activator of transcription 3 (STAT3), p-STAT3 and Poly (ADP-ribose) polymerase (PARP) were determined using Western blot. Cyclophosphamide administration significantly induced histopathological damages and increased MDA, p-p38/p38, p-STAT3/STAT3, and PARP levels compared with the saline group. Tropisetron treatment diminished histopathological injuries as well as MDA level, and STAT3 activity compared to cyclophosphamide treated rats. Co-administration of methyllycaconitine with tropisetron, partially or completely reversed the protective effects of tropisetron. Our results showed that prophylactic administration of tropisetron markedly ameliorated the cyclophosphamide-induced bladder hemorrhage and inflammation in rats. These effects of tropisetron were α7nAChR dependent.
Collapse
Affiliation(s)
- Mohammad Reza Zirak
- Pharmaceutical Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gholamreza Karimi
- Pharmaceutical Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Rahimian
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Amir Hossein Jafarian
- Cancer Molecular Pathology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - A Wallace Hayes
- University of South Florida College of Public Health, Tampa, FL, USA; Michigan State University, East Lansing, MI, USA
| | - Soghra Mehri
- Pharmaceutical Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
18
|
Naderi R, Pourheydar B, Ghiasi R, Shafiei F. Modulatory effect of tropisetron in the liver of streptozotocin-induced diabetes in rats: biochemical and histological evidence. Horm Mol Biol Clin Investig 2020; 41:hmbci-2020-0002. [PMID: 32364517 DOI: 10.1515/hmbci-2020-0002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 04/04/2020] [Indexed: 12/19/2022]
Abstract
Objectives There is an association between diabetes and liver disorders. Oxidative stress plays a crucial role in the pathology of hepatic abnormalities in diabetes. In this study, the effect of Tropisetron on the oxidative damage and histological alterations in the liver of type 1 diabetes mellitus (DM) were evaluated. Methods Thiry-five male Wistar rats were randomly divided into five experimental groups (n = 7): control (C), tropisetron (T), diabetes (D), diabetes + tropisetron (D + T) and diabetes + glibenclamide (D + G). A single injection of streptozotocin (STZ, 50 mg/kg; i.p) was used to induce diabetes. Tropisetron (3 mg/kg; i.p), as a 5-HT3 receptor antagonist and glibenclamide (1 mg/kg; i.p), as a positive control were given once daily for 2 weeks. Finally, animals were euthanized and liver samples were obtained for histopathological examination and biochemical measurements including malondialdehyde (MDA), superoxide dismutase (SOD), glutathione peroxidase (GPx) levels. Results There is a significant increase in MDA (p < 0.001) level and a significant decrease (p < 0.001) in SOD and GPx contents in diabetic animals. Tropisetron attenuated MDA levels (p < 0.001) and enhanced SOD (p < 0.05) and GPx (p < 0.01) activities accompanied by histopathological improvement in the diabetes liver. Similar results were achieved in the rats treated with the standard drug, namely: glibenclamide. Conclusions Our findings indicate that tropisetron mitigates liver damage in the diabetes rats in part by attenuation of oxidative stress.
Collapse
Affiliation(s)
- Roya Naderi
- Nephrology and Kidney Transplant Research Center, Urmia University of Medical Sciences, Urmia, Islamic Republic of Iran.,Department of Physiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Islamic Republic of Iran
| | - Bagher Pourheydar
- Neurophysiology Research Center, Urmia University of Medical Sciences, Urmia, Islamic Republic of Iran.,Department of Anatomical Sciences, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Islamic Republic of Iran
| | - Rafigheh Ghiasi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran.,Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran
| | - Fardin Shafiei
- Department of Physiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Islamic Republic of Iran
| |
Collapse
|
19
|
Wang Z, Sun W, Sun X, Wang Y, Zhou M. Kaempferol ameliorates Cisplatin induced nephrotoxicity by modulating oxidative stress, inflammation and apoptosis via ERK and NF-κB pathways. AMB Express 2020; 10:58. [PMID: 32219583 PMCID: PMC7098399 DOI: 10.1186/s13568-020-00993-w] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 03/16/2020] [Indexed: 02/01/2023] Open
Abstract
Anticancer drug like Cisplatin are associated with serious problem like nephrotoxicity. The effect of Kaempferol is a plant-derived flavonoid compound. The present work evaluated the effect of Kaempferol in mouse model of Cisplatin mediated nephrotoxicity also the involved mechanism. Oxidative stress, kidney function, histology, inflammation, apoptosis, level of proteins, Nrf2 translocation and its effect on cascades such as NF-κB and ERK were studied. It was observed that the pre-treatment of KPF reduced the Cisplatin mediated oxidative stress, inflammation, apoptosis and ameliorated renal injury and its functioning. Kaempferol suppressed the Cisplatin induced infiltration of mononuclear cells, levels of TNF-α, iNOS, IL-12, activation of NF-κB, phosphorylation of IκBα and nuclear translocation of p65 in renal tissues. Also KPF attenuated Cisplatin mediated phosphorylation of p38, ERK1/2 and JNK in renal tissues. KPF also corrected the levels of renal antioxidants and elevated the nuclear levels of HO-1 and Nrf2 in renal tissues. KPF attenuated the Cisplatin mediated apoptosis via down-regulating the levels of TP53, Bax/Bcl2 imbalance, activating caspase-3/9 and PARP. The outcomes conclude that KPF ameliorates Cisplatin-mediated nephrotoxicity by modulating oxidative stress, inflammation and apoptosis via ERK and NF-κB pathway.
Collapse
|
20
|
Tomar A, Kaushik S, Khan SI, Bisht K, Nag TC, Arya DS, Bhatia J. The dietary isoflavone daidzein mitigates oxidative stress, apoptosis, and inflammation in CDDP-induced kidney injury in rats: Impact of the MAPK signaling pathway. J Biochem Mol Toxicol 2019; 34:e22431. [PMID: 31833131 DOI: 10.1002/jbt.22431] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 10/04/2019] [Accepted: 12/02/2019] [Indexed: 01/14/2023]
Abstract
Cisplatin-induced nephrotoxicity persists as a clinical problem despite several supportive measures to alleviate renal damage. Daidzein (DZ), a dietary isoflavone having antioxidant and anti-inflammatory activity, is investigated in this study for protective effects against cisplatin-induced renal injury in rats. DZ (25, 50, or 100 mg/kg; intraperitoneally; 10 days) was administered along with Cisplatin, single dose, on the 7th day of the experiment. On the 11th day, the rats were euthanized, and different samples were collected for analysis. Biochemical, histopathological, and molecular parameters were assessed to evaluate the effect of daidzein. Cisplatin injection resulted in renal dysfunction, lipid peroxidation that led to consumption of antioxidants, exaggerated apoptosis, and inflammation. These changes were associated with increase in the signaling proteins. DZ attenuated the toxic effects of cisplatin on the kidney at 100 mg/kg dose. The study concludes with the finding that daidzein imparts protection against the nephrotoxic effect of Cisplatin and can be considered as a novel, potential therapy.
Collapse
Affiliation(s)
- Ameesha Tomar
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Swati Kaushik
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Sana Irfan Khan
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Khushboo Bisht
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Tapas Chandra Nag
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - Dharamvir Singh Arya
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Jagriti Bhatia
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
21
|
Liu X. Transporter-Mediated Drug-Drug Interactions and Their Significance. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1141:241-291. [PMID: 31571167 DOI: 10.1007/978-981-13-7647-4_5] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Drug transporters are considered to be determinants of drug disposition and effects/toxicities by affecting the absorption, distribution, and excretion of drugs. Drug transporters are generally divided into solute carrier (SLC) family and ATP binding cassette (ABC) family. Widely studied ABC family transporters include P-glycoprotein (P-GP), breast cancer resistance protein (BCRP), and multidrug resistance proteins (MRPs). SLC family transporters related to drug transport mainly include organic anion-transporting polypeptides (OATPs), organic anion transporters (OATs), organic cation transporters (OCTs), organic cation/carnitine transporters (OCTNs), peptide transporters (PEPTs), and multidrug/toxin extrusions (MATEs). These transporters are often expressed in tissues related to drug disposition, such as the small intestine, liver, and kidney, implicating intestinal absorption of drugs, uptake of drugs into hepatocytes, and renal/bile excretion of drugs. Most of therapeutic drugs are their substrates or inhibitors. When they are comedicated, serious drug-drug interactions (DDIs) may occur due to alterations in intestinal absorption, hepatic uptake, or renal/bile secretion of drugs, leading to enhancement of their activities or toxicities or therapeutic failure. This chapter will illustrate transporter-mediated DDIs (including food drug interaction) in human and their clinical significances.
Collapse
Affiliation(s)
- Xiaodong Liu
- China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
22
|
Rashidi M, Bazi A, Shiran MR, Bagheri A, Mehrabadi AR, Kalantar H, Ghafouri Z, Hosseini SM. Tropisetron attenuates tumor growth and progression in an experimental model of mouse lung cancer. J Cell Biochem 2019; 121:1610-1622. [PMID: 31535406 DOI: 10.1002/jcb.29395] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 08/28/2019] [Indexed: 12/25/2022]
Abstract
The antineoplastic effects of 5-hydroxytryptamine (5-HT) receptor antagonists have been shown in previous studies. However, the exact underlying mechanisms mediating these antineoplastic effects are unclear. In the present study, we assessed the antineoplastic effects of tropisetron, a 5-HT receptor antagonist, in an experimental model of lung cancer in BALB/c mouse. Lewis lung carcinoma cell line was used to induce lung cancer. Mice were divided into four groups (n = 6) as follows: tumor-bearing mice + tropisetron (5 mg/kg intraperitoneally [IP]), tumor-bearing mice + tropisetron (10 mg/kg IP), tumor-bearing mice + saline, healthy mice + tropisetron (10 mg/kg). Tumor burden, interferon-γ (IFN-γ), interleukin (IL)-4, pathological response, Ki-67, and E-cadherin were assessed using enzyme-linked immunosorbent assay, and real-time polymerase chain reaction. Comet assay was used to assess DNA toxicity. Tropisetrone-treated animals (either 5 or 10 mg/kg) showed significantly lower tumor sizes at the day 24th after tumor induction. Tropisetron received animals also showed significantly higher levels of IFN-γ, E-cadherin, pathologic response, and necrotic cells compared to the saline-treated counterparts. In addition, the levels of IL-4, and Ki-67 were significantly lower in tropisetrone treated mice in comparison with control. Furthermore, tropisteron coadministration signifcantly reduced H2 O2 -induced DNA toxicity while treatment with tropisteron alone showed no adverse effect on DNA. Tropisetrone can be used as a potential antineoplastic drug in lung cancer. This agent can promote its antineoplastic effects in part through modulating inflammatory and proliferating markers.
Collapse
Affiliation(s)
- Mohsen Rashidi
- Department of Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Molecular and Cell Biology Research Center, Faculty Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ali Bazi
- Department of Hematology, Faculty of Allied Medical Sciences, Zabol University of Medical Sciences, Zabol, Iran
| | - Mohammad R Shiran
- Department of Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Abouzar Bagheri
- Molecular and Cell Biology Research Center, Faculty Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Department of Clinical Biochemistry-Biophysics and Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Abbas R Mehrabadi
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Hadi Kalantar
- Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Zahra Ghafouri
- Department of Clinical Biochemistry-Biophysics and Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Sayed M Hosseini
- Human Genetic Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
23
|
Modulatory effect of zingerone against cisplatin or γ-irradiation induced hepatotoxicity by molecular targeting regulation. Appl Radiat Isot 2019; 154:108891. [PMID: 31536909 DOI: 10.1016/j.apradiso.2019.108891] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 08/16/2019] [Accepted: 09/09/2019] [Indexed: 12/19/2022]
Abstract
Zingerone (ZO) is an ingredient of ginger (Zingiber officinale) which has different pharmacological properties. The objective of this research was to evaluate the protective effect of ZO against Cisplatin (Cis) or γ-Irradiation (IR)-induced hepatotoxicity in rats. ZO was given orally for consecutive 14 days prior to the treatment with Cis or exposure to IR at 15th day. Animals were sacrificed at the 23rd day. Cis or IR induced a marked increase in MAPK signal transduction as evidenced by increased p38 MAPK, JNK and ErK1/2. CYP2E1 and NADPH oxidase were significantly up-regulated. Inflammatory markers (TLR4, iNOS, COX-2 and MPO) and liver enzymes (AST, ALT and ALP) activities were also increased. Administration of ZO significantly ameliorated the above mentioned parameters.
Collapse
|
24
|
Kandemir FM, Yildirim S, Caglayan C, Kucukler S, Eser G. Protective effects of zingerone on cisplatin-induced nephrotoxicity in female rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:22562-22574. [PMID: 31165450 DOI: 10.1007/s11356-019-05505-3] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 05/16/2019] [Indexed: 06/09/2023]
Abstract
Zingerone (ZO), one of the active components of ginger (Zingiber officinale), is a phenolic alkanone with antioxidant, antiapoptotic, and anti-inflammatory properties. Cisplatin (CP) is a widely used chemotherapeutic drug for solid tumors, but its therapeutic use is limited due to dose-dependent nephrotoxicity. In the present study, we investigated the ameliorative effect of ZO against CP-induced nephrotoxicity. Intraperitoneal administration of single-dose CP (7 mg/kg body weight) on the first day enhanced kidney lipid peroxidation and reduced antioxidant enzyme activities such as catalase (CAT), superoxide dismutase (SOD), glutathione peroxidase (GPx), and glutathione (GSH). CP increased serum urea and creatinine levels and disrupted histological integrity while causing a decrease aquaporin 1 (AQP1) level in the kidney tissues. CP induced inflammatory responses by elevating the levels of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interleukin-6 (IL-6), interleukin-33 (IL-33) and nuclear factor kappa B (NF-κB), and activities of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2). Moreover, it also caused oxidative DNA damage and activation of apoptotic pathway by increasing of 8-hydroxy-2'-deoxyguanosine (8-OHdG), p53, cysteine aspartate-specific protease-3 (caspase-3), and Bcl-2-associated x protein (bax) while decreasing B cell lymphoma-2 (Bcl-2). However, treatment with ZO at a dose of 25 and 50 mg/kg b.wt. for 7 days significantly decreased oxidative stress, apoptosis, inflammation, and histopathological alterations while increased AQP1 levels in the kidney tissue. The results of the current study suggested that ZO as an effective natural product attenuates CP-induced nephrotoxicity.
Collapse
Affiliation(s)
- Fatih Mehmet Kandemir
- Department of Biochemistry, Faculty of Veterinary Medicine, Ataturk University, Erzurum, Turkey
| | - Serkan Yildirim
- Department of Pathology, Faculty of Veterinary Medicine, Ataturk University, Erzurum, Turkey
| | - Cuneyt Caglayan
- Department of Biochemistry, Faculty of Veterinary Medicine, Bingol University, 12000, Bingol, Turkey.
| | - Sefa Kucukler
- Department of Biochemistry, Faculty of Veterinary Medicine, Ataturk University, Erzurum, Turkey
| | - Gizem Eser
- Department of Pathology, Faculty of Veterinary Medicine, Ataturk University, Erzurum, Turkey
| |
Collapse
|
25
|
Acute and 28-days repeated dose sub-acute toxicity study of gallic acid in albino mice. Regul Toxicol Pharmacol 2018; 101:71-78. [PMID: 30465803 DOI: 10.1016/j.yrtph.2018.11.010] [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: 04/12/2018] [Revised: 10/13/2018] [Accepted: 11/15/2018] [Indexed: 12/15/2022]
Abstract
Gallic acid is a phenolic acid ubiquitously present in numerous medicinal plants and food beverages. Gallic acid is also a potent anti-oxidant phytochemical possessing numerous medicinal potentials against various ailments such as diabetes, hypercholesterolemia and other life-threatening diseases including malignancy. Present study was aimed to evaluate acute and sub-acute toxicity of gallic acid in albino mice. The primary aim of the study was to investigate gallic acid prompted PPAR-α/γ activation associated adverse events. Acute toxicity of gallic acid was determined in albino mice and 28-days sub-acute toxicity study was carried out in male and female albino mice at three dose levels, 100, 300 and 900 mg/kg/day, p.o. LD50 of gallic acid was found to be greater than 2000 mg/kg in mice. Hematological investigation did not show any alteration in transaminases and other blood homeostasis parameters. Gross necropsy showed non-significant alteration upon gallic acid administration. Histopathological finding suggested no significant alteration in tissue histology with slight fatty cells in bone marrow indicating non-significant bone marrow suppression, also no obvious effect was observed on hematological parameters. High dose of gallic acid (900 mg/kg/day) for 28 days did not produce any significant alteration in morphological and behavioral parameters. Histopathological finding also supports safety of gallic acid in mice.
Collapse
|
26
|
Protective Effect of Sika Deer (Cervus nippon) Velvet Antler Extract against Cisplatin-Induced Kidney and Liver Injury in a Prostate Cancer PC-3 Cell Xenograft Model. J CHEM-NY 2018. [DOI: 10.1155/2018/6705156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We previously discovered the antioxidant and antiprostate cancer effects of antler extract (AE), but whether it inhibits cisplatin- (Cis-) induced toxicity has not been investigated. In this study, the effect of AE on Cis-induced side effects in the kidney and liver using 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide-based cytotoxicity and cell cycle assays in prostate cancer PC-3 cells in vitro is investigated. Furthermore, we used a xenograft mouse model of the same cells to examine the in vivo effects and mechanisms of action. Cis and Cis + AE treatment attenuated prostate cancer cell growth by inducing apoptosis in vitro. Cis + AE stimulated cleaved caspases 3, 7, and 9 and polyadenosine diphosphate ribose polymerase expression. Cis + AE treatment for 1 week significantly increased the superoxide dismutase and catalase antioxidant activity while thiobarbituric acid reactive substances decreased. The histopathological damage and tumor necrosis factor-α, interleukin- (IL-) 1β and IL-6, cyclooxygenase-2, and inducible nitric oxide synthase expression in the kidney and liver tissue decreased. Therefore, AE likely possesses antiprostate cancer activity and inhibits Cis toxicity.
Collapse
|
27
|
Inhibition of α2C-adrenoceptors ameliorates cisplatin-induced acute renal failure in rats. Eur J Pharmacol 2018; 838:113-119. [PMID: 30201375 DOI: 10.1016/j.ejphar.2018.09.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 09/05/2018] [Accepted: 09/05/2018] [Indexed: 11/23/2022]
Abstract
Nephrotoxicity is a major adverse reaction of the anticancer drug, cisplatin. We investigated the renoprotective effects of the α2-adrenoceptor antagonist, yohimbine and selective α2C-adrenoceptor antagonist, JP-1302, in cisplatin-treated Sprague Dawley rats. Rats were given a single intravenous dose of 7.5 mg/kg cisplatin and then yohimbine or JP-1302 was administered intraperitoneally at 0.1 or 3 mg/kg/day, respectively, for four days. Renal functional parameters, such as blood urea nitrogen, plasma creatinine, creatinine clearance and renal venous norepinephrine concentrations were measured. Kidney tissue damage and tumour necrosis factor-α (TNF-α) and monocyte chemoattractant protein-1 (MCP-1) mRNA levels were assessed after the animals were euthanized. Cisplatin treatment aggravated the kidney functional parameters of blood urea nitrogen, plasma creatinine and creatinine clearance. Renal venous norepinephrine concentrations were also elevated after cisplatin administration. Treatment with yohimbine or JP-1302 clearly ameliorated kidney function and cell apoptosis. These treatments suppressed elevated renal plasma norepinephrine, TNF-α, MCP-1 and cleaved caspase 3 expressions which occurred after administration of cisplatin. These results suggest that yohimbine can prevent cisplatin-induced renal toxicity associated with acute kidney injury by suppressing cytokine expression through α2C-adrenoceptors.
Collapse
|
28
|
Sun L, Liu J, Yuan Y, Zhang X, Dong Z. Protective effect of the BET protein inhibitor JQ1 in cisplatin-induced nephrotoxicity. Am J Physiol Renal Physiol 2018; 315:F469-F478. [PMID: 29767555 PMCID: PMC6172575 DOI: 10.1152/ajprenal.00527.2017] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 04/27/2018] [Accepted: 04/27/2018] [Indexed: 11/22/2022] Open
Abstract
As a potent chemotherapy drug, cisplatin is also notorious for its side-effects including nephrotoxicity in kidneys, presenting a pressing need to identify renoprotective agents. Cisplatin nephrotoxicity involves epigenetic regulations, including changes in histone acetylation. Bromodomain and extraterminal (BET) proteins are "readers" of the epigenetic code of histone acetylation. Here, we investigated the potential renoprotective effects of JQ1, a small molecule inhibitor of BET proteins. We show that JQ1 significantly ameliorated cisplatin-induced nephrotoxicity in mice as indicated by the measurements of kidney function, histopathology, and renal tubular apoptosis. JQ1 also partially prevented the body weight loss during cisplatin treatment in mice. Consistently, JQ1 inhibited cisplatin-induced apoptosis in renal proximal tubular cells. Mechanistically, JQ1 suppressed cisplatin-induced phosphorylation or activation of p53 and Chk2, key events in DNA damage response. JQ1 also attenuated cisplatin-induced MAP kinase (p38, ERK1/2, and JNK) activation. In addition, JQ1 enhanced the expression of antioxidant genes including nuclear factor erythroid 2-related factor 2 and heme oxygenase-1, while diminishing the expression of the nitrosative protein inducible nitric oxide synthase. JQ1 did not suppress cisplatin-induced apoptosis in A549 nonsmall cell lung cancer cells and AGS gastric cancer cells. These results suggest that JQ1 may protect against cisplatin nephrotoxicity by suppressing DNA damage response, p53, MAP kinases, and oxidative/nitrosative stress pathways.
Collapse
Affiliation(s)
- Liping Sun
- Key Renal Laboratory of Shenzhen, Department of Nephrology, The Second Clinical Medical College of Jinan University , Shenzhen , China
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood Veterans Affairs Medical Center , Augusta, Georgia
| | - Jing Liu
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood Veterans Affairs Medical Center , Augusta, Georgia
- Department of Nephrology, The Second Xiangya Hospital, Central South University , Changsha, Hunan , China
| | - Yanggang Yuan
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood Veterans Affairs Medical Center , Augusta, Georgia
- Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province People's Hospital , Nanjing , China
| | - Xinzhou Zhang
- Key Renal Laboratory of Shenzhen, Department of Nephrology, The Second Clinical Medical College of Jinan University , Shenzhen , China
| | - Zheng Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood Veterans Affairs Medical Center , Augusta, Georgia
- Department of Nephrology, The Second Xiangya Hospital, Central South University , Changsha, Hunan , China
| |
Collapse
|
29
|
Mercantepe F, Mercantepe T, Topcu A, Yılmaz A, Tumkaya L. Protective effects of amifostine, curcumin, and melatonin against cisplatin-induced acute kidney injury. Naunyn Schmiedebergs Arch Pharmacol 2018; 391:915-931. [DOI: 10.1007/s00210-018-1514-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 05/13/2018] [Indexed: 12/21/2022]
|
30
|
Li YZ, Ren S, Yan XT, Li HP, Li W, Zheng B, Wang Z, Liu YY. Improvement of Cisplatin-induced renal dysfunction by Schisandra chinensis stems via anti-inflammation and anti-apoptosis effects. JOURNAL OF ETHNOPHARMACOLOGY 2018; 217:228-237. [PMID: 29421595 DOI: 10.1016/j.jep.2018.01.033] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 01/24/2018] [Accepted: 01/27/2018] [Indexed: 06/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Schisandra chinensis (Turcz.) Baill is a frequently used traditional Chinese medicine, and modern pharmacological research has proven that S. chinensis has antioxidant, anti-hepatotoxity, anti-inflammatory, and anti-nephrotoxic effects. Cisplatin is widely used as antineoplastic drug at present, but the clinical application is limited owing to its nephrotoxicity. AIM OF THE STUDY To demonstrate the renoprotective activity of the extract of the stems of S. chinensis (SCE) in mice established by cisplatin-triggering acute kidney injury (AKI). The possible molecular mechanism of nephroprotection exhibited by SCE was evaluated for the first time. MATERIALS AND METHODS Mice in SCE groups were pre-treated with SCE for 10 consecutive days, and on 7th day 1 h after final administration, following intraperitoneal injection of cisplatin with 20 mg/kg was treated to cisplatin group and SCE groups. On the 10th day, renal function, histopathological change, and oxidative stress markers were investigated. RESULTS Renal oxidative stress level characterized by elevated heme oxygenase 1 (HO-1), cytochrome P450 E1 (CYP2E1) and 4-hydroxynonenal (4-HNE) expression was obviously reduced by SCE pre-treatment. In addition, SCE was found to suppress inflammatory response through the reduction of nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2) expression and nuclear factor-kappa B (NF-κB) p65 activation. SCE treatment also inhibited activation of apoptotic pathways through down-regulating Bax, cleaved caspase-3, 8, 9 and up-regulating Bcl-2 expression levels. CONCLUSION These findings illustrated that SCE possessed powerful protective effect on AKI caused by cisplatin via amelioration of oxidative stress, inflammation and apoptosis.
Collapse
Affiliation(s)
- Yan-Zi Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Shen Ren
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Xiao-Tong Yan
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Hui-Ping Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Wei Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Bing Zheng
- School of Business Administration, Zhejiang Gongshang University, Hangzhou 310018, China
| | - Zi Wang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China.
| | - Ying-Ying Liu
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun 130033, China.
| |
Collapse
|
31
|
Elkhoely A, Kamel R. Diallyl sulfide alleviates cisplatin-induced nephrotoxicity in rats via suppressing NF-κB downstream inflammatory proteins and p53/Puma signalling pathway. Clin Exp Pharmacol Physiol 2018; 45:591-601. [PMID: 29266336 DOI: 10.1111/1440-1681.12910] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 12/11/2017] [Accepted: 12/12/2017] [Indexed: 12/20/2022]
Abstract
Despite being a potent anticancer drug, nephrotoxicity is an adverse effect which renders the clinical use of cisplatin (Cis) limited. The protective role of diallyl sulfide (DAS); a naturally occurring organo-sulfide, present in garlic, in cisplatin-induced nephrotoxicity has been reported earlier. However, the mechanism through which DAS exerts its nephroprotective activity remains elusive. The aim of the current study was to elucidate the possible mechanisms underlying the reno-protective effect of DAS in cisplatin-induced nephrotoxicity in rats. DAS was given at 2 dose levels; 50 and 100 mg/kg, orally for 4 consecutive days, starting 1 hour after administration of single dose of cisplatin (3.5 mg/kg, intraperitoneally [i.p.]). The Cis-induced elevation in serum urea and creatinine, degree of histopathological alterations was significantly ameliorated in cisplatin groups co-treated with DAS. In addition, DAS significantly restored Cis-depleted glutathione (GSH) content and superoxide dismutase (SOD) activity and attenuated Cis-elevated Malondialdehyde (MDA) level. Also, DAS significantly reduced Cis-increased renal expression of nuclear factor kappa B (NF-κB) and subsequent pro-inflammatory mediators; tumour necrosis factor alpha (TNF-α), interleukin-1β (IL-1β), intercellular adhesion molecule-1 (ICAM-1) and inducible nitric oxide synthase (iNOS) in kidney tissues. Moreover, co-treatment with DAS significantly inhibited Cis-increased caspase-8 and -9 levels. Additionally, DAS significantly mitigated Cis-induced protein expression of p53, Puma, and Bax while, it significantly restored Cis-reduced protein expression of Bcl-xL compared to the Cis group. In conclusion, these results demonstrate that DAS ameliorates cisplatin-induced nephrotoxicity in rats through enhancement of antioxidant defense, reduction of inflammatory cytokine tissue levels as well as inhibition of apoptosis via p53/Puma signalling pathway.
Collapse
Affiliation(s)
- Abeer Elkhoely
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Helwan University, Helwan, Egypt
| | - Rehab Kamel
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Helwan University, Helwan, Egypt
| |
Collapse
|
32
|
Mercantepe T, Unal D, Tümkaya L, Yazici ZA. Protective effects of amifostine, curcumin and caffeic acid phenethyl ester against cisplatin-induced testis tissue damage in rats. Exp Ther Med 2018; 15:3404-3412. [PMID: 29545862 PMCID: PMC5840930 DOI: 10.3892/etm.2018.5819] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Accepted: 01/24/2018] [Indexed: 02/02/2023] Open
Abstract
Cisplatin is an effective antineoplastic drug that is usually used to treat a number of different types of cancer in the clinic. One of the most notable side effects of cisplatin use is infertility. The present study was designed to determine the non-oxidative testicular effects caused by the use of cisplatin in rats. The rats were randomly allocated to the experimental groups. The untreated rats represented the control group (group I) and the treatment groups were as follows: cisplatin alone (group II), cisplatin+amifostine (group III), cisplatin+curcumin (group IV), and cisplatin+caffeic acid phenethyl ester (CAPE; group V). The present study observed that following cisplatin administration, the expression of nuclear factor-κB (NF-κβ)/p65, caspase-3 and 8-deoxyguanosine (8-OHdG) increased in germinal epithelium and Leydig cells. However, the expression of these markers decreased in groups III–V, most notably in the group treated with amifostine. cisplatin induced-damage was countered by amifostine and curcumin. The results revealed that the activation of NF-κB, caspase-3 and 8-OHdG had a significant role in cisplatin-induced testicular toxicity. Thus, amifostine, curcumin and, to a lesser extent, CAPE have the potential for use as therapeutic adjuvants in cisplatin-induced testis injury.
Collapse
Affiliation(s)
- Tolga Mercantepe
- Department of Histology and Embryology, Faculty of Medicine, Recep Tayyip Erdoğan University, Rize 53010, Turkey
| | - Deniz Unal
- Department of Histology and Embryology, Faculty of Medicine, Atatürk University, Erzurum 25000, Turkey
| | - Levent Tümkaya
- Department of Histology and Embryology, Faculty of Medicine, Recep Tayyip Erdoğan University, Rize 53010, Turkey
| | - Zihni Acar Yazici
- Department of Medical Microbiology, Faculty of Medicine, Recep Tayyip Erdoğan University, Rize 53010, Turkey
| |
Collapse
|
33
|
Tomar A, Vasisth S, Khan SI, Malik S, Nag TC, Arya DS, Bhatia J. Galangin ameliorates cisplatin induced nephrotoxicity in vivo by modulation of oxidative stress, apoptosis and inflammation through interplay of MAPK signaling cascade. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2017; 34:154-161. [PMID: 28899498 DOI: 10.1016/j.phymed.2017.05.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 04/10/2017] [Accepted: 05/21/2017] [Indexed: 06/07/2023]
Abstract
BACKGROUND AND PURPOSE Cisplatin is a widely used chemotherapeutic agent but now-a-days its usage is limited in clinical chemotherapy because of its severe nephrotoxic effect on renal tissues. Galangin, a flavonoid obtained from ginger family has been demonstrated to have antioxidant, anti-apoptotic and anti-inflammatory properties. This study is aimed to investigate the possible ameliorative effect of galangin in a rodent model of cisplatin-induced nephrotoxicity. MATERIAL AND METHODS Adult male albino wistar rats were divided into six groups (n=6) viz normal, cisplatin-control, galangin (25, 50 and 100mg/kg p.o.) and per se (100mg/kg galangin, p.o.). Galangin was administrated orally to the rats for a period of 10 days. On the 7th day of the treatment, nephrotoxicity was induced in all the groups by a single dose of cisplatin (8mg/kg, i.p.) (except normal and per se group). On the 11th day, the rats were anaesthetized and blood was withdrawn via direct heart puncture for biochemical estimation. Rats were sacrificed and kidneys were isolated and preserved for evaluation of histopathological, ultra structural immunohistochemical studies and western blot analysis. RESULTS Cisplatin significantly impaired renal function and increased oxidative stress and inflammation. It also increased expression of pro-apoptotic proteins Bax and caspase-3 and decreased the expression of the anti-apoptotic protein Bcl-2. Histological and ultrastructural findings were also supportive of renal tubular damage. Pretreatment with galangin (100mg/kg p.o.) preserved renal function, morphology, suppressed oxidative stress, inflammation and the activation of apoptotic pathways. TUNEL assay showed decreased DNA fragmentation on galangin pre-treatment. Furthermore, galangin (100mg/kg) pre-treatment also reduced the expression of NFκB along with proteins MAPK pathway i.e. p38, JNK and ERK1/2. CONCLUSION In conclusion, Galangin (100mg/kg, p.o.) significantly ameliorated cisplatin induced nephrotoxicity by suppressing MAPK induced inflammation and apoptosis.
Collapse
Affiliation(s)
- Ameesha Tomar
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Swati Vasisth
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Sana Irfan Khan
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Salma Malik
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Tapas Chandra Nag
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Dharamveer Singh Arya
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Jagriti Bhatia
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi 110029, India.
| |
Collapse
|
34
|
Repeated dose 28-day oral toxicity study of DEAE-Dextran in mice: An advancement in safety chemotherapeutics. Regul Toxicol Pharmacol 2017; 88:262-272. [DOI: 10.1016/j.yrtph.2017.07.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 06/03/2017] [Accepted: 07/06/2017] [Indexed: 12/17/2022]
|
35
|
Nephroprotective Effects of Anthocyanin from the Fruits of Panax ginseng
(GFA) on Cisplatin-Induced Acute Kidney Injury in Mice. Phytother Res 2017; 31:1400-1409. [DOI: 10.1002/ptr.5867] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 06/22/2017] [Accepted: 06/22/2017] [Indexed: 12/31/2022]
|
36
|
Protective effects of tropisetron on cerulein-induced acute pancreatitis in mice. Biomed Pharmacother 2017; 93:589-595. [PMID: 28686973 DOI: 10.1016/j.biopha.2017.06.067] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 06/17/2017] [Accepted: 06/19/2017] [Indexed: 12/12/2022] Open
Abstract
Acute pancreatitis (AP) causes morbidity and mortality. The aim of the present study was to investigate the protective effect of tropisetron against AP induced by cerulein. Cerulein (50μg/kg, 5 doses) was used to induce AP in mice. Six hours after final cerulein injection, animals were decapitated. Hepatic/pancreatic enzymes in the serum, pancreatic content of malondialdehyde (MDA), pro-inflammatory cytokines and myeloperoxidase (MPO) activity were measured. Tropisetron significantly attenuated pancreatic injury markers and decreased the amount of elevated serum amylase, lipase, alanine aminotransferase (ALT), aspartate aminotransferase (AST), MPO activities and pro-inflammatory cytokines levels caused by AP in mice. Tropisetron didn't affect the pancreatic levels of MDA. Our results suggest that tropisetron could attenuate cerulein-induced AP by combating inflammatory signaling. Further clinical studies are needed to confirm its efficacy in patients with AP.
Collapse
|
37
|
Aminzadeh A. Protective effect of tropisetron on high glucose induced apoptosis and oxidative stress in PC12 cells: roles of JNK, P38 MAPKs, and mitochondria pathway. Metab Brain Dis 2017; 32:819-826. [PMID: 28243846 DOI: 10.1007/s11011-017-9976-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 02/15/2017] [Indexed: 12/16/2022]
Abstract
Tropisetron, a selective 5-HT3 receptor (5-HT3R) antagonist, is widely used to counteract chemotherapy-induced emesis. There is growing interest concerning the beneficial effects of tropisetron on the treatment of several diseases. This study was carried out to examine effects of tropisetron on high glucose (HG) induced apoptosis in PC12 cells as a suitable culture model for studying neuronal functions. Apoptosis was induced by HG, and cells were treated with HG in the absence and presence of tropisetron for varying periods of time. The viability of PC12 cells was measured by MTT assay. The ROS (reactive oxygen species) production, lipid peroxidation (LPO) levels and total antioxidant power (TAP) were measured. The expressions of proapoptotic Bax, antiapoptotic Bcl-2, caspase-3, total and phosphorylated JNK and P38 MAPKs were also examined by western blotting. The results indicated that pretreatment with tropisetron significantly improved the viability of the cells and protected PC12 cells against HG induced apoptotic cell death. It could increase the concentrations of TAP. HG induced ROS generation, Bax expression and caspase 3 activation, were prevented by tropisetron. HG also induced activation of JNK and P38 MAPKs. The phosphorylation of these kinases was inhibited by tropisetron. It may be concluded that tropisetron treatment protects PC12 cells against HG-induced apoptosis by preventing JNK, P38 activation and mitochondrial pathway.
Collapse
Affiliation(s)
- Azadeh Aminzadeh
- Department of Pharmacology and Toxicology, School of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran.
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
38
|
Mansour HH, El kiki SM, Galal SM. Metformin and low dose radiation modulates cisplatin-induced oxidative injury in rat via PPAR-γ and MAPK pathways. Arch Biochem Biophys 2017; 616:13-19. [DOI: 10.1016/j.abb.2017.01.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 01/11/2017] [Accepted: 01/12/2017] [Indexed: 11/28/2022]
|
39
|
Paeonol alleviates epirubicin-induced renal injury in mice by regulating Nrf2 and NF-κB pathways. Eur J Pharmacol 2017; 795:84-93. [DOI: 10.1016/j.ejphar.2016.12.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 11/29/2016] [Accepted: 12/05/2016] [Indexed: 01/04/2023]
|
40
|
Tropisetron Protects Against Acetaminophen-Induced Liver Injury via Suppressing Hepatic Oxidative Stress and Modulating the Activation of JNK/ERK MAPK Pathways. BIOMED RESEARCH INTERNATIONAL 2016; 2016:1952947. [PMID: 27891510 PMCID: PMC5116490 DOI: 10.1155/2016/1952947] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 09/09/2016] [Accepted: 10/03/2016] [Indexed: 12/26/2022]
Abstract
Objectives. To investigate the protective effects of tropisetron on acetaminophen- (APAP-) induced liver injury in a mice model. Methods. C57BL/6 male mice were given tropisetron (0.3 to 10 mg/kg) 30 minutes before a hepatotoxic dose of acetaminophen (300 mg/kg) intraperitoneally. Twenty hours after APAP intoxication, sera alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels, hepatic myeloperoxidase (MPO), malondialdehyde (MDA), glutathione (GSH), and superoxide dismutase (SOD) activities, and liver histopathological changes were examined. The MAP kinases were also detected by western blotting. Results. Our results showed that tropisetron pretreatment significantly attenuated the acute elevations of the liver enzyme ALT level, hepatic MPO activity, and hepatocytes necrosis in a dose-dependent manner (0.3-10 mg/kg) in APAP-induced hepatotoxicity mice. Tropisetron (1 and 3 mg/kg) suppressed APAP-induced hepatic lipid peroxidation expression and alleviated GSH and SOD depletion. Administration of tropisetron also attenuated the phosphorylation of c-Jun-NH2-terminal protein kinase (JNK) and extracellular signal-regulated kinase (ERK) caused by APAP. Conclusion. Our data demonstrated that tropisetron's hepatoprotective effect was in part correlated with the antioxidant, which were mediated via JNK and ERK pathways on acetaminophen-induced liver injury in mice.
Collapse
|
41
|
Malik S, Suchal K, Bhatia J, Khan SI, Vasisth S, Tomar A, Goyal S, Kumar R, Arya DS, Ojha SK. Therapeutic Potential and Molecular Mechanisms of Emblica officinalis Gaertn in Countering Nephrotoxicity in Rats Induced by the Chemotherapeutic Agent Cisplatin. Front Pharmacol 2016; 7:350. [PMID: 27752245 PMCID: PMC5045924 DOI: 10.3389/fphar.2016.00350] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 09/15/2016] [Indexed: 12/24/2022] Open
Abstract
Emblica officinalis Gaertn. belonging to family Euphorbiaceae is commonly known as Indian gooseberry or "Amla" in India. It is used as a 'rejuvenating herb' in traditional system of Indian medicine. It has been shown to possess antioxidant, anti-inflammatory and anti-apoptotic effects. Thus, on the basis of its biological effects, the present study was undertaken to evaluate the protective effect of the dried fruit extract of the E. Officinalis (EO) in cisplatin-induced nephrotoxicity in rats and also to evaluate the mechanism of its nephroprotection. The study was done on male albino Wistar rats. They were divided into six groups (n = 6) viz. control, cisplatin-control, cisplatin and EO (150, 300, and 600 mg/kg; p.o. respectively in different groups) and EO only (600 mg/kg; p.o. only). EO was administered orally to the rats for a period of 10 days and on the 7th day, a single injection of cisplatin (8 mg/kg; i.p.) was administered to the cisplatin-control and EO treatment groups. The rats were sacrificed on the 10th day. Cisplatin-control rats had deranged renal function parameters and the kidney histology confirmed the presence of acute tubular necrosis. Furthermore, there were increased oxidative stress, apoptosis and inflammation along with higher expression of MAPK pathway proteins in the rat kidney from the cisplatin-control group. Contrary to this, EO (600 mg/kg) significantly normalized renal function, bolstered antioxidant status and ameliorated histological alterations. The inflammation and apoptosis were markedly lower in comparison to cisplatin-control rats. Furthermore, EO (600 mg/kg) inhibited MAPK phosphorylation which was instrumental in preserving renal function and morphology. In conclusion, the results of our study demonstrated that EO attenuated cisplatin-induced nephrotoxicity in rats through suppression of MAPK induced inflammation and apoptosis.
Collapse
Affiliation(s)
- Salma Malik
- Cardiovascular Research Laboratory, Department of Pharmacology, All India Institute of Medical Sciences New Delhi, India
| | - Kapil Suchal
- Cardiovascular Research Laboratory, Department of Pharmacology, All India Institute of Medical Sciences New Delhi, India
| | - Jagriti Bhatia
- Cardiovascular Research Laboratory, Department of Pharmacology, All India Institute of Medical Sciences New Delhi, India
| | - Sana I Khan
- Cardiovascular Research Laboratory, Department of Pharmacology, All India Institute of Medical Sciences New Delhi, India
| | - Swati Vasisth
- Cardiovascular Research Laboratory, Department of Pharmacology, All India Institute of Medical Sciences New Delhi, India
| | - Ameesha Tomar
- Cardiovascular Research Laboratory, Department of Pharmacology, All India Institute of Medical Sciences New Delhi, India
| | - Sameer Goyal
- Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research Shirpur, India
| | - Rajeev Kumar
- Cardiovascular Research Laboratory, Department of Pharmacology, All India Institute of Medical Sciences New Delhi, India
| | - Dharamvir S Arya
- Cardiovascular Research Laboratory, Department of Pharmacology, All India Institute of Medical Sciences New Delhi, India
| | - Shreesh K Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University Al Ain, UAE
| |
Collapse
|
42
|
Rahimian R, Zirak MR, Keshavarz M, Fakhraei N, Mohammadi-Farani A, Hamdi H, Mousavizadeh K. Involvement of PPARγ in the protective action of tropisetron in an experimental model of ulcerative colitis. Immunopharmacol Immunotoxicol 2016; 38:432-440. [DOI: 10.1080/08923973.2016.1231202] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Reza Rahimian
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec City, Québec, Canada
| | - Mohammad Reza Zirak
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mojtaba Keshavarz
- Department of Pharmacology, School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nahid Fakhraei
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences Tehran, Tehran, Iran
| | - Ahmad Mohammadi-Farani
- Pharmaceutical Sciences Research Center, School of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Department of Pharmacology, Toxicology and Medical Services, School of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Hanan Hamdi
- Cellular and Molecular Research Center and Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine Iran University of Medical Sciences, Tehran, Iran
| | - Kazem Mousavizadeh
- Cellular and Molecular Research Center and Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
43
|
Haj-Mirzaian A, Amiri S, Amini-Khoei H, Rahimi-Balaei M, Kordjazy N, Olson CO, Rastegar M, Naserzadeh P, Marzban H, Dehpour AR, Hosseini MJ, Samiei E, Mehr SE. Attenuation of oxidative and nitrosative stress in cortical area associates with antidepressant-like effects of tropisetron in male mice following social isolation stress. Brain Res Bull 2016; 124:150-63. [DOI: 10.1016/j.brainresbull.2016.04.018] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 04/16/2016] [Accepted: 04/25/2016] [Indexed: 12/13/2022]
|
44
|
Yang C, Lv J, Lv T, Pan Y, Han Y, Zhao S, Wang J. Metal ion-assisted drug-loading model for novel delivery system of cisplatin solid lipid nanoparticles with improving loading efficiency and sustained release. J Microencapsul 2016; 33:292-8. [PMID: 27113257 DOI: 10.1080/02652048.2016.1176079] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 03/31/2016] [Indexed: 10/21/2022]
Abstract
Metal ion-assisted drug loading model, in which metal ion was used to modify the microstructure of lipid layer, has been developed to improve drug loading efficiency of solid lipid nanoparticles (SLNs). The microstructure and properties of metal ion-assisted cisplatin-loading SLNs were investigated by infra-red spectroscopy, fluorescence spectroscopy and zetasizer. The reactions of hydrogenated soybean lecithin with Zn(2+), Cu(2+), Mn(2+ )and Mg(2+ )have been detected; the mechanism for higher drug encapsulation efficiency (EE) has been investigated. In metal ion introduction SLNs, the compact degree of the lipid molecules was increased due to the electrostatic interaction between metal ions and phospholipid acyl and choline polarity groups, which result in increasing of drug EE. Meanwhile, these electrostatic interactions slowed the releasing rate of encapsulated drug. The study of cytotoxic activity in vitro indicated that the cell cytotoxicity of metal ions introduction SLNs depended on both cell uptake of SLNs and drug releasing from SLNs.
Collapse
Affiliation(s)
- Caiqin Yang
- a School of Pharmaceutical Sciences , Hebei Medical University , Shijiazhuang , P.R. China
| | - Jie Lv
- a School of Pharmaceutical Sciences , Hebei Medical University , Shijiazhuang , P.R. China
| | - Tao Lv
- a School of Pharmaceutical Sciences , Hebei Medical University , Shijiazhuang , P.R. China
| | - Yahui Pan
- a School of Pharmaceutical Sciences , Hebei Medical University , Shijiazhuang , P.R. China
| | - Yazhu Han
- a School of Pharmaceutical Sciences , Hebei Medical University , Shijiazhuang , P.R. China
| | - Sha Zhao
- a School of Pharmaceutical Sciences , Hebei Medical University , Shijiazhuang , P.R. China
| | - Jing Wang
- a School of Pharmaceutical Sciences , Hebei Medical University , Shijiazhuang , P.R. China
| |
Collapse
|
45
|
K V A, Madhana RM, Kasala ER, Samudrala PK, Lahkar M, Gogoi R. Morin Hydrate Mitigates Cisplatin-Induced Renal and Hepatic Injury by Impeding Oxidative/Nitrosative Stress and Inflammation in Mice. J Biochem Mol Toxicol 2016; 30:571-579. [DOI: 10.1002/jbt.21817] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 03/31/2016] [Accepted: 04/05/2016] [Indexed: 11/07/2022]
Affiliation(s)
- Athira K V
- Department of Pharmacology and Toxicology; National Institute of Pharmaceutical Education and Research; Guwahati 781 032 India
| | - Rajaram Mohanrao Madhana
- Department of Pharmacology and Toxicology; National Institute of Pharmaceutical Education and Research; Guwahati 781 032 India
| | - Eshvendar Reddy Kasala
- Department of Pharmacology and Toxicology; National Institute of Pharmaceutical Education and Research; Guwahati 781 032 India
| | - Pavan Kumar Samudrala
- Department of Pharmacology and Toxicology; National Institute of Pharmaceutical Education and Research; Guwahati 781 032 India
| | - Mangala Lahkar
- Department of Pharmacology and Toxicology; National Institute of Pharmaceutical Education and Research; Guwahati 781 032 India
- Department of Pharmacology; Gauhati Medical College; Guwahati 781 032 India
| | - Ranadeep Gogoi
- Department of Biotechnology; National Institute of Pharmaceutical Education and Research; Guwahati 781 032 India
| |
Collapse
|
46
|
Liu B, Ezeogu L, Zellmer L, Yu B, Xu N, Joshua Liao D. Protecting the normal in order to better kill the cancer. Cancer Med 2015; 4:1394-403. [PMID: 26177855 PMCID: PMC4567024 DOI: 10.1002/cam4.488] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 05/21/2015] [Accepted: 05/27/2015] [Indexed: 12/23/2022] Open
Abstract
Chemotherapy is the only option for oncologists when a cancer has widely spread to different body sites. However, almost all currently available chemotherapeutic drugs will eventually encounter resistance after their initial positive effect, mainly because cancer cells develop genetic alterations, collectively coined herein as mutations, to adapt to the therapy. Some patients may still respond to a second chemo drug, but few cases respond to a third one. Since it takes time for cancer cells to develop new mutations and then select those life-sustaining ones via clonal expansion, "run against time for mutations to emerge" should be a crucial principle for treatment of those currently incurable cancers. Since cancer cells constantly change to adapt to the therapy whereas normal cells are stable, it may be a better strategy to shift our focus from killing cancer cells per se to protecting normal cells from chemotherapeutic toxicity. This new strategy requires the development of new drugs that are nongenotoxic and can quickly, in just hours or days, kill cancer cells without leaving the still-alive cells with time to develop mutations, and that should have their toxicities confined to only one or few organs, so that specific protections can be developed and applied.
Collapse
Affiliation(s)
- Bingya Liu
- Shanghai Key Laboratory of Gastric Neoplasms, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghai, 200025, China
| | - Lewis Ezeogu
- Hormel Institute, University of MinnesotaAustin, Minnesota, 55912
| | - Lucas Zellmer
- Hormel Institute, University of MinnesotaAustin, Minnesota, 55912
| | - Baofa Yu
- Beijing Baofa Cancer Hospital, Shahe Wangzhuang Gong Ye YuanChang Pin Qu, Beijing, 102206, China
| | - Ningzhi Xu
- Laboratory of Cell and Molecular Biology, Cancer Institute, Chinese Academy of Medical ScienceBeijing, 100021, China
| | | |
Collapse
|
47
|
Ma X, Dang C, Kang H, Dai Z, Lin S, Guan H, Liu X, Wang X, Hui W. Saikosaponin-D reduces cisplatin-induced nephrotoxicity by repressing ROS-mediated activation of MAPK and NF-κB signalling pathways. Int Immunopharmacol 2015; 28:399-408. [DOI: 10.1016/j.intimp.2015.06.020] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 05/25/2015] [Accepted: 06/15/2015] [Indexed: 11/25/2022]
|
48
|
|
49
|
Sahu BD, Mahesh Kumar J, Sistla R. Baicalein, a Bioflavonoid, Prevents Cisplatin-Induced Acute Kidney Injury by Up-Regulating Antioxidant Defenses and Down-Regulating the MAPKs and NF-κB Pathways. PLoS One 2015. [PMID: 26222683 PMCID: PMC4519041 DOI: 10.1371/journal.pone.0134139] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Acute renal failure is a serious complication of the anticancer drug cisplatin. The potential role of baicalein, a naturally occurring bioflavonoid on cisplatin-induced renal injury is unknown. Here, we assessed the effect of baicalein against a murine model of cisplatin-induced acute renal failure and investigated the underlying possible mechanisms. Renal function, kidney histology, inflammation, oxidative stress, renal mitochondrial function, proteins involved in apoptosis, nuclear translocation of Nrf2 and effects on intracellular signaling pathways such as MAPKs, and NF-κB were assessed. Pretreatment with baicalein ameliorated the cisplatin-induced renal oxidative stress, apoptosis and inflammation and improved kidney injury and function. Baicalein inhibited the cisplatin-induced expression of iNOS, TNF-α, IL-6 and mononuclear cell infiltration and concealed redox-sensitive transcription factor NF-κB activation via reduced DNA-binding activity, IκBα phosphorylation and p65 nuclear translocation in kidneys. Further studies demonstrated baicalein markedly attenuated cisplatin-induced p38 MAPK, ERK1/2 and JNK phosphorylation in kidneys. Baicalein also restored the renal antioxidants and increased the amount of total and nuclear accumulation of Nrf2 and downstream target protein, HO-1 in kidneys. Moreover, baicalein preserved mitochondrial respiratory enzyme activities and inhibited cisplatin-induced apoptosis by suppressing p53 expression, Bax/Bcl-2 imbalance, cytochrome c release and activation of caspase-9, caspase-3 and PARP. Our findings suggest that baicalein ameliorates cisplatin-induced renal damage through up-regulation of antioxidant defense mechanisms and down regulation of the MAPKs and NF-κB signaling pathways.
Collapse
Affiliation(s)
- Bidya Dhar Sahu
- Medicinal Chemistry and Pharmacology Division, CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad, 500 007, India
| | - Jerald Mahesh Kumar
- Animal House Facility, CSIR-Centre for Cellular and Molecular Biology (CCMB), Hyderabad, 500 007, India
| | - Ramakrishna Sistla
- Medicinal Chemistry and Pharmacology Division, CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad, 500 007, India
| |
Collapse
|
50
|
Yin JX, Wei Z, Xu JJ, Sun ZQ. In vivo pharmacokinetic and tissue distribution investigation of sustained-release cisplatin implants in the normal esophageal submucosa of 12 beagle dogs. Cancer Chemother Pharmacol 2015; 76:525-36. [PMID: 26183605 DOI: 10.1007/s00280-015-2823-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 07/08/2015] [Indexed: 12/15/2022]
Abstract
PURPOSE The aim of this study was to clarify the pharmacokinetic, tissue distribution, hematologic, and histopathologic characteristics of sustained-release cisplatin from implants [CDDP-nanoparticle (NP) implants]. METHODS Eighteen dogs (six hybrids and twelve beagles) were divided into three groups. In Group A, the six hybrid dogs were intravenously administered 20 mg CDDP via a hind limb vein. In Groups B and C, CDDP-NP implants containing CDDP doses of 40 and 60 mg, respectively, were embedded into the esophageal submucosa of beagles via painless gastroscopy with an endoscopic booster. Graphite frameless atomic absorption spectrophotometry was used to measure total platinum in plasma and tissues at various timepoints. In addition, free platinum levels in Group B were determined using inductively coupled plasma mass spectrometry. Toxicologic evaluation was also conducted. RESULTS Pharmacokinetic results indicated that the CDDP-NP implant could achieve a smooth pharmacokinetic curve, with the plasma invalid concentration reached after almost 480 h, which is approximately ten times longer than that of standard CDDP (48 h). The peak time, peak concentration, clearance, elimination half-life, area under the curve, volume of distribution at steady state, and mean residence time of Groups B and C were 494 and 211, 0.39 and 0.42, 0.044 and 0.059, 80.11 and 87.70, 44 and 49, 38.8 and 57.9, and 12.29 and 12.39 times those of Group A, respectively (all P < 0.05). The ratio of free/total platinum concentration was 2.0-3.1% in plasma, 14.2% in liver tissue, and 14.3% in kidney tissue. Tissue distribution studies showed that the highest platinum concentrations were found in the esophagus, followed by the kidney and liver. Compared with pre-implantation (day 0), there were no significant differences in most hematological indicators in Groups B and C (P > 0.05). Furthermore, histopathologic examination of the kidneys of dogs from Group C revealed no significant kidney damage. Unlike the intravenous CDDP group (Group A), no animals in the implantation groups showed any clinical signs of toxicity. CONCLUSION CDDP-NP implants can be used to achieve a smooth pharmacokinetic curve and higher drug concentration, as well as a longer mean residence time at the implantation site, with reduced side effects compared with intravenous CDDP.
Collapse
Affiliation(s)
- Jia-Xue Yin
- Department of Gastroenterology, Jinan Military General Hospital, No 25 Shifan Road, Tianqiao District, Jinan, 250031, Shandong Province, China
| | | | | | | |
Collapse
|