1
|
Tao H, Wang C, Zou C, Zhu H, Zhang W. Unraveling the potential of neuroinflammation and autophagy in schizophrenia. Eur J Pharmacol 2025; 997:177469. [PMID: 40054715 DOI: 10.1016/j.ejphar.2025.177469] [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: 10/22/2024] [Revised: 02/03/2025] [Accepted: 03/04/2025] [Indexed: 03/17/2025]
Abstract
Schizophrenia (SCZ) is a complex and chronic psychiatric disorder that affects a significant proportion of the global population. Although the precise etiology of SCZ remains uncertain, recent studies have underscored the involvement of neuroinflammation and autophagy in its pathogenesis. Neuroinflammation, characterized by hyperactivated microglia and markedly elevated pro-inflammatory cytokines, has been observed in postmortem brain tissues of SCZ patients and is closely associated with disease severity. Autophagy, a cellular process responsible for eliminating damaged components and maintaining cellular homeostasis, is believed to play a pivotal role in neuronal health and the onset of SCZ. This review explores the roles and underlying mechanisms of neuroinflammation and autophagy in SCZ, with a particular focus on their intricate interplay. Additionally, we provide an overview of potential therapeutic strategies aimed at modulating neuroinflammation and autophagy, including nutritional interventions, anti-inflammatory drugs, antipsychotics, and plant-derived natural compounds. The review also addresses the dual effects of antipsychotics on autophagy. Our objective is to translate these insights into clinical practice, expanding the therapeutic options available to improve the overall health and well-being of individuals with SCZ.
Collapse
Affiliation(s)
- Hongxia Tao
- Mental Health Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Congyin Wang
- Department of Emergency Medicine, Chengdu Fifth People's Hospital, Chengdu, Sichuan, 611130, China
| | - Chuan Zou
- Department of General Practice, Chengdu Fifth People's Hospital, Chengdu, Sichuan, 611130, China
| | - Hongru Zhu
- Mental Health Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Wei Zhang
- Mental Health Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
2
|
Mostafa F, Mantawy EM, Said RS, Azab SS, El-Demerdash E. Captopril attenuates oxidative stress and neuroinflammation implicated in cisplatin-induced cognitive deficits in rats. Psychopharmacology (Berl) 2025; 242:563-578. [PMID: 39809925 PMCID: PMC11861019 DOI: 10.1007/s00213-024-06706-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 10/18/2024] [Indexed: 01/16/2025]
Abstract
RATIONALE One of the most debilitating drawbacks of cisplatin chemotherapy is neurotoxicity which elicits memory impairment and cognitive dysfunction (chemobrain). This is primarily triggered by oxidative stress and inflammation. Captopril, an angiotensin-converting enzyme inhibitor, has been reported as a neuroprotective agent owing to its antioxidant and anti-inflammatory effects. OBJECTIVE We examined the possible neuroprotective effect of captopril against cisplatin-induced neurological and behavioral abnormalities in rats. METHODS Chemobrain was induced in rats by cisplatin (5 mg/kg, i.p.) on the 7th and 14th days of the study while captopril was administered orally (25 mg/kg) daily for three weeks. The effects of captopril were assessed by performing behavioral tests, histological examination, and evaluation of oxidative stress and inflammatory markers. RESULTS Cisplatin caused learning/memory dysfunction assessed by passive avoidance and Y-maze tests, decline in locomotion, and rotarod motor balance loss which were further verified by neurodegeneration observed in histological examination. Also, cisplatin aggravated oxidative stress by elevating lipid peroxidation (MDA) levels and diminishing catalase activity. Moreover, cisplatin upregulated the neuroinflammatory markers (TNF, IL-6, GFAP, and NF-κB). Captopril successfully ameliorated cisplatin damage on the levels of neurobehavioral and histopathological changes. Mechanistically, captopril significantly diminished MDA production and preserved catalase antioxidant activity. Captopril also counteracted neuroinflammation through inhibiting NF-κB and its downstream proinflammatory cytokines besides repressing astrocyte activity by reducing GFAP expression. CONCLUSION Our findings revealed that captopril could abrogate cisplatin neurotoxicity via reducing oxidative stress and neuroinflammation thus enhancing cognitive and behavioral performance. This could suggest the repurposing of captopril as a neuroprotective agent, especially in hypertensive cancer patients receiving cisplatin.
Collapse
Affiliation(s)
- Fatma Mostafa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Eman M Mantawy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Riham S Said
- Department of Drug Radiation Research, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Samar S Azab
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Ebtehal El-Demerdash
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
3
|
Masoud MM, El-Laithy NA, Youness ER, Ahmed NM, Mahdy EME, Shousha WG. Effect of amifostine on apoptotic inflammatory makers in cisplatin induced brain damage in rats. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2025:jcim-2024-0250. [PMID: 39889275 DOI: 10.1515/jcim-2024-0250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 10/22/2024] [Indexed: 02/02/2025]
Abstract
OBJECTIVES To lessen the negative effects of the medication, we assessed the neuroprotective impact of amifostine nanoparticles against the neurotoxicity generated by cisplatin. METHODS 60 adult male albino Wistar rats were arranged into six groups. Group 1; received saline intraperitonealy (IP) and served as negative control. Group 2; received IP injection of silica nano-emulsion, Group 3 received cispatin for three consecutive days at the end of the study, Group 4 received amifostine intrapretonealy (IP) before cisplatin injection, Group 5 received silica nano-emulsion alone for one month, group 6 received silica nano-emulsion in combination with cisplatin for three consecutive days at the end of the study. Monocyte chemoattractant protein-1 (MCP-1) and glial fibrillary acidic protein (GFAP) were estimated by ELISA, butrylcholinesterase (BChE) by spectrophotometric method while caspase-3 as a marker of apoptosis by PCR. RESULTS The mean levels of brain GFAP, MCP-1, and caspase-3 in the cisplatin group were considerably higher than those in the control group. However, there was a drop in the average level of brain BChE activity. Additionally, the injection of (SiNPs@AMF + cisplatin) increased BChE activities while reducing GFAP, MCP-1, and caspase-3 levels, thereby reversing the negative effects of cisplatin on the brain tissue. On the other hand, the group treated with SiNPs@AMF + cisplatin showed improvement in overall brain structure and minimal pyknotic nuclei and apoptotic neurons were found. CONCLUSIONS These outcomes demonstrated amifostine's ability to lessen the histological changes brought on by cisplatin. To sum up, SiNPs@AMF may be a suitable and secure supplemental treatment agent to lessen cisplatin's toxicity in the brain and enhance the treatment's effects throughout chemotherapy.
Collapse
Affiliation(s)
- Mahmoud M Masoud
- 68787 Medical Biochemistry Department, Medical Research and Clinical Studies Institute - National Research Centre , Cairo, Egypt
| | - Nabila A El-Laithy
- 68787 Medical Biochemistry Department, Medical Research and Clinical Studies Institute - National Research Centre , Cairo, Egypt
| | - Eman R Youness
- 68787 Medical Biochemistry Department, Medical Research and Clinical Studies Institute - National Research Centre , Cairo, Egypt
| | - Nadia M Ahmed
- 68787 Medical Biochemistry Department, Medical Research and Clinical Studies Institute - National Research Centre , Cairo, Egypt
| | - Elsayed M E Mahdy
- Department of Chemistry, Faculty of Science, Helwan University, Helwan, Egypt
| | - Wafaa Gh Shousha
- Department of Chemistry, Faculty of Science, Helwan University, Helwan, Egypt
| |
Collapse
|
4
|
Cicek B, Danisman B, Bolat I, Kiliclioglu M, Kuzucu M, Suleyman H, Tsarouhas K, Tsatsakis A, Taghizadehghalehjoughi A. Effect of tangeretin on cisplatin-induced oxido-inflammatory brain damage in rats. J Cell Mol Med 2024; 28:e18565. [PMID: 39044287 PMCID: PMC11265995 DOI: 10.1111/jcmm.18565] [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: 12/08/2023] [Revised: 06/04/2024] [Accepted: 07/09/2024] [Indexed: 07/25/2024] Open
Abstract
Cisplatin (CIS) is a platinum-derived chemotherapeutic agent commonly utilized in the treatment of various malignant tumours. However, anticancer doses of the drug cause serious damage to the brain. This study aimed to determine the potential protective effects of tangeretin, which has antioxidant and anti-inflammatory properties, in cisplatin-induced neurotoxicity on BALB/c mice brains. Male BALB/c mice were randomized and separated into four groups. Tangeretin was given for 10 days by gavage. CIS was injected as a single dose of 10 mg/kg intraperitoneally (ip) on the 10th day. Brain tissues, malondialdehyde (MDA), total glutathione (tGSH), glutathione peroxidase (GPx), superoxide dismutase (SOD), catalase (CAT) and nitric oxide (NO) levels were measured to determine oxidative damage and myeloperoxidase, tumour necrosis factor-alpha (TNF-α), interleukin 1 beta (IL-1β), IL-6 and IL-10 were measured to determine inflammatory activity. In addition, 8-OHdG and caspase-3 were analysed by immunofluorescence methods. While CIS administration remarkably elevated reactive oxygen species, MDA, and NO levels in brain tissue compared to the control, tGSH, GPx, SOD and CAT levels were significantly decreased. Also, it has been detected that TNF-α, IL-1β and IL-6 obtained in CIS-treated groups increased as well as IL-10 decreased, thereby elevating the inflammatory response. In addition, 8-OHdG and caspase-3 immunoreactivity in neurons increased with CIS administration. Treatment with tangeretin ameliorated the deterioration in oxidant/antioxidant status, overpowered neuroinflammation and ameliorated neurotoxicity-induced apoptosis. This study shows that tangeretin has beneficial effects on CIS-induced neurodegeneration. Possible mechanisms underlying these beneficial effects include the antioxidant and anti-inflammatory properties of tangeretin.
Collapse
Affiliation(s)
- Betul Cicek
- Department of Physiology, Faculty of MedicineErzincan Binali Yildirim UniversityErzincanTurkey
| | - Betul Danisman
- Department of Biophysics, Faculty of MedicineAtaturk UniversityErzurumTurkey
| | - Ismail Bolat
- Department of Pathology, Faculty of VeterinaryAtatürk UniversityErzurumTurkey
| | - Metin Kiliclioglu
- Department of Pathology, Faculty of VeterinaryAtatürk UniversityErzurumTurkey
| | - Mehmet Kuzucu
- Department of Biology, Faculty of Arts and SciencesErzincan Binali Yildirim UniversityErzincanTurkey
| | - Halis Suleyman
- Department of Medical Pharmacology, Faculty of MedicineErzincan Binali Yildirim UniversityErzincanTurkey
| | - Konstantinos Tsarouhas
- Department of CardiologyUniversity General Hospital of Larissa, Terma MazourloLarissaGreece
| | - Aristidis Tsatsakis
- Department of Forensic Sciences and Toxicology, Faculty of MedicineUniversity of CreteHeraklionGreece
| | | |
Collapse
|
5
|
Mert H, Kerem Ö, Mıs L, Yıldırım S, Mert N. Effects of protocatechuic acid against cisplatin-induced neurotoxicity in rat brains: an experimental study. Int J Neurosci 2024; 134:725-734. [PMID: 36525373 DOI: 10.1080/00207454.2022.2147430] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 06/29/2022] [Accepted: 06/30/2022] [Indexed: 12/23/2022]
Abstract
Aims/Objectives: Cisplatin (CIS) is widely used in the treatment of various malignant tumors. The aim of study is to determine the potential protective effects of protocatechuic acid (PCA) on the brain in neurotoxicity induced by CIS in rats.Materials and methods: Forty rats were divided into four groups: 1-Control group: 2- PCA group: PCA was administered orally at a dose of 100 mg/kg/day for 5 weeks. 3-CIS group: 5 mg/kg/week of CIS was administered intraperiteonally 4-PCA + CIS group: The rats were given PCA orally daily for 5 weeks and CIS of 5 mg/kg/week. The brain tissues were used for histopathological examinations and for total antioxidant capacity (TAC), total oxidative state (TOS), oxidative stress index (OSI), tumornecrosis factor-alpha (T NF-α), interleukin 6 (IL-6) Interleukin 1 beta (IL-1β), acetylcholinesterase (AChE), glutamate, gamma aminobutyric acid (GABA), dopamine analyzes in ELISA. WBC, RBC, hemoglobin and hematocrit levels were measured.Results: PCA + CIS group compared to CIS group TOS, OSI, T NF-α, IL-6, IL-1β, AChE, glutamate, WBC levels decreased significantly, while TAC and GABA levels increased statistically significant. With this study, P CA corrected the deterioration in the oxidant / antioxidant status, suppressed neuro-inflammation, decreased AChE activity, partially normalized neurotransmitters, and decreased the increased WBC count. Necrosis seen in the CIS group in histopathological examinations was never seen in the PCA + CIS group.Conclusions: PCA may provide therapeutic benefit when used in conjunction with CIS.
Collapse
Affiliation(s)
- Handan Mert
- Department of Biochemistry, Faculty of Veterinary Medicine, Van Yuzuncu Yil University, Van, Turkey
| | - Özge Kerem
- Department of Biochemistry, Faculty of Veterinary Medicine, Van Yuzuncu Yil University, Van, Turkey
| | - Leyla Mıs
- Department of Physiology, Faculty of Veterinary Medicine, Van Yuzuncu Yil University, Van, Turkey
| | - Serkan Yıldırım
- Department of Pathology, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Nihat Mert
- Department of Biochemistry, Faculty of Veterinary Medicine, Van Yuzuncu Yil University, Van, Turkey
| |
Collapse
|
6
|
Üstüner E, Yıldırım E, Macun HC, Ekici H, Şahin Y, Güncüm E, Anteplioğlu T, Elifoğlu TB, Bozkaya E. Ultrasonographic and histopathological investigation of the effect of N-acetylcysteine on doxorubicin-induced ovarian and uterine toxicity in rats. J Ovarian Res 2024; 17:135. [PMID: 38943148 PMCID: PMC11214216 DOI: 10.1186/s13048-024-01459-4] [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: 04/29/2024] [Accepted: 06/17/2024] [Indexed: 07/01/2024] Open
Abstract
BACKGROUND This study aimed to investigate the mitigating effect of N-acetylcysteine (NAC) on doxorubicin (DOX)-induced ovarian and uterine toxicity in rats using laboratory tests, ultrasonographic (US) imaging, and histopathology analysis. METHODS Forty-eight rats were divided into six groups (n = 8) as follows: Group A (control) (0.5 mL saline administered intraperitoneally [IP]), Group B (a single 10 mg/kg dose of DOX administered IP on day 1), Group C (a single 10 mg/kg dose of DOX administered IP 24 h before sacrifice), Group D (100 mg/kg of NAC administered IP for 21 days), Group E ( a single 10 mg/kg dose of DOX administered IP on day 1 and 100 mg/kg of NAC administered IP for 21 days), and Group F (100 mg/kg of NAC administered IP for 21 days and a single 10 mg/kg dose of DOX administered IP 24 h before sacrifice). The ovaries were examined using B-mode US on days 1, 14, and 21, and the histopathological examinations of the ovaries and the uterus were undertaken after sacrifice on day 22. RESULTS Histomorphological analyses showed that ovarian weight decreased after DOX administration in Group B but not in Group E. US revealed a transient increase in ovarian size in Group B and E, reverting to baseline levels over time, as well as a progressive increase in peritoneal fluid in Groups B and E. Group B exhibited a significant decrease in the thickness of the endometrium and myometrium and uterine cornual length, which was not observed in Group E. Histopathological examination showed that DOX caused a decline in follicular count, especially in primordial, secondary, and Graafian follicles, and resulted in follicular atresia, predominantly in Group B. Destructive degeneration/necrosis and vascular changes were most prominently seen in the corpus luteum of Groups C and B. In NAC-treated rats (Groups E and F), although germ cell damage was present, atretic follicles and vascular changes, such as hyperemia and congestion, were reduced. The anti-müllerian hormone (AMH) level was the highest in Group F. CONCLUSIONS NAC, an antioxidant, attenuated DOX-induced gonadotoxicity in rats.
Collapse
Affiliation(s)
- Evren Üstüner
- Faculty of Medicine, Department of Radiology, Ankara University, Ankara, Turkey.
| | - Ebru Yıldırım
- Faculty of Veterinary Medicine, Department of Pharmacology and Toxicology, Kirikkale University, Kirikkale, Turkey
| | - Hasan Ceyhun Macun
- Faculty of Veterinary Medicine, Department of Obstetrics and Gynecology, Kirikkale University, Kirikkale, Turkey
| | - Hüsamettin Ekici
- Faculty of Veterinary Medicine, Department of Pharmacology and Toxicology, Kirikkale University, Kirikkale, Turkey
| | - Yaşar Şahin
- Faculty of Veterinary Medicine, Department of Pharmacology and Toxicology, Kirikkale University, Kirikkale, Turkey
| | - Enes Güncüm
- Faculty of Veterinary Medicine, Department of Pharmacology and Toxicology, Kirikkale University, Kirikkale, Turkey
| | - Tuğçe Anteplioğlu
- Faculty of Veterinary Medicine, Department of Pathology, Kirikkale University, Kirikkale, Turkey
| | - Taha Burak Elifoğlu
- Faculty of Veterinary Medicine, Department of Obstetrics and Gynecology, Kirikkale University, Kirikkale, Turkey
| | - Esra Bozkaya
- Scientific and Technological Research Application and Research Center, Kirikkale University, Kirikkale, Türkiye
| |
Collapse
|
7
|
Zaafar D, Elghazawy NH, Hassan A, Mahmoud MY, Bakr AF, Arafa RK. Unleashing new MTDL AChE and BuChE inhibitors as potential anti-AD therapeutic agents: In vitro, in vivo and in silico studies. Int J Biol Macromol 2024; 268:131740. [PMID: 38653428 DOI: 10.1016/j.ijbiomac.2024.131740] [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: 02/05/2024] [Revised: 03/31/2024] [Accepted: 04/19/2024] [Indexed: 04/25/2024]
Abstract
Alzheimer's disease (AD) is challenging due to its irreversible declining cognitive symptoms and multifactorial nature. This work tackles targeting both acetylcholinesterase (AChE) and BuChE with a multitarget-directed ligand (MTDL) through design, synthesis, and biological and in silico evaluation of a series of twenty eight new 5-substituted-2-anilino-1,3,4-oxadiazole derivatives 4a-g, 5a-g, 9a-g and 13a-g dual inhibitors of the target biomolecules. In vitro cholinesterases inhibition and selectivity assay of the synthesized derivatives showed excellent nanomolar level inhibitory activities. Compound 5a, the most potent inhibitor, elicited IC50s of 46.9 and 3.5 nM against AChE and BuChE, respectively (SI = 0.07), 5 folds better than the known dual inhibitor Rivastagmine. In vivo and ex vivo investigation showed that 5a significantly inhibited MDA levels and increased GSH contents, thus, attenuating the brain tissue oxidative stress. Additionally, 5a significantly decreased AChE and BuChE levels and inhibited self-mediated β-amyloid aggregation in brains of treated rats. Histopathological and immunohistochemical evaluation demonstrated lessened damage and decreased caspase-3 and VEGF expression levels. In silico prediction of 5a's pharmacokinetics and toxicity profiles reflected promising results. Finally, 5a demonstrated tight binding interactions with the two target biomolecules upon docking along with stable complex formation with its bio-targets throughout the 100 ns MD trajectories.
Collapse
Affiliation(s)
- Dalia Zaafar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Modern University for Technology and Information, Cairo 12974, Egypt.
| | - Nehal H Elghazawy
- Drug Design and Discovery Lab, Zewail City of Science and Technology, Cairo 12578, Egypt
| | - Afnan Hassan
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Modern University for Technology and Information, Cairo 12974, Egypt; Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Cairo 12578, Egypt.
| | - Mohamed Y Mahmoud
- Department of Toxicology and Forensic Medicine, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt.
| | - Alaa F Bakr
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| | - Reem K Arafa
- Drug Design and Discovery Lab, Zewail City of Science and Technology, Cairo 12578, Egypt; Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Cairo 12578, Egypt.
| |
Collapse
|
8
|
Oh HN, Yoo D, Park S, Lee S, Kim WK. Assessment of poly(hexamethylenebicyanoguanide-hexamethylenediamine) hydrochloride-induced developmental neurotoxicity via oxidative stress mechanism: Integrative approaches with neuronal cells and zebrafish. JOURNAL OF HAZARDOUS MATERIALS 2024; 465:133146. [PMID: 38064952 DOI: 10.1016/j.jhazmat.2023.133146] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/22/2023] [Accepted: 11/28/2023] [Indexed: 02/08/2024]
Abstract
Poly(hexamethylenebicyanoguanide-hexamethylenediamine) hydrochloride (PHMB) is a biocide with a broad spectrum of antibacterial activity. Its use as a disinfectant and preservative in consumer products results in human exposure to PHMB. Toxicity studies on PHMB mainly focus on systemic toxicity or skin irritation; however, its effects on developmental neurotoxicity (DNT) and the underlying mechanisms are poorly understood. In this study, the DNT effects of PHMB were evaluated using IMR-32 and SH-SY5Y cell lines and zebrafish. In both cell lines, PHMB concentrations ≥ 10 µM reduced neurite outgrowth, and cytotoxicity was observed at concentrations up to 40 µM. PHMB regulated expression of neurodevelopmental genes and induced reactive oxygen species (ROS) production and mitochondrial dysfunction. Treatment with N-acetylcysteine reversed the toxic effects of PHMB. Toxicity tests on zebrafish embryos showed that PHMB reduced viability and heart rate and caused irregular hatching. PHMB concentrations of 1-4 µM reduced the width of the brain and spinal cord of transgenic zebrafish and attenuated myelination processes. Furthermore, PHMB modulated expression of neurodevelopmental genes in zebrafish and induced ROS accumulation. These results suggested that PHMB exerted DNT effects in vitro and in vivo through a ROS-dependent mechanism, highlighting the risk of PHMB exposure.
Collapse
Affiliation(s)
- Ha-Na Oh
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Donggon Yoo
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea; Human and Environmental Toxicology, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Seungmin Park
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea; Human and Environmental Toxicology, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Sangwoo Lee
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea; Human and Environmental Toxicology, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Woo-Keun Kim
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea; Human and Environmental Toxicology, University of Science and Technology, Daejeon 34113, Republic of Korea.
| |
Collapse
|
9
|
Aslanlar DA, Vişneci EF, Oz M, Nurullahoglu Atalik KE. N-acetylcysteine ameliorates chemotherapy-induced impaired anxiety and depression-like behaviors by regulating inflammation, oxidative and cholinergic status, and BDNF release. Behav Brain Res 2024; 458:114740. [PMID: 37926333 DOI: 10.1016/j.bbr.2023.114740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/18/2023] [Accepted: 11/01/2023] [Indexed: 11/07/2023]
Abstract
Mood disorders caused by chemotherapy have become more important as the survival of cancer patients increases, and new studies in this field will contribute to the prevention of this disorder. For this purpose, we used methotrexate, a chemotherapeutic agent frequently preferred in oncological cases. Mtx was administered as a single dose of 100 mg/kg intraperitoneally to male Wistar albino rats. Since oxidative stress plays an important role in chemotherapy-induced emotional impairment, n-acetylcysteine (NAC), a potent antioxidant, was administered at 500 mg/kg in two doses before Mtx administration. We evaluated anxiety and depression-like behaviors 24 h after Mtx administration, as well as some oxidative and inflammatory markers in blood serum and hippocampal tissue, acetylcholinesterase activity (AChE), and brain-derived neurotrophic factor (BDNF) release in hippocampal tissue. In rats, Mtx induced anxiety and depression-like behaviors as well as abnormalities in oxidative and inflammatory markers in blood serum and hippocampal tissue, increased AChE activity in hippocampal tissue, and decreased BDNF release. NAC treatment was found to ameliorate Mtx-induced anxiety and depression-like behaviors, increase antioxidant capacity, reduce oxidative stress and inflammatory response, and regulate AChE activity and BDNF release. In conclusion, the fact that NAC treatment of Mtx was effective is important for revising the treatment strategies for individuals suffering from this disorder, and this effect is thought to be related to the antioxidant and anti-inflammatory power of NAC.
Collapse
Affiliation(s)
- Durmuş Ali Aslanlar
- Necmettin Erbakan University, Faculty of Medicine, Department of Medical Pharmacology, Konya, Turkiye
| | - Emin Fatih Vişneci
- Health Ministry of Turkish Rebublic, Konya City Hospital, Department of Emergency Medicine, Konya, Turkiye
| | - Mehmet Oz
- Aksaray University, Faculty of Medicine, Department of Physiology, Aksaray, Turkiye.
| | | |
Collapse
|
10
|
Zavala-Valencia AC, Velasco-Hidalgo L, Martínez-Avalos A, Castillejos-López M, Torres-Espíndola LM. Effect of N-Acetylcysteine on Cisplatin Toxicity: A Review of the Literature. Biologics 2024; 18:7-19. [PMID: 38250216 PMCID: PMC10799624 DOI: 10.2147/btt.s438150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 12/08/2023] [Indexed: 01/23/2024]
Abstract
N-acetylcysteine (NAC) is a membrane-permeable cysteine precursor capable of enhancing the intracellular cysteine pool, enhancing cellular glutathione (GSH) synthesis, and thus potentiating the endogenous antioxidant mechanism. Late administration of NAC after cisplatin has been shown in different in vivo studies to reduce the side effects caused by various toxicities at different levels without affecting the antitumor efficacy of platinum, improving total and enzymatic antioxidant capacity and decreasing oxidative stress markers. These characteristics provide NAC with a rationale as a potentially effective chemo protectant in cisplatin-based therapeutic cycles. NAC represents a potential candidate as a chemoprotective agent to decrease toxicities secondary to cisplatin treatment. It suggests that it could be used in clinical trials, whereby the effective dose, timing, and route should be adjusted to optimize chemoprotection. This review provides an overview of the effect of NAC on cisplatin toxicity, a drug widely used in the clinic in adults and children.
Collapse
Affiliation(s)
- Angeles Citlali Zavala-Valencia
- Laboratory of Pharmacology, National Institute of Pediatrics, Mexico City, Mexico
- Iztacala Faculty of Higher Studies, Tlalnepantla, México
| | | | | | - Manuel Castillejos-López
- Hospital Epidemiology and Infectology Unit, National Institute of Respiratory Diseases Ismael Cosío Villegas, Mexico City, Mexico
| | | |
Collapse
|
11
|
Tüfekci KK, Tatar M, Terzi F, Bakirhan EG. An investigation of the endoplasmic reticulum stress in obesity exposure in the prenatal period. J Chem Neuroanat 2023; 134:102348. [PMID: 37858742 DOI: 10.1016/j.jchemneu.2023.102348] [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: 09/18/2023] [Revised: 10/11/2023] [Accepted: 10/16/2023] [Indexed: 10/21/2023]
Abstract
OBJECTIVES Exposure to maternal obesity has been shown to make offspring more prone to cognitive and metabolic disorders later in life. Although the underlying mechanisms are unclear, the role of endoplasmic reticulum (ER) stress in the fetal programming process is remarkable. ER stress can be activated by many chronic diseases, including obesity and diabetes. Therefore, our study aimed to investigate the role of ER stress caused by maternal diet-induced obesity in the offspring hippocampus. We also evaluated the protective effect of N-acetylcysteine (NAC) against ER stress. METHODS A rat obesity model was created by providing a high-fat (60 % kcal) diet. N-acetylcysteine (NAC) was administered at a dosage of 150 mg/kg via the intragastric route. The animals were mated at the age of 12 weeks. The same diet was maintained during pregnancy and lactation. The experiment was terminated on the postnatal 28th day, and the offspring's brain tissues were examined. Immunohistochemical staining for ER stress markers was performed on sections taken from tissues after routine histological procedures. RESULTS The results revealed increased GRP78, PERK, and eIF2α immunoreactivities in the hippocampal dentate gyrus (DG) and cornu ammonis 1 (CA1) regions in the obese group offspring, while the expression of those markers in those regions normalized with NAC supplementation (p < 0.01). Statistical analysis of XBP1 immunoreactivity H-scores revealed no difference between the study groups (p > 0.05). DISCUSSION These results suggest that exposure to obesity during the prenatal period may cause increased ER stress in hippocampal neurons, which have an important role in the regulation of learning, memory and behavior, and this may contribute to decreased cognitive performance. On the other hand, NAC stands out as an effective agent that can counteract hippocampal ER stress.
Collapse
Affiliation(s)
- Kıymet Kübra Tüfekci
- Department of Histology and Embryology, Faculty of Medicine, Kastamonu University, Turkiye.
| | - Musa Tatar
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Kastamonu University, Turkiye
| | - Funda Terzi
- Department of Pathology, Faculty of Veterinary Medicine, Kastamonu University, Turkiye
| | - Elfide Gizem Bakirhan
- Department of Histology and Embryology, Faculty of Medicine, Adıyaman University, Turkiye
| |
Collapse
|
12
|
Hussain S, Alshahrani S, Siddiqui R, Khan A, Elhassan Taha MM, Ahmed RA, Jali AM, Qadri M, Khairat KHM, Ashafaq M. Cinnamon Oil Alleviates Acetaminophen-Induced Uterine Toxicity in Rats by Abrogation of Oxidative Stress, Apoptosis, and Inflammation. PLANTS (BASEL, SWITZERLAND) 2023; 12:2290. [PMID: 37375915 DOI: 10.3390/plants12122290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 05/18/2023] [Accepted: 06/01/2023] [Indexed: 06/29/2023]
Abstract
Paracetamol, or acetaminophen (APAP), is one of the first-line medications that is used for fever and pain. However, APAP can induce uterine toxicity when overused. The mode of action of APAP toxicity is due to the production of free radicals. The main goal of our study is to determine uterine toxicity from APAP overdose and the antioxidative activity of cinnamon oil (CO) in female rats. The effect of different doses of CO (50-200 mg/kg b.w.) was assessed in the uterus toxicity induced by APAP. Additionally, the imbalance in oxidative parameters, interleukins, and caspases was evaluated for the protective effects of CO. A single dose of APAP (2 g/kg b.w.) resulted in uterus toxicity, indicated by a significant increase in the level of lipid peroxidation (LPO), inflammatory interleukins cytokines (IL-1 and 6), expression of caspases 3 and 9, and a marked change in uterus tissue architecture evaluated by histopathology. Co-treatment of CO resulted in a significant amelioration of all the parameters such as LPO, interleukins IL-1β, IL-6, caspases 3 and 9 expression, and distortion of tissue architecture in a dose-dependent manner. Therefore, we can conclude that APAP-induced uterine injury due to oxidative stress can be restored by co-treatment with cinnamon oil (CO).
Collapse
Affiliation(s)
- Sohail Hussain
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Saeed Alshahrani
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Rahimullah Siddiqui
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Andleeb Khan
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | | | - Rayan A Ahmed
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Abdulmajeed M Jali
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Marwa Qadri
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Khairat H M Khairat
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Mohammad Ashafaq
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| |
Collapse
|
13
|
Hassanein EHM, Saleh FM, Ali FEM, Rashwan EK, Atwa AM, Abd El-Ghafar OAM. Neuroprotective effect of canagliflozin against cisplatin-induced cerebral cortex injury is mediated by regulation of HO-1/PPAR-γ, SIRT1/FOXO-3, JNK/AP-1, TLR4/iNOS, and Ang II/Ang 1-7 signals. Immunopharmacol Immunotoxicol 2023; 45:304-316. [PMID: 36326099 DOI: 10.1080/08923973.2022.2143371] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 10/30/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVES Canagliflozin (CAN), a sodium-glucose co-transporter 2 inhibitor, is an anti-hyperglycemic drug that has been approved to treat diabetes. This study evaluated the beneficial effects of CAN on cerebral cortex intoxication induced by cisplatin (CIS). MATERIALS AND METHODS Rats were allocated into four groups: normal control, CAN (10 mg/kg, P.O.) for 10 days, CIS (8 mg/kg, i.p.) as a single dose on the 5th day of the experiment, and CAN + CIS group. RESULTS In comparison with CIS control rats, CAN significantly mitigated CIS-induced cortical changes in rats' behavior in the open field and forced swimming assessment as well as histological structure. Biochemically, CAN administration efficiently decreased lipid peroxidation biomarkers MDA and boosted the antioxidant status via a remarkable increase in the cortical reduced glutathione (GSH) content as well as enzymatic activities of antioxidant enzymes superoxide dismutase (SOD), glutathione-S-transferase (GST), catalase (CAT), and glutathione peroxidase (GPx) mediated by up-regulation of heme oxygenase-1 (HO-1), peroxisome proliferator-activated receptors (PPARγ), and silent information regulator (SIRT1)/forkhead box-O3 (FOXO-3) signals. Additionally, pretreatment with CAN significantly decreased cortical myeloperoxidase (MPO), nitrite (NO2-), tumor necrosis factor-alpha (TNF-α), and interleukin-6 (IL-6) levels. At the same time, it elevated the IL-10 level associated with the downregulation of Jun N-terminal kinase (JNK)/activator protein 1 (AP-1), TLR4/inducible nitric oxide synthase (iNOS)/nitric oxide (NO), and Ang II/Ang 1-7 signals. CONCLUSIONS Due to the potent antioxidant and anti-inflammatory properties of CAN, our findings showed that CAN could be a good candidate for the protection against CIS-induced cortical intoxication in the patient receiving CIS.
Collapse
Affiliation(s)
- Emad H M Hassanein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt
| | - Fayez M Saleh
- Department of Medical Microbiology, Faculty of Medicine, University of Tabuk, Tabuk, Saudi Arabia
| | - Fares E M Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt
| | - Eman K Rashwan
- Department of Physiology, College of Medicine, Al-Azhar University, Assiut, Egypt
| | - Ahmed M Atwa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Omnia A M Abd El-Ghafar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Nahda University, Beni-Suef, Egypt
| |
Collapse
|
14
|
Vongsfak J, Apaijai N, Chunchai T, Pintana H, Arunsak B, Maneechote C, Singhanat K, Wu D, Liang G, Chattipakorn N, Chattipakorn SC. Acute administration of myeloid differentiation factor 2 inhibitor and N-acetyl cysteine attenuate brain damage in rats with cardiac ischemia/reperfusion injury. Arch Biochem Biophys 2023; 740:109598. [PMID: 37054769 DOI: 10.1016/j.abb.2023.109598] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 03/24/2023] [Accepted: 04/04/2023] [Indexed: 04/15/2023]
Abstract
Inflammation and oxidative stress are mechanisms which potentially underlie the brain damage that can occur after cardiac ischemic and reperfusion (I/R) injury. 2i-10 is a new anti-inflammatory agent, acting via direct inhibition of myeloid differentiation factor 2 (MD2). However, the effects of 2i-10 and the antioxidant N-acetylcysteine (NAC) on pathologic brain in cardiac I/R injury are unknown. We hypothesized that 2i-10 and NAC offer similar neuroprotection levels against dendritic spine reduction through attenuation of brain inflammation, loss of tight junction integrity, mitochondrial dysfunction, reactive gliosis, and suppression of AD protein expression in rats with cardiac I/R injury. Male rats were allocated to either sham or acute cardiac I/R group (30 min of cardiac ischemia and 120 min of reperfusion). Rats in cardiac I/R group were given one of following treatments intravenously at the onset of reperfusion: vehicle, 2i-10 (20 or 40 mg/kg), and NAC (75 or 150 mg/kg). The brain was then used to determine biochemical parameters. Cardiac I/R led to cardiac dysfunction with dendritic spine loss, loss of tight junction integrity, brain inflammation, and mitochondrial dysfunction. Treatment with 2i-10 (both doses) effectively reduced cardiac dysfunction, tau hyperphosphorylation, brain inflammation, mitochondrial dysfunction, dendritic spine loss, and improved tight junction integrity. Although both doses of NAC effectively reduced brain mitochondrial dysfunction, treatment using a high dose of NAC reduced cardiac dysfunction, brain inflammation, and dendritic spine loss. In conclusion, treatment with 2i-10 and a high dose of NAC at the onset of reperfusion alleviated brain inflammation and mitochondrial dysfunction, consequently reducing dendritic spine loss in rats with cardiac I/R injury.
Collapse
Affiliation(s)
- Jirapong Vongsfak
- Neurosurgery Unit, Department of Surgery, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nattayaporn Apaijai
- Neurosurgery Unit, Department of Surgery, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Titikorn Chunchai
- Neurosurgery Unit, Department of Surgery, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Hiranya Pintana
- Neurosurgery Unit, Department of Surgery, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Busarin Arunsak
- Neurosurgery Unit, Department of Surgery, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Chayodom Maneechote
- Neurosurgery Unit, Department of Surgery, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Kodchanan Singhanat
- Neurosurgery Unit, Department of Surgery, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Di Wu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Nipon Chattipakorn
- Neurosurgery Unit, Department of Surgery, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Siriporn C Chattipakorn
- Neurosurgery Unit, Department of Surgery, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
15
|
Mahmoud AMA, Mantawy EM, Wahdan SA, Ammar RM, El-Demerdash E. Vildagliptin restores cognitive function and mitigates hippocampal neuronal apoptosis in cisplatin-induced chemo-brain: Imperative roles of AMPK/Akt/CREB/ BDNF signaling cascades. Biomed Pharmacother 2023; 159:114238. [PMID: 36640673 DOI: 10.1016/j.biopha.2023.114238] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/31/2022] [Accepted: 01/09/2023] [Indexed: 01/13/2023] Open
Abstract
Cisplatin (CP) is a broad-spectrum antineoplastic agent used to treat many human cancers. Nonetheless, most patients receiving CP suffer from cognitive deficits, a phenomenon termed "chemo-brain". Recently, vildagliptin (Vilda), a DPP-4 inhibitor, has demonstrated promising neuroprotective properties against various neurological diseases. Therefore, the present study aims to investigate the potential neuroprotective properties of Vilda against CP-induced neurotoxicity and elucidate the underlying molecular mechanisms. Chemo-brain was induced in Sprague-Dawley rats by i.p injection of CP at a dose of 5 mg/kg once weekly for four weeks. Vilda was administered daily at a dose (10 mg/kg; P.O) for four weeks. The results revealed that Vilda restored the cognitive function impaired by CP, as assessed by the Morris water maze, Y-maze, and passive avoidance tests. Moreover, Vilda alleviated the CP-induced neurodegeneration, as shown by toluidine blue staining, besides markedly reduced amyloid plaque deposition, as evidenced by Congo red staining. Notably, Vilda boosted cholinergic neurotransmission through the downregulation of the acetylcholinesterase enzyme. In addition, the neuroprotective mechanisms of Vilda include diminishing oxidative stress by reducing MDA levels while raising GSH levels and SOD activity, repressing neuronal apoptosis as shown by elevated Bcl-2 levels together with diminished Bax and caspase-3 expressions, inhibiting neuroinflammation as shown by decreased GFAP expression, and finally boosting hippocampal neurogenesis and survival by upregulating expressions of BDNF and PCNA. These effects were mainly mediated by activating AMPK/Akt/CREB signaling cascades. In summary, Vilda can be considered a promising candidate for guarding against CP-induced chemo-brain and neurodegeneration, thus improving the quality of life of cancer patients.
Collapse
Affiliation(s)
- Abdulla M A Mahmoud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Badr University in Cairo (BUC), Cairo, Egypt
| | - Eman M Mantawy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Sara A Wahdan
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Ramy M Ammar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kafrelsheikh University, Kafr-Elsheikh, Egypt
| | - Ebtehal El-Demerdash
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
16
|
Saral S, Topçu A, Alkanat M, Mercantepe T, Şahin Z, Akyıldız K, Karataş KS, Yıldız L, Tümkaya L, Yazıcı ZA. Agomelatine attenuates cisplatin-induced cognitive impairment via modulation of BDNF/TrkB signaling in rat hippocampus. J Chem Neuroanat 2023; 130:102269. [PMID: 37001681 DOI: 10.1016/j.jchemneu.2023.102269] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 03/24/2023] [Accepted: 03/28/2023] [Indexed: 03/31/2023]
Abstract
Cisplatin is a drug used effectively in the treatment of malignant tumors. However, cisplatin has many side effects, including cognitive impairment. Agomelatine, a synthetic melatonin analogue, is an important antidepressant. Increasing evidence has shown that agomelatine may be a potential neuroprotective agent. The aim of this study was to investigate the effect of agomelatine on learning and memory functions in cisplatin-induced cognitive impairment in a rat model. Male rats were administered agomelatine and cisplatin for 4 weeks. Neurobehavioral tests were performed at the end of the 4th week. After behavioral tests, rats were euthanized and BDNF, TNF, IL-1β, MDA and GSH levels were measured in hippocampal homegenates by ELISA. In addition, nNOS and TrkB receptor activity were measured immunohistochemically. The results showed that agomelatine significantly improved cognitive functions in spatial memory tests in rats with cisplatin-induced cognitive impairment. In addition, agomelatine treatment positively affected the discrimination index (DI). On the other hand, agomelatine treatment elevated cisplatin-suppressed hippocampal BDNF levels. Agomelatine treatment reduced cisplatin-induced neuroinflammation by suppressing TNF and IL-1β levels. Similarly, agomelatine reduced oxidative stress in the hippocampus. Histological findings showed that agomelatine treatment reduced pyramidal neuron damage in hippocampal DG, CA1 and CA3. Cisplatin increased nNOS and TrkB positivity in DG, CA1 and CA3 neurons compared to control. In contrast, agomelatine treatment decreased both nNOS and TrkB positive scores. These findings indicate that agomelatine reduces cisplatin-related cognitive impairment by exerting anti-inflammatory action and possibly by the modulation of the BDNF/TrkB/nNOS pathways in the hippocampus.
Collapse
Affiliation(s)
- Sinan Saral
- Recep Tayyip Erdogan University, Faculty of Medicine, Department of Physiology, Rize, Turkey.
| | - Atilla Topçu
- Recep Tayyip Erdogan University, Faculty of Medicine, Department of Pharmacology, Rize, Turkey
| | - Mehmet Alkanat
- Giresun University, Faculty of Medicine, Department of Physiology, Giresun, Turkey
| | - Tolga Mercantepe
- Recep Tayyip Erdogan University, Faculty of Medicine, Department of Histology and Embryology, Rize, Turkey
| | - Zafer Şahin
- Karadeniz Technical University, Faculty of Medicine, Department of Physiology, Trabzon, Turkey
| | - Kerimali Akyıldız
- Recep Tayyip Erdogan University, School of Healh Care Services Vocational, Department of Medical Services and Techniques, Rize, Turkey
| | - Kader Semra Karataş
- Kutahya Health Sciences of University, Faculty of Medicine, Department of Mental Health and Diseases, Kütahya, Turkey
| | - Lamiye Yıldız
- Recep Tayyip Erdogan University, Faculty of Medicine, Department of Physiology, Rize, Turkey
| | - Levent Tümkaya
- Recep Tayyip Erdogan University, Faculty of Medicine, Department of Histology and Embryology, Rize, Turkey
| | - Zihni Açar Yazıcı
- Recep Tayyip Erdogan University, Faculty of Medicine, Department of Microbiology, Rize, Turkey
| |
Collapse
|
17
|
Kropp PA, Rogers P, Kelly SE, McWhirter R, Goff WD, Levitan IM, Miller DM, Golden A. Patient-specific variants of NFU1/NFU-1 disrupt cholinergic signaling in a model of multiple mitochondrial dysfunctions syndrome 1. Dis Model Mech 2023; 16:286662. [PMID: 36645076 PMCID: PMC9922734 DOI: 10.1242/dmm.049594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 01/05/2023] [Indexed: 01/17/2023] Open
Abstract
Neuromuscular dysfunction is a common feature of mitochondrial diseases and frequently presents as ataxia, spasticity and/or dystonia, all of which can severely impact individuals with mitochondrial diseases. Dystonia is one of the most common symptoms of multiple mitochondrial dysfunctions syndrome 1 (MMDS1), a disease associated with mutations in the causative gene (NFU1) that impair iron-sulfur cluster biogenesis. We have generated Caenorhabditis elegans strains that recreated patient-specific point variants in the C. elegans ortholog (nfu-1) that result in allele-specific dysfunction. Each of these mutants, Gly147Arg and Gly166Cys, have altered acetylcholine signaling at neuromuscular junctions, but opposite effects on activity and motility. We found that the Gly147Arg variant was hypersensitive to acetylcholine and that knockdown of acetylcholine release rescued nearly all neuromuscular phenotypes of this variant. In contrast, we found that the Gly166Cys variant caused predominantly postsynaptic acetylcholine hypersensitivity due to an unclear mechanism. These results are important for understanding the neuromuscular conditions of MMDS1 patients and potential avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Peter A Kropp
- Laboratory of Biochemistry and Genetics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA.,Biology Department, Kenyon College, Gambier, OH 43022, USA
| | - Philippa Rogers
- Laboratory of Biochemistry and Genetics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sydney E Kelly
- Laboratory of Biochemistry and Genetics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rebecca McWhirter
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37235, USA
| | - Willow D Goff
- Laboratory of Biochemistry and Genetics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA.,Biology Department, Colgate University, Hamilton, NY 13346, USA
| | - Ian M Levitan
- Laboratory of Biochemistry and Genetics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - David M Miller
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37235, USA.,Neuroscience Graduate Program, Vanderbilt University, Nashville, TN 37235, USA
| | - Andy Golden
- Laboratory of Biochemistry and Genetics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
18
|
Ghassemi-Barghi N, Ehsanfar Z, Mohammadrezakhani O, Ashari S, Ghiabi S, Bayrami Z. Mechanistic Approach for Protective Effect of ARA290, a Specific Ligand for the Erythropoietin/CD131 Heteroreceptor, against Cisplatin-Induced Nephrotoxicity, the Involvement of Apoptosis and Inflammation Pathways. Inflammation 2023; 46:342-358. [PMID: 36085231 DOI: 10.1007/s10753-022-01737-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 08/08/2022] [Accepted: 08/30/2022] [Indexed: 12/01/2022]
Abstract
ARA 290, an 11-amino acid linear nonhematopoietic peptide derived from the three-dimensional structure of helix B of the erythropoietin (EPO), interacts selectively with the innate repair receptor (IRR) that arbitrates tissue protection. The aim of this study was to investigate the protective effects of ARA290 against cisplatin-induced nephrotoxicity. For this purpose, HEK-293 and ACHN cells were treated with ARA290 (50-400 nM) and cisplatin (2.5 μM) in pretreatment condition. Then, cytotoxicity, genotoxicity, oxidative stress parameters (ROS, GPx, SOD, and MDA), and inflammatory markers (TNFα, IL6, and IL1β) were evaluated. Furthermore, apoptotic cell death was assessed via caspase-3 activity and tunnel assay. To determine the molecular mechanisms of the possible nephroprotective effects of ARA290, gene and protein expressions of TNFα, IL1β, IL6, Caspase-3, Bax, and Bcl2 were evaluated by real-time PCR and western blot assay, respectively. The findings indicated that ARA290 significantly reduced the DNA damage parameters of comet assay and the frequency of micronuclei induced by cisplatin. Besides, ARA290 improved cisplatin-induced oxidative stress by reducing MDA/ROS levels and enhancing antioxidant enzyme levels. In addition, reduced levels of pro-inflammatory cytokines indicated that cisplatin-induced renal inflammation was mitigated upon the treatment with ARA290. Besides, ARA290 ameliorates cisplatin-induced cell injury by antagonizing apoptosis. Furthermore, the molecular findings indicated that gene and protein levels of TNFα, IL1β, IL6, Caspase-3, and Bax were significantly decreased and gene and protein levels of Bcl2 significantly increased in the ARA290 plus cisplatin group compared with the cisplatin group. These findings revealed that ARA290 as a potent chemo-preventive agent exerted a protective effect on cisplatin-induced nephrotoxicity mostly through its anti-apoptotic, anti-inflammatory, and antioxidant potentials and also suggested that ARA290 might be a new therapeutic approach for patients with acute kidney injury.
Collapse
Affiliation(s)
- Nasrin Ghassemi-Barghi
- Toxicology and Diseases Group (TDG), Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| | | | - Omid Mohammadrezakhani
- Student Research Committee, Ramsar Campus, Mazandaran University of Medical Sciences, Ramsar, Iran
| | - Sorour Ashari
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shamim Ghiabi
- Department of Medical Chemistry, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Zahra Bayrami
- Toxicology and Diseases Group (TDG), Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
19
|
Mohammed AS, Al-Hassani AN, Alrawi RA, Tawfeeq RD. The protective effect of taurine, piracetam and vinpocetine on etoposide-induced inflammation and brain injury in the serum of female albino rats. Ecancermedicalscience 2023; 17:1499. [PMID: 36816786 PMCID: PMC9937074 DOI: 10.3332/ecancer.2023.1499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Indexed: 01/24/2023] Open
Abstract
Etoposide (ETP) is one of the leading antitumour agents in cancer chemotherapy. Many studies have reported on ETP-induced peripheral neuropathy; however, few reports have focused on its brain toxicity. The current research investigates the protective potential of taurine, piracetam and vinpocetine on serum biomarkers associated with inflammation and brain injury induced by ETP in a rodent model. A total of 30 female albino rats were equally divided into five groups; the 1st and 2nd groups were the control and ETP-treated groups, respectively, while the 3rd, 4th and 5th groups were ETP-treated rats cotreated with taurine, piracetam and vinpocetine, respectively. Administration of ETP reduced body weight significantly, enhanced production of serum proinflammatory cytokines including tumour necrosis factor-alpha, interleukin-1 beta (IL-1β) and IL-6 and decreased glutathione serum levels. Moreover, ETP treatment resulted in upregulation of glial fibrillary acidic protein expression and histopathological alterations in the rats' brain compared to the control group. Co-treatment with taurine, piracetam and vinpocetine counteracted ETP-induced brain injury and altered serum biomarkers levels. We concluded that co-treatment with vinpocetine could serve as a complementary therapeutic agent in reducing brain injury and toxicity induced by ETP.
Collapse
Affiliation(s)
- Arwa Salam Mohammed
- Department of Pharmacology and Toxicology, College of Pharmacy, Hawler Medical University, Erbil 44001, Iraq
| | - Ansam N Al-Hassani
- Department of Pharmacology and Toxicology, College of Pharmacy, Hawler Medical University, Erbil 44001, Iraq
| | - Rafal Abdulrazaq Alrawi
- Department of Clinical Analysis, College of Pharmacy, Hawler Medical University, Erbil 44001, Iraq
| | - Rawaz D Tawfeeq
- Department of Clinical Analysis, College of Pharmacy, Hawler Medical University, Erbil 44001, Iraq
| |
Collapse
|
20
|
Cisplatin-induced changes in calcitonin gene-related peptide or TNF-α release in rat dorsal root ganglia in vitro model of neurotoxicity are not reverted by rosiglitazone. Neurotoxicology 2022; 93:211-221. [DOI: 10.1016/j.neuro.2022.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 09/24/2022] [Accepted: 10/05/2022] [Indexed: 11/05/2022]
|
21
|
Afsar T, Razak S, Almajwal A. Reversal of cisplatin triggered neurotoxicity by Acacia hydaspica ethyl acetate fraction via regulating brain acetylcholinesterase activity, DNA damage, and pro-inflammatory cytokines in the rodent model. BMC Complement Med Ther 2022; 22:179. [PMID: 35790919 PMCID: PMC9254489 DOI: 10.1186/s12906-022-03657-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 06/24/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Cisplatin (CisPT) is a chemotherapeutic that outcome in adverse effects including neurotoxicity. We examined the efficacy of hydaspica ethyl acetate extract (AHE) against CisPT-prompted neurotoxicity.
Methods
Group I: Distilled water; Group II: CisPT (12 mg/kg b.w. i.p) on the 13th day of treatment. Group III: received AHE (400 mg/kg b.w) orally for 16 days. Group IV and V received 200 and 400 mg/kg b.w AHE orally for 16 days while CisPT injection on day 13, respectively. Group VI: received Silymarin (100 mg/kg b.w) orally for 16 days and CP (12 mg/kg b.w., i.p.) on day 13. TNF-α, IL6, brain acetylcholinesterase activity (AChE), oxidative trauma markers, genotoxicity, antioxidant enzymes, and morphological alterations in cerebral hemispheres were inspected.
Results
AHE administration before CisPT considerably reduced both tissue TNF-α and IL 6 expressions compared to CisPT treated group in a dose-dependent manner. AHE treatment (400 mg/kg b.w) significantly ameliorated brain AChE activity. Brain tissue MDA, H2O2, and NO content were markedly (p < 0.001) elevated after CisPT inoculation while a noticeable (p < 0.001) diminution was observed in AHE treatment groups. AHE treatment significantly (p < 0.001) improved brain antioxidant defense in a dose-dependent manner. Furthermore, AHE efficiently recused CisPT to induce DNA damage in brain tissue as revealed by ladder assay and DNA fragmentation patterns. Histopathological findings revealed severe neurodegenerations in CisPT treated group, however, AHE treatment noticeably precluded morphological alterations and neuron damages induced by CisPT.
Conclusion
A. hydaspica AHE extract may be provided as a prospective adjuvant that precludes CisPT-induced neurotoxicity due to its radical scavenging and antioxidant potential.
Collapse
|
22
|
Akomolafe SF, Asowata-Ayodele AM. Roasted cashew ( Anacardium occidentale L.) nut-enhanced diet forestalls cisplatin-initiated brain harm in rats. Heliyon 2022; 8:e11066. [PMID: 36276737 PMCID: PMC9578995 DOI: 10.1016/j.heliyon.2022.e11066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 03/16/2022] [Accepted: 10/07/2022] [Indexed: 11/30/2022] Open
Abstract
The incessant dose constraining symptom of the chemotherapeutic agent, cisplatin is neurotoxicity. This examination tried to explore the neuroprotective impact of roasted cashew nut-enhanced diet against brain deficits related with treatment with cisplatin. Rats were separated in to six groups: Control, CIS (cisplatin [7 mg/kg body weight, i.p]), CIS +10% CN (cisplatin plus 10% roasted cashew nut), CIS +20% CN (cisplatin plus 20% roasted cashew nut), 10% CN (10% roasted cashew nut) and 20% CN (20% roasted cashew nut) for 28 days. Key enzymes associated with brain function, including cholinesterases (AChE and BChE), monoaminergic enzyme (MAO), arginase, and adenosine deaminase (ADA), were investigated after the treatment. The following oxidative stress indicators were also measured in the rat brain: glutathione-S-transferase (GST), glutathione peroxidase (GPx), total antioxidant capacity (TAC), total thiol (T-SH), non-protein thiol (NPSH), thiobarbituric acid reactive substances (TBARS), reactive oxygen species (ROS), nitric oxide (NO), superoxide dismutase (SOD). Our outcomes demonstrated that roasted cashew nut enhanced diet showed inhibitory impact on activities of AChE, BChE, ADA, MAO and arginase in cisplatin-induced rats. The roasted cashew nut supplemented diet also boosted redox equilibrium and displayed protection against cispaltin-induced oxidative damage to rats' brains by an increase in SOD, CAT, GST and GPx activities, TAC, T-SH, NPSH and NO levels as well as a considerable drop in ROS and RBARS levels. Roasted cashew nut enhanced diet additionally forestalled neuronal degeneration in rat brain. Thus, roasted cashew nuts could be used as a nutraceutical or functional food to treat cisplatin-induced neurotoxicity. Practical applications The results show that increasing roasted cashew nut consumption can significantly improve antioxidant status, reduce lipid peroxidation, and suppress cholinesterase, adenosine deaminase, monoamine oxidase, and arginase activities in the brain under cisplatin-induced circumstances.
Collapse
Affiliation(s)
- Seun F. Akomolafe
- Department of Biochemistry, Ekiti State University, P.M.B. 5363, Ado Ekiti, Nigeria,Corresponding author.
| | | |
Collapse
|
23
|
Feng C, Li A, Yin C, Wang S, Jin W, Liu Y, Huo T, Jiang H. Realgar Alleviated Neuroinflammation Induced by High Protein and High Calorie Diet in Rats via the Microbiota-Gut-Brain Axis. Nutrients 2022; 14:nu14193958. [PMID: 36235611 PMCID: PMC9572528 DOI: 10.3390/nu14193958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/17/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Purpose: Gastrointestinal heat retention syndrome (GHRS) often occurs in adolescents, resulting into nervous system injury. Realgar, an arsenic mineral with neuroprotective effect, has been widely used to treat GHRS. However, its mechanism of action remains unknown. Methods: A GHRS rat model was established using a high protein and high calorie diet. We performed macroscopic characterization by assessing bowel sounds, hot/cold preference, anal temperature, and fecal features. Atomic fluorescence spectroscopy was employed to evaluate brain arsenic level while hippocampal ultrastructural changes were analyzed using transmission electron microscopy. In addition, inflammatory cytokines and BBB breakdown were analyzed by western blotting, immunofluorescence assays, and immunohistochemistry staining. We also evaluated hippocampal metabolites by LC-MS while fecal microorganisms were assessed by 16S rDNA sequencing. Results: Our data showed that the high protein and high calorie diet induced GHRS. The rat model depicted decreased bowel sounds, increased fecal characteristics score, preference for low temperature zone, and increased anal temperature. In addition, there was increase in inflammatory factors IL-6, Iba-1, and NF-κB p65 as well as reduced BBB structural protein Claudin-5 and Occludin. The data also showed appearance of hippocampus metabolites disorder and fecal microbial imbalance. Realgar treatment conferred a neuroprotective effect by inhibiting GHRS-specific characteristics, neuroinflammatory response, BBB impairment, metabolites disorder, and microbial imbalance in the GHRS rat model. Conclusion: Taken together, our analysis demonstrated that realgar confers a neuroprotective effect in GHRS rats through modulation of the microbiota-gut-brain axis.
Collapse
Affiliation(s)
- Cong Feng
- Department of Health Laboratory Technology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, China
- Laboratory of Research in Parkinson’s Disease and Related Disorders, Health Sciences Institute, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, China
| | - Aihong Li
- Department of Health Laboratory Technology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, China
| | - Chenhui Yin
- Department of Health Laboratory Technology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, China
| | - Siying Wang
- Department of Health Laboratory Technology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, China
| | - Weiyuan Jin
- Department of Health Laboratory Technology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, China
| | - Yi Liu
- Department of Health Laboratory Technology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, China
| | - Taoguang Huo
- Department of Health Laboratory Technology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, China
| | - Hong Jiang
- Department of Health Laboratory Technology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, China
- The Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, No. 77 Puhe Road, Shenyang North New Area, Shenyang 110122, China
- Correspondence:
| |
Collapse
|
24
|
Dhillon T, Kumar A, Kumar V. Neuroprotective Effect of N-acetylcysteine Against Monocrotophos-Induced Oxidative Stress in Different Brain Regions of Rats. Appl Biochem Biotechnol 2022; 194:4049-4065. [PMID: 35587328 DOI: 10.1007/s12010-022-03967-9] [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: 09/27/2021] [Accepted: 05/10/2022] [Indexed: 11/02/2022]
Abstract
Monocrotophos (MCP) is systemic organophosphate insecticide used against crop pests. It is reported to cause mammalian toxicity through both acute and chronic exposure. In the present study, we have shown the protective role of N-acetylcysteine (NAC) against MCP-induced oxidative stress in frontal cortex, corpus striatum and hippocampus brain regions of rats. Male Albino Wistar rats were divided into control, NAC-treated, MCP and NAC + MCP-treated groups. An oral dose of MCP (0.9 mg/kg b.wt) and NAC (200 mg/kg b.wt) was administered for 28 days. Results showed an increase in lipid peroxidation (LPO) and protein oxidation followed by decreased antioxidant enzymes after 28 days of MCP exposure. Histopathological analysis showed that monocrotophos exposure caused neurodegenerative changes as evident by neurons with dystrophic changes in the form of shrunken hyperchromatic nuclei in all the regions of the rat brain. N-acetylcysteine supplementation to MCP-treated rats showed a reduction in oxidative stress and ameliorated cellular alterations in all of the three regions. The results of the study indicate that N-acetylcysteine offers neuroprotection by improving antioxidant response and decreasing oxidative stress in different regions of the rat brain.
Collapse
Affiliation(s)
- Twinkle Dhillon
- Department of Biochemistry, Maharshi Dayanand University, Rohtak, 124001, Haryana, India
| | - Amit Kumar
- Department of Biochemistry, Maharshi Dayanand University, Rohtak, 124001, Haryana, India
| | - Vijay Kumar
- Department of Biochemistry, Maharshi Dayanand University, Rohtak, 124001, Haryana, India.
| |
Collapse
|
25
|
Wang XL, Lin FL, Xu W, Wang C, Wang QQ, Jiang RW. Silybin B exerts protective effect on cisplatin-induced neurotoxicity by alleviating DNA damage and apoptosis. JOURNAL OF ETHNOPHARMACOLOGY 2022; 288:114938. [PMID: 34999144 DOI: 10.1016/j.jep.2021.114938] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/10/2021] [Accepted: 12/21/2021] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Silybum marianum is a traditional Chinese medicine that has been used for treating liver disease. Silybin consisting of silybin A and silybin B, is a member of Silybum marianum, and exerts a therapeutic effect on many diseases. However, the protective effect of silybin on cisplatin-induced neurotoxicity and the stereoisomer contributing to the effect remain unknown. AIM OF THE STUDY The present study aimed to study the effect of silybin on cisplatin-induced neuronal injury, compare the difference of protective effect between silybin A and silybin B, and the potential mechanism. MATERIALS AND METHODS High performance liquid chromatography (HPLC) was used to separate silybin A and silybin B. X-ray crystallographic analysis in combination with experimental and calculated ECD were performed to identify the structure of silybin A and silybin B. The toxicity of the silybin or cisplatin against murine hippocampal neuronal HT22 cells was determined through MTT assay. The cell cycle and cell apoptosis were measured by PI staining and Annexin V-FITC/PI staining, respectively, and then subjected to flow cytometry. Western blot analysis was conducted to quantify the expression of proteins related to apoptosis and DNA damage. Immunofluorescence was used to evaluate the expression of DNA damage marker. In vivo experiment, the behavioral analysis was determined through pole test, swimming test and Morris water maze test. The index of superoxide dismutase (SOD), reduced glutathione (GSH), total antioxidant capacity (T-AOC) and lipid peroxidation (LPO) were examined to evaluate the antioxidant capacity in mice brain. Nissl staining and Tunel assay were used to detect the neuronal viability and apoptosis in hippocampus. RESULTS We successfully separated and identified silybin A and silybin B. We found both silybin A and silybin B alleviated cisplatin-induced apoptosis and cell cycle arrest in HT22 cells, and silybin B was more effective. We chose silybin B for further mechanism investigation, and found silybin B alleviated DNA damage by enhancing phosphorylation of ATR and decreasing expression of γ-H2AX. In the in vivo experiment, we observed that silybin B markedly improved the behavioral abnormalities in cisplatin-treated mice, reduced LPO level while increased SOD, GSH and T-AOC in mice brain tissue. Nissl staining and Tunel assay showed that silybin B alleviated cisplatin-induced hippocampal damage. CONCLUSIONS These results suggest that silybin B might serve as a promising drug candidate in mitigating cisplatin-induced neural injury in the brain and thereby improving the chemotherapeutic outcomes.
Collapse
Affiliation(s)
- Xiao-Lu Wang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, PR China.
| | - Fo-Lan Lin
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, PR China.
| | - Wei Xu
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, PR China.
| | - Chen Wang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, PR China.
| | - Qi-Qi Wang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, PR China.
| | - Ren-Wang Jiang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, PR China.
| |
Collapse
|
26
|
Bayraktar UA, Arıhan O, Atalay Ö, Gök M, Çiçek Ç, Bodur E, Tuncer M. Melatonin is effective in attenuating cisplatin-induced neurotoxicity. J Biochem Mol Toxicol 2022; 36:e23075. [PMID: 35451207 DOI: 10.1002/jbt.23075] [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] [Received: 12/17/2021] [Revised: 02/24/2022] [Accepted: 04/01/2022] [Indexed: 11/08/2022]
Abstract
Cisplatin (Cis) is a chemotherapeutic agent that has many side effects. Neurotoxicity is one of the most important of these side effects. Oxidative stress and neuroinflammation are the best-known mechanisms in the pathogenesis of neurotoxicity development. In this study, we aimed to determine whether melatonin (Mel), with antioxidant and anti-inflammatory effects, is effective in preventing Cis-induced neurotoxicity. Forty-eight male Sprague-Dawley rats were divided into six groups (n = 8) as follows: control (0.9% NaCl), vehicle (5% ethanol), Cis (6 mg/kg), Cis (6 mg/kg) + vehicle (5% ethanol), Mel (20 mg/kg), and Cis (6 mg/kg) + Mel (20 mg/kg) groups. Cis was administered as a single dose on the 3rd day of the experiment while Mel was given for 5 days. All administrations were performed via intraperitoneal injection. After injections, T-maze, rotarod, and hot plate tests were performed to evaluate cognitive, motor, and sensory functions, respectively. Following sacrification oxidative stress markers, cholinergic function, and proinflammatory cytokines were studied from brain homogenates. Cis impaired cognitive function and motor performance in the Cis and Cis+Vehicle groups. The drug also increased oxidative stress in the brain. Mel significantly improved brain oxidant/antioxidant status and also decreased the overproduction of proinflammatory cytokines (superoxide dismutase activities in Cis+Vehicle and Cis+Mel groups: 104.55 ± 9.50 µU/mg protein vs. 150.13 ± 4.70 µU/mg protein, respectively, p < 0.05; tumor necrosis factor-α levels in Cis and Cis+Mel groups: 40 pg/ml vs. 20 pg/ml, respectively, p < 0.05). It seems that Mel can improve Cis neurotoxicity. For a more firm conclusion, further studies using Mel at different doses with larger groups should be performed.
Collapse
Affiliation(s)
- U Arda Bayraktar
- Department of Physiology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Okan Arıhan
- Department of Physiology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Özbeyen Atalay
- Department of Physiology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Müslüm Gök
- Department of Biochemistry, Faculty of Medicine, Hacettepe University, Ankara, Turkey.,Department of Biochemistry, Faculty of Medicine, Muğla Sıtkı Koçman University, Muğla, Turkey
| | - Çiğdem Çiçek
- Department of Biochemistry, Faculty of Medicine, Hacettepe University, Ankara, Turkey.,Department of Biochemistry, Faculty of Medicine, Yüksek İhtisas University, Ankara, Turkey
| | - Ebru Bodur
- Department of Biochemistry, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Meltem Tuncer
- Department of Physiology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| |
Collapse
|
27
|
Elghazawy NH, Zaafar D, Hassan RR, Mahmoud MY, Bedda L, Bakr AF, Arafa RK. Discovery of New 1,3,4-Oxadiazoles with Dual Activity Targeting the Cholinergic Pathway as Effective Anti-Alzheimer Agents. ACS Chem Neurosci 2022; 13:1187-1205. [PMID: 35377601 DOI: 10.1021/acschemneuro.1c00766] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Finding an effective anti-Alzheimer agent is quite challenging due to its multifactorial nature. As such, multitarget directed ligands (MTDLs) could be a promising paradigm for finding potential therapeutically effective new small-molecule bioactive agents against Alzheimer's disease (AD). We herein present the design, synthesis, and biological evaluation of a new series of compounds based on a 5-pyrid-3-yl-1,3,4-oxadiazole scaffold. Our synthesized compounds displayed excellent in vitro enzyme inhibitory activity at nanomolar (nM) concentrations against two major AD disease-modifying targets, i.e., acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE). Among our compounds, 5e was considered the best dual inhibitor of both AChE (IC50 = 50.87 nM) and BuChE (IC50 = 4.77 nM), where these values surpassed those of rivastagmine (the only FDA-approved dual AChE and BuChE inhibitor) in our study. Furthermore, in vivo and ex vivo testing of the hit compound 5e highlighted its significant AD-biotargeting effects including reducing the elevated levels of lipid peroxidation and glutathione (GSH), normalizing levels of 8-OHdG, and, most importantly, decreasing the levels of the well-known AD hallmark β-amyloid protein. Finally, the binding ability of 5e to each of our targets, AChE and BuChE, was confirmed through additional molecular docking and molecular dynamics (MD) simulations that reflected good interactions of 5e to the active site of both targets. Hence, we herein present a series of new 1,3,4-oxadiazoles that are promising leads for the development of dual-acting AChE and BuChE inhibitors for the management of AD.
Collapse
Affiliation(s)
- Nehal H Elghazawy
- Drug Design and Discovery Lab, Zewail City of Science and Technology, Ahmed Zewail Road, October Gardens, Cairo 12578, Egypt
| | - Dalia Zaafar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Modern University for Technology and Information, Cairo 12055, Egypt
| | - Reham R Hassan
- Drug Design and Discovery Lab, Zewail City of Science and Technology, Ahmed Zewail Road, October Gardens, Cairo 12578, Egypt
| | - Mohamed Y Mahmoud
- Department of Toxicology, Forensic Medicine and Veterinary Regulations, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| | - Loay Bedda
- Drug Design and Discovery Lab, Zewail City of Science and Technology, Ahmed Zewail Road, October Gardens, Cairo 12578, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Ahmed Zewail Road, October Gardens, Cairo 12578, Egypt
| | - Alaa F Bakr
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| | - Reem K Arafa
- Drug Design and Discovery Lab, Zewail City of Science and Technology, Ahmed Zewail Road, October Gardens, Cairo 12578, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Ahmed Zewail Road, October Gardens, Cairo 12578, Egypt
| |
Collapse
|
28
|
Kim UJ, Lee KH. Neuroprotective effects of N-acetylcysteine amide against oxidative injury in an aging model of organotypic hippocampal slice cultures. Neuroreport 2022; 33:173-179. [DOI: 10.1097/wnr.0000000000001767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
29
|
Liu N, Zhou S, Olatunji OJ, Wu Y. Nucleosides rich extract from Cordyceps cicadae alleviated cisplatin-induced neurotoxicity in rats: A behavioral, biochemical and histopathological study. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2021.103476] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
30
|
Ribeiro AEAS, Ferreira EF, Leal JDS, Barberino RDS, Oliveira HPD, Palheta Junior RC. Involvement of MT2 receptors in protective effects of melatonin against cisplatin-induced gastrointestinal damage in mice. BRAZ J PHARM SCI 2022. [DOI: 10.1590/s2175-97902022e20476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
|
31
|
Melatonin Attenuates Cyclophosphamide-Induced Primordial Follicle Loss by Interaction with MT 1 Receptor and Modulation of PTEN/Akt/FOXO3a Proteins in the Mouse Ovary. Reprod Sci 2021; 29:2505-2514. [PMID: 34642909 DOI: 10.1007/s43032-021-00768-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 10/05/2021] [Indexed: 10/20/2022]
Abstract
This study evaluated the protective effect of melatonin before cyclophosphamide administration on ovarian function and its potential mechanism in a mouse model. Two studies were performed. In the first, mice were pretreated with melatonin (10, 20, or 30 mg/kg body weight, i.p.) once daily for 3 days, followed by injection with a single dose of cyclophosphamide (200 mg/kg body weight, i.p.) 30 min after the last melatonin injection. The second study analyzed whether melatonin type 1 and/or 2 receptors mediate the effects of melatonin on the ovary through administration of non-selective MT1/MT2 antagonist (luzindole) or selective MT2 antagonist (4-PPDOT) before the treatment with melatonin plus cyclophosphamide. After treatment groups, the ovaries were harvested and destined to histology, immunohistochemistry, and fluorescence analyses. Lastly, we examined the p-PTEN, p-Akt, and p-FOXO3a participation in the protective effect of melatonin in cyclophosphamide-induced ovarian damage. Results demonstrated that pretreatment with 20 mg/kg melatonin before cyclophosphamide administration showed more morphologically normal follicles, attenuated primordial follicle loss, decreased growing follicle atresia and mitochondrial damage, and increased GSH concentrations. Furthermore, treatment with luzindole blocked the protective effects of melatonin against the damage caused by cyclophosphamide. Additionally, pretreatment with 20 mg/kg melatonin regulated the PTEN/Akt/FOXO3a signaling pathway components after cyclophosphamide treatment. In conclusion, pretreatment with 20 mg/kg melatonin prevented primordial follicle loss and reduced apoptosis and oxidative damage in the mouse ovary during experimental chemotherapy with cyclophosphamide. Furthermore, the MT1 receptor and PTEN/Akt/FOXO3a proteins mediated these cytoprotective effects.
Collapse
|
32
|
Morgan AM, Hassanen EI, Ogaly HA, Al Dulmani SA, Al-Zahrani FAM, Galal MK, Kamel S, Rashad MM, Ibrahim MA, Hussien AM. The ameliorative effect of N-acetylcysteine against penconazole induced neurodegenerative and neuroinflammatory disorders in rats. J Biochem Mol Toxicol 2021; 35:e22884. [PMID: 34392569 DOI: 10.1002/jbt.22884] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/24/2021] [Accepted: 08/06/2021] [Indexed: 11/07/2022]
Abstract
Penconazole (PEN) is a widely used systemic fungicide to treat various fungal diseases in plants but it leaves residues in crops and food products causing serious environmental and health problems. N-acetylcysteine (NAC) is a precursor of the antioxidant glutathione in the body and exerts prominent antioxidant and anti-inflammatory effects. The present study aimed to explore the mechanistic way of NAC to ameliorate the PEN neurotoxicity in male rats. Twenty-eight male rats were randomly divided into four groups (n = 7) and given the treated material via oral gavage for 10 days as the following: Group I (distilled water), Group II (50 mg/kg body weight [bwt] PEN), Group III (200 mg/kg bwt NAC), and Group IV (NAC + PEN). After 10 days all rats were subjected to behavioral assessment and then euthanized to collect brain tissues to perform oxidative stress, molecular studies, and pathological examination. Our results revealed that PEN exhibits neurobehavioral toxicity manifested by alteration in the forced swim test, elevated plus maze test, and Y-maze test. There were marked elevations in malondialdehyde levels with reduction in total antioxidant capacity levels, upregulation of messenger RNA levels of bax, caspase 3, and caspase 9 genes with downregulation of bcl2 genes. In addition, brain sections showed marked histopathological alteration in the cerebrum and cerebellum with strong bax and inducible nitric oxide synthetase protein expression. On the contrary, cotreatment of rats with NAC had the ability to improve all the abovementioned neurotoxic parameters. The present study can conclude that NAC has a neuroprotective effect against PEN-induced neurotoxicity via its antioxidant, anti-inflammatory, and antiapoptotic effect. We recommend using NAC as a preventive and therapeutic agent for a wide variety of neurodegenerative and neuroinflammatory disorders.
Collapse
Affiliation(s)
- Ashraf M Morgan
- Toxicology and Forensic Medicine Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Eman I Hassanen
- Pathology Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Hanan A Ogaly
- Chemistry Department, Faculty of Science, King Khalid University, Abha, Saudi Arabia
| | - Sharah A Al Dulmani
- Chemistry Department, Faculty of Science, King Khalid University, Abha, Saudi Arabia
| | | | - Mona K Galal
- Biochemistry and Molecular Biology Department, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
| | - Shaimaa Kamel
- Biochemistry and Molecular Biology Department, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
| | - Maha M Rashad
- Biochemistry and Molecular Biology Department, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
| | - Marwa A Ibrahim
- Biochemistry and Molecular Biology Department, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
| | - Ahmed M Hussien
- Toxicology and Forensic Medicine Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
33
|
Rizor A, Pajarillo E, Nyarko-Danquah I, Digman A, Mooneyham L, Son DS, Aschner M, Lee E. Manganese-induced reactive oxygen species activate IκB kinase to upregulate YY1 and impair glutamate transporter EAAT2 function in human astrocytes in vitro. Neurotoxicology 2021; 86:94-103. [PMID: 34310962 DOI: 10.1016/j.neuro.2021.07.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/14/2021] [Accepted: 07/21/2021] [Indexed: 12/19/2022]
Abstract
Dysregulation of the astrocytic glutamate transporter excitatory amino acid transporter 2 (EAAT2) is associated with several neurological disorders, including Parkinson's disease, Alzheimer's disease, and manganism, the latter induced by chronic exposure to high levels of manganese (Mn). Mechanisms of Mn-induced neurotoxicity include impairment of EAAT2 function secondary to the activation of the transcription factor Yin Yang 1 (YY1) by nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). However, the upstream mechanisms by which Mn-induced NF-κB activates YY1 remain to be elucidated. In the present study, we used the H4 human astrocyte cell line to test if Mn activates YY1 through the canonical NF-κB signaling pathway, leading to EAAT2 repression. The results demonstrate that Mn exposure induced phosphorylation of the upstream kinase IκB kinase (IKK-β), leading to NF-κB p65 translocation, increased YY1 promoter activity, mRNA/protein levels, and consequently repressed EAAT2. Results also demonstrated that Mn-induced oxidative stress and subsequent TNF-α production were upstream of IKK-β activation, as antioxidants attenuated Mn-induced TNF-α production and IKK-β activation. Moreover, TNF-α inhibition attenuated the Mn-induced activation of IKK-β and YY1. Taken together, Mn-induced oxidative stress and TNF-α mediates activation of NF-κB signaling and YY1 upregulation, leading to repression of EAAT2. Thus, targeting reactive oxygen species (ROS), TNF-α and IKK-β may attenuate Mn-induced YY1 activation and consequent EAAT2 repression.
Collapse
Affiliation(s)
- Asha Rizor
- Department of Pharmaceutical Sciences, College of Pharmacy, Florida A&M University, Tallahassee, FL, 32301, USA
| | - Edward Pajarillo
- Department of Pharmaceutical Sciences, College of Pharmacy, Florida A&M University, Tallahassee, FL, 32301, USA
| | - Ivan Nyarko-Danquah
- Department of Pharmaceutical Sciences, College of Pharmacy, Florida A&M University, Tallahassee, FL, 32301, USA
| | - Alexis Digman
- Department of Pharmaceutical Sciences, College of Pharmacy, Florida A&M University, Tallahassee, FL, 32301, USA
| | - Leyah Mooneyham
- Department of Pharmaceutical Sciences, College of Pharmacy, Florida A&M University, Tallahassee, FL, 32301, USA
| | - Deok-Soo Son
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine Bronx, New York, NY, 10461, USA; Sechenov First Moscow State Medical University, Moscow, Russia
| | - Eunsook Lee
- Department of Pharmaceutical Sciences, College of Pharmacy, Florida A&M University, Tallahassee, FL, 32301, USA.
| |
Collapse
|
34
|
Neuroprotective Potential of Bone Marrow-Derived Mesenchymal Stem Cells Following Chemotherapy. Biomedicines 2021; 9:biomedicines9070750. [PMID: 34209542 PMCID: PMC8301303 DOI: 10.3390/biomedicines9070750] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/04/2021] [Accepted: 06/07/2021] [Indexed: 11/17/2022] Open
Abstract
Cisplatin (CP) is extensively used in the medical oncology field for malignancy treatment, but its use is associated with neurological side effects that compromise the patients' quality of life. Cytotherapy is a new treatment strategy for tissue damage that has recently emerged. The use of bone marrow-derived mesenchymal stem cells (BM-MSCs) was investigated for its therapeutic potential against CP-induced chemobrain as well as various models of brain damage. This study was carried out to elucidate, for the first time, the role of the intravenous injection (IV) of BM-MSCs against CP-induced neurotoxicity in a rat model through investigation of the parameters of oxidative stress, inflammation, and apoptosis in brain tissue. A rat model of neurotoxicity was generated by intraperitoneal injection of 7.5 mg/kg CP while 2 × 106 BM-MSCs was given by IV as a therapeutic dose. Injection of CP led to a significant rise in malondialdehyde and nitric oxide levels accompanied by a marked depletion of superoxide dismutase and reduced glutathione content in brain tissue in comparison to the normal control (NC) rats. Furthermore, a remarkable rise in the brain levels of inflammatory cytokines interleukin (IL)-1β and IL-6, together with the expression of apoptotic marker caspase-3, and the downregulation of the brain expression of proliferating marker Ki-67 in brain tissue were detected in the CP group compared to the NC group. Histopathological alterations were observed in the brain tissue of the CP group. BM-MSCs mitigated the biochemical and histopathological alterations induced by CP without affecting brain cell proliferation. BM-MSCs could be used as a promising neuroprotective agent against CP-induced neurotoxicity.
Collapse
|
35
|
Chemical and Green ZnO nanoparticles ameliorated adverse effects of cisplatin on histological structure, antioxidant defense system and neurotrophins expression in rat hippocampus. J Chem Neuroanat 2021; 116:101990. [PMID: 34146667 DOI: 10.1016/j.jchemneu.2021.101990] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 06/14/2021] [Accepted: 06/14/2021] [Indexed: 11/20/2022]
Abstract
Cisplatin (CP) is a chemotherapy agent used in the treatment of cancer, but it has various side effects, in particular, neurotoxicity. Zinc oxide nanoparticles (ZnO NPs) are a potent antioxidant. However, there is limited knowledge about the protective effects of ZnO NPs against CP-induced hippocampal toxicity. The present study aimed to explore the potential protective effects of ZnO NPs against CP-induced oxidative stress, loss of neurotrophins support, and tissue damage in the hippocampus of the rats. Eighty adult male Wistar rats were dividing into ten groups including: control (Con), sham, ZnO Bulk (ZnB), chemical ZnO NPs (ChZnO NPs), Green ZnO NPs (GrZnO NPs), CP, CP + ZnB, CP + ChZnO NPs, CP + GrZnO NPs and CP + AE. CP was administrated (5 mg/kg/weekly) for four weeks, and animals were treated simultaneously with different forms of ZnO (5 mg/kg/day). At the end of the experiment, the activities of superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), malondialdehyde (MDA), changes of reduced glutathione (GSH), oxidized glutathione (GSSG) and GSH/GSSG ratio, histological changes, expression of brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF) genes were assessed in the hippocampus. The results revealed that a decrease in BDNF and NGF mRNA expression, GSH concentration and GSH/GSSG ratio, increasing of GSSG and MDA levels, and neuronal loss in the CP-treated rats were reversed following the administration of different forms of ZnO, especially Gr ZnO NPs and ch ZnO NPs. Co-administration of ZnO NPs to CP-treated rats restored the suppressive effects of CP on activities of antioxidant enzymes (SOD, GPX, CAT). The results showed that in most of the evaluated factors, Gr ZnO NPs showed a greater protective effect than other forms of ZnO. The results suggest that ZnO NPs, in particular Green ZnO NPs (GrZnO NPs) had more potential protective effects against CP-induced oxidative stress, inadequate support neurotrophin and tissue damage in rat hippocampus.
Collapse
|
36
|
Sriphongphankul H, Liabsuetrakul T, Osatakul S. Clinical Outcomes of Children Diagnosed Dengue-Associated Acute Liver Failure with or without N-Acetylcysteine Treatment: A Retrospective Cohort Study. J Trop Pediatr 2021; 67:fmab039. [PMID: 34100091 DOI: 10.1093/tropej/fmab039] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVES N-acetylcysteine (NAC) has been shown to prevent hepatic damage and improve microcirculatory blood flow and oxygen delivery to the tissue. Previous studies have proposed the benefit of NAC in dengue-associated acute liver failure (ALF). However, most studies are descriptive and lack comparison between groups. We aimed to compare the ALF resolution rate and mortality rate of those who received and did not receive NAC treatment. METHODS A retrospective cohort study was conducted among children aged <15 years who were diagnosed with dengue-associated ALF at a tertiary hospital in Thailand, between January 2002 and July 2019. Demographic and clinical information were collected. Main outcomes were ALF resolution and mortality rate. RESULTS Thirty-three patients were included of which 16 received NAC treatment (48.5%). Mean ages were 8.5 years (SD 3.7) and mean onset of ALF was 6.3 days (SD 1.6) after onset of fever. The grading of hepatic encephalopathy (HE) and organ failure was not significantly different between the two groups. In the NAC group, 13/16 children were prescribed 100 mg/kg/day of NAC until INR <2 without HE or <1.5 with HE. NAC was initiated 1.1 days (SD 0.3) after the ALF diagnosis. The NAC group showed a higher rate of ALF resolution (75% vs. 53% in the non-NAC group, p = 0.34) with a lower mortality rate (31% vs. 53%, p = 0.36). Side effects of NAC were not found. CONCLUSION NAC may be beneficial in dengue-associated pediatric ALF. Further well-designed randomized control trials should be carried out.
Collapse
Affiliation(s)
- Hansa Sriphongphankul
- Department of Pediatrics, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| | - Tippawan Liabsuetrakul
- Epidemiology Unit, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| | - Seksit Osatakul
- Department of Pediatrics, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| |
Collapse
|
37
|
Saral S, Topçu A, Alkanat M, Mercantepe T, Akyıldız K, Yıldız L, Tümkaya L, Yazıcı ZA, Yılmaz A. Apelin-13 activates the hippocampal BDNF/TrkB signaling pathway and suppresses neuroinflammation in male rats with cisplatin-induced cognitive dysfunction. Behav Brain Res 2021; 408:113290. [PMID: 33845103 DOI: 10.1016/j.bbr.2021.113290] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 04/02/2021] [Accepted: 04/03/2021] [Indexed: 01/01/2023]
Abstract
It has been established that cisplatin causes neuronal damage and cognitive impairment. However, the mechanism is not sufficiently clear. Apelin-13 is an endogenous peptide with strong neuroprotective effects through the synthesis of neurotrophic factors and suppression of inflammation. The aim of this study was to investigate the role of brain-derived neurotrophic factor/tropomyosin receptor kinase B (BDNF/TrkB) signaling pathway and the potential inhibitory effects of apelin-13 in the mechanism of cisplatin-induced hippocampal damage and cognitive impairment. Apelin-13 was administered to adult sprague dawley male rats at a dose of 20 nmol/kg every day for 4 weeks, cisplatin was administered at a dose of 5 mg/kg once a week for 4 weeks. The spatial and recognition memory tests of the rats were performed on the 5th week. BDNF and the inflammatory cytokines tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) levels were measured by ELISA in hippocampal homogenates. Pyramidal neuron and glial cell damage in the hippocampal CA1, CA3 and dentate gyrus (DG) were analyzed histologically. TrkB activity in the hippocampus was determined by immunohistochemical methods. Cisplatin impaired spatial and recognition memory in rats, while apelin-13 improved spatial memory but did not affect recognition memory. Cisplatin suppressed BDNF in the hippocampus while increased IL-1β and TNF-α. In contrast, apelin-13 administration increased BDNF but significantly suppressed TNF-α and IL-1B. Cisplatin caused pyramidal neuron and glial cell damage in CA1, CA3 and DG. In the cisplatin + apelin-13 group, however, pyramidal neuron and glial cell damage was less than those without apelin-13. Cisplatin increased TrkB activity in the hippocampus, which was counteracted by apelin-13. In conclusion, apelin-13 reduced the cisplatin-induced cognitive deficiency, by suppressing inflammation and stimulating the synthesis and activation of neurotrophic factors in hippocampal tissue.
Collapse
Affiliation(s)
- Sinan Saral
- Recep Tayyip Erdogan University, Faculty of Medicine, Department of Physiology, Rize, Turkey.
| | - Atilla Topçu
- Recep Tayyip Erdogan University, Faculty of Medicine, Department of Medical Pharmacology, Rize, Turkey.
| | - Mehmet Alkanat
- Giresun University, Faculty of Medicine, Department of Physiology, Giresun, Turkey.
| | - Tolga Mercantepe
- Recep Tayyip Erdogan University, Faculty of Medicine, Department of Histology and Embryology, Rize, Turkey.
| | - Kerimali Akyıldız
- Recep Tayyip Erdogan University, School of Healh Care Services Vocational, Department of Medical Services and Techniques, Rize, Turkey.
| | - Lamiye Yıldız
- Recep Tayyip Erdogan University, Faculty of Medicine, Department of Physiology, Rize, Turkey.
| | - Levent Tümkaya
- Recep Tayyip Erdogan University, Faculty of Medicine, Department of Histology and Embryology, Rize, Turkey.
| | - Zihni Açar Yazıcı
- Recep Tayyip Erdogan University, Faculty of Medicine, Department of Microbiology, Rize, Turkey.
| | - Adnan Yılmaz
- Recep Tayyip Erdogan University, Faculty of Medicine, Department of Biochemistry, Rize, Turkey.
| |
Collapse
|
38
|
Wellenberg A, Weides L, Kurzke J, Hennecke T, Bornhorst J, Crone B, Karst U, Brinkmann V, Fritz G, Honnen S. Use of C. elegans as a 3R-compliant in vivo model for the chemoprevention of cisplatin-induced neurotoxicity. Exp Neurol 2021; 341:113705. [PMID: 33753139 DOI: 10.1016/j.expneurol.2021.113705] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 03/14/2021] [Accepted: 03/17/2021] [Indexed: 02/07/2023]
Abstract
Anticancer therapeutics can provoke severe side effects that impair the patient's quality of life. A frequent dose-limiting side effect of platinum-based anticancer therapy is neurotoxicity. Its pathophysiology is poorly understood, and effective preventive or therapeutic measures are missing. Therefore, elucidation of the molecular mechanism of platinating drug-induced neurotoxicity and the development of preventive strategies is urgently needed. To this end, we aim to use C. elegans as a 3R-compliant in vivo model. The 3R principles were conceived for animal welfare in science concerning animal experiments, which should be replaced, reduced or refined. We can analytically demonstrate dose-dependent uptake of cisplatin (CisPt) in C. elegans, as well as genotoxic and cytotoxic effects based on DNA adduct formation (i.e., 1,2-GpG intrastrand crosslinks), induction of apoptosis, and developmental toxicity. Measuring the impairment of pharyngeal pumping as a marker of neurotoxicity, we found that especially CisPt reduces the pumping frequency at concentrations where basal and touch-provoked movement were not yet affected. CisPt causes glutathione (GSH) depletion and RNAi-mediated knockdown of the glutamate-cysteine ligase GCS-1 aggravates the CisPt-induced inhibition of pharyngeal pumping. Moreover, N-acetylcysteine (NAC) mitigated CisPt-triggered toxicity, indicating that GSH depletion contributes to the CisPt-induced pharyngeal damage. In addition to NAC, amifostine (WR1065) also protected the pharynx of C. elegans from the toxic effects of CisPt. Measuring pharyngeal activity by the electrophysiological recording of neurotransmission in the pharynx, we confirmed that CisPt is neurotoxic in C. elegans and that NAC is neuroprotective in the nematode. The data support the hypothesis that monitoring the pharyngeal activity of C. elegans is a useful surrogate marker of CisPt-induced neurotoxicity. In addition, a low GSH pool reduces the resistance of neurons to CisPt treatment, and both NAC and WR1065 are capable of attenuating platinum-induced neurotoxicity during post-incubation in C. elegans. Overall, we propose C. elegans as a 3R-compliant in vivo model to study the molecular mechanisms of platinum-induced neurotoxicity and to explore novel neuroprotective therapeutic strategies to alleviate respective side effects of platinum-based cancer therapy.
Collapse
Affiliation(s)
- Anna Wellenberg
- Institute of Toxicology, Medical Faculty, Heinrich Heine University, Universitätsstraße 1, D-40225 Düsseldorf, Germany.
| | - Lea Weides
- Institute of Toxicology, Medical Faculty, Heinrich Heine University, Universitätsstraße 1, D-40225 Düsseldorf, Germany.
| | - Jennifer Kurzke
- Institute of Toxicology, Medical Faculty, Heinrich Heine University, Universitätsstraße 1, D-40225 Düsseldorf, Germany
| | - Till Hennecke
- Institute of Toxicology, Medical Faculty, Heinrich Heine University, Universitätsstraße 1, D-40225 Düsseldorf, Germany
| | - Julia Bornhorst
- Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, D-14558 Nuthetal, Germany; Faculty of Mathematics and Natural Sciences, Food Chemistry, University of Wuppertal, Gaußstr. 20, D-42119 Wuppertal, Germany.
| | - Barbara Crone
- Institute of Inorganic and Analytical Chemistry, University of Muenster, Corrensstraße 30, D-48149 Muenster, Germany.
| | - Uwe Karst
- Institute of Inorganic and Analytical Chemistry, University of Muenster, Corrensstraße 30, D-48149 Muenster, Germany.
| | - Vanessa Brinkmann
- Institute of Toxicology, Medical Faculty, Heinrich Heine University, Universitätsstraße 1, D-40225 Düsseldorf, Germany.
| | - Gerhard Fritz
- Institute of Toxicology, Medical Faculty, Heinrich Heine University, Universitätsstraße 1, D-40225 Düsseldorf, Germany.
| | - Sebastian Honnen
- Institute of Toxicology, Medical Faculty, Heinrich Heine University, Universitätsstraße 1, D-40225 Düsseldorf, Germany.
| |
Collapse
|
39
|
Kazak F, Akalın PP, Yarım GF, Başpınar N, Özdemir Ö, Ateş MB, Altuğ ME, Deveci MZY. Protective effects of nobiletin on cisplatin induced neurotoxicity in rats. Int J Neurosci 2021; 132:1-7. [PMID: 33650929 DOI: 10.1080/00207454.2021.1896507] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 01/11/2021] [Accepted: 01/30/2021] [Indexed: 12/26/2022]
Abstract
OBJECTIVES This study was designed to investigate the possible antioxidant, antiapoptotic and neuroprotective effects of nobiletin on cisplatin-induced neurotoxicity rat model by evaluating neurotrophins, antioxidants and histopathology. METHODS Forty male Wistar Albino rats were divided into four groups: control, cisplatin (CIS), cisplatin + nobiletin (CIS + NOB) and nobiletin + cisplatin (NOB + CIS). CIS + NOB was applied nobiletin (10 mg/kg, i.p.) during the last four days whereas NOB + CIS was applied nobiletin during the first four days of the study. Cisplatin (4 mg/kg, i.p. twice a day) was administered to the experimental groups on the 5th day of the study. All rats were sacrificed on the 10th day of the study. BDNF, NGF, G6PD, GPx, tGSH and MDA levels were determined in brain. In addition, routin histolopathological analysis and caspase-3 immunoreactivity assay were conducted. RESULTS BDNF concentrations increased in nobiletin-administered groups, compared to Control and CIS and that the increase was statistically significant in NOB + CIS (p < 0.05). It was also found that G6PD activity increased (p < 0.05) in the nobiletin-administered groups, compared to control and CIS. Histopathologically, neuronal degeneration, oedema and gliosis increased in CIS compared to Control, and nobiletin administration decreased neuronal degeneration and oedema compared to CIS (p < 0.05). Cisplatin increased (p < 0.05) caspase-3 immunoreactivity in cerebrovascular endothelium and neurons compared to Control, while nobiletin administration decreased caspase-3 immunoreactivity in cerebrovascular endothelium. Caspase-3 immunoreactivity in neurons decreased only in NOB + CIS (p < 0.05). CONCLUSION Nobiletin increased BDNF concentration and G6PD activity in brain and when evaluated together with histopathological and immunohistochemical findings, it may have antioxidant, antiapoptotic and neuroprotective effects against cisplatin.
Collapse
Affiliation(s)
- Filiz Kazak
- Department of Biochemistry, Veterinary Faculty, Hatay Mustafa Kemal University, Hatay, Turkey
| | - Pınar Peker Akalın
- Department of Biochemistry, Veterinary Faculty, Hatay Mustafa Kemal University, Hatay, Turkey
| | - Gül Fatma Yarım
- Department of Biochemistry, Veterinary Faculty, Ondokuz Mayıs University, Samsun, Turkey
| | - Nuri Başpınar
- Department of Biochemistry, Veterinary Faculty, Selçuk University, Konya, Turkey
| | - Özgür Özdemir
- Department of Pathology, Veterinary Faculty, Selçuk University, Konya, Turkey
| | - Mehmet Burak Ateş
- Department of Pathology, Veterinary Faculty, Selçuk University, Konya, Turkey
| | - Muhammed Enes Altuğ
- Department of Surgery, Veterinary Faculty, Hatay Mustafa Kemal University, Hatay, Turkey
| | | |
Collapse
|
40
|
Fumagalli G, Monza L, Cavaletti G, Rigolio R, Meregalli C. Neuroinflammatory Process Involved in Different Preclinical Models of Chemotherapy-Induced Peripheral Neuropathy. Front Immunol 2021; 11:626687. [PMID: 33613570 PMCID: PMC7890072 DOI: 10.3389/fimmu.2020.626687] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 12/21/2020] [Indexed: 12/12/2022] Open
Abstract
Peripheral neuropathies are characterized by nerves damage and axonal loss, and they could be classified in hereditary or acquired forms. Acquired peripheral neuropathies are associated with several causes, including toxic agent exposure, among which the antineoplastic compounds are responsible for the so called Chemotherapy-Induced Peripheral Neuropathy (CIPN). Several clinical features are related to the use of anticancer drugs which exert their action by affecting different mechanisms and structures of the peripheral nervous system: the axons (axonopathy) or the dorsal root ganglia (DRG) neurons cell body (neuronopathy/ganglionopathy). In addition, antineoplastic treatments may affect the blood brain barrier integrity, leading to cognitive impairment that may be severe and long-lasting. CIPN may affect patient quality of life leading to modification or discontinuation of the anticancer therapy. Although the mechanisms of the damage are not completely understood, several hypotheses have been proposed, among which neuroinflammation is now emerging to be relevant in CIPN pathophysiology. In this review, we consider different aspects of neuro-immune interactions in several CIPN preclinical studies which suggest a critical connection between chemotherapeutic agents and neurotoxicity. The features of the neuroinflammatory processes may be different depending on the type of drug (platinum derivatives, taxanes, vinca alkaloids and proteasome inhibitors). In particular, recent studies have demonstrated an involvement of the immune response (both innate and adaptive) and the stimulation and secretion of mediators (cytokines and chemokines) that may be responsible for the painful symptoms, whereas glial cells such as satellite and Schwann cells might contribute to the maintenance of the neuroinflammatory process in DRG and axons respectively. Moreover, neuroinflammatory components have also been shown in the spinal cord with microglia and astrocytes playing an important role in CIPN development. Taking together, better understanding of these aspects would permit the development of possible strategies in order to improve the management of CIPN.
Collapse
Affiliation(s)
- Giulia Fumagalli
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.,NeuroMI (Milan Center for Neuroscience), University of Milano-Bicocca, Monza, Italy
| | - Laura Monza
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.,NeuroMI (Milan Center for Neuroscience), University of Milano-Bicocca, Monza, Italy
| | - Guido Cavaletti
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.,NeuroMI (Milan Center for Neuroscience), University of Milano-Bicocca, Monza, Italy
| | - Roberta Rigolio
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.,NeuroMI (Milan Center for Neuroscience), University of Milano-Bicocca, Monza, Italy
| | - Cristina Meregalli
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.,NeuroMI (Milan Center for Neuroscience), University of Milano-Bicocca, Monza, Italy
| |
Collapse
|
41
|
Cankara FN, Günaydın C, Çelik ZB, Şahin Y, Pekgöz Ş, Erzurumlu Y, Gülle K. Agomelatine confers neuroprotection against cisplatin-induced hippocampal neurotoxicity. Metab Brain Dis 2021; 36:339-349. [PMID: 33165734 DOI: 10.1007/s11011-020-00634-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/14/2020] [Indexed: 01/17/2023]
Abstract
Neurotoxicity caused by cisplatin is a major obstacle during chemotherapy. Oxidative stress and inflammation are considered the primary mechanism behind neuronal damage which affects the continuing chemotherapy regimen. Agomelatine was recently described as a neuroprotective compound against toxic insults in the nervous systems. It is an analog of the well-known antioxidant and anti-inflammatory compound melatonin and currently used for depression and sleep disturbances. In the current study, we investigated the possible neuroprotective role of agomelatine against cisplatin-induced oxidative, inflammatory, and behavioral alterations in male rats. Our results show that agomelatine prevented cisplatin-induced neurotoxicity in the HT-22 mouse hippocampal neuronal cell line. Additionally, agomelatine treatment inhibited cisplatin-induced behavioral deficits and neuronal integrity in vivo. For the evaluation of the effect of agomelatine on oxidative stress and inflammation, GSH, MDA, TNF, and IL-6 levels were analyzed in HT-22 cells and hippocampal tissues. Agomelatine significantly attenuated oxidative stress and inflammation due to the cisplatin insult in vitro and in vivo. Also, agomelatine treatment ameliorated the neuronal pathology in the hippocampus, which is strongly related to cognition and memory. Taken together, our results indicate that in males, the neuroprotective effect of agomelatine is mediated through its antioxidant and anti-inflammatory actions abrogating functional deficits.
Collapse
Affiliation(s)
- Fatma Nihan Cankara
- Department of Pharmacology, Faculty of Medicine, Suleyman Demirel University, Isparta, 32260, Turkey.
| | - Caner Günaydın
- Department of Pharmacology, Faculty of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | - Zülfinaz Betül Çelik
- Department of Medical Biology, Faculty of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | - Yasemin Şahin
- Department of Pharmacology, Faculty of Medicine, Suleyman Demirel University, Isparta, 32260, Turkey
| | - Şakir Pekgöz
- Department of Pharmacology, Faculty of Medicine, Suleyman Demirel University, Isparta, 32260, Turkey
| | - Yalçın Erzurumlu
- Department of Biochemistry, Faculty of Pharmacy, Suleyman Demirel University, Isparta, Turkey
| | - Kanat Gülle
- Department of Histology and Embryology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| |
Collapse
|
42
|
Mantawy EM, Said RS, Kassem DH, Abdel-Aziz AK, Badr AM. Novel molecular mechanisms underlying the ameliorative effect of N-acetyl-L-cysteine against ϒ-radiation-induced premature ovarian failure in rats. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 206:111190. [PMID: 32871518 DOI: 10.1016/j.ecoenv.2020.111190] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 07/26/2020] [Accepted: 08/16/2020] [Indexed: 06/11/2023]
Abstract
Radiotherapy represents a critical component in cancer treatment. However, premature ovarian failure (POF) is a major hurdle of deleterious off-target effects in young females, which, therefore, call for an effective radioprotective agent. The present study aimed to explore the molecular mechanism underlying the protective effects of N-acetyl-L-cysteine (NAC) against γ-radiation-provoked POF. Immature female Sprague-Dawley rats were orally-administered NAC (50 mg/kg) and were exposed to a single whole-body dose of 3.2 Gy ϒ-radiation. NAC administration remarkably reversed abnormal serum estradiol and anti-Müllerian hormone levels by 73% and 40%, respectively while ameliorating the histopathological and ultrastructural alterations-triggered by γ-radiation. Mechanistically, NAC alleviated radiation-induced oxidative damage through significantly increased glutathione peroxidase activity by 102% alongside with decreasing NADPH oxidase subunits (p22 and NOX4) gene expressions by 48% and 38%, respectively compared to the irradiated untreated group. Moreover, NAC administration achieved its therapeutic effect by inhibiting ovarian apoptosis-induced by radiation through downregulating p53 and Bax levels by 33% and 16%, respectively while increasing the Bcl-2 mRNA expression by 135%. Hence, the Bax/Bcl2 ratio and cytochrome c expression were subsequently reduced leading to decreased caspase 3 activity by 43%. Importantly, the anti-apoptotic property of NAC could be attributed to inactivation of MAPK signaling molecules; p38 and JNK, and enhancement of the ovarian vascular endothelial growth factor (VEGF) expression. Taken together, our results suggest that NAC can inhibit radiotherapy-induced POF while preserving ovarian function and structure through upregulating VEGF expression and suppressing NOX4/MAPK/p53 apoptotic signaling.
Collapse
Affiliation(s)
- Eman M Mantawy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Riham S Said
- Department of Drug Radiation Research, National Center for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt.
| | - Dina H Kassem
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Amal Kamal Abdel-Aziz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Amira Mohamed Badr
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
43
|
Mansour HH, Omran MM, Hasan HF, El Kiki SM. Modulation of bleomycin-induced oxidative stress and pulmonary fibrosis by N-acetylcysteine in rats via AMPK/SIRT1/NF-κβ. Clin Exp Pharmacol Physiol 2020; 47:1943-1952. [PMID: 32658336 DOI: 10.1111/1440-1681.13378] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 07/07/2020] [Accepted: 07/08/2020] [Indexed: 12/12/2022]
Abstract
The efficacy of bleomycin (BLM) as an antineoplastic drug is limited to the development of dose and time-dependent pulmonary fibrosis. This study was intended to investigate the effect of N-acetylcysteine (NAC) on BLM-induced pulmonary fibrosis in rats. Twenty rats were randomly divided to the following four groups: Group one served as control; group two received BLM (15 mg/kg, intraperitoneal (ip)) for five consecutive days; group three received NAC (200 mg/kg, ip) for five consecutive days; and group four received NAC 1 hour before BLM for 5 days. The expression of connective tissue growth factor (CTGF), platelet-derived growth factor (PDGF), silent information regulator l (SIRT1), AMP-activated protein kinase (AMPK) were determined by qRT-PCR in lung tissues. The changes in transforming growth factor-beta1 (TGF-β1), tumour necrosis factor-α (TNF-α), interleukin-β1 (IL-β1) and nuclear factor kappa-β (NF-κβ) in serum were measured by ELISA. The tissue antioxidant status was determined biochemically. BLM administration caused pulmonary fibrosis as evidenced by increased levels of inflammatory mediators (TGF-β1, TNF-α, IL-β1 and NF-κβ) in serum (P < .05), elevated lipid peroxidation and nitric oxide and depleted endogenous antioxidants in lung tissue (P < .05). The expression levels of SIRT1 and AMPK were significantly decreased (P < .05), while the expression levels of CTGF and PDGF were increased significantly in the BLM group as compared to the control group (P < .05). These alterations were normalized by NAC intervention. NAC markedly attenuated the lung histopathological changes and reduced collagen deposition. These results suggest that NAC exerted an ameliorative effect against BLM-induced oxidative damage and pulmonary fibrosis via SIRT1/ AMPK/ NF-κβ pathways.
Collapse
Affiliation(s)
- Heba H Mansour
- Health Radiation Research Department, National Center for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| | - Mervat M Omran
- Pharmacology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Hesham F Hasan
- Radiation Biology Department, National Center for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| | - Shereen M El Kiki
- Health Radiation Research Department, National Center for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| |
Collapse
|
44
|
Liang KX, Vatne GH, Kristiansen CK, Ievglevskyi O, Kondratskaya E, Glover JC, Chen A, Sullivan GJ, Bindoff LA. N-acetylcysteine amide ameliorates mitochondrial dysfunction and reduces oxidative stress in hiPSC-derived dopaminergic neurons with POLG mutation. Exp Neurol 2020; 337:113536. [PMID: 33264635 DOI: 10.1016/j.expneurol.2020.113536] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 11/22/2020] [Accepted: 11/22/2020] [Indexed: 01/03/2023]
Abstract
The inability to reliably replicate mitochondrial DNA (mtDNA) by mitochondrial DNA polymerase gamma (POLG) leads to a subset of common mitochondrial diseases associated with neuronal death and depletion of neuronal mtDNA. Defining disease mechanisms in neurons remains difficult due to the limited access to human tissue. Using human induced pluripotent stem cells (hiPSCs), we generated functional dopaminergic (DA) neurons showing positive expression of dopaminergic markers TH and DAT, mature neuronal marker MAP2 and functional synaptic markers synaptophysin and PSD-95. These DA neurons were electrophysiologically characterized, and exhibited inward Na + currents, overshooting action potentials and spontaneous postsynaptic currents (sPSCs). POLG patient-specific DA neurons (POLG-DA neurons) manifested a phenotype that replicated the molecular and biochemical changes found in patient post-mortem brain samples namely loss of complex I and depletion of mtDNA. Compared to disease-free hiPSC-derived DA neurons, POLG-DA neurons exhibited loss of mitochondrial membrane potential, loss of complex I and loss of mtDNA and TFAM expression. POLG driven mitochondrial dysfunction also led to neuronal ROS overproduction and increased cellular senescence. This deficit was selectively rescued by treatment with N-acetylcysteine amide (NACA). In conclusion, our study illustrates the promise of hiPSC technology for assessing pathogenetic mechanisms associated with POLG disease, and that NACA can be a promising potential therapy for mitochondrial diseases such as those caused by POLG mutation.
Collapse
Affiliation(s)
- Kristina Xiao Liang
- Neuro-SysMed, Center of Excellence for Clinical Research in Neurological Diseases, Department of Neurology, Haukeland University Hospital, Jonas Lies vei 87, P. O. Box 7804, 5021 Bergen, Norway; Department of Clinical Medicine (K1), University of Bergen, Jonas Lies vei 87, P. O. Box 7804, 5021 Bergen, Norway.
| | - Guro Helén Vatne
- Department of Clinical Medicine (K1), University of Bergen, Jonas Lies vei 87, P. O. Box 7804, 5021 Bergen, Norway
| | - Cecilie Katrin Kristiansen
- Department of Clinical Medicine (K1), University of Bergen, Jonas Lies vei 87, P. O. Box 7804, 5021 Bergen, Norway
| | - Oleksandr Ievglevskyi
- The Intervention Centre, Oslo University Hospital, P. O. Box 4950, Nydalen, 0424 Oslo, Norway; Laboratory of Neural Development and Optical Recording (NDEVOR), Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, P. O. Box 1103, Blindern, 0317 Oslo, Norway
| | - Elena Kondratskaya
- Laboratory of Neural Development and Optical Recording (NDEVOR), Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, P. O. Box 1103, Blindern, 0317 Oslo, Norway
| | - Joel C Glover
- Laboratory of Neural Development and Optical Recording (NDEVOR), Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, P. O. Box 1103, Blindern, 0317 Oslo, Norway; Norwegian Center for Stem Cell Research, Department of Immunology and Transfusion Medicine, Oslo University Hospital, P. O. Box 4950, Nydalen, 0424 Oslo, Norway
| | - Anbin Chen
- Department of Clinical Medicine (K1), University of Bergen, Jonas Lies vei 87, P. O. Box 7804, 5021 Bergen, Norway; Department of Neurosurgery, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan 250012, Shandong Province, China; Shandong Key Laboratory of Brain Function Remodeling, Shandong University, 107 Wenhua Xi Road, Jinan 250012, Shandong Province, China
| | - Gareth John Sullivan
- Norwegian Center for Stem Cell Research, Department of Immunology and Transfusion Medicine, Oslo University Hospital, P. O. Box 4950, Nydalen, 0424 Oslo, Norway; Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, P. O. Box 1105, Blindern, 0317 Oslo, Norway; Institute of Immunology, Oslo University Hospital, PO Box 4950, 0424 Oslo, Norway; Hybrid Technology Hub - Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, P. O. Box 1110, Blindern, 0317 Oslo, Norway; Department of Pediatric Research, Oslo University Hospital, P. O. Box 4950, Nydalen, 0424 Oslo, Norway
| | - Laurence A Bindoff
- Neuro-SysMed, Center of Excellence for Clinical Research in Neurological Diseases, Department of Neurology, Haukeland University Hospital, Jonas Lies vei 87, P. O. Box 7804, 5021 Bergen, Norway; Department of Clinical Medicine (K1), University of Bergen, Jonas Lies vei 87, P. O. Box 7804, 5021 Bergen, Norway.
| |
Collapse
|
45
|
Afjal MA, Goswami P, Ahmad S, Dabeer S, Akhter J, Salman M, Mangla A, Raisuddin S. Tempol (4-hydroxy tempo) protects mice from cisplatin-induced acute kidney injury via modulation of expression of aquaporins and kidney injury molecule-1. Drug Chem Toxicol 2020; 45:1355-1363. [PMID: 33078650 DOI: 10.1080/01480545.2020.1831011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Tempol (4-hydroxy tempo), a pleiotropic antioxidant is reported to afford protection against cisplatin (CP)-induced nephrotoxicity. However, molecular mechanisms of action of tempol in improving the renal function in CP-induced nephrotoxicity are not fully understood. We investigated the attenuating effect of tempol against CP-induced alterations in kidney injury molecule-1 (KIM-1) and aquaporins (AQPs) in mice. Tempol (100 mg/kg, po) pretreatment with CP (20 mg/kg ip) showed restoration in renal function markers including electrolytes. CP treatment upregulated mRNA expression of KIM-1 and downregulated AQP and arginine vasopressin (AVP) expression which was attenuated by tempol. Immunoblotting analysis revealed that CP-induced alterations in KIM-1 and AQP expression were restored by tempol. Immunofluorocense study also showed restorative effect of tempol on the expression of AQP2 in CP-treated mice. In conclusion, this study provides experimental evidence that tempol resolved urinary concentration defect by the restoration of AQP, AVP and KIM-1 levels indicating a potential use of tempol in ameliorating the AKI in cancer patients under the treatment with CP.
Collapse
Affiliation(s)
- Mohd Amir Afjal
- Molecular Toxicology Laboratory, Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi, India
| | - Poonam Goswami
- Molecular Toxicology Laboratory, Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi, India
| | - Shahzad Ahmad
- Molecular Toxicology Laboratory, Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi, India
| | - Sadaf Dabeer
- Molecular Toxicology Laboratory, Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi, India
| | - Juheb Akhter
- Molecular Toxicology Laboratory, Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi, India
| | - Mohd Salman
- Molecular Neurobiology Laboratory, Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi, India
| | - Anuradha Mangla
- Molecular Toxicology Laboratory, Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi, India
| | - Sheikh Raisuddin
- Molecular Toxicology Laboratory, Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi, India
| |
Collapse
|
46
|
Park YJ, Kim KS, Park JH, Lee SH, Kim HR, Lee SH, Choi HB, Cao S, Kumar V, Kwak JH, Kim HS. Protective effects of dendropanoxide isolated from Dendropanax morbifera against cisplatin-induced acute kidney injury via the AMPK/mTOR signaling pathway. Food Chem Toxicol 2020; 145:111605. [PMID: 32750447 DOI: 10.1016/j.fct.2020.111605] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/07/2020] [Accepted: 07/09/2020] [Indexed: 12/11/2022]
Abstract
The aim of this study was to investigate the protective effects of dendropanoxide (DPx) isolated from Dendropanax morbifera against cis-diamminedichloroplatinum (II) (CDDP)-induced nephrotoxicity in NRK-52E cells and in Sprague-Dawley rats. DPx was administered to Sprague-Dawley rats by oral gavage (5 and 10 mg/kg) for 7 consecutive days, 24 h after intraperitoneal injection with CDDP (6 mg/kg). All rats were euthanized 24 h after the last DPx administration, and histopathological damage, acute kidney injury (AKI) biomarkers, inflammatory cytokines, and oxidative damages were evaluated. DPx (5 and 10 μg/mL) was found to protect against CDDP-induced cytotoxicity and apoptotic cell death in NRK-52E cells. CDDP-induced serum blood urea nitrogen (BUN), creatinine (sCr), and pro-inflammatory cytokines levels were significantly ameliorated by DPx in a dose-dependent manner. Furthermore, excretion of kidney injury molecules (KIM-1), selenium binding protein-1 (SBP-1), and neutrophil gelatinase-associated lipocalin (NGAL) in the urine was significantly reduced in response to DPx administration in CDDP-treated rats. Activities of antioxidant enzymes and lipid peroxidation levels were markedly altered in the kidney of CDDP-treated rats in response to DPx administration. Serum pro-inflammatory cytokine levels were dramatically suppressed by DPx in CDDP-treated rats. DPx also restored renal-cell apoptosis via regulation of AMPK/mTOR signaling in CDDP-treated rats. Our results clearly suggest that DPx ameliorates CDDP-induced nephrotoxicity in vitro and in vivo by inhibiting oxidative stress, inflammation, and apoptosis. Overall, our data demonstrates that DPx may serve as a therapeutic agent in patients with solid tumors to prevent CDDP-induced AKI.
Collapse
Affiliation(s)
- Yoo Jung Park
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Kyeong Seok Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jae Hyeon Park
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Song Hee Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hae Ri Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Su Hyun Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hye Been Choi
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Shugeng Cao
- Department of Pharmaceutical Sciences, Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, 200 West Kawili Street, Hilo, HI, 96720, USA
| | - Vikas Kumar
- Natural Product Drug Discovery Laboratory, Department of Pharmaceutical Sciences, Shalom Institute of Health Sciences, Sam Higginbottom University of Agriculture, Technology & Sciences, Allahabad, 211007, India
| | - Jong Hwan Kwak
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Hyung Sik Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
47
|
Welbat JU, Naewla S, Pannangrong W, Sirichoat A, Aranarochana A, Wigmore P. Neuroprotective effects of hesperidin against methotrexate-induced changes in neurogenesis and oxidative stress in the adult rat. Biochem Pharmacol 2020; 178:114083. [PMID: 32522593 DOI: 10.1016/j.bcp.2020.114083] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 06/03/2020] [Accepted: 06/04/2020] [Indexed: 02/06/2023]
Abstract
Methotrexate (MTX) induces the formation of reactive oxygen species (ROS) and leads to neurotoxicity. The drug also negatively impacts neurogenesis and memory. Hesperidin (Hsd) is a major flavanoid with multiple beneficial pharmacological effects such as anti-oxidation, anti-inflammation, and neuroprotective effects. The aim of our study was to investigate the neuroprotective effects of Hsd against MTX-induced alterations in oxidative stress and neurogenesis. Sprague Dawley rats were divided into four groups: 1) a vehicle group, which received saline and propylene glycol, 2) an Hsd group, which was orally administered with Hsd (100 mg/kg) for 21 days, 3) an MTX group, which received MTX (75 mg/kg) by intravenous injection on days 8 and 15, and 4) an MTX + Hsd group, which received both MTX and Hsd. After treatment with MTX, p21-positive cells had increased significantly and doublecortin (DCX) expression in the hippocampus had decreased significantly. Treatment with MTX also increased malondialdehyde (MDA) in both the hippocampus and prefrontal cortex and decreased levels of brain-derived neurotropic factor (BDNF) and nuclear factor erythroid 2-related factor 2 (Nrf2) in the hippocampus and prefrontal cortex. Additionally, there were significant decreases in superoxide dismutase (SOD), glutathione peroxidase (GPx), and catalase (CAT) in the hippocampus and prefrontal cortex in the MTX group. However, co-treatment with Hsd ameliorated the negative effects of MTX on neurogenesis, oxidative stress, and antioxidant enzymes. These findings suggest that Hsd may be able to prevent neurotoxic effects of MTX by reducing oxidative stress and enhancing hippocampal neurogenesis.
Collapse
Affiliation(s)
- Jariya Umka Welbat
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; Neuroscience Research and Development Group, Khon Kaen University, Khon Kaen 40002, Thailand.
| | - Salinee Naewla
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Wanassanan Pannangrong
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Apiwat Sirichoat
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Anusara Aranarochana
- Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Peter Wigmore
- School of Life Sciences, Medical School, Queen's Medical Centre, Nottingham University, Nottingham, United Kingdom
| |
Collapse
|
48
|
Ashafaq M, Hussain S, Alshahrani S, Madkhali O, Siddiqui R, Khuwaja G, Alam MI, Islam F. Role of cinnamon oil against acetaminophen overdose induced neurological aberrations through brain stress and cytokine upregulation in rat brain. Drug Chem Toxicol 2020; 45:633-640. [DOI: 10.1080/01480545.2020.1747484] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Mohammad Ashafaq
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Sohail Hussain
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Saeed Alshahrani
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Osama Madkhali
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Rahimullah Siddiqui
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Gulrana Khuwaja
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - M. Intakhab Alam
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Fakhrul Islam
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan, Kingdom of Saudi Arabia
| |
Collapse
|
49
|
Antioxidants as a Potential Target against Inflammation and Oxidative Stress in Attention-Deficit/Hyperactivity Disorder. Antioxidants (Basel) 2020; 9:antiox9020176. [PMID: 32098021 PMCID: PMC7070894 DOI: 10.3390/antiox9020176] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/29/2020] [Accepted: 01/30/2020] [Indexed: 12/22/2022] Open
Abstract
Psychostimulants and non-psychostimulants are the medications prescribed for the treatment of attention-deficit/hyperactivity disorder (ADHD). However, several adverse results have been linked with an increased risk of substance use and side effects. The pathophysiology of ADHD is not completely known, although it has been associated with an increase in inflammation and oxidative stress. This review presents an overview of findings following antioxidant treatment for ADHD and describes the potential amelioration of inflammation and oxidative stress using antioxidants that might have a future as multi-target adjuvant therapy in ADHD. The use of antioxidants against inflammation and oxidative conditions is an emerging field in the management of several neurodegenerative and neuropsychiatric disorders. Thus, antioxidants could be promising as an adjuvant ADHD therapy.
Collapse
|
50
|
Santos NAGD, Ferreira RS, Santos ACD. Overview of cisplatin-induced neurotoxicity and ototoxicity, and the protective agents. Food Chem Toxicol 2019; 136:111079. [PMID: 31891754 DOI: 10.1016/j.fct.2019.111079] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 12/11/2019] [Accepted: 12/23/2019] [Indexed: 12/15/2022]
Abstract
Cisplatin has dramatically improved the survival rate of cancer patients, but it has also increased the prevalence of hearing and neurological deficits in this population. Cisplatin induces ototoxicity, peripheral (most prevalent) and central (rare) neurotoxicity. This review addresses the ototoxicity and the neurotoxicity associated with cisplatin-based chemotherapy, providing an integrated view of the potential protective agents that have been evaluated in vitro, in vivo and in clinical trials, their targets and mechanisms of protection and their effects on the antitumor activity of cisplatin. So far, the findings are insufficient to support the use of any oto- or neuroprotective agent before, during or after cisplatin chemotherapy. Despite their promising effects in vitro and in animal studies, many agents have not been evaluated in clinical trials. Additionally, the clinical trials have limitations concerning the sample size, controls, measurement, heterogeneous groups, several arms of treatment, short follow-up or no blinding. Besides that, for most agents, the effects on the antitumor activity of cisplatin have not been evaluated in tumor-bearing animals, which discourages clinical trials. Further well-designed randomized controlled clinical trials are necessary to definitely demonstrate the effectiveness of the oto- or neuroprotective agents proposed by animal and in vitro studies.
Collapse
Affiliation(s)
- Neife Aparecida Guinaim Dos Santos
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Rafaela Scalco Ferreira
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Antonio Cardozo Dos Santos
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
| |
Collapse
|