1
|
Xu S, Chen Y, Zhang L, Lu W, Chen X, Wang T, Wang W. Neuroprotective effects of arctigenin on cerebral ischemia-reperfusion injury in rats via the EPO/EPOR-JAK2-STAT5 signaling pathway. Front Pharmacol 2025; 16:1503971. [PMID: 40206088 PMCID: PMC11979258 DOI: 10.3389/fphar.2025.1503971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 02/20/2025] [Indexed: 04/11/2025] Open
Abstract
Introduction Cerebral ischemia-reperfusion injury (CIRI) is a complex pathophysiological process with significant morbidity and mortality, and there is no specific agent. Previous studies have found that arctigenin can play an anti-CIRI role through anti-inflammatory and antioxidant effects. This study further explored the anti-CIRI mechanism of arctigenin via the EPO/EPOR-JAK2-STAT5 signaling pathway. Methods TTC and H&E staining were used to observe infarct volume and morphological changes in the brain, RT-PCR was used to detect EPO, EPOR, HIF, JAK2, STAT5, NF-κB mRNA expression, EPO/EPOR ratio was detected by immunofluorescence, and HIF was observed by immunohistochemical staining. The protein expression levels of JAK2 and STAT5 were detected, and the protein expression levels of EPO, EPOR, HIF, JAK2 and STAT5 were detected by western blot. Results Our results indicate that arctigenin significantly reduced infarct volume and improved histopathological changes in the brain tissues from CIRI rats at 24 h, 48 h, and 72 h after reperfusion by TTC and H&E staining. RT-PCR analysis showed that arctigenin could significantly upregulate the mRNA expressions of EPO, EPOR, and HIF and downregulate the mRNA expressions of JAK2, STAT5, and NF-κB in the brain tissues from CIRI rats at 24 h, 48 h, and 72 h after reperfusion. Immunofluorescence assay showed that the ratio of EPO/EPOR in CIRI rats at 24 h, 48 h, and 72 h post-reperfusion was significantly elevated by arctigenin. Arctigenin could upregulate the HIF protein expression while downregulate the protein expressions of JAK2, STAT5, and NFκB in the brain tissues from CIRI rats at 24 h, 48 h, and 72 h after reperfusion by immunohistochemical staining. The protein regulation results of EPO, EPOR, HIF, JAK2, and STAT5 were also confirmed by Western blot at 72 h after reperfusion, consistent with the above results. Discussion In conclusion, arctigenin demonstrated neuroprotective properties against CIRI potentially through the EPO/EPOR-JAK2-STAT5 signaling pathway. These findings provide a scientific rationale for further exploration of arctigenin as a therapeutic agent for stroke.
Collapse
Affiliation(s)
- Shanshan Xu
- Department of Pharmacy, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, China
| | - Yuting Chen
- Department of Pharmacy, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, China
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Lingling Zhang
- Department of Pharmacy, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, China
| | - Wei Lu
- Department of Pharmacy, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, China
| | - Xu Chen
- Department of Pharmacy, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, China
| | - Ting Wang
- Department of Pharmacy, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, China
- School of Medicine, Taizhou University, Taizhou, Zhejiang, China
| | - Wenjie Wang
- Department of Pharmacy, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, China
| |
Collapse
|
2
|
Ningning Y, Ying X, Xiang L, Yue S, Zhongda W, Ruoyu J, Hanwen S, Weiwei T, Yafeng Z, Junjie M, Xiaolan C. Danggui-Shaoyao San alleviates cognitive impairment via enhancing HIF-1α/EPO axis in vascular dementia rats. JOURNAL OF ETHNOPHARMACOLOGY 2024; 331:118306. [PMID: 38723920 DOI: 10.1016/j.jep.2024.118306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/05/2024] [Accepted: 05/06/2024] [Indexed: 05/15/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Invigorating blood circulation to remove blood stasis is a primary strategy in TCM for treating vascular dementia (VaD). Danggui-Shaoyao San (DSS), as a traditional prescription for neuroprotective activity, has been proved to be effective in VaD treatment. However, its precise molecular mechanisms remain incompletely understood. AIM OF THE STUDY The specific mechanism underlying the therapeutic effects of DSS on VaD was explored by employing network pharmacology as well as in vivo and in viro experiment validation. MATERIALS AND METHODS We downloaded components of DSS from the BATMAN-TCM database for target prediction. The intersection between the components of DSS and targets, PPI network, as well as GO and KEGG enrichment analysis were then performed. Subsequently, the potential mechanism of DSS predicted by network pharmacology was assessed and validated through VaD rat model induced by 2VO operation and CoCl2-treated PC12 cells. Briefly, the DSS extract were first quantified by HPLC. Secondly, the effect of DSS on VaD was studied using MWM test, HE staining and TUNEL assay. Finally, the molecular mechanism of DSS against VaD was validated by Western blot and RT-QPCR experiments. RESULTS Through network analysis, 137 active ingredients were obtained from DSS, and 67 potential targets associated with DSS and VaD were identified. GO and KEGG analysis indicated that the action of DSS on VaD primarily involves hypoxic terms and HIF-1 pathway. In vivo validation, cognitive impairment and neuron mortality were markedly ameliorated by DSS. Additionally, DSS significantly reduced the expression of proteins related to synaptic plasticity and neuron apoptosis including PSD-95, SYP, Caspase-3 and BCL-2. Mechanistically, we confirmed DSS positively modulated the expression of HIF-1α and its downstream proteins including EPO, p-EPOR, STAT5, EPOR, and AKT1 in the hippocampus of VaD rats as well as CoCl2-induced PC12 cells. HIF-1 inhibitor YC-1 significantly diminished the protection of DSS on CoCl2-induced PC12 cell damage, with decreased HIF-1α, EPO, EPOR expression. CONCLUSION Our results initially demonstrated DSS could exert neuroprotective effects in VaD. The pharmacological mechanism of DSS may be related to its positive regulation on HIF-1α/EPO pathway.
Collapse
Affiliation(s)
- Yuan Ningning
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xu Ying
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Li Xiang
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Su Yue
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Wang Zhongda
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jiang Ruoyu
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Shi Hanwen
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Tao Weiwei
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhang Yafeng
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, 214071, China.
| | - Ma Junjie
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Cheng Xiaolan
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
3
|
Abdelrahman A, Mahmoud AA, Lamie Fanous Y, Abd Elhaliem NG, Elalaf H. Impact of erythropoietin and myoinositol versus metformin on insulin resistance in a rat model of polycystic ovary syndrome. Arch Physiol Biochem 2024; 130:1-12. [PMID: 34297646 DOI: 10.1080/13813455.2021.1949023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 06/23/2021] [Indexed: 10/20/2022]
Abstract
This study aimed to evaluate the therapeutic role of erythropoietin (EPO) or myoinositol versus metformin (MET) in improving the reproductive functions and glucose tolerance in a rat model of polycystic ovary (PCOS). Oral letrozole (LTZ) was used for induction of PCOS in wester rats for 21 days, after that, MET, EPO and myoinositol were administered for the following 21 days. The LTZ-induced PCOS rats have lost their oestrous cyclicity and become fixed at the diestrus phase, developed insulin resistance, abnormal sex and gonadotrophin hormone serum levels, increased cystic follicles, decreased number of the growing follicles and very little or no corpora lutea on microscopic examination, which were reversed by the three drugs, MET, EPO and myoinositol. MET and myoinositol were mostly equally effective in improving the reproductive manifestations of the disease. However, EPO was most effective in decreasing the insulin level observed in this LTZ-induced model of PCOS.
Collapse
Affiliation(s)
- Amany Abdelrahman
- Physiology Department, Faculty of Medicine, Sohag University, Sohag, Egypt
| | - Aida Abdeen Mahmoud
- Biochemistry Department, Faculty of Medicine, Sohag University, Sohag, Egypt
| | | | | | - Hassan Elalaf
- Physiology Department, Faculty of Medicine, Sohag University, Sohag, Egypt
| |
Collapse
|
4
|
Xu G, Dong F, Su L, Tan ZX, Lei M, Li L, Wen D, Zhang F. The role and therapeutic potential of nuclear factor κB (NF-κB) in ischemic stroke. Biomed Pharmacother 2024; 171:116140. [PMID: 38211425 DOI: 10.1016/j.biopha.2024.116140] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 01/13/2024] Open
Abstract
Stroke is a prevalent cerebrovascular condition with a global impact, causing significant rates of illness and death. Despite extensive research, the available treatment options for stroke remain restricted. Hence, it is crucial to gain a deeper understanding of the molecular mechanisms associated with the onset and advancement of stroke in order to establish a theoretical foundation for novel preventive and therapeutic approaches. NF-κB, also known as nuclear factor κB, is a transcription factor responsible for controlling the expression of numerous genes and plays a crucial role in diverse physiological processes. NF-κB is triggered and regulates neuroinflammation and other processes after stroke, promoting the generation of cytokine storms and contributing to the advancement of ischemic stroke (IS). Therefore, NF-κB could potentially play a vital role in stroke by regulating diverse pathophysiological processes. This review provides an overview of the functions of NF-κB in stroke and its governing mechanisms. In addition, our attention is directed towards various potential therapies that aim to inhibit the NF-κB signaling pathway in order to offer valuable insights for the advancement of innovative treatment approaches for stroke.
Collapse
Affiliation(s)
- Guangyu Xu
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China
| | - Fang Dong
- Department of Clinical Laboratory Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China
| | - Lei Su
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding 071000, PR China
| | - Zi-Xuan Tan
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China
| | - Mingcheng Lei
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China
| | - Lina Li
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China
| | - Di Wen
- College of Forensic Medicine, Hebei Medical University, Shijiazhuang 050017, PR China; Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang 050017, PR China; Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang 050017, PR China.
| | - Feng Zhang
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China.
| |
Collapse
|
5
|
Insight into the Effects of High-Altitude Hypoxic Exposure on Learning and Memory. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4163188. [PMID: 36160703 PMCID: PMC9492407 DOI: 10.1155/2022/4163188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 08/22/2022] [Indexed: 02/05/2023]
Abstract
The earth land area is heterogeneous in terms of elevation; about 45% of its land area belongs to higher elevation with altitude above 500 meters compared to sea level. In most cases, oxygen concentration decreases as altitude increases. Thus, high-altitude hypoxic stress is commonly faced by residents in areas with an average elevation exceeding 2500 meters and those who have just entered the plateau. High-altitude hypoxia significantly affects advanced neurobehaviors including learning and memory (L&M). Hippocampus, the integration center of L&M, could be the most crucial target affected by high-altitude hypoxia exposure. Based on these points, this review thoroughly discussed the relationship between high-altitude hypoxia and L&M impairment, in terms of hippocampal neuron apoptosis and dysfunction, neuronal oxidative stress disorder, neurotransmitters and related receptors, and nerve cell energy metabolism disorder, which is of great significance to find potential targets for medical intervention. Studies illustrate that the mechanism of L&M damaged by high-altitude hypoxia should be further investigated based on the entire review of issues related to this topic.
Collapse
|
6
|
Merelli A, Repetto M, Lazarowski A, Auzmendi J. Hypoxia, Oxidative Stress, and Inflammation: Three Faces of Neurodegenerative Diseases. J Alzheimers Dis 2021; 82:S109-S126. [PMID: 33325385 DOI: 10.3233/jad-201074] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The cerebral hypoxia-ischemia can induce a wide spectrum of biologic responses that include depolarization, excitotoxicity, oxidative stress, inflammation, and apoptosis, and result in neurodegeneration. Several adaptive and survival endogenous mechanisms can also be activated giving an opportunity for the affected cells to remain alive, waiting for helper signals that avoid apoptosis. These signals appear to help cells, depending on intensity, chronicity, and proximity to the central hypoxic area of the affected tissue. These mechanisms are present not only in a large list of brain pathologies affecting commonly older individuals, but also in other pathologies such as refractory epilepsies, encephalopathies, or brain trauma, where neurodegenerative features such as cognitive and/or motor deficits sequelae can be developed. The hypoxia inducible factor 1α (HIF-1α) is a master transcription factor driving a wide spectrum cellular response. HIF-1α may induce erythropoietin (EPO) receptor overexpression, which provides the therapeutic opportunity to administer pharmacological doses of EPO to rescue and/or repair affected brain tissue. Intranasal administration of EPO combined with other antioxidant and anti-inflammatory compounds could become an effective therapeutic alternative, to avoid and/or slow down neurodegenerative deterioration without producing adverse peripheral effects.
Collapse
Affiliation(s)
- Amalia Merelli
- Universidad de Buenos Aires, Facultad de Farmacia y Bioqummica, Departamento de Bioquímica Clínica, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Argentina
| | - Marisa Repetto
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Química General e Inorgánica; Instituto de Bioquímica y Medicina Molecular, Consejo Nacional de Investigaciones Científicas y Técnicas (IBIMOL, UBA-CONICET), Argentina
| | - Alberto Lazarowski
- Universidad de Buenos Aires, Facultad de Farmacia y Bioqummica, Departamento de Bioquímica Clínica, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Argentina
| | - Jerónimo Auzmendi
- Universidad de Buenos Aires, Facultad de Farmacia y Bioqummica, Departamento de Bioquímica Clínica, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| |
Collapse
|
7
|
Drug delivery platforms for neonatal brain injury. J Control Release 2021; 330:765-787. [PMID: 33417984 DOI: 10.1016/j.jconrel.2020.12.056] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/30/2020] [Accepted: 12/31/2020] [Indexed: 12/18/2022]
Abstract
Hypoxic-ischemic encephalopathy (HIE), initiated by the interruption of oxygenated blood supply to the brain, is a leading cause of death and lifelong disability in newborns. The pathogenesis of HIE involves a complex interplay of excitotoxicity, inflammation, and oxidative stress that results in acute to long term brain damage and functional impairments. Therapeutic hypothermia is the only approved treatment for HIE but has limited effectiveness for moderate to severe brain damage; thus, pharmacological intervention is explored as an adjunct therapy to hypothermia to further promote recovery. However, the limited bioavailability and the side-effects of systemic administration are factors that hinder the use of the candidate pharmacological agents. To overcome these barriers, therapeutic molecules may be packaged into nanoscale constructs to enable their delivery. Yet, the application of nanotechnology in infants is not well examined, and the neonatal brain presents unique challenges. Novel drug delivery platforms have the potential to magnify therapeutic effects in the damaged brain, mitigate side-effects associated with high systemic doses, and evade mechanisms that remove the drugs from circulation. Encouraging pre-clinical data demonstrates an attenuation of brain damage and increased structural and functional recovery. This review surveys the current progress in drug delivery for treating neonatal brain injury.
Collapse
|
8
|
Patel AMR, Apaijai N, Chattipakorn N, Chattipakorn SC. The Protective and Reparative Role of Colony-Stimulating Factors in the Brain with Cerebral Ischemia/Reperfusion Injury. Neuroendocrinology 2021; 111:1029-1065. [PMID: 33075777 DOI: 10.1159/000512367] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 10/19/2020] [Indexed: 11/19/2022]
Abstract
Stroke is a debilitating disease and has the ability to culminate in devastating clinical outcomes. Ischemic stroke followed by reperfusion entrains cerebral ischemia/reperfusion (I/R) injury, which is a complex pathological process and is associated with serious clinical manifestations. Therefore, the development of a robust and effective poststroke therapy is crucial. Granulocyte colony-stimulating factor (GCSF) and erythropoietin (EPO), originally discovered as hematopoietic growth factors, are versatile and have transcended beyond their traditional role of orchestrating the proliferation, differentiation, and survival of hematopoietic progenitors to one that fosters brain protection/neuroregeneration. The clinical indication regarding GCSF and EPO as an auspicious therapeutic strategy is conferred in a plethora of illnesses, including anemia and neutropenia. EPO and GCSF alleviate cerebral I/R injury through a multitude of mechanisms, involving antiapoptotic, anti-inflammatory, antioxidant, neurogenic, and angiogenic effects. Despite bolstering evidence from preclinical studies, the multiple brain protective modalities of GCSF and EPO failed to translate in clinical trials and thereby raises several questions. The present review comprehensively compiles and discusses key findings from in vitro, in vivo, and clinical data pertaining to the administration of EPO, GCSF, and other drugs, which alter levels of colony-stimulating factor (CSF) in the brain following cerebral I/R injury, and elaborates on the contributing factors, which led to the lost in translation of CSFs from bench to bedside. Any controversial findings are discussed to enable a clear overview of the role of EPO and GCSF as robust and effective candidates for poststroke therapy.
Collapse
Affiliation(s)
- Aysha Mohamed Rafik Patel
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Nattayaporn Apaijai
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand,
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand,
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand,
| |
Collapse
|
9
|
Insights into Potential Targets for Therapeutic Intervention in Epilepsy. Int J Mol Sci 2020; 21:ijms21228573. [PMID: 33202963 PMCID: PMC7697405 DOI: 10.3390/ijms21228573] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/04/2020] [Accepted: 11/11/2020] [Indexed: 02/06/2023] Open
Abstract
Epilepsy is a chronic brain disease that affects approximately 65 million people worldwide. However, despite the continuous development of antiepileptic drugs, over 30% patients with epilepsy progress to drug-resistant epilepsy. For this reason, it is a high priority objective in preclinical research to find novel therapeutic targets and to develop effective drugs that prevent or reverse the molecular mechanisms underlying epilepsy progression. Among these potential therapeutic targets, we highlight currently available information involving signaling pathways (Wnt/β-catenin, Mammalian Target of Rapamycin (mTOR) signaling and zinc signaling), enzymes (carbonic anhydrase), proteins (erythropoietin, copine 6 and complement system), channels (Transient Receptor Potential Vanilloid Type 1 (TRPV1) channel) and receptors (galanin and melatonin receptors). All of them have demonstrated a certain degree of efficacy not only in controlling seizures but also in displaying neuroprotective activity and in modifying the progression of epilepsy. Although some research with these specific targets has been done in relation with epilepsy, they have not been fully explored as potential therapeutic targets that could help address the unsolved issue of drug-resistant epilepsy and develop new antiseizure therapies for the treatment of epilepsy.
Collapse
|
10
|
Li J, Wu DM, Yu Y, Deng SH, Liu T, Zhang T, He M, Zhao YY, Xu Y. Amifostine ameliorates induction of experimental autoimmune encephalomyelitis: Effect on reactive oxygen species/NLRP3 pathway. Int Immunopharmacol 2020; 88:106998. [PMID: 33182064 DOI: 10.1016/j.intimp.2020.106998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 09/04/2020] [Accepted: 09/08/2020] [Indexed: 10/23/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune disease for which conventional treatments have limited efficacy or side effects. Free radicals are primarily involved in blood-brain barrier disruption and induce neuronal and axonal damage, thus promoting the development of MS. Amifostine, a radioprotective drug used as a cytoprotective agent, attenuates oxidative stress and improves radiation damage by acting as a direct scavenger of reactive oxygen and nitrogen species. The aim of this study was to evaluate the effects of amifostine on MS in a mouse model of experimental autoimmune encephalomyelitis (EAE), which was developed by immunizing C57BL/6 mice with myelin oligodendrocyte glycoprotein and pertussis toxin. EAE mice received intraperitoneal injections of amifostine prior to onset of clinical symptoms and were monitored up to day 15 post induction. We observed abnormal clinical behavioral scores and a decrease in body weight. Histological analysis showed severe inflammatory infiltration and demyelination in the brain and spinal cord lumbar enlargements where significant upregulation of the mRNA expression of the pro-inflammatory cytokines interleukin-6 and interleukin-8, downregulation of the anti-inflammatory cytokine interleukin-10, and obvious microgliosis were also observed. Amifostine treatment potently reversed these abnormal changes. The anti-inflammatory effect of amifostine was associated with the inhibition of reactive oxygen species generation. Furthermore, the expression of proteins involved in the NLRP3 signaling pathway and pyroptosis was decreased. In conclusion, our study showed that amifostine ameliorates induction of experimental autoimmune encephalomyelitis via anti-inflammatory and anti-pyroptosis effects, providing further insights into the use of amifostine for the treatment of MS.
Collapse
Affiliation(s)
- Jing Li
- Clinical Laboratory, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, 278 Baoguang Road, Chengdu, Sichuan 610500, PR China; Collaborative Innovation Center of Sichuan for Elderly Care and Health of Chengdu Medical College, Baoguang Road, Chengdu, Sichuan 610041, PR China
| | - Dong-Ming Wu
- Clinical Laboratory, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, 278 Baoguang Road, Chengdu, Sichuan 610500, PR China; Collaborative Innovation Center of Sichuan for Elderly Care and Health of Chengdu Medical College, Baoguang Road, Chengdu, Sichuan 610041, PR China
| | - Ye Yu
- Clinical Laboratory, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, 278 Baoguang Road, Chengdu, Sichuan 610500, PR China; Collaborative Innovation Center of Sichuan for Elderly Care and Health of Chengdu Medical College, Baoguang Road, Chengdu, Sichuan 610041, PR China
| | - Shi-Hua Deng
- Clinical Laboratory, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, 278 Baoguang Road, Chengdu, Sichuan 610500, PR China; Collaborative Innovation Center of Sichuan for Elderly Care and Health of Chengdu Medical College, Baoguang Road, Chengdu, Sichuan 610041, PR China
| | - Teng Liu
- Clinical Laboratory, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, 278 Baoguang Road, Chengdu, Sichuan 610500, PR China; Collaborative Innovation Center of Sichuan for Elderly Care and Health of Chengdu Medical College, Baoguang Road, Chengdu, Sichuan 610041, PR China
| | - Ting Zhang
- Clinical Laboratory, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, 278 Baoguang Road, Chengdu, Sichuan 610500, PR China; Collaborative Innovation Center of Sichuan for Elderly Care and Health of Chengdu Medical College, Baoguang Road, Chengdu, Sichuan 610041, PR China
| | - Miao He
- Clinical Laboratory, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, 278 Baoguang Road, Chengdu, Sichuan 610500, PR China; Collaborative Innovation Center of Sichuan for Elderly Care and Health of Chengdu Medical College, Baoguang Road, Chengdu, Sichuan 610041, PR China
| | - Yang-Yang Zhao
- Clinical Laboratory, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, 278 Baoguang Road, Chengdu, Sichuan 610500, PR China; Collaborative Innovation Center of Sichuan for Elderly Care and Health of Chengdu Medical College, Baoguang Road, Chengdu, Sichuan 610041, PR China
| | - Ying Xu
- Clinical Laboratory, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, 278 Baoguang Road, Chengdu, Sichuan 610500, PR China; Collaborative Innovation Center of Sichuan for Elderly Care and Health of Chengdu Medical College, Baoguang Road, Chengdu, Sichuan 610041, PR China.
| |
Collapse
|