1
|
Sun J, Yan T, Wang X, Li Y, Yang J. Signaling Transduction Network Elucidation of ACE 2 Regulating Apostichopus japonicus Autolysis by Using Integrative TMT Proteomics and Transcriptomics. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:2197-2214. [PMID: 39786724 DOI: 10.1021/acs.jafc.4c10292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
This study aims to reveal the transduction signaling network that triggers sea cucumber (Apostichopus japonicus) autolysis. The tandem mass tag (TMT) proteomics and transcriptomic techniques were used to analyze expression differences between inhibited and activated sea cucumber autolysis. Flow cytometry was used to identify apoptosis. Western blotting and RT-PCR verified the signaling pathway. The results showed that the angiotensin-converting enzyme 2 (ACE 2) activator (diminazene, DIZE) maintained the health of A. japonicus. The ACE 2 inhibitor (captopril, Capt) accelerated the autolysis. Based on the multiomics analysis, the ACE 2 activator activated the downstream NF-κB pathway to prevent the sea cucumber apoptosis. The Capt activated apoptosis initiation. Apoptosis occurred through the regulation of TNF and PI3K-Akt signaling pathways, which downregulate NF-κB. Akt was identified as an intermediate signaling protein downstream of ACE 2 that regulates autolysis in A. japonicus. Compared to A. japonicus in the ultraviolet-irradiated and Capt groups, the Akt inhibitor (perifosine, KRX) significantly reduced the expression of the PI3K-Akt and NF-κB pathways, thereby inhibiting the autolysis process. Additionally, DIZE attenuated ROS levels, thereby inhibiting the autolysis of A. japonicus. This study provides better insight into the autolysis mechanism of A. japonicus.
Collapse
Affiliation(s)
- Jinghe Sun
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Tingting Yan
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Xiaoyan Wang
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Yimeng Li
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Jingfeng Yang
- National & Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application, Dalian Polytechnic University, Dalian 116034, PR China
- SKL of Marine Food Processing & Safety Control, School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, China
| |
Collapse
|
2
|
Yang L, Hou H, Lu L, Sun Y, Chen R, Deng Q, Chen H. Effects of natural source polysaccharides on neurological diseases: A review. Int J Biol Macromol 2025; 296:139697. [PMID: 39805435 DOI: 10.1016/j.ijbiomac.2025.139697] [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: 11/19/2024] [Revised: 01/05/2025] [Accepted: 01/07/2025] [Indexed: 01/16/2025]
Abstract
With the aging of society and changes in lifestyle, the incidence of neurological diseases (NDs) has been increasing year by year, bringing a heavy burden to patients and society. Although the efficacy of chemical drugs in the treatment of NDs is remarkable, there are problems such as high side effects and high costs. Therefore, finding mild and efficient drugs for NDs treatment has become an urgent clinical need. Natural source polysaccharides (NSPs) are macromolecules with unique bioactivity and low toxicity characteristics, which have great potential to become novel therapeutic agents for NDs. In the present study, the pharmacological activities and potential molecular mechanisms of NSPs to alleviate NDs are systematically reviewed from the perspectives of inflammation, oxidative stress, apoptosis, neuronal cell autophagy, neurotoxicity, and sedation-hypnosis. In addition, the limitations of the existing studies were analyzed and discussed, and the future research direction was suggested. This study may provide scientific basis for the research and development of therapeutic agents for NDs based on NSPs.
Collapse
Affiliation(s)
- Luyuan Yang
- Key Laboratory for Information System of Mountainous Areas and Protection of Ecological Environment, Guizhou Normal University, Guiyang 550001, China; Guizhou Engineering Laboratory for Quality Control & Evaluation Technology of Medicine, Guizhou Normal University, Guiyang 550001, China
| | - Hailu Hou
- Key Laboratory for Information System of Mountainous Areas and Protection of Ecological Environment, Guizhou Normal University, Guiyang 550001, China; Guizhou Engineering Laboratory for Quality Control & Evaluation Technology of Medicine, Guizhou Normal University, Guiyang 550001, China
| | - Liping Lu
- Guizhou Dalong Pharmaceutical Co., Ltd., Guiyang 550001, China
| | - Yu Sun
- Key Laboratory for Information System of Mountainous Areas and Protection of Ecological Environment, Guizhou Normal University, Guiyang 550001, China; Guizhou Engineering Laboratory for Quality Control & Evaluation Technology of Medicine, Guizhou Normal University, Guiyang 550001, China
| | - Ruhai Chen
- Key Laboratory for Information System of Mountainous Areas and Protection of Ecological Environment, Guizhou Normal University, Guiyang 550001, China; Guizhou Engineering Laboratory for Quality Control & Evaluation Technology of Medicine, Guizhou Normal University, Guiyang 550001, China
| | - Qingfang Deng
- Key Laboratory for Information System of Mountainous Areas and Protection of Ecological Environment, Guizhou Normal University, Guiyang 550001, China; Guizhou Engineering Laboratory for Quality Control & Evaluation Technology of Medicine, Guizhou Normal University, Guiyang 550001, China
| | - Huaguo Chen
- Key Laboratory for Information System of Mountainous Areas and Protection of Ecological Environment, Guizhou Normal University, Guiyang 550001, China; Guizhou Engineering Laboratory for Quality Control & Evaluation Technology of Medicine, Guizhou Normal University, Guiyang 550001, China.
| |
Collapse
|
3
|
Jiang Y, Li R, Ban Y, Zhang W, Kong N, Tang J, Ma B, Shao Y, Jin R, Sun L, Yue H, Zhang H. EPO modified MSCs protects SH-SY5Y cells against ischemia/hypoxia-induced apoptosis via REST-dependent epigenetic remodeling. Sci Rep 2024; 14:23252. [PMID: 39370424 PMCID: PMC11456618 DOI: 10.1038/s41598-024-74261-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 09/24/2024] [Indexed: 10/08/2024] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a diffuse brain tissue injury caused by acute ischemia and hypoxia, and it is most commonly found in newborn infants but can also occur in adults. Mesenchymal stem cell (MSC) therapies have showed improved outcomes for treating HIE-induced neuronal defects. However, many key issues associated with poor cell viability and tolerance of grafted MSCs after HIE remain to be resolved. Genetic engineering could endow MSCs with more robust regenerative capacities. Our research, along with that of other scientists, has found that the expression of intracellular erythropoietin (EPO) in human umbilical cord MSCs (hUC-MSCs) increases proportionally with the duration of hypoxia exposure. Furthermore, we observed that EPO, when introduced into the EPO gene-modified hUC-MSCs, can be secreted into the extracellular space. However, the underlying mechanisms that support the neuroprotective effects of EPO-MSCs remain unclear. EPO-MSCs, hUC-MSCs, and NC-MSCs were identified by flow cytometry, osteogenic, and adipogenic differentiation assays. The oxygen-glucose deprivation (OGD)-induced SH-SY5Y cell-line was established, and five groups were set up: control, 24-h ischemia-hypoxia, co-cultured with hUC-MSCs, NC-MSCs, and EPO-MSCs after hypoxia. LEGENDplex™ multi-factor flow cytometry was used to detect the secretion of inflammatory factors in cell supernatants and cerebrospinal fluid. Chromosome-targeted excision and tagging (CUT&Tag) sequencing was applied to detect genomic H3K4me2 modifications, and conjoint analysis with transcriptome sequencing (RNA-seq) was performed. Lentiviral vector infection was used to construct SH-SY5Y cells with stable knockdown of RE1-silencing transcription factor (REST), and flow cytometry was used to detect alterations in apoptosis. Finally, the molecular mechanism underlying the neuroprotective and anti-apoptotic effects of EPO-MSCs was investigated using RNA sequencing, qRT-PCR, and western blot assays. Our results suggest that EPO-MSCs are genetically engineered to secrete significantly more EPO. EPO-MSCs treatment has anti-apoptotic properties and offers neuronal protection during ischemic-hypoxic injury. Furthermore, RNA-seq results suggest that multiple inflammation-related genes were down-regulated after EPO-MSCs treatment. Application of RNA-seq and CUT&Tag combined analysis found that the expressions of REST were significantly up-regulated. Lentiviral vector infection to construct REST knockdown SH-SY5Y failed to rescue apoptosis after hypoxia and co-culture with EPO-MSCs, and SETD2-mediated H3K36me3 protein level expression was reduced. EPO-MSCs may promote neuronal survival by affecting H3K4me2 and thus activating the expression of REST and TET3. EPO-MSCs also upregulated the modification level of SETD2-mediated H3K36me3 and regulated the expression of inflammation-related genes such as PLCG2, as well as apoptosis genes BCL2A1. To investigate the neuroprotective effects of EPO-modified hUC-MSCs and the underlying epigenetic regulatory mechanisms, this study aims to provide a theoretical foundation for the potential application of EPO gene-modified hUC-MSCs in the treatment of HIE.
Collapse
Affiliation(s)
- Yu Jiang
- Department of Neurosurgery, Zhengzhou Central Hospital Affiliated Zhengzhou University, Zhengzhou, 450000, China
| | - Ruibo Li
- Department of Neurosurgery, Zhengzhou Central Hospital Affiliated Zhengzhou University, Zhengzhou, 450000, China
| | - Yueyao Ban
- Department of Neurosurgery, Zhengzhou Central Hospital Affiliated Zhengzhou University, Zhengzhou, 450000, China
| | - Wenjin Zhang
- Department of Neurosurgery, Zhengzhou Central Hospital Affiliated Zhengzhou University, Zhengzhou, 450000, China
| | - Ning Kong
- Department of Neurosurgery, Zhengzhou Central Hospital Affiliated Zhengzhou University, Zhengzhou, 450000, China
| | - Jixiang Tang
- Department of Neurosurgery, Zhengzhou Central Hospital Affiliated Zhengzhou University, Zhengzhou, 450000, China
| | - Baodong Ma
- Department of Neurosurgery, Zhengzhou Central Hospital Affiliated Zhengzhou University, Zhengzhou, 450000, China
| | - Yiming Shao
- Department of Neurosurgery, Zhengzhou Central Hospital Affiliated Zhengzhou University, Zhengzhou, 450000, China
| | - Ranran Jin
- Department of Neurosurgery, Zhengzhou Central Hospital Affiliated Zhengzhou University, Zhengzhou, 450000, China
| | - Lei Sun
- Department of Neurosurgery, Zhengzhou Central Hospital Affiliated Zhengzhou University, Zhengzhou, 450000, China.
| | - Han Yue
- Stem Cell Research Center, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, Henan, China.
| | - Hui Zhang
- Department of Neurosurgery, Zhengzhou Central Hospital Affiliated Zhengzhou University, Zhengzhou, 450000, China.
| |
Collapse
|
4
|
Kang K, Wu Y, Gan H, Yang B, Xiao H, Wang D, Qiu H, Dong X, Tang H, Zhai X. Pathophysiological mechanisms underlying the development of focal cortical dysplasia and their association with epilepsy: Experimental models as a research approach. Seizure 2024; 121:176-185. [PMID: 39191070 DOI: 10.1016/j.seizure.2024.08.013] [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: 03/03/2024] [Revised: 08/16/2024] [Accepted: 08/16/2024] [Indexed: 08/29/2024] Open
Abstract
Focal cortical dysplasia (FCD) is a structural lesion that is the most common anatomical lesion identified in children, and the second most common in adults with drug-resistant focal-onset epilepsy. These lesions vary in size, location, and histopathological manifestations. FCDs are classified into three subtypes associated with loss-of-function mutations in PI3K/AKT, TSC1/TSC2, RHEB, and DEPDC/NPRL2/NPRL3. During the decades of research into FCD, experimental models have played an irreplaceable role in the research design of studies investigating disease pathogenesis, pathophysiology, and treatment. Further, the establishment of FCD experimental models has moved the field forward by (1) revealing the cellular processes and signaling pathways underlying FCD pathogenesis and (2) varying the methods and materials to study the function of FCD proteins. Currently, FCD experimental models are predominantly murine, with each model providing unique insights into FCD lesions. This review briefly summarizes the pathology and molecular functions of FCD, further comparing the available modeling methods and indexes, as well as the utilization of models, followed by an analysis of the similarities, advantages, and disadvantages between these models and human FCD.
Collapse
Affiliation(s)
- Kaiyi Kang
- Department of Neurosurgery, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing 400000, China
| | - Yuxin Wu
- Department of Neurosurgery, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing 400000, China
| | - Hui Gan
- Department of Neurosurgery, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing 400000, China
| | - Baohui Yang
- Department of Neurosurgery, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing 400000, China; Department of Neurosurgery, Laboratory of Neurosurgery, Institute of Neurology, Lanzhou University, Lanzhou 730000, China
| | - Han Xiao
- Department of Neurosurgery, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing 400000, China
| | - Difei Wang
- Department of Neurosurgery, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing 400000, China
| | - Hanli Qiu
- Department of Neurosurgery, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing 400000, China
| | - Xinyu Dong
- Department of Neurosurgery, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing 400000, China
| | - Haotian Tang
- Department of Neurosurgery, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing 400000, China
| | - Xuan Zhai
- Department of Neurosurgery, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing 400000, China.
| |
Collapse
|
5
|
Wang Y, Ping Z, Gao H, Liu Z, Xv Q, Jiang X, Yu W. LYC inhibits the AKT signaling pathway to activate autophagy and ameliorate TGFB-induced renal fibrosis. Autophagy 2024; 20:1114-1133. [PMID: 38037248 PMCID: PMC11135866 DOI: 10.1080/15548627.2023.2287930] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 11/17/2023] [Accepted: 11/20/2023] [Indexed: 12/02/2023] Open
Abstract
Renal fibrosis is a typical pathological change in chronic kidney disease (CKD). Epithelial-mesenchymal transition (EMT) is the predominant stage. Activation of macroautophagy/autophagy plays a crucial role in the process of EMT. Lycopene (LYC) is a highly antioxidant carotenoid with pharmacological effects such as anti-inflammation, anti-apoptosis and mediation of autophagy. In this study, we demonstrated the specific mechanism of LYC in activating mitophagy and improving renal fibrosis. The enrichment analysis results of GO and KEGG showed that LYC had high enrichment values with autophagy. In this study, we showed that LYC alleviated aristolochic acid I (AAI)-induced intracellular expression of PINK1, TGFB/TGF-β, p-SMAD2, p-SMAD3, and PRKN/Parkin, recruited expression of MAP1LC3/LC3-II and SQSTM1/p62, decreased mitochondrial membrane potential (MMP), and ameliorated renal fibrosis in mice. When we simultaneously intervened NRK52E cells using bafilomycin A1 (Baf-A1), AAI, and LYC, intracellular MAP1LC3-II and SQSTM1 expression was significantly increased. A similar result was seen in renal tissue and cells when treated in vitro and in vivo with CQ, AAI, and LYC, and the inhibitory effect of LYC on the AAI-activated SMAD2-SMAD3 signaling pathway was attenuated. Molecular docking simulation experiments showed that LYC stably bound to the AKT active site. After intervention of cells with AAI and GSK-690693, the expression of PINK1, PRKN, MAP1LC3-II, BECN1, p-SMAD2 and p-SMAD3 was increased, and the expression of SQSTM1 was decreased. However, SC79 inhibited autophagy and reversed the inhibitory effect of LYC on EMT. The results showed that LYC could inhibit the AKT signaling pathway to activate mitophagy and reduce renal fibrosis.Abbreviation: AA: aristolochic acid; ACTA2/α-SMA: actin alpha 2, smooth muscle, aorta; ACTB: actin beta; AKT/protein kinase B: thymoma viral proto-oncogene; BAF-A1: bafilomycin A1; BECN1: beclin 1, autophagy related; CCN2/CTGF: cellular communication network factor 2; CDH1/E-Cadherin: cadherin 1; CKD: chronic kidney disease; COL1: collagen, type I; COL3: collagen, type III; CQ: chloroquine; ECM: extracellular matrix; EMT: epithelial-mesenchymal transition; FN1: fibronectin 1; LYC: lycopene; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; MMP: mitochondrial membrane potential; MTOR: mechanistic target of rapamycin kinase ; PI3K: phosphoinositide 3-kinase; PINK1: PTEN induced putative kinase 1; PRKN/Parkin: parkin RBR E3 ubiquitin protein ligase; PPI: protein-protein interaction; SMAD2: SMAD family member 2; SMAD3: SMAD family member 3; SQSTM1/p62: sequestosome 1; TGFB/TGFβ: transforming growth factor, beta; VIM: vimentin.
Collapse
Affiliation(s)
- Yu Wang
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Zhenlei Ping
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Hongxin Gao
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Zhihui Liu
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Qingyang Xv
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xiaowen Jiang
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Wenhui Yu
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Institute of Chinese Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Key Laboratory of Animal Pathogenesis and Comparative Medicine in Heilongjiang Province, Northeast Agricultural University, Harbin, China
| |
Collapse
|
6
|
Turkistani A, Al-kuraishy HM, Al-Gareeb AI, Albuhadily AK, Elhussieny O, AL-Farga A, Aqlan F, Saad HM, Batiha GES. The functional and molecular roles of p75 neurotrophin receptor (p75 NTR) in epilepsy. J Cent Nerv Syst Dis 2024; 16:11795735241247810. [PMID: 38655152 PMCID: PMC11036928 DOI: 10.1177/11795735241247810] [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: 12/08/2023] [Accepted: 03/14/2024] [Indexed: 04/26/2024] Open
Abstract
Epilepsy is a chronic neurological disorder manifested by recurring unprovoked seizures resulting from an imbalance in the inhibitory and excitatory neurotransmitters in the brain. The process of epileptogenesis involves a complex interplay between the reduction of inhibitory gamma-aminobutyric acid (GABA) and the enhancement of excitatory glutamate. Pro-BDNF/p75NTR expression is augmented in both glial cells and neurons following epileptic seizures and status epileptics (SE). Over-expression of p75NTR is linked with the pathogenesis of epilepsy, and augmentation of pro-BDNF/p75NTR is implicated in the pathogenesis of epilepsy. However, the precise mechanistic function of p75NTR in epilepsy has not been completely elucidated. Therefore, this review aimed to revise the mechanistic pathway of p75NTR in epilepsy.
Collapse
Affiliation(s)
- Areej Turkistani
- Department of pharmacology and toxicology, Collage of Medicine, Taif University, Taif, Kingdom of Saudi
| | - Hayder M. Al-kuraishy
- Professor in department of clinical pharmacology and medicine, college of medicine, Mustansiriyah University, Baghdad, Iraq
| | - Ali I. Al-Gareeb
- Professor in department of clinical pharmacology and medicine, college of medicine, Mustansiriyah University, Baghdad, Iraq
| | - Ali K. Albuhadily
- Professor in department of clinical pharmacology and medicine, college of medicine, Mustansiriyah University, Baghdad, Iraq
| | - Omnya Elhussieny
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matruh, Egypt
| | - Ammar AL-Farga
- Biochemistry Department, College of Sciences, University of Jeddah, Jeddah, Saudia Arbia
| | - Faisal Aqlan
- Department of Chemistry, College of Sciences, Ibb University, Ibb Governorate, Yemen
| | - Hebatallah M. Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Matrouh, Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| |
Collapse
|
7
|
Chen Y, Guan W, Wang ML, Lin XY. PI3K-AKT/mTOR Signaling in Psychiatric Disorders: A Valuable Target to Stimulate or Suppress? Int J Neuropsychopharmacol 2024; 27:pyae010. [PMID: 38365306 PMCID: PMC10888523 DOI: 10.1093/ijnp/pyae010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 02/08/2024] [Indexed: 02/18/2024] Open
Abstract
Economic development and increased stress have considerably increased the prevalence of psychiatric disorders in recent years, which rank as some of the most prevalent diseases globally. Several factors, including chronic social stress, genetic inheritance, and autogenous diseases, lead to the development and progression of psychiatric disorders. Clinical treatments for psychiatric disorders include psychotherapy, chemotherapy, and electric shock therapy. Although various achievements have been made researching psychiatric disorders, the pathogenesis of these diseases has not been fully understood yet, and serious adverse effects and resistance to antipsychotics are major obstacles to treating patients with psychiatric disorders. Recent studies have shown that the mammalian target of rapamycin (mTOR) is a central signaling hub that functions in nerve growth, synapse formation, and plasticity. The PI3K-AKT/mTOR pathway is a critical target for mediating the rapid antidepressant effects of these pharmacological agents in clinical and preclinical research. Abnormal PI3K-AKT/mTOR signaling is closely associated with the pathogenesis of several neurodevelopmental disorders. In this review, we focused on the role of mTOR signaling and the related aberrant neurogenesis in psychiatric disorders. Elucidating the neurobiology of the PI3K-AKT/mTOR signaling pathway in psychiatric disorders and its actions in response to antidepressants will help us better understand brain development and quickly identify new therapeutic targets for the treatment of these mental illnesses.
Collapse
Affiliation(s)
- Yan Chen
- Department of Neurology, Nantong Third People’s Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, Jiangsu, China
| | - Wei Guan
- Department of Pharmacology, Pharmacy College, Nantong University, Nantong, Jiangsu, China
| | - Mei-Lan Wang
- Department of Neurology, Nantong Third People’s Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, Jiangsu, China
| | - Xiao-Yun Lin
- Department of Neurology, Nantong Third People’s Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
8
|
Xiao Y, Yang J, Deng Y, Zhang L, Xu Q, Li H. Tartary buckwheat protein-derived peptide AFYRW alleviates H 2O 2-induced vascular injury via the PI3K/AKT/NF-κB pathway. Prostaglandins Other Lipid Mediat 2023; 169:106768. [PMID: 37597762 DOI: 10.1016/j.prostaglandins.2023.106768] [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: 07/02/2023] [Revised: 08/09/2023] [Accepted: 08/14/2023] [Indexed: 08/21/2023]
Abstract
Tartary buckwheat protein-derived peptide (Ala-Phe-Tyr-Arg-Trp, AFYRW) is a natural active peptide that hampers the atherosclerosis process, but the underlying role of AFYRW in angiogenesis remains unknown. Here, we present a system-based study to evaluate the effects of AFYRW on H2O2-induced vascular injury in human umbilical vein endothelial cells (HUVECs). HUVECs were co-incubated with H2O2 for 2 h in the vascular injury model, and AFYRW was added 24 h in advance to investigate the protective mechanism of vascular injury. We identified that AFYRW inhibits oxidative stress, cell migration, cell invasion, and angiogenesis in H2O2-treated HUVECs. In addition, we found H2O2-induced upregulation of phosphoinositide 3-kinase (PI3K), protein kinase B (AKT), phosphorylation of nuclear factor-κB (NF-κB) p65 and nuclear translocation of NF-κB decreased by AFYRW. Taken together, AFYRW attenuated H2O2-induced vascular injury through the PI3K/AKT/NF-κB pathway. Thereby, AFYRW may serve as a therapeutic option for vascular injuries.
Collapse
Affiliation(s)
- Yi Xiao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Guizhou Medical University, Guiyang 550004, PR China
| | - Jiajun Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Guizhou Medical University, Guiyang 550004, PR China; Key Laboratory of Endemic and Ethenic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang 550004, Guizhou, China
| | - Yan Deng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Guizhou Medical University, Guiyang 550004, PR China
| | - Lilin Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Guizhou Medical University, Guiyang 550004, PR China
| | - Qingzhong Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Guizhou Medical University, Guiyang 550004, PR China
| | - Hongmei Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Guizhou Medical University, Guiyang 550004, PR China; Key Laboratory of Endemic and Ethenic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang 550004, Guizhou, China.
| |
Collapse
|
9
|
Su X, Huang L, Li S, Ying J, Zhao F, Wang S, Liu Q, Qu Y, Mu D. The RNA m6A modification might participate in microglial activation during hypoxic-ischemic brain damage in neonatal mice. Hum Genomics 2023; 17:78. [PMID: 37626401 PMCID: PMC10463984 DOI: 10.1186/s40246-023-00527-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND The RNA m6A modification has been implicated in multiple neurological diseases as well as macrophage activation. However, whether it regulates microglial activation during hypoxic-ischemic brain damage (HIBD) in neonates remains unknown. Here, we aim to examine whether the m6A modification is involved in modulating microglial activation during HIBD. We employed an oxygen and glucose deprivation microglial model for in vitro studies and a neonatal mouse model of HIBD. The brain tissue was subjected to RNA-seq to screen for significant changes in the mRNA m6A regulator. Thereafter, we performed validation and bioinformatics analysis of the major m6A regulators. RESULTS RNA-seq analysis revealed that, among 141 m6A regulators, 31 exhibited significant differential expression (FC (abs) ≥ 2) in HIBD mice. We then subjected the major m6A regulators Mettl3, Mettl14, Fto, Alkbh5, Ythdf1, and Ythdf2 to further validation, and the results showed that all were significantly downregulated in vitro and in vivo. GO analysis reveals that regulators are mainly involved in the regulation of cellular and metabolic processes. The KEGG results indicate the involvement of the signal transduction pathway. CONCLUSIONS Our findings demonstrate that m6A modification of mRNA plays a crucial role in the regulation of microglial activation in HIBD, with m6A-associated regulators acting as key modulators of microglial activation.
Collapse
Affiliation(s)
- Xiaojuan Su
- Department of Pediatrics/Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Lingyi Huang
- Department of Pediatrics/Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- West China College of Stomatology/State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, 610041, China
| | - Shiping Li
- Department of Pediatrics/Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Junjie Ying
- Department of Pediatrics/Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Fengyan Zhao
- Department of Pediatrics/Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Shaopu Wang
- Department of Pediatrics/Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Qian Liu
- Department of Pediatrics/Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Yi Qu
- Department of Pediatrics/Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Dezhi Mu
- Department of Pediatrics/Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
10
|
Maász A, Bodó T, Till Á, Molnár G, Masszi G, Labossa G, Herbert Z, Bene J, Hadzsiev K. Three-Year Follow-Up after Intrauterine mTOR Inhibitor Administration for Fetus with TSC-Associated Rhabdomyoma. Int J Mol Sci 2023; 24:12886. [PMID: 37629066 PMCID: PMC10454323 DOI: 10.3390/ijms241612886] [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: 06/30/2023] [Revised: 08/14/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
Tuberous sclerosis complex (TSC) is a multisystem disorder characterized by seizures, neuropsychiatric disorders, and tumors of the heart, brain, skin, lungs, and kidneys. We present a three-year follow-up of a patient with TSC-associated rhabdomyoma detected in utero. Genetic examination of the fetus and the parents revealed a de novo variant in the TSC2 gene (c.3037delG, p.Asp1013IlefsTer3). Oral everolimus was initiated in the pregnant mother to regress the fetal tumor, which was successful. To the best of our knowledge, there is very little information regarding the use of everolimus therapy during pregnancy. West-syndrome was diagnosed when the proband was four months old. The symptoms were well-manageable, however temporarily. Therapy-resistant focal seizures were frequent. The patient had good vitals and was under regular cardiological control, showed a balanced circulation, and did not require any medication. Subependymal giant cell astrocytoma (SEGA) identified by regular neuroimaging examinations remained unchanged, which may be a consequence of early intrauterine treatment. Early detection of the pathogenic TSC2 variant, followed by in utero administration of everolimus and early vigabatrin therapy, allowed the detection of a milder developmental delay of the proband. Our study emphasizes how early genetic testing and management of epilepsy are pivotal for proper neurodevelopmental impacts and therapeutic strategies.
Collapse
Affiliation(s)
- Anita Maász
- Department of Medical Genetics, Medical School and Clinical Centre, University of Pécs, H-7624 Pécs, Hungary
| | - Tímea Bodó
- Bethesda Children’s Hospital, H-1146 Budapest, Hungary
| | - Ágnes Till
- Department of Medical Genetics, Medical School and Clinical Centre, University of Pécs, H-7624 Pécs, Hungary
| | - Gábor Molnár
- Department of Obstetrics and Gynaecology, Medical School and Clinical Centre, University of Pécs, H-7624 Pécs, Hungary
| | - György Masszi
- Department of Paediatrics, Medical School and Clinical Centre, University of Pécs, H-7624 Pécs, Hungary
| | - Gusztáv Labossa
- Department of Obstetrics and Gynaecology, Medical School and Clinical Centre, University of Pécs, H-7624 Pécs, Hungary
| | - Zsuzsanna Herbert
- Department of Medical Imaging, Medical School and Clinical Centre, University of Pécs, H-7624 Pécs, Hungary
| | - Judit Bene
- Department of Medical Genetics, Medical School and Clinical Centre, University of Pécs, H-7624 Pécs, Hungary
| | - Kinga Hadzsiev
- Department of Medical Genetics, Medical School and Clinical Centre, University of Pécs, H-7624 Pécs, Hungary
| |
Collapse
|
11
|
Shan Y, Chen Y, Gu H, Wang Y, Sun Y. Regulatory Basis of Adipokines Leptin and Adiponectin in Epilepsy: from Signaling Pathways to Glucose Metabolism. Neurochem Res 2023; 48:2017-2028. [PMID: 36797447 PMCID: PMC10181973 DOI: 10.1007/s11064-023-03891-2] [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: 12/21/2022] [Revised: 01/25/2023] [Accepted: 02/07/2023] [Indexed: 02/18/2023]
Abstract
Epilepsy is a common and severe neurological disorder in which impaired glucose metabolism leads to changes in neuronal excitability that slow or promote the development of epilepsy. Leptin and adiponectin are important mediators regulating glucose metabolism in the peripheral and central nervous systems. Many studies have reported a strong association between epilepsy and these two adipokines involved in multiple signaling cascades and glucose metabolism. Due to the complex regulatory mechanisms between them and various signal activation networks, their role in epilepsy involves many aspects, including the release of inflammatory mediators, oxidative damage, and neuronal apoptosis. This paper aims to summarize the signaling pathways involved in leptin and adiponectin and the regulation of glucose metabolism from the perspective of the pathogenesis of epilepsy. In particular, we discuss the dual effects of leptin in epilepsy and the relationship between antiepileptic drugs and changes in the levels of these two adipokines. Clinical practitioners may need to consider these factors in evaluating clinical drugs. Through this review, we can better understand the specific involvement of leptin and adiponectin in the pathogenesis of epilepsy, provide ideas for further exploration, and bring about practical significance for the treatment of epilepsy, especially for the development of personalized treatment according to individual metabolic characteristics.
Collapse
Affiliation(s)
- Yisi Shan
- Department of Neurology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, 215600, China.,Translational Medical Innovation Center, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, 215600, China
| | - Yeting Chen
- Department of Acupuncture, Zhangjiagang Second People's Hospital, Zhangjiagang, 215600, China
| | - Haiping Gu
- Department of Neurology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, 215600, China
| | - Yadong Wang
- Department of Neurology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, 215600, China
| | - Yaming Sun
- Department of Neurology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, 215600, China.
| |
Collapse
|
12
|
Yang CZ, Wang SH, Zhang RH, Lin JH, Tian YH, Yang YQ, Liu J, Ma YX. Neuroprotective effect of astragalin via activating PI3K/Akt-mTOR-mediated autophagy on APP/PS1 mice. Cell Death Dis 2023; 9:15. [PMID: 36681681 PMCID: PMC9867706 DOI: 10.1038/s41420-023-01324-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 01/07/2023] [Accepted: 01/11/2023] [Indexed: 01/22/2023]
Abstract
As a small molecule flavonoid, astragalin (AST) has anti-inflammatory, anti-cancer, and anti-oxidation effects. However, the impact and molecular mechanism of AST in Alzheimer's disease (AD) are still not clear. This study aims to investigate the neuroprotective effect and mechanism of AST on APP/PS1 mice and Aβ25-35-injured HT22 cells. In this study, we found that AST ameliorated cognitive dysfunction, reduced hippocampal neuronal damage and loss, and Aβ pathology in APP/PS1 mice. Subsequently, AST activated autophagy and up-regulated the levels of autophagic flux-related protein in APP/PS1 mice and Aβ25-35-induced injury in HT22 cells. Interestingly, AST down-regulated the phosphorylation level of PI3K/Akt-mTOR pathway-related proteins, which was reversed by autophagy inhibitors 3-Methyladenine (3-MA) or Bafilomycin A1 (Baf A1). At the same time, consistent with the impacts of Akt inhibitor MK2206 and mTOR inhibitor rapamycin, inhibited levels of autophagy in Aβ25-35-injured HT22 cells were activated by the administration of AST. Taken together, these results suggested that AST played key neuroprotective roles on AD via stimulating PI3K/Akt-mTOR pathway-mediated autophagy and autophagic flux. This study revealed a new mechanism of autophagy regulation behind the neuroprotection impact of AST for AD treatment.
Collapse
Affiliation(s)
- Cui-Zhu Yang
- grid.411847.f0000 0004 1804 4300Department of Anatomy, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Shu-Han Wang
- grid.411847.f0000 0004 1804 4300Department of Anatomy, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Run-Heng Zhang
- grid.411847.f0000 0004 1804 4300Department of Anatomy, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jia-Hong Lin
- grid.411847.f0000 0004 1804 4300Department of Anatomy, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Ying-Hong Tian
- grid.284723.80000 0000 8877 7471Experiment Teaching & Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Ya-Qi Yang
- grid.411847.f0000 0004 1804 4300Department of Anatomy, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jing Liu
- grid.411847.f0000 0004 1804 4300Department of Anatomy, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yu-Xin Ma
- grid.411847.f0000 0004 1804 4300Department of Anatomy, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China ,grid.411847.f0000 0004 1804 4300Guangdong Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
13
|
Zavala-Tecuapetla C, Luna-Munguia H, López-Meraz ML, Cuellar-Herrera M. Advances and Challenges of Cannabidiol as an Anti-Seizure Strategy: Preclinical Evidence. Int J Mol Sci 2022; 23:ijms232416181. [PMID: 36555823 PMCID: PMC9783044 DOI: 10.3390/ijms232416181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/24/2022] [Accepted: 12/16/2022] [Indexed: 12/23/2022] Open
Abstract
The use of Cannabis for medicinal purposes has been documented since ancient times, where one of its principal cannabinoids extracted from Cannabis sativa, cannabidiol (CBD), has emerged over the last few years as a promising molecule with anti-seizure potential. Here, we present an overview of recent literature pointing out CBD's pharmacological profile (solubility, metabolism, drug-drug interactions, etc.,), CBD's interactions with multiple molecular targets as well as advances in preclinical research concerning its anti-seizure effect on both acute seizure models and chronic models of epilepsy. We also highlight the recent attention that has been given to other natural cannabinoids and to synthetic derivatives of CBD as possible compounds with therapeutic anti-seizure potential. All the scientific research reviewed here encourages to continue to investigate the probable therapeutic efficacy of CBD and its related compounds not only in epilepsy but also and specially in drug-resistant epilepsy, since there is a dire need for new and effective drugs to treat this disease.
Collapse
Affiliation(s)
- Cecilia Zavala-Tecuapetla
- Laboratory of Physiology of Reticular Formation, National Institute of Neurology and Neurosurgery, Insurgentes Sur 3877, La Fama, Mexico City 14269, Mexico
- Correspondence:
| | - Hiram Luna-Munguia
- Departamento de Neurobiologia Conductual y Cognitiva, Instituto de Neurobiologia, Universidad Nacional Autonoma de Mexico, Campus UNAM-Juriquilla, Queretaro 76230, Mexico
| | - María-Leonor López-Meraz
- Instituto de Investigaciones Cerebrales, Universidad Veracruzana, Luis Castelazo Ayala s/n, Col. Industrial Ánimas, Xalapa 91190, Mexico
| | - Manola Cuellar-Herrera
- Epilepsy Clinic, Hospital General de México Dr. Eduardo Liceaga, Dr. Balmis 148, Doctores, Mexico City 06720, Mexico
| |
Collapse
|
14
|
FDA-Approved Kinase Inhibitors in Preclinical and Clinical Trials for Neurological Disorders. Pharmaceuticals (Basel) 2022; 15:ph15121546. [PMID: 36558997 PMCID: PMC9784968 DOI: 10.3390/ph15121546] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/09/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022] Open
Abstract
Cancers and neurological disorders are two major types of diseases. We previously developed a new concept termed "Aberrant Cell Cycle Diseases" (ACCD), revealing that these two diseases share a common mechanism of aberrant cell cycle re-entry. The aberrant cell cycle re-entry is manifested as kinase/oncogene activation and tumor suppressor inactivation, which are hallmarks of both tumor growth in cancers and neuronal death in neurological disorders. Therefore, some cancer therapies (e.g., kinase inhibition, tumor suppressor elevation) can be leveraged for neurological treatments. The United States Food and Drug Administration (US FDA) has so far approved 74 kinase inhibitors, with numerous other kinase inhibitors in clinical trials, mostly for the treatment of cancers. In contrast, there are dire unmet needs of FDA-approved drugs for neurological treatments, such as Alzheimer's disease (AD), intracerebral hemorrhage (ICH), ischemic stroke (IS), traumatic brain injury (TBI), and others. In this review, we list these 74 FDA-approved kinase-targeted drugs and identify those that have been reported in preclinical and/or clinical trials for neurological disorders, with a purpose of discussing the feasibility and applicability of leveraging these cancer drugs (FDA-approved kinase inhibitors) for neurological treatments.
Collapse
|
15
|
Kagitani-Shimono K, Kato H, Soeda F, Iwatani Y, Mukai M, Ogawa K, Tominaga K, Nabatame S, Taniike M. Extension of microglial activation is associated with epilepsy and cognitive dysfunction in Tuberous sclerosis complex: A TSPO-PET study. Neuroimage Clin 2022; 37:103288. [PMID: 36521371 PMCID: PMC9758490 DOI: 10.1016/j.nicl.2022.103288] [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: 09/17/2022] [Revised: 12/07/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND OBJECTIVES Neuroinflammation contributes to the severity of various neurological disorders, including epilepsy. Tuberous sclerosis complex (TSC) is a condition that results in the overactivation of the mammalian target of rapamycin (mTOR) pathway, which has been linked to the activation of microglia responsible for neuroinflammation. To clarify the involvement of neuroinflammation in the neuropathophysiology of TSC, we performed a positron emission tomography (PET) study using the translocator protein (TSPO) radioligand, [11C] DPA713, and investigated microglial activation in relation to neurological manifestations, especially epilepsy and cognitive function. METHODS This cross-sectional study included 18 patients with TSC (6 in the no-seizure group, 6 in the refractory seizure group, and 6 in the mTOR-inhibitor [mTOR-i] group). All participants underwent [11C] DPA713-PET. PET results were superimposed with a 3D T2-weighted fluid-attenuated inversion-recovery (FLAIR) and T1-weighted image (T1WI) to evaluate the location of cortical tubers. Microglial activation was assessed using the standardized uptake value ratio (SUVr) of DPA713 binding. The volume ratio of the DPA713-positive area to the intracranial volume (volume ratio of DPA713/ICV) was calculated to evaluate the extent of microglial activation. A correlation analysis was performed to examine the relationship between volume ratio of DPA713/ICV and severity of epilepsy and cognitive function. RESULTS Most cortical tubers with hyperintensity on FLAIR and hypo- or isointensity on T1WI showed microglial activation. The extent of microglial activation was significantly greater in the refractory seizure group than in the no-seizure or mTOR-i groups (p < 0.001). The extent of microglial activation in subjects without mTOR-i treatment correlated positively with epilepsy severity (r = 0.822, P = 0.001) and negatively with cognitive function (r = -0.846, p = 0.001), but these correlations were not present in the mTOR-i group (r = 0.232, P = 0.658, r = 0.371, P = 0.469, respectively). CONCLUSION Neuroinflammation is associated with the severity of epilepsy and cognitive dysfunction in brains with TSC. mTOR-i may suppress the extent of neuroinflammation in TSC. Investigating the spread of microglial activation using TSPO-PET in these patients may help to predict the progression of neuropathy by assessing the degree of neuroinflammation and therefore be useful for determining how aggressive the treatment should be and in assessing the effectiveness of such treatment in patients with TSC.
Collapse
Affiliation(s)
- Kuriko Kagitani-Shimono
- Department of Child Development, United Graduate School of Child Development, Osaka University, Osaka, Japan; Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan.
| | - Hiroki Kato
- Department of Nuclear Medicine and Tracer Kinetics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Fumihiko Soeda
- Department of Nuclear Medicine and Tracer Kinetics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshiko Iwatani
- Department of Child Development, United Graduate School of Child Development, Osaka University, Osaka, Japan; Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Masashi Mukai
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Katsuhiro Ogawa
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Koji Tominaga
- Department of Child Development, United Graduate School of Child Development, Osaka University, Osaka, Japan; Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shin Nabatame
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Masako Taniike
- Department of Child Development, United Graduate School of Child Development, Osaka University, Osaka, Japan; Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
16
|
Hu Y, Meng B, Yin S, Yang M, Li Y, Liu N, Li S, Liu Y, Sun D, Wang S, Wang Y, Fu Z, Wu Y, Pang A, Sun J, Wang Y, Yang X. Scorpion venom peptide HsTx2 suppressed PTZ-induced seizures in mice via the circ_0001293/miR-8114/TGF-β2 axis. J Neuroinflammation 2022; 19:284. [PMID: 36457055 PMCID: PMC9713996 DOI: 10.1186/s12974-022-02647-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 11/17/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Due to the complexity of the mechanisms involved in epileptogenesis, the available antiseizure drugs (ASDs) do not meet clinical needs; hence, both the discovery of new ASDs and the elucidation of novel molecular mechanisms are very important. METHODS BALB/c mice were utilized to establish an epilepsy model induced by pentylenetetrazol (PTZ) administration. The peptide HsTx2 was administered for treatment. Primary astrocyte culture, immunofluorescence staining, RNA sequencing, identification and quantification of mouse circRNAs, cell transfection, bioinformatics and luciferase reporter analyses, enzyme-linked immunosorbent assay, RNA extraction and reverse transcription-quantitative PCR, Western blot and cell viability assays were used to explore the potential mechanism of HsTx2 via the circ_0001293/miR-8114/TGF-β2 axis. RESULTS The scorpion venom peptide HsTx2 showed an anti-epilepsy effect, reduced the inflammatory response, and improved the circular RNA circ_0001293 expression decrease caused by PTZ in the mouse brain. Mechanistically, in astrocytes, circ_0001293 acted as a sponge of endogenous microRNA-8114 (miR-8114), which targets transforming growth factor-beta 2 (TGF-β2). The knockdown of circ_0001293, overexpression of miR-8114, and downregulation of TGF-β2 all reversed the anti-inflammatory effects and the influence of HsTx2 on the MAPK and NF-κB signaling pathways in astrocytes. Moreover, both circ_0001293 knockdown and miR-8114 overexpression reversed the beneficial effects of HsTx2 on inflammation, epilepsy progression, and the MAPK and NF-κB signaling pathways in vivo. CONCLUSIONS HsTx2 suppressed PTZ-induced epilepsy by ameliorating inflammation in astrocytes via the circ_0001293/miR-8114/TGF-β2 axis. Our results emphasized that the use of exogenous peptide molecular probes as a novel type of ASD, as well as to explore the novel endogenous noncoding RNA-mediated mechanisms of epilepsy, might be a promising research area.
Collapse
Affiliation(s)
- Yan Hu
- grid.285847.40000 0000 9588 0960Department of Anatomy and Histology and Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500 Yunnan China ,grid.452826.fDepartment of Gynecology, Third Affiliated Hospital of Kunming Medical University, Kunming, 650118 Yunnan China
| | - Buliang Meng
- grid.285847.40000 0000 9588 0960Department of Anatomy and Histology and Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500 Yunnan China
| | - Saige Yin
- grid.285847.40000 0000 9588 0960Department of Anatomy and Histology and Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500 Yunnan China
| | - Meifeng Yang
- grid.285847.40000 0000 9588 0960Department of Anatomy and Histology and Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500 Yunnan China
| | - Yilin Li
- grid.285847.40000 0000 9588 0960Department of Anatomy and Histology and Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500 Yunnan China
| | - Naixin Liu
- grid.285847.40000 0000 9588 0960Department of Anatomy and Histology and Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500 Yunnan China
| | - Shanshan Li
- grid.285847.40000 0000 9588 0960Department of Anatomy and Histology and Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500 Yunnan China
| | - Yixiang Liu
- grid.413059.a0000 0000 9952 9510Key Laboratory of Chemistry in Ethnic Medicine Resource, State Ethnic Affairs Commission & Ministry of Education, School of Ethno-Medicine and Ethno-Pharmacy, Yunnan Minzu University, Kunming, 650504 Yunnan China
| | - Dandan Sun
- grid.285847.40000 0000 9588 0960Department of Anatomy and Histology and Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500 Yunnan China
| | - Siyu Wang
- grid.285847.40000 0000 9588 0960Department of Anatomy and Histology and Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500 Yunnan China
| | - Yinglei Wang
- grid.285847.40000 0000 9588 0960Department of Anatomy and Histology and Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500 Yunnan China
| | - Zhe Fu
- grid.285847.40000 0000 9588 0960Department of Anatomy and Histology and Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500 Yunnan China
| | - Yutong Wu
- grid.285847.40000 0000 9588 0960Department of Anatomy and Histology and Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500 Yunnan China
| | - Ailan Pang
- grid.414902.a0000 0004 1771 3912Department of Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, 650031 Yunnan China
| | - Jun Sun
- grid.285847.40000 0000 9588 0960Department of Anatomy and Histology and Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500 Yunnan China
| | - Ying Wang
- grid.413059.a0000 0000 9952 9510Key Laboratory of Chemistry in Ethnic Medicine Resource, State Ethnic Affairs Commission & Ministry of Education, School of Ethno-Medicine and Ethno-Pharmacy, Yunnan Minzu University, Kunming, 650504 Yunnan China
| | - Xinwang Yang
- grid.285847.40000 0000 9588 0960Department of Anatomy and Histology and Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, 650500 Yunnan China
| |
Collapse
|
17
|
Kline C, Jain P, Kilburn L, Bonner ER, Gupta N, Crawford JR, Banerjee A, Packer RJ, Villanueva-Meyer J, Luks T, Zhang Y, Kambhampati M, Zhang J, Yadavilli S, Zhang B, Gaonkar KS, Rokita JL, Kraya A, Kuhn J, Liang W, Byron S, Berens M, Molinaro A, Prados M, Resnick A, Waszak SM, Nazarian J, Mueller S. Upfront Biology-Guided Therapy in Diffuse Intrinsic Pontine Glioma: Therapeutic, Molecular, and Biomarker Outcomes from PNOC003. Clin Cancer Res 2022; 28:3965-3978. [PMID: 35852795 PMCID: PMC9475246 DOI: 10.1158/1078-0432.ccr-22-0803] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/22/2022] [Accepted: 07/15/2022] [Indexed: 01/07/2023]
Abstract
PURPOSE PNOC003 is a multicenter precision medicine trial for children and young adults with newly diagnosed diffuse intrinsic pontine glioma (DIPG). PATIENTS AND METHODS Patients (3-25 years) were enrolled on the basis of imaging consistent with DIPG. Biopsy tissue was collected for whole-exome and mRNA sequencing. After radiotherapy (RT), patients were assigned up to four FDA-approved drugs based on molecular tumor board recommendations. H3K27M-mutant circulating tumor DNA (ctDNA) was longitudinally measured. Tumor tissue and matched primary cell lines were characterized using whole-genome sequencing and DNA methylation profiling. When applicable, results were verified in an independent cohort from the Children's Brain Tumor Network (CBTN). RESULTS Of 38 patients enrolled, 28 patients (median 6 years, 10 females) were reviewed by the molecular tumor board. Of those, 19 followed treatment recommendations. Median overall survival (OS) was 13.1 months [95% confidence interval (CI), 11.2-18.4] with no difference between patients who followed recommendations and those who did not. H3K27M-mutant ctDNA was detected at baseline in 60% of cases tested and associated with response to RT and survival. Eleven cell lines were established, showing 100% fidelity of key somatic driver gene alterations in the primary tumor. In H3K27-altered DIPGs, TP53 mutations were associated with worse OS (TP53mut 11.1 mo; 95% CI, 8.7-14; TP53wt 13.3 mo; 95% CI, 11.8-NA; P = 3.4e-2), genome instability (P = 3.1e-3), and RT resistance (P = 6.4e-4). The CBTN cohort confirmed an association between TP53 mutation status, genome instability, and clinical outcome. CONCLUSIONS Upfront treatment-naïve biopsy provides insight into clinically relevant molecular alterations and prognostic biomarkers for H3K27-altered DIPGs.
Collapse
Affiliation(s)
- Cassie Kline
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Payal Jain
- Division of Neurosurgery, Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Lindsay Kilburn
- Department of Hematology and Oncology, Children's National Hospital, Washington, DC
| | - Erin R. Bonner
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC
- Institute for Biomedical Sciences, The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Nalin Gupta
- Department of Neurological Surgery, University of California, San Francisco, California
| | - John R. Crawford
- Department of Neuroscience, University of California, San Diego, California
- Rady Children's Hospital San Diego, San Diego, California
| | - Anu Banerjee
- Department of Neurological Surgery, University of California, San Francisco, California
- Department of Pediatrics, University of California, San Francisco, California
| | - Roger J. Packer
- Center for Neuroscience and Behavioral Medicine, Children's National Hospital, Washington, DC
| | - Javier Villanueva-Meyer
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Tracy Luks
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Yalan Zhang
- Department of Neurological Surgery, University of California, San Francisco, California
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California
| | - Madhuri Kambhampati
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC
| | - Jie Zhang
- Department of Neurology, University of California, San Francisco, California
| | - Sridevi Yadavilli
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC
| | - Bo Zhang
- Division of Neurosurgery, Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Krutika S. Gaonkar
- Division of Neurosurgery, Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Bioinformatics and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Jo Lynne Rokita
- Division of Neurosurgery, Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Bioinformatics and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Adam Kraya
- Division of Neurosurgery, Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - John Kuhn
- College of Pharmacy, University of Texas Health Science Center, San Antonio, Texas
| | - Winnie Liang
- Translational Genomic Research Institute (TGEN), Phoenix, Arizona
| | - Sara Byron
- Translational Genomic Research Institute (TGEN), Phoenix, Arizona
| | - Michael Berens
- Translational Genomic Research Institute (TGEN), Phoenix, Arizona
| | - Annette Molinaro
- Department of Neurological Surgery, University of California, San Francisco, California
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California
| | - Michael Prados
- Department of Neurological Surgery, University of California, San Francisco, California
| | - Adam Resnick
- Division of Neurosurgery, Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Sebastian M. Waszak
- Department of Neurology, University of California, San Francisco, California
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo and Oslo University Hospital, Oslo, Norway
- Division of Pediatric and Adolescent Medicine, Department of Pediatric Research, Rikshospitalet, Oslo University Hospital, Oslo, Norway
| | - Javad Nazarian
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC
- Institute for Biomedical Sciences, The George Washington University School of Medicine and Health Sciences, Washington, DC
- Department of Oncology, University Children's Hospital Zürich, Zürich, Switzerland
| | - Sabine Mueller
- Department of Neurological Surgery, University of California, San Francisco, California
- Department of Pediatrics, University of California, San Francisco, California
- Department of Neurology, University of California, San Francisco, California
- Department of Oncology, University Children's Hospital Zürich, Zürich, Switzerland
| |
Collapse
|
18
|
Wang X, Fu Y, Botchway BOA, Zhang Y, Zhang Y, Jin T, Liu X. Quercetin Can Improve Spinal Cord Injury by Regulating the mTOR Signaling Pathway. Front Neurol 2022; 13:905640. [PMID: 35669881 PMCID: PMC9163835 DOI: 10.3389/fneur.2022.905640] [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: 03/27/2022] [Accepted: 04/06/2022] [Indexed: 11/16/2022] Open
Abstract
The pathogenesis of spinal cord injury (SCI) is complex. At present, there is no effective treatment for SCI, with most current interventions focused on improving the symptoms. Inflammation, apoptosis, autophagy, and oxidative stress caused by secondary SCI may instigate serious consequences in the event of SCI. The mammalian target of rapamycin (mTOR), as a key signaling molecule, participates in the regulation of inflammation, apoptosis, and autophagy in several processes associated with SCI. Quercetin can reduce the loss of myelin sheath, enhance the ability of antioxidant stress, and promote axonal regeneration. Moreover, quercetin is also a significant player in regulating the mTOR signaling pathway that improves pathological alterations following neuronal injury. Herein, we review the therapeutic effects of quercetin in SCI through its modulation of the mTOR signaling pathway and elaborate on how it can be a potential interventional agent for SCI.
Collapse
Affiliation(s)
- Xichen Wang
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Zhejiang, China
| | - Yuke Fu
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Zhejiang, China
| | | | - Yufeng Zhang
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Zhejiang, China
| | - Yong Zhang
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Zhejiang, China
| | - Tian Jin
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Zhejiang, China
| | - Xuehong Liu
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Zhejiang, China
| |
Collapse
|
19
|
Song L, Liu S, Zhao S. Everolimus (RAD001) combined with programmed death-1 (PD-1) blockade enhances radiosensitivity of cervical cancer and programmed death-ligand 1 (PD-L1) expression by blocking the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR)/S6 kinase 1 (S6K1) pathway. Bioengineered 2022; 13:11240-11257. [PMID: 35485300 PMCID: PMC9208494 DOI: 10.1080/21655979.2022.2064205] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cervical cancer (CC) is the 4th most prevalent malignancy in females. This study explored the mechanism of everolimus (RAD001) combined with programmed death-1 (PD-1) blockade on radiosensitivity by phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) pathway and autophagy in CC cells. Low-radiosensitive CaSki cells were selected as study objects. After RAD001 treatment, PI3K/AKT/mTOR pathway activation, autophagy, migration and invasion abilities, autophagy-related proteins (LC3-I, LC3-II, and p62), and PD-L1 expression in CC cells were detected. After triple treatment of radiotherapy (RT), RAD001, and PD-1 blockade to the CC mouse models, tumor weight and volume were recorded. Ki67 expression, the number of CD8 + T cells, and the ability to produce IFN-γ and TNF-α in tumor tissues were determined. RAD001 promoted autophagy by repressing PI3K/AKT/mTOR pathway, augmented RT-induced apoptosis, and weakened migration and invasion, thereby increasing CC cell radiosensitivity. RAD001 elevated RT-induced PD-L1 level. RT combined with RAD001 and PD-1 blockade intensified the inhibitory effect of RT on tumor growth, reduced the amount of Ki67-positive cells, enhanced radiosensitivity of CC mice, and increased the quantity and killing ability of CD8 + T cells. Briefly, RAD001 combined with PD-1 blockade increases radiosensitivity of CC by impeding the PI3K/AKT/mTOR pathway and potentiating cell autophagy.
Collapse
Affiliation(s)
- Lili Song
- Department of Obstetrics and Gynecology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shikai Liu
- Department of Obstetrics and Gynecology, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Sufen Zhao
- Department of Obstetrics and Gynecology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
20
|
Senousy MA, Hanafy ME, Shehata N, Rizk SM. Erythropoietin and Bacillus Calmette-Guérin Vaccination Mitigate 3-Nitropropionic Acid-Induced Huntington-like Disease in Rats by Modulating the PI3K/Akt/mTOR/P70S6K Pathway and Enhancing the Autophagy. ACS Chem Neurosci 2022; 13:721-732. [PMID: 35226456 DOI: 10.1021/acschemneuro.1c00523] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Oxidative stress and mitochondrial dysfunction are among the mechanisms expected to explain the pathogenesis of Huntington's disease. Erythropoietin (EPO) and the Bacillus Calmette-Guérin (BCG) vaccine have neuroprotective effects against neurodegenerative diseases; however, the full mechanisms of their action are currently unclear. Here, for the first time, we investigated the neuroprotective effect of BCG vaccination in Huntington-like disease induced by 3-nitropropionic acid (3-NP) and its combination with EPO. Male Wistar rats were randomized into five groups: saline-treated control; 3-NP group (20 mg/kg/day, i.p.) for 7 days; EPO-treated group (5000 IU/kg/day, i.p.) for 14 days after 3-NP administration; live BCG vaccine prophylactic group (5000 cfu/g, i.p.) 10 days prior to 3-NP administration; and live BCG vaccine (5000 cfu/g, i.p.) 10 days before 3-NP administration, followed by EPO treatment (5000 IU/kg/day, i.p.) for 14 days. In a histopathological examination, striatum neurodegeneration was evidenced in the 3-NP injected rats. Administration of 3-NP elevated the levels of p-PI3K, p-Akt, p-mTOR, p-P70S6K, BAX, malondialdehyde, nitric oxide, and cytochrome oxidase while reduced the levels of BCL-2, superoxide dismutase, reduced glutathione, and the autophagy marker microtubule-associated protein light chain 3 in the striatum. EPO and BCG ameliorated the biochemical, histopathological, and behavioral derangements induced by 3-NP, with prominent neuroprotection observed in rats administered the BCG prophylactic combined with EPO treatment. These results highlight the role played by EPO and BCG in the management of 3-NP-induced Huntington-like disease by inhibiting the PI3K/Akt/mTOR/P70S6K pathway and enhancing the autophagy.
Collapse
Affiliation(s)
- Mahmoud A. Senousy
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Mona Essam Hanafy
- Central Administration of Biological and Innovative Products and Clinical Studies, Egyptian Drug Authority, Giza 00202, Egypt
| | - Nahla Shehata
- Central Administration of Biological and Innovative Products and Clinical Studies, Egyptian Drug Authority, Giza 00202, Egypt
| | - Sherine M. Rizk
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| |
Collapse
|
21
|
Cai M, Lin W. The Function of NF-Kappa B During Epilepsy, a Potential Therapeutic Target. Front Neurosci 2022; 16:851394. [PMID: 35360161 PMCID: PMC8961383 DOI: 10.3389/fnins.2022.851394] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 02/22/2022] [Indexed: 01/04/2023] Open
Abstract
The transcriptional regulator nuclear factor kappa B (NF-κB) modulates cellular biological activity by binding to promoter regions in the nucleus and transcribing various protein-coding genes. The NF-κB pathway plays a major role in the expressing genes related to inflammation, including chemokines, interleukins, and tumor necrosis factor. It also transcribes genes that can promote neuronal survival or apoptosis. Epilepsy is one of the most common brain disorders and it not only causes death worldwide but also affects the day-to-day life of affected individuals. While epilepsy has diverse treatment options, there remain patients who are not sensitive to the existing treatment methods. Recent studies have implicated the critical role of NF-κB in epilepsy. It is upregulated in neurons, glial cells, and endothelial cells, due to neuronal loss, glial cell proliferation, blood-brain barrier dysfunction, and hippocampal sclerosis through the glutamate and γ-aminobutyric acid imbalance, ion concentration changes, and other mechanisms. In this review, we summarize the functional changes caused by the upregulation of NF-κB in the central nervous system during different periods after seizures. This review is the first to deconvolute the complicated functions of NF-κB, and speculate that the regulation of NF-κB can be a safe and effective treatment strategy for epilepsy.
Collapse
|
22
|
Park S, Zhu J, Jeong KH, Kim WJ. Adjudin prevents neuronal damage and neuroinflammation via inhibiting mTOR activation against pilocarpine-induced status epilepticus. Brain Res Bull 2022; 182:80-89. [PMID: 35182690 DOI: 10.1016/j.brainresbull.2022.02.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/23/2022] [Accepted: 02/14/2022] [Indexed: 11/02/2022]
Abstract
Inflammatory responses in the brain play an etiological role in the development of epilepsy, suggesting that finding novel molecules for controlling neuroinflammation may have clinical value in developing the disease-modifying strategies for epileptogenesis. Adjudin, a multi-functional small molecule compound, has pleiotropic effects, including anti-inflammatory properties. In the present study, we aimed to investigate the effects of adjudin on pilocarpine-induced status epilepticus (SE) and its role in the regulation of reactive gliosis and neuroinflammation. SE was induced in male C57BL/6 mice that were then treated with adjudin (50mg/kg) for 3 days after SE onset. Immunofluorescence staining, terminal deoxynucleotidyl transferase dUTP nick end labeling staining, and western blot analysis were used to evaluate the effects of adjudin treatment in the hippocampus after SE. Our results showed that adjudin treatment significantly mitigated apoptotic cell death in the hippocampus after SE onset. Moreover, adjudin treatment suppressed SE-induced glial activation and activation of mammalian target of rapamycin signaling in the hippocampus. Concomitantly, adjudin treatment significantly reduced SE-induced inflammatory processes, as confirmed by changes in the expression of inflammatory mediators such as tumor necrosis factor-α, interleukin-1β, and arginase-1. In conclusion, these findings suggest that adjudin may serve as a potential neuroprotective agent for preventing pathological mechanisms implicated in epileptogenesis.
Collapse
Affiliation(s)
- Soojin Park
- Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea; Brain Korea 21 Plus Project for Medical Science, Yonsei University, Seoul, Republic of Korea
| | - Jing Zhu
- Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea; Brain Korea 21 Plus Project for Medical Science, Yonsei University, Seoul, Republic of Korea
| | - Kyoung Hoon Jeong
- Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea; Epilepsy Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Won-Joo Kim
- Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
23
|
Lee JW, Chun W, Lee HJ, Kim SM, Min JH, Kim DY, Kim MO, Ryu HW, Lee SU. The Role of Microglia in the Development of Neurodegenerative Diseases. Biomedicines 2021; 9:biomedicines9101449. [PMID: 34680566 PMCID: PMC8533549 DOI: 10.3390/biomedicines9101449] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/04/2021] [Accepted: 10/08/2021] [Indexed: 01/15/2023] Open
Abstract
Microglia play an important role in the maintenance and neuroprotection of the central nervous system (CNS) by removing pathogens, damaged neurons, and plaques. Recent observations emphasize that the promotion and development of neurodegenerative diseases (NDs) are closely related to microglial activation. In this review, we summarize the contribution of microglial activation and its associated mechanisms in NDs, such as epilepsy, Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD), based on recent observations. This review also briefly introduces experimental animal models of epilepsy, AD, PD, and HD. Thus, this review provides a better understanding of microglial functions in the development of NDs, suggesting that microglial targeting could be an effective therapeutic strategy for these diseases.
Collapse
Affiliation(s)
- Jae-Won Lee
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea; (S.-M.K.); (J.-H.M.); (D.-Y.K.)
- Correspondence: (J.-W.L.); (M.-O.K.); (H.W.R.); (S.U.L.); Tel.: +82-43-240-6135 (J.-W.L.)
| | - Wanjoo Chun
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon 24341, Korea; (W.C.); (H.J.L.)
| | - Hee Jae Lee
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon 24341, Korea; (W.C.); (H.J.L.)
| | - Seong-Man Kim
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea; (S.-M.K.); (J.-H.M.); (D.-Y.K.)
| | - Jae-Hong Min
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea; (S.-M.K.); (J.-H.M.); (D.-Y.K.)
| | - Doo-Young Kim
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea; (S.-M.K.); (J.-H.M.); (D.-Y.K.)
| | - Mun-Ock Kim
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea; (S.-M.K.); (J.-H.M.); (D.-Y.K.)
- Correspondence: (J.-W.L.); (M.-O.K.); (H.W.R.); (S.U.L.); Tel.: +82-43-240-6135 (J.-W.L.)
| | - Hyung Won Ryu
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea; (S.-M.K.); (J.-H.M.); (D.-Y.K.)
- Correspondence: (J.-W.L.); (M.-O.K.); (H.W.R.); (S.U.L.); Tel.: +82-43-240-6135 (J.-W.L.)
| | - Su Ui Lee
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea; (S.-M.K.); (J.-H.M.); (D.-Y.K.)
- Correspondence: (J.-W.L.); (M.-O.K.); (H.W.R.); (S.U.L.); Tel.: +82-43-240-6135 (J.-W.L.)
| |
Collapse
|