1
|
Zhang X, Zheng Y, Wang Z, Zhang G, Yang L, Gan J, Jiang X. Calpain: The regulatory point of cardiovascular and cerebrovascular diseases. Biomed Pharmacother 2024; 179:117272. [PMID: 39153432 DOI: 10.1016/j.biopha.2024.117272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 07/24/2024] [Accepted: 08/05/2024] [Indexed: 08/19/2024] Open
Abstract
Calpain, a key member of the Calpain cysteine protease superfamily, performs limited protein hydrolysis in a calcium-dependent manner. Its activity is tightly regulated due to the potential for non-specific cleavage of various intracellular proteins upon aberrant activation. A thorough review of the literature from 2010 to 2023 reveals 121 references discussing cardiovascular and cerebrovascular diseases. Dysregulation of the Calpain system is associated with various pathological phenomena, including lipid metabolism disorders, inflammation, apoptosis, and excitotoxicity. Although recent studies have revealed the significant role of Calpain in cardiovascular and cerebrovascular diseases, the precise mechanisms remain incompletely understood. Exploring the potential of Calpain inhibition as a therapeutic approach for the treatment of cardiovascular and cerebrovascular diseases may emerge as a compelling area of interest for future calpain research.
Collapse
Affiliation(s)
- Xiaolu Zhang
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Yujia Zheng
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Ziyu Wang
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Guangming Zhang
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Lin Yang
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Jiali Gan
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Xijuan Jiang
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China.
| |
Collapse
|
2
|
Wu M, Xu S, Mi K, Yang S, Xu Y, Li J, Chen J, Zhang X. GluN2B-containing NMDA receptor attenuated neuronal apoptosis in the mouse model of HIBD through inhibiting endoplasmic reticulum stress-activated PERK/eIF2α signaling pathway. Front Mol Neurosci 2024; 17:1375843. [PMID: 38638600 PMCID: PMC11024425 DOI: 10.3389/fnmol.2024.1375843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 03/22/2024] [Indexed: 04/20/2024] Open
Abstract
Introduction Neonatal hypoxic-ischemic brain damage (HIBD) refers to brain damage in newborns caused by hypoxia and reduced or even stopped cerebral blood flow during the perinatal period. Currently, there are no targeted treatments for neonatal ischemic hypoxic brain damage, primarily due to the incomplete understanding of its pathophysiological mechanisms. Especially, the role of NMDA receptors is less studied in HIBD. Therefore, this study explored the molecular mechanism of endogenous protection mediated by GluN2B-NMDAR in HIBD. Method Hypoxic ischemia was induced in mice aged 9-11 days. The brain damage was examined by Nissl staining and HE staining, while neuronal apoptosis was examined by Hoechst staining and TTC staining. And cognitive deficiency of mice was examined by various behavior tests including Barnes Maze, Three Chamber Social Interaction Test and Elevated Plus Maze. The activation of ER stress signaling pathways were evaluated by Western blot. Results We found that after HIBD induction, the activation of GluN2B-NMDAR attenuated neuronal apoptosis and brain damage. Meanwhile, the ER stress PERK/eIF2α signaling pathway was activated in a time-dependent manner after HIBE. Furthermore, after selective inhibiting GluN2B-NMDAR in HIBD mice with ifenprodil, the PERK/eIF2α signaling pathway remains continuously activated, leading to neuronal apoptosis, morphological brain damage. and aggravating deficits in spatial memory, cognition, and social abilities in adult mice. Discussion The results of this study indicate that, unlike its role in adult brain damage, GluN2B in early development plays a neuroprotective role in HIBD by inhibiting excessive activation of the PERK/eIF2α signaling pathway. This study provides theoretical support for the clinical development of targeted drugs or treatment methods for HIBD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xiaomin Zhang
- Department of Physiology, School of Basic Medicine, Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
3
|
Zhang X, Peng KZ, Xu SL, Wu MX, Sun HJ, Zhao J, Yang S, Liu SJ, Lia CY, Zhang XM. The GluN2B-Containing NMDA Receptor Alleviates Neuronal Apoptosis in Neonatal Hypoxic-Ischemic Encephalopathy by Activating PI3K-Akt-CREB Signaling Pathwa. Physiol Res 2023; 72:669-680. [PMID: 38015765 PMCID: PMC10751047 DOI: 10.33549/physiolres.935044] [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: 11/11/2022] [Accepted: 07/18/2023] [Indexed: 01/05/2024] Open
Abstract
Neonatal hypoxic-ischemic encephalopathy (HIE) is a disease caused by insufficient blood supply in the brain in newborns during the perinatal period. Severe HIE leads to patient death, and patients with mild HIE are at increased risk of cognitive deficits and behavioral abnormalities. The NMDA receptor is an important excitatory receptor in the central nervous system, and in adult hypoxic-ischemic injury both subtypes of the NMDA receptor play important but distinct roles. The GluN2A-containing NMDA receptor (GluN2A-NMDAR) could activate neuronal protective signaling pathway, while the GluN2B-NMDAR subtype is coupled to the apoptosis-inducing signaling pathway and leads to neuronal death. However, the expression level of GluN2B is higher in newborns than in adults, while the expression of GluN2A is lower. Therefore, it is not clear whether the roles of different NMDA receptor subtypes in HIE are consistent with those in adults. We investigated this issue in this study and found that in HIE, GluN2B plays a protective role by mediating the protective pathway through binding with PSD95, which is quite different to that in adults. The results of this study provided new theoretical support for the clinical treatment of neonatal hypoxic ischemia.
Collapse
Affiliation(s)
- X Zhang
- Department of Physiology, School of Basic Medicine, Kunming Medical University, Kunming, Yunnan Province, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
4
|
He Y, Ying J, Tang J, Zhou R, Qu H, Qu Y, Mu D. Neonatal Arterial Ischaemic Stroke: Advances in Pathologic Neural Death, Diagnosis, Treatment, and Prognosis. Curr Neuropharmacol 2022; 20:2248-2266. [PMID: 35193484 PMCID: PMC9890291 DOI: 10.2174/1570159x20666220222144744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 02/04/2022] [Accepted: 02/18/2022] [Indexed: 12/29/2022] Open
Abstract
Neonatal arterial ischaemic stroke (NAIS) is caused by focal arterial occlusion and often leads to severe neurological sequelae. Neural deaths after NAIS mainly include necrosis, apoptosis, necroptosis, autophagy, ferroptosis, and pyroptosis. These neural deaths are mainly caused by upstream stimulations, including excitotoxicity, oxidative stress, inflammation, and death receptor pathways. The current clinical approaches to managing NAIS mainly focus on supportive treatments, including seizure control and anticoagulation. In recent years, research on the pathology, early diagnosis, and potential therapeutic targets of NAIS has progressed. In this review, we summarise the latest progress of research on the pathology, diagnosis, treatment, and prognosis of NAIS and highlight newly potential diagnostic and treatment approaches.
Collapse
Affiliation(s)
- Yang He
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Junjie Ying
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Jun Tang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Ruixi Zhou
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Haibo Qu
- Department of Radiology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yi Qu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Dezhi Mu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| |
Collapse
|
5
|
Turcu AL, Companys-Alemany J, Phillips MB, Patel DS, Griñán-Ferré C, Loza MI, Brea JM, Pérez B, Soto D, Sureda FX, Kurnikova MG, Johnson JW, Pallàs M, Vázquez S. Design, synthesis, and in vitro and in vivo characterization of new memantine analogs for Alzheimer's disease. Eur J Med Chem 2022; 236:114354. [PMID: 35453065 PMCID: PMC9106868 DOI: 10.1016/j.ejmech.2022.114354] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 12/28/2022]
Abstract
Currently, of the few accessible symptomatic therapies for Alzheimer's disease (AD), memantine is the only N-methyl-d-aspartate receptor (NMDAR) blocker approved by the FDA. This work further explores a series of memantine analogs featuring a benzohomoadamantane scaffold. Most of the newly synthesized compounds block NMDARs in the micromolar range, but with lower potency than previously reported hit IIc, results that were supported by molecular dynamics simulations. Subsequently, electrophysiological studies with the more potent compounds allowed classification of IIc, a low micromolar, uncompetitive, voltage-dependent, NMDAR blocker, as a memantine-like compound. The excellent in vitro DMPK properties of IIc made it a promising candidate for in vivo studies in Caenorhabditis elegans (C. elegans) and in the 5XFAD mouse model of AD. Administration of IIc or memantine improved locomotion and rescues chemotaxis behavior in C. elegans. Furthermore, both compounds enhanced working memory in 5XFAD mice and modified NMDAR and CREB signaling, which may prevent synaptic dysfunction and modulate neurodegenerative progression.
Collapse
Affiliation(s)
- Andreea L Turcu
- Laboratori de Química Farmacèutica (Unitat Associada al CSIC), Facultat de Farmàcia i Ciències de l'Alimentació i Institut de Biomedicina (IBUB), Universitat de Barcelona, Av. Joan XXIII, 27-31, 08028, Barcelona, Spain; Neurophysiology Laboratory, Department of Biomedicine, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08036, Barcelona, Spain
| | - Júlia Companys-Alemany
- Pharmacology Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Neurosciences (NeuroUB), Universitat de Barcelona, Av. Joan XXIII 27-31, 08028, Barcelona, Spain
| | - Matthew B Phillips
- Department of Neuroscience and Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Dhilon S Patel
- Chemistry Department, Carnegie Mellon University, 4400 Fifth Ave, Pittsburgh, PA, 15213, USA
| | - Christian Griñán-Ferré
- Pharmacology Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Neurosciences (NeuroUB), Universitat de Barcelona, Av. Joan XXIII 27-31, 08028, Barcelona, Spain
| | - M Isabel Loza
- Innopharma Screening Platform, Biofarma Research Group, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas, Universidad de Santiago de Compostela, Edificio CIMUS, Av. Barcelona, S/N, E, 15706, Santiago de Compostela, Spain
| | - José M Brea
- Innopharma Screening Platform, Biofarma Research Group, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas, Universidad de Santiago de Compostela, Edificio CIMUS, Av. Barcelona, S/N, E, 15706, Santiago de Compostela, Spain
| | - Belén Pérez
- Department of Pharmacology, Therapeutics and Toxicology, Autonomous University of Barcelona, E-08193, Bellaterra, Spain
| | - David Soto
- Neurophysiology Laboratory, Department of Biomedicine, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08036, Barcelona, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Francesc X Sureda
- Pharmacology Unit, Faculty of Medicine and Health Sciences, Universitat Rovira i Virgili, C./ St. Llorenç 21, 43201, Reus, Tarragona, Spain
| | - Maria G Kurnikova
- Chemistry Department, Carnegie Mellon University, 4400 Fifth Ave, Pittsburgh, PA, 15213, USA
| | - Jon W Johnson
- Department of Neuroscience and Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Mercè Pallàs
- Pharmacology Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Neurosciences (NeuroUB), Universitat de Barcelona, Av. Joan XXIII 27-31, 08028, Barcelona, Spain
| | - Santiago Vázquez
- Laboratori de Química Farmacèutica (Unitat Associada al CSIC), Facultat de Farmàcia i Ciències de l'Alimentació i Institut de Biomedicina (IBUB), Universitat de Barcelona, Av. Joan XXIII, 27-31, 08028, Barcelona, Spain.
| |
Collapse
|
6
|
Kovač V, Čurin Šerbec V. Prion Protein: The Molecule of Many Forms and Faces. Int J Mol Sci 2022; 23:ijms23031232. [PMID: 35163156 PMCID: PMC8835406 DOI: 10.3390/ijms23031232] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/10/2022] [Accepted: 01/21/2022] [Indexed: 02/06/2023] Open
Abstract
Cellular prion protein (PrPC) is a glycosylphosphatidylinositol (GPI)-anchored protein most abundantly found in the outer membrane of neurons. Due to structural characteristics (a flexible tail and structured core), PrPC interacts with a wide range of partners. Although PrPC has been proposed to be involved in many physiological functions, only peripheral nerve myelination homeostasis has been confirmed as a bona fide function thus far. PrPC misfolding causes prion diseases and PrPC has been shown to mediate β-rich oligomer-induced neurotoxicity in Alzheimer’s and Parkinson’s disease as well as neuroprotection in ischemia. Upon proteolytic cleavage, PrPC is transformed into released and attached forms of PrP that can, depending on the contained structural characteristics of PrPC, display protective or toxic properties. In this review, we will outline prion protein and prion protein fragment properties as well as overview their involvement with interacting partners and signal pathways in myelination, neuroprotection and neurodegenerative diseases.
Collapse
|
7
|
Qiu H, Qian T, Wu T, Gao T, Xing Q, Wang L. Src Family Kinases Inhibition Ameliorates Hypoxic-Ischemic Brain Injury in Immature Rats. Front Cell Neurosci 2022; 15:746130. [PMID: 34992524 PMCID: PMC8724194 DOI: 10.3389/fncel.2021.746130] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 11/24/2021] [Indexed: 01/06/2023] Open
Abstract
Hypoxic-ischemic (HI) injury is one of the initial factors contributing to neonatal brain injury. Src family kinases (SFKs) are considered to act as molecular hubs for N-methyl-d-aspartate receptor (NMDAR) regulation and participate in the HI injury process. The objectives of this study were to evaluate the levels of phospho-Src (p-Src), the relationship between NMDARs and SFKs, and the effects of SFK inhibition on an immature rat HI brain injury model. The model was induced in 3-day-old Sprague–Dawley rats using the Rice-Vannucci model operation. The level of p-Src was evaluated using Western blotting. The association of NMDARs with SFKs was detected using Western blotting and coimmunoprecipitation. After intraperitoneal injection of 4-amino-5-(4-chlorophenyl)-7-(t-butyl) pyrazolo [3,4-d] pyrimidine (PP2), an SFK-selective inhibitor, neuropathological changes were observed by performing H&E and immunofluorescence staining, and the neurological functions were assessed using the following behavioral tests: modified neurological severity score, open field test, and Morris water maze test. The levels of p-Src first decreased at 0 h after injury, increased at 2 h after injury, and continuously decreased from 6 h to 3 days. Along with the increased p-Src levels observed at 2 h after injury, the phosphorylation of NMDAR subunit NR2B at tyrosine 1472 was increased. Following the administration of PP2, the increased p-Src and NMDAR-2B levels detected at 2 h after injury were decreased, and tissue injury and myelin basic protein expression were improved at 7 days after injury. The PP2 intervention improved the performance of injured rats on behavioral tests. In conclusion, we determined the patterns of p-Src expression after HI brain injury in immature rats and showed a relationship with the activated NMDA receptor. The inhibition of p-Src ameliorates neuropathological changes and damages neurological functions induced by HI injury.
Collapse
Affiliation(s)
- Han Qiu
- National Health Commission Key Laboratory of Neonatal Diseases, Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
| | - Tianyang Qian
- National Health Commission Key Laboratory of Neonatal Diseases, Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
| | - Tong Wu
- National Health Commission Key Laboratory of Neonatal Diseases, Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
| | - Ting Gao
- National Health Commission Key Laboratory of Neonatal Diseases, Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
| | - Qinghe Xing
- Department of Neonatology, Children's Hospital of Fudan University and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Laishuan Wang
- National Health Commission Key Laboratory of Neonatal Diseases, Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
| |
Collapse
|
8
|
Zhao M, Yao Y, Du J, Kong L, Zhao T, Wu D, Man L, Zhou W. 6-Gingerol Alleviates Neonatal Hypoxic-Ischemic Cerebral and White Matter Injury and Contributes to Functional Recovery. Front Pharmacol 2021; 12:707772. [PMID: 34630084 PMCID: PMC8492979 DOI: 10.3389/fphar.2021.707772] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 09/08/2021] [Indexed: 12/19/2022] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) is one main cause of neonatal death and disability, causing substantial injury to white and gray matter, which can lead to severe neurobehavioral dysfunction, including intellectual disability and dyskinesia. Inflammation, nerve cell death, and white matter injury are important factors in the pathological process of HIE. 6-Gingerol is a ginger extract, which reduces inflammatory response and cell death. However, the role of 6-Gingerol in neonatal hypoxic-ischemic brain injury (HIBI) remains unknown. In this study, we constructed a mouse HIBI model and analyzed the protective effect of 6-Gingerol on HIBI by using behavioral tests, histological staining, qPCR and western blot. Here, we found that 6-Gingerol treatment could alleviate HIBI and improve short-term reflex performance, which is closely related to cell death and neuroinflammation. Additionally, 6-Gingerol reduced neuronal apoptosis, pro-inflammatory factor release, as well as microglial activation. Furthermore, 6-Gingerol significantly improved motor disability, which is associated with white matter damage. Thus, our results showed that 6-Gingerol could reduce the loss of myelin sheaths, alleviate cell death of oligodendrocytes, and stimulate the maturation of oligodendrocytes. In terms of mechanism, we found that 6-Gingerol decreased histone H3K27me3 levels, activated AKT pathway and inhibited the activation of ERK and NF-κB pathway at 3 days post-HIBI. Taken together, our data clearly indicate that 6-Gingerol plays a neuroprotective role against HIBI by epigenetic modification and regulation of AKT, ERK, and NF-κB pathways, inhibiting inflammatory responses and reducing cell death.
Collapse
Affiliation(s)
- Man Zhao
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yuan Yao
- Centre for Sports and Exercise Science, School of Sport, Rehabilitation and Exercise Sciences, University of Essex, Colchester, United Kingdom
| | - Jingyi Du
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Liang Kong
- Department of Clinical Laboratory, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Tiantian Zhao
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Dong Wu
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lajie Man
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wenjuan Zhou
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
9
|
Zhang L, Wang L, Xiao H, Gan H, Chen H, Zheng S, Jian D, Zhai X, Jiang N, Jing Z, Liang P. Tyrosine kinase Fyn promotes apoptosis after intracerebral hemorrhage in rats by activating Drp1 signaling. J Mol Med (Berl) 2021; 99:359-371. [PMID: 33409551 DOI: 10.1007/s00109-020-02022-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 11/30/2020] [Accepted: 12/08/2020] [Indexed: 02/07/2023]
Abstract
Tyrosine kinase Fyn is a member of the Src kinase family, which is involved in neuroinflammation, apoptosis, and oxidative stress. Its role in intracerebral hemorrhage (ICH) is not fully understood. In this study, we found that Fyn was significantly elevated in human brain tissue after ICH. Accordingly, we investigated the role of Fyn in a rat ICH model, which was constructed by injecting blood into the right basal ganglia. In this model, Fyn expression was significantly upregulated in brain tissue adjacent to the hematoma. SiRNA-induced Fyn knockdown was neuroprotective for secondary cerebral damage, as demonstrated by reduced brain edema, suppression of the modified neurological severity score, and mitigation of blood-brain barrier permeability and neuronal damage. Fyn downregulation reduced apoptosis following ICH, as indicated by downregulation of apoptosis-related proteins AIF, Cyt.c, caspase 3, and Bax; upregulation of anti-apoptosis-related protein Bcl-2; and decreased tunnel staining. Mdivi-1, a Drp1 inhibitor, reversed Fyn overexpression induced pro-apoptosis. However, Fyn did not significantly affect inflammation-related proteins NF-κB, TNF-α, caspase 1, MPO, IL-1β, or IL-18 after ICH. Fyn activated Drp1 signaling by phosphorylating Drp1 at serine 616, which increased apoptosis after ICH in rats. This study clarifies the relationship between Fyn, apoptosis, and inflammation following ICH and provides a new strategy for exploring the prevention and treatment of ICH. KEY MESSAGES: ICH induced an increase in Fyn expression in human and rat cerebral tissues. Knockdown of Fyn prevented cerebral damage following ICH. Inhibition of Fyn had no significant effects on inflammatory responses. However, the downregulation of Fyn exerted neuroprotective effects on apoptosis. Fyn perturbed ICH-induced cell apoptosis by interacting with and phosphorylating (Ser616) Drp1 in a rat ICH model.
Collapse
Affiliation(s)
- Li Zhang
- Department of Neurosurgery, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, People's Republic of China.,National Clinical Research Center for Child Health and Disorders, Chongqing, People's Republic of China.,China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing, People's Republic of China
| | - Lu Wang
- Department of Neurosurgery, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, People's Republic of China.,National Clinical Research Center for Child Health and Disorders, Chongqing, People's Republic of China.,China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing, People's Republic of China
| | - Han Xiao
- Department of Neurosurgery, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, People's Republic of China.,National Clinical Research Center for Child Health and Disorders, Chongqing, People's Republic of China.,China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing, People's Republic of China
| | - Hui Gan
- Department of Neurosurgery, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, People's Republic of China.,National Clinical Research Center for Child Health and Disorders, Chongqing, People's Republic of China.,China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing, People's Republic of China
| | - Hui Chen
- Department of Neurosurgery, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, People's Republic of China.,National Clinical Research Center for Child Health and Disorders, Chongqing, People's Republic of China.,China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing, People's Republic of China
| | - Shuyue Zheng
- Department of Neurosurgery, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, People's Republic of China.,National Clinical Research Center for Child Health and Disorders, Chongqing, People's Republic of China.,China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing, People's Republic of China
| | - Dan Jian
- Department of Neurosurgery, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, People's Republic of China.,National Clinical Research Center for Child Health and Disorders, Chongqing, People's Republic of China.,China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing, People's Republic of China
| | - Xuan Zhai
- Department of Neurosurgery, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China. .,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, People's Republic of China. .,National Clinical Research Center for Child Health and Disorders, Chongqing, People's Republic of China. .,China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing, People's Republic of China.
| | - Ning Jiang
- Institute of Neuroscience, School of Basic Medicine, Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Zhao Jing
- Institute of Neuroscience, School of Basic Medicine, Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Ping Liang
- Department of Neurosurgery, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, People's Republic of China.,National Clinical Research Center for Child Health and Disorders, Chongqing, People's Republic of China.,China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing, People's Republic of China
| |
Collapse
|
10
|
Zhang X, Peng K, Zhang X. The Function of the NMDA Receptor in Hypoxic-Ischemic Encephalopathy. Front Neurosci 2020; 14:567665. [PMID: 33117117 PMCID: PMC7573650 DOI: 10.3389/fnins.2020.567665] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 08/28/2020] [Indexed: 12/17/2022] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) is one of the main forms of neonatal brain injury which could lead to neonatal disability or even cause neonatal death. Therefore, HIE strongly affects the health of newborns and brings heavy burden to the family and society. It has been well studied that N-methyl-D-aspartate (NMDA) receptors are involved in the excitotoxicity induced by hypoxia ischemia in adult brain. Recently, it has been shown that the NMDA receptor also plays important roles in HIE. In the present review, we made a summary of the molecular mechanism of NMDA receptor in the pathological process of HIE, focusing on the distinct role of GluN2A- and GluN2B-containing NMDA receptor subtypes and aiming to provide some insights into the clinical treatment and drug development of HIE.
Collapse
|
11
|
Niu Q, Sun W, Chen Q, Long Y, Cao W, Wen S, Li A, Dong F, Shi H. Protective Effects of Ischemic Postconditioning on Livers in Rats with Limb Ischemia-Reperfusion via Glycogen Synthase Kinase 3 beta (GSK-3β)/Fyn/Nuclear Receptor-Erythroid-2-Related Factor (Nrf2) Pathway. Med Sci Monit 2020; 26:e923049. [PMID: 32686659 PMCID: PMC7392060 DOI: 10.12659/msm.923049] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background Ischemia/reperfusion (I/R) injury not only exists in ischemic tissues and organs, but also can cause damage to distant tissues and organs. As the largest metabolic organ of the human body, the liver is very vulnerable to injury after limb I/R. However, the mechanism of liver injury caused by limb I/R injury has not been fully elucidated. This study investigated the effect and mechanism of ischemic postconditioning (IPO) on the liver after hindlimb I/R in rats. Material/Methods A rat model of hindlimb I/R was established and treated by IPO. Liver function, changes of oxidative stress index and inflammation, Bcl-2 and Bax proteins, and apoptosis were assessed. The structural changes were observed by electron microscopy. GSK-3β/Fyn/Nrf2 levels were detected by quantitative PCR and Western blot. Results IPO significantly reduced serum AST, ALP, LDH, and ALT levels induced by I/R. Compared with the I/R group, the levels of SOD, GSH-Px, and CAT in the IPO group were significantly increased, while the levels of MDA, MPO, and ROS were significantly decreased. The IPO group had significantly higher Bcl-2 level and significantly lower Bax level compared to the I/R group. Consistently, IPO decreased the apoptosis rate induced by I/R. Furthermore, IPO lowered the levels of TNF-α, IL-1β, IL-10, and INF-γ and alleviated the ultrastructural changes of hepatocytes. Finally, Nrf2, Fyn, and GSK-3β mRNA and protein levels in the IPO group were significantly higher than in the I/R group. Conclusions IPO protects against liver injury caused by I/R injury of the hindlimb, possibly via the GSK-3β/Fyn/Nrf2 pathway.
Collapse
Affiliation(s)
- Qibing Niu
- Department of Vascular Surgery, People's Hospital in Gansu Province, Gansu University of Chinese Medicine, Lanzhou, Gansu, China (mainland)
| | - Wanli Sun
- Department of Vascular Surgery, People's Hospital in Gansu Province, Gansu University of Chinese Medicine, Lanzhou, Gansu, China (mainland)
| | - Quan Chen
- Department of Vascular Surgery, People's Hospital in Gansu Province, Gansu University of Chinese Medicine, Lanzhou, Gansu, China (mainland)
| | - Yang Long
- Department of Vascular Surgery, People's Hospital in Gansu Province, Gansu University of Chinese Medicine, Lanzhou, Gansu, China (mainland)
| | - Wanjun Cao
- Department of Vascular Surgery, People's Hospital in Gansu Province, Gansu University of Chinese Medicine, Lanzhou, Gansu, China (mainland)
| | - Shiqi Wen
- Department of Vascular Surgery, People's Hospital in Gansu Province, Gansu University of Chinese Medicine, Lanzhou, Gansu, China (mainland)
| | - Anqiang Li
- Department of Vascular Surgery, People's Hospital in Gansu Province, Gansu University of Chinese Medicine, Lanzhou, Gansu, China (mainland)
| | - Fang Dong
- Department of Vascular Surgery, People's Hospital in Gansu Province, Gansu University of Chinese Medicine, Lanzhou, Gansu, China (mainland)
| | - Hao Shi
- Department of Vascular Surgery, People's Hospital in Gansu Province, Gansu University of Chinese Medicine, Lanzhou, Gansu, China (mainland)
| |
Collapse
|
12
|
Puig B, Yang D, Brenna S, Altmeppen HC, Magnus T. Show Me Your Friends and I Tell You Who You Are: The Many Facets of Prion Protein in Stroke. Cells 2020; 9:E1609. [PMID: 32630841 PMCID: PMC7407975 DOI: 10.3390/cells9071609] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 06/24/2020] [Accepted: 06/27/2020] [Indexed: 12/12/2022] Open
Abstract
Ischemic stroke belongs to the leading causes of mortality and disability worldwide. Although treatments for the acute phase of stroke are available, not all patients are eligible. There is a need to search for therapeutic options to promote neurological recovery after stroke. The cellular prion protein (PrPC) has been consistently linked to a neuroprotective role after ischemic damage: it is upregulated in the penumbra area following stroke in humans, and animal models of stroke have shown that lack of PrPC aggravates the ischemic damage and lessens the functional outcome. Mechanistically, these effects can be linked to numerous functions attributed to PrPC: (1) as a signaling partner of the PI3K/Akt and MAPK pathways, (2) as a regulator of glutamate receptors, and (3) promoting stem cell homing mechanisms, leading to angio- and neurogenesis. PrPC can be cleaved at different sites and the proteolytic fragments can account for the manifold functions. Moreover, PrPC is present on extracellular vesicles (EVs), released membrane particles originating from all types of cells that have drawn attention as potential therapeutic tools in stroke and many other diseases. Thus, identification of the many mechanisms underlying PrPC-induced neuroprotection will not only provide further understanding of the physiological functions of PrPC but also new ideas for possible treatment options after ischemic stroke.
Collapse
Affiliation(s)
- Berta Puig
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (D.Y.); (S.B.); (T.M.)
| | - Denise Yang
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (D.Y.); (S.B.); (T.M.)
| | - Santra Brenna
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (D.Y.); (S.B.); (T.M.)
| | | | - Tim Magnus
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (D.Y.); (S.B.); (T.M.)
| |
Collapse
|
13
|
Fyn Tyrosine Kinase as Harmonizing Factor in Neuronal Functions and Dysfunctions. Int J Mol Sci 2020; 21:ijms21124444. [PMID: 32580508 PMCID: PMC7352836 DOI: 10.3390/ijms21124444] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/19/2020] [Accepted: 06/20/2020] [Indexed: 12/25/2022] Open
Abstract
Fyn is a non-receptor or cytoplasmatic tyrosine kinase (TK) belonging to the Src family kinases (SFKs) involved in multiple transduction pathways in the central nervous system (CNS) including synaptic transmission, myelination, axon guidance, and oligodendrocyte formation. Almost one hundred years after the original description of Fyn, this protein continues to attract extreme interest because of its multiplicity of actions in the molecular signaling pathways underlying neurodevelopmental as well as neuropathologic events. This review highlights and summarizes the most relevant recent findings pertinent to the role that Fyn exerts in the brain, emphasizing aspects related to neurodevelopment and synaptic plasticity. Fyn is a common factor in healthy and diseased brains that targets different proteins and shapes different transduction signals according to the neurological conditions. We will primarily focus on Fyn-mediated signaling pathways involved in neuronal differentiation and plasticity that have been subjected to considerable attention lately, opening the fascinating scenario to target Fyn TK for the development of potential therapeutic interventions for the treatment of CNS injuries and certain neurodegenerative disorders like Alzheimer’s disease.
Collapse
|
14
|
Barks AK, Beeson MM, Matveeva T, Gale JJ, Rao R, Tran PV. Perinatal Ischemia Alters Global Expression of Synaptosomal Proteins Critical for Neural Plasticity in the Developing Mouse Brain. Dev Neurosci 2019; 40:1-13. [PMID: 31207599 DOI: 10.1159/000499126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 02/25/2019] [Indexed: 11/19/2022] Open
Abstract
Ischemic perinatal stroke (IPS) affects 1 in 2,300-5,000 live births. Despite a survival rate >95%, approximately 60% of IPS infants develop motor and cognitive impairments. Given the importance of axonal growth and synaptic plasticity in neurocognitive development, our objective was to identify the molecular pathways underlying IPS-associated synaptic dysfunction using a mouse model. IPS was induced by unilateral ligation of the common carotid artery of postnatal day 10 (P10) mice. Five days after ischemia, sensorimotor and motor functions were assessed by vibrissae-evoked forepaw placement and the tail suspension test respectively, showing evidence of greater impairments in male pups than in female pups. Twenty-four hours after ischemia, both hemispheres were collected and synaptosomal proteins then prepared for quantification, using isobaric tags for relative and absolute quantitation. Seventy-two of 1,498 qualified proteins were altered in the ischemic hemisphere. Ingenuity Pathway Analysis was used to map these proteins onto molecular networks indicative of reduced neuronal proliferation, survival, and synaptic plasticity, accompanied by reduced PKCα signaling in male, but not female, pups. These effects also occurred in the non-ischemic hemisphere when compared with sham controls. The altered signaling effects may contribute to the sex-specific neurodevelopmental dysfunction following IPS, highlighting potential pathways for targeting during treatment.
Collapse
Affiliation(s)
- Amanda K Barks
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Montana M Beeson
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Tatyana Matveeva
- Department of Psychology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jonathan J Gale
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Raghavendra Rao
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Phu V Tran
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA,
| |
Collapse
|
15
|
Rodent Models of Developmental Ischemic Stroke for Translational Research: Strengths and Weaknesses. Neural Plast 2019; 2019:5089321. [PMID: 31093271 PMCID: PMC6476045 DOI: 10.1155/2019/5089321] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 12/19/2018] [Accepted: 02/06/2019] [Indexed: 12/25/2022] Open
Abstract
Cerebral ischemia can occur at any stage in life, but clinical consequences greatly differ depending on the developmental stage of the affected brain structures. Timing of the lesion occurrence seems to be critical, as it strongly interferes with neuronal circuit development and determines the way spontaneous plasticity takes place. Translational stroke research requires the use of animal models as they represent a reliable tool to understand the pathogenic mechanisms underlying the generation, progression, and pathological consequences of a stroke. Moreover, in vivo experiments are instrumental to investigate new therapeutic strategies and the best temporal window of intervention. Differently from adults, very few models of the human developmental stroke have been characterized, and most of them have been established in rodents. The models currently used provide a better understanding of the molecular factors involved in the effects of ischemia; however, they still hold many limitations due to matching developmental stages across different species and the complexity of the human disorder that hardly can be described by segregated variables. In this review, we summarize the key factors contributing to neonatal brain vulnerability to ischemic strokes and we provide an overview of the advantages and limitations of the currently available models to recapitulate different aspects of the human developmental stroke.
Collapse
|
16
|
Hara Y, Crimins JL, Puri R, Wang ACJ, Motley SE, Yuk F, Ramos TM, Janssen WGM, Rapp PR, Morrison JH. Estrogen Alters the Synaptic Distribution of Phospho-GluN2B in the Dorsolateral Prefrontal Cortex While Promoting Working Memory in Aged Rhesus Monkeys. Neuroscience 2019; 394:303-315. [PMID: 30482274 DOI: 10.1016/j.neuroscience.2018.09.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 09/05/2018] [Accepted: 09/17/2018] [Indexed: 01/06/2023]
Abstract
Age- and menopause-related deficits in working memory can be partially restored with estradiol replacement in women and female nonhuman primates. Working memory is a cognitive function reliant on persistent firing of dorsolateral prefrontal cortex (dlPFC) neurons that requires the activation of GluN2B-containing glutamate NMDA receptors. We tested the hypothesis that the distribution of phospho-Tyr1472-GluN2B (pGluN2B), a predominant form of GluN2B seen at the synapse, is sensitive to aging or estradiol treatment and coupled to working memory performance. First, ovariectomized young and aged rhesus monkeys (Macaca mulatta) received long-term cyclic vehicle (V) or estradiol (E) treatment and were tested on the delayed response (DR) test of working memory. Then, serial section electron microscopic immunocytochemistry was performed to quantitatively assess the subcellular distribution of pGluN2B. While the densities of pGluN2B immunogold particles in dlPFC dendritic spines were not different across age or treatment groups, the percentage of gold particles located within the synaptic compartment was significantly lower in aged-E monkeys compared to young-E and aged-V monkeys. On the other hand, the percentage of pGluN2B gold particles in the spine cytoplasm was decreased with E treatment in young, but increased with E in aged monkeys. In aged monkeys, DR average accuracy inversely correlated with the percentage of synaptic pGluN2B, while it positively correlated with the percentage of cytoplasmic pGluN2B. Together, E replacement may promote cognitive health in aged monkeys, in part, by decreasing the relative representation of synaptic pGluN2B and potentially protecting the dlPFC from calcium toxicity.
Collapse
Affiliation(s)
- Yuko Hara
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Johanna L Crimins
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Rishi Puri
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Athena C J Wang
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Sarah E Motley
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; California National Primate Research Center, Davis, CA 95616, United States
| | - Frank Yuk
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Tiffany M Ramos
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - William G M Janssen
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Peter R Rapp
- National Institute on Aging, Laboratory of Behavioral Neuroscience, Baltimore, MD 21224, United States
| | - John H Morrison
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Department of Geriatrics and Palliative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; California National Primate Research Center, Davis, CA 95616, United States; Department of Neurology, School of Medicine, University of California, Davis, CA 95616, United States.
| |
Collapse
|
17
|
Liang X, Yao Y, Lin Y, Kong L, Xiao H, Shi Y, Yang J. Panaxadiol inhibits synaptic dysfunction in Alzheimer's disease and targets the Fyn protein in APP/PS1 mice and APP-SH-SY5Y cells. Life Sci 2019; 221:35-46. [PMID: 30735733 DOI: 10.1016/j.lfs.2019.02.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 02/02/2019] [Accepted: 02/04/2019] [Indexed: 12/12/2022]
Abstract
AIM Alzheimer's disease (AD), a neurodegenerative disease, is characterized by memory loss and synaptic damage. Up to now, there are limited drugs to cure or delay the state of this illness. Recently, the Fyn tyrosine kinase is implicated in AD pathology triggered by synaptic damage. Thus, Fyn inhibition may prevent or delay the AD progression. Therefore, in this paper, we investigated whether Panaxadiol could decrease synaptic damage in AD and the underlying mechanism. MAIN METHODS The ability of learning and memory of mice has detected by Morris Water Maze. The pathological changes detected by H&E staining and Nissl staining. The percentage of cell apoptosis and the calcium concentration were detected by Flow Cytometry in vitro. The amount of synaptic protein and related proteins in the Fyn/GluN2B/CaMKIIα signaling pathway were detected by Western Blot. KEY FINDINGS In the present article, Panaxadiol could significantly improve the ability of learning and memory of mice and reduce its synaptic dysfunction. Panaxadiol could down-regulate GluN2B's phosphorylation level by inhibition Fyn kinase activity, Subsequently, decrease Ca2+-mediated synaptic damage, reducing LDH leakage, inhibiting apoptosis in AD, resulting in facilitating the cells survival. For the underlying molecular mechanism, we used PP2 to block the Fyn/GluN2B/CaMKIIα signaling pathway. The results from WB showed that the expression of related proteins in the Fyn signaling pathway decreased with PP2 treated. SIGNIFICANCE Our results indicate that Panaxadiol could decrease synaptic damage, which will cause AD via inhibition of the Fyn/GluN2B/CaMKIIα signaling pathway. Thus, the Panaxadiol is a best promising candidate to test as a potential therapy for AD.
Collapse
Affiliation(s)
- Xicai Liang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China
| | - Yingjia Yao
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China
| | - Ying Lin
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China
| | - Liang Kong
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China
| | - Honghe Xiao
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China
| | - Yue Shi
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China
| | - Jingxian Yang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China.
| |
Collapse
|
18
|
Neuroprotection of Cytisine Against Cerebral Ischemia–Reperfusion Injury in Mice by Regulating NR2B-ERK/CREB Signal Pathway. Neurochem Res 2018; 43:1575-1586. [DOI: 10.1007/s11064-018-2572-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 05/09/2018] [Accepted: 06/08/2018] [Indexed: 10/28/2022]
|
19
|
Al Mamun A, Yu H, Romana S, Liu F. Inflammatory Responses are Sex Specific in Chronic Hypoxic-Ischemic Encephalopathy. Cell Transplant 2018; 27:1328-1339. [PMID: 29692197 PMCID: PMC6168990 DOI: 10.1177/0963689718766362] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Neonatal hypoxic-ischemic encephalopathy (HIE) is increasingly recognized as a sexually dimorphic disease. Male infants are not only more vulnerable to ischemic insult; they also suffer more long-term cognitive deficits compared with females with comparable brain damage. The innate immune response plays a fundamental role in mediating acute neonatal HIE injury. However, the mechanism underlying the sex difference in chronic HIE is still elusive. The present study investigated the sex difference in HIE outcomes and inflammatory response in the chronic stage (30 days after HIE). Postnatal day 10 (P10) male and female C57BL/6 pups were subjected to 60-min Rice-Vanucci model (RVM) to induce HIE. Brain atrophy and behavioral deficits were analyzed to measure stroke outcomes at 30 days of HIE. Flow cytometry (FC) was performed to examine central (microglial activation) and peripheral immune responses. Serum levels of cytokines and sex hormones were determined by enzyme-linked immunosorbent assay (ELISA). Neurogenesis was quantified by 5-Bromo-2'-deoxyuridine (BrdU) incorporation with neurons. Results showed males had worse HIE outcomes than females at the endpoint. Female microglia exhibited a more robust anti-inflammatory response that was corresponding to an enhanced expression of CX3C chemokine receptor 1 (CX3CR1) than males. More infiltration of peripheral lymphocytes was seen in male vs. female HIE brains. Cytokine levels of tumor necrosis factor (TNF)-α and interleukin (IL)-10 were more upregulated in males and females respectively than their counterparts. Neurogenesis was more highly induced in females vs. males. No significant difference in circulating hormonal level was found between males and females after HIE. We conclude that a sex dichotomy in pro- and anti-inflammatory response underlies the sex-specific chronic HIE outcomes, and an enhanced neurogenesis in females also contribute to the sex difference.
Collapse
Affiliation(s)
- Abdullah Al Mamun
- 1 Department of Neurology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Haifu Yu
- 1 Department of Neurology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA.,2 Department of Neurology, Shanghai Jiaotong University Sixth People's Hospital South Campus, Shanghai Fengxian District Central Hospital, Shanghai, China
| | - Sharmeen Romana
- 1 Department of Neurology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Fudong Liu
- 1 Department of Neurology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| |
Collapse
|
20
|
Effects of Src kinase inhibition on expression of pro-caspase-2 after brain hypoxia in a piglet animal model. Neuroreport 2018; 28:770-773. [PMID: 28658050 DOI: 10.1097/wnr.0000000000000835] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Caspase-2 has features of both initiator and effector caspases. Previously, we have shown that brain hypoxia-induced production of caspases 1, 3, 8, and 9 is Src kinase mediated, a nonreceptor intracellular family of kinases. The present study tests the hypothesis that hypoxia results in increased expression of caspase-2 and this effect is mediated by Src kinase. Two to three days old newborn piglets were subjected to normoxia, hypoxia (Hx, FiO2 7%), and Src kinase inhibition (using PP2, 1 mg/kg, intravenous), followed by 30 min of acute hypoxia (Hx+PP2). ATP and phosphocreatine were determined biochemically to verify energy molecule depletion in the hypoxic groups. The cytosolic brain function was isolated and a western blot analysis was carried out using an antibody specific for the caspase-2. The immune-complex band density was expressed as OD/mm. Caspase-2 expression was increased two-fold in the Hx group. After Src kinase inhibition followed by hypoxia, caspase-2 expression was similar to normoxia levels. We conclude that hypoxia results in increased expression of caspase-2 protein in the cytosolic fraction of the cerebral cortex of the newborn piglets. This increase is mediated by Src kinase.
Collapse
|
21
|
Sun Y, Chen Y, Zhan L, Zhang L, Hu J, Gao Z. The role of non-receptor protein tyrosine kinases in the excitotoxicity induced by the overactivation of NMDA receptors. Rev Neurosci 2018; 27:283-9. [PMID: 26540220 DOI: 10.1515/revneuro-2015-0037] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 09/17/2015] [Indexed: 11/15/2022]
Abstract
Protein tyrosine phosphorylation is one of the primary modes of regulation of N-methyl-d-aspartate (NMDA) receptors. The non-receptor tyrosine kinases are one of the two types of protein tyrosine kinases that are involved in this process. The overactivation of NMDA receptors is a primary reason for neuron death following cerebral ischemia. Many studies have illustrated the important role of non-receptor tyrosine kinases in ischemia insults. This review introduces the roles of Src, Fyn, focal adhesion kinase, and proline-rich tyrosine kinase 2 in the excitotoxicity induced by the overactivation of NMDA receptors following cerebral ischemia.
Collapse
|
22
|
Lu F, Shao G, Wang Y, Guan S, Burlingame AL, Liu X, Liang X, Knox R, Ferriero DM, Jiang X. Hypoxia-ischemia modifies postsynaptic GluN2B-containing NMDA receptor complexes in the neonatal mouse brain. Exp Neurol 2017; 299:65-74. [PMID: 28993251 DOI: 10.1016/j.expneurol.2017.10.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 09/09/2017] [Accepted: 10/05/2017] [Indexed: 01/08/2023]
Abstract
The N-methyl-d-aspartate-type glutamate receptor (NMDAR)-associated multiprotein complexes are indispensable for synaptic plasticity and cognitive functions. While purification and proteomic analyses of these signaling complexes have been performed in adult rodent and human brain, much less is known about the protein composition of NMDAR complexes in the developing brain and their modifications by neonatal hypoxic-ischemic (HI) brain injury. In this study, the postsynaptic density proteins were prepared from postnatal day 9 naïve, sham-operated and HI-injured mouse cortex. The GluN2B-containing NMDAR complexes were purified by immunoprecipitation with a mouse GluN2B antibody and subjected to mass spectrometry analysis for determination of the GluN2B binding partners. A total of 71 proteins of different functional categories were identified from the naïve animals as native GluN2B-interacting partners in the developing mouse brain. Neonatal HI reshaped the postsynaptic GluN2B interactome by recruiting new proteins, including multiple kinases, into the complexes; and modifying the existing associations within 1h of reperfusion. The early responses of postsynaptic NMDAR complexes and their related signaling networks may contribute to molecular processes leading to cell survival or death, brain damage and/or neurological disorders in term infants with neonatal encephalopathy.
Collapse
Affiliation(s)
- Fuxin Lu
- Department of Pediatrics, University of California San Francisco, CA, USA
| | - Guo Shao
- Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Baotou, China
| | - Yongqiang Wang
- Department of Cellular & Molecular Pharmacology, University of California San Francisco, CA, USA; Howard Hughes Medical Institute, University of California, San Francisco, CA, USA
| | - Shenheng Guan
- Department of Pharmaceutical Chemistry, University of California San Francisco, CA, USA
| | - Alma L Burlingame
- Department of Pharmaceutical Chemistry, University of California San Francisco, CA, USA
| | - Xuemei Liu
- Central Laboratory, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiao Liang
- Central Laboratory, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Renatta Knox
- Department of Pediatrics, Weill Cornell Medical College, New York, NY, USA
| | - Donna M Ferriero
- Department of Pediatrics, University of California San Francisco, CA, USA; Department of Neurology, University of California San Francisco, CA, USA
| | - Xiangning Jiang
- Department of Pediatrics, University of California San Francisco, CA, USA.
| |
Collapse
|
23
|
Zhao F, Qu Y, Zhu J, Zhang L, Huang L, Liu H, Li S, Mu D. miR-30d-5p Plays an Important Role in Autophagy and Apoptosis in Developing Rat Brains After Hypoxic–Ischemic Injury. J Neuropathol Exp Neurol 2017; 76:709-719. [DOI: 10.1093/jnen/nlx052] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
24
|
BDNF Contributes to Spinal Long-Term Potentiation and Mechanical Hypersensitivity Via Fyn-Mediated Phosphorylation of NMDA Receptor GluN2B Subunit at Tyrosine 1472 in Rats Following Spinal Nerve Ligation. Neurochem Res 2017; 42:2712-2729. [DOI: 10.1007/s11064-017-2274-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 04/01/2017] [Accepted: 04/18/2017] [Indexed: 11/26/2022]
|
25
|
Sun Y, Zhan L, Cheng X, Zhang L, Hu J, Gao Z. The Regulation of GluN2A by Endogenous and Exogenous Regulators in the Central Nervous System. Cell Mol Neurobiol 2017; 37:389-403. [PMID: 27255970 DOI: 10.1007/s10571-016-0388-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 05/25/2016] [Indexed: 12/25/2022]
Abstract
The NMDA receptor is the most widely studied ionotropic glutamate receptor, and it is central to many physiological and pathophysiological processes in the central nervous system. GluN2A is one of the two main types of GluN2 NMDA receptor subunits in the forebrain. The proper activity of GluN2A is important to brain function, as the abnormal regulation of GluN2A may induce some neuropsychiatric disorders. This review will examine the regulation of GluN2A by endogenous and exogenous regulators in the central nervous system.
Collapse
Affiliation(s)
- Yongjun Sun
- Department of Pharmacy, Hebei University of Science and Technology, Yuhua East Road 70, Shijiazhuang, 050018, People's Republic of China
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, 050018, People's Republic of China
| | - Liying Zhan
- Department of Pharmacy, Hebei University of Science and Technology, Yuhua East Road 70, Shijiazhuang, 050018, People's Republic of China
| | - Xiaokun Cheng
- North China Pharmaceutical Group New Drug Research and Development Co., Ltd, Shijiazhuang, 050015, People's Republic of China
| | - Linan Zhang
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, 050017, People's Republic of China
| | - Jie Hu
- School of Nursing, Hebei Medical University, Shijiazhuang, 050017, People's Republic of China
| | - Zibin Gao
- Department of Pharmacy, Hebei University of Science and Technology, Yuhua East Road 70, Shijiazhuang, 050018, People's Republic of China.
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, 050018, People's Republic of China.
- State Key Laboratory Breeding Base-Hebei Province Key Laboratory of Molecular Chemistry for Drug, Shijiazhuang, 050018, People's Republic of China.
| |
Collapse
|
26
|
Shao G, Wang Y, Guan S, Burlingame AL, Lu F, Knox R, Ferriero DM, Jiang X. Proteomic Analysis of Mouse Cortex Postsynaptic Density following Neonatal Brain Hypoxia-Ischemia. Dev Neurosci 2017; 39:66-81. [PMID: 28315865 PMCID: PMC5519436 DOI: 10.1159/000456030] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 01/11/2017] [Indexed: 02/02/2023] Open
Abstract
Proteomics of the synapses and postsynaptic densities (PSDs) have provided a deep understanding of protein composition and signal networks in the adult brain, which underlie neuronal plasticity and neurodegenerative or psychiatric disorders. However, there is a paucity of knowledge about the architecture and organization of PSDs in the immature brain, and how it is modified by brain injury in an early developing stage. Mass spectrometry (MS)-based proteomic analysis was performed on PSDs prepared from cortices of postnatal day 9 naïve mice or pups which had suffered hypoxic-ischemic (HI) brain injury. 512 proteins of different functional groups were identified from PSDs collected 1 h after HI injury, among which 60 have not been reported previously. Seven newly identified proteins involved in neural development were highlighted. HI injury increased the yield of PSDs at early time points upon reperfusion, and multiple proteins were recruited into PSDs following the insult. Quantitative analysis was performed using spectral counting, and proteins whose relative expression was more than 50% up- or downregulated compared to the sham animals 1 h after HI insult were reported. Validation with Western blotting demonstrated changes in expression and phosphorylation of the N-methyl-D-aspartate receptor, activation of a series of postsynaptic protein kinases and dysregulation of scaffold and adaptor proteins in response to neonatal HI insult. This work, along with other recent studies of synaptic protein profiling in the immature brain, builds a foundation for future investigation on the molecular mechanisms underlying developing plasticity. Furthermore, it provides insights into the biochemical changes of PSDs following early brain hypoxia-ischemia, which is helpful for understanding not only the injury mechanisms, but also the process of repair or replenishment of neuronal circuits during recovery from brain damage.
Collapse
Affiliation(s)
- Guo Shao
- Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Baotou, China
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Anguita E, Villalobo A. Src-family tyrosine kinases and the Ca 2+ signal. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1864:915-932. [PMID: 27818271 DOI: 10.1016/j.bbamcr.2016.10.022] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 10/25/2016] [Accepted: 10/30/2016] [Indexed: 01/08/2023]
Abstract
In this review, we shall describe the rich crosstalk between non-receptor Src-family kinases (SFKs) and the Ca2+ transient generated in activated cells by a variety of extracellular and intracellular stimuli, resulting in diverse signaling events. The exchange of information between SFKs and Ca2+ is reciprocal, as it flows in both directions. These kinases are main actors in pathways leading to the generation of the Ca2+ signal, and reciprocally, the Ca2+ signal modulates SFKs activity and functions. We will cover how SFKs participate in the generation of the cytosolic Ca2+ rise upon activation of a series of receptors and the mechanism of clearance of this Ca2+ signal. The role of SFKs modulating Ca2+-translocating channels participating in these events will be amply discussed. Finally, the role of the Ca2+ sensor protein calmodulin on the activity of c-Src, and potentially on other SFKs, will be outlined as well. This article is part of a Special Issue entitled: ECS Meeting edited by Claus Heizmann, Joachim Krebs and Jacques Haiech.
Collapse
Affiliation(s)
- Estefanía Anguita
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, c/ Arturo Duperier 4, E-28029 Madrid, Spain
| | - Antonio Villalobo
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, c/ Arturo Duperier 4, E-28029 Madrid, Spain.
| |
Collapse
|
28
|
Seo HY, Jeon JH, Jung YA, Jung GS, Lee EJ, Choi YK, Park KG, Choe MS, Jang BK, Kim MK, Lee IK. Fyn deficiency attenuates renal fibrosis by inhibition of phospho-STAT3. Kidney Int 2016; 90:1285-1297. [PMID: 27616741 DOI: 10.1016/j.kint.2016.06.038] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 06/24/2016] [Accepted: 06/30/2016] [Indexed: 01/30/2023]
Abstract
The hallmark of renal tubulointerstitial fibrosis is the accumulation of myofibroblasts and extracellular matrix proteins. Fyn, a member of the Src family of kinases, has diverse biological functions including regulation of mitogenic signaling and proliferation and integrin-mediated interaction. Src family proteins promote pulmonary fibrosis by augmenting transforming growth factor-β signaling, but their role in renal fibrosis is less understood. We observed upregulation of Fyn in a renal fibrosis model induced by unilateral ureteral obstruction. Upon ureteral obstruction, Fyn-deficient mice exhibited attenuated renal fibrosis relative to wild-type mice. Furthermore, obstruction-induced renal expression of type I collagen, fibronectin, α-smooth muscle actin, and plasminogen activator inhibitor-1 was suppressed. Pharmacologic inhibition of Fyn blocked induction of extracellular matrix proteins in kidney cell lines. Importantly, the attenuation of renal fibrosis by Fyn deficiency was not accompanied by changes in the Smad pathway. Rather, the antifibrotic effect of Fyn deficiency was associated with downregulation of signal transducer and activator of transcription 3 (STAT3). Small, interfering RNA targeting STAT3 in Fyn-deficient cells further suppressed α-smooth muscle actin expression, whereas a STAT3 activator partially restored plasminogen activator inhibitor-1 expression, indicating that STAT3 signaling is critically involved in this process. Thus, Fyn plays an important role in renal fibrosis. Hence, Fyn kinase inhibitors may be therapeutically useful against renal fibrosis.
Collapse
Affiliation(s)
- Hye-Young Seo
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu, South Korea; Institute for Medical Science, Keimyung University School of Medicine, Daegu, South Korea
| | - Jae-Han Jeon
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, South Korea
| | - Yun-A Jung
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu, South Korea; Institute for Medical Science, Keimyung University School of Medicine, Daegu, South Korea
| | - Gwon-Soo Jung
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, South Korea
| | - Eun Ju Lee
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu, South Korea; Institute for Medical Science, Keimyung University School of Medicine, Daegu, South Korea
| | - Young-Keun Choi
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, South Korea
| | - Keun-Gyu Park
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, South Korea
| | - Mi Sun Choe
- Department of Pathology, Keimyung University School of Medicine, Daegu, South Korea
| | - Byoung Kuk Jang
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu, South Korea; Institute for Medical Science, Keimyung University School of Medicine, Daegu, South Korea
| | - Mi-Kyung Kim
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu, South Korea; Institute for Medical Science, Keimyung University School of Medicine, Daegu, South Korea.
| | - In-Kyu Lee
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, South Korea.
| |
Collapse
|
29
|
Duan ZZ, Zhang F, Li FY, Luan YF, Guo P, Li YH, Liu Y, Qi SH. Protease activated receptor 1 (PAR1) enhances Src-mediated tyrosine phosphorylation of NMDA receptor in intracerebral hemorrhage (ICH). Sci Rep 2016; 6:29246. [PMID: 27385592 PMCID: PMC4935874 DOI: 10.1038/srep29246] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 06/14/2016] [Indexed: 12/20/2022] Open
Abstract
It has been demonstrated that Src could modulate NMDA receptor, and PAR1 could also affect NMDAR signaling. However, whether PAR1 could regulate NMDAR through Src under ICH has not yet been investigated. In this study, we demonstrated the role of Src-PSD95-GluN2A signaling cascades in rat ICH model and in vitro thrombin challenged model. Using the PAR1 agonist SFLLR, antagonist RLLFS and Src inhibitor PP2, electrophysiological analysis showed that PAR1 regulated NMDA-induced whole-cell currents (INMDA) though Src in primary cultured neurons. Both in vivo and in vitro results showed the elevated phosphorylation of tyrosine in Src and GluN2A and enhanced interaction of the Src-PSD95-GluN2A under model conditions. Treatment with the PAR1 antagonist RLLFS, AS-PSD95 (Antisense oligonucleotide against PSD95) and Src inhibitor PP2 inhibited the interaction among Src-PSD95-GluN2A, and p-Src, p-GluN2A. Co-application of SFLLR and AS-PSD95, PP2, or MK801 (NMDAR inhibitor) abolished the effect of SF. In conclusion, our results demonstrated that activated thrombin receptor PAR1 induced Src activation, enhanced the interaction among Src-PSD95-GluN2A signaling modules, and up-regulated GluN2A phosphorylation after ICH injury. Elucidation of such signaling cascades would possibly provide novel targets for ICH treatment.
Collapse
Affiliation(s)
- Zhen-Zhen Duan
- Research Center for Biochemistry and Molecular Biology and Provincial Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical College, Xuzhou, 221002, P. R. China
| | - Feng Zhang
- Research Center for Biochemistry and Molecular Biology and Provincial Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical College, Xuzhou, 221002, P. R. China
| | - Feng-Ying Li
- Research Center for Biochemistry and Molecular Biology and Provincial Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical College, Xuzhou, 221002, P. R. China
| | - Yi-Fei Luan
- Research Center for Biochemistry and Molecular Biology and Provincial Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical College, Xuzhou, 221002, P. R. China
| | - Peng Guo
- Research Center for Biochemistry and Molecular Biology and Provincial Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical College, Xuzhou, 221002, P. R. China
| | - Yi-Hang Li
- Research Center for Biochemistry and Molecular Biology and Provincial Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical College, Xuzhou, 221002, P. R. China
| | - Yong Liu
- Research Center for Biochemistry and Molecular Biology and Provincial Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical College, Xuzhou, 221002, P. R. China
| | - Su-Hua Qi
- Research Center for Biochemistry and Molecular Biology and Provincial Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical College, Xuzhou, 221002, P. R. China
| |
Collapse
|
30
|
Curcio M, Salazar IL, Mele M, Canzoniero LMT, Duarte CB. Calpains and neuronal damage in the ischemic brain: The swiss knife in synaptic injury. Prog Neurobiol 2016; 143:1-35. [PMID: 27283248 DOI: 10.1016/j.pneurobio.2016.06.001] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 03/22/2016] [Accepted: 05/09/2016] [Indexed: 12/26/2022]
Abstract
The excessive extracellular accumulation of glutamate in the ischemic brain leads to an overactivation of glutamate receptors with consequent excitotoxic neuronal death. Neuronal demise is largely due to a sustained activation of NMDA receptors for glutamate, with a consequent increase in the intracellular Ca(2+) concentration and activation of calcium- dependent mechanisms. Calpains are a group of Ca(2+)-dependent proteases that truncate specific proteins, and some of the cleavage products remain in the cell, although with a distinct function. Numerous studies have shown pre- and post-synaptic effects of calpains on glutamatergic and GABAergic synapses, targeting membrane- associated proteins as well as intracellular proteins. The resulting changes in the presynaptic proteome alter neurotransmitter release, while the cleavage of postsynaptic proteins affects directly or indirectly the activity of neurotransmitter receptors and downstream mechanisms. These alterations also disturb the balance between excitatory and inhibitory neurotransmission in the brain, with an impact in neuronal demise. In this review we discuss the evidence pointing to a role for calpains in the dysregulation of excitatory and inhibitory synapses in brain ischemia, at the pre- and post-synaptic levels, as well as the functional consequences. Although targeting calpain-dependent mechanisms may constitute a good therapeutic approach for stroke, specific strategies should be developed to avoid non-specific effects given the important regulatory role played by these proteases under normal physiological conditions.
Collapse
Affiliation(s)
- Michele Curcio
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Ivan L Salazar
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; Doctoral Programme in Experimental Biology and Biomedicine, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; Institute for Interdisciplinary Research, University of Coimbra (IIIUC), 3030-789 Coimbra, Portugal
| | - Miranda Mele
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | | | - Carlos B Duarte
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; Department of Life Sciences, University of Coimbra, 3000-456 Coimbra, Portugal.
| |
Collapse
|
31
|
Zamzow DR, Elias V, Acosta VA, Escobedo E, Magnusson KR. Higher levels of phosphorylated Y1472 on GluN2B subunits in the frontal cortex of aged mice are associated with good spatial reference memory, but not cognitive flexibility. AGE (DORDRECHT, NETHERLANDS) 2016; 38:50. [PMID: 27094400 PMCID: PMC5005925 DOI: 10.1007/s11357-016-9913-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Accepted: 04/06/2016] [Indexed: 06/05/2023]
Abstract
The N-methyl-D-aspartate receptor (NMDAr) is particularly vulnerable to aging. The GluN2B subunit of the NMDAr, compared to other NMDAr subunits, suffers the greatest losses of expression in the aging brain, especially in the frontal cortex. While expression levels of GluN2B mRNA and protein in the aged brain are well documented, there has been little investigation into age-related posttranslational modifications of the subunit. In this study, we explored some of the mechanisms that may promote differences in the NMDAr complex in the frontal cortex of aged animals. Two ages of mice, 3 and 24 months, were behaviorally tested in the Morris water maze. The frontal cortex and hippocampus from each mouse were subjected to differential centrifugation followed by solubilization in Triton X-100. Proteins from Triton-insoluble membranes, Triton-soluble membranes, and intracellular membranes/cytosol were examined by Western blot. Higher levels of GluN2B tyrosine 1472 phosphorylation in frontal cortex synaptic fractions of old mice were associated with better reference learning but poorer cognitive flexibility. Levels of GluN2B phosphotyrosine 1336 remained steady, but there were greater levels of the calpain-induced 115 kDa GluN2B cleavage product on extrasynaptic membranes in these old good learners. There was an age-related increase in calpain activity, but it was not associated with better learning. These data highlight a unique aging change for aged mice with good spatial learning that might be detrimental to cognitive flexibility. This study also suggests that higher levels of truncated GluN2B on extrasynaptic membranes are not deleterious to spatial memory in aged mice.
Collapse
Affiliation(s)
| | - Val Elias
- Oregon State University, Corvallis, OR, USA
| | | | | | | |
Collapse
|
32
|
A comprehensive assessment of networks and pathways of hypoxia-associated proteins and identification of responsive protein modules. ACTA ACUST UNITED AC 2016. [DOI: 10.1007/s13721-016-0123-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
33
|
Li Y, Sun W, Han S, Li J, Ding S, Wang W, Yin Y. IGF-1-Involved Negative Feedback of NR2B NMDA Subunits Protects Cultured Hippocampal Neurons Against NMDA-Induced Excitotoxicity. Mol Neurobiol 2016; 54:684-696. [DOI: 10.1007/s12035-015-9647-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Accepted: 12/16/2015] [Indexed: 11/28/2022]
|
34
|
Min JW, Hu JJ, He M, Sanchez RM, Huang WX, Liu YQ, Bsoul NB, Han S, Yin J, Liu WH, He XH, Peng BW. Vitexin reduces hypoxia-ischemia neonatal brain injury by the inhibition of HIF-1alpha in a rat pup model. Neuropharmacology 2015; 99:38-50. [PMID: 26187393 DOI: 10.1016/j.neuropharm.2015.07.007] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 06/16/2015] [Accepted: 07/07/2015] [Indexed: 01/25/2023]
Abstract
Previous studies have demonstrated that the early suppression of HIF-1α after hypoxia-ischemia (HI) injury provides neuroprotection. Vitexin (5, 7, 4-trihydroxyflavone-8-glucoside), an HIF-1α inhibitor, is a c-glycosylated flavone that has been identified in medicinal plants. Therefore, we hypothesized that treatment with vitexin would protect against HI brain injury. Newborn rat pups were subjected to unilateral carotid artery ligation followed by 2.5 h of hypoxia (8% O2 at 37 °C). Vitexin (30, 45 or 60 mg/kg) was administered intraperitoneally at 5 min or 3 h after HI. Vitexin, administered 5 min after HI, was neuroprotective as seen by decreased infarct volume evaluated at 48 h post-HI. This neuroprotection was removed when vitexin was administered 3 h after HI. Neuronal cell death, blood-brain barrier (BBB) integrity, brain edema, HIF-1α and VEGF protein levels were evaluated using a combination of Nissl staining, IgG staining, brain water content, immunohistochemistry and Western blot at 24 and 48 h after HI. The long-term effects of vitexin were evaluated by brain atrophy measurement, Nissl staining and neurobehavioral tests. Vitexin (45 mg/kg) ameliorated brain edema, BBB disruption and neuronal cell death; Upregulation of HIF-1α by dimethyloxalylglycine (DMOG) increased the BBB permeability and brain edema compared to HI alone. Vitexin attenuated the increase in HIF-1α and VEGF. Vitexin also had long-term effects of protecting against the loss of ipsilateral brain and improveing neurobehavioral outcomes. In conclusion, our data indicate early HIF-1α inhibition with vitexin provides both acute and long-term neuroprotection in the developing brain after neonatal HI injury.
Collapse
Affiliation(s)
- Jia-Wei Min
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Jiang-Jian Hu
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Miao He
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Russell M Sanchez
- Department of Surgery, College of Medicine, Texas A&M Health Science Center, Neuroscience Institute, Scott & White Hospital, & Central Texas Veterans Health Care System, Temple, TX, USA
| | - Wen-Xian Huang
- Department of Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Yu-Qiang Liu
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Najeeb Bassam Bsoul
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Song Han
- Department of Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Jun Yin
- Department of Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Wan-Hong Liu
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Xiao-Hua He
- Department of Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China.
| | - Bi-Wen Peng
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China.
| |
Collapse
|
35
|
Titomanlio L, Fernández-López D, Manganozzi L, Moretti R, Vexler ZS, Gressens P. Pathophysiology and neuroprotection of global and focal perinatal brain injury: lessons from animal models. Pediatr Neurol 2015; 52:566-584. [PMID: 26002050 PMCID: PMC4720385 DOI: 10.1016/j.pediatrneurol.2015.01.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 01/16/2015] [Accepted: 01/24/2015] [Indexed: 12/14/2022]
Abstract
BACKGROUND Arterial ischemic stroke occurs more frequently in term newborns than in the elderly, and brain immaturity affects mechanisms of ischemic injury and recovery. The susceptibility to injury of the brain was assumed to be lower in the perinatal period as compared with childhood. This concept was recently challenged by clinical studies showing marked motor disabilities after stroke in neonates, with the severity of motor and cortical sensory deficits similar in both perinatal and childhood ischemic stroke. Our understanding of the triggers and the pathophysiological mechanisms of perinatal stroke has greatly improved in recent years, but many factors remain incompletely understood. METHODS In this review, we focus on the pathophysiology of perinatal stroke and on therapeutic strategies that can protect the immature brain from the consequences of stroke by targeting inflammation and brain microenvironment. RESULTS Studies in neonatal rodent models of cerebral ischemia have suggested a potential role for soluble inflammatory molecules as important modulators of injury and recovery. A great effort is underway to investigate neuroprotective molecules based on our increasing understanding of the pathophysiology. CONCLUSION In this review, we provide a comprehensive summary of new insights concerning pathophysiology of focal and global perinatal brain injury and their implications for new therapeutic approaches.
Collapse
Affiliation(s)
- Luigi Titomanlio
- Pediatric Emergency Department, APHP, Robert Debré Hospital, Paris, France
- Inserm, U1141, F-75019 Paris, France
| | - David Fernández-López
- Department of Neurology, University of California San Francisco, San Francisco, CA, 94158-0663, USA
| | - Lucilla Manganozzi
- Pediatric Emergency Department, APHP, Robert Debré Hospital, Paris, France
- Inserm, U1141, F-75019 Paris, France
| | | | - Zinaida S. Vexler
- Department of Neurology, University of California San Francisco, San Francisco, CA, 94158-0663, USA
| | - Pierre Gressens
- Inserm, U1141, F-75019 Paris, France
- Univ Paris Diderot, Sorbonne Paris Cité, UMRS 676, F-75019 Paris, France
- PremUP, Paris, France
- Centre for the Developing Brain, King’s College, St Thomas’ Campus, London SE1 7EH, UK
| |
Collapse
|
36
|
Mirza MA, Ritzel R, Xu Y, McCullough LD, Liu F. Sexually dimorphic outcomes and inflammatory responses in hypoxic-ischemic encephalopathy. J Neuroinflammation 2015; 12:32. [PMID: 25889641 PMCID: PMC4359482 DOI: 10.1186/s12974-015-0251-6] [Citation(s) in RCA: 123] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 01/16/2015] [Indexed: 12/11/2022] Open
Abstract
Background Neonatal hypoxic-ischemic encephalopathy (HIE) is an important cause of motor and cognitive impairment in children. Clinically, male infants are more vulnerable to ischemic insults and suffer more long-term deficits than females; however, the mechanisms underlying this sex difference remain elusive. Inflammatory processes initiated by microglial activation are fundamental in the pathophysiology of ischemia. Recent studies report a sexual dimorphism in microglia numbers and expression of activation markers in the neonatal brain under normal conditions. How these basal sex differences in microglia affect HIE remains largely unexplored. This study investigated sex differences in ischemic outcomes and inflammation triggered by HIE. We hypothesize that ischemia induces sex-specific brain injury in male and female neonates and that microglial activation and inflammatory responses play an important role in this sexual dimorphism. Methods Male and female C57BL6 mice were subjected to 60-min Rice-Vanucci modeling (RVM) at post-natal day 10 (P10) to induce HIE. Stroke outcomes were measured 1, 3, 7, and 30 days after stroke. Microglial activation and inflammatory responses were evaluated by flow cytometry and cytokine analysis. Results On day 1 of HIE, no difference in infarct volumes or seizure scores was seen between male and female neonates. However, female neonates exhibited significantly smaller infarct size and fewer seizures compared to males 3 days after HIE. Females also had less brain tissue loss and behavioral deficits compared to males at the chronic stage of HIE. Male animals demonstrated increased microglial activation and up-regulated inflammatory response compared to females at day 3. Conclusions HIE leads to an equivalent primary brain injury in male and female neonates at the acute stage that develops into sexually dimorphic outcomes at later time points. An innate immune response secondary to the primary injury may contribute to sexual dimorphism in HIE.
Collapse
Affiliation(s)
- Mehwish A Mirza
- Department of Neuroscience, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030, USA.
| | - Rodney Ritzel
- Department of Neuroscience, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030, USA.
| | - Yan Xu
- Department of Neuroscience, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030, USA.
| | - Louise D McCullough
- Department of Neuroscience, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030, USA. .,Department of Neurology, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030, USA.
| | - Fudong Liu
- Department of Neuroscience, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030, USA.
| |
Collapse
|
37
|
Knox R, Jiang X. Fyn in Neurodevelopment and Ischemic Brain Injury. Dev Neurosci 2015; 37:311-20. [PMID: 25720756 DOI: 10.1159/000369995] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 11/18/2014] [Indexed: 12/11/2022] Open
Abstract
The Src family kinases (SFKs) are nonreceptor protein tyrosine kinases that are implicated in many normal and pathological processes in the nervous system. The SFKs Fyn, Src, Yes, Lyn, and Lck are expressed in the brain. This review will focus on Fyn, as Fyn mutant mice have striking phenotypes in the brain and Fyn has been shown to be involved in ischemic brain injury in adult rodents and, with our work, in neonatal animals. An understanding of Fyn's role in neurodevelopment and disease will allow researchers to target pathological pathways while preserving protective ones.
Collapse
Affiliation(s)
- Renatta Knox
- Department of Pediatrics, Weill Cornell Medical College, New York, N.Y., USA
| | | |
Collapse
|
38
|
López-Pérez SJ, Morales-Villagrán A, Medina-Ceja L. Effect of perinatal asphyxia and carbamazepine treatment on cortical dopamine and DOPAC levels. J Biomed Sci 2015; 22:14. [PMID: 25889791 PMCID: PMC4335632 DOI: 10.1186/s12929-015-0117-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 01/23/2015] [Indexed: 01/29/2023] Open
Abstract
Background One of the most important manifestations of perinatal asphyxia is the occurrence of seizures, which are treated with antiepileptic drugs, such as carbamazepine. These early seizures, combined with pharmacological treatments, may influence the development of dopaminergic neurotransmission in the frontal cortex. This study aimed to determine the extracellular levels of dopamine and its main metabolite DOPAC in 30-day-old rats that had been asphyxiated for 45 min in a low (8%) oxygen chamber at a perinatal age and treated with daily doses of carbamazepine. Quantifications were performed using microdialysis coupled to a high-performance liquid chromatography (HPLC) system in basal conditions and following the use of the chemical stimulus. Results Significant decreases in basal and stimulated extracellular dopamine and DOPAC content were observed in the frontal cortex of the asphyxiated group, and these decreases were partially recovered in the animals administered daily doses of carbamazepine. Greater basal dopamine concentrations were also observed as an independent effect of carbamazepine. Conclusions Perinatal asphyxia plus carbamazepine affects extracellular levels of dopamine and DOPAC in the frontal cortex and stimulated the release of dopamine, which provides evidence for the altered availability of dopamine in cortical brain areas during brain development.
Collapse
Affiliation(s)
- Silvia J López-Pérez
- Laboratorio de Neurofisiología y Neuroquímica, Departamento de Biología Celular y Molecular, CUCBA, Universidad de Guadalajara, Camino Ing. Ramón Padilla Sánchez #2100, Predio Las Agujas, Zapopan, Jalisco, C.P 44600, Mexico.
| | - Alberto Morales-Villagrán
- Laboratorio de Neurofisiología y Neuroquímica, Departamento de Biología Celular y Molecular, CUCBA, Universidad de Guadalajara, Camino Ing. Ramón Padilla Sánchez #2100, Predio Las Agujas, Zapopan, Jalisco, C.P 44600, Mexico.
| | - Laura Medina-Ceja
- Laboratorio de Neurofisiología y Neuroquímica, Departamento de Biología Celular y Molecular, CUCBA, Universidad de Guadalajara, Camino Ing. Ramón Padilla Sánchez #2100, Predio Las Agujas, Zapopan, Jalisco, C.P 44600, Mexico.
| |
Collapse
|
39
|
Zhu QJ, Kong FS, Xu H, Wang Y, Du CP, Sun CC, Liu Y, Li T, Hou XY. Tyrosine phosphorylation of GluK2 up-regulates kainate receptor-mediated responses and downstream signaling after brain ischemia. Proc Natl Acad Sci U S A 2014; 111:13990-5. [PMID: 25201974 PMCID: PMC4183319 DOI: 10.1073/pnas.1403493111] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Although kainate receptors play important roles in ischemic stroke, the molecular mechanisms underlying postischemic regulation of kainate receptors remain unclear. In this study we demonstrate that Src family kinases contribute to the potentiation of kainate receptor function. Brain ischemia and reperfusion induce rapid and sustained phosphorylation of the kainate receptor subunit GluK2 by Src in the rat hippocampus, implicating a critical role for Src-mediated GluK2 phosphorylation in ischemic brain injury. The NMDA and kainate receptors are involved in the tyrosine phosphorylation of GluK2. GluK2 binds to Src, and the tyrosine residue at position 590 (Y590) on GluK2 is a major site of phosphorylation by Src kinases. GluK2 phosphorylation at Y590 is responsible for increases in whole-cell currents and calcium influx in response to transient kainate stimulation. In addition, GluK2 phosphorylation at Y590 facilitates the endocytosis of GluK2 subunits, and the activation of JNK3 and its substrate c-Jun after long-term kainate treatment. Thus, Src phosphorylation of GluK2 plays an important role in the opening of kainate receptor channels and downstream proapoptosis signaling after brain ischemia. The present study reveals an additional mechanism for the regulation of GluK2-containing kainate receptors by Src family kinases, which may be of pathological significance in ischemic stroke.
Collapse
Affiliation(s)
- Qiu-Ju Zhu
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical College, Jiangsu 221004, China
| | - Fan-Shu Kong
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical College, Jiangsu 221004, China
| | - Hao Xu
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical College, Jiangsu 221004, China
| | - Yi Wang
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical College, Jiangsu 221004, China
| | - Cai-Ping Du
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical College, Jiangsu 221004, China
| | - Chang-Cheng Sun
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical College, Jiangsu 221004, China
| | - Yong Liu
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical College, Jiangsu 221004, China
| | - Ting Li
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical College, Jiangsu 221004, China
| | - Xiao-Yu Hou
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical College, Jiangsu 221004, China
| |
Collapse
|
40
|
Knox R, Brennan-Minnella AM, Lu F, Yang D, Nakazawa T, Yamamoto T, Swanson RA, Ferriero DM, Jiang X. NR2B phosphorylation at tyrosine 1472 contributes to brain injury in a rodent model of neonatal hypoxia-ischemia. Stroke 2014; 45:3040-7. [PMID: 25158771 DOI: 10.1161/strokeaha.114.006170] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND AND PURPOSE The NR2B subunit of the N-methyl-d-aspartate (NMDA) receptor is phosphorylated by the Src family kinase Fyn in brain, with tyrosine (Y) 1472 as the major phosphorylation site. Although Y1472 phosphorylation is important for synaptic plasticity, it is unknown whether it is involved in NMDA receptor-mediated excitotoxicity in neonatal brain hypoxia-ischemia (HI). This study was designed to elucidate the specific role of Y1472 phosphorylation of NR2B in neonatal HI in vivo and in NMDA-mediated neuronal death in vitro. METHODS Neonatal mice with a knockin mutation of Y1472 to phenylalanine (YF-KI) and their wild-type littermates were subjected to HI using the Vannucci model. Brains were scored 5 days later for damage using cresyl violet and iron staining. Western blotting and immunoprecipitation were performed to determine NR2B tyrosine phosphorylation. Expression of NADPH oxidase subunits and superoxide production were measured in vivo. NMDA-induced calcium response, superoxide formation, and cell death were evaluated in primary cortical neurons. RESULTS After neonatal HI, YF-KI mice have reduced expression of NADPH oxidase subunit gp91phox and p47phox and superoxide production, lower activity of proteases implicated in necrotic and apoptotic cell death, and less brain damage when compared with the wild-type mice. In vitro, YF-KI mutation diminishes superoxide generation in response to NMDA without effect on calcium accumulation and inhibits NMDA and glutamate-induced cell death. CONCLUSIONS Upregulation of NR2B phosphorylation at Y1472 after neonatal HI is involved in superoxide-mediated oxidative stress and contributes to brain injury.
Collapse
Affiliation(s)
- Renatta Knox
- From the Department of Pediatrics (R.K., F.L., D.Y., D.M.F., X.J.), Biomedical Sciences Graduate Program (R.K., D.M.F.), Medical Scientist Training Program (R.K.), Department of Neurology (A.M.B.-M., R.A.S., D.M.F.), and San Francisco Veterans Affairs Medical Center (A.M.B.-M., R.A.S.), University of California, San Francisco; and Division of Oncology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan (T.N., T.Y.)
| | - Angela M Brennan-Minnella
- From the Department of Pediatrics (R.K., F.L., D.Y., D.M.F., X.J.), Biomedical Sciences Graduate Program (R.K., D.M.F.), Medical Scientist Training Program (R.K.), Department of Neurology (A.M.B.-M., R.A.S., D.M.F.), and San Francisco Veterans Affairs Medical Center (A.M.B.-M., R.A.S.), University of California, San Francisco; and Division of Oncology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan (T.N., T.Y.)
| | - Fuxin Lu
- From the Department of Pediatrics (R.K., F.L., D.Y., D.M.F., X.J.), Biomedical Sciences Graduate Program (R.K., D.M.F.), Medical Scientist Training Program (R.K.), Department of Neurology (A.M.B.-M., R.A.S., D.M.F.), and San Francisco Veterans Affairs Medical Center (A.M.B.-M., R.A.S.), University of California, San Francisco; and Division of Oncology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan (T.N., T.Y.)
| | - Diana Yang
- From the Department of Pediatrics (R.K., F.L., D.Y., D.M.F., X.J.), Biomedical Sciences Graduate Program (R.K., D.M.F.), Medical Scientist Training Program (R.K.), Department of Neurology (A.M.B.-M., R.A.S., D.M.F.), and San Francisco Veterans Affairs Medical Center (A.M.B.-M., R.A.S.), University of California, San Francisco; and Division of Oncology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan (T.N., T.Y.)
| | - Takanobu Nakazawa
- From the Department of Pediatrics (R.K., F.L., D.Y., D.M.F., X.J.), Biomedical Sciences Graduate Program (R.K., D.M.F.), Medical Scientist Training Program (R.K.), Department of Neurology (A.M.B.-M., R.A.S., D.M.F.), and San Francisco Veterans Affairs Medical Center (A.M.B.-M., R.A.S.), University of California, San Francisco; and Division of Oncology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan (T.N., T.Y.)
| | - Tadashi Yamamoto
- From the Department of Pediatrics (R.K., F.L., D.Y., D.M.F., X.J.), Biomedical Sciences Graduate Program (R.K., D.M.F.), Medical Scientist Training Program (R.K.), Department of Neurology (A.M.B.-M., R.A.S., D.M.F.), and San Francisco Veterans Affairs Medical Center (A.M.B.-M., R.A.S.), University of California, San Francisco; and Division of Oncology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan (T.N., T.Y.)
| | - Raymond A Swanson
- From the Department of Pediatrics (R.K., F.L., D.Y., D.M.F., X.J.), Biomedical Sciences Graduate Program (R.K., D.M.F.), Medical Scientist Training Program (R.K.), Department of Neurology (A.M.B.-M., R.A.S., D.M.F.), and San Francisco Veterans Affairs Medical Center (A.M.B.-M., R.A.S.), University of California, San Francisco; and Division of Oncology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan (T.N., T.Y.)
| | - Donna M Ferriero
- From the Department of Pediatrics (R.K., F.L., D.Y., D.M.F., X.J.), Biomedical Sciences Graduate Program (R.K., D.M.F.), Medical Scientist Training Program (R.K.), Department of Neurology (A.M.B.-M., R.A.S., D.M.F.), and San Francisco Veterans Affairs Medical Center (A.M.B.-M., R.A.S.), University of California, San Francisco; and Division of Oncology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan (T.N., T.Y.)
| | - Xiangning Jiang
- From the Department of Pediatrics (R.K., F.L., D.Y., D.M.F., X.J.), Biomedical Sciences Graduate Program (R.K., D.M.F.), Medical Scientist Training Program (R.K.), Department of Neurology (A.M.B.-M., R.A.S., D.M.F.), and San Francisco Veterans Affairs Medical Center (A.M.B.-M., R.A.S.), University of California, San Francisco; and Division of Oncology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan (T.N., T.Y.).
| |
Collapse
|
41
|
Ko HM, Lee SH, Kim KC, Joo SH, Choi WS, Shin CY. The Role of TLR4 and Fyn Interaction on Lipopolysaccharide-Stimulated PAI-1 Expression in Astrocytes. Mol Neurobiol 2014; 52:8-25. [DOI: 10.1007/s12035-014-8837-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 07/28/2014] [Indexed: 01/05/2023]
|
42
|
The Role of Src Kinase in the Caspase-1 Pathway After Hypoxia in the Brain of Newborn Piglets. Neurochem Res 2014; 39:2118-26. [DOI: 10.1007/s11064-014-1404-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 06/28/2014] [Accepted: 07/28/2014] [Indexed: 01/06/2023]
|
43
|
Mechanisms of perinatal arterial ischemic stroke. J Cereb Blood Flow Metab 2014; 34:921-32. [PMID: 24667913 PMCID: PMC4050239 DOI: 10.1038/jcbfm.2014.41] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 12/30/2013] [Accepted: 01/02/2014] [Indexed: 01/21/2023]
Abstract
The incidence of perinatal stroke is high, similar to that in the elderly, and produces a significant morbidity and severe long-term neurologic and cognitive deficits, including cerebral palsy, epilepsy, neuropsychological impairments, and behavioral disorders. Emerging clinical data and data from experimental models of cerebral ischemia in neonatal rodents have shown that the pathophysiology of perinatal brain damage is multifactorial. These studies have revealed that, far from just being a smaller version of the adult brain, the neonatal brain is unique with a very particular and age-dependent responsiveness to hypoxia-ischemia and focal arterial stroke. In this review, we discuss fundamental clinical aspects of perinatal stroke as well as some of the most recent and relevant findings regarding the susceptibility of specific brain cell populations to injury, the dynamics and the mechanisms of neuronal cell death in injured neonates, the responses of neonatal blood-brain barrier to stroke in relation to systemic and local inflammation, and the long-term effects of stroke on angiogenesis and neurogenesis. Finally, we address translational strategies currently being considered for neonatal stroke as well as treatments that might effectively enhance repair later after injury.
Collapse
|
44
|
Mirza MA, Capozzi LA, Xu Y, McCullough LD, Liu F. Knockout of vascular early response gene worsens chronic stroke outcomes in neonatal mice. Brain Res Bull 2013; 98:111-21. [PMID: 23973431 DOI: 10.1016/j.brainresbull.2013.07.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 07/11/2013] [Accepted: 07/24/2013] [Indexed: 01/19/2023]
Abstract
Vascular early response gene (Verge) is a novel immediate early gene that is highly expressed during developmental angiogenesis and after ischemic insults in adult brain. However, the role of Verge after neonatal injury is not known. In the present study, we investigated the hypothesis that Verge contributes to vascular remodeling and tissue repair after neonatal ischemic injury. The Rice-Vanucci model (RVM) was employed to induce neonatal stroke in both Verge knockout (KO) and wild-type (WT) postnatal day 10 (P10) mice. Histological and behavioral outcomes at acute (24h), subacute (7 days) and chronic (30 days) phases were evaluated. Angiogenesis, neurogenesis, and glial scar formation were also examined in the ischemic brain. No significant differences in outcomes were found between WT and Verge mice at 24h or 7 days after stroke. However genetic deletion of Verge led to pronounced cystic cavitation, decreased angiogenensis and glial scar formation in the ischemic hemisphere compared to WT mice at 30 days. Verge KO mice also had significantly worse functional outcomes at 30 days which was accompanied by decreased neurogenesis and angiogenesis in the ischemic hemisphere. Our study suggests that Verge plays an important role in the induction of neurogenesis and angiogenesis after ischemia, contributes to improved tissue repair, and enhances chronic functional recovery.
Collapse
Affiliation(s)
- Mehwish A Mirza
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT 06030, United States
| | | | | | | | | |
Collapse
|