1
|
Zhong J, Yu X, Lin Z. Phosphodiesterase 4 inhibition as a novel treatment for stroke. PeerJ 2025; 13:e18905. [PMID: 39897494 PMCID: PMC11786714 DOI: 10.7717/peerj.18905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 01/06/2025] [Indexed: 02/04/2025] Open
Abstract
The incidence of stroke ranks third among the leading causes of mortality worldwide. It has the characteristics of high morbidity, high disability rate and high recurrence rate. The current risk associated with stroke surgery is exceedingly high. It may potentially outweigh the benefits and fail to ameliorate the cerebral tissue damage following ischemia. Therefore, pharmacological intervention assumes paramount importance. The use of thrombolytic drugs is most common in the treatment of stroke; however, its efficacy is limited due to its time-sensitive nature and propensity for increased bleeding. Over the past few years, the treatment of stroke has witnessed a surge in interest towards neuroprotective drugs that possess the potential to enhance neurological function. The PDE4D gene has been demonstrated to have a positive correlation with the risk of ischemic stroke. Additionally, the utilization of phosphodiesterase 4 inhibitors can enhance synaptic plasticity within the neural circuitry, regulate cellular metabolism, and prevent secondary brain injury caused by impaired blood flow. These mechanisms collectively facilitate the recovery of functional neurons, thereby serving as potential therapeutic interventions. Therefore, the comprehensive investigation of phosphodiesterase 4 as an innovative pharmacological target for stroke injury provides valuable insights into the development of therapeutic interventions in stroke treatment. This review is intended for, but not limited to, pharmacological researchers, drug target researchers, neurologists, neuromedical researchers, and behavioral scientists.
Collapse
Affiliation(s)
- Jiahong Zhong
- Department of Clinical Pharmacy, Meizhou People’s Hospital, Meizhou, Guangdong, China
| | - Xihui Yu
- Department of Pharmacy, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Zhuomiao Lin
- Department of Clinical Pharmacy, Meizhou People’s Hospital, Meizhou, Guangdong, China
| |
Collapse
|
2
|
Han Z, Song Y, Qin C, Zhou H, Han D, Yan S, Ni H. S-Nitrosylation of Dexras1 Controls Post-Stroke Recovery via Regulation of Neuronal Excitability and Dendritic Remodeling. CNS Neurosci Ther 2025; 31:e70199. [PMID: 39749632 PMCID: PMC11696243 DOI: 10.1111/cns.70199] [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: 10/09/2024] [Revised: 11/29/2024] [Accepted: 12/15/2024] [Indexed: 01/04/2025] Open
Abstract
AIMS Stroke is a major public health concern leading to high rates of death and disability worldwide, unfortunately with no effective treatment available for stroke recovery during the repair phase. METHODS Photothrombotic stroke was induced in mice. Adeno-associated viruses (AAV) were microinjected into the peri-infarct cortex immediately after photothrombotic stroke. Grid-walking task and cylinder task were used to assess motor function. Western blotting, Golgi staining, and electrophysiology recordings were performed to uncover the mechanisms. RESULTS The ternary complex of neuronal nitric oxide synthase (nNOS), carboxy-terminal PDZ ligand of nNOS (CAPON) and dexamethasone-induced ras protein 1 (Dexras1) is structurally beneficial for S-nitrosylation of Dexras1 (SNO-Dexras1). In our previous study, uncoupling nNOS-CAPON interaction by Tat-CAPON-12C promoted functional recovery after stroke. Here, we show that ischemia elevated the levels of nNOS-Dexras1 complex and SNO-Dexras1 in the peri-infarct cortex in the days 4-10 after stroke induction, and as excepted, Tat-CAPON-12C, a peptide disrupting nNOS-CAPON interaction, significantly reversed these changes. The above information implies that repressed SNO-Dexras1 may mediate functional-promoting effects of Tat-CAPON-12C and SNO-Dexras1 could be the vital molecular substrate for post-stroke functional recovery in the repair phage. Inhibiting the ischemia-induced SNO-Dexras1 by AAV vector-mediated knockdown of Dexras1 or over-expression of dominant negative Dexras1 (Dexras1-C11S) produced sustained recovery of motor function from stroke. In contrast, up-regulation of SNO-Dexras1 by over-expressing Dexras1 worsened stroke outcome. Using electrophysiology recordings, we also observed that silence of Dexras1 in the peri-infarct cortex increased the spike number and the miniature excitatory postsynaptic currents (mEPSCs) frequency, suggesting enhancement of neuronal excitability. In addition, silence of Dexras1 increased dendritic complexity in cultured neuron and more importantly enhanced dendritic spine density in the peri-infarct cortex, implying dendritic remodeling. CONCLUSION Thus, inhibition of SNO-Dexras1 positively regulates post-stroke functional recovery via enhanced neuronal excitability and dendritic remodeling. Our results identify that SNO-Dexras1 may serve as a novel target for promoting motor functional restoration from stroke in the delayed phase, shedding light on stroke treatment.
Collapse
Affiliation(s)
- Zhou Han
- Department of Pharmacy, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingJiangsuChina
- Nanjing Medical Center for Clinical PharmacyNanjingJiangsuChina
| | - Yixuan Song
- Department of Histoembryology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive MedicineShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Cheng Qin
- School of Life Sciences and Chemical EngineeringJiangsu Second Normal UniversityNanjingChina
| | - Haihui Zhou
- Department of Pharmacy, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingJiangsuChina
- Nanjing Medical Center for Clinical PharmacyNanjingJiangsuChina
| | - Dan Han
- Department of Pharmacy, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingJiangsuChina
- Nanjing Medical Center for Clinical PharmacyNanjingJiangsuChina
| | - Simin Yan
- Department of Pharmacy, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingJiangsuChina
- Nanjing Medical Center for Clinical PharmacyNanjingJiangsuChina
| | - Huanyu Ni
- Department of Pharmacy, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingJiangsuChina
- Nanjing Medical Center for Clinical PharmacyNanjingJiangsuChina
| |
Collapse
|
3
|
Gui Y, Kim Y, Brenna S, Wilmes M, Zaghen G, Goulbourne CN, Kuchenbecker-Pöls L, Siebels B, Voß H, Gocke A, Schlüter H, Schweizer M, Altmeppen HC, Magnus T, Levy E, Puig B. Cystatin C loaded in brain-derived extracellular vesicles rescues synapses after ischemic insult in vitro and in vivo. Cell Mol Life Sci 2024; 81:224. [PMID: 38769196 PMCID: PMC11106054 DOI: 10.1007/s00018-024-05266-4] [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: 02/12/2024] [Revised: 04/12/2024] [Accepted: 05/05/2024] [Indexed: 05/22/2024]
Abstract
Synaptic loss is an early event in the penumbra area after an ischemic stroke. Promoting synaptic preservation in this area would likely improve functional neurological recovery. We aimed to detect proteins involved in endogenous protection mechanisms of synapses in the penumbra after stroke and to analyse potential beneficial effects of these candidates for a prospective stroke treatment. For this, we performed Liquid Chromatography coupled to Mass Spectrometry (LC-MS)-based proteomics of synaptosomes isolated from the ipsilateral hemispheres of mice subjected to experimental stroke at different time points (24 h, 4 and 7 days) and compared them to sham-operated mice. Proteomic analyses indicated that, among the differentially expressed proteins between the two groups, cystatin C (CysC) was significantly increased at 24 h and 4 days following stroke, before returning to steady-state levels at 7 days, thus indicating a potential transient and intrinsic rescue mechanism attempt of neurons. When CysC was applied to primary neuronal cultures subjected to an in vitro model of ischemic damage, this treatment significantly improved the preservation of synaptic structures. Notably, similar effects were observed when CysC was loaded into brain-derived extracellular vesicles (BDEVs). Finally, when CysC contained in BDEVs was administered intracerebroventricularly to stroked mice, it significantly increased the expression of synaptic markers such as SNAP25, Homer-1, and NCAM in the penumbra area compared to the group supplied with empty BDEVs. Thus, we show that CysC-loaded BDEVs promote synaptic protection after ischemic damage in vitro and in vivo, opening the possibility of a therapeutic use in stroke patients.
Collapse
Affiliation(s)
- Yuqi Gui
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI) Group, University Medical Center Hamburg-Eppendorf (UKE), Martinistraße, 52, 20246, Hamburg, Germany
- Department of Intensive Care Unit, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yohan Kim
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY, 10962, USA
- Departments of Psychiatry, Biochemistry and Molecular Pharmacology, and the Neuroscience Institute, NYU Langone Medical Center, New York, NY, 10016, USA
| | - Santra Brenna
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI) Group, University Medical Center Hamburg-Eppendorf (UKE), Martinistraße, 52, 20246, Hamburg, Germany
| | - Maximilian Wilmes
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI) Group, University Medical Center Hamburg-Eppendorf (UKE), Martinistraße, 52, 20246, Hamburg, Germany
| | - Giorgio Zaghen
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY, 10962, USA
- Departments of Psychiatry, Biochemistry and Molecular Pharmacology, and the Neuroscience Institute, NYU Langone Medical Center, New York, NY, 10016, USA
| | - Chris N Goulbourne
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY, 10962, USA
| | - Lennart Kuchenbecker-Pöls
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI) Group, University Medical Center Hamburg-Eppendorf (UKE), Martinistraße, 52, 20246, Hamburg, Germany
| | - Bente Siebels
- Section for Mass Spectrometry and Proteomics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hannah Voß
- Section for Mass Spectrometry and Proteomics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Antonia Gocke
- Section for Mass Spectrometry and Proteomics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Hartmut Schlüter
- Section for Mass Spectrometry and Proteomics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michaela Schweizer
- Morphology and Electron Microscopy Core Facility, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hermann C Altmeppen
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tim Magnus
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI) Group, University Medical Center Hamburg-Eppendorf (UKE), Martinistraße, 52, 20246, Hamburg, Germany
| | - Efrat Levy
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY, 10962, USA
- Departments of Psychiatry, Biochemistry and Molecular Pharmacology, and the Neuroscience Institute, NYU Langone Medical Center, New York, NY, 10016, USA
| | - Berta Puig
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI) Group, University Medical Center Hamburg-Eppendorf (UKE), Martinistraße, 52, 20246, Hamburg, Germany.
| |
Collapse
|
4
|
Montero-Almagro G, Bernal-Utrera C, Geribaldi-Doldán N, Nunez-Abades P, Castro C, Rodriguez-Blanco C. Influence of High-Intensity Interval Training on Neuroplasticity Markers in Post-Stroke Patients: Systematic Review. J Clin Med 2024; 13:1985. [PMID: 38610750 PMCID: PMC11012260 DOI: 10.3390/jcm13071985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/19/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Background: Exercise has shown beneficial effects on neuronal neuroplasticity; therefore, we want to analyze the influence of high-intensity interval training (HIIT) on neuroplasticity markers in post-stroke patients. Methods: A systematic review of RCTs including studies with stroke participants was conducted using the following databases (PubMed, LILACS, ProQuest, PEDro, Web of Science). Searches lasted till (20/11/2023). Studies that used a HIIT protocol as the main treatment or as a coadjutant treatment whose outcomes were neural plasticity markers were used and compared with other exercise protocols, controls or other kinds of treatment. Studies that included other neurological illnesses, comorbidities that interfere with stroke or patients unable to complete a HIIT protocol were excluded. HIIT protocol, methods to assess intensity, neuroplasticity markers (plasmatic and neurophysiological) and other types of assessments such as cognitive scales were extracted to make a narrative synthesis. Jadad and PEDro scales were used to assess bias. Results: Eight articles were included, one included lacunar stroke (less than 3 weeks) and the rest had chronic stroke. The results found here indicate that HIIT facilitates neuronal recovery in response to an ischemic injury. This type of training increases the plasma concentrations of lactate, BDNF and VEGF, which are neurotrophic and growth factors involved in neuroplasticity. HIIT also positively regulates other neurophysiological measurements that are directly associated with a better outcome in motor learning tasks. Conclusions: We conclude that HIIT improves post-stroke recovery by increasing neuroplasticity markers. However, a limited number of studies have been found indicating that future studies are needed that assess this effect and include the analysis of the number of intervals and their duration in order to maximize this effect.
Collapse
Affiliation(s)
- Gines Montero-Almagro
- Physiotherapy Department, Faculty of Nursing, Physiotherapy and Podiatry, University of Seville, 41013 Seville, Spain; (G.M.-A.); (C.R.-B.)
| | - Carlos Bernal-Utrera
- Physiotherapy Department, Faculty of Nursing, Physiotherapy and Podiatry, University of Seville, 41013 Seville, Spain; (G.M.-A.); (C.R.-B.)
- Institute for Biomedical Research and Innovation of Cadiz (INiBICA), 11009 Cadiz, Spain; (P.N.-A.); (C.C.)
| | - Noelia Geribaldi-Doldán
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Cadiz, 11002 Cadiz, Spain;
| | - Pedro Nunez-Abades
- Institute for Biomedical Research and Innovation of Cadiz (INiBICA), 11009 Cadiz, Spain; (P.N.-A.); (C.C.)
- Department of Physiology, Faculty of Pharmacy, University of Seville, 41013 Seville, Spain
| | - Carmen Castro
- Institute for Biomedical Research and Innovation of Cadiz (INiBICA), 11009 Cadiz, Spain; (P.N.-A.); (C.C.)
- Department of Biomedicine, Biotechnology and Public Health, Area of Physiology, Faculty of Medicine, University of Cadiz, 11002 Cadiz, Spain
| | - Cleofas Rodriguez-Blanco
- Physiotherapy Department, Faculty of Nursing, Physiotherapy and Podiatry, University of Seville, 41013 Seville, Spain; (G.M.-A.); (C.R.-B.)
| |
Collapse
|
5
|
Wang J, Zhu M, Sun J, Feng L, Yang M, Sun B, Mao L. Gene therapy of adeno-associated virus (AAV) vectors in preclinical models of ischemic stroke. CNS Neurosci Ther 2023; 29:3725-3740. [PMID: 37551863 PMCID: PMC10651967 DOI: 10.1111/cns.14392] [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: 06/09/2023] [Revised: 07/15/2023] [Accepted: 07/27/2023] [Indexed: 08/09/2023] Open
Abstract
Stroke has been associated with devastating clinical outcomes, with current treatment strategies proving largely ineffective. Therefore, there is a need to explore alternative treatment options for addressing post-stroke functional deficits. Gene therapy utilizing adeno-associated viruses (AAVs) as a critical gene vector delivering genes to the central nervous system (CNS) gene delivery has emerged as a promising approach for treating various CNS diseases. This review aims to provide an overview of the biological characteristics of AAV vectors and the therapeutic advancements observed in preclinical models of ischemic stroke. The study further investigates the potential of manipulating AAV vectors in preclinical applications, emphasizing the challenges and prospects in the selection of viral vectors, drug delivery strategies, immune reactions, and clinical translation.
Collapse
Affiliation(s)
- Jing Wang
- Medical College of Qingdao UniversityQingdaoChina
- Institute for Neurological Research, The Second Affiliated HospitalSchool of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical SciencesTaianChina
| | - Mengna Zhu
- Institute for Neurological Research, The Second Affiliated HospitalSchool of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical SciencesTaianChina
| | - Jingyi Sun
- Department of Spinal SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Lina Feng
- Institute for Neurological Research, The Second Affiliated HospitalSchool of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical SciencesTaianChina
| | - Mingfeng Yang
- Institute for Neurological Research, The Second Affiliated HospitalSchool of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical SciencesTaianChina
| | - Baoliang Sun
- Medical College of Qingdao UniversityQingdaoChina
- Institute for Neurological Research, The Second Affiliated HospitalSchool of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical SciencesTaianChina
| | - Leilei Mao
- Institute for Neurological Research, The Second Affiliated HospitalSchool of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical SciencesTaianChina
| |
Collapse
|
6
|
Inoue Y, Sawano T, Yamaguchi N, Inoue S, Takayama A, Nakazawa S, Inagaki S, Nakatani J, Tanaka H. Comparative distribution of
Arcadlin/Protocadherin‐8
mRNA in the intact and ischemic brains of adult mice. J Comp Neurol 2022; 530:2033-2055. [DOI: 10.1002/cne.25319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 02/24/2022] [Accepted: 03/07/2022] [Indexed: 11/12/2022]
Affiliation(s)
- Yosuke Inoue
- Pharmacology Laboratory Department of Biomedical Sciences, College of Life Sciences Ritsumeikan University Shiga Japan
| | - Toshinori Sawano
- Pharmacology Laboratory Department of Biomedical Sciences, College of Life Sciences Ritsumeikan University Shiga Japan
| | - Natsumi Yamaguchi
- Pharmacology Laboratory Department of Biomedical Sciences, College of Life Sciences Ritsumeikan University Shiga Japan
| | - Shota Inoue
- Pharmacology Laboratory Department of Biomedical Sciences, College of Life Sciences Ritsumeikan University Shiga Japan
| | - Akinori Takayama
- Pharmacology Laboratory Department of Biomedical Sciences, College of Life Sciences Ritsumeikan University Shiga Japan
| | - Shuma Nakazawa
- Pharmacology Laboratory Department of Biomedical Sciences, College of Life Sciences Ritsumeikan University Shiga Japan
| | - Shinobu Inagaki
- United Graduate School of Child Development Osaka University Suita Japan
- Department of Physical Therapy Osaka Yukioka College of Health Science Ibaraki Japan
| | - Jin Nakatani
- Pharmacology Laboratory Department of Biomedical Sciences, College of Life Sciences Ritsumeikan University Shiga Japan
| | - Hidekazu Tanaka
- Pharmacology Laboratory Department of Biomedical Sciences, College of Life Sciences Ritsumeikan University Shiga Japan
| |
Collapse
|
7
|
Merino-Serrais P, Plaza-Alonso S, Hellal F, Valero-Freitag S, Kastanauskaite A, Muñoz A, Plesnila N, DeFelipe J. Microanatomical study of pyramidal neurons in the contralesional somatosensory cortex after experimental ischemic stroke. Cereb Cortex 2022; 33:1074-1089. [PMID: 35353195 PMCID: PMC9930620 DOI: 10.1093/cercor/bhac121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 11/13/2022] Open
Abstract
At present, many studies support the notion that after stroke, remote regions connected to the infarcted area are also affected and may contribute to functional outcome. In the present study, we have analyzed possible microanatomical alterations in pyramidal neurons from the contralesional hemisphere after induced stroke. We performed intracellular injections of Lucifer yellow in pyramidal neurons from layer III in the somatosensory cortex of the contralesional hemisphere in an ischemic stroke mouse model. A detailed 3-dimensional analysis of the neuronal complexity and morphological alterations of dendritic spines was then performed. Our results demonstrate that pyramidal neurons from layer III in the somatosensory cortex of the contralesional hemisphere show selective changes in their dendritic arbors, namely, less dendritic complexity of the apical dendritic arbor-but no changes in the basal dendritic arbor. In addition, we found differences in spine morphology in both apical and basal dendrites comparing the contralesional hemisphere with the lesional hemisphere. Our results show that pyramidal neurons of remote areas connected to the infarct zone exhibit a series of selective changes in neuronal complexity and morphological distribution of dendritic spines, supporting the hypothesis that remote regions connected to the peri-infarcted area are also affected after stroke.
Collapse
Affiliation(s)
- Paula Merino-Serrais
- Corresponding author: Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Campus Montegancedo S/N, Pozuelo de Alarcón, Madrid 28223/Instituto Cajal (CSIC), Avenida Doctor Arce, 37, Madrid 28002, Spain.
| | - Sergio Plaza-Alonso
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid 28223, Spain,Departamento de Neurobiología Funcional y de Sistemas, Instituto Cajal, CSIC, Madrid 28002, Spain
| | - Farida Hellal
- Institute for Stroke and Dementia Research (ISD), University of Munich, Munich 81337, Germany,iTERM, Helmholtz center, Munich 85764, Germany
| | - Susana Valero-Freitag
- Institute for Stroke and Dementia Research (ISD), University of Munich, Munich 81337, Germany
| | - Asta Kastanauskaite
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid 28223, Spain,Departamento de Neurobiología Funcional y de Sistemas, Instituto Cajal, CSIC, Madrid 28002, Spain
| | - Alberto Muñoz
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid 28223, Spain,Departamento de Neurobiología Funcional y de Sistemas, Instituto Cajal, CSIC, Madrid 28002, Spain,Departamento de Biología Celular, Universidad Complutense, Madrid 28040, Spain
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research (ISD), University of Munich, Munich 81337, Germany,Munich Cluster of Systems Neurology (Synergy), Munich 85764, Germany
| | - Javier DeFelipe
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid 28223, Spain,Departamento de Neurobiología Funcional y de Sistemas, Instituto Cajal, CSIC, Madrid 28002, Spain,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas. (CIBERNED), ISCIII, Madrid 28031, Spain
| |
Collapse
|
8
|
Wang B, Li X, Yu N, Yang L, Nan C, Guo L, Zhao Z. Intracerebral hemorrhage alters α2δ1 and thrombospondin expression in rats. Exp Ther Med 2022; 23:327. [PMID: 35386622 PMCID: PMC8972837 DOI: 10.3892/etm.2022.11256] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 01/19/2022] [Indexed: 12/02/2022] Open
Abstract
Calcium voltage-gated channel auxiliary subunit (α2δ1) is a non-essential subunit of the voltage-gated calcium channel complex and is ubiquitously expressed in a number of tissues, including the brain. Thrombospondin (TSP)1 and TSP2 are extracellular matrix proteins and belong to the multi-domain glycoprotein family of macromolecular oligomers. TSP1/2 and α2δ1 promote synapse formation and functional recovery following cerebral infarction. However, to the best of our knowledge, the expression levels of α2δ1 and TSP1/2 in brain tissues, and the effects of intracerebral hemorrhage (ICH) on these levels have not yet been elucidated. The present study established a rat model of hemorrhage induced by injecting collagenase IV into the striatum to determine the changes in α2δ1 and TSP1/2 expression following ICH. The protein expression levels of α2δ1 and TSP1 in the striatum after hemorrhage were significantly increased on day 5 and returned to baseline levels on day 21; however, the protein expression levels of TSP2 were decreased on day 5, whereas they increased on day 14, subsequently returning to baseline levels. In addition, using proteomics analysis of tissues from the sham group (saline injection) and at 24 h post-ICH, it was found that both α2δ1 and TSP1 interacted with neural EGFL like 2. Taken together, these findings suggested that the expression levels of α2δ1 and TSP1/2 were altered in brain tissues in response to ICH.
Collapse
Affiliation(s)
- Bingqian Wang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Xiaopeng Li
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Ning Yu
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Liang Yang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Chengrui Nan
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Lisi Guo
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Zongmao Zhao
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| |
Collapse
|
9
|
Neuronal cell life, death, and axonal degeneration as regulated by the BCL-2 family proteins. Cell Death Differ 2020; 28:108-122. [PMID: 33162554 PMCID: PMC7852532 DOI: 10.1038/s41418-020-00654-2] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 10/13/2020] [Accepted: 10/15/2020] [Indexed: 12/17/2022] Open
Abstract
Axonal degeneration and neuronal cell death are fundamental processes in development and contribute to the pathology of neurological disease in adults. Both processes are regulated by BCL-2 family proteins which orchestrate the permeabilization of the mitochondrial outer membrane (MOM). MOM permeabilization (MOMP) results in the activation of pro-apoptotic molecules that commit neurons to either die or degenerate. With the success of small-molecule inhibitors targeting anti-apoptotic BCL-2 proteins for the treatment of lymphoma, we can now envision the use of inhibitors of apoptosis with exquisite selectivity for BCL-2 family protein regulation of neuronal apoptosis in the treatment of nervous system disease. Critical to this development is deciphering which subset of proteins is required for neuronal apoptosis and axon degeneration, and how these two different outcomes are separately regulated. Moreover, noncanonical BCL-2 family protein functions unrelated to the regulation of MOMP, including impacting necroptosis and other modes of cell death may reveal additional potential targets and/or confounders. This review highlights our current understanding of BCL-2 family mediated neuronal cell death and axon degeneration, while identifying future research questions to be resolved to enable regulating neuronal survival pharmacologically.
Collapse
|
10
|
Lange Canhos L, Chen M, Falk S, Popper B, Straub T, Götz M, Sirko S. Repetitive injury and absence of monocytes promote astrocyte self-renewal and neurological recovery. Glia 2020; 69:165-181. [PMID: 32744730 DOI: 10.1002/glia.23893] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/07/2020] [Accepted: 07/08/2020] [Indexed: 12/24/2022]
Abstract
Unlike microglia and NG2 glia, astrocytes are incapable of migrating to sites of injury in the posttraumatic cerebral cortex, instead relying on proliferation to replenish their numbers and distribution in the affected region. However, neither the spectrum of their proliferative repertoire nor their postinjury distribution has been examined in vivo. Using a combination of different thymidine analogs and clonal analysis in a model of repetitive traumatic brain injury, we show for the first time that astrocytes that are quiescent following an initial injury can be coerced to proliferate after a repeated insult in the cerebral cortex grey matter. Interestingly, this process is promoted by invasion of monocytes to the injury site, as their genetic ablation (using CCR2-/- mice) increased the number of repetitively dividing astrocytes at the expense of newly proliferating astrocytes in repeatedly injured parenchyma. These differences profoundly affected both the distribution of astrocytes and recovery period for posttraumatic behavior deficits suggesting key roles of astrocyte self-renewal in brain repair after injury.
Collapse
Affiliation(s)
- Luisa Lange Canhos
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University Munich, Munich, Germany.,Institute of Stem Cell Research, Helmholtz Zentrum Munich, Neuherberg, Germany.,Graduate School of Systemic Neurosciences (GSN-LMU), Ludwig-Maximilians-University Munich, Munich, Germany
| | - Muxin Chen
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Sven Falk
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University Munich, Munich, Germany.,Institute of Stem Cell Research, Helmholtz Zentrum Munich, Neuherberg, Germany
| | - Bastian Popper
- Core Facility Animal Models, Biomedical Center, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Tobias Straub
- Core Facility Bioinformatics, Biomedical Center, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Magdalena Götz
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University Munich, Munich, Germany.,Institute of Stem Cell Research, Helmholtz Zentrum Munich, Neuherberg, Germany.,Excellence Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Swetlana Sirko
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University Munich, Munich, Germany.,Institute of Stem Cell Research, Helmholtz Zentrum Munich, Neuherberg, Germany
| |
Collapse
|
11
|
Hoffmann L, Rust MB, Culmsee C. Actin(g) on mitochondria - a role for cofilin1 in neuronal cell death pathways. Biol Chem 2020; 400:1089-1097. [PMID: 31256058 DOI: 10.1515/hsz-2019-0120] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 04/16/2019] [Indexed: 11/15/2022]
Abstract
Actin dynamics, the coordinated assembly and disassembly of actin filaments (F-actin), are essential for fundamental cellular processes, including cell shaping and motility, cell division or organelle transport. Recent studies highlighted a novel role for actin dynamics in the regulation of mitochondrial morphology and function, for example, through mitochondrial recruitment of dynamin-related protein 1 (Drp1), a key factor in the mitochondrial fission machinery. Mitochondria are dynamic organelles, and permanent fission and fusion is essential to maintain their function in energy metabolism, calcium homeostasis and regulation of reactive oxygen species (ROS). Here, we summarize recent insights into the emerging role of cofilin1, a key regulator of actin dynamics, for mitochondrial shape and function under physiological conditions and during cellular stress, respectively. This is of peculiar importance in neurons, which are particularly prone to changes in actin regulation and mitochondrial integrity and function. In neurons, cofilin1 may contribute to degenerative processes through formation of cofilin-actin rods, and through enhanced mitochondrial fission, mitochondrial membrane permeabilization, and the release of cytochrome c. Overall, mitochondrial impairment induced by dysfunction of actin-regulating proteins such as cofilin1 emerge as important mechanisms of neuronal death with relevance to acute brain injury and neurodegenerative diseases, such as Parkinson's or Alzheimer's disease.
Collapse
Affiliation(s)
- Lena Hoffmann
- Institute of Pharmacology and Clinical Pharmacy, Biochemical-Pharmacological Center Marburg, University of Marburg, Karl-von-Frisch Straße 2, D-35043 Marburg, Germany.,Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus-Liebig-University Gießen, Hans-Meerwein-Straße 6, D-35032 Marburg, Germany
| | - Marco B Rust
- Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus-Liebig-University Gießen, Hans-Meerwein-Straße 6, D-35032 Marburg, Germany.,Molecular Neurobiology Group, Institute of Physiological Chemistry, Biochemical-Pharmacological Center Marburg, University of Marburg, Karl-von-Frisch Straße 2, D-35043 Marburg, Germany
| | - Carsten Culmsee
- Institute of Pharmacology and Clinical Pharmacy, Biochemical-Pharmacological Center Marburg, University of Marburg, Karl-von-Frisch Straße 2, D-35043 Marburg, Germany.,Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus-Liebig-University Gießen, Hans-Meerwein-Straße 6, D-35032 Marburg, Germany.,Third Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
12
|
Ni HY, Song YX, Lin YH, Cao B, Wang DL, Zhang Y, Dong J, Liang HY, Xu K, Li TY, Chang L, Wu HY, Luo CX, Zhu DY. Dissociating nNOS (Neuronal NO Synthase)-CAPON (Carboxy-Terminal Postsynaptic Density-95/Discs Large/Zona Occludens-1 Ligand of nNOS) Interaction Promotes Functional Recovery After Stroke via Enhanced Structural Neuroplasticity. Stroke 2019; 50:728-737. [PMID: 30727847 DOI: 10.1161/strokeaha.118.022647] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Background and Purpose- Stroke is a major public health concern worldwide. Although clinical treatments have improved in the acute period after stroke, long-term therapeutics remain limited to physical rehabilitation in the delayed phase. This study is aimed to determine whether nNOS (neuronal NO synthase)-CAPON (carboxy-terminal postsynaptic density-95/discs large/zona occludens-1 ligand of nNOS) interaction may serve as a new therapeutic target in the delayed phase for stroke recovery. Methods- Photothrombotic stroke and transient middle cerebral artery occlusion were induced in mice. Adeno-associated virus (AAV)-cytomegalovirus (CMV)-CAPON-125C-GFP (green fluorescent protein)-3Flag and the other 2 drugs (Tat-CAPON-12C and ZLc-002) were microinjected into the peri-infarct cortex immediately and 4 to 10 days after photothrombotic stroke, respectively. ZLc-002 was also systemically injected 4 to 10 days after transient middle cerebral artery occlusion. Grid-walking task and cylinder task were conducted to assess motor function. Western blotting, immunohistochemistry, Golgi staining, and electrophysiology recordings were performed to uncover the mechanisms. Results- Stroke increased nNOS-CAPON association in the peri-infarct cortex in the delayed period. Inhibiting the ischemia-induced nNOS-CAPON association substantially decreased the number of foot faults in the grid-walking task and forelimb asymmetry in the cylinder task, suggesting the promotion of functional recovery from stroke. Moreover, dissociating nNOS-CAPON significantly facilitated dendritic remodeling and synaptic transmission, indicated by increased dendritic spine density, dendritic branching, and length and miniature excitatory postsynaptic current frequency but did not affect stroke-elicited neuronal loss, infarct size, or cerebral edema, suggesting that nNOS-CAPON interaction may function via regulating structural neuroplasticity, rather than neuroprotection. Furthermore, ZLc-002 reversed the transient middle cerebral artery occlusion-induced impairment of motor function. Conclusions- Our results reveal that nNOS-CAPON coupling can serve as a novel pharmacological target for functional restoration after stroke.
Collapse
Affiliation(s)
- Huan-Yu Ni
- From the Institution of Stem Cells and Neuroregeneration (H.-Y.N., Y.-X.S., Y.-H.L., B.C., D.-L.W., Y.Z., J.D., H.-Y.L., K.X., T.-Y.L., L.C., H.-Y.W., C.-X.L., D.-Y.Z.), Nanjing Medical University, China.,Department of Pharmacology, School of Pharmacy (H.-Y.N., Y.-X.S., Y.-H.L., B.C., D.-L.W., Y.Z., J.D., H.-Y.L., K.X., T.-Y.L., L.C., H.-Y.W., C.-X.L., D.-Y.Z.), Nanjing Medical University, China
| | - Yi-Xuan Song
- From the Institution of Stem Cells and Neuroregeneration (H.-Y.N., Y.-X.S., Y.-H.L., B.C., D.-L.W., Y.Z., J.D., H.-Y.L., K.X., T.-Y.L., L.C., H.-Y.W., C.-X.L., D.-Y.Z.), Nanjing Medical University, China.,Department of Pharmacology, School of Pharmacy (H.-Y.N., Y.-X.S., Y.-H.L., B.C., D.-L.W., Y.Z., J.D., H.-Y.L., K.X., T.-Y.L., L.C., H.-Y.W., C.-X.L., D.-Y.Z.), Nanjing Medical University, China
| | - Yu-Hui Lin
- From the Institution of Stem Cells and Neuroregeneration (H.-Y.N., Y.-X.S., Y.-H.L., B.C., D.-L.W., Y.Z., J.D., H.-Y.L., K.X., T.-Y.L., L.C., H.-Y.W., C.-X.L., D.-Y.Z.), Nanjing Medical University, China.,Department of Pharmacology, School of Pharmacy (H.-Y.N., Y.-X.S., Y.-H.L., B.C., D.-L.W., Y.Z., J.D., H.-Y.L., K.X., T.-Y.L., L.C., H.-Y.W., C.-X.L., D.-Y.Z.), Nanjing Medical University, China
| | - Bo Cao
- From the Institution of Stem Cells and Neuroregeneration (H.-Y.N., Y.-X.S., Y.-H.L., B.C., D.-L.W., Y.Z., J.D., H.-Y.L., K.X., T.-Y.L., L.C., H.-Y.W., C.-X.L., D.-Y.Z.), Nanjing Medical University, China.,Department of Pharmacology, School of Pharmacy (H.-Y.N., Y.-X.S., Y.-H.L., B.C., D.-L.W., Y.Z., J.D., H.-Y.L., K.X., T.-Y.L., L.C., H.-Y.W., C.-X.L., D.-Y.Z.), Nanjing Medical University, China
| | - Dong-Liang Wang
- From the Institution of Stem Cells and Neuroregeneration (H.-Y.N., Y.-X.S., Y.-H.L., B.C., D.-L.W., Y.Z., J.D., H.-Y.L., K.X., T.-Y.L., L.C., H.-Y.W., C.-X.L., D.-Y.Z.), Nanjing Medical University, China.,Department of Pharmacology, School of Pharmacy (H.-Y.N., Y.-X.S., Y.-H.L., B.C., D.-L.W., Y.Z., J.D., H.-Y.L., K.X., T.-Y.L., L.C., H.-Y.W., C.-X.L., D.-Y.Z.), Nanjing Medical University, China
| | - Yu Zhang
- From the Institution of Stem Cells and Neuroregeneration (H.-Y.N., Y.-X.S., Y.-H.L., B.C., D.-L.W., Y.Z., J.D., H.-Y.L., K.X., T.-Y.L., L.C., H.-Y.W., C.-X.L., D.-Y.Z.), Nanjing Medical University, China.,Department of Pharmacology, School of Pharmacy (H.-Y.N., Y.-X.S., Y.-H.L., B.C., D.-L.W., Y.Z., J.D., H.-Y.L., K.X., T.-Y.L., L.C., H.-Y.W., C.-X.L., D.-Y.Z.), Nanjing Medical University, China
| | - Jian Dong
- From the Institution of Stem Cells and Neuroregeneration (H.-Y.N., Y.-X.S., Y.-H.L., B.C., D.-L.W., Y.Z., J.D., H.-Y.L., K.X., T.-Y.L., L.C., H.-Y.W., C.-X.L., D.-Y.Z.), Nanjing Medical University, China.,Department of Pharmacology, School of Pharmacy (H.-Y.N., Y.-X.S., Y.-H.L., B.C., D.-L.W., Y.Z., J.D., H.-Y.L., K.X., T.-Y.L., L.C., H.-Y.W., C.-X.L., D.-Y.Z.), Nanjing Medical University, China
| | - Hai-Ying Liang
- From the Institution of Stem Cells and Neuroregeneration (H.-Y.N., Y.-X.S., Y.-H.L., B.C., D.-L.W., Y.Z., J.D., H.-Y.L., K.X., T.-Y.L., L.C., H.-Y.W., C.-X.L., D.-Y.Z.), Nanjing Medical University, China.,Department of Pharmacology, School of Pharmacy (H.-Y.N., Y.-X.S., Y.-H.L., B.C., D.-L.W., Y.Z., J.D., H.-Y.L., K.X., T.-Y.L., L.C., H.-Y.W., C.-X.L., D.-Y.Z.), Nanjing Medical University, China
| | - Ke Xu
- From the Institution of Stem Cells and Neuroregeneration (H.-Y.N., Y.-X.S., Y.-H.L., B.C., D.-L.W., Y.Z., J.D., H.-Y.L., K.X., T.-Y.L., L.C., H.-Y.W., C.-X.L., D.-Y.Z.), Nanjing Medical University, China.,Department of Pharmacology, School of Pharmacy (H.-Y.N., Y.-X.S., Y.-H.L., B.C., D.-L.W., Y.Z., J.D., H.-Y.L., K.X., T.-Y.L., L.C., H.-Y.W., C.-X.L., D.-Y.Z.), Nanjing Medical University, China
| | - Ting-You Li
- From the Institution of Stem Cells and Neuroregeneration (H.-Y.N., Y.-X.S., Y.-H.L., B.C., D.-L.W., Y.Z., J.D., H.-Y.L., K.X., T.-Y.L., L.C., H.-Y.W., C.-X.L., D.-Y.Z.), Nanjing Medical University, China.,Department of Pharmacology, School of Pharmacy (H.-Y.N., Y.-X.S., Y.-H.L., B.C., D.-L.W., Y.Z., J.D., H.-Y.L., K.X., T.-Y.L., L.C., H.-Y.W., C.-X.L., D.-Y.Z.), Nanjing Medical University, China.,Department of Medicinal Chemistry, School of Pharmacy (T.-Y.L.), Nanjing Medical University, China.,Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing, China (T.-Y.L., C.-X.L., D.-Y.Z.)
| | - Lei Chang
- From the Institution of Stem Cells and Neuroregeneration (H.-Y.N., Y.-X.S., Y.-H.L., B.C., D.-L.W., Y.Z., J.D., H.-Y.L., K.X., T.-Y.L., L.C., H.-Y.W., C.-X.L., D.-Y.Z.), Nanjing Medical University, China.,Department of Pharmacology, School of Pharmacy (H.-Y.N., Y.-X.S., Y.-H.L., B.C., D.-L.W., Y.Z., J.D., H.-Y.L., K.X., T.-Y.L., L.C., H.-Y.W., C.-X.L., D.-Y.Z.), Nanjing Medical University, China
| | - Hai-Yin Wu
- From the Institution of Stem Cells and Neuroregeneration (H.-Y.N., Y.-X.S., Y.-H.L., B.C., D.-L.W., Y.Z., J.D., H.-Y.L., K.X., T.-Y.L., L.C., H.-Y.W., C.-X.L., D.-Y.Z.), Nanjing Medical University, China.,Department of Pharmacology, School of Pharmacy (H.-Y.N., Y.-X.S., Y.-H.L., B.C., D.-L.W., Y.Z., J.D., H.-Y.L., K.X., T.-Y.L., L.C., H.-Y.W., C.-X.L., D.-Y.Z.), Nanjing Medical University, China
| | - Chun-Xia Luo
- From the Institution of Stem Cells and Neuroregeneration (H.-Y.N., Y.-X.S., Y.-H.L., B.C., D.-L.W., Y.Z., J.D., H.-Y.L., K.X., T.-Y.L., L.C., H.-Y.W., C.-X.L., D.-Y.Z.), Nanjing Medical University, China.,Department of Pharmacology, School of Pharmacy (H.-Y.N., Y.-X.S., Y.-H.L., B.C., D.-L.W., Y.Z., J.D., H.-Y.L., K.X., T.-Y.L., L.C., H.-Y.W., C.-X.L., D.-Y.Z.), Nanjing Medical University, China.,Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing, China (T.-Y.L., C.-X.L., D.-Y.Z.)
| | - Dong-Ya Zhu
- From the Institution of Stem Cells and Neuroregeneration (H.-Y.N., Y.-X.S., Y.-H.L., B.C., D.-L.W., Y.Z., J.D., H.-Y.L., K.X., T.-Y.L., L.C., H.-Y.W., C.-X.L., D.-Y.Z.), Nanjing Medical University, China.,Department of Pharmacology, School of Pharmacy (H.-Y.N., Y.-X.S., Y.-H.L., B.C., D.-L.W., Y.Z., J.D., H.-Y.L., K.X., T.-Y.L., L.C., H.-Y.W., C.-X.L., D.-Y.Z.), Nanjing Medical University, China.,Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing, China (T.-Y.L., C.-X.L., D.-Y.Z.)
| |
Collapse
|
13
|
Kanazawa M, Takahashi T, Ishikawa M, Onodera O, Shimohata T, Del Zoppo GJ. Angiogenesis in the ischemic core: A potential treatment target? J Cereb Blood Flow Metab 2019; 39:753-769. [PMID: 30841779 PMCID: PMC6501515 DOI: 10.1177/0271678x19834158] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The ischemic penumbra is both a concept in understanding the evolution of cerebral tissue injury outcome of focal ischemia and a potential therapeutic target for ischemic stroke. In this review, we examine the evidence that angiogenesis can contribute to beneficial outcomes following focal ischemia in model systems. Several studies have shown that, following cerebral ischemia, endothelial proliferation and subsequent angiogenesis can be detected beginning four days after cerebral ischemia in the border of the ischemic core, or in the ischemic periphery, in rodent and non-human primate models, although initial signals appear within hours of ischemia onset. Components of the neurovascular unit, its participation in new vessel formation, and the nature of the core and penumbra responses to experimental focal cerebral ischemia, are considered here. The potential co-localization of vascular remodeling and axonal outgrowth following focal cerebral ischemia based on the definition of tissue remodeling and the processes that follow ischemic stroke are also considered. The region of angiogenesis in the ischemic core and its surrounding tissue (ischemic periphery) may be a novel target for treatment. We summarize issues that are relevant to model studies of focal cerebral ischemia looking ahead to potential treatments.
Collapse
Affiliation(s)
- Masato Kanazawa
- 1 Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Tetsuya Takahashi
- 1 Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Masanori Ishikawa
- 1 Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Osamu Onodera
- 1 Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Takayoshi Shimohata
- 2 Department of Neurology and Geriatrics, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Gregory J Del Zoppo
- 3 Department of Medicine (Division of Hematology), University of Washington, Seattle, WA, USA.,4 Department of Neurology, University of Washington, Seattle, WA, USA
| |
Collapse
|
14
|
Treadmill exercise ameliorates focal cerebral ischemia/reperfusion-induced neurological deficit by promoting dendritic modification and synaptic plasticity via upregulating caveolin-1/VEGF signaling pathways. Exp Neurol 2019; 313:60-78. [DOI: 10.1016/j.expneurol.2018.12.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 12/02/2018] [Accepted: 12/10/2018] [Indexed: 11/18/2022]
|
15
|
Henley R, Chandrasekaran V, Giulivi C. Computing neurite outgrowth and arborization in superior cervical ganglion neurons. Brain Res Bull 2018; 144:194-199. [PMID: 30529562 DOI: 10.1016/j.brainresbull.2018.12.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/30/2018] [Accepted: 12/04/2018] [Indexed: 11/18/2022]
Abstract
Dendrites are the primary site of synaptic activity in neurons and changes in synapses are often the first pathological stage in neurodegenerative diseases. Molecular studies of these changes rely on morphological analysis of the imaging of somas and dendritic arbors of cultured or primary neurons. As research on preventing or reversing synaptic degeneration develops, demands increase for user-friendly 2D neurite analyzers without undermining accuracy and reproducibility. The most common method of 2D neurite analysis is manual by using ImageJ. This method relies completely on the user's ability to distinguish the shape and size of dendrites and trace morphology with a series of straight connected lines. Semi-automatic methods have also been developed, such as the NeuronJ plugin for ImageJ. These methods still rely on the user to identify the start and end of the dendrites, but automatically determine the shape, reducing the likelihood of user bias and speeding the process. Some automatic methods have been developed through image processing software, like ImagePro. These programs tend to be expensive, but have been shown to be fast and effective, limiting user interaction. In this study, we compare three methods of neurite analysis-ImageJ, NeuronJ, and ImagePro-in measuring the soma size, number of dendrites, and length of dendrites per cell of embryonic sympathetic rat neurons with BMP-7-induced dendritic growth. Our results indicate that ImageJ and NeuronJ measurements were of similar effectiveness and consistent throughout various images and multiple trials. NeuronJ required less user interaction in measuring the length of dendrites than the manual method and therefore, was faster and less labor intensive. Conversely, ImagePro tended to be inconsistent across images, overestimating both soma size and the number of dendrites per cell while underestimating the length of dendrites. Overall, NeuronJ, in conjunction with ImageJ, is the most reliable and efficient method of 2D neurite analysis tested in the present study.
Collapse
Affiliation(s)
- Rachel Henley
- Department of Biology, Saint Mary's College of California, Moraga, CA, 94575, United States
| | - Vidya Chandrasekaran
- Department of Biology, Saint Mary's College of California, Moraga, CA, 94575, United States
| | - Cecilia Giulivi
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, United States; Medical Investigations of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, CA 95817, United States.
| |
Collapse
|
16
|
Wang X, Xuan W, Zhu ZY, Li Y, Zhu H, Zhu L, Fu DY, Yang LQ, Li PY, Yu WF. The evolving role of neuro-immune interaction in brain repair after cerebral ischemic stroke. CNS Neurosci Ther 2018; 24:1100-1114. [PMID: 30350341 DOI: 10.1111/cns.13077] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 09/23/2018] [Accepted: 09/25/2018] [Indexed: 12/13/2022] Open
Abstract
Stroke is the world's leading cause of disability with limited brain repair treatments which effectively improve long-term neurological deficits. The neuroinflammatory responses persist into the late repair phase of stroke and participate in all brain repair elements, including neurogenesis, angiogenesis, synaptogenesis, remyelination and axonal sprouting, shedding new light on post-stroke brain recovery. Resident brain glial cells, such as astrocytes not only contribute to neuroinflammation after stroke, but also secrete a wide range of trophic factors that can promote post-stroke brain repair. Alternatively, activated microglia, monocytes, and neutrophils in the innate immune system, traditionally considered as major damaging factors after stroke, have been suggested to be extensively involved in brain repair after stroke. The adaptive immune system may also have its bright side during the late regenerative phase, affecting the immune suppressive regulatory T cells and B cells. This review summarizes the recent findings in the evolving role of neuroinflammation in multiple post-stroke brain repair mechanisms and poses unanswered questions that may generate new directions for future research and give rise to novel therapeutic targets to improve stroke recovery.
Collapse
Affiliation(s)
- Xin Wang
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Wei Xuan
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Zi-Yu Zhu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yan Li
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Hao Zhu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Ling Zhu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Dan-Yun Fu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Li-Qun Yang
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Pei-Ying Li
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Wei-Feng Yu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
17
|
Imaging the Transformation of Ipsilateral Internal Capsule Following Focal Cerebral Ischemia in Rat by Diffusion Kurtosis Imaging. J Stroke Cerebrovasc Dis 2017; 26:42-48. [DOI: 10.1016/j.jstrokecerebrovasdis.2016.08.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 08/05/2016] [Accepted: 08/17/2016] [Indexed: 12/13/2022] Open
|
18
|
Nair VA, Beniwal-Patel P, Mbah I, Young BM, Prabhakaran V, Saha S. Structural Imaging Changes and Behavioral Correlates in Patients with Crohn's Disease in Remission. Front Hum Neurosci 2016; 10:460. [PMID: 27695405 PMCID: PMC5025433 DOI: 10.3389/fnhum.2016.00460] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 08/31/2016] [Indexed: 11/27/2022] Open
Abstract
Background: Crohn’s disease (CD) is a subtype of inflammatory bowel disease caused by immune-mediated inflammation in the gastrointestinal tract. The extent of morphologic brain alterations and their associated cognitive and affective impairments remain poorly characterized. Aims: We used magnetic resonance imaging to identify structural brain differences between patients with Crohn’s disease in remission compared to age-matched healthy controls and evaluated for structural-behavioral correlates. Methods: Nineteen patients and 20 healthy, age-matched controls were recruited in the study. Group differences in brain morphometric measures and correlations between brain measures and performance on a cognitive task, the verbal fluency (VF) task, were examined. Correlations between brain measures and cognitive measures as well as self-reported measures of depression, personality, and affective scales were examined. Results: Patients showed significant cortical thickening in the left superior frontal region compared to controls. Significant group differences were observed in sub-cortical volume measures in both hemispheres. Investigation of brain-behavior correlations revealed significant group differences in the correlation between cortical surface area and VF performance, although behavioral performance was equivalent between the two groups. The left middle temporal surface area was a significant predictor of VF performance with controls showing a significant positive correlation between these measures, and patients showing the opposite effect. Conclusion: Our results indicate key differences in structural brain measures in patients with CD compared to controls. Additionally, correlation between brain measures and behavioral responses suggest there may be a neural basis to the alterations in patients’ cognitive and affective responses.
Collapse
Affiliation(s)
- Veena A Nair
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin - Madison, Madison WI, USA
| | - Poonam Beniwal-Patel
- Division of Gastroenterology and Hepatology, University of Wisconsin - Madison, Madison WI, USA
| | - Ifeanyi Mbah
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin - Madison, MadisonWI, USA; Division of Gastroenterology and Hepatology, University of Wisconsin - Madison, MadisonWI, USA
| | - Brittany M Young
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin - Madison, MadisonWI, USA; Medical Scientist Training Program, University of Wisconsin - Madison, MadisonWI, USA; Neuroscience Training Program, University of Wisconsin - Madison, MadisonWI, USA
| | - Vivek Prabhakaran
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin - Madison, MadisonWI, USA; Medical Scientist Training Program, University of Wisconsin - Madison, MadisonWI, USA; Neuroscience Training Program, University of Wisconsin - Madison, MadisonWI, USA; Department of Neurology, University of Wisconsin - Madison, MadisonWI, USA; Department of Psychology and Department of Psychiatry, University of Wisconsin - Madison, MadisonWI, USA
| | - Sumona Saha
- Division of Gastroenterology and Hepatology, University of Wisconsin - Madison, Madison WI, USA
| |
Collapse
|
19
|
Nikolakopoulou AM, Koeppen J, Garcia M, Leish J, Obenaus A, Ethell IM. Astrocytic Ephrin-B1 Regulates Synapse Remodeling Following Traumatic Brain Injury. ASN Neuro 2016; 8:1-18. [PMID: 26928051 PMCID: PMC4774052 DOI: 10.1177/1759091416630220] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 12/31/2015] [Indexed: 01/06/2023] Open
Abstract
Traumatic brain injury (TBI) can result in tissue alterations distant from the site of the initial injury, which can trigger pathological changes within hippocampal circuits and are thought to contribute to long-term cognitive and neuropsychological impairments. However, our understanding of secondary injury mechanisms is limited. Astrocytes play an important role in brain repair after injury and astrocyte-mediated mechanisms that are implicated in synapse development are likely important in injury-induced synapse remodeling. Our studies suggest a new role of ephrin-B1, which is known to regulate synapse development in neurons, in astrocyte-mediated synapse remodeling following TBI. Indeed, we observed a transient upregulation of ephrin-B1 immunoreactivity in hippocampal astrocytes following moderate controlled cortical impact model of TBI. The upregulation of ephrin-B1 levels in hippocampal astrocytes coincided with a decline in the number of vGlut1-positive glutamatergic input to CA1 neurons at 3 days post injury even in the absence of hippocampal neuron loss. In contrast, tamoxifen-induced ablation of ephrin-B1 from adult astrocytes in ephrin-B1loxP/yERT2-CreGFAP mice accelerated the recovery of vGlut1-positive glutamatergic input to CA1 neurons after TBI. Finally, our studies suggest that astrocytic ephrin-B1 may play an active role in injury-induced synapse remodeling through the activation of STAT3-mediated signaling in astrocytes. TBI-induced upregulation of STAT3 phosphorylation within the hippocampus was suppressed by astrocyte-specific ablation of ephrin-B1 in vivo, whereas the activation of ephrin-B1 in astrocytes triggered an increase in STAT3 phosphorylation in vitro. Thus, regulation of ephrin-B1 signaling in astrocytes may provide new therapeutic opportunities to aid functional recovery after TBI.
Collapse
Affiliation(s)
| | - Jordan Koeppen
- Biomedical Sciences Division, School of Medicine, University of California Riverside, CA, USA Cell, Molecular, and Developmental Biology graduate program, University of California Riverside, CA, USA
| | - Michael Garcia
- Biomedical Sciences Division, School of Medicine, University of California Riverside, CA, USA
| | - Joshua Leish
- Biomedical Sciences Division, School of Medicine, University of California Riverside, CA, USA
| | - Andre Obenaus
- Department of Pediatrics, School of Medicine, Loma Linda University, CA, USA
| | - Iryna M Ethell
- Biomedical Sciences Division, School of Medicine, University of California Riverside, CA, USA Cell, Molecular, and Developmental Biology graduate program, University of California Riverside, CA, USA
| |
Collapse
|
20
|
Xu Y, Hou QH, Russell SD, Bennett BC, Sellers AJ, Lin Q, Huang DF. Neuroplasticity in post-stroke gait recovery and noninvasive brain stimulation. Neural Regen Res 2016; 10:2072-80. [PMID: 26889202 PMCID: PMC4730838 DOI: 10.4103/1673-5374.172329] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Gait disorders drastically affect the quality of life of stroke survivors, making post-stroke rehabilitation an important research focus. Noninvasive brain stimulation has potential in facilitating neuroplasticity and improving post-stroke gait impairment. However, a large inter-individual variability in the response to noninvasive brain stimulation interventions has been increasingly recognized. We first review the neurophysiology of human gait and post-stroke neuroplasticity for gait recovery, and then discuss how noninvasive brain stimulation techniques could be utilized to enhance gait recovery. While post-stroke neuroplasticity for gait recovery is characterized by use-dependent plasticity, it evolves over time, is idiosyncratic, and may develop maladaptive elements. Furthermore, noninvasive brain stimulation has limited reach capability and is facilitative-only in nature. Therefore, we recommend that noninvasive brain stimulation be used adjunctively with rehabilitation training and other concurrent neuroplasticity facilitation techniques. Additionally, when noninvasive brain stimulation is applied for the rehabilitation of gait impairment in stroke survivors, stimulation montages should be customized according to the specific types of neuroplasticity found in each individual. This could be done using multiple mapping techniques.
Collapse
Affiliation(s)
- Yi Xu
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China; Guangdong Provincial Engineering Technology Research Center for Rehabilitation Medicine and Clinical Translation, Guangzhou, Guangdong Province, China; Motion Analysis and Motor Performance Laboratory, Department of Orthopedics and Mechanical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Qing-Hua Hou
- Department of Neurology, Guangdong No.2 Provincial People's Hospital, Guangzhou, Guangdong Province, China
| | - Shawn D Russell
- Motion Analysis and Motor Performance Laboratory, Department of Orthopedics and Mechanical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Bradford C Bennett
- H.C Sweere Center for Clinical Biomechanics and Applied Ergonomics, Northwestern Health Science University, Bloomington, MN, USA
| | - Andrew J Sellers
- Department of Radiology, Naval Medical Center Portsmouth, Portsmouth, VA, USA
| | - Qiang Lin
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China; Guangdong Provincial Engineering Technology Research Center for Rehabilitation Medicine and Clinical Translation, Guangzhou, Guangdong Province, China
| | - Dong-Feng Huang
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China; Guangdong Provincial Engineering Technology Research Center for Rehabilitation Medicine and Clinical Translation, Guangzhou, Guangdong Province, China
| |
Collapse
|
21
|
Liu Y, Sun Z, Sun S, Duan Y, Shi J, Qi Z, Meng R, Sun Y, Zeng X, Chui D, Ji X. Effects of hypoxic preconditioning on synaptic ultrastructure in mice. Synapse 2014; 69:7-14. [PMID: 25155519 DOI: 10.1002/syn.21777] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Revised: 07/21/2014] [Accepted: 08/08/2014] [Indexed: 11/06/2022]
Abstract
Hypoxic preconditioning (HPC) elicits resistance to more drastic subsequent insults, which potentially provide neuroprotective therapeutic strategy, but the underlying mechanisms remain to be fully elucidated. Here, we examined the effects of HPC on synaptic ultrastructure in olfactory bulb of mice. Mice underwent up to five cycles of repeated HPC treatments, and hypoxic tolerance was assessed with a standard gasp reflex assay. As expected, HPC induced an increase in tolerance time. To assess synaptic responses, Western blots were used to quantify protein levels of representative markers for glia, neuron, and synapse, and transmission electron microscopy was used to examine synaptic ultrastructure and mitochondrial density. HPC did not significantly alter the protein levels of astroglial marker (GFAP), neuron-specific markers (GAP43, Tuj-1, and OMP), synaptic number markers (synaptophysin and SNAP25) or the percentage of excitatory synapses versus inhibitory synapses. However, HPC significantly affected synaptic curvature and the percentage of synapses with presynaptic mitochondria, which showed concomitant change pattern. These findings demonstrate that HPC is associated with changes in synaptic ultrastructure.
Collapse
Affiliation(s)
- Yi Liu
- China-America Joint Institute of Neuroscience, CAJIN, Xuanwu Hospital, Capital Medical University, Beijing, China; Neuroprotection Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Neuroprotection Research Laboratory, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Effects of activated protein C on the size of modeled ischemic focus and morphometric parameters of neurons and neuroglia in its perifocal zone. Bull Exp Biol Med 2014; 157:530-4. [PMID: 25110099 DOI: 10.1007/s10517-014-2607-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Indexed: 10/24/2022]
Abstract
The effects of activated protein C (APC) on the quantitative parameters of neurons and neuroglia in the perifocal zone of infarction induced in the left hemispheric cortex were studied in two groups of rats. Group 1 animals served as control (control infarction). Group 2 rats were injected with APC (50 μg/kg) in the right lateral cerebral ventricle 3 h after infarction was induced, and after 72 h the infarction size was evaluated and the neurons and neuroglia in the perifocal zone were counted. APC reduced the infarction size 2.5 times in comparison with the control and reduced by 16% the neuronal death in the perifocal zone layer V, causing no appreciable changes in layer III, and did not change the size of neuronal bodies but increased (by 11%) the size of neuronal nuclei in layer III. The protein maintained the sharply increased count of gliocytes in the perifocal zone of infarction and promoted their growth. Hence, APC protected the neurons from death in the ischemic focus by increasing the gliocyte count and stimulating the compensatory reparative processes.
Collapse
|
23
|
Adult cortical plasticity following injury: Recapitulation of critical period mechanisms? Neuroscience 2014; 283:4-16. [PMID: 24791715 DOI: 10.1016/j.neuroscience.2014.04.029] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 04/16/2014] [Accepted: 04/17/2014] [Indexed: 12/12/2022]
Abstract
A primary goal of research on developmental critical periods (CPs) is the recapitulation of a juvenile-like state of malleability in the adult brain that might enable recovery from injury. These ambitions are often framed in terms of the simple reinstatement of enhanced plasticity in the growth-restricted milieu of an injured adult brain. Here, we provide an analysis of the similarities and differences between deprivation-induced and injury-induced cortical plasticity, to provide for a nuanced comparison of these remarkably similar processes. As a first step, we review the factors that drive ocular dominance plasticity in the primary visual cortex of the uninjured brain during the CP and in adults, to highlight processes that might confer adaptive advantage. In addition, we directly compare deprivation-induced cortical plasticity during the CP and plasticity following acute injury or ischemia in mature brain. We find that these two processes display a biphasic response profile following deprivation or injury: an initial decrease in GABAergic inhibition and synapse loss transitions into a period of neurite expansion and synaptic gain. This biphasic response profile emphasizes the transition from a period of cortical healing to one of reconnection and recovery of function. Yet while injury-induced plasticity in adult shares several salient characteristics with deprivation-induced plasticity during the CP, the degree to which the adult injured brain is able to functionally rewire, and the time required to do so, present major limitations for recovery. Attempts to recapitulate a measure of CP plasticity in an adult injury context will need to carefully dissect the circuit alterations and plasticity mechanisms involved while measuring functional behavioral output to assess their ultimate success.
Collapse
|
24
|
Ito U, Hakamata Y, Watabe K, Oyanagi K. Mannitol infusion immediately after reperfusion suppresses the development of focal cortical infarction after temporary cerebral ischemia in gerbils. Neuropathology 2014; 34:360-9. [PMID: 24661099 PMCID: PMC4238828 DOI: 10.1111/neup.12113] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Accepted: 02/19/2014] [Indexed: 11/28/2022]
Abstract
Previously we found that, after temporary cerebral ischemia, microvasculogenic secondary focal cerebral cortical ischemia occurred, caused by microvascular obstruction due to compression by swollen astrocytic end-feet, resulting in focal infarction. Herein, we examined whether mannitol infusion immediately after restoration of blood flow could protect the cerebral cortex against the development of such an infarction. If so, the infusion of mannitol might improve the results of vascular reperfusion therapy. We selected stroke-positive animals during the first 10 min after left carotid occlusion performed twice with a 5-h interval, and allocated them into four groups: sham-operated control, no-treatment, mannitol-infusion, and saline-infusion groups. Light- and electron-microscopic studies were performed on cerebral cortices of coronal sections prepared at the chiasmatic level, where the focal infarction develops abruptly in the area where disseminated selective neuronal necrosis is maturing. Measurements were performed to determine the following: (A) infarct size in HE-stained specimens from all groups at 72 and 120 h after return of blood flow; (B) number of carbon-black-suspension-perfused microvessels in the control and at 0.5, 3, 5, 8, 12 and 24 h in the no-treatment and mannitol-infusion groups; (C) area of astrocytic end-feet; and (D) number of mitochondria in the astrocytic end-feet in electron microscopic pictures taken at 5 h. The average decimal fraction area ratio of infarct size in the mannitol group was significantly reduced at 72 and 120 h, associated with an increased decimal fraction number ratio of carbon-black-suspension-perfused microvessels at 3, 5 and 8 h, and a marked reduction in the size of the end-feet at 5 h. Mannitol infusion performed immediately after restitution of blood flow following temporary cerebral ischemia remarkably reduced the size of the cerebral cortical focal infarction by decreasing the swelling of the end-feet, thus preventing the microvascular compression and stasis and thereby microvasculogenic secondary focal cerebral ischemia.
Collapse
Affiliation(s)
- Umeo Ito
- Laboratory for Neurodegenerative Pathology, Department of Sensory and Motor Systems, Tokyo Metropolitan Institute of Medical Science; Division of Neuropathology, Department of Brain Disease Research, Shinshu University School of Medicine, Matsumoto, Japan
| | | | | | | |
Collapse
|
25
|
Ito U, Hakamata Y, Watabe K, Oyanagi K. Astrocytic involvement in the maturation phenomenon after temporary cerebral ischemia. ACTA NEUROCHIRURGICA. SUPPLEMENT 2013; 118:23-9. [PMID: 23564099 DOI: 10.1007/978-3-7091-1434-6_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2023]
Abstract
Astrocytes support neuronal functions by regulating the extracellular ion homeostasis and levels of neurotransmitters, and by providing fuel such as lactate to the neurons via their processes (APs). After two 10-min unilateral carotid occlusions with a 5-h interval in gerbils, we investigated maturing disseminated selective neuronal necrosis (DSNN) on the coronal surface sectioned at the infundibular level. We chronologically counted the normal appearing, degenerated, and dead neurons and astrocytes in the cerebral cortex; observed the ultrastructure of APs, and counted the number of their cut-ends and mitochondria in the neuropil; determined the percentage volume of APs according to Weibel's point-counting method; compared the number of cut-ends and mitochondria and percentage volume of APs around the astrocytes and around the normal-appearing, degenerated, and dead -neurons. Heterogeneous degeneration of APs was concluded to be closely associated with the maturation of DSNN.Using the same model, at the coronally sectioned surface on the chiasmatic level, we investigated the mechanism of development of focal infarction in the maturing DSNN. Same as in the above study, we chronologically counted various neurons and astrocytes; observed and measured the area of the ultrastructure of astrocytic end-feet; counted the number of carbon-black-suspension-perfused microvessels. We concluded that after temporary cerebral ischemia, secondary focal ischemia was induced by microvascular obstruction compressed by swollen astrocytic end-feet, resulting in delayed focal infarction.
Collapse
Affiliation(s)
- Umeo Ito
- Department of Sensory and Motor Systems, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.
| | | | | | | |
Collapse
|
26
|
Abstract
Changes in brain circuits occur within specific paradigms of action in the adult brain. These paradigms include changes in behavioral activity patterns, alterations in environmental experience, and direct brain injury. Each of these paradigms can produce axonal sprouting, dendritic morphology changes, and alterations in synaptic connectivity. Activity-, experience-, and injury-dependent plasticity alter neuronal network function and behavioral output, and in the case of brain injury, may produce neurological recovery. The molecular substrate for adult neuronal plasticity overlaps in these three paradigms in key signaling pathways. These common pathways for adult plasticity suggest common mechanisms for activity-, experience-, and injury-dependent plasticity. These common pathways may also interact to enhance or impede each other during adult recovery of function after injury. This review focuses on common molecular changes evoked during the process of adult neuronal plasticity, with a focus on neural repair in stroke.
Collapse
|
27
|
Thalamic T-type Ca²+ channels mediate frontal lobe dysfunctions caused by a hypoxia-like damage in the prefrontal cortex. J Neurosci 2011; 31:4063-73. [PMID: 21411648 DOI: 10.1523/jneurosci.4493-10.2011] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Hypoxic damage to the prefrontal cortex (PFC) has been implicated in the frontal lobe dysfunction found in various neuropsychiatric disorders. The underlying subcortical mechanisms, however, have not been well explored. In this study, we induced a PFC-specific hypoxia-like damage by cobalt-wire implantation to demonstrate that the role of the mediodorsal thalamus (MD) is critical for the development of frontal lobe dysfunction, including frontal lobe-specific seizures and abnormal hyperactivity. Before the onset of these abnormalities, the cross talk between the MD and PFC nuclei at theta frequencies was enhanced. During the theta frequency interactions, burst spikes, known to depend on T-type Ca(2+) channels, were increased in MD neurons. In vivo knockout or knockdown of the T-type Ca(2+) channel gene (Ca(V)3.1) in the MD substantially reduced the theta frequency MD-PFC cross talk, frontal lobe-specific seizures, and locomotor hyperactivity in this model. These results suggest a two-step model of prefrontal dysfunction in which the response to a hypoxic lesion in the PFC results in abnormal thalamocortical feedback driven by thalamic T-type Ca(2+) channels, which, in turn, leads to the onset of neurological and behavioral abnormalities. This study provides valuable insights into preventing the development of neuropsychiatric disorders arising from irreversible PFC damage.
Collapse
|
28
|
|
29
|
Mostany R, Chowdhury TG, Johnston DG, Portonovo SA, Carmichael ST, Portera-Cailliau C. Local hemodynamics dictate long-term dendritic plasticity in peri-infarct cortex. J Neurosci 2010; 30:14116-26. [PMID: 20962232 PMCID: PMC6634780 DOI: 10.1523/jneurosci.3908-10.2010] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2010] [Revised: 08/16/2010] [Accepted: 08/19/2010] [Indexed: 11/21/2022] Open
Abstract
Changes in dendritic spine turnover are a major mechanism of experience-dependent plasticity in the adult neocortex. Dendritic spine plasticity may also contribute to functional recovery after stroke, but in that setting its expression may be complicated by alterations in local tissue perfusion, especially around the infarct. Using adult Thy-1 GFP-M mice, we simultaneously recorded long-term spine dynamics in apical dendrites from layer 5 pyramidal cells and blood flow from surrounding capillaries with in vivo two-photon microscopy in peri-infarct cortex before and after unilateral middle cerebral artery occlusion. Blood flow in peri-infarct cortex decreased significantly immediately after stroke and improved gradually over time, in a distance-dependent manner from the epicenter of the infarct. However, local tissue perfusion was never fully restored even after a 3 month recovery period. On average, surviving layer 5 pyramidal neurons experienced a ∼20% decrease in spine density acutely after stroke but eventually recovered. The dynamics of this improvement were different depending on the degree of tissue perfusion acutely after arterial occlusion. Cells in ischemic areas closer to the infarct returned to normal spine density levels slowly by retaining spines, while cells in more remote regions with preserved blood flow recovered faster by adding more spines, eventually surpassing baseline spine density by 15%. Our data suggest that maintaining tissue perfusion in the area surrounding the infarct could hasten or augment synaptic plasticity and functional recovery after stroke.
Collapse
Affiliation(s)
- Ricardo Mostany
- Departments of Neurology and Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California 90095, USA.
| | | | | | | | | | | |
Collapse
|
30
|
Abstract
Brain plasticity describes the potential of the organ for adaptive changes involved in various phenomena in health and disease. A substantial amount of experimental evidence, received in animal and cell models, shows that a cascade of plastic changes at the molecular, cellular, and tissue levels, is initiated in different regions of the postischemic brain. Underlying mechanisms include neurochemical alterations, functional changes in excitatory and inhibitory synapses, axonal and dendritic sprouting, and reorganization of sensory and motor central maps. Multiple lines of evidence indicate numerous points in which the process of postischemic recovery may be influenced with the aim to restore the full capacity of the brain tissue injured by an ischemic episode.
Collapse
Affiliation(s)
- Galyna G Skibo
- Department of Cytology, Bogomoletz Institute of Physiology, Kiev, Ukraine
| | | |
Collapse
|
31
|
Nikonenko AG, Radenovic L, Andjus PR, Skibo GG. Structural Features of Ischemic Damage in the Hippocampus. Anat Rec (Hoboken) 2009; 292:1914-21. [DOI: 10.1002/ar.20969] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
32
|
Ito U, Hakamata Y, Kawakami E, Oyanagi K. Degeneration of astrocytic processes and their mitochondria in cerebral cortical regions peripheral to the cortical infarction: heterogeneity of their disintegration is closely associated with disseminated selective neuronal necrosis and maturation of injury. Stroke 2009; 40:2173-81. [PMID: 19359621 DOI: 10.1161/strokeaha.108.534990] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Astrocytes support neuronal functions by regulating the extracellular ion-homeostasis and levels of neurotransmitters, and by providing fuel such as lactate to the neurons via their astrocytic processes (APs). Whether injured APs are associated with neuronal survival/death is still an unanswered question. We investigated APs in the neuropil, especially those around astrocytes and normal-appearing, degenerating, and dead neurons in cerebral cortical regions peripheral to the cortical infarction (RPI). METHODS Stroke-positive gerbils were euthanized at various times after the ischemic insult. Ultrathin sections were obtained from the RPI sectioned coronally at the infundibular level. We counted the number of normal-appearing, degenerated, and dead neurons and astrocytes in paraffin sections, the number of cut-ends and mitochondria in APs in the neuropil on electron-microscopic photographs, and determined the percent-volume of APs by Weibel point-counting method. We compared the number of cut-ends and mitochondria and percent-volume of APs around astrocytes at 5 hours and 48 hours, and around normal-appearing, degenerated, and dead neurons at 12 hours. RESULTS Although the number of astrocytes did not change (average of 12.3+/-0.20%) during 0 to 48 hours, that of the dead neurons increased from 9.71+/-1.34 to 44.39+/-1.40% during 5 to 48 hours postischemia. The number of normal-appearing APs and mitochondria in APs decreased respectively from 13.49+/-0.65 to 1.61+/-0.14/28.20 microm(2) and from 1.86+/-0.18 to 0.61+/-0.07/28.20 microm(2) in the neuropil during 0 to 48 hours. The number of normal-appearing APs around astrocytes decreased from 12.3+/-0.19 to 1.7+/-0.05/38.33 microm(2) with an increase in percent-volume of degenerated APs from 1.17+/-0.04 to 11.45+/-0.23%, from 5 to 48 hours postischemia. The number of normal-appearing APs decreased from 4.36+/-0.52 to 1.56+/-0.17/38.33 microm(2) with an increase in percent-volume of degenerated APs, from 2.41+/-0.52 to 12.55+/-1.0%, from around the normal-appearing to dead neurons, at 12 hours. CONCLUSIONS In the RPI, heterogeneous degeneration of APs was closely associated with disseminated selective neuronal necrosis and the maturation phenomenon seen in ischemic neuronal injury.
Collapse
Affiliation(s)
- Umeo Ito
- Department of Neuropathology, Tokyo Metropolitan Institute for Neuroscience, 2-6 Musashidai, Fuchu-shi, Tokyo 183-8526, Japan.
| | | | | | | |
Collapse
|
33
|
Chu CT, Plowey ED, Dagda RK, Hickey RW, Cherra SJ, Clark RSB. Autophagy in neurite injury and neurodegeneration: in vitro and in vivo models. Methods Enzymol 2009; 453:217-49. [PMID: 19216909 DOI: 10.1016/s0076-6879(08)04011-1] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Recent advances indicate that maintaining a balanced level of autophagy is critically important for neuronal health and function. Pathologic dysregulation of macroautophagy has been implicated in synaptic dysfunction, cellular stress, and neuronal cell death. Autophagosomes and autolysosomes are induced in acute and chronic neurological disorders including stroke, brain trauma, neurotoxin injury, Parkinson's, Alzheimer's, Huntington's, motor neuron, prion, lysosomal storage, and other neurodegenerative diseases. Compared to other cell types, neuronal autophagy research presents particular challenges that may be addressed through still evolving techniques. Neuronal function depends upon maintenance of axons and dendrites (collectively known as neurites) that extend for great distances from the cell body. Both autophagy and mitochondrial content have been implicated in regulation of neurite length and function in physiological (plasticity) and pathological remodeling. Here, we highlight several molecular cell biological and imaging methods to study autophagy and mitophagy in neuritic and somatic compartments of differentiated neuronal cell lines and primary neuron cultures, using protocols developed in toxic and genetic models of parkinsonian neurodegeneration. In addition, mature neurons can be studied using in vivo protocols for modeling ischemic and traumatic injuries. Future challenges include application of automated computer-assisted image analysis to the axodendritic tree of individual neurons and improving methods for measuring neuronal autophagic flux.
Collapse
Affiliation(s)
- Charleen T Chu
- Department of Pathology, Division of Neuropathology, University of Pittsburgh School of Medicine and Center for Neuroscience, Pittsburgh, Pennsylvania, USA
| | | | | | | | | | | |
Collapse
|
34
|
Cao F, Hata R, Zhu P, Niinobe M, Sakanaka M. Up-regulation of syntaxin1 in ischemic cortex after permanent focal ischemia in rats. Brain Res 2009; 1272:52-61. [PMID: 19344701 DOI: 10.1016/j.brainres.2009.03.047] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2008] [Revised: 03/18/2009] [Accepted: 03/22/2009] [Indexed: 11/25/2022]
Abstract
Syntaxin1 and synaptotagmin are located in the pre-synaptic terminals and play central roles in Ca(2+)-triggered neurotransmitter release. Because excessive synaptic transmission has been implicated in neuronal cell death after ischemia, we investigated the effects of cerebral ischemia on the levels of these proteins using a rat permanent focal ischemia model. Western blot analysis revealed that the protein level of syntaxin1 was significantly up-regulated in the ischemic core cortex and peri-ischemic cortex at 1 day after ischemia, while the protein level of synaptotagmin was not. Immunohistochemical analysis revealed that the protein level of syntaxin1 was markedly up-regulated in the ischemic areas where immunoreaction for MAP2 was lost. Furthermore, we showed that resident microglial cells were quite vulnerable to ischemia. Our data provide novel insights into the molecular mechanism of cerebral ischemia at the pre-synaptic terminals.
Collapse
Affiliation(s)
- Fang Cao
- Department of Functional Histology, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime 791-0295, Japan
| | | | | | | | | |
Collapse
|
35
|
Long-term evaluation of cytoarchitectonic characteristics of prefrontal cortex pyramidal neurons, following global cerebral ischemia and neuroprotective melatonin treatment, in rats. Neurosci Lett 2008; 448:148-52. [DOI: 10.1016/j.neulet.2008.10.043] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2008] [Revised: 10/10/2008] [Accepted: 10/14/2008] [Indexed: 11/16/2022]
|
36
|
The development of stroke therapeutics: promising mechanisms and translational challenges. Neuropharmacology 2008; 56:329-41. [PMID: 19007799 DOI: 10.1016/j.neuropharm.2008.10.006] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2008] [Revised: 09/29/2008] [Accepted: 10/06/2008] [Indexed: 12/13/2022]
Abstract
Ischemic stroke is the second most common cause of death worldwide and a major cause of disability. Intravenous thrombolysis with rt-PA remains the only available acute therapy in patients who present within 3h of stroke onset other than the recently approved mechanical MERCI device, substantiating the high unmet need in available stroke therapeutics. The development of successful therapeutic strategies remains challenging, as evidenced by the continued failures of new therapies in clinical trials. However, significant lessons have been learned and this knowledge is currently being incorporated into improved pre-clinical and clinical design. Furthermore, advancements in imaging technologies and continued progress in understanding biological pathways have established a prolonged presence of salvageable penumbral brain tissue and have begun to elucidate the natural repair response initiated by ischemic insult. We review important past and current approaches to drug development with an emphasis on implementing principles of translational research to achieve a rigorous conversion of knowledge from bench to bedside. We highlight current strategies to protect and repair brain tissue with the promise to provide longer therapeutic windows, preservation of multiple tissue compartments and improved clinical success.
Collapse
|
37
|
DaSilva AF, Becerra L, Pendse G, Chizh B, Tully S, Borsook D. Colocalized structural and functional changes in the cortex of patients with trigeminal neuropathic pain. PLoS One 2008; 3:e3396. [PMID: 18923647 PMCID: PMC2561059 DOI: 10.1371/journal.pone.0003396] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2008] [Accepted: 09/11/2008] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Recent data suggests that in chronic pain there are changes in gray matter consistent with decreased brain volume, indicating that the disease process may produce morphological changes in the brains of those affected. However, no study has evaluated cortical thickness in relation to specific functional changes in evoked pain. In this study we sought to investigate structural (gray matter thickness) and functional (blood oxygenation dependent level - BOLD) changes in cortical regions of precisely matched patients with chronic trigeminal neuropathic pain (TNP) affecting the right maxillary (V2) division of the trigeminal nerve. The model has a number of advantages including the evaluation of specific changes that can be mapped to known somatotopic anatomy. METHODOLOGY/PRINCIPAL FINDINGS Cortical regions were chosen based on sensory (Somatosensory cortex (SI and SII), motor (MI) and posterior insula), or emotional (DLPFC, Frontal, Anterior Insula, Cingulate) processing of pain. Both structural and functional (to brush-induced allodynia) scans were obtained and averaged from two different imaging sessions separated by 2-6 months in all patients. Age and gender-matched healthy controls were also scanned twice for cortical thickness measurement. Changes in cortical thickness of TNP patients were frequently colocalized and correlated with functional allodynic activations, and included both cortical thickening and thinning in sensorimotor regions, and predominantly thinning in emotional regions. CONCLUSIONS Overall, such patterns of cortical thickness suggest a dynamic functionally-driven plasticity of the brain. These structural changes, which correlated with the pain duration, age-at-onset, pain intensity and cortical activity, may be specific targets for evaluating therapeutic interventions.
Collapse
Affiliation(s)
- Alexandre F DaSilva
- PAIN Group, Brain Imaging Center, Mclean Hospital, Harvard Medical School, Boston, MA, USA.
| | | | | | | | | | | |
Collapse
|
38
|
Liauw J, Hoang S, Choi M, Eroglu C, Choi M, Sun GH, Percy M, Wildman-Tobriner B, Bliss T, Guzman RG, Barres BA, Steinberg GK. Thrombospondins 1 and 2 are necessary for synaptic plasticity and functional recovery after stroke. J Cereb Blood Flow Metab 2008; 28:1722-32. [PMID: 18594557 DOI: 10.1038/jcbfm.2008.65] [Citation(s) in RCA: 207] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Thrombospondins 1 and 2 (TSP-1/2) belong to a family of extracellular glycoproteins with angiostatic and synaptogenic properties. Although TSP-1/2 have been postulated to drive the resolution of postischemic angiogenesis, their role in synaptic and functional recovery is unknown. We investigated whether TSP-1/2 are necessary for synaptic and motor recovery after stroke. Focal ischemia was induced in 8- to 12-week-old wild-type (WT) and TSP-1/2 knockout (KO) mice by unilateral occlusion of the distal middle cerebral artery and the common carotid artery (CCA). Thrombospondins 1 and 2 increased after stroke, with both TSP-1 and TSP-2 colocalizing mostly to astrocytes. Wild-type and TSP-1/2 KO mice were compared in angiogenesis, synaptic density, axonal sprouting, infarct size, and functional recovery at different time points after stroke. Using the tongue protrusion test of motor function, we observed that TSP-1/2 KO mice exhibited significant deficit in their ability to recover function (P<0.05) compared with WT mice. No differences were found in infarct size and blood vessel density between the two groups after stroke. However, TSP-1/2 KO mice exhibited significant synaptic density and axonal sprouting deficits. Deficiency of TSP-1/2 leads to impaired recovery after stroke mainly due to the role of these proteins in synapse formation and axonal outgrowth.
Collapse
Affiliation(s)
- Jason Liauw
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California 94305-5327, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Park SJ, Jung YJ, Kim YA, Lee-Kang JH, Lee KE. Glucose/oxygen deprivation and reperfusion upregulate SNAREs and complexin in organotypic hippocampal slice cultures. Neuropathology 2008; 28:612-20. [PMID: 18503508 DOI: 10.1111/j.1440-1789.2008.00927.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Brain ischemia activates Ca(2+)-dependent synaptic vesicle exocytosis. The synaptosomal-associated protein 25 (SNAP-25) and syntaxin proteins, located on presynaptic terminals, are components of the SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) complex and play a key role in regulating exocytosis. Changes in the expression of SNAREs could affect SNARE complex formation, fusion of vesicles with the presynaptic membrane, and release of neurotransmitters through exocytosis. To investigate the relationship of glucose/oxygen deprivation (GOD)/reperfusion-induced neuronal damage and alteration of presynaptic function, we examined the expression of SNAREs and complexin during GOD and reperfusion using organotypic hippocampal slice cultures. Microtubule-associated protein 2 (MAP-2) staining and transmission electron microscopy showed that neuronal damage increased in a time-dependent manner and both types of neuronal death can occur at different times during GOD and reperfusion. The immunoreactivity of SNAREs such as SNAP-25, vesicle-associated membrane protein and syntaxin and complexin increased in pyramidal cell bodies in the CA1 and CA3 areas in a time-dependent manner following reperfusion. Our data suggest that alteration of presynaptic function may play a partial role in delayed neuronal death during GOD and reperfusion in organotypic hippocampal slice cultures.
Collapse
Affiliation(s)
- Su Jin Park
- Department of Pharmacology, School of Medicine, Ewha Woman's University, Yangcheon-Gu, Seoul, South Korea
| | | | | | | | | |
Collapse
|
40
|
Manganese-enhanced MRI of brain plasticity in relation to functional recovery after experimental stroke. J Cereb Blood Flow Metab 2008; 28:832-40. [PMID: 17987047 DOI: 10.1038/sj.jcbfm.9600576] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Restoration of function after stroke may be associated with structural remodeling of neuronal connections outside the infarcted area. However, the spatiotemporal profile of poststroke alterations in neuroanatomical connectivity in relation to functional recovery is still largely unknown. We performed in vivo magnetic resonance imaging (MRI)-based neuronal tract tracing with manganese in combination with immunohistochemical detection of the neuronal tracer wheat-germ agglutinin horseradish peroxidase (WGA-HRP), to assess changes in intra- and interhemispheric sensorimotor network connections from 2 to 10 weeks after unilateral stroke in rats. In addition, functional recovery was measured by repetitive behavioral testing. Four days after tracer injection in perilesional sensorimotor cortex, manganese enhancement and WGA-HRP staining were decreased in subcortical areas of the ipsilateral sensorimotor network at 2 weeks after stroke, which was restored at later time points. At 4 to 10 weeks after stroke, we detected significantly increased manganese enhancement in the contralateral hemisphere. Behaviorally, sensorimotor functions were initially disturbed but subsequently recovered and plateaued 17 days after stroke. This study shows that manganese-enhanced MRI can provide unique in vivo information on the spatiotemporal pattern of neuroanatomical plasticity after stroke. Our data suggest that the plateau stage of functional recovery is associated with restoration of ipsilateral sensorimotor pathways and enhanced interhemispheric connectivity.
Collapse
|
41
|
Di Filippo M, Tozzi A, Costa C, Belcastro V, Tantucci M, Picconi B, Calabresi P. Plasticity and repair in the post-ischemic brain. Neuropharmacology 2008; 55:353-62. [PMID: 18359495 DOI: 10.1016/j.neuropharm.2008.01.012] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2007] [Revised: 01/29/2008] [Accepted: 01/30/2008] [Indexed: 12/22/2022]
Abstract
Stroke is the second commonest cause of death and the principal cause of adult disability in the world. In most cases ischemic injuries have been reported to induce mild to severe permanent deficits. Nevertheless, recovery is often dynamic, reflecting the ability of the injured neuronal networks to adapt. Plastic phenomena occurring in the cerebral cortex and in subcortical structures after ischemic injuries have been documented at the synaptic, cellular, and network level and several findings suggest that they may play a key role during neurorehabilitation in human stroke survivors. In particular, in vitro studies have demonstrated that oxygen and glucose deprivation (in vitro ischemia) exerts long-term effects on the efficacy of synaptic transmission via the induction of a post-ischemic long-term potentiation (i-LTP). i-LTP may deeply influence the plastic reorganization of cortical representational maps occurring after cerebral ischemia, inducing a functional connection of previously non-interacting neurons. On the other hand, there is evidence that i-LTP may exert a detrimental effect in the peri-infarct area, facilitating excitotoxic processes via the sustained, long-term enhancement of glutamate mediated neurotransmission. In the present work we will review the molecular and synaptic mechanisms underlying ischemia-induced synaptic plastic changes taking into account their potential adaptive and/or detrimental effects on the neuronal network in which they occur. Thereafter, we will consider the implications of brain plastic phenomena in the post-stroke recovery phase as well as during the rehabilitative and therapeutic intervention in human subjects.
Collapse
Affiliation(s)
- Massimiliano Di Filippo
- Clinica Neurologica, Università degli Studi di Perugia, Ospedale S Maria della Misericordia, Via S Andrea delle Fratte, Perugia, Italy
| | | | | | | | | | | | | |
Collapse
|
42
|
Giffard C, Landeau B, Kerrouche N, Young AR, Barré L, Baron JC. Decreased chronic-stage cortical 11C-flumazenil binding after focal ischemia-reperfusion in baboons: a marker of selective neuronal loss? Stroke 2008; 39:991-9. [PMID: 18239185 DOI: 10.1161/strokeaha.107.489419] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Although the penumbra can be saved by early reperfusion, in the rat it is consistently affected by selective neuronal loss. Mapping selective neuronal loss in the living primate would be desirable. METHODS Five young adult baboons underwent (15)O positron emission tomography for cerebral blood flow, cerebral oxygen consumption, and oxygen extraction fraction mapping at baseline and serially during and after 20-hour temporary middle cerebral artery occlusion. At approximately day 30, (11)C-flumazenil (FMZ), a potential positron emission tomography marker of selective neuronal loss, and structural magnetic resonance-based infarct mapping were obtained, and the brain was perfused-fixed. Reduced FMZ binding in noninfarcted cortical middle cerebral artery areas was searched voxel-wise, and specific binding was assessed using compartmental modeling of FMZ time-activity curves. RESULTS Visual inspection revealed reduced late FMZ uptake in the affected cortical territory, extending well beyond the infarct. Accordingly, the incidence of selected voxels was greater than chance, documenting mildly but significantly reduced FMZ uptake and specific binding. Serial (15)O positron emission tomography revealed moderately severe acute ischemia followed by reperfusion. Histopathology documented only mild neuronal changes in or near the affected areas. CONCLUSIONS We document moderate but definite late FMZ binding decrements in noninfarcted cortical areas in the baboon, consistent with previous rat and human studies. These were acutely characterized by moderate ischemia followed by reperfusion, consistent with neuronal damage from ischemic or reperfusion injury in the salvaged at-risk tissue. Only mild histopathological changes subtended these FMZ alterations suggesting subtle processes such as isolated dendrite or synapse loss. Whether these changes impact on clinical outcome deserves studying because they may be targeted by specific neuroprotection.
Collapse
Affiliation(s)
- Cyril Giffard
- INSERM U 320, Centre CYCERON, University of Caen, France
| | | | | | | | | | | |
Collapse
|
43
|
Brown CE, Murphy TH. Livin' on the edge: imaging dendritic spine turnover in the peri-infarct zone during ischemic stroke and recovery. Neuroscientist 2007; 14:139-46. [PMID: 18039977 DOI: 10.1177/1073858407309854] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The spontaneous recovery of sensory, motor, and cognitive functions after stroke is thought to be mediated primarily through the reorganization and rewiring of surviving brain circuits. Given that dendritic spine turnover underlies rewiring during normal development and plasticity, this process is likely to play a key role in mediating functional changes that occur during and after stroke. Recently, a new approach has been taken using two-photon microscopy to monitor, in real time, the temporal and spatial progression of dendritic plasticity in the living animal, both while it is experiencing the initial ischemic episode as well as during long-term recovery from stroke damage. Here, we highlight recent evidence showing that stroke can trigger extensive changes in the relatively hardwired adult brain. For example, when dendrites are challenged by acute ischemia, they can disintegrate within minutes of ischemia and rapidly reassemble during reperfusion. Over longer time scales, dendrites in the surviving peri-infarct zone show heightened levels of spine turnover for many weeks after stroke, thereby raising the possibility that future stroke therapies may be able to facilitate or optimize dendritic rewiring to improve functional recovery.
Collapse
Affiliation(s)
- Craig E Brown
- Department of Psychiatry, Brain Research Center University of British Columbia, Vancouver, British Columbia, Canada.
| | | |
Collapse
|
44
|
Rodríguez-González R, Hurtado O, Sobrino T, Castillo J. Neuroplasticity and cellular therapy in cerebral infarction. Cerebrovasc Dis 2007; 24 Suppl 1:167-80. [PMID: 17971653 DOI: 10.1159/000107393] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Stroke is the second to third most common cause of death in adults, and more than a third of people who survive a stroke will have severe disability. Therapeutic options currently centre on fibrinolytic treatment, but its limitations restrict use to a small proportion of patients. Although a wide range of neuroprotective substances has been effective in experimental models, they have repeatedly failed in clinical trials because of toxicity or loss of effectiveness. Recent strategies based on neuroplasticity and cellular therapy have shown significant efficacy in improving functional recovery in experimental models, although further study is still necessary to clarify how the brain responds to ischaemic damage and is able to reorganize itself in the long term. Although steps must still be taken to ensure the safety and feasibility of treatments based on neuroplasticity and cellular therapy, neurorepair strategies provide promising future therapeutic options for stroke.
Collapse
Affiliation(s)
- Raquel Rodríguez-González
- Clinical Neuroscience Research Laboratory, Division of Vascular Neurology, Department of Neurology, Hospital Clínico Universitario, University of Santiago de Compostela, Santiago de Compostela, Spain
| | | | | | | |
Collapse
|
45
|
Ito U, Nagasao J, Kawakami E, Oyanagi K. Fate of Disseminated Dead Neurons in the Cortical Ischemic Penumbra. Stroke 2007; 38:2577-83. [PMID: 17673709 DOI: 10.1161/strokeaha.107.484394] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background and Purpose—
Because the mechanism for scavenging acidophilic electron-dense dead neurons disseminated among the neuritic networks of surviving neurons in the ischemic penumbra of the cerebral cortex is still obscure, we investigated the fate of them up to 24 weeks after the ischemic insult.
Methods—
Stroke-positive animals were selected according to their stroke index score during the first 10-minute left carotid occlusion done twice with a 5-hour interval. The animals were killed at various times after the second ischemic insult. Ultrathin sections including the second through fourth cortical layers were obtained from the neocortex coronally sectioned at the infundibular level in which the penumbra appeared and was observed by electron microscopy. We determined the percentages of resting, activated, and phagocytic microglia and astrocytes in the specimens obtained at various times postischemia.
Results—
The electron-dense neurons had been fragmented into granular pieces by invading astrocytic processes from the periphery of the dead neurons and only the central portion remained. These granular pieces were dispersed along the extracellular spaces in the neuropil. By 8 to 24 weeks, the central core portion became a tiny vesicular particle (3.5 to 5.5 μm in diameter) with a central dot. Microglia and astrocytes phagocytized these dispersed granular pieces.
Conclusions—
We found a novel scavenger mechanism in the ischemic penumbra, one by which dead neurons were fragmented by invading small astrocytic processes and only a thinned-out core portion remained, which finally became a tiny vesicular particle. The dispersed fragmented pieces were phagocytized by the microglia and astrocytes late, at 8 to 24 weeks postischemia.
Collapse
Affiliation(s)
- Umeo Ito
- Department of Neuropathology, Tokyo Metropolitan Institute for Neuroscience, Musashidai, Fuchu-shi, Tokyo, Japan.
| | | | | | | |
Collapse
|
46
|
Brown CE, Li P, Boyd JD, Delaney KR, Murphy TH. Extensive turnover of dendritic spines and vascular remodeling in cortical tissues recovering from stroke. J Neurosci 2007; 27:4101-9. [PMID: 17428988 PMCID: PMC6672555 DOI: 10.1523/jneurosci.4295-06.2007] [Citation(s) in RCA: 274] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Recovery of function after stroke is thought to be dependent on the reorganization of adjacent, surviving areas of the brain. Macroscopic imaging studies (functional magnetic resonance imaging, optical imaging) have shown that peri-infarct regions adopt new functional roles to compensate for damage caused by stroke. To better understand the process by which these regions reorganize, we used in vivo two-photon imaging to examine changes in dendritic and vascular structure in cortical regions recovering from stroke. In adult control mice, dendritic arbors were relatively stable with very low levels of spine turnover (<0.5% turnover over 6 h). After stroke, however, the organization of dendritic arbors in peri-infarct cortex was fundamentally altered with both apical dendrites and blood vessels radiating in parallel from the lesion. On a finer scale, peri-infarct dendrites were exceptionally plastic, manifested by a dramatic increase in the rate of spine formation that was maximal at 1-2 weeks (5-8-fold increase), and still evident 6 weeks after stroke. These changes were selective given that turnover rates were not significantly altered in ipsilateral cortical regions more distant to the lesion (>1.5 mm). These data provide a structural framework for understanding functional and behavioral changes that accompany brain injury and suggest new targets that could be exploited by future therapies to rebuild and rewire neuronal circuits lost to stroke.
Collapse
Affiliation(s)
- Craig E. Brown
- Department of Psychiatry, Brain Research Center, University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3, and
| | - Ping Li
- Department of Psychiatry, Brain Research Center, University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3, and
| | - Jamie D. Boyd
- Department of Biology, University of Victoria, Victoria, British Columbia, Canada V8W 3N5
| | - Kerry R. Delaney
- Department of Biology, University of Victoria, Victoria, British Columbia, Canada V8W 3N5
| | - Timothy H. Murphy
- Department of Psychiatry, Brain Research Center, University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3, and
| |
Collapse
|