1
|
Yang H, Xiang Y, Wang J, Ke Z, Zhou W, Yin X, Zhang M, Chen Z. Modulating the blood-brain barrier in CNS disorders: A review of the therapeutic implications of secreted protein acidic and rich in cysteine (SPARC). Int J Biol Macromol 2025; 288:138747. [PMID: 39674451 DOI: 10.1016/j.ijbiomac.2024.138747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 12/09/2024] [Accepted: 12/11/2024] [Indexed: 12/16/2024]
Abstract
Secreted protein acidic and rich in cysteine (SPARC), an essential stromal cell protein, plays a crucial role in angiogenesis and maintaining endothelial barrier function. This protein is expressed by diverse cell types, including endothelial cells, fibroblasts, and macrophages, with increased expression found in regions of tissues undergoing active remodeling, repair, and proliferation. The role of SPARC in non-neural tissues is of significant interest. In the central nervous system (CNS), SPARC is highly expressed in blood vessels during early development. It becomes down-regulated as the brain matures, a pattern consistent with its role in angiogenesis and blood-brain barrier (BBB) establishment. In this review, we explore the multifaceted roles of SPARC in regulating CNS disorders, particularly its action in angiogenesis, inflammatory responses, neural system development and repair, barrier establishment, maintenance of BBB function, and the pathogenesis of CNS disorders triggered by BBB dysfunction.
Collapse
Affiliation(s)
- Hui Yang
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China; School of Basic Medicine, Jiujiang University, Jiujiang, Jiangxi 332000, China
| | - Yuanyuan Xiang
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China
| | - Jiaxuan Wang
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China; School of Basic Medicine, Jiujiang University, Jiujiang, Jiangxi 332000, China
| | - Zunliang Ke
- Department of Neurosurgery, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, China
| | - Weixin Zhou
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China
| | - Xiaoping Yin
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China
| | - Manqing Zhang
- School of Basic Medicine, Jiujiang University, Jiujiang, Jiangxi 332000, China.
| | - Zhiying Chen
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China.
| |
Collapse
|
2
|
Shu H, Parada I, Delgado A, Prince DA, Gu F. Increased excitatory connectivity and epileptiform activity in thrombospondin1/2 knockout mice following cortical trauma. Neurobiol Dis 2024; 200:106634. [PMID: 39122122 DOI: 10.1016/j.nbd.2024.106634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 07/23/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024] Open
Abstract
Thrombospondins (TSPs) are astrocyte-secreted extracellular matrix proteins that play key roles as regulators of synaptogenesis in the central nervous system. We previously showed that TSP1/2 are upregulated in the partial neocortical isolation model ("undercut" or "UC" below) of posttraumatic epileptogenesis and may contribute to abnormal axonal sprouting, aberrant synaptogenesis and epileptiform discharges in the UC cortex. These results led to the hypothesis that posttraumatic epileptogeneis would be reduced in TSP1/2 knockout (TSP1/2 KO) mice. To test the hypothesis, we made UC lesions at P21, and subsequent experiments were conducted 14d later at P35. Ex vivo extracellular single or multi-electrode field potential recordings were obtained from layer V in cortical slices at P35 and in vivo video-EEGs of spontaneous epileptiform bursts were recorded to examine the effect of TSP1/2 deletion on epileptogenesis following cortical injury. Immunohistochemical experiments were performed to assess the effect of TSP1/2 KO + UC on the number of putative excitatory synapses and the expression of TSP4 and HEVIN, other astrocytic proteins known to up-regulate excitatory synapse formation. Unexpectedly, our results showed that, compared with WT + UC mice, TSP1/2 KO + UC mice displayed increased epileptiform activity, as indicated by 1) increased incidence and more rapid propagation of evoked and spontaneous epileptiform discharges in UC neocortical slices; 2) increased occurrence of spontaneous epileptiform discharges in vivo. There was an associated increase in the density of VLUT1/PSD95-IR colocalizations (putative excitatory synapses) and significantly upregulated TSP4- and HEVIN-IR in TSP1/2 KO + UC versus WT + UC mice. Results suggest that TSP1/2 deletion plays a potential epileptogenic role following neocortical injury, associated with compensatory upregulation of TSP4 and HEVIN, which may contribute to the increase in the density of excitatory synapses and resulting neural network hyperexcitability.
Collapse
Affiliation(s)
- Haifeng Shu
- Department of Neurosurgery, General Hospital of Western Theatre Command, College of Medicine of Southwest Jiaotong University, China
| | - Isabel Parada
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Alex Delgado
- Department of Biological Sciences, University of North Texas, Denton, TX 76205, USA
| | - David A Prince
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Feng Gu
- Department of Biological Sciences, University of North Texas, Denton, TX 76205, USA.
| |
Collapse
|
3
|
Davletshin AI, Matveeva AA, Poletaeva II, Evgen'ev MB, Garbuz DG. The role of molecular chaperones in the mechanisms of epileptogenesis. Cell Stress Chaperones 2023; 28:599-619. [PMID: 37755620 PMCID: PMC10746656 DOI: 10.1007/s12192-023-01378-1] [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: 07/17/2023] [Revised: 08/30/2023] [Accepted: 09/08/2023] [Indexed: 09/28/2023] Open
Abstract
Epilepsy is a group of neurological diseases which requires significant economic costs for the treatment and care of patients. The central point of epileptogenesis stems from the failure of synaptic signal transmission mechanisms, leading to excessive synchronous excitation of neurons and characteristic epileptic electroencephalogram activity, in typical cases being manifested as seizures and loss of consciousness. The causes of epilepsy are extremely diverse, which is one of the reasons for the complexity of selecting a treatment regimen for each individual case and the high frequency of pharmacoresistant cases. Therefore, the search for new drugs and methods of epilepsy treatment requires an advanced study of the molecular mechanisms of epileptogenesis. In this regard, the investigation of molecular chaperones as potential mediators of epileptogenesis seems promising because the chaperones are involved in the processing and regulation of the activity of many key proteins directly responsible for the generation of abnormal neuronal excitation in epilepsy. In this review, we try to systematize current data on the role of molecular chaperones in epileptogenesis and discuss the prospects for the use of chemical modulators of various chaperone groups' activity as promising antiepileptic drugs.
Collapse
Affiliation(s)
| | - Anna A Matveeva
- Engelhardt Institute of Molecular Biology RAS, 119991, Moscow, Russia
- Moscow Institute of Physics and Technology, 141700, Dolgoprudny, Moscow Region, Russia
| | - Inga I Poletaeva
- Biology Department, Lomonosov Moscow State University, 119991, Moscow, Russia
| | | | - David G Garbuz
- Engelhardt Institute of Molecular Biology RAS, 119991, Moscow, Russia
| |
Collapse
|
4
|
Zveik O, Fainstein N, Rechtman A, Haham N, Ganz T, Lavon I, Brill L, Vaknin‐Dembinsky A. Cerebrospinal fluid of progressive multiple sclerosis patients reduces differentiation and immune functions of oligodendrocyte progenitor cells. Glia 2022; 70:1191-1209. [PMID: 35266197 PMCID: PMC9314832 DOI: 10.1002/glia.24165] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 02/20/2022] [Accepted: 02/21/2022] [Indexed: 12/31/2022]
Abstract
Oligodendrocyte progenitor cells (OPCs) are responsible for remyelination in the central nervous system (CNS) in health and disease. For patients with multiple sclerosis (MS), remyelination is not always successful, and the mechanisms differentiating successful from failed remyelination are not well-known. Growing evidence suggests an immune role for OPCs, in addition to their regenerative role; however, it is not clear if this helps or hinders the regenerative process. We studied the effect of cerebrospinal fluid (CSF) from relapsing MS (rMS) and progressive MS (pMS) patients on primary OPC differentiation and immune gene expression and function. We observed that CSF from either rMS or pMS patients has a differential effect on the ability of mice OPCs to differentiate into mature oligodendrocytes and to express immune functions. CSF of pMS patients impaired differentiation into mature oligodendrocytes. In addition, it led to decreased major histocompatibility complex class (MHC)-II expression, tumor necrosis factor (TNF)-α secretion, nuclear factor kappa-B (NFκB) activation, and less activation and proliferation of T cells. Our findings suggest that OPCs are not only responsible for remyelination, but they may also play an active role as innate immune cells in the CNS.
Collapse
Affiliation(s)
- Omri Zveik
- Faculty of Medicine, Hebrew University of Jerusalem, Department of Neurology and Laboratory of NeuroimmunologyThe Agnes‐Ginges Center for Neurogenetics, Hadassah – Hebrew University Medical CenterJerusalemIsrael
| | - Nina Fainstein
- Faculty of Medicine, Hebrew University of Jerusalem, Department of Neurology and Laboratory of NeuroimmunologyThe Agnes‐Ginges Center for Neurogenetics, Hadassah – Hebrew University Medical CenterJerusalemIsrael
| | - Ariel Rechtman
- Faculty of Medicine, Hebrew University of Jerusalem, Department of Neurology and Laboratory of NeuroimmunologyThe Agnes‐Ginges Center for Neurogenetics, Hadassah – Hebrew University Medical CenterJerusalemIsrael
| | - Nitzan Haham
- Faculty of Medicine, Hebrew University of Jerusalem, Department of Neurology and Laboratory of NeuroimmunologyThe Agnes‐Ginges Center for Neurogenetics, Hadassah – Hebrew University Medical CenterJerusalemIsrael
| | - Tal Ganz
- Faculty of Medicine, Hebrew University of Jerusalem, Department of Neurology and Laboratory of NeuroimmunologyThe Agnes‐Ginges Center for Neurogenetics, Hadassah – Hebrew University Medical CenterJerusalemIsrael
| | - Iris Lavon
- Faculty of Medicine, Hebrew University of Jerusalem, Department of Neurology and Laboratory of NeuroimmunologyThe Agnes‐Ginges Center for Neurogenetics, Hadassah – Hebrew University Medical CenterJerusalemIsrael
- Leslie and Michael Gaffin Center for Neuro‐OncologyHadassah‐Hebrew University Medical CenterJerusalemIsrael
| | - Livnat Brill
- Faculty of Medicine, Hebrew University of Jerusalem, Department of Neurology and Laboratory of NeuroimmunologyThe Agnes‐Ginges Center for Neurogenetics, Hadassah – Hebrew University Medical CenterJerusalemIsrael
| | - Adi Vaknin‐Dembinsky
- Faculty of Medicine, Hebrew University of Jerusalem, Department of Neurology and Laboratory of NeuroimmunologyThe Agnes‐Ginges Center for Neurogenetics, Hadassah – Hebrew University Medical CenterJerusalemIsrael
| |
Collapse
|
5
|
Characterization of Hevin (SPARCL1) Immunoreactivity in Postmortem Human Brain Homogenates. Neuroscience 2021; 467:91-109. [PMID: 34033869 DOI: 10.1016/j.neuroscience.2021.05.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 11/22/2022]
Abstract
Hevin is a matricellular glycoprotein that plays important roles in neural developmental processes such as neuronal migration, synaptogenesis and synaptic plasticity. In contrast to other matricellular proteins whose expression decreases when development is complete, hevin remains highly expressed, suggesting its involvement in adult brain function. In vitro studies have shown that hevin can have different post-translational modifications. However, the glycosylation pattern of hevin in the human brain remains unknown, as well as its relative distribution and localization. The present study provides the first thorough characterization of hevin protein expression by Western blot in postmortem adult human brain. Our results demonstrated two major specific immunoreactive bands for hevin: an intense band migrating around 130 kDa, and a band migrating around 100 kDa. Biochemical assays revealed that both hevin bands have a different glycosylation pattern. Subcellular fractionation showed greater expression in membrane-enriched fraction than in cytosolic preparation, and a higher expression in prefrontal cortex (PFC) compared to hippocampus (HIP), caudate nucleus (CAU) and cerebellum (CB). We confirmed that a disintegrin and metalloproteinase with thrombospondin motifs 4 (ADAMTS4) and matrixmetalloproteinase 3 (MMP-3) proteases digestion led to an intense double band with similar molecular weight to that described as SPARC-like fragment (SLF). Finally, hevin immunoreactivity was also detected in human astrocytoma, meningioma, cerebrospinal fluid and serum samples, but was absent from any blood cell type.
Collapse
|
6
|
Chen S, Zou Q, Guo Q, Chen Y, Kuang X, Zhang Y, Liu Y, Wu W, Li G, Tu L, Tong J, Li S, Ma L, Li Q. SPARC Knockdown Reduces Glutamate-Induced HT22 Hippocampal Nerve Cell Damage by Regulating Autophagy. Front Neurosci 2021; 14:581441. [PMID: 33584170 PMCID: PMC7874057 DOI: 10.3389/fnins.2020.581441] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 12/07/2020] [Indexed: 12/31/2022] Open
Abstract
Secreted protein acidic and rich in cysteine (SPARC) is a matricellular protein involved in the extracellular matrix and interactions between cells during neural development of the central nervous system (CNS). Oxidative glutamate toxicity is involved in CNS diseases, including epilepsy, Alzheimer’s disease, and ischemic stroke. However, the molecular mechanism of nerve injury is not fully understood in CNS diseases. Herein, the glutamate-induced nerve damage model was used to explore the molecular mechanisms affecting nerve damage. The levels of SPARC and autophagy were increased in glutamate-induced HT22 hippocampal nerve injury. In summary, the current study confirmed that SPARC regulates autophagy in HT22 hippocampal nerve cells, and its knockdown reduces the glutamate-induced HT22 hippocampal nerve injury by inhibiting autophagy. These findings suggested that SPARC plays a crucial role in nerve injury of CNS diseases.
Collapse
Affiliation(s)
- Shuang Chen
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Qin Zou
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Qiang Guo
- Epilepsy Center, Guangdong Sanjiu Brain Hospital, Guangzhou, China
| | - Yongmin Chen
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Xi Kuang
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Hainan Health Vocational College, Haikou, China
| | - Yukang Zhang
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Yan Liu
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Wengang Wu
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Ge Li
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Linzhi Tu
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Jingyi Tong
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Songrong Li
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Lin Ma
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Qifu Li
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| |
Collapse
|
7
|
Chen S, Zou Q, Chen Y, Kuang X, Wu W, Guo M, Cai Y, Li Q. Regulation of SPARC family proteins in disorders of the central nervous system. Brain Res Bull 2020; 163:178-189. [DOI: 10.1016/j.brainresbull.2020.05.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/29/2020] [Accepted: 05/05/2020] [Indexed: 12/14/2022]
|
8
|
Heat Shock Proteins Accelerate the Maturation of Brain Endothelial Cell Glucocorticoid Receptor in Focal Human Drug-Resistant Epilepsy. Mol Neurobiol 2020; 57:4511-4529. [PMID: 32748370 DOI: 10.1007/s12035-020-02043-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 07/24/2020] [Indexed: 12/21/2022]
Abstract
Pharmacoresistance in epilepsy is a major challenge to successful clinical therapy. Glucocorticoid receptor (GR) dysregulation can affect the underlying disease pathogenesis. We recently reported that local drug biotransformation at the blood-brain barrier is upregulated by GR, which controls drug-metabolizing enzymes (e.g., cytochrome P450s, CYPs) and efflux drug transporters (MDR1) in human epileptic brain endothelial cells (EPI-ECs). Here, we establish that this mechanism is influenced upstream by GR and its association with heat shock proteins/co-chaperones (Hsps) during maturation, which differentially affect human epileptic (EPI) tissue and brain endothelial cells. Overexpressed GR, Hsp90, Hsp70, and Hsp40 were found in EPI vs. NON-EPI brain regions. Elevated neurovascular GR expression and co-localization with Hsps was evident in the EPI regions with cortical dysplasia, predominantly in the brain micro-capillaries and neurons. A corresponding increase in ATPase activity (*p < 0.05) was found in the EPI regions. The GR-Hsp90/Hsp70 binding patterns indicated a faster chaperone-promoted maturation of GR, leading to its overactivation in both the tissue and EPI-ECs derived from EPI/focal regions and GR silencing in EPI-ECs slowed such GR-Hsp interactions. Significantly accelerated GR nuclear translocation was determined in EPI-ECs following treatment with GR modulators/ligands dexamethasone, rifampicin, or phenytoin. Our findings reveal that overexpressed GR co-localizes with Hsps in the neurovasculature of EPI brain, increased GR maturation by Hsps accelerates EPI GR machinery, and furthermore this change in EPI and NON-EPI GR-Hsp interaction alters with the age of seizure onset in epileptic patients, together affecting the pathophysiology and drug regulation in the epileptic brain endothelium.
Collapse
|
9
|
Wang Z, Tao Y, Song C, Liu P, Wang C, Li Y, Cui W, Xie K, Zhang L, Wang G. Spinal hevin mediates membrane trafficking of GluA1-containing AMPA receptors in remifentanil-induced postoperative hyperalgesia in mice. Neurosci Lett 2020; 722:134855. [PMID: 32088196 DOI: 10.1016/j.neulet.2020.134855] [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: 12/17/2019] [Revised: 02/17/2020] [Accepted: 02/19/2020] [Indexed: 10/25/2022]
Abstract
INTRODUCTION Hevin, a matricellular protein involved in tissue repair and remodeling, is crucial for initiation and development of excitatory synapses. Besides, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPA) is an ionotropic transmembrane receptor for glutamate that mediates fast synaptic transmission in the central nervous system (CNS). This study aimed to investigate the correlation between spinal Hevin and AMPA receptors in remifentanil-induced postoperative hyperalgesia in mice. METHODS Remifentanil (1.33 μg/kg/min for 60 min) was subcutaneously injected into a mouse model of postoperative pain. The von Frey and hot plate tests were performed to assess mechanical and thermal hyperalgesia. The gene and protein expression of Hevin and the membrane trafficking of GluA1-containing AMPA receptors in the dorsal horn of spinal cord were detected by quantitative reverse transcription polymerase chain reaction (RT-qPCR) and Western blot analysis. In addition, Hevin-shRNA, exogenous Hevin, and 1-naphtylacetyl-spermine (NASPM) were administrated intrathecally to assess the relationship between spinal Hevin and AMPA receptors. RESULTS Perioperative administration of remifentanil can aggravate and prolong incision-induced mechanical and thermal hyperalgesia. Treatment with remifentanil increased the expression of spinal Hevin and the membrane trafficking of AMPA receptors. Additionally, knockdown of spinal Hevin attenuated pain hypersensitivity and downregulated membrane trafficking of AMPA receptors after treatment with remifentanil. Meanwhile, preadministration of NASPM reversed spontaneous pain and membrane trafficking of spinal GluA1-containing AMPA receptors induced by exogenous Hevin in naïve mice. CONCLUSIONS Spinal Hevin was involved in the maintenance of remifentanil-induced postoperative hyperalgesia via modulating membrane trafficking of AMPA receptors.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China
| | - Yuzhu Tao
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China
| | - Chengcheng Song
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China
| | - Peng Liu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Chunyan Wang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China
| | - Yize Li
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China
| | - Wei Cui
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China
| | - Keliang Xie
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China
| | - Linlin Zhang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China.
| | - Guolin Wang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China.
| |
Collapse
|
10
|
Cartography of hevin-expressing cells in the adult brain reveals prominent expression in astrocytes and parvalbumin neurons. Brain Struct Funct 2019; 224:1219-1244. [PMID: 30656447 DOI: 10.1007/s00429-019-01831-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 01/08/2019] [Indexed: 02/03/2023]
Abstract
Hevin, also known as SPARC-like 1, is a member of the secreted protein acidic and rich in cysteine family of matricellular proteins, which has been implicated in neuronal migration and synaptogenesis during development. Unlike previously characterized matricellular proteins, hevin remains strongly expressed in the adult brain in both astrocytes and neurons, but its precise pattern of expression is unknown. The present study provides the first systematic description of hevin mRNA distribution in the adult mouse brain. Using isotopic in situ hybridization, we showed that hevin is strongly expressed in the cortex, hippocampus, basal ganglia complex, diverse thalamic nuclei and brainstem motor nuclei. To identify the cellular phenotype of hevin-expressing cells, we used double fluorescent in situ hybridization in mouse and human adult brains. In the mouse, hevin mRNA was found in the majority of astrocytes but also in specific neuronal populations. Hevin was expressed in almost all parvalbumin-positive projection neurons and local interneurons. In addition, hevin mRNA was found in: (1) subsets of other inhibitory GABAergic neuronal subtypes, including calbindin, cholecystokinin, neuropeptide Y, and somatostatin-positive neurons; (2) subsets of glutamatergic neurons, identified by the expression of the vesicular glutamate transporters VGLUT1 and VGLUT2; and (3) the majority of cholinergic neurons from motor nuclei. Hevin mRNA was absent from all monoaminergic neurons and cholinergic neurons of the ascending pathway. A similar cellular profile of expression was observed in human, with expression of hevin in parvalbumin interneurons and astrocytes in the cortex and caudate nucleus as well as in cortical glutamatergic neurons. Furthermore, hevin transcript was enriched in ribosomes of astrocytes and parvalbumin neurons providing a direct evidence of hevin mRNAs translation in these cell types. This study reveals the unique and complex expression profile of the matricellular protein hevin in the adult brain. This distribution is compatible with a role of hevin in astrocytic-mediated adult synaptic plasticity and in the regulation of network activity mediated by parvalbumin-expressing neurons.
Collapse
|
11
|
Bridel C, Koel-Simmelink MJA, Peferoen L, Derada Troletti C, Durieux S, Gorter R, Nutma E, Gami P, Iacobaeus E, Brundin L, Kuhle J, Vrenken H, Killestein J, Piersma SR, Pham TV, De Vries HE, Amor S, Jimenez CR, Teunissen CE. Brain endothelial cell expression of SPARCL-1 is specific to chronic multiple sclerosis lesions and is regulated by inflammatory mediators in vitro. Neuropathol Appl Neurobiol 2017; 44:404-416. [PMID: 28543098 DOI: 10.1111/nan.12412] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 05/18/2017] [Accepted: 05/24/2017] [Indexed: 02/05/2023]
Abstract
AIMS Cell matrix modulating protein SPARCL-1 is highly expressed by astrocytes during CNS development and following acute CNS damage. Applying NanoLC-MS/MS to CSF of RRMS and SPMS patients, we identified SPARCL-1 as differentially expressed between these two stages of MS, suggesting a potential as CSF biomarker to differentiate RRMS from SPMS and a role in MS pathogenesis. METHODS This study examines the potential of SPARCL-1 as CSF biomarker discriminating RRMS from SPMS in three independent cohorts (n = 249), analyses its expression pattern in MS lesions (n = 26), and studies its regulation in cultured human brain microvasculature endothelial cells (BEC) after exposure to MS-relevant inflammatory mediators. RESULTS SPARCL-1 expression in CSF was significantly higher in SPMS compared to RRMS in a Dutch cohort of 76 patients. This finding was not replicated in 2 additional cohorts of MS patients from Sweden (n = 81) and Switzerland (n = 92). In chronic MS lesions, but not active lesions or NAWM, a vessel expression pattern of SPARCL-1 was observed in addition to the expression by astrocytes. EC were found to express SPARCL-1 in chronic MS lesions, and SPARCL-1 expression was regulated by MS-relevant inflammatory mediators in cultured human BEC. CONCLUSIONS Conflicting results of SPARCL-1's differential expression in CSF of three independent cohorts of RRMS and SPMS patients precludes its use as biomarker for disease progression. The expression of SPARCL-1 by BEC in chronic MS lesions together with its regulation by inflammatory mediators in vitro suggest a role for SPARCL-1 in MS neuropathology, possibly at the brain vascular level.
Collapse
Affiliation(s)
- C Bridel
- Department of Clinical Chemistry, Neurochemistry Lab and Biobank, VU Medical Centre, Amsterdam, The Netherlands
| | - M J A Koel-Simmelink
- Department of Clinical Chemistry, Neurochemistry Lab and Biobank, VU Medical Centre, Amsterdam, The Netherlands
| | - L Peferoen
- Department of Pathology, VU Medical Centre, Amsterdam, The Netherlands
| | - C Derada Troletti
- Department of Molecular Cell Biology and Immunology, Neuroscience Campus Amsterdam, VU University Medical Centre, Amsterdam, The Netherlands
| | - S Durieux
- Department of Clinical Chemistry, Neurochemistry Lab and Biobank, VU Medical Centre, Amsterdam, The Netherlands
| | - R Gorter
- Department of Pathology, VU Medical Centre, Amsterdam, The Netherlands
| | - E Nutma
- Department of Pathology, VU Medical Centre, Amsterdam, The Netherlands
| | - P Gami
- Department of Pathology, VU Medical Centre, Amsterdam, The Netherlands
| | - E Iacobaeus
- Department of Clinical Neuroscience, Neuroimmunology Unit, Karolinska Institute, Solna, Sweden.,Center for Molecular Medicine, Stockholm, Sweden
| | - L Brundin
- Department of Clinical Neuroscience, Neuroimmunology Unit, Karolinska Institute, Solna, Sweden.,Center for Molecular Medicine, Stockholm, Sweden
| | - J Kuhle
- Neurology, Department of Medicine, Biomedicine and Clinical Research, University Hospital Basel, Basel, Switzerland
| | - H Vrenken
- Department of Radiology and Nuclear Medicine and Department of Physics and Medical Technology, VU University Medical Center, Amsterdam, The Netherlands
| | - J Killestein
- Department of Neurology, MS Centre Amsterdam, VU Medical Centre, Amsterdam, The Netherlands
| | - S R Piersma
- Department of Medical Oncology, OncoProteomics Laboratory, VU Medical Centre, Amsterdam, The Netherlands
| | - T V Pham
- Department of Medical Oncology, OncoProteomics Laboratory, VU Medical Centre, Amsterdam, The Netherlands
| | - H E De Vries
- Department of Molecular Cell Biology and Immunology, Neuroscience Campus Amsterdam, VU University Medical Centre, Amsterdam, The Netherlands
| | - S Amor
- Department of Pathology, VU Medical Centre, Amsterdam, The Netherlands.,Queen Mary University of London, Blizard Institute, Barts and The London School of Medicine and Dentistry, London, UK
| | - C R Jimenez
- Department of Medical Oncology, OncoProteomics Laboratory, VU Medical Centre, Amsterdam, The Netherlands
| | - C E Teunissen
- Department of Clinical Chemistry, Neurochemistry Lab and Biobank, VU Medical Centre, Amsterdam, The Netherlands
| |
Collapse
|
12
|
Kim JE, Hyun HW, Min SJ, Kang TC. Sustained HSP25 Expression Induces Clasmatodendrosis via ER Stress in the Rat Hippocampus. Front Cell Neurosci 2017; 11:47. [PMID: 28275338 PMCID: PMC5319974 DOI: 10.3389/fncel.2017.00047] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 02/13/2017] [Indexed: 12/16/2022] Open
Abstract
Heat shock protein (HSP) 25 (murine/rodent 25 kDa, human 27 kDa) is one of the major astroglial HSP families, which has a potent anti-apoptotic factor contributing to a higher resistance of astrocytes to the stressful condition. However, impaired removals of HSP25 decrease astroglial viability. In the present study, we investigated whether HSP25 is involved in astroglial apoptosis or clasmatodendrosis (autophagic astroglial death) in the rat hippocampus induced by status epilepticus (SE). Following SE, HSP25 expression was transiently increased in astrocytes within the dentate gyrus (DG), while it was sustained in CA1 astrocytes until 4 weeks after SE. HSP25 knockdown exacerbated SE-induced apoptotic astroglial degeneration, but mitigated clasmatodendrosis accompanied by abrogation of endoplasmic reticulum (ER) stress without changed seizure susceptibility or severity. These findings suggest that sustained HSP25 induction itself may result in clasmatodendrosis via prolonged ER stress. To the best of our knowledge, the present study demonstrates for the first time the double-edge properties of HSP25 in astroglial death induced by SE.
Collapse
Affiliation(s)
- Ji-Eun Kim
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University Chuncheon, South Korea
| | - Hye-Won Hyun
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University Chuncheon, South Korea
| | - Su-Ji Min
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University Chuncheon, South Korea
| | - Tae-Cheon Kang
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University Chuncheon, South Korea
| |
Collapse
|
13
|
Miller SJ, Zhang PW, Glatzer J, Rothstein JD. Astroglial transcriptome dysregulation in early disease of an ALS mutant SOD1 mouse model. J Neurogenet 2016; 31:37-48. [DOI: 10.1080/01677063.2016.1260128] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Sean J. Miller
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Brain Science Institute, Johns Hopkins University, Baltimore, MD, USA
- Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ping-wu Zhang
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Brain Science Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Jenna Glatzer
- The Brain Science Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Jeffrey D. Rothstein
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Brain Science Institute, Johns Hopkins University, Baltimore, MD, USA
- Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
14
|
Kim SY, Porter BE, Friedman A, Kaufer D. A potential role for glia-derived extracellular matrix remodeling in postinjury epilepsy. J Neurosci Res 2016; 94:794-803. [PMID: 27265805 DOI: 10.1002/jnr.23758] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 04/07/2016] [Accepted: 04/07/2016] [Indexed: 01/04/2023]
Abstract
Head trauma and vascular injuries are known risk factors for acquired epilepsy. The sequence of events that lead from the initial injury to the development of epilepsy involves complex plastic changes and circuit rewiring. In-depth, comprehensive understanding of the epileptogenic process is critical for the identification of disease-modifying targets. Here we review the complex interactions of cellular and extracellular components that may promote epileptogenesis, with an emphasis on the role of astrocytes. Emerging evidence demonstrates that astrocytes promptly respond to brain damage and play a critical role in the development of postinjury epilepsy. Astrocytes have been shown to regulate extracellular matrix (ECM) remodeling, which can affect plasticity and stability of synapses and, in turn, contribute to the epileptogenic process. From these separate lines of evidence, we present a hypothesis suggesting a possible role for astrocyte-regulated remodeling of ECM and perineuronal nets, a specialized ECM structure around fast-spiking inhibitory interneurons, in the development and progression of posttraumatic epilepsies. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Soo Young Kim
- Department of Integrative Biology, University of California, Berkeley, Berkeley, California.,Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California
| | - Brenda E Porter
- Department of Neurology, Stanford University School of Medicine, Palo Alto, California
| | - Alon Friedman
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Daniela Kaufer
- Department of Integrative Biology, University of California, Berkeley, Berkeley, California.,Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California.,Canadian Institute for Advanced Research Program in Child and Brain Development, Toronto, Ontario, Canada
| |
Collapse
|
15
|
Parseghian MH, Hobson ST, Richieri RA. Targeted heat shock protein 72 for pulmonary cytoprotection. Ann N Y Acad Sci 2016; 1374:78-85. [PMID: 27152638 DOI: 10.1111/nyas.13059] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 02/29/2016] [Accepted: 03/09/2016] [Indexed: 12/12/2022]
Abstract
Heat shock protein 72 (HSP72) is perhaps the most important member of the HSP70 family of proteins, given that it is induced in a wide variety of tissues and cells to combat stress, particularly oxidative stress. Here, we review independent observations of the critical role this protein plays as a pulmonary cytoprotectant and discuss the merits of developing HSP72 as a therapeutic for rapid delivery to cells and tissues after a traumatic event. We also discuss the fusion of HSP72 to a cell-penetrating single-chain Fv antibody fragment derived from mAb 3E10, referred to as Fv-HSP70. This fusion construct has been validated in vivo in a cerebral infarction model and is currently in testing as a clinical therapeutic to treat ischemic events and as a fieldable medical countermeasure to treat inhalation of toxicants caused by terrorist actions or industrial accidents.
Collapse
Affiliation(s)
| | - Stephen T Hobson
- Rubicon Biotechnology, Lake Forest, California.,Seacoast Science, Inc, Carlsbad, California
| | | |
Collapse
|
16
|
Blakely PK, Hussain S, Carlin LE, Irani DN. Astrocyte matricellular proteins that control excitatory synaptogenesis are regulated by inflammatory cytokines and correlate with paralysis severity during experimental autoimmune encephalomyelitis. Front Neurosci 2015; 9:344. [PMID: 26500475 PMCID: PMC4598482 DOI: 10.3389/fnins.2015.00344] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 09/14/2015] [Indexed: 11/29/2022] Open
Abstract
The matricellular proteins, secreted protein acidic and rich in cysteine (SPARC) and SPARC-like 1 (SPARCL1), are produced by astrocytes and control excitatory synaptogenesis in the central nervous system. While SPARCL1 directly promotes excitatory synapse formation in vitro and in the developing nervous system in vivo, SPARC specifically antagonizes the synaptogenic actions of SPARCL1. We hypothesized these proteins also help maintain existing excitatory synapses in adult hosts, and that local inflammation in the spinal cord alters their production in a way that dynamically modulates motor synapses and impacts the severity of paralysis during experimental autoimmune encephalomyelitis (EAE) in mice. Using a spontaneously remitting EAE model, paralysis severity correlated inversely with both expression of synaptic proteins and the number of synapses in direct contact with the perikarya of motor neurons in spinal gray matter. In both remitting and non-remitting EAE models, paralysis severity also correlated inversely with sparcl1:sparc transcript and SPARCL1:SPARC protein ratios directly in lumbar spinal cord tissue. In vitro, astrocyte production of both SPARCL1 and SPARC was regulated by T cell-derived cytokines, causing dynamic modulation of the SPARCL1:SPARC expression ratio. Taken together, these data support a model whereby proinflammatory cytokines inhibit SPARCL1 and/or augment SPARC expression by astrocytes in spinal gray matter that, in turn, cause either transient or sustained synaptic retraction from lumbar spinal motor neurons thereby regulating hind limb paralysis during EAE. Ongoing studies seek ways to alter this SPARCL1:SPARC expression ratio in favor of synapse reformation/maintenance and thus help to modulate neurologic deficits during times of inflammation. This could identify new astrocyte-targeted therapies for diseases such as multiple sclerosis.
Collapse
Affiliation(s)
- Pennelope K Blakely
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology, University of Michigan Medical School Ann Arbor, MI, USA
| | - Shabbir Hussain
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology, University of Michigan Medical School Ann Arbor, MI, USA
| | - Lindsey E Carlin
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology, University of Michigan Medical School Ann Arbor, MI, USA
| | - David N Irani
- Holtom-Garrett Program in Neuroimmunology, Department of Neurology, University of Michigan Medical School Ann Arbor, MI, USA
| |
Collapse
|
17
|
Kandratavicius L, Hallak JE, Carlotti CG, Assirati JA, Leite JP. Hippocampal expression of heat shock proteins in mesial temporal lobe epilepsy with psychiatric comorbidities and their relation to seizure outcome. Epilepsia 2014; 55:1834-43. [DOI: 10.1111/epi.12787] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2014] [Indexed: 01/03/2023]
Affiliation(s)
- Ludmyla Kandratavicius
- Department of Neurosciences and Behavior; Ribeirao Preto School of Medicine; University of Sao Paulo (USP); Ribeirao Preto Brazil
- Center for Interdisciplinary Research on Applied Neurosciences (NAPNA); University of Sao Paulo (USP); Ribeirao Preto Brazil
| | - Jaime E. Hallak
- Department of Neurosciences and Behavior; Ribeirao Preto School of Medicine; University of Sao Paulo (USP); Ribeirao Preto Brazil
- Center for Interdisciplinary Research on Applied Neurosciences (NAPNA); University of Sao Paulo (USP); Ribeirao Preto Brazil
- National Institute of Science and Technology in Translational Medicine (INCT-TM - CNPq); Ribeirao Preto Brazil
| | - Carlos G. Carlotti
- Department of Surgery; Ribeirao Preto School of Medicine; University of Sao Paulo (USP); Ribeirao Preto Brazil
| | - Joao A. Assirati
- Department of Surgery; Ribeirao Preto School of Medicine; University of Sao Paulo (USP); Ribeirao Preto Brazil
| | - Joao P. Leite
- Department of Neurosciences and Behavior; Ribeirao Preto School of Medicine; University of Sao Paulo (USP); Ribeirao Preto Brazil
- Center for Interdisciplinary Research on Applied Neurosciences (NAPNA); University of Sao Paulo (USP); Ribeirao Preto Brazil
| |
Collapse
|
18
|
Chang CC, Chen SD, Lin TK, Chang WN, Liou CW, Chang AYW, Chan SHH, Chuang YC. Heat shock protein 70 protects against seizure-induced neuronal cell death in the hippocampus following experimental status epilepticus via inhibition of nuclear factor-κB activation-induced nitric oxide synthase II expression. Neurobiol Dis 2013; 62:241-9. [PMID: 24141017 DOI: 10.1016/j.nbd.2013.10.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 09/17/2013] [Accepted: 10/07/2013] [Indexed: 10/26/2022] Open
Abstract
Status epilepticus induces subcellular changes that may eventually lead to neuronal cell death in the hippocampus. Based on an animal model of status epilepticus, our laboratory showed previously that sustained hippocampal seizure activity activates nuclear factor-κB (NF-κB) and upregulates nitric oxide synthase (NOS) II gene expression, leading to apoptotic neuronal cell death in the hippocampus. The present study examined the potential modulatory role of heat shock protein 70 (HSP70) on NF-κB signaling in the hippocampus following experimental status epilepticus. In Sprague-Dawley rats, kainic acid (KA) was microinjected unilaterally into the hippocampal CA3 subfield to induce prolonged bilateral seizure activity. Expression of HSP70 was elevated as early as 1h after the elicitation of sustained seizure activity, followed by a progressive elevation that peaked at 24h. Pretreatment with an antisense oligonucleotide against hsp70 decreased the HSP70 expression, and significantly augmented IκB kinase (IKK) activity and phosphorylation of IκBα, alongside enhanced nuclear translocation and DNA binding activity of NF-κB in the hippocampal CA3 neurons and glial cells. These cellular events were followed by enhanced upregulation of NOS II and peroxynitrite expression 3h after sustained seizure activity that led to an increase of caspase-3 and DNA fragmentation in the hippocampal CA3 neurons 7days after experimental status epilepticus. We concluded that HSP70 protects against apoptotic cell death induced by NF-κB activation and NOS II-peroxynitrite signaling cascade in the hippocampal CA3 and glial cells following experimental status epilepticus via suppression of IKK activity and deactivation of IκBα.
Collapse
Affiliation(s)
- Chiung-Chih Chang
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; Chang Gung University College of Medicine, Kaohsiung, Taiwan; Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Shang-Der Chen
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; Chang Gung University College of Medicine, Kaohsiung, Taiwan; Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Tsu-Kung Lin
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Wen-Neng Chang
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chia-Wei Liou
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Alice Y W Chang
- Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Samuel H H Chan
- Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Yao-Chung Chuang
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; Chang Gung University College of Medicine, Kaohsiung, Taiwan; Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; Department of Biological Science, National Sun Yet-sen University, Kaohsiung, Taiwan.
| |
Collapse
|
19
|
Kim YJ, Kim JY, Ko AR, Kang TC. Reduction in heat shock protein 90 correlates to neuronal vulnerability in the rat piriform cortex following status epilepticus. Neuroscience 2013; 255:265-77. [PMID: 24096135 DOI: 10.1016/j.neuroscience.2013.09.050] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Revised: 09/24/2013] [Accepted: 09/25/2013] [Indexed: 12/20/2022]
Abstract
In the present study, we addressed the question of whether the distinct patterns of heat shock protein (HSP) 70 and HSP90 expressions in the brain region represents the regional specific responses to status epilepsticus (SE) in an effort to better understand the role of HSPs in epileptogenic insult. HSP70 immunoreactivity was increased in CA3 pyramidal cells as well as dentate granule cells at 12h-1week after SE. HSP70 immunoreactivity was transiently increased in neurons within the piriform cortex (PC) following SE. Linear regression analysis showed no correlation between the intensity of NeuN and that of HSP70. In contrast to HSP70, HSP90 immunoreactivity was decreased in CA1-3 pyramidal cells at 4days-4weeks after SE. In addition, HSP90 immunoreactivity was decreased in PC neurons at 12h-4weeks after SE. linear regression analysis showed a direct proportional relationship between the intensity of NeuN and that of HSP90. Therefore, these findings suggest that HSP90 degradation may be closely related to neuronal vulnerability to SE insult.
Collapse
Affiliation(s)
- Y-J Kim
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chunchon 200-702, South Korea; Institute of Epilepsy Research, College of Medicine, Hallym University, Chunchon 200-702, South Korea
| | | | | | | |
Collapse
|
20
|
Hashimoto R, Voigt B, Ishimaru Y, Hokao R, Chiba S, Serikawa T, Sasa M, Kuramoto T. Identification of QTLs involved in the development of amygdala kindling in the rat. Exp Anim 2013; 62:181-7. [PMID: 23903052 PMCID: PMC4160946 DOI: 10.1538/expanim.62.181] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Amygdala kindling is useful for modeling human epilepsy development. It has been known that genetic factors are involved in the development of amygdala kindling. The purpose of this study was to identify the loci that are responsible for the development of amygdala kindling. To achieve this, rat strains from a LEXF/FXLE recombinant inbred (RI) strain panel were used. The phenotypes of amygdala kindling-related parameters for seven RI strains and parental LE/Stm and F344/Stm strains were determined. They included the afterdischarge threshold (ADT), the afterdischarge duration (ADD), and the kindling rate, an incidence of development of kindling. Quantitative trait loci (QTL) analysis was performed to identify linkage relationships between these phenotypes and 1,033 SNP markers. Although no significant differences in pre-kindling ADT and ADD were observed, a significant difference in the kindling rate was found for the LEXF/FXLE RI strain. Two QTLs for the amygdala kindling rate (Agkr1 and Agkr2) were identified on rat chromosome 2. These findings clearly prove the existence of genetic influences that are involved in kindling development and suggest that substantial genetic components contribute to the progression of partial seizures into generalized seizures.
Collapse
Affiliation(s)
- Ryoko Hashimoto
- Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Yoshidakonoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Lloyd-Burton S, Roskams AJ. SPARC-like 1 (SC1) is a diversely expressed and developmentally regulated matricellular protein that does not compensate for the absence of SPARC in the CNS. J Comp Neurol 2013; 520:2575-90. [PMID: 22173850 DOI: 10.1002/cne.23029] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
SPARC-like 1 (SC1) is a member of the SPARC family of matricellular proteins that has been implicated in the regulation of processes such as cell migration, proliferation, and differentiation. Here we show that SC1 exhibits remarkably diverse and dynamic expression in the developing and adult nervous system. During development, SC1 localizes to radial glia and pial-derived structures, including the vasculature, choroid plexus, and pial membranes. SC1 is not downregulated in postnatal development, but its expression shifts to distinct time windows in subtypes of glia and neurons, including astrocytes, large projection neurons, Bergmann glia, Schwann cells, and ganglionic satellite cells. In addition, SC1 expression levels and patterns are not altered in the SPARC null mouse, suggesting that SC1 does not compensate for the absence of SPARC. We conclude that SC1 and SPARC may share significant homology, but are likely to have distinct but complementary roles in nervous system development.
Collapse
Affiliation(s)
- Samantha Lloyd-Burton
- Department of Zoology, Life Sciences Institute and Brain Research Centre, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | | |
Collapse
|
22
|
Lee C, Hu J, Ralls S, Kitamura T, Loh YP, Yang Y, Mukouyama YS, Ahn S. The molecular profiles of neural stem cell niche in the adult subventricular zone. PLoS One 2012; 7:e50501. [PMID: 23209762 PMCID: PMC3510163 DOI: 10.1371/journal.pone.0050501] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Accepted: 10/23/2012] [Indexed: 11/19/2022] Open
Abstract
Neural stem cells (NSCs) reside in a unique microenvironment called the neurogenic niche and generate functional new neurons. The neurogenic niche contains several distinct types of cells and interacts with the NSCs in the subventricular zone (SVZ) of the lateral ventricle. While several molecules produced by the niche cells have been identified to regulate adult neurogenesis, a systematic profiling of autocrine/paracrine signaling molecules in the neurogenic regions involved in maintenance, self-renewal, proliferation, and differentiation of NSCs has not been done. We took advantage of the genetic inducible fate mapping system (GIFM) and transgenic mice to isolate the SVZ niche cells including NSCs, transit-amplifying progenitors (TAPs), astrocytes, ependymal cells, and vascular endothelial cells. From the isolated cells and microdissected choroid plexus, we obtained the secretory molecule expression profiling (SMEP) of each cell type using the Signal Sequence Trap method. We identified a total of 151 genes encoding secretory or membrane proteins. In addition, we obtained the potential SMEP of NSCs using cDNA microarray technology. Through the combination of multiple screening approaches, we identified a number of candidate genes with a potential relevance for regulating the NSC behaviors, which provide new insight into the nature of neurogenic niche signals.
Collapse
Affiliation(s)
- Cheol Lee
- Program in Genomics of Differentiation, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jingqiong Hu
- Laboratory of Stem Cell and Neuro-Vascular Biology, Genetics and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Sherry Ralls
- Program in Genomics of Differentiation, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Toshio Kitamura
- Division of Cellular Therapy, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Y. Peng Loh
- Section on Cellular Neurobiology, Program on Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Yanqin Yang
- DNA Sequencing and Genomics Core Facility, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Yoh-suke Mukouyama
- Laboratory of Stem Cell and Neuro-Vascular Biology, Genetics and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail: (YM); (SA)
| | - Sohyun Ahn
- Program in Genomics of Differentiation, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail: (YM); (SA)
| |
Collapse
|
23
|
Rajasekaran K, Todorovic M, Kapur J. Calcium-permeable AMPA receptors are expressed in a rodent model of status epilepticus. Ann Neurol 2012; 72:91-102. [PMID: 22829271 DOI: 10.1002/ana.23570] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVE A study was undertaken to characterize the plasticity of AMPA receptor (AMPAR)-mediated neurotransmission in the hippocampus during status epilepticus (SE). METHODS SE was induced by pilocarpine, and animals were studied 10 minutes (refractory SE) or 60 minutes (late SE) after the onset of the first grade 5 seizures. AMPAR-mediated currents were recorded from CA1 pyramidal neurons and dentate granule cells (DGCs) by voltage clamp technique. The surface expression of GluA2 subunit on hippocampal membranes was determined using a biotinylation assay. GluA2 internalization and changes in intracellular calcium ([Ca](i)) levels were studied in hippocampal cultures using immunocytochemical and live-imaging techniques. AMPAR antagonist treatment of SE was evaluated by video and electroencephalography. RESULTS AMPAR-mediated currents recorded from CA1 neurons from refractory and late SE animals were inwardly rectifying, and philanthotoxin-sensitive; similar changes were observed in recordings obtained from DGCs from refractory SE animals. GluA2 subunit surface expression was reduced in the hippocampus during refractory and late SE. In cultured hippocampal pyramidal neurons, recurrent bursting diminished surface expression of the GluA2 subunit and enhanced its internalization rate. Recurrent bursting-induced increase in [Ca](i) levels was reduced by selective inhibition of GluA2-lacking AMPARs. GYKI-52466 terminated diazepam-refractory SE. INTERPRETATION During SE, there is rapid, ongoing plasticity of AMPARs with the expression of GluA2-lacking AMPARs. These receptors provide another source of Ca(2+) entry into the principal neurons. Benzodiazepam-refractory SE can be terminated by AMPAR antagonism. The data identify AMPARs as a potential therapeutic target for the treatment of SE.
Collapse
|
24
|
SC1/hevin identifies early white matter injury after ischemia and intracerebral hemorrhage in young and aged rats. J Neuropathol Exp Neurol 2012; 71:480-93. [PMID: 22588386 DOI: 10.1097/nen.0b013e318256901c] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The progression of white matter damage after ischemic and hemorrhagic strokes can exacerbate the initial injury, but little is known about the processes involved. We show that the antiadhesive matricellular glycoprotein SC1 is a novel early marker of white matter damage in 3 models of acute injury in the rat striatum: transient focal ischemia, intracerebral hemorrhage, and a needle penetration wound. SC1 was restricted to the damaged portions of axon bundles that bordered stroke lesions in young-adult and aged rats. SC1 peaked at 1 and 3 days after intracerebral hemorrhage and at 7 days after ischemia. The SC1-positive bundles usually expressed degraded myelin basic protein and amyloid precursor protein, a marker of axonal injury. At the hematoma edge, SC1 was seen in a few axon bundles that retained myelin basic protein staining. In these bundles, punctate SC1 staining filled individual axons, extended beyond a core of pan-axonal neurofilament and NF200 and was inside or overlapped with myelin basic protein staining when it was present. Aged rats had less SC1 (and amyloid precursor protein) after both types of stroke, suggesting a reduced axonal response. SC1 also labeled amyloid precursor protein-positive axon bundles along the needle penetration tract of saline-injected rats; thus, SC1 appears to characterize damaged striatal white matter damage after multiple types of injury.
Collapse
|
25
|
Lopes MW, Soares FMS, de Mello N, Nunes JC, de Cordova FM, Walz R, Leal RB. Time-Dependent Modulation of Mitogen Activated Protein Kinases and AKT in Rat Hippocampus and Cortex in the Pilocarpine Model of Epilepsy. Neurochem Res 2012; 37:1868-78. [DOI: 10.1007/s11064-012-0797-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Revised: 04/11/2012] [Accepted: 05/07/2012] [Indexed: 01/14/2023]
|
26
|
The 27-kDa heat shock protein (HSP27) is a reliable hippocampal marker of full development of pilocarpine-induced status epilepticus. Epilepsy Res 2012; 98:35-43. [DOI: 10.1016/j.eplepsyres.2011.08.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2010] [Revised: 08/18/2011] [Accepted: 08/19/2011] [Indexed: 11/18/2022]
|
27
|
Wu X, Reddy DS. Integrins as receptor targets for neurological disorders. Pharmacol Ther 2011; 134:68-81. [PMID: 22233753 DOI: 10.1016/j.pharmthera.2011.12.008] [Citation(s) in RCA: 117] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Accepted: 12/15/2011] [Indexed: 12/18/2022]
Abstract
This review focuses on the neurobiology of integrins, pathophysiological roles of integrins in neuroplasticity and nervous system disorders, and therapeutic implications of integrins as potential drug targets and possible delivery pathways. Neuroplasticity is a central phenomenon in many neurological conditions such as seizures, trauma, and traumatic brain injury. During the course of many brain diseases, in addition to intracellular compartment changes, alterations in non-cell compartments such as extracellular matrix (ECM) are recognized as an essential process in forming and reorganizing neural connections. Integrins are heterodimeric transmembrane receptors that mediate cell-ECM and cell-cell adhesion events. Although the mechanisms of neuroplasticity remain unclear, it has been suggested that integrins undergo plasticity including clustering through interactions with ECM proteins, modulating ion channels, intracellular Ca(2+) and protein kinase signaling, and reorganization of cytoskeletal filaments. As cell surface receptors, integrins are central to the pathophysiology of many brain diseases, such as epilepsy, and are potential targets for the development of new drugs for neurological disorders.
Collapse
Affiliation(s)
- Xin Wu
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center College of Medicine, Bryan, TX 77807, USA
| | | |
Collapse
|
28
|
Distribution and Time-Course of 4-Hydroxynonenal, Heat Shock Protein 110/105 Family Members and Cyclooxygenase-2 Expression in the Hippocampus of Rat During Trimethyltin-Induced Neurodegeneration. Neurochem Res 2011; 36:1490-500. [DOI: 10.1007/s11064-011-0478-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2011] [Indexed: 10/18/2022]
|
29
|
Kharlamov EA, Lepsveridze E, Meparishvili M, Solomonia RO, Lu B, Miller ER, Kelly KM, Mtchedlishvili Z. Alterations of GABA(A) and glutamate receptor subunits and heat shock protein in rat hippocampus following traumatic brain injury and in posttraumatic epilepsy. Epilepsy Res 2011; 95:20-34. [PMID: 21439793 DOI: 10.1016/j.eplepsyres.2011.02.008] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Revised: 12/20/2010] [Accepted: 02/18/2011] [Indexed: 12/26/2022]
Abstract
Traumatic brain injury (TBI) can result in the development of posttraumatic epilepsy (PTE). Recently, we reported differential alterations in tonic and phasic GABA(A) receptor (GABA(A)R) currents in hippocampal dentate granule cells 90 days after controlled cortical impact (CCI) (Mtchedlishvili et al., 2010). In the present study, we investigated long-term changes in the protein expression of GABA(A)R α1, α4, γ2, and δ subunits, NMDA (NR2B) and AMPA (GluR1) receptor subunits, and heat shock proteins (HSP70 and HSP90) in the hippocampus of Sprague-Dawley rats evaluated by Western blotting in controls, CCI-injured animals without PTE (CCI group), and CCI-injured animals with PTE (PTE group). No differences were found among all three groups for α1 and α4 subunits. Significant reduction of γ2 protein was observed in the PTE group compared to control. CCI caused a 194% and 127% increase of δ protein in the CCI group compared to control (p<0.0001), and PTE (p<0.0001) groups, respectively. NR2B protein was increased in CCI and PTE groups compared to control (p=0.0001, and p=0.011, respectively). GluR1 protein was significantly decreased in CCI and PTE groups compared to control (p=0.003, and p=0.001, respectively), and in the PTE group compared to the CCI group (p=0.036). HSP70 was increased in CCI and PTE groups compared to control (p=0.014, and p=0.005, respectively); no changes were found in HSP90 expression. These results provide for the first time evidence of long-term alterations of GABA(A) and glutamate receptor subunits and a HSP following CCI.
Collapse
Affiliation(s)
- Elena A Kharlamov
- Center for Neuroscience Research, Allegheny-Singer Research Institute, Allegheny General Hospital, 320 East North Avenue, Pittsburgh, PA 15212-4772, United States.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Weaver MS, Workman G, Cardo-Vila M, Arap W, Pasqualini R, Sage EH. Processing of the matricellular protein hevin in mouse brain is dependent on ADAMTS4. J Biol Chem 2009; 285:5868-77. [PMID: 20018883 DOI: 10.1074/jbc.m109.070318] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The matricellular SPARC family member hevin (SPARC-like 1/SPARCL-1/SC1/Mast9) contributes to neural development and alters tumor progression in a range of mammalian models. The distribution of hevin in mouse tissues was reexamined with a novel monoclonal antibody that discriminates between hevin and its ortholog SPARC. We now report proteolysis of hevin in many tissues, with the most extensive processing in the brain. We demonstrate a cleavage site within the hevin sequence for the neural tissue proteinase ADAMTS4. Digestion of hevin by ADAMTS4 in vitro produced fragments similar to those present in brain lysates. Monoclonal antibodies revealed a SPARC-like fragment generated from hevin that was co-localized with ADAMTS4 in vivo. We show that proteolysis of hevin by ADAMTS4 in the mouse cerebellum is important for the normal development of this tissue. In conclusion, we have identified the fragmentation of hevin by ADAMTS4 in the mouse brain and propose that this specific proteolysis is integral to cell morphology and extracellular matrix deposition in the developing brain.
Collapse
Affiliation(s)
- Matt S Weaver
- Hope Heart Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington 98101-2795, USA
| | | | | | | | | | | |
Collapse
|
31
|
Eroglu C. The role of astrocyte-secreted matricellular proteins in central nervous system development and function. J Cell Commun Signal 2009; 3:167-76. [PMID: 19904629 PMCID: PMC2778595 DOI: 10.1007/s12079-009-0078-y] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2009] [Accepted: 10/01/2009] [Indexed: 11/28/2022] Open
Abstract
Matricellular proteins, such as thrombospondins (TSPs1-4), SPARC, SPARC-like1 (hevin) and tenascin C are expressed by astrocytes in the central nervous system (CNS) of rodents. The spatial and temporal expression patterns of these proteins suggest that they may be involved in important developmental processes such as cell proliferation and maturation, cell migration, axonal guidance and synapse formation. In addition, upon injury to the nervous system the expression of these proteins is upregulated, suggesting that they play a role in tissue remodeling and repair in the adult CNS. The genes encoding these proteins have been disrupted in mice. Interestingly, none of these proteins are required for survival, and furthermore, there are no evident abnormalities at the gross anatomical level in the CNS. However, detailed analyses of some of these mice in the recent years have revealed interesting CNS phenotypes. Here we will review the expression of these proteins in the CNS. We will discuss a newly described function for thrombospondins in synapse formation in the CNS in detail, and speculate whether other matricellular proteins could play similar roles in nervous system development and function.
Collapse
Affiliation(s)
- Cagla Eroglu
- Cell Biology, Duke University Medical Center, 333A Nanaline Duke Bldg., Box 3709, Durham, NC 27710 USA
| |
Collapse
|
32
|
Lively S, Brown IR. The extracellular matrix protein SC1/Hevin localizes to multivesicular bodies in Bergmann glial fibers in the adult rat cerebellum. Neurochem Res 2009; 35:315-22. [PMID: 19757034 DOI: 10.1007/s11064-009-0057-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2009] [Accepted: 08/28/2009] [Indexed: 02/06/2023]
Abstract
SC1 is an extracellular matrix molecule prominent in the mammalian brain. In the cerebellum, SC1 localizes to Bergmann glial cells and perisynaptic glial processes that envelop synapses in the molecular layer. In the present study, confocal microscopy revealed a punctate distribution of SC1 along Bergmann glial fibers that colocalized with the intermediate filament GFAP when fibers were viewed in cross-section. Immunoelectron microscopy showed that the punctate SC1 pattern corresponded to the localization of SC1 in multivesicular bodies situated within Bergmann glial fibers. The pattern of SC1 localization was not disrupted following hyperthermia or pilocarpine-induced status epilepticus. The present study suggests that SC1 protein may reach its destination in perisynaptic glial processes and glial endfeet by transport along Bergmann glial fibers in multivesicular bodies and that this process is preserved following stress.
Collapse
Affiliation(s)
- Starlee Lively
- Center for the Neurobiology of Stress, University of Toronto at Scarborough, 1265 Military Trail, Toronto, ON M1C 1A4, Canada
| | | |
Collapse
|