1
|
Hernández-Ortega K, Canul-Euan AA, Solis-Paredes JM, Borboa-Olivares H, Reyes-Muñoz E, Estrada-Gutierrez G, Camacho-Arroyo I. S100B actions on glial and neuronal cells in the developing brain: an overview. Front Neurosci 2024; 18:1425525. [PMID: 39027325 PMCID: PMC11256909 DOI: 10.3389/fnins.2024.1425525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 06/20/2024] [Indexed: 07/20/2024] Open
Abstract
The S100B is a member of the S100 family of "E" helix-loop- "F" helix structure (EF) hand calcium-binding proteins expressed in diverse glial, selected neuronal, and various peripheral cells, exerting differential effects. In particular, this review compiles descriptions of the detection of S100B in different brain cells localized in specific regions during the development of humans, mice, and rats. Then, it summarizes S100B's actions on the differentiation, growth, and maturation of glial and neuronal cells in humans and rodents. Particular emphasis is placed on S100B regulation of the differentiation and maturation of astrocytes, oligodendrocytes (OL), and the stimulation of dendritic development in serotoninergic and cerebellar neurons during embryogenesis. We also summarized reports that associate morphological alterations (impaired neurite outgrowth, neuronal migration, altered radial glial cell morphology) of specific neural cell groups during neurodevelopment and functional disturbances (slower rate of weight gain, impaired spatial learning) with changes in the expression of S100B caused by different conditions and stimuli as exposure to stress, ethanol, cocaine and congenital conditions such as Down's Syndrome. Taken together, this evidence highlights the impact of the expression and early actions of S100B in astrocytes, OL, and neurons during brain development, which is reflected in the alterations in differentiation, growth, and maturation of these cells. This allows the integration of a spatiotemporal panorama of S100B actions in glial and neuronal cells in the developing brain.
Collapse
Affiliation(s)
- Karina Hernández-Ortega
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, México City, Mexico
| | - Arturo Alejandro Canul-Euan
- Department of Developmental Neurobiology, National Institute of Perinatology Isidro Espinosa de los Reyes (INPer), Mexico City, Mexico
| | | | | | | | | | - Ignacio Camacho-Arroyo
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, México City, Mexico
| |
Collapse
|
2
|
Sharma SD, Reddy BK, Pal R, Ritakari TE, Cooper JD, Selvaraj BT, Kind PC, Chandran S, Wyllie DJA, Chattarji S. Astrocytes mediate cell non-autonomous correction of aberrant firing in human FXS neurons. Cell Rep 2023; 42:112344. [PMID: 37018073 PMCID: PMC10157295 DOI: 10.1016/j.celrep.2023.112344] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 12/12/2022] [Accepted: 03/21/2023] [Indexed: 04/06/2023] Open
Abstract
Pre-clinical studies of fragile X syndrome (FXS) have focused on neurons, with the role of glia remaining largely underexplored. We examined the astrocytic regulation of aberrant firing of FXS neurons derived from human pluripotent stem cells. Human FXS cortical neurons, co-cultured with human FXS astrocytes, fired frequent short-duration spontaneous bursts of action potentials compared with less frequent, longer-duration bursts of control neurons co-cultured with control astrocytes. Intriguingly, bursts fired by FXS neurons co-cultured with control astrocytes are indistinguishable from control neurons. Conversely, control neurons exhibit aberrant firing in the presence of FXS astrocytes. Thus, the astrocyte genotype determines the neuronal firing phenotype. Strikingly, astrocytic-conditioned medium, and not the physical presence of astrocytes, is capable of determining the firing phenotype. The mechanistic basis of this effect indicates that the astroglial-derived protein, S100β, restores normal firing by reversing the suppression of a persistent sodium current in FXS neurons.
Collapse
Affiliation(s)
- Shreya Das Sharma
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, India; University of Trans-Disciplinary Health Science and Technology, Bangalore 560064, India; Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, Bangalore 560065, India; Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor's Building, Edinburgh EH16 4SB, UK; UK Dementia Research Institute at the University of Edinburgh, Edinburgh Medical School, Chancellor's Building, Edinburgh EH16 4SB, UK
| | - Bharath Kumar Reddy
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, India; Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, Bangalore 560065, India
| | - Rakhi Pal
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, India; Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, Bangalore 560065, India
| | - Tuula E Ritakari
- Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor's Building, Edinburgh EH16 4SB, UK; UK Dementia Research Institute at the University of Edinburgh, Edinburgh Medical School, Chancellor's Building, Edinburgh EH16 4SB, UK
| | - James D Cooper
- Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor's Building, Edinburgh EH16 4SB, UK; UK Dementia Research Institute at the University of Edinburgh, Edinburgh Medical School, Chancellor's Building, Edinburgh EH16 4SB, UK
| | - Bhuvaneish T Selvaraj
- Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor's Building, Edinburgh EH16 4SB, UK; UK Dementia Research Institute at the University of Edinburgh, Edinburgh Medical School, Chancellor's Building, Edinburgh EH16 4SB, UK
| | - Peter C Kind
- Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, Bangalore 560065, India; Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK; Patrick Wild Centre, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK; Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
| | - Siddharthan Chandran
- Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, Bangalore 560065, India; Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor's Building, Edinburgh EH16 4SB, UK; UK Dementia Research Institute at the University of Edinburgh, Edinburgh Medical School, Chancellor's Building, Edinburgh EH16 4SB, UK; Patrick Wild Centre, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK; Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
| | - David J A Wyllie
- Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, Bangalore 560065, India; Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK; Patrick Wild Centre, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK; Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK.
| | - Sumantra Chattarji
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, India; Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, Bangalore 560065, India; Patrick Wild Centre, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK; Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK.
| |
Collapse
|
3
|
Rodríguez JJ, Terzieva S, Yeh CY, Gardenal E, Zallo F, Verkhratsky A, Busquets X. Neuroanatomical and morphometric study of S100β positive astrocytes in the entorhinal cortex during ageing in the 3xTg-Alzehimer's disease mouse model. Neurosci Lett 2023; 802:137167. [PMID: 36894021 DOI: 10.1016/j.neulet.2023.137167] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/03/2023] [Accepted: 03/03/2023] [Indexed: 03/09/2023]
Abstract
Astrocytes contribute to the progression of neurodegenerative diseases, including Alzheimer's disease (AD). Here, we report the neuroanatomical and morphometric analysis of astrocytes in the entorhinal cortex (EC) of the aged wild type (WT) and triple transgenic (3xTg-AD) mouse model of AD. Using 3D confocal microscopy, we determined the surface area and volume of positive astrocytic profiles in male mice (WT and 3xTg-AD) from 1 to 18 months of age. We showed that S100β-positive astrocytes were equally distributed throughout the entire EC in both animal types and showed no changes in Nv (number of cells/mm3) nor in their distribution at the different ages studied. These positive astrocytes, demonstrated an age-dependent gradual increase in their surface area and in their volume starting at 3 months of age, in both WT and 3xTg-AD mice. This last group demonstrated a large increase in both surface area and volume at 18 months of age when the burden of pathological hallmarks of AD is present (69.74% to 76.73% in the surface area and the volume, for WT and 3xTg-AD mice respectively). We observed that these changes were due to the enlargement of the cell processes and to less extend the somata. In fact, the volume of the cell body was increased by 35.82% in 18-month-old 3xTg-AD compared to WT. On the other hand, the increase on the astrocytic processes were detected as soon as 9 months of age where we found an increase of surface area and volume (36.56% and 43.73%, respectively) sustained till 18 month of age (93.6% and 113.78%, respectively) when compared age-matched non-Tg mice. Moreover, we demonstrated that these hypertrophic S100β-positive astrocytes were mainly associated with Aβ plaques. Our results show a severe atrophy in GFAP cytoskeleton in all cognitive areas; whilst within the EC astrocytes independent to this atrophy show no changes in GS and S100β; which can play a key role in the memory impairment.
Collapse
Affiliation(s)
- J J Rodríguez
- Biocruces Health Research Institute, Functional Neuroanatomy Group, IKERBASQUE, Basque Foundation for Science, Dept. of Neurosciences, Medical Faculty, University of the Basque Country (UPV/EHU), Barakaldo, Spain.
| | - S Terzieva
- Biocruces Health Research Institute, Functional Neuroanatomy Group, IKERBASQUE, Basque Foundation for Science, Dept. of Neurosciences, Medical Faculty, University of the Basque Country (UPV/EHU), Barakaldo, Spain
| | - C Y Yeh
- Biocruces Health Research Institute, Functional Neuroanatomy Group, IKERBASQUE, Basque Foundation for Science, Dept. of Neurosciences, Medical Faculty, University of the Basque Country (UPV/EHU), Barakaldo, Spain
| | - E Gardenal
- Biocruces Health Research Institute, Functional Neuroanatomy Group, IKERBASQUE, Basque Foundation for Science, Dept. of Neurosciences, Medical Faculty, University of the Basque Country (UPV/EHU), Barakaldo, Spain
| | - F Zallo
- Biocruces Health Research Institute, Functional Neuroanatomy Group, IKERBASQUE, Basque Foundation for Science, Dept. of Neurosciences, Medical Faculty, University of the Basque Country (UPV/EHU), Barakaldo, Spain
| | - A Verkhratsky
- Faculty of Life Sciences, The University of Manchester, Manchester, United Kingdom
| | - X Busquets
- Laboratory of Molecular Cell Biomedicine, Department of Biology, University of the Balearic Islands, 07122 Palma, Spain
| |
Collapse
|
4
|
Increased cerebrospinal fluid S100B protein levels in patients with trigeminal neuralgia and hemifacial spasm. Acta Neurochir (Wien) 2022; 165:959-965. [PMID: 36459237 DOI: 10.1007/s00701-022-05434-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 11/19/2022] [Indexed: 12/04/2022]
Abstract
BACKGROUND The pathophysiology of neurovascular compression syndrome has not been fully elucidated, and cerebrospinal fluid levels of nerve tissue-related markers involved in this disorder have not yet been reported. METHODS We measured cerebrospinal fluid levels of S100B protein, neuron-specific enolase, and myelin basic protein in 21 patients with trigeminal neuralgia, 9 patients with hemifacial spasms, and 10 patients with non-ruptured intracranial aneurysms (control). Cerebrospinal fluid levels of these markers were determined using commercially available assay kits. RESULTS Both trigeminal neuralgia and hemifacial spasm groups showed significantly increased cerebrospinal fluid levels of S100B compared with the control group (1120 [IQR 391-1420], 766 [IQR 583-1500], and 255 [IQR 190-285] pg/mL, respectively; p = 0.001). There were no statistically significant differences in cerebrospinal fluid levels of neuron-specific enolase or myelin basic protein among the groups. CONCLUSION Cerebrospinal fluid S100B levels were significantly higher in patients with trigeminal neuralgia and hemifacial spasm than in controls, which suggests the involvement of S100B in the underlying pathophysiology of neurovascular compression syndrome.
Collapse
|
5
|
Janigro D, Mondello S, Posti JP, Unden J. GFAP and S100B: What You Always Wanted to Know and Never Dared to Ask. Front Neurol 2022; 13:835597. [PMID: 35386417 PMCID: PMC8977512 DOI: 10.3389/fneur.2022.835597] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/03/2022] [Indexed: 12/19/2022] Open
Abstract
Traumatic brain injury (TBI) is a major global health issue, with outcomes spanning from intracranial bleeding, debilitating sequelae, and invalidity with consequences for individuals, families, and healthcare systems. Early diagnosis of TBI by testing peripheral fluids such as blood or saliva has been the focus of many research efforts, leading to FDA approval for a bench-top assay for blood GFAP and UCH-L1 and a plasma point-of-care test for GFAP. The biomarker S100B has been included in clinical guidelines for mTBI (mTBI) in Europe. Despite these successes, several unresolved issues have been recognized, including the robustness of prior data, the presence of biomarkers in tissues beyond the central nervous system, and the time course of biomarkers in peripheral body fluids. In this review article, we present some of these issues and provide a viewpoint derived from an analysis of existing literature. We focus on two astrocytic proteins, S100B and GFAP, the most commonly employed biomarkers used in mTBI. We also offer recommendations that may translate into a broader acceptance of these clinical tools.
Collapse
Affiliation(s)
- Damir Janigro
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH, United States.,FloTBI, Cleveland, OH, United States
| | - Stefania Mondello
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Jussi P Posti
- Department of Neurosurgery, Neurocenter, Turku Brain Injury Center, Turku University Hospital, University of Turku, Turku, Finland
| | - Johan Unden
- Department of Operation and Intensive Care, Hallands Hospital Halmstad, Lund University, Lund, Sweden
| |
Collapse
|
6
|
Mello e Souza T. Unraveling molecular and system processes for fear memory. Neuroscience 2022; 497:14-29. [DOI: 10.1016/j.neuroscience.2022.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 03/02/2022] [Accepted: 03/14/2022] [Indexed: 11/26/2022]
|
7
|
Leo M, Schmitt LI, Steffen R, Kutritz A, Kleinschnitz C, Hagenacker T. Modulation of Glutamate Transporter EAAT1 and Inward-Rectifier Potassium Channel K ir4.1 Expression in Cultured Spinal Cord Astrocytes by Platinum-Based Chemotherapeutics. Int J Mol Sci 2021; 22:6300. [PMID: 34208258 PMCID: PMC8230757 DOI: 10.3390/ijms22126300] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/06/2021] [Accepted: 06/09/2021] [Indexed: 12/26/2022] Open
Abstract
Platinum-based chemotherapeutics still play an essential role in cancer treatment. Despite their high effectiveness, severe side effects such as chemotherapy-induced neuropathy (CIPN) occur frequently. The pathophysiology of CIPN by platinum-based chemotherapeutics is not fully understood yet, but primarily the disturbance of dorsal root ganglion cells is discussed. However, there is increasing evidence of central nervous system involvement with activation of spinal cord astrocytes after treatment with chemotherapeutics. We investigated the influence of cis- or oxaliplatin on the functionality of cultured rat spinal cord astrocytes by using immunocytochemistry and patch-clamp electrophysiology. Cis- or oxaliplatin activated spinal astrocytes and led to downregulation of the excitatory amino acid transporter 1 (EAAT1) expression. Furthermore, the expression and function of potassium channel Kir4.1 were modulated. Pre-exposure to a specific Kir4.1 blocker in control astrocytes led to a reduced immune reactivity (IR) of EAAT1 and a nearly complete block of the current density. When spinal astrocytes were pre-exposed to antibiotic minocycline, all effects of cis- or oxaliplatin were abolished. Taken together, the modulation of Kir4.1 and EAAT1 proteins in astrocytes could be linked to the direct impact of cis- or oxaliplatin, identifying spinal astrocytes as a potential target in the prevention and treatment of chemotherapy-induced neuropathy.
Collapse
Affiliation(s)
- Markus Leo
- Department of Neurology, Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, 45147 Essen, Germany; (L.-I.S.); (R.S.); (A.K.); (C.K.); (T.H.)
| | | | | | | | | | | |
Collapse
|
8
|
Ryczko D, Hanini‐Daoud M, Condamine S, Bréant BJB, Fougère M, Araya R, Kolta A. S100β‐mediated astroglial control of firing and input processing in layer 5 pyramidal neurons of the mouse visual cortex. J Physiol 2020; 599:677-707. [DOI: 10.1113/jp280501] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 11/23/2020] [Indexed: 12/18/2022] Open
Affiliation(s)
- Dimitri Ryczko
- Département de Neurosciences Université de Montréal Montréal QC Canada
- Département de Pharmacologie‐Physiologie Université de Sherbrooke Sherbrooke QC Canada
- Centre de recherche du CHUS Sherbrooke QC Canada
- Institut de Pharmacologie de Sherbrooke Sherbrooke QC Canada
- Centre d'excellence en neurosciences de l'Université de Sherbrooke Sherbrooke QC Canada
| | | | - Steven Condamine
- Département de Neurosciences Université de Montréal Montréal QC Canada
| | | | - Maxime Fougère
- Département de Pharmacologie‐Physiologie Université de Sherbrooke Sherbrooke QC Canada
| | - Roberto Araya
- Département de Neurosciences Université de Montréal Montréal QC Canada
| | - Arlette Kolta
- Département de Neurosciences Université de Montréal Montréal QC Canada
- Faculté de Médecine Dentaire Université de Montréal Montréal QC Canada
| |
Collapse
|
9
|
Wang J, Liu J, Li R, Wang C. Research and progress on biomarkers of neuromyelitis optica spectrum disorders. J Recept Signal Transduct Res 2020; 41:417-424. [PMID: 33019871 DOI: 10.1080/10799893.2020.1830109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Neuromyelitis optica spectrum disorders (NMOSD) are a demyelinating disorder of the central nervous system based on the involvement of the optic nerve and/or spinal cord. The disease is characterized by high recurrence and disability. NMOSD is mainly diagnosed by AQP4-IgG and MOG-IgG. However, there are still some patients with negative or undetermined double-antibody, and AQP4-IgG and MOG-IgG cannot indicate the clinical disease activity. Therefore, it is urgent to explore interesting biomarkers in serum and cerebrospinal fluid to promote early clinical diagnosis and/or as a target for diagnosis and treatment. This article summarized the research progress in serum and cerebrospinal fluid biomarkers of astrocytes, neurons, myelin sheath, and other damage after the onset of NMOSD. Besides the value of microglial activation-related proteins in the diagnosis and treatment of NMOSD was prospected, so as to promote the research progress of NMOSD.
Collapse
Affiliation(s)
- Jinyang Wang
- School of Laboratory Medicine, Weifang Medical College, Weifang, P. R. China.,Department of Laboratory Medicine, the First Medical Centre, Chinese PLA General Hospital, Beijing, P. R. China
| | - Jiayu Liu
- Department of Laboratory Medicine, the First Medical Centre, Chinese PLA General Hospital, Beijing, P. R. China
| | - Ruibing Li
- Department of Laboratory Medicine, the First Medical Centre, Chinese PLA General Hospital, Beijing, P. R. China
| | - Chengbin Wang
- School of Laboratory Medicine, Weifang Medical College, Weifang, P. R. China.,Department of Laboratory Medicine, the First Medical Centre, Chinese PLA General Hospital, Beijing, P. R. China
| |
Collapse
|
10
|
Illouz T, Madar R, Biragyn A, Okun E. Restoring microglial and astroglial homeostasis using DNA immunization in a Down Syndrome mouse model. Brain Behav Immun 2019; 75:163-180. [PMID: 30389461 PMCID: PMC6358279 DOI: 10.1016/j.bbi.2018.10.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 09/22/2018] [Accepted: 10/23/2018] [Indexed: 12/20/2022] Open
Abstract
Down Syndrome (DS), the most common cause of genetic intellectual disability, is characterized by over-expression of the APP and DYRK1A genes, located on the triplicated chromosome 21. This chromosomal abnormality leads to a cognitive decline mediated by Amyloid-β (Aβ) overproduction and tau hyper-phosphorylation as early as the age of 40. In this study, we used the Ts65Dn mouse model of DS to evaluate the beneficial effect of a DNA vaccination against the Aβ1-11 fragment, in ameliorating Aβ-related neuropathology and rescue of cognitive and behavioral abilities. Anti-Aβ1-11 vaccination induced antibody production and facilitated clearance of soluble oligomers and small extracellular inclusions of Aβ from the hippocampus and cortex of Ts65Dn mice. This was correlated with reduced neurodegeneration and restoration of the homeostatic phenotype of microglial and astroglial cells. Vaccinated Ts65Dn mice performed better in spatial-learning tasks, exhibited reduced motor hyperactivity typical for this strain, and restored short-term memory abilities. Our findings support the hypothesis that DS individuals may benefit from active immunotherapy against Aβ from a young age by slowing the progression of dementia.
Collapse
Affiliation(s)
- Tomer Illouz
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan 5290002, Israel; The Paul Feder Laboratory on Alzheimer's Disease Research, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Ravit Madar
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan 5290002, Israel; The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel; The Paul Feder Laboratory on Alzheimer's Disease Research, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Arya Biragyn
- Laboratory of Molecular Biology and Immunology, NIA, NIH, MD 21224, USA
| | - Eitan Okun
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan 5290002, Israel; The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel; The Paul Feder Laboratory on Alzheimer's Disease Research, Bar-Ilan University, Ramat Gan 5290002, Israel.
| |
Collapse
|
11
|
Michetti F, D'Ambrosi N, Toesca A, Puglisi MA, Serrano A, Marchese E, Corvino V, Geloso MC. The S100B story: from biomarker to active factor in neural injury. J Neurochem 2018; 148:168-187. [DOI: 10.1111/jnc.14574] [Citation(s) in RCA: 128] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 07/19/2018] [Accepted: 08/15/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Fabrizio Michetti
- Institute of Anatomy and Cell Biology; Università Cattolica del Sacro Cuore; Rome Italy
- IRCCS San Raffaele Scientific Institute; Università Vita-Salute San Raffaele; Milan Italy
| | - Nadia D'Ambrosi
- Department of Biology; Università degli Studi di Roma Tor Vergata; Rome Italy
| | - Amelia Toesca
- Institute of Anatomy and Cell Biology; Università Cattolica del Sacro Cuore; Rome Italy
| | | | - Alessia Serrano
- Institute of Anatomy and Cell Biology; Università Cattolica del Sacro Cuore; Rome Italy
| | - Elisa Marchese
- Institute of Anatomy and Cell Biology; Università Cattolica del Sacro Cuore; Rome Italy
| | - Valentina Corvino
- Institute of Anatomy and Cell Biology; Università Cattolica del Sacro Cuore; Rome Italy
| | - Maria Concetta Geloso
- Institute of Anatomy and Cell Biology; Università Cattolica del Sacro Cuore; Rome Italy
| |
Collapse
|
12
|
Impaired oligodendrogenesis and myelination by elevated S100B levels during neurodevelopment. Neuropharmacology 2018; 129:69-83. [DOI: 10.1016/j.neuropharm.2017.11.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 10/22/2017] [Accepted: 11/03/2017] [Indexed: 11/23/2022]
|
13
|
Hao MM, Capoccia E, Cirillo C, Boesmans W, Vanden Berghe P. Arundic Acid Prevents Developmental Upregulation of S100B Expression and Inhibits Enteric Glial Development. Front Cell Neurosci 2017; 11:42. [PMID: 28280459 PMCID: PMC5322270 DOI: 10.3389/fncel.2017.00042] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 02/08/2017] [Indexed: 12/31/2022] Open
Abstract
S100B is expressed in various types of glial cells and is involved in regulating many aspects of their function. However, little is known about its role during nervous system development. In this study, we investigated the effect of inhibiting the onset of S100B synthesis in the development of the enteric nervous system, a network of neurons and glia located in the wall of the gut that is vital for control of gastrointestinal function. Intact gut explants were taken from embryonic day (E)13.5 mice, the day before the first immunohistochemical detection of S100B, and cultured in the presence of arundic acid, an inhibitor of S100B synthesis, for 48 h. The effects on Sox10-immunoreactive enteric neural crest progenitors and Hu-immunoreactive enteric neurons were then analyzed. Culture in arundic acid reduced the proportion of Sox10+ cells and decreased cell proliferation. There was no change in the density of Hu+ enteric neurons, however, a small population of cells exhibited atypical co-expression of both Sox10 and Hu, which was not observed in control cultures. Addition of exogenous S100B to the cultures did not change Sox10+ cell numbers. Overall, our data suggest that cell-intrinsic intracellular S100B is important for maintaining Sox10 and proliferation of the developing enteric glial lineage.
Collapse
Affiliation(s)
- Marlene M Hao
- Laboratory for Enteric Neuroscience, Translational Research in GastroIntestinal Disorders, KU Leuven Leuven, Belgium
| | - Elena Capoccia
- Laboratory for Enteric Neuroscience, Translational Research in GastroIntestinal Disorders, KU LeuvenLeuven, Belgium; Department of Physiology and Pharmacology, Sapienza University of RomeRome, Italy
| | - Carla Cirillo
- Laboratory for Enteric Neuroscience, Translational Research in GastroIntestinal Disorders, KU Leuven Leuven, Belgium
| | - Werend Boesmans
- Laboratory for Enteric Neuroscience, Translational Research in GastroIntestinal Disorders, KU Leuven Leuven, Belgium
| | - Pieter Vanden Berghe
- Laboratory for Enteric Neuroscience, Translational Research in GastroIntestinal Disorders, KU Leuven Leuven, Belgium
| |
Collapse
|
14
|
Long-Term Intake of Uncaria rhynchophylla Reduces S100B and RAGE Protein Levels in Kainic Acid-Induced Epileptic Seizures Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 2017:9732854. [PMID: 28386293 PMCID: PMC5343263 DOI: 10.1155/2017/9732854] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 01/13/2017] [Accepted: 01/24/2017] [Indexed: 12/19/2022]
Abstract
Epileptic seizures are crucial clinical manifestations of recurrent neuronal discharges in the brain. An imbalance between the excitatory and inhibitory neuronal discharges causes brain damage and cell loss. Herbal medicines offer alternative treatment options for epilepsy because of their low cost and few side effects. We established a rat epilepsy model by injecting kainic acid (KA, 12 mg/kg, i.p.) and subsequently investigated the effect of Uncaria rhynchophylla (UR) and its underlying mechanisms. Electroencephalogram and epileptic behaviors revealed that the KA injection induced epileptic seizures. Following KA injection, S100B levels increased in the hippocampus. This phenomenon was attenuated by the oral administration of UR and valproic acid (VA, 250 mg/kg). Both drugs significantly reversed receptor potentiation for advanced glycation end product proteins. Rats with KA-induced epilepsy exhibited no increase in the expression of metabotropic glutamate receptor 3, monocyte chemoattractant protein 1, and chemokine receptor type 2, which play a role in inflammation. Our results provide novel and detailed mechanisms, explaining the role of UR in KA-induced epileptic seizures in hippocampal CA1 neurons.
Collapse
|
15
|
Secretagogin-dependent matrix metalloprotease-2 release from neurons regulates neuroblast migration. Proc Natl Acad Sci U S A 2017; 114:E2006-E2015. [PMID: 28223495 DOI: 10.1073/pnas.1700662114] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The rostral migratory stream (RMS) is viewed as a glia-enriched conduit of forward-migrating neuroblasts in which chemorepulsive signals control the pace of forward migration. Here we demonstrate the existence of a scaffold of neurons that receive synaptic inputs within the rat, mouse, and human fetal RMS equivalents. These neurons express secretagogin, a Ca2+-sensor protein, to execute an annexin V-dependent externalization of matrix metalloprotease-2 (MMP-2) for reconfiguring the extracellular matrix locally. Mouse genetics combined with pharmacological probing in vivo and in vitro demonstrate that MMP-2 externalization occurs on demand and that its loss slows neuroblast migration. Loss of function is particularly remarkable upon injury to the olfactory bulb. Cumulatively, we identify a signaling cascade that provokes structural remodeling of the RMS through recruitment of MMP-2 by a previously unrecognized neuronal constituent. Given the life-long presence of secretagogin-containing neurons in human, this mechanism might be exploited for therapeutic benefit in rescue strategies.
Collapse
|
16
|
The role and potential mechanism of resveratrol in the prevention and control of epilepsy. Future Med Chem 2015; 7:2005-18. [PMID: 26505553 DOI: 10.4155/fmc.15.130] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Epilepsy is one of the most common diseases affecting the nervous system, with more than 50 million patients suffering from epilepsy worldwide. Although epilepsy has been prevalent for thousands of years, it is still not possible to completely control the disease. Despite an increase in the number of available antiepileptic drugs, the incidence of epilepsy and its cure rate have not been substantially improved; thus, there is an urgent need to identify new drugs that treat, cure or protect against epilepsy. Resveratrol is a polyphenol compound with a broad range of biological activity; not only it has considerable antiepileptic effects, but it is also neuroprotective and has functions to counter epileptic depression. Resveratrol has the potential to be a new antiepileptic drug, thus further studies are needed to better investigate its potential.
Collapse
|
17
|
Meng XJ, Wang F, Li CK. Resveratrol is Neuroprotective and Improves Cognition in Pentylenetetrazole-kindling Model of Epilepsy in Rats. Indian J Pharm Sci 2014; 76:125-31. [PMID: 24843185 PMCID: PMC4023281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 01/19/2014] [Accepted: 01/25/2014] [Indexed: 12/03/2022] Open
Abstract
S100B protein in serum and cerebral spinal fluid is increasingly used as a biochemical marker in early examinations after seizure to assess brain damage. Resveratrol, a nonflavonoid polyphenol, has been identified as a potent antiepileptic agent. However, a potential association between epilepsy with S100B protein in the cerebral spinal fluid and the sera of animal models lacks investigation. In this study, we evaluated the effects of resveratrol on behaviour and S100B protein levels in cerebral spinal fluid and serum in a rat model of chronic epilepsy induced via pentylenetetrazole kindling. By Morris water maze experiment analysis, we found that recovery of cognitive function in the resveratrol group (15 mg/kg/day), was significantly better than that of either the untreated or the vehicle groups. Further Nissl staining revealed that resveratrol significantly reduced pentylenetetrazole-induced death of neurons in the CA1 and CA3 regions of the hippocampus. Moreover, S100B protein levels in the cerebral spinal fluid and serum of rats treated with resveratrol were significantly reduced compared with the untreated and vehicle groups. These novel findings suggest an important mechanism of resveratrol and contribute to the treatment of epilepsy.
Collapse
Affiliation(s)
- X. J. Meng
- Department of Neurosurgery, School of Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - F. Wang
- Department of Neurosurgery, School of Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - C. K. Li
- Department of Neurosurgery, School of Medicine, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| |
Collapse
|
18
|
Bock N, Koc E, Alter H, Roessner V, Becker A, Rothenberger A, Manzke T. Chronic fluoxetine treatment changes S100B expression during postnatal rat brain development. J Child Adolesc Psychopharmacol 2013; 23:481-9. [PMID: 24024533 PMCID: PMC3779020 DOI: 10.1089/cap.2011.0065] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Fluoxetine, a selective serotonin reuptake inhibitor, is approved for treatment of childhood depression. In rats, fluoxetine influences neuronal development, but it is unclear whether it also influences glia development. S100B is a glia-derived calcium-binding protein, which may influence the development of serotonergic fibers and, vice versa, serotonin may influence the expression of S100B. OBJECTIVES The purpose of this study was to investigate whether fluoxetine treatment influences the expression of S100B during postnatal development, and whether potential changes are regionally dependent upon the time frame of drug administration. METHODS S100B gene expression and S100B protein expression in three different brain regions (frontal cortex, hippocampus, and striatum) were studied by real-time polymerase chain reaction (PCR) and immunohistochemistry, respectively. First, a short-term effect, 24 hours after a 14 day fluoxetine treatment (5 mg/kg/bw s.c.) of rats either from postnatal day (PD) 1 to 15, 21 to 35, or 50 to 64, was investigated. Then, the same treatment was used to analyze S100B gene and protein levels at PD 90 (long-term effect). RESULTS At PD 90, a significant increase of gene and protein expression was observed in all regions if rats were treated during PDs 21-35, whereas treatment during other periods had no long-term effects. A short-term effect 24 hours after fluoxetine treatment was found for almost all development stages and regions, demonstrated by a significant increase of S100B. CONCLUSIONS These results support recent research indicating a highly drug-sensitive period (i.e., periadolescence) of rat brain development. Therefore, further clinical studies should be performed to clarify whether such a sensitive period also exists in children.
Collapse
Affiliation(s)
- Nathalie Bock
- Department of Child and Adolescent Psychiatry, Georg-August-University of Göttingen, Göttingen, Germany
| | - Emre Koc
- Department of Child and Adolescent Psychiatry, Georg-August-University of Göttingen, Göttingen, Germany
| | - Hannah Alter
- Department of Child and Adolescent Psychiatry, Georg-August-University of Göttingen, Göttingen, Germany
| | - Veit Roessner
- Department of Child and Adolescent Psychiatry, Technische Universität Dresden, Dresden, Germany
| | - Andreas Becker
- Department of Child and Adolescent Psychiatry, Georg-August-University of Göttingen, Göttingen, Germany
| | - Aribert Rothenberger
- Department of Child and Adolescent Psychiatry, Georg-August-University of Göttingen, Göttingen, Germany
| | - Till Manzke
- Department of Neuro- and Sensory Physiology, Georg-August-University of Göttingen, Göttingen, Germany.,DFG Research Center Molecular Physiology of the Brain, Göttingen, Germany
| |
Collapse
|
19
|
Kleindienst A, Grünbeck F, Buslei R, Emtmann I, Buchfelder M. Intraperitoneal treatment with S100B enhances hippocampal neurogenesis in juvenile mice and after experimental brain injury. Acta Neurochir (Wien) 2013; 155:1351-60. [PMID: 23649988 DOI: 10.1007/s00701-013-1720-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2012] [Accepted: 04/08/2013] [Indexed: 11/26/2022]
Abstract
BACKGROUND Neurogenesis is documented in adult mammals including humans, is promoted by neurotrophic factors, and constitutes an innate repair mechanism following brain injury. The glial neurotrophic protein S100B is released following various types of brain injuries, enhances hippocampal neurogenesis and improves cognitive function following brain injury in rats when applied intrathecally. The present study was designed to elucidate whether the beneficial effect of S100B on injury-induced neurogenesis can be confirmed in mice when applied intraperitoneally (i.p.), and whether this effect is dose-dependent. METHODS Male juvenile mice were subjected to a unilateral parietal cryolesion or sham injury, and treated with S100B at 20nM, 200nM or vehicle i.p. once daily. Hippocampal progenitor cell proliferation was quantified following labelling with bromo-deoxyuridine (BrdU, 50 mg/KG i.p.) in the germinative area of the dentate gyrus, the subgranular zone (SGZ), on day 4 as well as on cell survival and migration to the granular cell layer (GCL) on day 28. Progenitor cell differentiation was assessed following colabelling with the glial marker GFAP and the neuronal marker NeuN. RESULTS S100B enhanced significantly the early progenitor cell proliferation in the SGZ as well as cell survival and migration to the GCL, and promoted neuronal differentiation. While these effects were predominately dose-dependent, 200nM S100B failed to enhance the proliferation in the SGZ on day 4 post-injury. CONCLUSION We conclude that S100B participates in hippocampal neurogenesis after injury at lower nanomolar concentrations. Therefore S100B may serve as a potential adjunct treatment to promote neuroregeneration following brain damage.
Collapse
Affiliation(s)
- Andrea Kleindienst
- Departments of Neurosurgery and Neuropathology, Friedrich-Alexander University of Erlangen-Nürnberg, Schwabachanlage 6, 91054, Erlangen, Germany.
| | | | | | | | | |
Collapse
|
20
|
Kim J, Wan CK, J O'Carroll S, Shaikh SB, Nicholson LFB. The role of receptor for advanced glycation end products (RAGE) in neuronal differentiation. J Neurosci Res 2012; 90:1136-47. [PMID: 22344976 DOI: 10.1002/jnr.23014] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Revised: 11/18/2011] [Accepted: 12/01/2011] [Indexed: 02/01/2023]
Abstract
The receptor for advanced glycation end products (RAGE) is a multiligand receptor protein thought to play an important role in neuronal differentiation. RAGE can bind a number of ligands and activate a variety of signalling pathways that lead to diverse downstream effects. Amphoterin and S100B are endogenous ligands, the interaction of which with RAGE is known to be involved in defined physiological processes. The present study investigated the spatiotemporal pattern of the expression for RAGE and its ligands, amphoterin and S100B, during neuronal differentiation of NT2/D1 cells. In this study, all three proteins were shown to increase with progression of neuronal differentiation as determined by Western blotting, raising the possibility that both amphoterin and S100B may interact with RAGE and have important functions during the process of cell differentiation. Moreover, blocking the activation of RAGE with neutralizing antibody in the presence of retinoic acid disrupted the progression of normal neuronal differentiation. Immunocytochemistry (ICC) studies showed that amphoterin partially colocalized with RAGE within differentiating NT2 cells, whereas S100B showed a high degree of colocalization. This result suggests that S100B is more likely to be the principal ligand for RAGE during the differentiation process and that RAGE and amphoterin might have both independent and combined roles. Moreover, RAGE was expressed only in cells that were committed to a neuronal phenotype, suggesting direct involvement of RAGE in mediating cellular changes within differentiating neuronal cells. Further detailed studies are now required to characterize fully the role of RAGE during the neuronal differentiation period.
Collapse
Affiliation(s)
- Joanne Kim
- Department of Anatomy with Radiology, Centre for Brain Research, Faculty of Medical and Health Science, University of Auckland, Auckland, New Zealand.
| | | | | | | | | |
Collapse
|
21
|
Michetti F, Corvino V, Geloso MC, Lattanzi W, Bernardini C, Serpero L, Gazzolo D. The S100B protein in biological fluids: more than a lifelong biomarker of brain distress. J Neurochem 2012; 120:644-59. [PMID: 22145907 DOI: 10.1111/j.1471-4159.2011.07612.x] [Citation(s) in RCA: 173] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
S100B is a calcium-binding protein concentrated in glial cells, although it has also been detected in definite extra-neural cell types. Its biological role is still debated. When secreted, S100B is believed to have paracrine/autocrine trophic effects at physiological concentrations, but toxic effects at higher concentrations. Elevated S100B levels in biological fluids (CSF, blood, urine, saliva, amniotic fluid) are thus regarded as a biomarker of pathological conditions, including perinatal brain distress, acute brain injury, brain tumors, neuroinflammatory/neurodegenerative disorders, psychiatric disorders. In the majority of these conditions, high S100B levels offer an indicator of cell damage when standard diagnostic procedures are still silent. The key question remains as to whether S100B is merely leaked from injured cells or is released in concomitance with both physiological and pathological conditions, participating at high concentrations in the events leading to cell injury. In this respect, S100B levels in biological fluids have been shown to increase in physiological conditions characterized by stressful physical and mental activity, suggesting that it may be physiologically regulated and raised during conditions of stress, with a putatively active role. This possibility makes this protein a candidate not only for a biomarker but also for a potential therapeutic target.
Collapse
Affiliation(s)
- Fabrizio Michetti
- Institute of Anatomy and Cell Biology, Università Cattolica Sacro Cuore, Roma, Italy.
| | | | | | | | | | | | | |
Collapse
|
22
|
Mattusch C, Diederich KW, Schmidt A, Scheinert D, Thiele H, Schuler G, Desch S. Effect of Carotid Artery Stenting on the Release of S-100B and Neurone-Specific Enolase. Angiology 2011; 62:376-80. [DOI: 10.1177/0003319710387920] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Serum levels of S-100B and neurone-specific enolase (NSE) reflect cerebral injury in a variety of neurological conditions such as stroke, traumatic brain injury, and cardiac arrest. There are limited data on the release of S-100B and NSE following carotid artery stenting (CAS). In 22 patients undergoing CAS, serial blood samples for S-100B and NSE were collected before and 2, 4, and 6 to 8 hours after the procedure. A group of 20 patients with significant CAS undergoing purely diagnostic angiography served as controls. A significant increase in S-100B levels was observed 2 hours after the procedure in patients with CAS (P = .001) with a gradual decline over the next hours. In contrast, patients who underwent purely diagnostic angiography did not show significant changes in S-100B levels up to 8 hours after the procedure. Neither patients with CAS nor those undergoing diagnostic angiography displayed any significant changes in serial NSE levels.
Collapse
Affiliation(s)
- Christiane Mattusch
- University of Leipzig-Heart Center, Department of Internal Medicine/Cardiology, Leipzig, Germany
| | - Klaus-Werner Diederich
- University of Leipzig-Heart Center, Department of Internal Medicine/Cardiology, Leipzig, Germany
| | - Andrej Schmidt
- Park Hospital and Heart Center, Department of Angiology, Leipzig, Germany
| | - Dierk Scheinert
- Park Hospital and Heart Center, Department of Angiology, Leipzig, Germany
| | - Holger Thiele
- University of Leipzig-Heart Center, Department of Internal Medicine/ Cardiology, Leipzig, Germany
| | - Gerhard Schuler
- University of Leipzig-Heart Center, Department of Internal Medicine/ Cardiology, Leipzig, Germany
| | - Steffen Desch
- University of Leipzig-Heart Center, Department of Internal Medicine/ Cardiology, Leipzig, Germany,
| |
Collapse
|
23
|
Lin YW, Hsieh CL. Oral Uncaria rhynchophylla (UR) reduces kainic acid-induced epileptic seizures and neuronal death accompanied by attenuating glial cell proliferation and S100B proteins in rats. JOURNAL OF ETHNOPHARMACOLOGY 2011; 135:313-320. [PMID: 21402140 DOI: 10.1016/j.jep.2011.03.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Revised: 12/15/2010] [Accepted: 03/05/2011] [Indexed: 05/30/2023]
Abstract
AIM OF THE STUDY Epilepsy is a common clinical syndrome with recurrent neuronal discharges in cerebral cortex and hippocampus. Here we aim to determine the protective role of Uncaria rhynchophylla (UR), an herbal drug belong to Traditional Chinese Medicine (TCM), on epileptic rats. MATERIALS AND METHODS To address this issue, we tested the effect of UR on kainic acid (KA)-induced epileptic seizures and further investigate the underlying mechanisms. RESULTS Oral UR successfully decreased neuronal death and discharges in hippocampal CA1 pyramidal neurons. The population spikes (PSs) were decreased from 4.1 ± 0.4 mV to 2.1 ± 0.3 mV in KA-induced epileptic seizures and UR-treated groups, respectively. Oral UR protected animals from neuronal death induced by KA treatment (from 34 ± 4.6 to 191.7 ± 48.6 neurons/field) through attenuating glial cell proliferation and S100B protein expression but not GABAA and TRPV1 receptors. CONCLUSIONS The above results provide detail mechanisms underlying the neuroprotective action of UR on KA-induced epileptic seizure in hippocampal CA1 neurons.
Collapse
Affiliation(s)
- Yi-Wen Lin
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan
| | | |
Collapse
|
24
|
Cooke MJ, Wang Y, Morshead CM, Shoichet MS. Controlled epi-cortical delivery of epidermal growth factor for the stimulation of endogenous neural stem cell proliferation in stroke-injured brain. Biomaterials 2011; 32:5688-97. [PMID: 21550655 DOI: 10.1016/j.biomaterials.2011.04.032] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Accepted: 04/12/2011] [Indexed: 01/06/2023]
Abstract
One of the challenges in treating central nervous system (CNS) disorders with biomolecules is achieving local delivery while minimizing invasiveness. For the treatment of stroke, stimulation of endogenous neural stem/progenitor cells (NSPCs) by growth factors is a promising strategy for tissue regeneration. Epidermal growth factor (EGF) enhances proliferation of endogenous NSPCs in the subventricular zone (SVZ) when delivered directly to the ventricles of the brain; however, this strategy is highly invasive. We designed a biomaterials-based strategy to deliver molecules directly to the brain without tissue damage. EGF or poly(ethylene glycol)-modified EGF (PEG-EGF) was dispersed in a hyaluronan and methylcellulose (HAMC) hydrogel and placed epi-cortically on both uninjured and stroke-injured mouse brains. PEG-modification decreased the rate of EGF degradation by proteases, leading to a significant increase in protein accumulation at greater tissue depths than previously shown. Consequently, EGF and PEG-EGF increased NSPC proliferation in uninjured and stroke-injured brains; and in stroke-injured brains, PEG-EGF significantly increased NSPC stimulation. Our epi-cortical delivery system is a minimally-invasive method for local delivery to the brain, providing a new paradigm for local delivery to the brain.
Collapse
Affiliation(s)
- Michael J Cooke
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, ON, Canada M5S 3E5
| | | | | | | |
Collapse
|
25
|
Sorci G, Bianchi R, Riuzzi F, Tubaro C, Arcuri C, Giambanco I, Donato R. S100B Protein, A Damage-Associated Molecular Pattern Protein in the Brain and Heart, and Beyond. Cardiovasc Psychiatry Neurol 2010; 2010:656481. [PMID: 20827421 PMCID: PMC2933911 DOI: 10.1155/2010/656481] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2010] [Accepted: 06/08/2010] [Indexed: 12/15/2022] Open
Abstract
S100B belongs to a multigenic family of Ca(2+)-binding proteins of the EF-hand type and is expressed in high abundance in the brain. S100B interacts with target proteins within cells thereby altering their functions once secreted/released with the multiligand receptor RAGE. As an intracellular regulator, S100B affects protein phosphorylation, energy metabolism, the dynamics of cytoskeleton constituents (and hence, of cell shape and migration), Ca(2+) homeostasis, and cell proliferation and differentiation. As an extracellular signal, at low, physiological concentrations, S100B protects neurons against apoptosis, stimulates neurite outgrowth and astrocyte proliferation, and negatively regulates astrocytic and microglial responses to neurotoxic agents, while at high doses S100B causes neuronal death and exhibits properties of a damage-associated molecular pattern protein. S100B also exerts effects outside the brain; as an intracellular regulator, S100B inhibits the postinfarction hypertrophic response in cardiomyocytes, while as an extracellular signal, (high) S100B causes cardiomyocyte death, activates endothelial cells, and stimulates vascular smooth muscle cell proliferation.
Collapse
Affiliation(s)
- Guglielmo Sorci
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Via del Giochetto, 06122 Perugia, Italy
| | - Roberta Bianchi
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Via del Giochetto, 06122 Perugia, Italy
| | - Francesca Riuzzi
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Via del Giochetto, 06122 Perugia, Italy
| | - Claudia Tubaro
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Via del Giochetto, 06122 Perugia, Italy
| | - Cataldo Arcuri
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Via del Giochetto, 06122 Perugia, Italy
| | - Ileana Giambanco
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Via del Giochetto, 06122 Perugia, Italy
| | - Rosario Donato
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Via del Giochetto, 06122 Perugia, Italy
| |
Collapse
|
26
|
Mollenhauer B, Trenkwalder C. Neurochemical biomarkers in the differential diagnosis of movement disorders. Mov Disord 2009; 24:1411-26. [PMID: 19412961 DOI: 10.1002/mds.22510] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In recent years, the neurochemical analysis of neuronal proteins in cerebrospinal fluid (CSF) has become increasingly accepted for the diagnosis of neurodegenerative dementia diseases such as Alzheimer's disease and Creutzfeldt-Jakob disease. CSF surrounds the central nervous system, and in the composition of CSF proteins one finds brain-specific proteins that are prioritized from blood-derived proteins. Levels of specific CSF proteins could be very promising biomarkers for central nervous system diseases. We need the development of more easily accessible biomarkers, in the blood. In neurodegenerative diseases with and without dementia, studies on CSF and blood proteins have investigated the usefulness of biomarkers in differential diagnosis. The clinical diagnoses of Parkinson's disease, dementia with Lewy bodies, multiple system atrophy, progressive supranuclear palsy, and corticobasal degeneration still rely mainly on clinical symptoms as defined by international classification criteria. In this article, we review CSF biomarkers in these movement disorders and discuss recent published reports on the neurochemical intra vitam diagnosis of neurodegenerative disorders (including recent CSF alpha-synuclein findings).
Collapse
|
27
|
S100B secretion in acute brain slices: modulation by extracellular levels of Ca(2+) and K (+). Neurochem Res 2009; 34:1603-11. [PMID: 19288274 DOI: 10.1007/s11064-009-9949-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2008] [Accepted: 03/04/2009] [Indexed: 01/16/2023]
Abstract
Hippocampal slices have been widely used to investigate electrophysiological and metabolic neuronal parameters, as well as parameters of astroglial activity including protein phosphorylation and glutamate uptake. S100B is an astroglial-derived protein, which extracellularly plays a neurotrophic activity during development and excitotoxic insult. Herein, we characterized S100B secretion in acute hippocampal slices exposed to different concentrations of K(+) and Ca(2+) in the extracellular medium. Absence of Ca(2+) and/or low K(+) (0.2 mM KCl) caused an increase in S100B secretion, possibly by mobilization of internal stores of Ca(2+). In contrast, high K(+) (30 mM KCl) or calcium channel blockers caused a decrease in S100B secretion. This study suggests that exposure of acute hippocampal slices to low- and high-K(+) could be used as an assay to evaluate astrocyte activity by S100B secretion: positively regulated by low K(+) (possibly involving mobilization of internal stores of Ca(2+)) and negatively regulated by high-K(+) (likely secondary to influx of K(+)).
Collapse
|
28
|
Brozzi F, Arcuri C, Giambanco I, Donato R. S100B Protein Regulates Astrocyte Shape and Migration via Interaction with Src Kinase: IMPLICATIONS FOR ASTROCYTE DEVELOPMENT, ACTIVATION, AND TUMOR GROWTH. J Biol Chem 2009; 284:8797-811. [PMID: 19147496 DOI: 10.1074/jbc.m805897200] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
S100B is a Ca(2+)-binding protein of the EF-hand type that is abundantly expressed in astrocytes and has been implicated in the regulation of several intracellular activities, including proliferation and differentiation. We show here that reducing S100B levels in the astrocytoma cell line GL15 and the Müller cell line MIO-M1 by small interference RNA technique results in a rapid disassembly of stress fibers, collapse of F-actin onto the plasma membrane and reduced migration, and acquisition of a stellate shape. Also, S100B-silenced GL15 and MIO-M1 Müller cells show a higher abundance of glial fibrillary acidic protein filaments, which mark differentiated astrocytes, compared with control cells. These effects are dependent on reduced activation of the phosphatidylinositol 3-kinase (PI3K) downstream effectors, Akt and RhoA, and consequently elevated activity of GSK3beta and Rac1 and decreased activity of the RhoA-associated kinase. Also, rat primary astrocytes transiently down-regulate S100B expression when exposed to the differentiating agent dibutyryl cyclic AMP and re-express S100B at later stages of dibutyryl cyclic AMP-induced differentiation. Moreover, reducing S100B levels results in a remarkably slow resumption of S100B expression, suggesting the S100B might regulate its own expression. Finally, we show that S100B interacts with Src kinase, thereby stimulating the PI3K/Akt and PI3K/RhoA pathways. These results suggest that S100B might contribute to reduce the differentiation potential of cells of the astrocytic lineage and participate in the astrocyte activation process in the case of brain insult and in invasive properties of glioma cells.
Collapse
Affiliation(s)
- Flora Brozzi
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, C.P. 81 Succ. 3, 06122 Perugia, Italy
| | | | | | | |
Collapse
|
29
|
Donato R, Sorci G, Riuzzi F, Arcuri C, Bianchi R, Brozzi F, Tubaro C, Giambanco I. S100B's double life: intracellular regulator and extracellular signal. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2008; 1793:1008-22. [PMID: 19110011 DOI: 10.1016/j.bbamcr.2008.11.009] [Citation(s) in RCA: 537] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2008] [Revised: 11/12/2008] [Accepted: 11/21/2008] [Indexed: 12/22/2022]
Abstract
The Ca2+-binding protein of the EF-hand type, S100B, exerts both intracellular and extracellular functions. Recent studies have provided more detailed information concerning the mechanism(s) of action of S100B as an intracellular regulator and an extracellular signal. Indeed, intracellular S100B acts as a stimulator of cell proliferation and migration and an inhibitor of apoptosis and differentiation, which might have important implications during brain, cartilage and skeletal muscle development and repair, activation of astrocytes in the course of brain damage and neurodegenerative processes, and of cardiomyocyte remodeling after infarction, as well as in melanomagenesis and gliomagenesis. As an extracellular factor, S100B engages RAGE (receptor for advanced glycation end products) in a variety of cell types with different outcomes (i.e. beneficial or detrimental, pro-proliferative or pro-differentiative) depending on the concentration attained by the protein, the cell type and the microenvironment. Yet, RAGE might not be the sole S100B receptor, and S100B's ability to engage RAGE might be regulated by its interaction with other extracellular factors. Future studies using S100B transgenic and S100B null mice might shed more light on the functional role(s) of the protein.
Collapse
Affiliation(s)
- Rosario Donato
- Department of Experimental Medicine and Biochemical Sciences, Section Anatomy, University of Perugia, Via del Giochetto C.P. 81 Succ. 3, 06122 Perugia, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Neural-activity-dependent release of S100B from astrocytes enhances kainate-induced gamma oscillations in vivo. J Neurosci 2008; 28:10928-36. [PMID: 18945900 DOI: 10.1523/jneurosci.3693-08.2008] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
S100B is the principal calcium-binding protein of astrocytes and known to be secreted to extracellular space. Although secreted S100B has been reported to promote neurite extension and cell survival via its receptor [receptor for advanced glycation end products (RAGE)], effects of extracellular S100B on neural activity have been mostly unexplored. Here, we demonstrate that secreted S100B enhances kainate-induced gamma oscillations. Local infusion of S100B in S100B(-/-) mice enhanced hippocampal kainate-induced gamma oscillations in vivo. In a complementary set of experiments, local application of anti-S100B antibody in wild-type mice attenuated the gamma oscillations. Both results indicate that the presence of extracellular S100B enhances the kainate-induced gamma oscillations. In acutely isolated hippocampal slices, kainate application increased S100B secretion in a neural-activity-dependent manner. Further pharmacological experiments revealed that S100B secretion was critically dependent on presynaptic release of neurotransmitter and activation of metabotropic glutamate receptor 3. Moreover, the kainate-induced gamma oscillations were attenuated by the genetic deletion or antibody blockade of RAGE in vivo. These results suggest RAGE activation by S100B enhances the gamma oscillations. Together, we propose a novel pathway of neuron-glia communications--astrocytic release of S100B modulates neural network activity through RAGE activation.
Collapse
|
31
|
Yang K, Xie GR, Hu YQ, Mao FQ, Su LY. The effects of gender and numbers of depressive episodes on serum S100B levels in patients with major depression. J Neural Transm (Vienna) 2008; 115:1687-94. [PMID: 18982242 DOI: 10.1007/s00702-008-0130-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2008] [Accepted: 10/04/2008] [Indexed: 11/24/2022]
Abstract
S100B protein is a calcium-binding protein mostly derived from glial cells, which exerts trophic or toxic effects on neural cell depending on its concentration. It has been reported that S100B played an important role as a potential marker in psychiatric disorders. Thus, we will explore the clinical implication of S100B in major depression, especially the effect of gender and numbers of depressive episodes on S100B. The levels of serum S100B were measured with enzyme-linked immunosorbent assay (ELISA) in 54 patients with major depression and 35 age-matched healthy controls. The S100B levels in major depressed patients were significantly higher than those in controls. The serum S100B levels in female patients were significantly higher than those in male patients. Patients with recurrent depressive episodes had significantly higher S100B levels than those in first-episode depression. Serum S100B levels were significantly positive related with the numbers of depressive episode, family history and cognitive disturbance scores. These findings confirmed an increase in serum S100B levels in major depressive patients and presence of a sexual dimorphism. Moreover, numbers of depressive episodes in depression seemed to have an additional increasing effect on S100B levels.
Collapse
Affiliation(s)
- Kun Yang
- Department of Psychiatry, Tianjin Medical University, 300070, Tianjin, People's Republic of China
| | | | | | | | | |
Collapse
|
32
|
Malup TK, Kobzev VF, Zhdanova LG, Slobodyanyuk SY, Sviridov SM. Methylation of CpG dinucleotides in the promoter region of the gene encoding the S100b protein in BALB/cLac mice. DOKL BIOCHEM BIOPHYS 2008; 412:1-3. [PMID: 17506341 DOI: 10.1134/s1607672907010012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- T K Malup
- Institute of Cytology and Genetics, Siberian Division, Russian Academy of Sciences, pr. Akademika Lavrent'eva 10, Novosibirsk 630090, Russia
| | | | | | | | | |
Collapse
|
33
|
Bloomfield SM, McKinney J, Smith L, Brisman J. Reliability of S100B in predicting severity of central nervous system injury. Neurocrit Care 2007; 6:121-38. [PMID: 17522796 DOI: 10.1007/s12028-007-0008-x] [Citation(s) in RCA: 142] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
S100B is a protein biomarker that reflects CNS injury. It can be measured in the CSF or serum with readily available immunoassay kits. The excellent sensitivity of S100B has enabled it to confirm the existence of subtle brain injury in patients with mild head trauma, strokes, and after successful resuscitation from cardiopulmonary arrest. The extent of S100B elevation has been found to be useful in predicting clinical outcome after brain injury. Elevations of S100B above certain threshold levels might be able to reliably predict brain death or mortality. A normal S100B level reliably predicts the absence of significant CNS injury. The specificity of S100B levels as a reflection of CNS injury is compromised by the findings that extra-cranial injuries can lead to elevations in the absence of brain injury. This potential problem can most likely be avoided by measuring serial S100B levels along with other biomarkers and carefully noting peripheral injuries. Serum markers GFAP and NSE are both more specific for CNS injury and have little to no extra-cranial sources. Sustained elevations of S100B over 24 h along with elevations of GFAP and NSE can more reliably predict the extent of brain injury and clinical outcomes. In the future, S100B measurements might reliably predict secondary brain injury and enable physicians to initiate therapeutic interventions in a timelier manner. S100B levels have been shown to rise hours to days before changes in ICP, neurological examinations, and neuroimaging tests. S100B levels may also be used to monitor the efficacy of treatments.
Collapse
Affiliation(s)
- Stephen M Bloomfield
- New Jersey Neuroscience, Institute JFK Hospital and Medical Center, Edison, NJ 08818, USA.
| | | | | | | |
Collapse
|
34
|
Aleksic M, Heckenkamp J, Reichert V, Gawenda M, Brunkwall J. S-100B Release during Carotid Endarterectomy under Local Anesthesia. Ann Vasc Surg 2007; 21:571-5. [PMID: 17521874 DOI: 10.1016/j.avsg.2007.04.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2006] [Revised: 04/02/2007] [Accepted: 04/02/2007] [Indexed: 11/23/2022]
Abstract
The neuronal protein S-100B has been found to be an indicator of cellular brain damage. The aim of the study was to evaluate whether cross-clamping of the carotid artery for carotid endarterectomy (CEA) under local anesthesia is associated with the same S-100B release pattern as during general anesthesia, where an increase in S-100B concentration in the jugular vein blood of 120% has been reported. In 45 consecutive patients undergoing CEA under local anesthesia, serum S-100B samples were drawn before surgery (T1), before carotid cross-clamping (T2), before cerebral reperfusion (T3), after reperfusion but before the end of surgery (T4), and 6 hr postoperatively (T5). At T1 and T5, blood samples were drawn only from the radial artery. Intraoperatively (T2-T4), samples were collected from the internal jugular vein additionally. S-100B levels were determined using an immunoluminometric assay (LIAISON) Sangtec 100; Sangtec, Bromma, Sweden). In eight patients, it was necessary to insert an intraluminal shunt because of signs of cerebral ischemia. In the remaining 37 patients, median carotid clamping time was 40 min. There were no neurological complications. There were no differences in baseline S-100B levels regarding gender and symptomatology. Median baseline (T1) and postoperative (T5) S-100B levels were identical (0.077 microg/L). All blood samples from the jugular vein showed significantly higher median S-100B levels than the corresponding arterial blood samples. Only slight increases of 13% and 18% were found during cross-clamping (T3) compared to the first intraoperative measurement (T2) in the venous and arterial samples, respectively, which was followed by decreases of 5% and 18%, respectively (T3-T4). S-100B release did not differ at any time point between patients who needed and patients who did not need a shunt, in either the arterial or the venous blood samples. During uncomplicated CEA under local anesthesia, there is no relevant increase of S-100B. These results are different from those reported when CEA is done under general anesthesia.
Collapse
Affiliation(s)
- Marko Aleksic
- Division of Vascular Surgery, Department of Visceral and Vascular Surgery, University of Cologne, Cologne, Germany.
| | | | | | | | | |
Collapse
|
35
|
Leclerc E, Fritz G, Weibel M, Heizmann CW, Galichet A. S100B and S100A6 differentially modulate cell survival by interacting with distinct RAGE (receptor for advanced glycation end products) immunoglobulin domains. J Biol Chem 2007; 282:31317-31. [PMID: 17726019 DOI: 10.1074/jbc.m703951200] [Citation(s) in RCA: 214] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
S100 proteins are EF-hand calcium-binding proteins with various intracellular functions including cell proliferation, differentiation, migration, and apoptosis. Some S100 proteins are also secreted and exert extracellular paracrine and autocrine functions. Experimental results suggest that the receptor for advanced glycation end products (RAGE) plays important roles in mediating S100 protein-induced cellular signaling. Here we compared the interaction of two S100 proteins, S100B and S100A6, with RAGE by in vitro assay and in culture of human SH-SY5Y neuroblastoma cells. Our in vitro binding data showed that S100B and S100A6, although structurally very similar, interact with different RAGE extracellular domains. Our cell assay data demonstrated that S100B and S100A6 differentially modulate cell survival. At micromolar concentration, S100B increased cellular proliferation, whereas at the same concentration, S100A6 triggered apoptosis. Although both S100 proteins induced the formation of reactive oxygen species, S100B recruited phosphatidylinositol 3-kinase/AKT and NF-kappaB, whereas S100A6 activated JNK. More importantly, we showed that S100B and S100A6 modulate cell survival in a RAGE-dependent manner; S100B specifically interacted with the RAGE V and C(1) domains and S100A6 specifically interacted with the C(1) and C(2) RAGE domains. Altogether these results highlight the complexity of S100/RAGE cellular signaling.
Collapse
MESH Headings
- Apoptosis
- Blotting, Western
- Caspase 3/metabolism
- Caspase 7/metabolism
- Cell Line, Tumor
- Cell Survival/physiology
- Culture Media, Serum-Free
- Electrophoresis, Polyacrylamide Gel
- Enzyme-Linked Immunosorbent Assay
- Escherichia coli/genetics
- Fluorescent Antibody Technique, Direct
- Glioblastoma/pathology
- Humans
- In Situ Nick-End Labeling
- Luminescent Measurements
- Models, Biological
- NF-kappa B/metabolism
- Neuroblastoma/pathology
- Protein Structure, Tertiary
- Reactive Oxygen Species/metabolism
- Receptor for Advanced Glycation End Products
- Receptors, Immunologic/immunology
- Recombinant Proteins/isolation & purification
- Recombinant Proteins/metabolism
- S100 Proteins/genetics
- S100 Proteins/metabolism
- S100 Proteins/physiology
- Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
- Time Factors
- Transfection
Collapse
Affiliation(s)
- Estelle Leclerc
- Division of Clinical Chemistry and Biochemistry, Department of Pediatrics, University of Zurich, Steinwiesstrasse 75, 8032 Zurich, Switzerland
| | | | | | | | | |
Collapse
|
36
|
Abstract
In recent years there has been a proliferation of interest in the brain-specific protein S100B, its many physiologic roles, and its behaviour in various neuropathologic conditions. Since the mid-1960s, its wide variety of intracellular and extracellular activities has been elucidated, and it has also been implicated in an increasing number of central nervous system (CNS) disorders. S100B is part of a superfamily of proteins, some of which (including S100B) have been implicated as calcium-dependent regulatory proteins that modulate the activity of effector proteins or cells. S100B is primarily an astrocytic protein. Within cells, it may have a role in signal transduction, and it is involved in calcium homeostasis. Information about the functional implication of S100B secretion by astrocytes into the extracellular space is scant but there is substantial evidence that secreted glial S100B exerts trophic or toxic effects depending on its concentration. This review summarises the historic development and current knowledge of S100B, including recent interesting findings relating S100B to a diversity of CNS pathologies such as traumatic brain injury, Alzheimer's disease, Down's syndrome, schizophrenia, and Tourette's syndrome. These broad implications have led some workers to describe S100B as 'the CRP (C-reactive protein) of the brain.' This review also examines S100B's potential role as a neurologic screening tool, or biomarker of CNS injury, analogous to the role of CRP as a marker of systemic inflammation.
Collapse
Affiliation(s)
- Jon Sen
- Institute of Neurology, University College London, Queen Square, London, United Kingdom.
| | | |
Collapse
|
37
|
Kleindienst A, Hesse F, Bullock MR, Buchfelder M. The neurotrophic protein S100B: value as a marker of brain damage and possible therapeutic implications. PROGRESS IN BRAIN RESEARCH 2007; 161:317-25. [PMID: 17618987 DOI: 10.1016/s0079-6123(06)61022-4] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
We provide a critical analysis of the value of S100B as a marker of brain damage and possible therapeutic implications. The early assessment of the injury severity and the consequent prognosis are of major concern for physicians treating patients suffering from traumatic brain injury (TBI). A reliable indicator to accurately determine the extent of the brain damage has to meet certain requirements: (i) to originate in the central nervous system (CNS) with no contribution from extracerebral sources; (ii) a passive release from damaged neurons and/or glial cells without any stimulated active release; (iii) a lack of specific effects on neurons and/or glial cells interfering with the initial injury; (iv) an unlimited passage through the blood-brain barrier (BBB). The measurement of putative biochemical markers, such as the S100B protein, has been proposed in this role. Over the past decade, numerous studies have reported a positive correlation of S100B serum levels with a poor outcome following TBI. However, some studies raise doubt whether the serum measurement of S100B is a valid biochemical marker of brain damage. We summarize the specific properties of S100B and analyze whether they support or counteract the necessary requirements to designate this protein as an indicator of brain damage. Finally, we report recent experimental findings suggesting a possible therapeutic potential of S100B.
Collapse
Affiliation(s)
- Andrea Kleindienst
- Department of Neurosurgery, Friedrich-Alexander-University, Erlangen-Nuremberg, Germany.
| | | | | | | |
Collapse
|
38
|
Kleindienst A, Ross Bullock M. A Critical Analysis of the Role of the Neurotrophic Protein S100B in Acute Brain Injury. J Neurotrauma 2006; 23:1185-200. [PMID: 16928177 DOI: 10.1089/neu.2006.23.1185] [Citation(s) in RCA: 120] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
We provide a critical analysis of the relevance of S100B in acute brain injury emphazising the beneficial effect of its biological properties. S100B is a calcium-binding protein, primarily produced by glial cells, and exerts auto- and paracrine functions. Numerous reports indicate, that S100B is released after brain insults and serum levels are positively correlated with the degree of injury and negatively correlated with outcome. However, new data suggest that the currently held view, that serum measurement of S100B is a valid "biomarker" of brain damage in traumatic brain injury (TBI), does not acknowlege the multifaceted release pattern and effect of the blood-brain barrier disruption upon S100B levels in serum. In fact, serum and brain S100B levels are poorly correlated, with serum levels dependent primarily on the integrity of the blood-brain barrier, and not the level of S100B in the brain. The time profile of S100B release following experimental TBI, both in vitro and in vivo, suggests a role of S100B in delayed reparative processes. Further, recent findings provide evidence, that S100B may decrease neuronal injury and/or contribute to repair following TBI. Hence, S100B, far from being a negative determinant of outcome, as suggested previously in the human TBI and ischemia literature, is of potential therapeutic value that could improve outcome in patients who sustain various forms of acute brain damage.
Collapse
Affiliation(s)
- Andrea Kleindienst
- Department of Neurosurgery, Georg August University, Göttingen, Germany.
| | | |
Collapse
|
39
|
Kleindienst A, McGinn MJ, Harvey HB, Colello RJ, Hamm RJ, Bullock MR. Enhanced hippocampal neurogenesis by intraventricular S100B infusion is associated with improved cognitive recovery after traumatic brain injury. J Neurotrauma 2005; 22:645-55. [PMID: 15941374 DOI: 10.1089/neu.2005.22.645] [Citation(s) in RCA: 144] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Evidence of injury-induced neurogenesis in the adult hippocampus suggests that an endogenous repair mechanism exists for cognitive dysfunction following traumatic brain injury (TBI). One factor that may be associated with this restoration is S100B, a neurotrophic/mitogenic protein produced by astrocytes, which has been shown to improve memory function. Therefore, we examined whether an intraventricular S100B infusion enhances neurogenesis within the hippocampus following experimental TBI and whether the biological response can be associated with a measurable cognitive improvement. Following lateral fluid percussion or sham injury in male rats (n = 60), we infused S100B (50 ng/h) or vehicle into the lateral ventricle for 7 days using an osmotic micro-pump. Cell proliferation was assessed by injecting the mitotic marker bromodeoxyuridine (BrdU) on day 2 postinjury. Quantification of BrdU-immunoreactive cells in the dentate gyrus revealed an S100B-enhanced proliferation as assessed on day 5 post-injury (p < 0.05), persisting up to 5 weeks (p < 0.05). Using cell-specific markers, we determined the relative numbers of these progenitor cells that became neurons or glia and found that S100B profoundly increased hippocampal neurogenesis 5 weeks after TBI (p < 0.05). Furthermore, spatial learning ability, as assessed by the Morris water maze on day 30-34 post-injury, revealed an improved cognitive performance after S100B infusion (p < 0.05). Collectively, our findings indicate that an intraventricular S100B infusion induces neurogenesis within the hippocampus, which can be associated with an enhanced cognitive function following experimental TBI. These observations provide compelling evidence for the therapeutic potential of S100B in improving functional recovery following TBI.
Collapse
Affiliation(s)
- Andrea Kleindienst
- Department of Neurosurgery, Virginia Commonwealth University Medical Center, Richmond, Virginia, USA.
| | | | | | | | | | | |
Collapse
|
40
|
Kleindienst A, Tolias CM, Corwin FD, Müller C, Marmarou A, Fatouros P, Bullock MR. Assessment of cerebral S100B levels by proton magnetic resonance spectroscopy after lateral fluid-percussion injury in the rat. J Neurosurg 2005; 102:1115-21. [PMID: 16028772 DOI: 10.3171/jns.2005.102.6.1115] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Object. After traumatic brain injury (TBI), S100B protein is released by astrocytes. Furthermore, cerebrospinal fluid (CSF) and serum S100B levels have been correlated to outcome. Given that no data exist about the temporal profile of cerebral S100B levels following TBI and their correlation to serum levels, the authors examined whether proton magnetic resonance (MR) spectroscopy is capable of measuring S100B.
Methods. Results of in vitro proton MR spectroscopy experiments (2.35-tesla magnet, 25 G/cm, point-resolved spatially localized spectroscopy) revealed an S100B-specific peak at 4.5 ppm and confirmed a positive correlation between different S100B concentrations (10 nM–1 µM) and the area under the curve (AUC) for the S100B peak (r = 0.991, p < 0.001). Thereafter, proton MR spectroscopy was performed in male Sprague—Dawley rats (7 × 5 × 5—mm voxel in each hemisphere, TR 3000 msec, TE 30 msec, 256 acquisitions). Exogenously increased CSF S100B levels (∼ 200 ng/ml) through the intraventricular infusion of S100B increased the AUC of the S100B peak from 0.06 ± 0.02 to 0.44 ± 0.06 (p < 0.05), whereas serum S100B levels remained normal. Two hours after lateral fluid-percussion injury, serum S100B levels increased to 0.61 ± 0.09 ng/ml (p < 0.01) and rapidly returned to normal levels, whereas the AUC of the S100B peak increased to 0.19 ± 0.04 at 2 hours postinjury and 0.41 ± 0.07 (p < 0.05) on Day 5 postinjury.
Conclusions. Proton MR spectroscopy proves a strong correlation between the AUC of the S100B peak and S100B concentrations. Following experimental TBI, serum S100B levels increased for only a very short period, whereas cerebral S100B levels were increased up to Day 5 postinjury. Given that experimental data indicate that S100B is actively released following TBI, proton MR spectroscopy may represent a new tool to identify increased cerebral S100B levels in patients after injury, thus allowing its biological function to be better understood.
Collapse
Affiliation(s)
- Andrea Kleindienst
- Department of Neurosurgery, Medical College of Virginia, Virginia Commonwealth University, Richmond, Virginia 23298-0508, USA.
| | | | | | | | | | | | | |
Collapse
|
41
|
Shirasaki Y, Edo N, Sato T. Serum S-100b protein as a biomarker for the assessment of neuroprotectants. Brain Res 2004; 1021:159-66. [PMID: 15342263 DOI: 10.1016/j.brainres.2004.06.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2004] [Indexed: 11/25/2022]
Abstract
The study of biomarkers associated with stroke has proved to be of considerable utility. The astroglial protein S-100b is a candidate marker for cerebral tissue damage. We used a rat embolic model produced by injection of microspheres to demonstrate that serum S-100b is a useful biochemical marker for ischemic brain injury. Serum S-100b levels were significantly increased following microsphere injection, which was closely correlated with the development of brain edema. We found that structurally and mechanistically independent neuroprotective agents, such as 3-[2-[4-(3-chloro-2-methylphenylmethyl)-1-piperazinyl]ethyl]-5,6-dimethoxy-1-(4-imidazolylmethyl)-1H-indazole dihydrochloride 3.5 hydrate (DY-9760e), a novel calmodulin antagonist, and the N-methyl-d-aspartate (NMDA) receptor antagonist MK-801, are capable of attenuating increased serum S-100b levels and brain edema. In contrast, the hyperosmolar agent glycerol, which has no direct neuroprotective action, had little effect on serum S-100b levels, despite a significant decrease in brain water content. These results suggest that lowering of serum S-100b is mediated by neuroprotection against ischemic brain injury. Thus, serum S-100b reflects the extent of brain damage following cerebral ischemia and serves as a useful biomarker for the assessment of neuroprotectants.
Collapse
Affiliation(s)
- Yasufumi Shirasaki
- New Product Research Laboratories II, Daiichi Pharmaceutical Co., Ltd., 1-16-13 Kitakasai 1-Chome, Edogawa-ku, Tokyo 134-8630, Japan.
| | | | | |
Collapse
|
42
|
Kleindienst A, Harvey HB, Rice AC, Müller C, Hamm RJ, Gaab MR, Bullock MR. Intraventricular Infusion of the Neurotrophic Protein S100B Improves Cognitive Recovery after Fluid Percussion Injury in the Rat. J Neurotrauma 2004; 21:541-7. [PMID: 15165362 DOI: 10.1089/089771504774129874] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Elevated serum S100B levels have been shown to be a predictor of poor outcome after traumatic brain injury (TBI). Experimental data, on the other hand, demonstrate a neuroprotective and neurotrophic effect of this calcium-binding protein. The purpose of this study was to examine the role of increased S100B levels on functional outcome after TBI. Following lateral fluid percussion or sham injury in male Sprague Dawley rats (n = 56), we infused S100B (50 ng/h) or vehicle into the cerebrospinal fluid of the ipsilateral ventricle for 7 days using an osmotic mini-pump. Assessment of cognitive performance by the Morris water maze on days 30-34 after injury revealed an improved performance of injured animals after S100B infusion (p < 0.05), when compared to vehicle infusion. Blood samples for analysis of clinical markers of brain damage, S100B and neuron specific enolase, taken at 30 min, 3 h, 4 h, 2 days, or 5 days showed a typical peak 3 h after injury (p < 0.01), and higher serum levels correlated significantly with an impaired cognitive recovery (p < 0.01). The correlation of higher serum S100B levels with poor water maze performance may result from injury induced opening of the blood-brain barrier, allowing the passage of S100B into serum. Thus while higher serum levels of S100B seem to reflect the degree of blood-brain barrier opening and severity of injury, a beneficial effect of intraventricular S100B administration on long-term functional recovery after TBI has been demonstrated for the first time. The exact mechanism by which S100B exerts its neuroprotective or neurotrophic influence remains unknown and needs to be elucidated by further investigation.
Collapse
Affiliation(s)
- Andrea Kleindienst
- Division of Neurosurgery, Medical College of Virginia, Virginia Commonwealth University, Richmond, Virginia 23298-0508, USA.
| | | | | | | | | | | | | |
Collapse
|
43
|
Sahlein DH, Heyer EJ, Rampersad A, Winfree CJ, Solomon RA, Benvenisty AI, Quest DO, Du E, Connolly ES. Failure of intraoperative jugular bulb S-100B and neuron-specific enolase sampling to predict cognitive injury after carotid endarterectomy. Neurosurgery 2004; 53:1243-9 discussion 1249-50. [PMID: 14633290 PMCID: PMC2663381 DOI: 10.1227/01.neu.0000093493.16850.11] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Cognitive decline occurs in 25% of patients after carotid endarterectomy (CEA). Elevated serum concentrations of S-100B and neuron-specific enolase (NSE) occur after stroke, and serum S-100B levels at 24 hours are associated with clinical outcome after both stroke and CEA. We hypothesized that we could detect acute elevations in serum levels of these markers obtained intraoperatively from the jugular bulb (JB) and that these elevations would predict cognitive dysfunction postoperatively as measured by neuropsychometric test performance. METHODS Forty-three patients scheduled for elective CEA were assessed with a battery of neuropsychometric tests before and 1 day after surgery. Before the carotid artery was clamped, a 6-French Fogarty catheter was inserted into the facial vein and threaded 6 cm rostrally into the JB. Serum samples were withdrawn from this catheter and simultaneously from a radial arterial catheter (A-line) at three time points: before clamping, 15 minutes into clamping, and after unclamping the carotid artery. Concentrations between groups were compared by analysis of variance and paired t tests. RESULTS Total deficit scores were significantly worse in 13 (30%) of the 43 patients 1 day after surgery. There was a trend toward elevations in JB concentrations of S-100B relative to A-line levels 15 minutes after cross-clamping (11% elevation, P = 0.079, paired t test). In addition, 15 minutes after clamping of the carotid artery, levels of S-100B from the JB were significantly elevated compared with levels at baseline (P = 0.040, one-way analysis of variance). No significant changes were found between any time point in levels of S-100B from the A-line blood or of NSE from either the JB or the A-line. Subtle cognitive decline after CEA was not correlated with intraoperative levels of S-100B or NSE, but there was a weak, statistically nonsignificant, association between a rise in 15-minute S-100B levels and cognitive injury that was not seen with JB samples. CONCLUSION Although intraoperative levels of S-100B and NSE from the JB failed to predict cognitive injury, carotid cross-clamping, independent of injury, seems to be associated with early elevations in S-100B.
Collapse
Affiliation(s)
- Daniel H Sahlein
- Department of Anesthesiology, Columbia University, New York, New York, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Zahs KR, Kofuji P, Meier C, Dermietzel R. Connexin immunoreactivity in glial cells of the rat retina. J Comp Neurol 2003; 455:531-46. [PMID: 12508325 DOI: 10.1002/cne.10524] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The rat retina contains two types of macroglial cells, Müller cells, radial glial cells that are the principal macroglial cells of vertebrate retinas, and astrocytes associated with the surface vasculature. In addition to the often-described gap-junctional coupling between astrocytes, coupling also occurs between astrocytes and Müller cells. Immunohistochemistry and confocal microscopy were used to identify connexins in the retinas of pigmented rats. Several antibodies directed against connexin43 stained astrocytes, identified using antibodies directed against glial fibrillary acidic protein (GFAP). In addition, two connexin43 antibodies stained Müller cells, identified with antibodies directed against S100 or glutamine synthetase. Connexin30-immunoreactive puncta were confined to the vitreal surface of the retina and colocalized with GFAP-immunoreactive astrocyte processes. Connexin45 immunoreactivity was associated with both astrocytes and Müller cells. We conclude that retinal glial cells express multiple connexins, and the patterns of immunostaining that we observe in this study are consistent with the expression of connexins30, -43, and possibly -45 by astrocytes and the expression of connexins43 and -45 by Müller cells. As gap-junction channels may be formed by both homotypic and heterotypic hemichannels, and the hemichannels may themselves be homomeric or heteromeric, there exists a multitude of possible gap-junction channels that could underlie the homotypic coupling between retinal astrocytes and the heterotypic coupling between astrocytes and Müller cells.
Collapse
Affiliation(s)
- Kathleen R Zahs
- Department of Physiology, University of Minnesota Medical School, Minneapolis, Minnesota 55455, USA.
| | | | | | | |
Collapse
|
45
|
Dyck RH, Bogoch II, Marks A, Melvin NR, Teskey GC. Enhanced epileptogenesis in S100B knockout mice. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2002; 106:22-9. [PMID: 12393261 DOI: 10.1016/s0169-328x(02)00406-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
S100B is a small calcium- and zinc-binding protein expressed by astrocytes in the central nervous system. Here, we examined the role of S100B in epileptogenesis using an amygdala kindling paradigm comparing S100B knockout mice with their wild-type counterparts. Astrocyte activation following kindling, assessed by glial fibrillary acidic protein expression in the hippocampus and amygdala, was similar in wild-type and knockout mice. In addition, wild-type and knockout mice did not have substantially different afterdischarge thresholds. However, knockout mice kindled more rapidly and exhibited more severe seizures. These results implicate normal levels of S100B in the attenuation of epileptogenesis.
Collapse
Affiliation(s)
- Richard H Dyck
- Behavioural Neuroscience Research Group, Department of Psychology, University of Calgary, Alberta, Canada T2N 1N4.
| | | | | | | | | |
Collapse
|
46
|
Nomura H, Furuta A, Iwaki T. Dorsal root rupture injury induces extension of astrocytic processes into the peripheral nervous system and expression of GDNF in astrocytes. Brain Res 2002; 950:21-30. [PMID: 12231225 DOI: 10.1016/s0006-8993(02)02982-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Preganglionic brachial plexus injuries fall into two categories according to the lesion site, root avulsion injury and root rupture injury. The latter type of injury involves part of the peripheral nervous system (PNS) component at the injured spinal cord surface. Previous investigators have used rhizotomy of experimental animals as a model for dorsal root rupture injury. However, the effect on the central nervous system (CNS)-PNS junction accompanied by the mechanical stress from traction force is hard to estimate in this model. The current study aimed to demonstrate temporal molecular alterations from the CNS-PNS junction to the ruptured dorsal root after traction injury by immunohistochemical procedures. At 28 days after dorsal rupture injury, GFAP-positive structures could be clearly identified showing rather straight lines from the centro-peripheral junction toward the peripheral stump in the ruptured dorsal root. Immunoelectron microscopy for GFAP verified GFAP IR within the astrocytic processes at the injured dorsal root at 28 days after dorsal rupture injury. Glial cell line-derived neurotrophic factor immunoreactivity (GDNF IR) was slightly upregulated within the Schwann cell bodies on the injured dorsal root at 24-48 h after rupture injury. However, GDNF IR had appeared showing a process-like profile on the ruptured dorsal root by 28 days, and it was closely related with GFAP-positive structures. In contrast, a small increase in GFAP IR was only detected on the proximal side on the rhizotomized dorsal root at 28 days after rhizotomy. A marked decrease in NF IR and S-100 IR was observed at the ruptured dorsal root from 7 days. On the other hand, laminin IR was strongly upregulated on the ruptured dorsal root from 48 h to 7 days, and was still evident at 28 days. We therefore conclude that the astrocytes show a unique ability to extend their processes toward the stump. This ability may provide a new medium for the study of axonal regeneration in future clinical experiments.
Collapse
Affiliation(s)
- Hiroshi Nomura
- Department of Neuropathology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 812-8582, Fukuoka, Japan.
| | | | | |
Collapse
|
47
|
Nishiyama H, Knopfel T, Endo S, Itohara S. Glial protein S100B modulates long-term neuronal synaptic plasticity. Proc Natl Acad Sci U S A 2002; 99:4037-42. [PMID: 11891290 PMCID: PMC122644 DOI: 10.1073/pnas.052020999] [Citation(s) in RCA: 195] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Glial cells are traditionally regarded as elements for structural support and ionic homeostasis, but have recently attracted attention as putative integral elements of the machinery involved in synaptic transmission and plasticity. Here, we demonstrate that calcium-binding protein S100B, which is synthesized in considerable amounts in astrocytes (a major glial cell subtype), modulates long-term synaptic plasticity. Mutant mice devoid of S100B developed normally and had no detectable abnormalities in the cytoarchitecture of the brain. These mutant mice, however, had strengthened synaptic plasticity as identified by enhanced long-term potentiation (LTP) in the hippocampal CA1 region. Perfusion of hippocampal slices with recombinant S100B proteins reversed the levels of LTP in the mutant slices to those of the wild-type slices, indicating that S100B might act extracellularly. In addition to enhanced LTP, mutant mice had enhanced spatial memory in the Morris water maze test and enhanced fear memory in the contextual fear conditioning. The results indicate that S100B is a glial modulator of neuronal synaptic plasticity and strengthen the notion that glial-neuronal interaction is important for information processing in the brain.
Collapse
Affiliation(s)
- Hiroshi Nishiyama
- Laboratories for Behavioral Genetics and Neuronal Circuit Dynamics, and Neuronal Circuit Mechanisms Research Group, Brain Science Institute (BSI), Institute of Physical and Chemical Research (RIKEN), 2-1 Hirosawa, Wako-shi, Saitama 351-0198, Japan
| | | | | | | |
Collapse
|
48
|
Nishiyama H, Takemura M, Takeda T, Itohara S. Normal development of serotonergic neurons in mice lacking S100B. Neurosci Lett 2002; 321:49-52. [PMID: 11872254 DOI: 10.1016/s0304-3940(01)02549-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
S100B, a glia-derived calcium binding protein, exhibits strong neurite extension activity in cultured serotonergic neurons. Using S100B-knockout mice, we examined whether this protein possesses in vivo serotonergic trophic activity. The distribution of serotonergic fibers, determined by immunohistochemistry, in the brains of S100B-knockout mice was quite similar to that of wild-type mice. Furthermore, the content of serotonin and its metabolite 5-hydroxyindole-3-acetic acid in knockout mice was also indistinguishable from those of wild-type mice. Our findings argue against the hypothesis that S100B has a crucial role in neurite extension of serotonergic neurons.
Collapse
Affiliation(s)
- Hiroshi Nishiyama
- Laboratory for Behavioral Genetics, Brain Science Institute (BSI), RIKEN, 2-1 Hirosawa, Wako-shi, Saitama 351-0198, Japan
| | | | | | | |
Collapse
|
49
|
Peskind ER, Griffin WS, Akama KT, Raskind MA, Van Eldik LJ. Cerebrospinal fluid S100B is elevated in the earlier stages of Alzheimer's disease. Neurochem Int 2001; 39:409-13. [PMID: 11578776 DOI: 10.1016/s0197-0186(01)00048-1] [Citation(s) in RCA: 154] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Postmortem demonstration of increased expression of biologically active S100B in Alzheimer's disease (AD) and its relation to progression of neuropathological changes across the cortical regions suggests involvement of this astrocytic cytokine in the pathophysiology of AD. The hypothesis that the overexpression of S100B in Alzheimer brain is related to the progression of clinical symptoms was addressed in living persons by measuring S100B concentrations in cerebrospinal fluid (CSF) from AD patients with a broad range of clinical dementia severity and from healthy older persons. The effect of normal aging on CSF S100B concentrations also was estimated. CSF S100B did not differ between all 68 AD subjects (0.98+/-0.09 ng/ml (mean+/-S.E.M.)) and 25 healthy older subjects (0.81+/-0.13 ng/ml). When AD subjects were divided into mild/moderate stage and advanced stage clinical dementia severity by the established Clinical Dementia Rating Scale (CDR) criteria, S100B was significantly higher in the 46 mild/moderate stage AD subjects (1.17+/-0.11 ng/ml) than in either the 22 advanced stage AD subjects (0.60+/-0.12 ng/ml) or the healthy older subjects. Consistent with higher CSF S100B in mild to moderate AD, there was a significant correlation among all AD subjects between CSF S100B and cognitive status as measured by the Mini Mental State Exam (MMSE) score. CSF S100B did not differ between healthy older subjects and healthy young subjects. These results suggest increased CNS expression of S100B in the earlier stages of AD, and are consistent with a role for S100B in the initiation and/or facilitation of neuritic plaque formation in AD brain.
Collapse
Affiliation(s)
- E R Peskind
- Mental Illness Research, Education, and Clinical Center, Veterans Affairs Puget Sound Health Care System, and Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA 98108, USA.
| | | | | | | | | |
Collapse
|
50
|
Donato R. S100: a multigenic family of calcium-modulated proteins of the EF-hand type with intracellular and extracellular functional roles. Int J Biochem Cell Biol 2001; 33:637-68. [PMID: 11390274 DOI: 10.1016/s1357-2725(01)00046-2] [Citation(s) in RCA: 1176] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
S100 is a multigenic family of non-ubiquitous Ca(2+)-modulated proteins of the EF-hand type expressed in vertebrates exclusively and implicated in intracellular and extracellular regulatory activities. Within cells, most of S100 members exist in the form of antiparallelly packed homodimers (in some cases heterodimers), capable of functionally crossbridging two homologous or heterologous target proteins in a Ca(2+)-dependent (and, in some instances, Ca(2+)-independent) manner. S100 oligomers can also form, under the non-reducing conditions found in the extracellular space and/or within cells upon changes in the cell redox status. Within cells, S100 proteins have been implicated in the regulation of protein phosphorylation, some enzyme activities, the dynamics of cytoskeleton components, transcription factors, Ca(2+) homeostasis, and cell proliferation and differentiation. Certain S100 members are released into the extracellular space by an unknown mechanism. Extracellular S100 proteins stimulate neuronal survival and/or differentiation and astrocyte proliferation, cause neuronal death via apoptosis, and stimulate (in some cases) or inhibit (in other cases) the activity of inflammatory cells. A cell surface receptor, RAGE, has been identified on inflammatory cells and neurons for S100A12 and S100B, which transduces S100A12 and S100B effects. It is not known whether RAGE is a universal S100 receptor, S100 members interact with other cell surface receptors, or S100 protein interaction with other extracellular factors specifies the biological effects of a given S100 protein on a target cell. The variety of intracellular target proteins of S100 proteins and, in some cases, of a single S100 protein, and the cell specificity of expression of certain S100 members suggest that these proteins might have a role in the fine regulation of effector proteins and/or specific steps of signaling pathways/cellular functions. Future analyses should discriminate between functionally relevant S100 interactions with target proteins and in vitro observations devoid of physiological importance.
Collapse
Affiliation(s)
- R Donato
- Department of Experimental and Biochemical Sciences, Section of Anatomy, University of Perugia, Via del Giochetto, C.P. 81 Succ. 3, 06122, Perugia, Italy.
| |
Collapse
|