1
|
Dean T, Ghaemmaghami J, Corso J, Gallo V. The cortical NG2-glia response to traumatic brain injury. Glia 2023; 71:1164-1175. [PMID: 36692058 PMCID: PMC10404390 DOI: 10.1002/glia.24342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/21/2022] [Accepted: 01/03/2023] [Indexed: 01/25/2023]
Abstract
Traumatic brain injury (TBI) is a significant worldwide cause of morbidity and mortality. A chronic neurologic disease bearing the moniker of "the silent epidemic," TBI currently has no targeted therapies to ameliorate cellular loss or enhance functional recovery. Compared with those of astrocytes, microglia, and peripheral immune cells, the functions and mechanisms of NG2-glia following TBI are far less understood, despite NG2-glia comprising the largest population of regenerative cells in the mature cortex. Here, we synthesize the results from multiple rodent models of TBI, with a focus on cortical NG2-glia proliferation and lineage potential, and propose future avenues for glia researchers to address this unique cell type in TBI. As the molecular mechanisms that regulate NG2-glia regenerative potential are uncovered, we posit that future therapeutic strategies may exploit cortical NG2-glia to augment local cellular recovery following TBI.
Collapse
Affiliation(s)
- Terry Dean
- Center for Neuroscience Research, Children's National Hospital, Washington, District of Columbia, USA
- Division of Critical Care Medicine, Children's National Hospital, Washington, District of Columbia, USA
| | - Javid Ghaemmaghami
- Center for Neuroscience Research, Children's National Hospital, Washington, District of Columbia, USA
| | - John Corso
- Center for Neuroscience Research, Children's National Hospital, Washington, District of Columbia, USA
| | - Vittorio Gallo
- Center for Neuroscience Research, Children's National Hospital, Washington, District of Columbia, USA
| |
Collapse
|
2
|
Tewari BP, Chaunsali L, Prim CE, Sontheimer H. A glial perspective on the extracellular matrix and perineuronal net remodeling in the central nervous system. Front Cell Neurosci 2022; 16:1022754. [PMID: 36339816 PMCID: PMC9630365 DOI: 10.3389/fncel.2022.1022754] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 09/23/2022] [Indexed: 11/18/2022] Open
Abstract
A structural scaffold embedding brain cells and vasculature is known as extracellular matrix (ECM). The physical appearance of ECM in the central nervous system (CNS) ranges from a diffused, homogeneous, amorphous, and nearly omnipresent matrix to highly organized distinct morphologies such as basement membranes and perineuronal nets (PNNs). ECM changes its composition and organization during development, adulthood, aging, and in several CNS pathologies. This spatiotemporal dynamic nature of the ECM and PNNs brings a unique versatility to their functions spanning from neurogenesis, cell migration and differentiation, axonal growth, and pathfinding cues, etc., in the developing brain, to stabilizing synapses, neuromodulation, and being an active partner of tetrapartite synapses in the adult brain. The malleability of ECM and PNNs is governed by both intrinsic and extrinsic factors. Glial cells are among the major extrinsic factors that facilitate the remodeling of ECM and PNN, thereby acting as key regulators of diverse functions of ECM and PNN in health and diseases. In this review, we discuss recent advances in our understanding of PNNs and how glial cells are central to ECM and PNN remodeling in normal and pathological states of the CNS.
Collapse
|
3
|
Bijelić D, Adžić M, Perić M, Reiss G, Milošević M, Andjus PR, Jakovčevski I. Tenascin-C fibronectin D domain is involved in the fine-tuning of glial response to CNS injury in vitro. Front Cell Dev Biol 2022; 10:952208. [PMID: 36092707 PMCID: PMC9462431 DOI: 10.3389/fcell.2022.952208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/18/2022] [Indexed: 11/19/2022] Open
Abstract
Understanding processes that occur after injuries to the central nervous system is essential in order to gain insight into how the restoration of function can be improved. Extracellular glycoprotein tenascin-C (TnC) has numerous functions in wound healing process depending on the expression time, location, isoform and binding partners which makes it interesting to study in this context. We used an in vitro injury model, the mixed culture of cortical astrocytes and microglia, and observed that without TnC microglial cells tend to populate gap area in greater numbers and proliferate more, whereas astrocytes build up in the border region to promote faster gap closure. Alternatively spliced domain of TnC, fibronectin type III-like repeat D (FnD) strongly affected physiological properties and morphology of both astrocytes and microglia in this injury model. The rate of microglial proliferation in the injury region decreased significantly with the addition of FnD. Additionally, density of microglia also decreased, in part due to reduced proliferation, and possibly due to reduced migration and increased contact inhibition between enlarged FnD-treated cells. Overall morphology of FnD-treated microglia resembled the activated pro-inflammatory cells, and elevated expression of iNOS was in accordance with this phenotype. The effect of FnD on astrocytes was different, as it did not affect their proliferation, but stimulated migration of reactivated astrocytes into the scratched area 48 h after the lesion. Elevated expression and secretion of TNF-α and IL-1β upon FnD treatment indicated the onset of inflammation. Furthermore, on Western blots we observed increased intensity of precursor bands of β1 integrin and appearance of monomeric bands of P2Y12R after FnD treatment which substantiates and clarifies its role in cellular shape and motility changes. Our results show versatile functions of TnC and in particular FnD after injury, mostly contributing to ongoing inflammation in the injury region. Based on our findings, FnD might be instrumental in limiting immune cell infiltration, and promoting astrocyte migration within the injury region, thus influencing spaciotemporal organization of the wound and surrounding area.
Collapse
Affiliation(s)
- Dunja Bijelić
- Centre for Laser Microscopy, Institute of Physiology and Biochemistry “Ivan Djaja”, Faculty of Biology, University of Belgrade, Belgrade, Serbia
- *Correspondence: Dunja Bijelić, ; Igor Jakovčevski,
| | - Marija Adžić
- Centre for Laser Microscopy, Institute of Physiology and Biochemistry “Ivan Djaja”, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Mina Perić
- Centre for Laser Microscopy, Institute of Physiology and Biochemistry “Ivan Djaja”, Faculty of Biology, University of Belgrade, Belgrade, Serbia
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Gebhard Reiss
- Institute for Anatomy and Clinical Morphology, University Witten / Herdecke, Witten, Germany
| | - Milena Milošević
- Centre for Laser Microscopy, Institute of Physiology and Biochemistry “Ivan Djaja”, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Pavle R. Andjus
- Centre for Laser Microscopy, Institute of Physiology and Biochemistry “Ivan Djaja”, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Igor Jakovčevski
- Institute for Anatomy and Clinical Morphology, University Witten / Herdecke, Witten, Germany
| |
Collapse
|
4
|
Sanchez-Gonzalez R, Koupourtidou C, Lepko T, Zambusi A, Novoselc KT, Durovic T, Aschenbroich S, Schwarz V, Breunig CT, Straka H, Huttner HB, Irmler M, Beckers J, Wurst W, Zwergal A, Schauer T, Straub T, Czopka T, Trümbach D, Götz M, Stricker SH, Ninkovic J. Innate Immune Pathways Promote Oligodendrocyte Progenitor Cell Recruitment to the Injury Site in Adult Zebrafish Brain. Cells 2022; 11:520. [PMID: 35159329 PMCID: PMC8834209 DOI: 10.3390/cells11030520] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 01/18/2022] [Accepted: 01/18/2022] [Indexed: 01/13/2023] Open
Abstract
The oligodendrocyte progenitors (OPCs) are at the front of the glial reaction to the traumatic brain injury. However, regulatory pathways steering the OPC reaction as well as the role of reactive OPCs remain largely unknown. Here, we compared a long-lasting, exacerbated reaction of OPCs to the adult zebrafish brain injury with a timely restricted OPC activation to identify the specific molecular mechanisms regulating OPC reactivity and their contribution to regeneration. We demonstrated that the influx of the cerebrospinal fluid into the brain parenchyma after injury simultaneously activates the toll-like receptor 2 (Tlr2) and the chemokine receptor 3 (Cxcr3) innate immunity pathways, leading to increased OPC proliferation and thereby exacerbated glial reactivity. These pathways were critical for long-lasting OPC accumulation even after the ablation of microglia and infiltrating monocytes. Importantly, interference with the Tlr1/2 and Cxcr3 pathways after injury alleviated reactive gliosis, increased new neuron recruitment, and improved tissue restoration.
Collapse
Affiliation(s)
- Rosario Sanchez-Gonzalez
- Institute of Stem Cell Research, Helmholtz Center Munich, 85764 Oberschleißheim, Germany; (R.S.-G.); (C.K.); (T.L.); (A.Z.); (K.T.N.); (T.D.); (S.A.); (V.S.); (M.G.)
- Department Biology II, University of Munich, 80539 München, Germany;
| | - Christina Koupourtidou
- Institute of Stem Cell Research, Helmholtz Center Munich, 85764 Oberschleißheim, Germany; (R.S.-G.); (C.K.); (T.L.); (A.Z.); (K.T.N.); (T.D.); (S.A.); (V.S.); (M.G.)
- Biomedical Center (BMC), Division of Cell Biology and Anatomy, Faculty of Medicine, LMU Munich, 80539 München, Germany
- Graduate School Systemic Neurosciences, LMU, 80539 Munich, Germany
| | - Tjasa Lepko
- Institute of Stem Cell Research, Helmholtz Center Munich, 85764 Oberschleißheim, Germany; (R.S.-G.); (C.K.); (T.L.); (A.Z.); (K.T.N.); (T.D.); (S.A.); (V.S.); (M.G.)
- Biomedical Center (BMC), Division of Cell Biology and Anatomy, Faculty of Medicine, LMU Munich, 80539 München, Germany
- Graduate School Systemic Neurosciences, LMU, 80539 Munich, Germany
| | - Alessandro Zambusi
- Institute of Stem Cell Research, Helmholtz Center Munich, 85764 Oberschleißheim, Germany; (R.S.-G.); (C.K.); (T.L.); (A.Z.); (K.T.N.); (T.D.); (S.A.); (V.S.); (M.G.)
- Biomedical Center (BMC), Division of Cell Biology and Anatomy, Faculty of Medicine, LMU Munich, 80539 München, Germany
- Graduate School Systemic Neurosciences, LMU, 80539 Munich, Germany
| | - Klara Tereza Novoselc
- Institute of Stem Cell Research, Helmholtz Center Munich, 85764 Oberschleißheim, Germany; (R.S.-G.); (C.K.); (T.L.); (A.Z.); (K.T.N.); (T.D.); (S.A.); (V.S.); (M.G.)
- Biomedical Center (BMC), Division of Cell Biology and Anatomy, Faculty of Medicine, LMU Munich, 80539 München, Germany
- Graduate School Systemic Neurosciences, LMU, 80539 Munich, Germany
| | - Tamara Durovic
- Institute of Stem Cell Research, Helmholtz Center Munich, 85764 Oberschleißheim, Germany; (R.S.-G.); (C.K.); (T.L.); (A.Z.); (K.T.N.); (T.D.); (S.A.); (V.S.); (M.G.)
- Biomedical Center (BMC), Division of Cell Biology and Anatomy, Faculty of Medicine, LMU Munich, 80539 München, Germany
- Graduate School Systemic Neurosciences, LMU, 80539 Munich, Germany
| | - Sven Aschenbroich
- Institute of Stem Cell Research, Helmholtz Center Munich, 85764 Oberschleißheim, Germany; (R.S.-G.); (C.K.); (T.L.); (A.Z.); (K.T.N.); (T.D.); (S.A.); (V.S.); (M.G.)
- Biomedical Center (BMC), Division of Cell Biology and Anatomy, Faculty of Medicine, LMU Munich, 80539 München, Germany
- Graduate School Systemic Neurosciences, LMU, 80539 Munich, Germany
| | - Veronika Schwarz
- Institute of Stem Cell Research, Helmholtz Center Munich, 85764 Oberschleißheim, Germany; (R.S.-G.); (C.K.); (T.L.); (A.Z.); (K.T.N.); (T.D.); (S.A.); (V.S.); (M.G.)
- Biomedical Center (BMC), Division of Cell Biology and Anatomy, Faculty of Medicine, LMU Munich, 80539 München, Germany
- Graduate School Systemic Neurosciences, LMU, 80539 Munich, Germany
| | - Christopher T. Breunig
- Reprogramming and Regeneration, Biomedical Center (BMC), Physiological Genomics, Faculty of Medicine, LMU Munich, 80539 München, Germany; (C.T.B.); (S.H.S.)
- Epigenetic Engineering, Institute of Stem Cell Research, Helmholtz Center Munich, 85764 Oberschleißheim, Germany
| | - Hans Straka
- Department Biology II, University of Munich, 80539 München, Germany;
| | - Hagen B. Huttner
- Department of Neurology, Justus-Liebig-University Giessen, Klinikstrasse 33, 35392 Giessen, Germany;
| | - Martin Irmler
- Institute of Experimental Genetics, Helmholtz Center Munich, 85764 Oberschleißheim, Germany; (M.I.); (J.B.)
| | - Johannes Beckers
- Institute of Experimental Genetics, Helmholtz Center Munich, 85764 Oberschleißheim, Germany; (M.I.); (J.B.)
- German Center for Diabetes Research (DZD e.V.), 85764 Neuherberg, Germany
- Chair of Experimental Genetics, School of Life Sciences Weihenstephan, Technical University Munich, 80333 München, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Center Munich, 85764 Oberschleißheim, Germany; (W.W.); (D.T.)
- Munich Cluster for Systems Neurology SYNERGY, LMU, 80539 Munich, Germany
- Chair of Developmental Genetics c/o Helmholtz Zentrum München, School of Life Sciences Weihenstephan, Technical University Munich, 80333 München, Germany
- German Center for Neurodegenerative Diseases (DZNE), Site Munich, 80539 Munich, Germany
| | - Andreas Zwergal
- Department of Neurology, Ludwig-Maximilians University, Campus Grosshadern, 81377 Munich, Germany;
| | - Tamas Schauer
- Biomedical Center (BMC), Bioinformatic Core Facility, Faculty of Medicine, LMU Munich, 80539 München, Germany; (T.S.); (T.S.)
| | - Tobias Straub
- Biomedical Center (BMC), Bioinformatic Core Facility, Faculty of Medicine, LMU Munich, 80539 München, Germany; (T.S.); (T.S.)
| | - Tim Czopka
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH8 9YL, UK;
| | - Dietrich Trümbach
- Institute of Developmental Genetics, Helmholtz Center Munich, 85764 Oberschleißheim, Germany; (W.W.); (D.T.)
| | - Magdalena Götz
- Institute of Stem Cell Research, Helmholtz Center Munich, 85764 Oberschleißheim, Germany; (R.S.-G.); (C.K.); (T.L.); (A.Z.); (K.T.N.); (T.D.); (S.A.); (V.S.); (M.G.)
- Munich Cluster for Systems Neurology SYNERGY, LMU, 80539 Munich, Germany
- Biomedical Center (BMC), Division of Physiological Genomics, Faculty of Medicine, LMU Munich, 80539 München, Germany
| | - Stefan H. Stricker
- Reprogramming and Regeneration, Biomedical Center (BMC), Physiological Genomics, Faculty of Medicine, LMU Munich, 80539 München, Germany; (C.T.B.); (S.H.S.)
- Epigenetic Engineering, Institute of Stem Cell Research, Helmholtz Center Munich, 85764 Oberschleißheim, Germany
| | - Jovica Ninkovic
- Institute of Stem Cell Research, Helmholtz Center Munich, 85764 Oberschleißheim, Germany; (R.S.-G.); (C.K.); (T.L.); (A.Z.); (K.T.N.); (T.D.); (S.A.); (V.S.); (M.G.)
- Biomedical Center (BMC), Division of Cell Biology and Anatomy, Faculty of Medicine, LMU Munich, 80539 München, Germany
- Munich Cluster for Systems Neurology SYNERGY, LMU, 80539 Munich, Germany
| |
Collapse
|
5
|
MicroRNA-488 inhibits neural inflammation and apoptosis in spinal cord injury through restraint on the HMGB1/TLR4/NF-κB signaling pathway. Neuroreport 2021; 32:1017-1026. [PMID: 34102644 DOI: 10.1097/wnr.0000000000001680] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Secondary spinal cord injury (SCI), a reversible pathological change, involves neural inflammation and apoptosis. This study explored how microRNA (miR)-488, an inflammatory regulator as reported affected secondary SCI. METHODS In vivo, Wistar rats were clipped on the spinal cord for SCI induction. In vitro, PC-12 cells were treated with lipopolysaccharide (LPS) to induce cell injuries to mimic the environment during the secondary SCI. Cell viability and apoptosis were measured by CCK-8 assay and flow cytometry. The levels of inflammation-related factors (interleukin (IL)-6, IL-1β and tumor necrosis factor (TNF)-α) in the serum and PC-12 cells were determined by ELISA. The expressions of miR-488, high mobility group box 1 (HMGB1), B-cell lymphoma 2 (Bcl-2), Bcl-2-associated X protein (Bax), cleaved caspase-3, toll-like receptor 4 (TLR4), phosphorylated (p)-p65 and total-p65 in rat spinal cord or PC-12 cells were analyzed by quantitative reverse transcription PCR or western blot. RESULTS After SCI induction, rats exhibited low Basso-Beattie-Bresnahan scores, promoted the release of inflammation-related factors and downregulated miR-488. LPS treatment decreased cell viability, enhanced apoptosis and downregulated miR-488. Upregulating miR-488 neutralized LPS-induced releases of inflammation-related factors and expressions of Bax and cleaved caspase-3 and counteracted LPS-induced inhibition on Bcl-2 expression. MiR-488 directly targeted HMGB1 and miR-488 mimic decreased LPS-induced HMGB1 expression. Overexpressing HMGB1 counteracted miR-488 mimic-induced decreases in the expressions of TLR4 and p-p65 and the ratio of p-p65 to Total-p65 in LPS-treated PC-12 cells. CONCLUSION MiR-488 inhibited neural inflammation and apoptosis in SCI via its binding with HMGB1-mediated restraint on the TLR4/NF-κB signaling pathway.
Collapse
|
6
|
The Long Non-coding RNA NEAT1/miR-224-5p/IL-33 Axis Modulates Macrophage M2a Polarization and A1 Astrocyte Activation. Mol Neurobiol 2021; 58:4506-4519. [PMID: 34076838 DOI: 10.1007/s12035-021-02405-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 04/26/2021] [Indexed: 02/07/2023]
Abstract
To identify potential regulators and investigate the molecular mechanism of macrophage polarization affecting astrocyte activation from the perspective of non-coding RNA regulation, we isolated mouse bone marrow mononuclear cells (BMMNCs)-induced macrophages toward M1 or M2a polarization. Long non-coding RNA NEAT1 and IL-33 expression levels were significantly upregulated in M2a macrophages; NEAT1 knockdown in M2a macrophages markedly reduced the protein levels of IL-33 and M2a markers, IL-4 and IL-13 concentrations, and the bacterial killing capacity of M2a macrophages. NEAT1 acted as a competing endogenous RNA (ceRNA) to regulate IL-33 expression by sponging miR-224-5p in M2a macrophages; NEAT1 knockdown upregulated miR-224-5p expression, while miR-224-5p inhibition increased the protein content and concentration of IL-33. miR-224-5p inhibition exerted the opposite effects on the protein levels of IL-33 and M2a markers, IL-4 and IL-13 concentrations, and the bacterial killing capacity of M2a macrophages compared to NEAT1 knockdown; the effects of NEAT1 knockdown were significantly reversed by miR-224-5p inhibition. M2a macrophage conditioned medium (CM) significantly suppressed the activation of A1 astrocytes. NEAT1 knockdown M2a macrophage CM led to enhanced A1 astrocyte activation while miR-224-5p-silenced M2a macrophage CM led to a blockade of A1 astrocyte activation; the effects of NEAT1 knockdown M2a macrophage CM on A1 astrocyte activation were significantly reversed by miR-224-5p inhibition in M2a macrophages. The NEAT1/miR-224-5p/IL-33 axis modulates macrophage M2a polarization, therefore affecting A1 astrocyte activation.
Collapse
|
7
|
von Streitberg A, Jäkel S, Eugenin von Bernhardi J, Straube C, Buggenthin F, Marr C, Dimou L. NG2-Glia Transiently Overcome Their Homeostatic Network and Contribute to Wound Closure After Brain Injury. Front Cell Dev Biol 2021; 9:662056. [PMID: 34012966 PMCID: PMC8128074 DOI: 10.3389/fcell.2021.662056] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 03/12/2021] [Indexed: 12/27/2022] Open
Abstract
In the adult brain, NG2-glia represent a cell population that responds to injury. To further investigate if, how and why NG2-glia are recruited to the injury site, we analyzed in detail the long-term reaction of NG2-glia after a lesion by time-lapse two-photon in vivo microscopy. Live imaging over several weeks of GFP-labeled NG2-glia in the stab wounded cerebral cortex revealed their fast and heterogeneous reaction, including proliferation, migration, polarization, hypertrophy, or a mixed response, while a small subset of cells remained unresponsive. At the peak of the reaction, 2-4 days after the injury, NG2-glia accumulated around and within the lesion core, overcoming the homeostatic control of their density, which normalized back to physiological conditions only 4 weeks after the insult. Genetic ablation of proliferating NG2-glia demonstrated that this accumulation contributed beneficially to wound closure. Thus, NG2-glia show a fast response to traumatic brain injury (TBI) and participate in tissue repair.
Collapse
Affiliation(s)
- Axel von Streitberg
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Sarah Jäkel
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Jaime Eugenin von Bernhardi
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University Munich, Munich, Germany.,Molecular and Translational Neuroscience, Department of Neurology, Ulm University, Ulm, Germany
| | - Christoph Straube
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Felix Buggenthin
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Carsten Marr
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Leda Dimou
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University Munich, Munich, Germany.,Molecular and Translational Neuroscience, Department of Neurology, Ulm University, Ulm, Germany
| |
Collapse
|
8
|
Yang H, An J, Choi I, Lee K, Park SM, Jou I, Joe EH. Region-specific astrogliosis: differential vessel formation contributes to different patterns of astrogliosis in the cortex and striatum. Mol Brain 2020; 13:103. [PMID: 32698847 PMCID: PMC7374828 DOI: 10.1186/s13041-020-00642-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 07/14/2020] [Indexed: 11/10/2022] Open
Abstract
Brain injury causes astrocytes to become reactive (astrogliosis). In this study, we compared astrogliosis in acutely injured cortex and striatum of adult FVB/N mice induced by stereotaxic injection of ATP, a component of danger-associated molecular patterns (DAMPs). Interestingly, MR analysis showed that same amount of ATP induced smaller damage in the cortex than in the striatum. However, in histological analysis, thick and dense scar-like astrogliosis was found in the injured cortex near meninges within 2 wk., but not in other regions, including the striatum and even the cortex near the corpus callosum for up to 30 d. There was little regional difference in the number of Ki67(+)-proliferating astrocytes or mRNA expression of inflammatory cytokines. The most prominent difference between regions with and without scar-like astrogliosis was blood vessel formation. Blood vessels highly expressing collagen 1A1 formed densely near meninges, and astrocytes converged on them. In other regions, however, both blood vessels and astrocytes were relatively evenly distributed. Consistent with this, inhibition of blood vessel formation with the vascular endothelial growth factor (VEGF)-blocking antibody, Avastin, attenuated scar-like astrogliosis near meninges. These results indicate that region-specific astrogliosis occurs following brain injury, and that blood vessel formation plays a critical role in scar formation.
Collapse
Affiliation(s)
- Haijie Yang
- Department of Pharmacology/Neuroscience Graduate Program, National Research Lab of Brain Inflammation, Ajou University School of Medicine, Worldcup-ro 164, Youngtong-gu, Suwon, Kyunggi-do, 16499, South Korea.,Department of Brain Science, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, South Korea
| | - Jiawei An
- Department of Pharmacology/Neuroscience Graduate Program, National Research Lab of Brain Inflammation, Ajou University School of Medicine, Worldcup-ro 164, Youngtong-gu, Suwon, Kyunggi-do, 16499, South Korea.,Department of Brain Science, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, South Korea.,Department of Biomedical Sciences, Neuroscience Graduate Program, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, South Korea
| | - Insup Choi
- Department of Pharmacology/Neuroscience Graduate Program, National Research Lab of Brain Inflammation, Ajou University School of Medicine, Worldcup-ro 164, Youngtong-gu, Suwon, Kyunggi-do, 16499, South Korea.,Department of Brain Science, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, South Korea.,Department of Biomedical Sciences, Neuroscience Graduate Program, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, South Korea
| | - Kihwang Lee
- Department of Ophthalmology, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, South Korea
| | - Sang-Myun Park
- Department of Pharmacology/Neuroscience Graduate Program, National Research Lab of Brain Inflammation, Ajou University School of Medicine, Worldcup-ro 164, Youngtong-gu, Suwon, Kyunggi-do, 16499, South Korea.,Department of Biomedical Sciences, Neuroscience Graduate Program, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, South Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, South Korea
| | - Ilo Jou
- Department of Pharmacology/Neuroscience Graduate Program, National Research Lab of Brain Inflammation, Ajou University School of Medicine, Worldcup-ro 164, Youngtong-gu, Suwon, Kyunggi-do, 16499, South Korea.,Department of Biomedical Sciences, Neuroscience Graduate Program, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, South Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, South Korea
| | - Eun-Hye Joe
- Department of Pharmacology/Neuroscience Graduate Program, National Research Lab of Brain Inflammation, Ajou University School of Medicine, Worldcup-ro 164, Youngtong-gu, Suwon, Kyunggi-do, 16499, South Korea. .,Department of Brain Science, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, South Korea. .,Department of Biomedical Sciences, Neuroscience Graduate Program, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, South Korea. .,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, South Korea.
| |
Collapse
|
9
|
Zhong K, Wang RX, Qian XD, Yu P, Zhu XY, Zhang Q, Ye YL. Neuroprotective effects of saffron on the late cerebral ischemia injury through inhibiting astrogliosis and glial scar formation in rats. Biomed Pharmacother 2020; 126:110041. [PMID: 32113053 DOI: 10.1016/j.biopha.2020.110041] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 02/18/2020] [Accepted: 02/19/2020] [Indexed: 12/11/2022] Open
Abstract
This study is to explore the neuroprotective effects and involved glial scar of saffron (Crocus sativus L.) on the late cerebral ischemia in rats. Focal cerebral ischemia was induced by middle cerebral artery occlusion (MCAO) in Sprague Dawley rats that were randomly divided into sham group, MCAO group, edaravone group (as a positive control) and saffron groups (saffron extract 30, 100, 300 mg/kg). Saffron was administered orally at 2 h at the first day and once daily from day 2 to 42 after ischemia. Behavioral changes were detected from day 43 to 46 after ischemia to evaluate the effects of saffron. Infarct volume, survival neuron density, activated astrocyte, and the thickness of glial scar were also detected. GFAP, neurocan, phosphocan, neurofilament expressions and inflammatory cytokine contents were detected by Western-blotting and ELISA methods, respectively. Saffron improved the body weight loss, neurological deficit and spontaneous activity. It also ameliorated anxiety-like state and cognitive dysfunction, which were detected by elevated plus maze (EPM), marble burying test (MBT) and novel object recognition test (NORT). Toluidine blue staining found that saffron treatment decreased the infarct volume and increased the neuron density in cortex in the ischemic boundary zone. The activated astrocyte number and the thickness of glial scar in the penumbra zone reduced after saffron treatment. Additionally, saffron decreased the contents of IL-6 and IL-1β, increased the content of IL-10 in the ischemic boundary zone. GFAP, neurocan, and phosphocan expressions in ischemic boundary zone and ischemic core zone all decreased after saffron treatment. Saffron exerted neuroprotective effects on late cerebral ischemia, associating with attenuating astrogliosis and glial scar formation after ischemic injury.
Collapse
Affiliation(s)
- Kai Zhong
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Rou-Xin Wang
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | | | - Ping Yu
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xin-Ying Zhu
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Qi Zhang
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yi-Lu Ye
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
10
|
Ji YR, Homaeigohar S, Wang YH, Lin C, Su TY, Cheng CC, Yang SH, Young TH. Selective Regulation of Neurons, Glial Cells, and Neural Stem/Precursor Cells by Poly(allylguanidine)-Coated Surfaces. ACS APPLIED MATERIALS & INTERFACES 2019; 11:48381-48392. [PMID: 31845571 DOI: 10.1021/acsami.9b17143] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Poly(allylguanidine) (PAG) was synthesized and characterized as a polycationic coating material for culturing neurons, glial cells, and neural stem/precursor cells (NSPCs) to apply PAG for neural tissue engineering. For comparison, poly-d-lysine (PDL), the golden benchmark of the neuron cell culture system, was also used in this study. When PAG was subjected to a mixed culture of neurons and glial cells, cell adhesion and neurite extension of neuronal cells were clearly observed but only few glial cells could be found alongside the neurons. Compared to PDL, the significantly lower density of the glial fibrillary acidic protein-positive cells implied that PAG suppressed the glial cell development. Likewise, PAG was demonstrated to dominate the differentiation of NSPCs principally into neurons. To investigate whether the different effects of PAG and PDL on neuron and glial cell behaviors resulted from the difference of guanidinium cations and ammonium cations, poly-l-arginine (PLA) was included and compared in this study. Similar to PDL, PLA supported high neuron and glial cell viability simultaneously. Consequently, glial cell growth and viability restrained on PAG was not only affected by the side-chain guanidino groups but also by the backbone structure property. The absence of the peptide structure in the backbone of PAG and the conformation of coated PAG on tissue culture polystyrene possibly determined the polycationic biomaterial to limit the growth of glial cells.
Collapse
Affiliation(s)
- You-Ren Ji
- Department of Biomedical Engineering , National Taiwan University , Taipei 100 , Taiwan
| | - Shahin Homaeigohar
- Nanochemistry and Nanoengineering, School of Chemical Engineering, Department of Chemistry and Materials Science , Aalto University , Kemistintie 1 , 00076 Aalto , Finland
| | - Yu-Hsin Wang
- Department of Biomedical Engineering , National Taiwan University , Taipei 100 , Taiwan
| | - Chen Lin
- Department of Biomedical Engineering , National Taiwan University , Taipei 100 , Taiwan
| | - Tai-Yuan Su
- Department of Electrical Engineering , Yuan-Ze University , Taoyuan 320 , Taiwan
| | - Ching-Chia Cheng
- Department of Biomedical Engineering , National Taiwan University , Taipei 100 , Taiwan
| | | | - Tai-Horng Young
- Department of Biomedical Engineering , National Taiwan University , Taipei 100 , Taiwan
| |
Collapse
|
11
|
García-Bernal F, Geribaldi-Doldán N, Domínguez-García S, Carrasco M, Murillo-Carretero M, Delgado-Ariza A, Díez-Salguero M, Verástegui C, Castro C. Protein Kinase C Inhibition Mediates Neuroblast Enrichment in Mechanical Brain Injuries. Front Cell Neurosci 2018; 12:462. [PMID: 30542270 PMCID: PMC6277931 DOI: 10.3389/fncel.2018.00462] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 11/13/2018] [Indexed: 01/08/2023] Open
Abstract
Brain injuries of different etiologies lead to irreversible neuronal loss and persisting neuronal deficits. New therapeutic strategies are emerging to compensate neuronal damage upon brain injury. Some of these strategies focus on enhancing endogenous generation of neurons from neural stem cells (NSCs) to substitute the dying neurons. However, the capacity of the injured brain to produce new neurons is limited, especially in cases of extensive injury. This reduced neurogenesis is a consequence of the effect of signaling molecules released in response to inflammation, which act on intracellular pathways, favoring gliogenesis and preventing recruitment of neuroblasts from neurogenic regions. Protein kinase C (PKC) is a family of intracellular kinases involved in several of these gliogenic signaling pathways. The aim of this study was to analyze the role of PKC isozymes in the generation of neurons from neural progenitor cells (NPCs) in vitro and in vivo in brain injuries. PKC inhibition in vitro, in cultures of NPC isolated from the subventricular zone (SVZ) of postnatal mice, leads differentiation towards a neuronal fate. This effect is not mediated by classical or atypical PKC. On the contrary, this effect is mediated by novel PKCε, which is abundantly expressed in NPC cultures under differentiation conditions. PKCε inhibition by siRNA promotes neuronal differentiation and reduces glial cell differentiation. On the contrary, inhibition of PKCθ exerts a small anti-gliogenic effect and reverts the effect of PKCε inhibition on neuronal differentiation when both siRNAs are used in combination. Interestingly, in cortical brain injuries we have found expression of almost all PKC isozymes found in vitro. Inhibition of PKC activity in this type of injuries leads to neuronal production. In conclusion, these findings show an effect of PKCε in the generation of neurons from NPC in vitro, and they highlight the role of PKC isozymes as targets to produce neurons in brain lesions.
Collapse
Affiliation(s)
- Francisco García-Bernal
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigación e Innovación en Biomedicina de Cádiz (INIBICA), Universidad de Cadiz, Cádiz, Spain
| | - Noelia Geribaldi-Doldán
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigación e Innovación en Biomedicina de Cádiz (INIBICA), Universidad de Cadiz, Cádiz, Spain
| | - Samuel Domínguez-García
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigación e Innovación en Biomedicina de Cádiz (INIBICA), Universidad de Cadiz, Cádiz, Spain
| | - Manuel Carrasco
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigación e Innovación en Biomedicina de Cádiz (INIBICA), Universidad de Cadiz, Cádiz, Spain
| | - Maribel Murillo-Carretero
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigación e Innovación en Biomedicina de Cádiz (INIBICA), Universidad de Cadiz, Cádiz, Spain
| | | | - Mónica Díez-Salguero
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigación e Innovación en Biomedicina de Cádiz (INIBICA), Universidad de Cadiz, Cádiz, Spain
| | - Cristina Verástegui
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain.,Departamento de Anatomía, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain
| | - Carmen Castro
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigación e Innovación en Biomedicina de Cádiz (INIBICA), Universidad de Cadiz, Cádiz, Spain
| |
Collapse
|
12
|
Geribaldi-Doldán N, Carrasco M, Murillo-Carretero M, Domínguez-García S, García-Cózar FJ, Muñoz-Miranda JP, Del Río-García V, Verástegui C, Castro C. Specific inhibition of ADAM17/TACE promotes neurogenesis in the injured motor cortex. Cell Death Dis 2018; 9:862. [PMID: 30154402 PMCID: PMC6113335 DOI: 10.1038/s41419-018-0913-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 07/03/2018] [Accepted: 07/25/2018] [Indexed: 11/12/2022]
Abstract
Brain injuries in the adult mammalian brain are accompanied by a fast neurogenic response inside neurogenic niches. However, this response does not contribute to the generation of new neurons within damaged tissues like the cerebral cortex, which are essentially non-neurogenic. This occurs because injuries create a hostile environment that favors gliogenesis. Overexpression and sequential activation of the ADAM17/TGFα/EGFR signaling cascade are crucial for the generation of this gliogenic/non-neurogenic environment. Here, we demonstrate that chronic local infusion of a general metalloprotease inhibitor in areas of traumatic cortical injury in adult mice moderately increased the number of neuroblasts around the lesion, by facilitating the survival of neuroblasts and undifferentiated progenitors, which had migrated to the perilesional area from the subventricular zone. Next, we generated a dominant-negative version of ADAM17 metalloprotease, consisting of a truncated protein containing only the pro-domain (ADAM17-Pro). Specific inhibition of ADAM17 activity by ADAM17-Pro overexpression increased the generation of new neurons in vitro. Local overexpression of ADAM17-Pro in injured cortex in vivo, mediated by lentiviral vectors, dramatically increased the number of neuroblasts observed at the lesion 14 days after injury. Those neuroblasts were able to differentiate into cholinergic and GABAergic neurons 28 days after injury. We conclude that ADAM17 is a putative target to develop new therapeutic tools for the treatment of traumatic brain injury.
Collapse
Affiliation(s)
- Noelia Geribaldi-Doldán
- Área de Fisiología, Facultad de Medicina and Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), Universidad de Cádiz, Cádiz, Spain
| | - Manuel Carrasco
- Área de Fisiología, Facultad de Medicina and Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), Universidad de Cádiz, Cádiz, Spain
| | - Maribel Murillo-Carretero
- Área de Fisiología, Facultad de Medicina and Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), Universidad de Cádiz, Cádiz, Spain
| | - Samuel Domínguez-García
- Área de Fisiología, Facultad de Medicina and Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), Universidad de Cádiz, Cádiz, Spain
| | - Francisco J García-Cózar
- Área de Inmunología, Facultad de Medicina and Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), Universidad de Cádiz, Cádiz, Spain
| | - Juan Pedro Muñoz-Miranda
- Área de Inmunología, Facultad de Medicina and Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), Universidad de Cádiz, Cádiz, Spain
| | - Valme Del Río-García
- Área de Fisiología, Facultad de Medicina and Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), Universidad de Cádiz, Cádiz, Spain
| | - Cristina Verástegui
- Departamento de Anatomía, Facultad de Medicina and Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), Universidad de Cádiz, Cádiz, Spain
| | - Carmen Castro
- Área de Fisiología, Facultad de Medicina and Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), Universidad de Cádiz, Cádiz, Spain.
| |
Collapse
|
13
|
Ye J, Qin Y, Tang Y, Ma M, Wang P, Huang L, Yang R, Chen K, Chai C, Wu Y, Shen H. Methylprednisolone inhibits the proliferation of endogenous neural stem cells in nonhuman primates with spinal cord injury. J Neurosurg Spine 2018; 29:199-207. [DOI: 10.3171/2017.12.spine17669] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVEThe aim of this work was to investigate the effects of methylprednisolone on the proliferation of endogenous neural stem cells (ENSCs) in nonhuman primates with spinal cord injury (SCI).METHODSA total of 14 healthy cynomolgus monkeys (Macaca fascicularis) (4–5 years of age) were randomly divided into 3 groups: the control group (n = 6), SCI group (n = 6), and methylprednisolone therapy group (n = 2). Only laminectomy was performed in the control animals at T-10. SCI was induced in monkeys using Allen’s weight-drop method (50 mm × 50 g) to injure the posterior portion of the spinal cord at T-10. In the methylprednisolone therapy group, monkeys were intravenously infused with methylprednisolone (30 mg/kg) immediately after SCI. All animals were intravenously infused with 5-bromo-2-deoxyuridine (BrdU) (50 mg/kg/day) for 3 days prior to study end point. The small intestine was dissected for immunohistochemical examination. After 3, 7, and 14 days, the spinal cord segments of the control and SCI groups were dissected to prepare frozen and paraffin sections. The proliferation of ENSCs was evaluated using BrdU and nestin immunofluorescence staining.RESULTSHistological examination showed that a larger number of mucosa epithelial cells in the small intestine of all groups were BrdU positive. Nestin-positive ependymal cells are increased around the central canal after SCI. After 3, 7, and 14 days of SCI, BrdU-positive ependymal cells in the SCI group were significantly increased compared with the control group, and the percentage of BrdU-positive cells in the left/right ventral horns and dorsal horn was significantly higher than that of the control group. Seven days after SCI, the percentages of both BrdU-positive ependymal cells around the central canal and BrdU– and nestin–double positive cells in the left/right ventral horns and dorsal horn were significantly lower in the methylprednisolone therapy group than in the SCI group.CONCLUSIONSWhile ENSCs proliferate significantly after SCI in nonhuman primates, methylprednisolone can inhibit the proliferation of ependymal cells after SCI.
Collapse
Affiliation(s)
- Jichao Ye
- 1Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
| | - Yi Qin
- 2Department of Orthopedics, Zhuhai People’s Hospital; and
| | - Yong Tang
- 1Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
| | - Mengjun Ma
- 1Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
| | - Peng Wang
- 1Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
| | - Lin Huang
- 1Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
| | - Rui Yang
- 1Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
| | - Keng Chen
- 1Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
| | - Chaopeng Chai
- 1Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
| | - Yanfeng Wu
- 3Biotherapy Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Huiyong Shen
- 1Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
| |
Collapse
|
14
|
Environmental enrichment reduces brain damage in hydrocephalic immature rats. Childs Nerv Syst 2017; 33:921-931. [PMID: 28382436 DOI: 10.1007/s00381-017-3403-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 03/27/2017] [Indexed: 01/12/2023]
Abstract
PURPOSE We investigate the effects of environmental enrichment (EE) on morphological alterations in different brain structures of pup rats submitted to hydrocephalus condition. METHODS Hydrocephalus was induced in 7-day-old pup rats by injection of 20% kaolin into the cisterna magna. Ventricular dilatation and magnetization transfer to analyze myelin were assessed by magnetic resonance. Hydrocephalic and control rats exposed to EE (n = 10 per group) were housed in cages with a tunnel, ramp, and colored plastic balls that would emit sound when touched. The walls of the housing were decorated with colored adhesive tape. Moreover, tactile and auditory stimulation was performed daily throughout the experiment. Hydrocephalic and control rats not exposed to EE (n = 10 per group) were allocated singly in standard cages. All animals were weighed daily and exposed to open-field conditions every 2 days until the end of the experiment when they were sacrificed and the brains removed for histology and immunohistochemistry. Solochrome cyanine staining was performed to assess the thickness of the corpus callosum. The glial fibrillary acidic protein method was used to evaluate reactive astrocytes, and the Ki67 method to assess cellular proliferation in the subventricular zone. RESULTS The hydrocephalic animals exposed to EE showed better performance in Open Field tests (p < 0.05), while presenting lower weight gain. In addition, these animals showed better myelination as revealed by magnetization transfer (p < 0.05). Finally, the EE group showed a reduction in reactive astrocytes by means of glial fibrillary acidic protein immunostaining and preservation of the proliferation potential of progenitor cells. CONCLUSION The results suggest that EE can protect the developing brain against damaging effects caused by hydrocephalus.
Collapse
|
15
|
Whetstone WD, Walker B, Trivedi A, Lee S, Noble-Haeusslein LJ, Hsu JYC. Protease-Activated Receptor-1 Supports Locomotor Recovery by Biased Agonist Activated Protein C after Contusive Spinal Cord Injury. PLoS One 2017; 12:e0170512. [PMID: 28122028 PMCID: PMC5266300 DOI: 10.1371/journal.pone.0170512] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 01/05/2017] [Indexed: 12/11/2022] Open
Abstract
Thrombin-induced secondary injury is mediated through its receptor, protease activated receptor-1 (PAR-1), by "biased agonism." Activated protein C (APC) acts through the same PAR-1 receptor but functions as an anti-coagulant and anti-inflammatory protein, which counteracts many of the effects of thrombin. Although the working mechanism of PAR-1 is becoming clear, the functional role of PAR-1 and its correlation with APC in the injured spinal cord remains to be elucidated. Here we investigated if PAR-1 and APC are determinants of long-term functional recovery after a spinal cord contusive injury using PAR-1 null and wild-type mice. We found that neutrophil infiltration and disruption of the blood-spinal cord barrier were significantly reduced in spinal cord injured PAR-1 null mice relative to the wild-type group. Both locomotor recovery and ability to descend an inclined grid were significantly improved in the PAR-1 null group 42 days after injury and this improvement was associated with greater long-term sparing of white matter and a reduction in glial scarring. Wild-type mice treated with APC acutely after injury showed a similar level of improved locomotor recovery to that of PAR-1 null mice. However, improvement of APC-treated PAR-1 null mice was indistinguishable from that of vehicle-treated PAR-1 null mice, suggesting that APC acts through PAR-1. Collectively, our findings define a detrimental role of thrombin-activated PAR-1 in wound healing and further validate APC, also acting through the PAR-1 by biased agonism, as a promising therapeutic target for spinal cord injury.
Collapse
Affiliation(s)
- William D. Whetstone
- Department of Emergency Medicine, University of California, San Francisco, California, United States of America
| | - Breset Walker
- Department of Neurological Surgery, University of California, San Francisco, California, United States of America
| | - Alpa Trivedi
- Department of Neurological Surgery, University of California, San Francisco, California, United States of America
| | - Sangmi Lee
- Department of Neurological Surgery, University of California, San Francisco, California, United States of America
| | - Linda J. Noble-Haeusslein
- Department of Neurological Surgery, University of California, San Francisco, California, United States of America
- Department of Physical Therapy and Rehabilitation Science, University of California, San Francisco, California, United States of America
| | - Jung-Yu C. Hsu
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| |
Collapse
|
16
|
Fernandez-Castaneda A, Gaultier A. Adult oligodendrocyte progenitor cells - Multifaceted regulators of the CNS in health and disease. Brain Behav Immun 2016; 57:1-7. [PMID: 26796621 PMCID: PMC4940337 DOI: 10.1016/j.bbi.2016.01.005] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 12/28/2015] [Accepted: 01/11/2016] [Indexed: 01/17/2023] Open
Abstract
Oligodendrocyte progenitor cells (OPCs) are the often-overlooked fourth glial cell type in the central nervous system (CNS), comprising about 5% of the CNS. For a long time, our vision of OPC function was limited to the generation of mature oligodendrocytes. However, new studies have highlighted the multifaceted nature of OPCs. During homeostatic and pathological conditions, OPCs are the most proliferative cell type in the CNS, a property not consistent with the need to generate new oligodendrocytes. Indeed, OPCs modulate neuronal activity and OPC depletion in the brain can trigger depressive-like behavior. More importantly, OPCs are actively recruited to injury sites, where they orchestrate glial scar formation and contribute to the immune response. The following is a comprehensive analysis of the literature on OPC function beyond myelination, in the context of the healthy and diseased adult CNS.
Collapse
Affiliation(s)
- Anthony Fernandez-Castaneda
- Center for Brain Immunology and Glia, Department of Neuroscience, Graduate Program in Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Alban Gaultier
- Center for Brain Immunology and Glia, Department of Neuroscience, Graduate Program in Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
17
|
Okolie O, Bago JR, Schmid RS, Irvin DM, Bash RE, Miller CR, Hingtgen SD. Reactive astrocytes potentiate tumor aggressiveness in a murine glioma resection and recurrence model. Neuro Oncol 2016; 18:1622-1633. [PMID: 27298311 DOI: 10.1093/neuonc/now117] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 05/04/2016] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Surgical resection is a universal component of glioma therapy. Little is known about the postoperative microenvironment due to limited preclinical models. Thus, we sought to develop a glioma resection and recurrence model in syngeneic immune-competent mice to understand how surgical resection influences tumor biology and the local microenvironment. METHODS We genetically engineered cells from a murine glioma mouse model to express fluorescent and bioluminescent reporters. Established allografts were resected using image-guided microsurgery. Postoperative tumor recurrence was monitored by serial imaging, and the peritumoral microenvironment was characterized by histopathology and immunohistochemistry. Coculture techniques were used to explore how astrocyte injury influences tumor aggressiveness in vitro. Transcriptome and secretome alterations in injured astrocytes was examined by RNA-seq and Luminex. RESULTS We found that image-guided resection achieved >90% reduction in tumor volume but failed to prevent both local and distant tumor recurrence. Immunostaining for glial fibrillary acidic protein and nestin showed that resection-induced injury led to temporal and spatial alterations in reactive astrocytes within the peritumoral microenvironment. In vitro, we found that astrocyte injury induced transcriptome and secretome alterations and promoted tumor proliferation, as well as migration. CONCLUSIONS This study demonstrates a unique syngeneic model of glioma resection and recurrence in immune-competent mice. Furthermore, this model provided insights into the pattern of postsurgical tumor recurrence and changes in the peritumoral microenvironment, as well as the impact of injured astrocytes on glioma growth and invasion. A better understanding of the postsurgical tumor microenvironment will allow development of targeted anticancer agents that improve surgery-mediated effects on tumor biology.
Collapse
Affiliation(s)
- Onyinyechukwu Okolie
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (O.O., J.R.B., S.D.H.); Division of Neuropathology, Department of Pathology and Laboratory Medicine, Department of Neurology, and Neuroscience Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.R.M.); Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.S.S., D.M.I., R.E.B., C.R.M., S.D.H.); Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (S.D.H.)
| | - Juli R Bago
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (O.O., J.R.B., S.D.H.); Division of Neuropathology, Department of Pathology and Laboratory Medicine, Department of Neurology, and Neuroscience Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.R.M.); Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.S.S., D.M.I., R.E.B., C.R.M., S.D.H.); Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (S.D.H.)
| | - Ralf S Schmid
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (O.O., J.R.B., S.D.H.); Division of Neuropathology, Department of Pathology and Laboratory Medicine, Department of Neurology, and Neuroscience Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.R.M.); Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.S.S., D.M.I., R.E.B., C.R.M., S.D.H.); Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (S.D.H.)
| | - David M Irvin
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (O.O., J.R.B., S.D.H.); Division of Neuropathology, Department of Pathology and Laboratory Medicine, Department of Neurology, and Neuroscience Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.R.M.); Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.S.S., D.M.I., R.E.B., C.R.M., S.D.H.); Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (S.D.H.)
| | - Ryan E Bash
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (O.O., J.R.B., S.D.H.); Division of Neuropathology, Department of Pathology and Laboratory Medicine, Department of Neurology, and Neuroscience Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.R.M.); Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.S.S., D.M.I., R.E.B., C.R.M., S.D.H.); Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (S.D.H.)
| | - C Ryan Miller
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (O.O., J.R.B., S.D.H.); Division of Neuropathology, Department of Pathology and Laboratory Medicine, Department of Neurology, and Neuroscience Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.R.M.); Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.S.S., D.M.I., R.E.B., C.R.M., S.D.H.); Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (S.D.H.)
| | - Shawn D Hingtgen
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (O.O., J.R.B., S.D.H.); Division of Neuropathology, Department of Pathology and Laboratory Medicine, Department of Neurology, and Neuroscience Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.R.M.); Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.S.S., D.M.I., R.E.B., C.R.M., S.D.H.); Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (S.D.H.)
| |
Collapse
|
18
|
A. Elawady M, M. Elmaghrabi M, Ebrahim N, A. Elawady M, Sabry D, Shamaa A, Ragaei A. Therapeutic Potential of Bone Marrow Derived Mesenchymal Stem Cells in Modulating Astroglyosis of Surgical Induced Experimental Spinal Cord Injury. ACTA ACUST UNITED AC 2016. [DOI: 10.4236/abb.2016.76024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
19
|
Mohn TC, Koob AO. Adult Astrogenesis and the Etiology of Cortical Neurodegeneration. J Exp Neurosci 2015; 9:25-34. [PMID: 26568684 PMCID: PMC4634839 DOI: 10.4137/jen.s25520] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 10/06/2015] [Accepted: 10/08/2015] [Indexed: 01/09/2023] Open
Abstract
As more evidence points to a clear role for astrocytes in synaptic processing, synaptogenesis and cognition, continuing research on astrocytic function could lead to strategies for neurodegenerative disease prevention. Reactive astrogliosis results in astrocyte proliferation early in injury and disease states and is considered neuroprotective, indicating a role for astrocytes in disease etiology. This review describes the different types of human cortical astrocytes and the current evidence regarding adult cortical astrogenesis in injury and degenerative disease. A role for disrupted astrogenesis as a cause of cortical degeneration, with a focus on the tauopathies and synucleinopathies, will also be considered.
Collapse
Affiliation(s)
- Tal C. Mohn
- Biology Department, University of Wisconsin—River Falls, River Falls, Wisconsin, USA
| | - Andrew O. Koob
- Biology Department, University of Wisconsin—River Falls, River Falls, Wisconsin, USA
| |
Collapse
|
20
|
Astrocytic CCAAT/Enhancer-Binding Protein Delta Contributes to Glial Scar Formation and Impairs Functional Recovery After Spinal Cord Injury. Mol Neurobiol 2015; 53:5912-5927. [PMID: 26510742 PMCID: PMC5085997 DOI: 10.1007/s12035-015-9486-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 10/12/2015] [Indexed: 02/06/2023]
Abstract
After spinal cord injury, inflammatory reaction induces the aggregation of astrocytes to form a glial scar that eventually blocks axonal regeneration. Transcription factor CCAAT/enhancer-binding protein delta (C/EBPδ) is a regulatory protein of genes responsive to inflammatory factors, but its role in glial scar formation after spinal cord injury remains unknown. By using a model of moderate spinal cord contusion injury at the mid-thoracic level, we found that C/EBPδ was expressed mostly in the reactive astrocytes bordering the lesion in wild-type mice from 7 days after the injury. C/EBPδ-deficient mice showed reduced glial scar formation, more residual white matter, and better motor function recovery compared with wild-type mice 28 days after the injury. Upon interleukin (IL)-1β stimulation in vitro, the increased expression of C/EBPδ in reactive astrocytes inhibited RhoA expression and, subsequently, the ability of astrocyte migration. However, these reactive astrocytes also produced an increased amount of matrix metalloproteinase-3, which promoted the migration of non-IL-1β-treated, inactive astrocytes. Although the involvement of other non-astroglial C/EBPδ cannot be entirely excluded, our studies suggest that astrocytic C/EBPδ is integral to the inflammatory cascades leading to glial scar formation after spinal cord injury.
Collapse
|
21
|
Dimou L, Gallo V. NG2-glia and their functions in the central nervous system. Glia 2015; 63:1429-51. [PMID: 26010717 DOI: 10.1002/glia.22859] [Citation(s) in RCA: 184] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 05/04/2015] [Indexed: 12/12/2022]
Abstract
In the central nervous system, NG2-glia represent a neural cell population that is distinct from neurons, astrocytes, and oligodendrocytes. While in the past the main role ascribed to these cells was that of progenitors for oligodendrocytes, in the last years it has become more obvious that they have further functions in the brain. Here, we will discuss some of the most current and highly debated issues regarding NG2-glia: Do these cells represent a heterogeneous population? Can they give rise to different progenies, and does this change under pathological conditions? How do they respond to injury or pathology? What is the role of neurotransmitter signaling between neurons and NG2-glia? We will first give an overview on the developmental origin of NG2-glia, and then discuss whether their distinct properties in different brain regions are the result of environmental influences, or due to intrinsic differences. We will then review and discuss their in vitro differentiation potential and in vivo lineage under physiological and pathological conditions, together with their electrophysiological properties in distinct brain regions and at different developmental stages. Finally, we will focus on their potential to be used as therapeutic targets in demyelinating and neurodegenerative diseases. Therefore, this review article will highlight the importance of NG2-glia not only in the healthy, but also in the diseased brain.
Collapse
Affiliation(s)
- L Dimou
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians University, Munich, 80336, Germany.,Institute for Stem Cell Research, Helmholtz Zentrum Munich, Neuherberg, 85764, Germany
| | - V Gallo
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, District of Columbia
| |
Collapse
|
22
|
Yan H, Hong P, Jiang M, Li H. MicroRNAs as potential therapeutics for treating spinal cord injury. Neural Regen Res 2015; 7:1352-9. [PMID: 25657667 PMCID: PMC4308808 DOI: 10.3969/j.issn.1673-5374.2012.17.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Accepted: 05/03/2012] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs are a class of recently discovered, small non-coding RNAs that have been shown to play essential roles in a vast majority of biological processes. Very little is known about the role of microRNAs during spinal cord injury. This review summarizes the changes in expression levels of microRNAs after spinal cord injury. These aberrant changes suggest that microRNAs play an important role in inflammation, oxidative stress, apoptosis, glial scar formation and axonal regeneration. Given their small size and specificity of action, microRNAs could be potential therapeutics for treating spinal cord injury in the future. There are rapidly developing techniques for manipulating microRNA levels in animals; we review different chemical modification and delivery strategies. These may provide platforms for designing efficient microRNA delivery protocols for use in the clinic.
Collapse
Affiliation(s)
- Hualin Yan
- West China Developmental & Stem Cell Institute, Department of Obstetric & Gynecologic and Pediatric, Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China ; West China Medical School, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Peiwei Hong
- West China Developmental & Stem Cell Institute, Department of Obstetric & Gynecologic and Pediatric, Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Mei Jiang
- West China Developmental & Stem Cell Institute, Department of Obstetric & Gynecologic and Pediatric, Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Hedong Li
- West China Developmental & Stem Cell Institute, Department of Obstetric & Gynecologic and Pediatric, Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
23
|
Sun D, Daniels TE, Rolfe A, Waters M, Hamm R. Inhibition of injury-induced cell proliferation in the dentate gyrus of the hippocampus impairs spontaneous cognitive recovery after traumatic brain injury. J Neurotrauma 2015; 32:495-505. [PMID: 25242459 DOI: 10.1089/neu.2014.3545] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Neurogenesis persists throughout life in the neurogenic regions of the mature mammalian brain, and this response is enhanced after traumatic brain injury (TBI). In the hippocampus, adult neurogenesis plays an important role in hippocampal-dependent learning and memory functions and is thought to contribute to the spontaneous cognitive recovery observed after TBI. Utilizing an antimitotic agent, arabinofuranosyl cytidine (Ara-C), the current study investigated the direct association of injury-induced hippocampal neurogenesis with cognitive recovery. In this study, adult rats received a moderate lateral fluid percussion injury followed by a 7-day intraventricular infusion of 2% Ara-C or vehicle. To examine the effect of Ara-C on cell proliferation, animals received intraperitoneal injections of 5-bromo-2-deoxyuridine (BrdU), to label dividing cells, and were sacrificed at 7 days after injury. Brain sections were immunostained for BrdU or doublecortin (DCX), and the total number of BrdU(+) or DCX(+) cells in the hippocampus was quantified. To examine the outcome of inhibiting the injury-induced cell proliferative response on cognitive recovery, animals were assessed on Morris water maze (MWM) tasks at 21-25 or 56-60 days postinjury. We found that a 7-day infusion of Ara-C significantly reduced the total number of BrdU(+) and DCX(+) cells in the dentate gyrus (DG) in both hemispheres. Moreover, inhibition of the injury-induced cell proliferative response in the DG completely abolished the innate cognitive recovery on MWM performance at 56-60 days postinjury. These results support the causal relationship of injury-induced hippocampal neurogenesis on cognitive functional recovery and suggest the importance of this endogenous repair mechanism on restoration of hippocampal function.
Collapse
Affiliation(s)
- Dong Sun
- 1 Department of Neurosurgery, Medical College of Virginia Campus, Virginia Commonwealth University , Richmond, Virginia
| | | | | | | | | |
Collapse
|
24
|
Zhang C, He X, Li H, Wang G. Chondroitinase ABC plus bone marrow mesenchymal stem cells for repair of spinal cord injury. Neural Regen Res 2014; 8:965-74. [PMID: 25206389 PMCID: PMC4145889 DOI: 10.3969/j.issn.1673-5374.2013.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2012] [Accepted: 01/20/2013] [Indexed: 01/09/2023] Open
Abstract
As chondroitinase ABC can improve the hostile microenvironment and cell transplantation is proven to be effective after spinal cord injury, we hypothesized that their combination would be a more effective treatment option. At 5 days after T8 spinal cord crush injury, rats were injected with bone marrow mesenchymal stem cell suspension or chondroitinase ABC 1 mm from the edge of spinal cord damage zone. Chondroitinase ABC was first injected, and bone marrow mesenchymal stem cell suspension was injected on the next day in the combination group. At 14 days, the mean Basso, Beattie and Bresnahan score of the rats in the combination group was higher than other groups. Hematoxylin-eosin staining showed that the necrotic area was significantly reduced in the combination group compared with other groups. Glial fibrillary acidic protein-chondroitin sulfate proteoglycan double staining showed that the damage zone of astrocytic scars was significantly reduced without the cavity in the combination group. Glial fibrillary acidic protein/growth associated protein-43 double immunostaining revealed that positive fibers traversed the damage zone in the combination group. These results suggest that the combination of chondroitinase ABC and bone marrow mesenchymal stem cell transplantation contributes to the repair of spinal cord injury.
Collapse
Affiliation(s)
- Chun Zhang
- Department of Orthopedics, Second Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Xijing He
- Department of Orthopedics, Second Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Haopeng Li
- Department of Orthopedics, Second Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Guoyu Wang
- Department of Orthopedics, Second Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| |
Collapse
|
25
|
Temporal patterns of cortical proliferation of glial cell populations after traumatic brain injury in mice. ASN Neuro 2014; 6:159-70. [PMID: 24670035 PMCID: PMC4013687 DOI: 10.1042/an20130034] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
TBI (traumatic brain injury) triggers an inflammatory cascade, gliosis and cell proliferation following cell death in the pericontusional area and surrounding the site of injury. In order to better understand the proliferative response following CCI (controlled cortical impact) injury, we systematically analyzed the phenotype of dividing cells at several time points post-lesion. C57BL/6 mice were subjected to mild to moderate CCI over the left sensory motor cortex. At different time points following injury, mice were injected with BrdU (bromodeoxyuridine) four times at 3-h intervals and then killed. The greatest number of proliferating cells in the pericontusional region was detected at 3 dpi (days post-injury). At 1 dpi, NG2+ cells were the most proliferative population, and at 3 and 7 dpi the Iba-1+ microglial cells were proliferating more. A smaller, but significant number of GFAP+ (glial fibrillary acidic protein) astrocytes proliferated at all three time points. Interestingly, at 3 dpi we found a small number of proliferating neuroblasts [DCX+ (doublecortin)] in the injured cortex. To determine the cell fate of proliferative cells, mice were injected four times with BrdU at 3 dpi and killed at 28 dpi. Approximately 70% of proliferative cells observed at 28 dpi were GFAP+ astrocytes. In conclusion, our data suggest that the specific glial cell types respond differentially to injury, suggesting that each cell type responds to a specific pattern of growth factor stimulation at each time point after injury.
Collapse
|
26
|
Analgesic effect of minocycline in rat model of inflammation-induced visceral pain. Eur J Pharmacol 2014; 727:87-98. [PMID: 24485889 DOI: 10.1016/j.ejphar.2014.01.026] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 01/08/2014] [Accepted: 01/12/2014] [Indexed: 12/30/2022]
Abstract
The present study investigates the analgesic effect of minocycline, a semi-synthetic tetracycline antibiotic, in a rat model of inflammation-induced visceral pain. Inflammation was induced in male rats by intracolonic administration of tri-nitrobenzenesulphonic acid (TNBS). Visceral hyperalgesia was assessed by comparing the viscero-motor response (VMR) to graded colorectal distension (CRD) prior and post 7 days after TNBS treatment. Electrophysiology recordings from CRD-sensitive pelvic nerve afferents (PNA) and lumbo-sacral (LS) spinal neurons were performed in naïve and inflamed rats. Colonic inflammation produced visceral hyperalgesia characterized by increase in the VMRs to CRD accompanied with simultaneous activation of microglia in the spinal cord and satellite glial cells (SGCs) in the dorsal root ganglions (DRGs). Selectively inhibiting the glial activation following inflammation by araC (Arabinofuranosyl Cytidine) prevented the development of visceral hyperalgesia. Intrathecal minocycline significantly attenuated the VMR to CRD in inflamed rats, whereas systemic minocycline produced a delayed effect. In electrophysiology experiments, minocycline significantly attenuated the mechanotransduction of CRD-sensitive PNAs and the responses of CRD-sensitive LS spinal neurons in TNBS-treated rats. While the spinal effect of minocycline was observed within 5min of administration, systemic injection of the drug produced a delayed effect (60min) in inflamed rats. Interestingly, minocycline did not exhibit analgesic effect in naïve, non-inflamed rats. The results demonstrate that intrathecal injection of minocycline can effectively attenuate inflammation-induced visceral hyperalgesia. Minocycline might as well act on neuronal targets in the spinal cord of inflamed rats, in addition to the widely reported glial inhibitory action to produce analgesia.
Collapse
|
27
|
Gao Z, Zhu Q, Zhang Y, Zhao Y, Cai L, Shields CB, Cai J. Reciprocal modulation between microglia and astrocyte in reactive gliosis following the CNS injury. Mol Neurobiol 2013; 48:690-701. [PMID: 23613214 DOI: 10.1007/s12035-013-8460-4] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 04/09/2013] [Indexed: 12/16/2022]
Abstract
Reactive gliosis, also known as glial scar formation, is an inflammatory response characterized by the proliferation of microglia and astrocytes as well as astrocytic hypertrophy following injury in the central nervous system (CNS). The glial scar forms a physical and molecular barrier to isolate the injured area from adjacent normal nervous tissue for re-establishing the integrity of the CNS. It prevents the further spread of cellular damage but represents an obstacle to regrowing axons. In this review, we integrated the current findings to elucidate the tightly reciprocal modulation between activated microglia and astrocytes in reactive gliosis and proposed that modification of cellular response to the injury or cellular reprogramming in the glial scar could lead advances in axon regeneration and functional recovery after the CNS injury.
Collapse
Affiliation(s)
- Zhongwen Gao
- Department of Spine Surgery, the First Hospital of Jilin University, 71 Xinmin Street, Changchun, Jilin, 130021, China
| | | | | | | | | | | | | |
Collapse
|
28
|
p53 Regulates the neuronal intrinsic and extrinsic responses affecting the recovery of motor function following spinal cord injury. J Neurosci 2013; 32:13956-70. [PMID: 23035104 DOI: 10.1523/jneurosci.1925-12.2012] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Following spinal trauma, the limited physiological axonal sprouting that contributes to partial recovery of function is dependent upon the intrinsic properties of neurons as well as the inhibitory glial environment. The transcription factor p53 is involved in DNA repair, cell cycle, cell survival, and axonal outgrowth, suggesting p53 as key modifier of axonal and glial responses influencing functional recovery following spinal injury. Indeed, in a spinal cord dorsal hemisection injury model, we observed a significant impairment in locomotor recovery in p53(-/-) versus wild-type mice. p53(-/-) spinal cords showed an increased number of activated microglia/macrophages and a larger scar at the lesion site. Loss- and gain-of-function experiments suggested p53 as a direct regulator of microglia/macrophages proliferation. At the axonal level, p53(-/-) mice showed a more pronounced dieback of the corticospinal tract (CST) and a decreased sprouting capacity of both CST and spinal serotoninergic fibers. In vivo expression of p53 in the sensorimotor cortex rescued and enhanced the sprouting potential of the CST in p53(-/-) mice, while, similarly, p53 expression in p53(-/-) cultured cortical neurons rescued a defect in neurite outgrowth, suggesting a direct role for p53 in regulating the intrinsic sprouting ability of CNS neurons. In conclusion, we show that p53 plays an important regulatory role at both extrinsic and intrinsic levels affecting the recovery of motor function following spinal cord injury. Therefore, we propose p53 as a novel potential multilevel therapeutic target for spinal cord injury.
Collapse
|
29
|
Wang J, Ma C, Rong W, Jing H, Hu X, Liu X, Jiang L, Wei F, Liu Z. Bog bilberry anthocyanin extract improves motor functional recovery by multifaceted effects in spinal cord injury. Neurochem Res 2012; 37:2814-25. [PMID: 23001399 DOI: 10.1007/s11064-012-0883-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Revised: 08/09/2012] [Accepted: 08/13/2012] [Indexed: 11/29/2022]
Abstract
The aim of this study was to determine the therapeutic efficiency of bog bilberry anthocyanin extract (BBAE) treatment starting 1 d after spinal cord injury (SCI) in rats and to investigate the underlying mechanism. The BBAE contained cyanidin-3-glucoside, malvidin-3-galactoside and malvidin-3-glucoside. SCI models were induced using the weight-drop method in Sprague-Dawley rats and additionally with sham group (laminectomy only). The animals were divided into four groups: vehicle-treated group; 10 mg/kg BBAE-treated group; 20 mg/kg BBAE-treated group; sham group. BBAE-treated or vehicle-treated group was administered orally at one day after SCI and then daily for 8 weeks. Locomotor functional recovery was assessed during the 8 weeks post operation period by performing a Basso, Beattie, and Bresnahan (BBB) locomotor score test. At the end of study, the animals were killed, and 1.5 cm segments of spinal cord encompassing the injury site were removed for immunohistochemistry, histopathological and western blotting analysis. Immunohistochemistry for GFAP, aggrecan, neurocan and NeuN was used to assess the degree of astrocytic glial scar formation and neuron survival. Immunohistochemistry and western blotting analysis for TNF-α, IL-6, IL-1β was used to evaluate the anti-inflammation effect of BBAE. To evaluate its inhibition effect on the astrocytes, we performed the MTT assay and immunohistochemistry for Ki67 in vitro. Results show that the BBAE-treated animals showed significantly better locomotor functional recovery, neuron death and smaller glial scar formation after spinal cord injury in vivo. In addition, BBAE administration could inhibit astrocyte proliferation in vivo and vitro. Therefore, BBAE may be useful as a promising therapeutic agent for SCI.
Collapse
Affiliation(s)
- Jun Wang
- Department of Orthopedics, Peking University Third Hospital, 49 North Garden Rd, Haidian, Beijing 100191, People's Republic of China.
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Meng F, Hlady V, Tresco PA. Inducing alignment in astrocyte tissue constructs by surface ligands patterned on biomaterials. Biomaterials 2011; 33:1323-35. [PMID: 22100982 DOI: 10.1016/j.biomaterials.2011.10.034] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Accepted: 10/13/2011] [Indexed: 11/24/2022]
Abstract
Planar substrates with patterned ligands were used to induce astrocyte alignment whereas substrates with uniform fields of ligand were used to produce random cell orientation. DRG neurons plated on top of oriented astrocyte monolayers exhibited directional outgrowth along aligned astrocytes, demonstrating that purely biological cues provided by the oriented astrocytes were sufficient to provide guidance cues. Antibody blocking studies demonstrated that astrocyte associated FN played a major mechanistic role in directing engineered neurite extension. Our results show that nanometer level surface cues are sufficient to direct nerve outgrowth through an intervening organized astrocyte cell layer. In other studies, we showed that patterned ligands were able to transmit organization cues through multiple cell layers to control the overall alignment of an astrocyte tissue construct, demonstrating how natural scar tissue may develop in situ into potent barriers. In such constructs the spatial organization of astrocyte derived FN maintained its organizational anisotropy throughout the thickness of multilayered astrocyte constructs. These in vitro studies suggest possible roles for such constructs as bridging substrates for neuroregenerative applications.
Collapse
Affiliation(s)
- Fanwei Meng
- The Keck Center for Tissue Engineering, Department of Bioengineering, University of Utah, Salt Lake City, UT 84112, USA
| | | | | |
Collapse
|
31
|
Delivopoulos E, Murray AF. Controlled adhesion and growth of long term glial and neuronal cultures on Parylene-C. PLoS One 2011; 6:e25411. [PMID: 21966523 PMCID: PMC3178637 DOI: 10.1371/journal.pone.0025411] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Accepted: 09/02/2011] [Indexed: 11/19/2022] Open
Abstract
This paper explores the long term development of networks of glia and neurons on patterns of Parylene-C on a SiO2 substrate. We harvested glia and neurons from the Sprague-Dawley (P1–P7) rat hippocampus and utilized an established cell patterning technique in order to investigate cellular migration, over the course of 3 weeks. This work demonstrates that uncontrolled glial mitosis gradually disrupts cellular patterns that are established early during culture. This effect is not attributed to a loss of protein from the Parylene-C surface, as nitrogen levels on the substrate remain stable over 3 weeks. The inclusion of the anti-mitotic cytarabine (Ara-C) in the culture medium moderates glial division and thus, adequately preserves initial glial and neuronal conformity to underlying patterns. Neuronal apoptosis, often associated with the use of Ara-C, is mitigated by the addition of brain derived neurotrophic factor (BDNF). We believe that with the right combination of glial inhibitors and neuronal promoters, the Parylene-C based cell patterning method can generate structured, active neural networks that can be sustained and investigated over extended periods of time. To our knowledge this is the first report on the concurrent application of Ara-C and BDNF on patterned cell cultures.
Collapse
Affiliation(s)
- Evangelos Delivopoulos
- Nanoscience Centre Department of Engineering, The University of Cambridge, Cambridge, Cambridgeshire, United Kingdom.
| | | |
Collapse
|
32
|
Khaing ZZ, Milman BD, Vanscoy JE, Seidlits SK, Grill RJ, Schmidt CE. High molecular weight hyaluronic acid limits astrocyte activation and scar formation after spinal cord injury. J Neural Eng 2011; 8:046033. [DOI: 10.1088/1741-2560/8/4/046033] [Citation(s) in RCA: 137] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
33
|
Simon C, Götz M, Dimou L. Progenitors in the adult cerebral cortex: cell cycle properties and regulation by physiological stimuli and injury. Glia 2011; 59:869-81. [PMID: 21446038 DOI: 10.1002/glia.21156] [Citation(s) in RCA: 241] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Accepted: 01/19/2011] [Indexed: 02/06/2023]
Abstract
The adult brain parenchyma contains a widespread population of progenitors generating different cells of the oligodendrocyte lineage such as NG2+ cells and some mature oligodendrocytes. However, it is still largely unknown how proliferation and lineage decisions of these progenitors are regulated. Here, we first characterized the cell cycle length, proliferative fraction, and progeny of dividing cells in the adult cerebral cortex and then compared these proliferation characteristics after two distinct stimuli, invasive acute brain injury and increased physiological activity by voluntary physical exercise. Our data show that adult parenchymal progenitors have a very long cell cycle due to an extended G1 phase, many of them can divide at least twice and only a limited proportion of the progeny differentiates into mature oligodendrocytes. After stab wound injury, however, many of these progenitors re-enter the cell cycle very fast, suggesting that the normally long G1 phase is subject to regulation and can be abruptly shortened. In striking contrast, voluntary physical exercise shows the opposite effect with increased exit of the cell cycle followed by an enhanced and fast differentiation into mature oligodendrocytes. Taken together, our data demonstrate that the endogenous population of adult brain parenchymal progenitors is subject to profound modulation by environmental stimuli in both directions, either faster proliferation or faster differentiation.
Collapse
Affiliation(s)
- Christiane Simon
- Physiological Genomics, Institute of Physiology, Ludwig-Maximilians University Munich, Germany
| | | | | |
Collapse
|
34
|
Chong ZZ, Shang YC, Zhang L, Wang S, Maiese K. Mammalian target of rapamycin: hitting the bull's-eye for neurological disorders. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2010; 3:374-91. [PMID: 21307646 PMCID: PMC3154047 DOI: 10.4161/oxim.3.6.14787] [Citation(s) in RCA: 119] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The mammalian target of rapamycin (mTOR) and its associated cell signaling pathways have garnered significant attention for their roles in cell biology and oncology. Interestingly,the explosion of information in this field has linked mTOR to neurological diseases with promising initial studies. mTOR, a 289 kDa serine/threonine protein kinase, plays an important role in cell growth and proliferation and is activated through phosphorylation in response to growth factors, mitogens and hormones. Growth factors, amino acids, cellular nutrients and oxygen deficiency can downregulate mTOR activity. The function of mTOR signaling is mediated primarily through two mTOR complexes: mTORC1 and mTORC2. mTORC1 initiates cap-dependent protein translation, a rate-limiting step of protein synthesis, through the phosphorylation of the targets eukaryotic initiation factor 4E-binding protein 1 (4EBP1) and p70 ribosomal S6 kinase (p70S6K). In contrast, mTORC2 regulates development of the cytoskeleton and also controls cell survival. Although closely tied to tumorigenesis, mTOR and the downstream signaling pathways are significantly involved in the central nervous system (CNS) with synaptic plasticity, memory retention, neuroendocrine regulation associated with food intake and puberty and modulation of neuronal repair following injury. The signaling pathways of mTOR also are believed to be a significant component in a number of neurological diseases, such as Alzheimer disease, Parkinson disease and Huntington disease, tuberous sclerosis, neurofibromatosis, fragile X syndrome, epilepsy, traumatic brain injury and ischemic stroke. Here we describe the role of mTOR in the CNS and illustrate the potential for new strategies directed against neurological disorders.
Collapse
Affiliation(s)
- Zhao Zhong Chong
- Department of Neurology and Neurosciences, Cancer Center, University of Medicine and Dentistry - New Jersey Medical School, Newark, NJ, USA
| | | | | | | | | |
Collapse
|
35
|
Watzlawik J, Warrington AE, Rodriguez M. Importance of oligodendrocyte protection, BBB breakdown and inflammation for remyelination. Expert Rev Neurother 2010; 10:441-57. [PMID: 20187865 DOI: 10.1586/ern.10.13] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the CNS. A better understanding of why remyelination fails in MS is necessary to improve remyelination strategies. Remyelination is mediated by oligodendrocyte precursor cells (OPCs), which are widely distributed throughout the adult CNS. However, it is still unclear whether OPCs detectable in MS lesions survive the inflammatory response but are unable to myelinate or whether OPC and oligodendrocyte death is primarily responsible for remyelination failure and detectable OPCs enter demyelinated areas from adjacent tissue as the lesion evolves. Remyelination strategies should, therefore, focus on stimulation of differentiation or prevention of apoptosis, as well as establishment of a supportive environment for OPC-mediated remyelination, which may be especially important in chronically demyelinated lesions.
Collapse
Affiliation(s)
- Jens Watzlawik
- Departments of Neurology and Immunology, Mayo Clinic College of Medicine, 200 First Street, SW, Rochester, MN 55905, USA
| | | | | |
Collapse
|
36
|
Seira O, Gavín R, Gil V, Llorens F, Rangel A, Soriano E, del Río JA. Neurites regrowth of cortical neurons by GSK3beta inhibition independently of Nogo receptor 1. J Neurochem 2010; 113:1644-58. [PMID: 20374426 DOI: 10.1111/j.1471-4159.2010.06726.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Lesioned axons do not regenerate in the adult mammalian CNS, owing to the over-expression of inhibitory molecules such as myelin-derived proteins or chondroitin sulphate proteoglycans. In order to overcome axon inhibition, strategies based on extrinsic and intrinsic treatments have been developed. For myelin-associated inhibition, blockage with NEP1-40, receptor bodies or IN-1 antibodies has been used. In addition, endogenous blockage of cell signalling mechanisms induced by myelin-associated proteins is a potential tool for overcoming axon inhibitory signals. We examined the participation of glycogen synthase kinase 3beta (GSK3beta) and extracellular-related kinase (ERK) 1/2 in axon regeneration failure in lesioned cortical neurons. We also investigated whether pharmacological blockage of GSK3beta and ERK1/2 activities facilitates regeneration after myelin-directed inhibition in two models: (i) cerebellar granule cells and (ii) lesioned entorhino-hippocampal pathway in slice cultures, and whether the regenerative effects are mediated by Nogo Receptor 1 (NgR1). We demonstrate that, in contrast to ERK1/2 inhibition, the pharmacological treatment of GSK3beta inhibition strongly facilitated regrowth of cerebellar granule neurons over myelin independently of NgR1. Finally, these regenerative effects were corroborated in the lesioned entorhino-hippocampal pathway in NgR1-/- mutant mice. These results provide new findings for the development of new assays and strategies to enhance axon regeneration in injured cortical connections.
Collapse
Affiliation(s)
- Oscar Seira
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia, Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
37
|
Dou F, Huang L, Yu P, Zhu H, Wang X, Zou J, Lu P, Xu XM. Temporospatial expression and cellular localization of oligodendrocyte myelin glycoprotein (OMgp) after traumatic spinal cord injury in adult rats. J Neurotrauma 2010; 26:2299-311. [PMID: 19580419 DOI: 10.1089/neu.2009.0954] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Traumatic spinal cord injury (SCI) leads to permanent neurological deficits, which, in part, is due to the inability of mature axons to regenerate in the mammalian central nervous system (CNS). The oligodendrocyte myelin glycoprotein (OMgp) is one of the myelin-associated inhibitors of neurite outgrowth in the CNS. To date, limited information is available concerning its expression following SCI, possibly due to the lack of a reliable antibody against it. Here we report the generation of a highly specific OMgp polyclonal antibody from the rabbit. Using this antibody, we found that OMgp was almost exclusively expressed in the CNS. Following a moderately contusive SCI using a New York University impactor (10 g rod dropped from a height of 12.5 mm), both OMgp mRNA and protein levels were elevated at 1 and 7 days post-SCI, respectively, and peaked at 28 days compared to those of the sham-operated controls. Spatially, OMgp was expressed throughout the entire rostrocaudal extension of a 10 mm long spinal segment with the highest expression seen at the injury epicenter. OMgp was exclusively localized in neurons and oligodendrocytes in the normal and sham-operated controls with an increased expression found in these cells following SCI. OMgp was not expressed in astrocytes or microglia in all groups. Thus, our study has provided evidence for temporospatial expression and cellular localization of OMgp following SCI and suggested that this molecule may contribute to the overall inhibition of axonal regeneration.
Collapse
Affiliation(s)
- Fangfang Dou
- Department of Neurobiology, Shanghai Jiaotong University School of Medicine, Shanghai, P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Norman LL, Stroka K, Aranda-Espinoza H. Guiding Axons in the Central Nervous System: A Tissue Engineering Approach. TISSUE ENGINEERING PART B-REVIEWS 2009; 15:291-305. [DOI: 10.1089/ten.teb.2009.0114] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Affiliation(s)
- Leann L. Norman
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| | - Kimberly Stroka
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| | - Helim Aranda-Espinoza
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| |
Collapse
|
39
|
The glia-derived extracellular matrix glycoprotein tenascin-C promotes embryonic and postnatal retina axon outgrowth via the alternatively spliced fibronectin type III domain TNfnD. ACTA ACUST UNITED AC 2009; 4:271-83. [DOI: 10.1017/s1740925x09990020] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Tenascin-C (Tnc) is an astrocytic multifunctional extracellular matrix (ECM) glycoprotein that potentially promotes or inhibits neurite outgrowth. To investigate its possible functions for retinal development, explants from embryonic day 18 (E18) rat retinas were cultivated on culture substrates composed of poly-d-lysine (PDL), or PDL additionally coated with Tnc or laminin (LN)-1, which significantly increased fiber length. When combined with LN, Tnc induced axon fasciculation that reduced the apparent number of outgrowing fibers. In order to circumscribe the stimulatory region, Tnc-derived fibronectin type III (TNfn) domains fused to the human Ig-Fc-fragment TNfnD6-Fc, TNfnBD-Fc, TNFnA1A2-Fc and TNfnA1D-Fc were studied. The fusion proteins TNfnBD-Fc and to a lesser degree TNfnA1D-Fc were stimulatory when compared with the Ig-Fc-fragment protein without insert. In contrast, the combination TNfnA1A2-Fc reduced fiber outgrowth beneath the values obtained for the Ig-Fc domain, indicating potential inhibitory properties. The monoclonal J1/tn2 antibody (clone 578) that is specific for domain TNfnD blocked the stimulatory properties of the TNfn-Fc fusions. When postnatal day 7 retinal ganglion cells were used rather that explants, Tnc and Tnc-derived proteins proved permissive for neurite outgrowth. The present study highlights a strong retinal axon growth-promoting activity of the Tnc domain TNfnD, which is modulated by neighboring domains.
Collapse
|
40
|
Rolls A, Shechter R, Schwartz M. The bright side of the glial scar in CNS repair. Nat Rev Neurosci 2009; 10:235-41. [PMID: 19229242 DOI: 10.1038/nrn2591] [Citation(s) in RCA: 485] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Following CNS injury, in an apparently counterintuitive response, scar tissue formation inhibits axonal growth, imposing a major barrier to regeneration. Accordingly, scar-modulating treatments have become a leading therapeutic goal in the field of spinal cord injury. However, increasing evidence suggests a beneficial role for this scar tissue as part of the endogenous local immune regulation and repair process. How can these opposing effects be reconciled? Perhaps it is all a matter of timing.
Collapse
Affiliation(s)
- Asya Rolls
- Asya Rolls, Ravid Shechter and Michal Schwartz are at the Department of Neurobiology, The Weizmann Institute of Science, 76100 Rehovot, Israel
| | | | | |
Collapse
|
41
|
Gervasi NM, Kwok JC, Fawcett JW. Role of extracellular factors in axon regeneration in the CNS: implications for therapy. Regen Med 2009; 3:907-23. [PMID: 18947312 DOI: 10.2217/17460751.3.6.907] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The glial scar that forms after an injury to the CNS contains molecules that are inhibitory to axon growth. Understanding of the mechanisms of inhibition has allowed the development of therapeutic strategies aimed at promoting axon regeneration. Promising results have been obtained in animal models, and some therapies are undergoing clinical trials. This offers great hope for achievement of functional recovery after CNS injury.
Collapse
Affiliation(s)
- Noreen M Gervasi
- Cambridge University Centre for Brain Repair, ED Adrian Building, Forvie Site, Robinson Way, Cambridge CB22PY, UK.
| | | | | |
Collapse
|
42
|
Verma P, Garcia-Alias G, Fawcett JW. Spinal Cord Repair: Bridging the Divide. Neurorehabil Neural Repair 2008; 22:429-37. [DOI: 10.1177/1545968307313500] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The normal spinal cord coordinates movement and sensation in the body. It is a complex organ containing nerve cells, supporting cells, and nerve fibers to and from the brain. The spinal cord is arranged in segments, with higher segments controlling movement and sensation in the upper parts of the body and lower segments controlling the lower parts of the body. Recent notable discoveries in the fields of neuroscience and cell biology have ensured that many more people survive injuries to the brain and spinal cord. The consequences of injury reflect this organization. Although these developments have been mirrored by significant strides in our understanding of the evolution and pathology of spinal injuries, complete repair of structure and hence function remain elusive. Most spinal cord injuries still cause lifelong disability, and continued research is critically needed. Here we review the molecular and cellular processes that occur during the evolution of an injury to the central nervous system. Throughout, we highlight several promising therapies aimed to restore the disrupted connections in the brain and spinal cord. These, used in combination with supportive care and rehabilitation strategies, may help patients to achieve significant long-term recovery.
Collapse
Affiliation(s)
- Poonam Verma
- Cambridge University Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom,
| | - Guillermo Garcia-Alias
- Cambridge University Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - James W. Fawcett
- Cambridge University Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
43
|
Costanzo RM, Perrino LA. Peak in matrix metaloproteinases-2 levels observed during recovery from olfactory nerve injury. Neuroreport 2008; 19:327-31. [PMID: 18303576 DOI: 10.1097/wnr.0b013e3282f50c7b] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Matrix metalloproteineases are associated with extracellular remodeling that occurs in injury and repair processes in the central nervous system (CNS). We examined the role of MMP-2 in a model of olfactory nerve injury and found that MMP-2 levels increased several hours following injury, peaked at day 7 and then decreased rapidly. We previously reported a rapid increase in MMP-9, within 5 h after nerve injury, corresponding to neuronal degeneration and increased glial activity. In this study, we show that MMP-2 peaks later than MMP-9, at the onset of neuronal regeneration and repair. Using MMP-9 knockout mice, we determined that the MMP-2 increase is independent of MMP-9. Our data suggest that MMP-2 and MMP-9 may play different roles in the injury and repair processes.
Collapse
Affiliation(s)
- Richard M Costanzo
- Department of Physiology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA.
| | | |
Collapse
|
44
|
Abstract
The subject of central nervous system damage includes a wide variety of problems, from the slow selective 'picking off' of characteristic sub-populations of neurons typical of neurodegenerative diseases, to the wholesale destruction of areas of brain and spinal cord seen in traumatic injury and stroke. Experimental repair strategies are diverse and the type of pathology dictates which approach will be appropriate. Damage may be to grey matter (loss of neurons), white matter (cutting of axons, leaving neurons otherwise intact, at least initially) or both. This review will consider four possible forms of treatment for repair of the human central nervous system.
Collapse
Affiliation(s)
- J Fitzgerald
- Cambridge University Centre for Brain Repair, Cambridge CB2 2PY, UK.
| | | |
Collapse
|
45
|
Hampton DW, Asher RA, Kondo T, Steeves JD, Ramer MS, Fawcett JW. A potential role for bone morphogenetic protein signalling in glial cell fate determination following adult central nervous system injury in vivo. Eur J Neurosci 2008; 26:3024-35. [PMID: 18028109 DOI: 10.1111/j.1460-9568.2007.05940.x] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Bone morphogenetic proteins (BMPs) and their endogenous inhibitors, including noggin, chordin and follistatin, have roles in pattern formation and fate specification of neuronal and glial cells during nervous system development. We have examined their influence on glial reactions in the injured central nervous system (CNS). We show that penetrating injuries to the brain and spinal cord resulted in the upregulation of BMP-2/4, BMP-7, and noggin, with the latter being expressed almost exclusively by reactive astrocytes at the injury site, and we show that astrocytes in vitro produce noggin. As BMPs have been shown to drive cultured NG2-positive oligodendrocyte precursors (OPCs) towards a multipotential phenotype (type II astrocytes), we investigated the effects of inhibiting noggin with a function-blocking antibody (noggin-FbAb). In vitro, BMP-driven conversion of OPCs to type 2 astrocytes was inhibited by noggin, an effect that was reversed by noggin-FbAb. Noggin-FbAb also increased the number of type 2 astrocytes generated from cultured OPCs exposed to an astrocyte feeder layer, consistent with astrocytes producing both BMPs and noggin. In knife cut injuries in vivo, noggin-FbAb treatment resulted in an increase in the number of NG2-positive cells and small GFAP-positive cells in the injury site, and the appearance of glial cells with the morphological and antigenic characteristics of type 2 astrocytes (as generated in vitro), with coexpression of both GFAP and NG2. This potential conversion of inhibitory OPCs to type 2 astrocyte-like cells in vivo suggests that endogenous BMPs, unmasked by noggin antagonism, might be exploited to manipulate cell fate following CNS trauma.
Collapse
Affiliation(s)
- David W Hampton
- ICORD, University of British Columbia, Vancouver, BC, Canada.
| | | | | | | | | | | |
Collapse
|
46
|
Pattern of chondroitin sulfate proteoglycan expression after ablation of the sensorimotor cortex of the neonatal and adult rat brain. ARCH BIOL SCI 2008. [DOI: 10.2298/abs0804581d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
The central nervous system has a limited capacity for self-repair after damage. However, the neonatal brain has agreater capacity for recovery than the adult brain. These differences in the regenerative capability depend on local environmental factors and the maturational stage of growing axons. Among molecules which have both growth-promoting and growth-inhibiting activities is the heterogeneous class of chondroitin sulfate proteoglycans (CSPGs). In this paper, we investigated the chondroitin-4 and chondroitin-6 sulfate proteoglycan expression profile after left sensorimotor cortex ablation of the neonatal and adult rat brain. Immunohistochemical analysis revealed that compared to the normal uninjured cortex, lesion provoked up regulation of CSPGs showing a different pattern of expression in the neonatal vs. the adult brain. Punctuate and membrane-bound labeling was predominate after neonatal lesion, where as heavy deposition of staining in the extracellular matrix was observed after adult lesion. Heavy deposition of CSPG immunoreactivity around the lesionsite in adult rats, in contrast to a less CSPG-rich environment in neonatal rats, indicated that enhancement of the recovery process after neonatal injury is due to amore permissive environment.
Collapse
|
47
|
Webber DJ, Bradbury EJ, McMahon SB, Minger SL. Transplanted neural progenitor cells survive and differentiate but achieve limited functional recovery in the lesioned adult rat spinal cord. Regen Med 2007; 2:929-45. [DOI: 10.2217/17460751.2.6.929] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Endogenous repair after injury in the adult CNS is limited by a number of factors including cellular loss, inflammation, cavitation and glial scarring. Spinal cord neural progenitor cells (SCNPCs) may provide a valuable cellular source for promoting repair following spinal cord injury. SCNPCs are multipotent, can be expanded in vitro, have the capacity to differentiate into CNS cell lineages and are capable of long-term survival following transplantation. Aims & Method: To determine the extent to which SCNPCs may contribute to spinal cord repair SCNPCs isolated from rat fetal spinal cord were expanded ex vivo and transplanted into the adult rat spinal cord after a dorsal column crush lesion. Results: The survival and distribution of transplanted cells were examined at 24 h, 1, 2 and 6 weeks after injury. Transplanted cells were identified at all time points, located mainly at the lesion perimeter, indicating good post-transplant cell survival. Furthermore, SCNPCs maintained their ability to differentiate in vivo, with approximately 40% differentiating into cells with a glial morphology, whilst 8% displayed a neural morphology. Transplanted animals were also assessed on a number of behavioral tasks measuring sensorimotor and proprioceptive function to determine the extent to which SCNPC transplants might attenuate lesion-induced functional deficits. SCNPCs failed to promote significant functional recovery, with a small improvement observed in only one of the four tasks employed, primarily related to improvements in sensory function. Tracing of the corticospinal tract and ascending dorsal column pathway revealed no regeneration of the axons beyond the lesion site. Conclusions: These data indicate that, although transplanted SCNPCs show good survival in the spinal cord injury environment, combination with other treatment strategies is likely to be required for these cells to fully exert their therapeutic potential.
Collapse
Affiliation(s)
- Daniel J Webber
- University of Cambridge, Centre for Brain Repair, Forvie Site, Robinson Way, Cambridge, CB2 2PY, UK
- King’s College London, Neurorestoration Group, Wolfson Centre for Age Related Disease, Guy’s Campus, London, SE1 1UL, UK
- King’s College London, Stem Cell Biology Laboratory, Wolfson Centre, Guy’s Campus, London, SE1 1UL, UK
| | - Elizabeth J Bradbury
- King’s College London, Neurorestoration Group, Wolfson Centre for Age Related Disease, Guy’s Campus, London, SE1 1UL, UK
| | - Stephen B McMahon
- King’s College London, Neurorestoration Group, Wolfson Centre for Age Related Disease, Guy’s Campus, London, SE1 1UL, UK
| | - Stephen L Minger
- King’s College London, Stem Cell Biology Laboratory, Wolfson Centre, Guy’s Campus, London, SE1 1UL, UK
| |
Collapse
|
48
|
Miller JM, McAllister II JP. Reduction of astrogliosis and microgliosis by cerebrospinal fluid shunting in experimental hydrocephalus. Cerebrospinal Fluid Res 2007; 4:5. [PMID: 17555588 PMCID: PMC1899521 DOI: 10.1186/1743-8454-4-5] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2006] [Accepted: 06/07/2007] [Indexed: 11/23/2022] Open
Abstract
Background Reactive gliosis has the potential to alter biomechanical properties of the brain, impede neuronal regeneration and affect plasticity. Determining the onset and progression of reactive astrogliosis and microgliosis due to hydrocephalus is important for designing better clinical treatments. Methods Reactive astrogliosis and microgliosis were evaluated as the severity of hydrocephalus increased with age in hydrocephalic H-Tx rats and control littermates. Previous studies have suggested that gliosis may persist after short-term drainage (shunt treatment) of the cerebrospinal fluid. Therefore shunts were placed in 15d hydrocephalic rats that were sacrificed after 6d (21d of age) or after 21d (36d of age). Tissue was processed for Western blot procedures and immunohistochemistry, and probed for the astrocytic protein, Glial Fibrillary Acidic Protein (GFAP) and for microglial protein, Isolectin B4 (ILB4). Results In the parietal cortex of untreated hydrocephalic animals, GFAP levels increased significantly at 5d and at 12d compared to age-matched control rats. There was a continued increase in GFAP levels over control at 21d and at 36d. Shunting prevented some of the increase in GFAP levels in the parietal cortex. In the occipital cortex of untreated hydrocephalic animals, there was a significant increase over control in levels of GFAP at 5d. This trend continued in the 12d animals, although not significantly. Significant increases in GFAP levels were present in 21d and in 36d animals. Shunting significantly reduced GFAP levels in the 36d shunted group. Quantitative grading of immuno-stained sections showed similar changes in GFAP stained astrocytes. Immuno-stained microglia were altered in shape in hydrocephalic animals. At 5d and 12d, they appeared to be developmentally delayed with a lack of processes. Older 21d and 36d hydrocephalic animals exhibited the characteristics of activated microglia, with thicker processes and enlarged cell bodies. Following shunting, fewer activated microglia were present. Histologic examination of the periventricular area and the periaqueductal area showed similar findings with the 21d and 36d animals having increased populations of both astrocytes and microglia which were reduced following shunting with a more dramatic reduction in the long term shunted animals. Conclusion Overall, these results suggest that reactive astrocytosis and microgliosis are associated with progressive untreated ventriculomegaly, but that shunt treatment can reduce the gliosis occurring with hydrocephalus.
Collapse
Affiliation(s)
- Janet M Miller
- Department of Neurosurgery, Wayne State University, Detroit, Michigan, USA
- Department of Physiology, Wayne State University, Detroit, Michigan, USA
- Department of Pediatric Neurosurgery, Children's Hospital of Michigan, Detroit, Michigan USA
| | - James P McAllister II
- Department of Neurosurgery, Wayne State University, Detroit, Michigan, USA
- Department of Physiology, Wayne State University, Detroit, Michigan, USA
- Department of Anatomy and Cell Biology, Wayne State University, Detroit Michigan, USA
- Department of Pediatric Neurosurgery, Children's Hospital of Michigan, Detroit, Michigan USA
| |
Collapse
|
49
|
Hayashi S, Inoue A. Cardiomyocytes re-enter the cell cycle and contribute to heart development after differentiation from cardiac progenitors expressing Isl1 in chick embryo. Dev Growth Differ 2007; 49:229-39. [PMID: 17394601 DOI: 10.1111/j.1440-169x.2007.00923.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Cardiomyocytes are generated from the precardiac mesoderm and the size of the heart increases dramatically during embryogenesis. However, it is unclear how differentiation and proliferation correlate in the cardiac cell line during development. Here, we show that cardiomyocytes re-entered into a proliferative state after differentiation with a concomitant cell cycle arrest in chick embryo. The cells in the course of differentiation from Isl1-positive cardiac precursors to cardiomyocytes did not proliferate, but differentiated cardiomyocytes proliferated even after the acquisition of contractile function. After differentiation, cardiomyocytes developed a proliferative potential to contribute to the increase in cell numbers during heart development. Almost all differentiated cardiomyocytes (82.8%) incorporated bromodeoxyuridine (BrdU) in vitro, indicating the ability of DNA replication. Furthermore, mitotic chromosomes were observed in the cardiomyocytes in which a sarcomeric structure was sustained in the cytoplasm. We conclude that the sequential events of the differentiation to contractile myocytes and the re-entry into the cell cycle are strictly regulated during cardiac cell maturation. These results provide an insight into the maturation mechanism of the cardiac cell line.
Collapse
Affiliation(s)
- Shinichi Hayashi
- Department of Biology, Graduate School of Science, Osaka University, 1-1 Machikaneyama-cho, Toyonaka, Osaka 560-0043, Japan.
| | | |
Collapse
|
50
|
Bourguignon LYW, Gilad E, Peyrollier K, Brightman A, Swanson RA. Hyaluronan-CD44 interaction stimulates Rac1 signaling and PKN gamma kinase activation leading to cytoskeleton function and cell migration in astrocytes. J Neurochem 2007; 101:1002-17. [PMID: 17403031 DOI: 10.1111/j.1471-4159.2007.04485.x] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Both hyaluronan [HA, the major glycosaminoglycans in the extracellular matrix (ECM)] and CD44 (a primary HA receptor) are associated with astrocyte activation and tissue repair following central nervous system (CNS) injury. In this study we investigated the question of whether HA-CD44 interaction influences astrocyte signaling and migration. Our data indicated that HA binding to the cultured astrocytes stimulated Rac1 signaling and cytoskeleton-mediated migration. To determine the cellular and molecular basis of these events, we focused on PKN gamma, a Rac1-activated serine/threonine kinase in astrocytes. We determined that HA binding to astrocytes stimulated Rac1-dependent PKN gamma kinase activity which, in turn, up-regulated the phosphorylation of the cytoskeletal protein, cortactin, and attenuated the ability of cortactin to cross-link F-actin. Further analyses indicated that the N-terminal antiparallel coiled-coil (ACC) domains of PKN gamma interacted with Rac1, and transfection of astrocytes with PKN gamma-ACCcDNA inhibited PKN gamma activity. Over-expression of the PKN gamma-ACC domain also functions as a dominant-negative mutant to block HA/CD44-mediated PKN gamma activation of cortactin and astrocyte migration. Taken together, these findings strongly suggest that hyaluronan/CD44 interaction with Rac1-PKN gamma plays a pivotal role in cytoskeleton activation and astrocyte migration. These newly discovered HA/CD44-induced astrocyte function may provide important insight into novel therapeutic treatments for tissue repair following CNS injury.
Collapse
Affiliation(s)
- Lilly Y W Bourguignon
- Department of Medicine, University of California San Francisco, Endocrine Unit (111 N), San Francisco VA Medical Center, San Francisco, CA 94121, USA.
| | | | | | | | | |
Collapse
|