1
|
The Time Course of MHC-I Expression in C57BL/6J and A/J Mice Correlates with the Degree of Retrograde Gliosis in the Spinal Cord following Sciatic Nerve Crush. Cells 2022; 11:cells11233710. [PMID: 36496969 PMCID: PMC9740909 DOI: 10.3390/cells11233710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 11/23/2022] Open
Abstract
The pleiotropic role of the major histocompatibility complex class I (MHC-I) reflects the close association between the nervous and immune systems. In turn, MHC-I upregulation postinjury is associated with a better regenerative outcome in isogenic mice following peripheral nerve damage. In the present work, we compared the time course of neuronal, glial, and sensorimotor recovery (1, 3, 5, 7, and 28 days after lesion—dal) following unilateral sciatic nerve crush in A/J and C57BL/6J mice. The A/J strain showed higher expression of MHC-I (7 dal, ** p < 0.01), Iba-1 (microglial reaction, 7 dal, *** p < 0.001), and GFAP (astrogliosis, 5 dal, * p < 0.05) than the C57BL/6J counterpart. Synaptic coverage (synaptophysin) was equivalent in both strains over time. In addition, mRNA expression of microdissected spinal motoneurons revealed an increase in cytoskeleton-associated molecules (cofilin, shp2, and crmp2, * p < 0.05), but not trkB, in C57BL/6J mice. Gait recovery, studied by the sciatic functional index, was faster in the A/J strain, despite the equivalent results of C57BL/6J at 28 days after injury. A similar recovery was also seen for the nociceptive threshold (von Frey test). Interestingly, when evaluating proprioceptive recovery, C57BL/6J animals showed an enlarged base of support, indicating abnormal ambulation postinjury. Overall, the present results reinforce the role of MHC-I expression in the plasticity of the nervous system following axotomy, which in turn correlates with the variable recovery capacity among strains of mice.
Collapse
|
2
|
Geier DA, Kern JK, Hooker BS, King PG, Sykes LK, Homme KG, Geier MR. Thimerosal exposure and increased risk for diagnosed tic disorder in the United States: a case-control study. Interdiscip Toxicol 2016; 8:68-76. [PMID: 27486363 PMCID: PMC4961900 DOI: 10.1515/intox-2015-0011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 04/10/2015] [Accepted: 04/23/2015] [Indexed: 01/08/2023] Open
Abstract
A hypothesis testing, case-control study evaluated automated medical records for exposure to organic-Hg from Thimerosal-containing hepatitis B vaccines (TM-HepB) administered at specific intervals in the first six-months-of-life among cases diagnosed with a tic disorder (TD) or cerebral degeneration (CD) (an outcome not biologically plausibly linked to TM exposure) in comparison to controls; both cases and controls were continuously enrolled from birth (born from 1991-2000) within the Vaccine Safety Datalink (VSD) database. TD cases were significantly more likely than controls to have received increased organic-Hg from TM-HepB administered within the first month-of-life (odds ratio (OR)=1.59, p<0.00001), first two-months-of-life (OR=1.59, p<0.00001), and first six-months-of-life (OR=2.97, p<0.00001). Male TD cases were significantly more likely than male controls to have received increased organic-Hg from TM-HepB administered within the first month-of-life (OR =1.65, p<0.0001), first two-months-of-life (OR=1.64, p<0.0001), and first six months-of-life (OR=2.47, p<0.05), where as female TD were significantly more likely than female controls to have received increased organic-Hg from TM-HepB administered within the first six-months-of-life (OR=4.97, p<0.05). By contrast, CD cases were no more likely than controls to have received increased organic-Hg exposure from TM-HepB administered at any period studied within the first six-months-of-life. Although routine childhood vaccination is considered an important public health tool to combat infectious diseases, the present study associates increasing organic-Hg exposure from TM-HepB and the subsequent risk of a TD diagnosis.
Collapse
Affiliation(s)
- David A Geier
- Institute of Chronic Illnesses, Inc., 14 Redgate Ct, Silver Spring, MD, USA
| | - Janet K Kern
- Institute of Chronic Illnesses, Inc., 14 Redgate Ct, Silver Spring, MD, USA
| | - Brian S Hooker
- Institute of Chronic Illnesses, Inc., 14 Redgate Ct, Silver Spring, MD, USA
| | - Paul G King
- Institute of Chronic Illnesses, Inc., 14 Redgate Ct, Silver Spring, MD, USA
| | - Lisa K Sykes
- Institute of Chronic Illnesses, Inc., 14 Redgate Ct, Silver Spring, MD, USA
| | - Kristin G Homme
- Institute of Chronic Illnesses, Inc., 14 Redgate Ct, Silver Spring, MD, USA
| | - Mark R Geier
- Institute of Chronic Illnesses, Inc., 14 Redgate Ct, Silver Spring, MD, USA
| |
Collapse
|
3
|
Lewandowska MK, Radivojević M, Jäckel D, Müller J, Hierlemann AR. Cortical Axons, Isolated in Channels, Display Activity-Dependent Signal Modulation as a Result of Targeted Stimulation. Front Neurosci 2016; 10:83. [PMID: 27013945 PMCID: PMC4779934 DOI: 10.3389/fnins.2016.00083] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 02/19/2016] [Indexed: 12/01/2022] Open
Abstract
Mammalian cortical axons are extremely thin processes that are difficult to study as a result of their small diameter: they are too narrow to patch while intact, and super-resolution microscopy is needed to resolve single axons. We present a method for studying axonal physiology by pairing a high-density microelectrode array with a microfluidic axonal isolation device, and use it to study activity-dependent modulation of axonal signal propagation evoked by stimulation near the soma. Up to three axonal branches from a single neuron, isolated in different channels, were recorded from simultaneously using 10-20 electrodes per channel. The axonal channels amplified spikes such that propagations of individual signals along tens of electrodes could easily be discerned with high signal to noise. Stimulation from 10 up to 160 Hz demonstrated similar qualitative results from all of the cells studied: extracellular action potential characteristics changed drastically in response to stimulation. Spike height decreased, spike width increased, and latency increased, as a result of reduced propagation velocity, as the number of stimulations and the stimulation frequencies increased. Quantitatively, the strength of these changes manifested itself differently in cells at different frequencies of stimulation. Some cells' signal fidelity fell to 80% already at 10 Hz, while others maintained 80% signal fidelity at 80 Hz. Differences in modulation by axonal branches of the same cell were also seen for different stimulation frequencies, starting at 10 Hz. Potassium ion concentration changes altered the behavior of the cells causing propagation failures at lower concentrations and improving signal fidelity at higher concentrations.
Collapse
Affiliation(s)
- Marta K Lewandowska
- Bio Engineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich Basel, Switzerland
| | - Miloš Radivojević
- Bio Engineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich Basel, Switzerland
| | - David Jäckel
- Bio Engineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich Basel, Switzerland
| | - Jan Müller
- Bio Engineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich Basel, Switzerland
| | - Andreas R Hierlemann
- Bio Engineering Laboratory, Department of Biosystems Science and Engineering, ETH Zürich Basel, Switzerland
| |
Collapse
|
4
|
Abstract
Integrins are adhesion and survival molecules involved in axon growth during CNS development, as well as axon regeneration after injury in the peripheral nervous system (PNS). Adult CNS axons do not regenerate after injury, partly due to a low intrinsic growth capacity. We have previously studied the role of integrins in axon growth in PNS axons; in the present study, we investigate whether integrin mechanisms involved in PNS regeneration may be altered or lacking from mature CNS axons by studying maturing CNS neurons in vitro. In rat cortical neurons, we find that integrins are present in axons during initial growth but later become restricted to the somato-dendritic domain. We investigated how this occurs and whether it can be altered to enhance axonal growth potential. We find a developmental change in integrin trafficking; transport becomes predominantly retrograde throughout axons, but not dendrites, as neurons mature. The directionality of transport is controlled through the activation state of ARF6, with developmental upregulation of the ARF6 GEF ARNO enhancing retrograde transport. Lowering ARF6 activity in mature neurons restores anterograde integrin flow, allows transport into axons, and increases axon growth. In addition, we found that the axon initial segment is partly responsible for exclusion of integrins and removal of this structure allows integrins into axons. Changing posttranslational modifications of tubulin with taxol also allows integrins into the proximal axon. The experiments suggest that the developmental loss of regenerative ability in CNS axons is due to exclusion of growth-related molecules due to changes in trafficking.
Collapse
|
5
|
Kern JK, Geier DA, King PG, Sykes LK, Mehta JA, Geier MR. Shared Brain Connectivity Issues, Symptoms, and Comorbidities in Autism Spectrum Disorder, Attention Deficit/Hyperactivity Disorder, and Tourette Syndrome. Brain Connect 2015; 5:321-35. [PMID: 25602622 DOI: 10.1089/brain.2014.0324] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The prevalence of neurodevelopmental disorders, including autism spectrum disorder (ASD), attention deficit/hyperactivity disorder (ADHD), and Tourette syndrome (TS), has increased over the past two decades. Currently, about one in six children in the United States is diagnosed as having a neurodevelopmental disorder. Evidence suggests that ASD, ADHD, and TS have similar neuropathology, which includes long-range underconnectivity and short-range overconnectivity. They also share similar symptomatology with considerable overlap in their core and associated symptoms and a frequent overlap in their comorbid conditions. Consequently, it is apparent that ASD, ADHD, and TS diagnoses belong to a broader spectrum of neurodevelopmental illness. Biologically, long-range underconnectivity and short-range overconnectivity are plausibly related to neuronal insult (e.g., neurotoxicity, neuroinflammation, excitotoxicity, sustained microglial activation, proinflammatory cytokines, toxic exposure, and oxidative stress). Therefore, these disorders may a share a similar etiology. The main purpose of this review is to critically examine the evidence that ASD, ADHD, and TS belong to a broader spectrum of neurodevelopmental illness, an abnormal connectivity spectrum disorder, which results from neural long-range underconnectivity and short-range overconnectivity. The review also discusses the possible reasons for these neuropathological connectivity findings. In addition, this review examines the role and issue of axonal injury and regeneration in order to better understand the neuropathophysiological interplay between short- and long-range axons in connectivity issues.
Collapse
Affiliation(s)
- Janet K Kern
- 1 Institute of Chronic Illnesses, Inc. , Silver Spring, Maryland
| | - David A Geier
- 1 Institute of Chronic Illnesses, Inc. , Silver Spring, Maryland
| | | | | | - Jyutika A Mehta
- 3 Communication Sciences & Disorders, Texas Woman's University , Denton, Texas
| | - Mark R Geier
- 1 Institute of Chronic Illnesses, Inc. , Silver Spring, Maryland
| |
Collapse
|
6
|
Williams RR, Venkatesh I, Pearse DD, Udvadia AJ, Bunge MB. MASH1/Ascl1a leads to GAP43 expression and axon regeneration in the adult CNS. PLoS One 2015; 10:e0118918. [PMID: 25751153 PMCID: PMC4353704 DOI: 10.1371/journal.pone.0118918] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 01/16/2015] [Indexed: 12/20/2022] Open
Abstract
Unlike CNS neurons in adult mammals, neurons in fish and embryonic mammals can regenerate their axons after injury. These divergent regenerative responses are in part mediated by the growth-associated expression of select transcription factors. The basic helix-loop-helix (bHLH) transcription factor, MASH1/Ascl1a, is transiently expressed during the development of many neuronal subtypes and regulates the expression of genes that mediate cell fate determination and differentiation. In the adult zebrafish (Danio rerio), Ascl1a is also transiently expressed in retinal ganglion cells (RGCs) that regenerate axons after optic nerve crush. Utilizing transgenic zebrafish with a 3.6 kb GAP43 promoter that drives expression of an enhanced green fluorescent protein (EGFP), we observed that knock-down of Ascl1a expression reduces both regenerative gap43 gene expression and axonal growth after injury compared to controls. In mammals, the development of noradrenergic brainstem neurons requires MASH1 expression. In contrast to zebrafish RGCs, however, MASH1 is not expressed in the mammalian brainstem after spinal cord injury (SCI). Therefore, we utilized adeno-associated viral (AAV) vectors to overexpress MASH1 in four month old rat (Rattus norvegicus) brainstem neurons in an attempt to promote axon regeneration after SCI. We discovered that after complete transection of the thoracic spinal cord and implantation of a Schwann cell bridge, animals that express MASH1 exhibit increased noradrenergic axon regeneration and improvement in hindlimb joint movements compared to controls. Together these data demonstrate that MASH1/Ascl1a is a fundamental regulator of axonal growth across vertebrates and can induce modifications to the intrinsic state of neurons to promote functional regeneration in response to CNS injury.
Collapse
Affiliation(s)
- Ryan R. Williams
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Ishwariya Venkatesh
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI, United States of America
| | - Damien D. Pearse
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States of America
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Ava J. Udvadia
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI, United States of America
| | - Mary Bartlett Bunge
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States of America
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States of America
- Department of Cell Biology, University of Miami Miller School of Medicine, Miami, FL, United States of America
- * E-mail:
| |
Collapse
|
7
|
Stoyanova II, van Wezel RJA, Rutten WLC. In vivo testing of a 3D bifurcating microchannel scaffold inducing separation of regenerating axon bundles in peripheral nerves. J Neural Eng 2013; 10:066018. [PMID: 24280623 DOI: 10.1088/1741-2560/10/6/066018] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Artificial nerve guidance channels enhance the regenerative effectiveness in an injured peripheral nerve but the existing design so far has been limited to basic straight tubes simply guiding the growth to bridge the gap. Hence, one of the goals in development of more effective neuroprostheses is to create bidirectional highly selective neuro-electronic interface between a prosthetic device and the severed nerve. A step towards improving selectivity for both recording and stimulation have been made with some recent in vitro studies which showed that three-dimensional (3D) bifurcating microchannels can separate neurites growing on a planar surface and bring them into contact with individual electrodes. Since the growing axons in vivo have the innate tendency to group in bundles surrounded by connective tissue, one of the big challenges in neuro-prosthetic interface design is how to overcome it. Therefore, we performed experiments with 3D bifurcating guidance scaffolds implanted in the sciatic nerve of rats to test if this new channel architecture could trigger separation pattern of ingrowth also in vivo. Our results showed that this new method enabled the re-growth of neurites into channels with gradually diminished width (80, 40 and 20 µm) and facilitated the separation of the axonal bundles with 91% success. It seems that the 3D bifurcating scaffold might contribute towards conveying detailed neural control and sensory feedback to users of prosthetic devices, and thus could improve the quality of their daily life.
Collapse
Affiliation(s)
- Irina I Stoyanova
- Neurotechnology Group, Biomedical Signals and Systems, Faculty of Electrical Engineering, Mathematics and Computer Sciences, Institute for Biomedical Engineering and Technical Medicine MIRA, University of Twente, Enschede, The Netherlands
| | | | | |
Collapse
|
8
|
Acid fibroblast growth factor and peripheral nerve grafts regulate Th2 cytokine expression, macrophage activation, polyamine synthesis, and neurotrophin expression in transected rat spinal cords. J Neurosci 2011; 31:4137-47. [PMID: 21411654 DOI: 10.1523/jneurosci.2592-10.2011] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Spinal cord injury elicits an inflammatory response that recruits macrophages to the injured spinal cord. Quantitative real-time PCR results have shown that a repair strategy combining peripheral nerve grafts with acidic fibroblast growth factor (aFGF) induced higher interleukin-4 (IL-4), IL-10, and IL-13 levels in the graft areas of rat spinal cords compared with transected spinal cords at 10 and 14 d. This led to higher arginase I-positive alternatively activated macrophage (M2 macrophage) responses. The gene expression of several enzymes involved in polyamine biosynthesis pathways was also upregulated in the graft areas of repaired spinal cords. The treatment induced a twofold upregulation of polyamine levels at 14 d, as confirmed by HPLC. Polyamines are important for the repair process, as demonstrated by the observation that treatment with inhibitors of arginase I and ornithine decarboxylase attenuates the functional recoveries of repaired rats. After 14 d, the treatment also induced the expression of neurotrophin nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF), as well as M2 macrophages within grafted nerves expressing BDNF. IL-4 was upregulated in the injury sites of transected rats that received aFGF alone compared with those that received nerve grafts alone at 10 d. Conversely, nerve graft treatment induced NGF and BDNF expression at 14 d. Macrophages expressing polyamines and BDNF may benefit axonal regeneration at 14 d. These results indicate that aFGF and nerve grafts regulate different macrophage responses, and M2 macrophages may play an important role in axonal regeneration after spinal cord injury in rats.
Collapse
|
9
|
Wang M, Zhai P, Chen X, Schreyer DJ, Sun X, Cui F. Bioengineered scaffolds for spinal cord repair. TISSUE ENGINEERING PART B-REVIEWS 2011; 17:177-94. [PMID: 21338266 DOI: 10.1089/ten.teb.2010.0648] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Spinal cord injury can lead to devastating and permanent loss of neurological function, affecting all levels below the site of trauma. Unfortunately, the injured adult mammalian spinal cord displays little regenerative capacity and little functional recovery in large part due to a tissue environment that is nonpermissive for regenerative axon growth. Artificial tissue repair scaffolds may provide a physical guide to allow regenerative axon growth that bridges the lesion cavity and restores functional neural connectivity. By integrating different strategies, including the use of various biomaterials and microstructures as well as incorporation of bioactive molecules and living cells, combined or synergistic effects for spinal cord repair through regenerative axon growth may be achieved. This article briefly reviews the development of bioengineered scaffolds for spinal cord repair, focusing on spinal cord injury and the subsequent cellular response, scaffold materials, fabrication techniques, and current therapeutic strategies. Key issues and challenges are also identified and discussed along with recommendations for future research.
Collapse
Affiliation(s)
- Mindan Wang
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | | | | | | | | | | |
Collapse
|
10
|
Flynn JR, Graham BA, Galea MP, Callister RJ. The role of propriospinal interneurons in recovery from spinal cord injury. Neuropharmacology 2011; 60:809-22. [PMID: 21251920 DOI: 10.1016/j.neuropharm.2011.01.016] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Revised: 12/23/2010] [Accepted: 01/10/2011] [Indexed: 11/29/2022]
Abstract
Over one hundred years ago, Sir Charles Sherrington described a population of spinal cord interneurons (INs) that connect multiple spinal cord segments and participate in complex or 'long' motor reflexes. These neurons were subsequently termed propriospinal neurons (PNs) and are known to play a crucial role in motor control and sensory processing. Recent work has shown that PNs may also be an important substrate for recovery from spinal cord injury (SCI) as they contribute to plastic reorganisation of spinal circuits. The location, inter-segmental projection pattern and sheer number of PNs mean that after SCI, a significant number of them are capable of 'bridging' an incomplete spinal cord lesion. When these properties are combined with the capacity of PNs to activate and coordinate locomotor central pattern generators (CPGs), it is clear they are ideally placed to assist locomotor recovery. Here we summarise the anatomy, organisation and function of PNs in the uninjured spinal cord, briefly outline the pathophysiology of SCI, describe how PNs contribute to recovery of motor function, and finally, we discuss the mechanisms that underlie PN plasticity. We propose there are two major challenges for PN research. The first is to learn more about ways we can promote PN plasticity and manipulate the 'hostile' micro-environment that limits regeneration in the damaged spinal cord. The second is to study the cellular/intrinsic properties of PNs to better understand their function in both the normal and injured spinal cord. This article is part of a Special Issue entitled 'Synaptic Plasticity & Interneurons'.
Collapse
Affiliation(s)
- Jamie R Flynn
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Hunter Medical Research Institute, The University of Newcastle, Callaghan, NSW 2308, Australia.
| | | | | | | |
Collapse
|
11
|
Harris NG, Mironova YA, Hovda DA, Sutton RL. Pericontusion axon sprouting is spatially and temporally consistent with a growth-permissive environment after traumatic brain injury. J Neuropathol Exp Neurol 2010; 69:139-54. [PMID: 20084019 PMCID: PMC2821052 DOI: 10.1097/nen.0b013e3181cb5bee] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
We previously reported that pericontusional extracellular chondroitin sulfate proteoglycans (CSPGs) are profoundly reduced for 3 weeks after experimental traumatic brain injury, indicating a potential growth-permissive window for plasticity. Here, we investigate the extracellular environment of sprouting neurons after controlled cortical impact injury in adult rats to determine the spatial and temporal arrangement of inhibitory and growth-promoting molecules in relation to growth-associated protein 43-positive (GAP43+) neurons. Spontaneous cortical sprouting was maximal in pericontused regions at 7 and 14 days after injury but absent by 28 days. Perineuronal nets containing CSPGs were reduced at 7 days after injury in the pericontused region (p < 0.05), which was commensurate with a reduction in extracellular CSPGs. Sprouting was restricted to the perineuronal nets and CSPG-deficient regions at 7 days, indicating that the pericontused region is temporarily and spatially permissive to new growth. At this time point,GAP43+ neurons were associated with brain regions containing cells positive for polysialic acid neural cell adhesion molecule but not with fibronectin-positive cells. Brain-derived neurotrophic factor was reduced in the immediate pericontused region at 7 days. Along with prior Western blot evidence, these data suggest that a lowered intrinsic growth stimulus, together with a later return of growth-inhibitory CSPGs, may contribute to the ultimate disappearance of sprouting neurons after traumatic brain injury.
Collapse
Affiliation(s)
- Neil G Harris
- UCLA Brain Injury Research Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-7039, USA.
| | | | | | | |
Collapse
|
12
|
Spatiotemporal patterns of dexamethasone-induced Ras protein 1 expression in the central nervous system of rats with experimental autoimmune encephalomyelitis. J Mol Neurosci 2010; 41:198-209. [PMID: 20084551 DOI: 10.1007/s12031-009-9322-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2009] [Accepted: 12/15/2009] [Indexed: 10/20/2022]
Abstract
Dexamethasone-induced Ras protein 1 (Dexras 1), a brain-enriched member of Ras subfamily of guanosine triphosphatases, as a novel physiologic nitric oxide (NO) effector, anchor neuronal nitric oxide synthase (nNOS) that could form a ternary complex with carboxy-terminal PDZ ligand of nNOS (CAPON) and nNOS, to specific targets to enhance NO signaling. The present study was to explore the expression pattern of Dexras 1 in the development of experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis. Western blot and immunochemistry analysis showed that the gene and protein expression of Dexras 1 in the central nervous system (CNS) of rats increased significantly during the process of EAE compared with control groups (p < 0.01) and maintain a high level in the remission period. The protein expressions of nNOS and CAPON in hippocampus were approximately paralleled Dexras 1. Immunofluorescence revealed that both neurons and glial cells expressed the Dexras 1 in EAE CNS. Importantly, the damaged CNS in EAE-affected rats showed the codistribution between Dexras 1 and caspase 3, indicating the role of Dexras 1 played in the apoptotic process in EAE. Furthermore, colocalizations of Dexras 1 were observed in neurons and glial cells in CNS with nNOS or CAPON, supporting the ternary complex in this model. Thus, these findings suggest the postulation that Dexras 1 might participate into CNS neuronal cell death and demyelination in the whole process of EAE through regulating the NO signaling by binding to nNOS and CAPON.
Collapse
|
13
|
Munderloh C, Solis GP, Bodrikov V, Jaeger FA, Wiechers M, Málaga-Trillo E, Stuermer CAO. Reggies/flotillins regulate retinal axon regeneration in the zebrafish optic nerve and differentiation of hippocampal and N2a neurons. J Neurosci 2009; 29:6607-15. [PMID: 19458231 PMCID: PMC6665911 DOI: 10.1523/jneurosci.0870-09.2009] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2009] [Revised: 04/15/2009] [Accepted: 04/20/2009] [Indexed: 11/21/2022] Open
Abstract
The reggies/flotillins--proteins upregulated during axon regeneration in retinal ganglion cells (RGCs)--are scaffolding proteins of microdomains and involved in neuronal differentiation. Here, we show that reggies regulate axon regeneration in zebrafish (ZF) after optic nerve section (ONS) in vivo as well as axon/neurite extension in hippocampal and N2a neurons in vitro through signal transduction molecules modulating actin dynamics. ZF reggie-1a, -2a, and -2b downregulation by reggie-specific morpholino (Mo) antisense oligonucleotides directly after ONS significantly reduced ZF RGC axon regeneration: RGC axons from reggie Mo retinas were markedly reduced. Moreover, the number of axon-regenerating RGCs, identified by insertion of A488-coupled dextran, decreased by 69% in retinas 7 d after Mo application. At 10 and 14 d, RGCs decreased by 53 and 33%, respectively, in correlation with the gradual inactivation of the Mos. siRNA-mediated knockdown of reggie-1 and -2 inhibited the differentiation and axon/neurite extension in hippocampal and N2a neurons. N2a cells had significantly shorter filopodia, more cells had lamellipodia and fewer neurites, defects which were rescued by a reggie-1 construct without siRNA-binding sites. Furthermore, reggie knockdown strongly perturbed the balanced activation of the Rho family GTPases Rac1, RhoA, and cdc42, influenced the phosphorylation of cortactin and cofilin, the formation of the N-WASP, cortactin and Arp3 complex, and affected p38, Ras, ERK1/2 (extracellular signal-regulated kinases 1 and 2), and focal adhesion kinase activation. Thus, as suggested by their prominent re-expression after lesion, the reggies represent neuron-intrinsic factors for axon outgrowth and regeneration, being crucial for the coordinated assembly of signaling complexes regulating cytoskeletal remodeling.
Collapse
Affiliation(s)
| | - Gonzalo P. Solis
- Department of Biology, University of Konstanz, D-78457 Konstanz, Germany
| | - Vsevolod Bodrikov
- Department of Biology, University of Konstanz, D-78457 Konstanz, Germany
| | | | - Marianne Wiechers
- Department of Biology, University of Konstanz, D-78457 Konstanz, Germany
| | | | | |
Collapse
|
14
|
Langhorst MF, Jaeger FA, Mueller S, Sven Hartmann L, Luxenhofer G, Stuermer CA. Reggies/flotillins regulate cytoskeletal remodeling during neuronal differentiation via CAP/ponsin and Rho GTPases. Eur J Cell Biol 2008; 87:921-31. [DOI: 10.1016/j.ejcb.2008.07.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2008] [Revised: 06/29/2008] [Accepted: 07/03/2008] [Indexed: 10/21/2022] Open
|
15
|
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
|
16
|
Sabha M, Emirandetti A, Cullheim S, De Oliveira ALR. MHC I expression and synaptic plasticity in different mice strains after axotomy. Synapse 2008; 62:137-48. [PMID: 18000810 DOI: 10.1002/syn.20475] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The success of axonal regeneration has been attributed to a co-operation between the severed neurons and the surrounding environment, including non-neuronal cells and the extracellular matrix. Important differences regarding the regeneration potential after injury have been described among inbred mice strains. To date, there is only limited knowledge of how such variation can be linked with the genetic background. It has recently been demonstrated that MHC class I molecules have an influence on the spinal cord synaptic plasticity elicited by a peripheral lesion, and the regenerative capacity following such a lesion. Therefore, in the present work we compared the MHC I expression after axotomy in three isogenic mice strains, namely C57BL/6J, Balb/cJ, and A/J, and investigated the fine ultrastructure of the synaptic elimination process that follows such lesion. The results show that C57BL/6J mice, that have a comparatively poor regenerative potential, display a lower upregulation of MHC I in the spinal cord, coupled with a slower synaptic stripping. On the other hand, A/J mice, which have been shown to have a stronger axonal regrowth potential, showed a clear upregulation of MHC I and a sharp acute loss of afferents, at 1 week after lesion. Our results suggest that a more prominent expression of MHC I in the first week after lesion may positively influence the regenerative outcome associated with a more effective axonal regrowth.
Collapse
Affiliation(s)
- Mario Sabha
- Department of Anatomy, State University of Campinas/SP - Brazil
| | | | | | | |
Collapse
|
17
|
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
|
18
|
Shen A, Chen M, Niu S, Sun L, Gao S, Shi S, Li X, Lv Q, Guo Z, Cheng C. Changes in mRNA for CAPON and Dexras1 in adult rat following sciatic nerve transection. J Chem Neuroanat 2008; 35:85-93. [PMID: 17768032 DOI: 10.1016/j.jchemneu.2007.07.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2007] [Revised: 07/09/2007] [Accepted: 07/14/2007] [Indexed: 12/13/2022]
Abstract
Peripheral nerve transection has been implicated to cause a production of neuronal nitric oxide synthase (nNOS), which may influence a range of post-axotomy processes necessary for neuronal survival and nerve regeneration. Carboxy-terminal post synaptic density protein/Drosophila disc large tumor suppressor/zonula occuldens-1 protein (PDZ) ligand of neuronal nitric oxide synthase (CAPON), as an adaptor, interacts with nNOS via the PDZ domain helping regulate nNOS activity at postsynaptic sites in neurons. And Dexras1, a small G protein mediating multiple signal transductions, has been reported to form a complex with CAPON and nNOS. A role for the physiologic linkage by CAPON of nNOS to Dexras1 has suggested that NO-mediated activation of Dexras1 is markedly enhanced by CAPON. We investigated the changes in mRNA for CAPON, Dexras1 and nNOS in the sciatic nerve, dorsal root ganglia and lumbar spinal cord of adult rat following sciatic axotomy by TaqMan quantitative real-time PCR and in situ hybridization combined with immunofluorescence. Signals of mRNA for CAPON and Dexras1 were initially expressed in these neural tissues mentioned, transiently increased at certain time periods after sciatic axotomy and finally recovered to the basal level. It was also found that nNOS mRNA underwent a similar change pattern during this process. These results suggest that CAPON as well as Dexras1 may be involved in the different pathological conditions including nerve regeneration, neuron loss or survival and even pain process, possibly via regulating the nNOS activity or through the downstream targets of Dexras1.
Collapse
Affiliation(s)
- Aiguo Shen
- The Jiangsu Province Key Laboratory of Neuroregeneration, Nantong University, Nantong 226001, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Kuo HS, Tsai MJ, Huang MC, Huang WC, Lee MJ, Kuo WC, You LH, Szeto KC, Tsai IL, Chang WC, Chiu CW, Ma H, Chak KF, Cheng H. The combination of peripheral nerve grafts and acidic fibroblast growth factor enhances arginase I and polyamine spermine expression in transected rat spinal cords. Biochem Biophys Res Commun 2007; 357:1-7. [PMID: 17418108 DOI: 10.1016/j.bbrc.2007.02.167] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2007] [Accepted: 02/27/2007] [Indexed: 11/23/2022]
Abstract
Treatment with a combination of peripheral nerve grafts and acidic fibroblast growth factor improves hind limb locomotor function after spinal cord transection. This study examined the effect of treatment on expression of arginase I (Arg I) and polyamines. Arg I expression was low in the spinal cords of normal rats but increased following spinal injury. Only fully repaired spinal cords expressed higher Arg I levels 6-14 days following repair. In 10-day repaired spinal cords, high Arg I immunoreactivity was detected in motoneurons and alternatively activated macrophages in the graft area and graft-stump edges, and high levels of the polyamine spermine were expressed by macrophages within the intercostal nerve graft. Thus, in addition to enhancing the expression of Arg I and spermine in repaired spinal cords, our treatment may recruit activated macrophages and create a more favorable environment for axonal regrowth.
Collapse
Affiliation(s)
- Huai-Sheng Kuo
- Neural Regeneration Laboratory, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Deller T, Haas CA, Freiman TM, Phinney A, Jucker M, Frotscher M. Lesion-Induced Axonal Sprouting in the Central Nervous System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 557:101-21. [PMID: 16955706 DOI: 10.1007/0-387-30128-3_6] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Injury or neuronal death often come about as a result of brain disorders. Inasmuch as the damaged nerve cells are interconnected via projections to other regions of the brain, such lesions lead to axonal loss in distal target areas. The central nervous system responds to deafferentation by means of plastic remodeling processes, in particular by inducing outgrowth of new axon collaterals from surviving neurons (collateral sprouting). These sprouting processes result in a partial reinnervation, new circuitry, and functional changes within the deafferented brain regions. Lesioning of the entorhinal cortex is an established model system for studying the phenomenon of axonal sprouting. Using this model system, it could be shown that the sprouting process respects the pre-existing lamination pattern of the deafferented fascia dentata, i. e., it is layer-specific. A variety of different molecules are involved in regulating this reorganization process (extracellular matrix molecules, cell adhesion molecules, transcription factors, neurotrophic factors, growth-associated proteins). It is proposed here that molecules of the extracellular matrix define the boundaries of the laminae following entorhinal lesioning and in so doing limit the sprouting process to the deafferented zone. To illustrate the role of axonal sprouting in disease processes, special attention is given to its significance for neurodegenerative disorders, particularly Alzheimer's disease (AD), and temporal lobe epilepsy. Finally, we discuss both the beneficial as well as disadvantageous functional implications of axonal sprouting for the injured organism in question.
Collapse
Affiliation(s)
- Thomas Deller
- Institute of Clinical Neuroanatomy, Johann Wolfgang Goether-University, Frankfurt am Main, Germany
| | | | | | | | | | | |
Collapse
|
21
|
Rossi F, Gianola S, Corvetti L. Regulation of intrinsic neuronal properties for axon growth and regeneration. Prog Neurobiol 2006; 81:1-28. [PMID: 17234322 DOI: 10.1016/j.pneurobio.2006.12.001] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2006] [Revised: 11/04/2006] [Accepted: 12/05/2006] [Indexed: 01/29/2023]
Abstract
Regulation of neuritic growth is crucial for neural development, adaptation and repair. The intrinsic growth potential of nerve cells is determined by the activity of specific molecular sets, which sense environmental signals and sustain structural extension of neurites. The expression and function of these molecules are dynamically regulated by multiple mechanisms, which adjust the actual growth properties of each neuron population at different ontogenetic stages or in specific conditions. The neuronal potential for axon elongation and regeneration are restricted at the end of development by the concurrent action of several factors associated with the final maturation of neurons and of the surrounding tissue. In the adult, neuronal growth properties can be significantly modulated by injury, but they are also continuously tuned in everyday life to sustain physiological plasticity. Strict regulation of structural remodelling and neuritic elongation is thought to be required to maintain specific patterns of connectivity in the highly complex mammalian CNS. Accordingly, procedures that neutralize such mechanisms effectively boost axon growth in both intact and injured nervous system. Even in these conditions, however, aberrant connections are only formed in the presence of unusual external stimuli or experience. Therefore, growth regulatory mechanisms play an essentially permissive role by setting the responsiveness of neural circuits to environmental stimuli. The latter exert an instructive action and determine the actual shape of newly formed connections. In the light of this notion, efficient therapeutic interventions in the injured CNS should combine targeted manipulations of growth control mechanisms with task-specific training and rehabilitation paradigms.
Collapse
Affiliation(s)
- Ferdinando Rossi
- Rita Levi Montalcini Centre for Brain Repair, Department of Neuroscience, University of Turin, Corso Raffaello 30, I-10125 Turin, Italy.
| | | | | |
Collapse
|
22
|
Gianola S, Rossi F. Neurite-myelin interaction in the control of purkinje axon growth and regeneration. Ann N Y Acad Sci 2006; 1048:141-8. [PMID: 16154928 DOI: 10.1196/annals.1342.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The limited regenerative capability of adult central neurons results from an interplay between the intrinsic growth potential of injured nerve cells and environmental growth-inhibitory signals, such as myelin-associated molecules. These molecules, which are known to inhibit axonal regeneration, are expressed in the adult intact central nervous system, suggesting that they also exert a constitutive regulatory function on uninjured neurites. However, most of the mechanisms underlying this function of myelin-associated factors are still unclear. To address this issue, we examined Purkinje cells, which show extremely poor regenerative capabilities. Such a weak intrinsic growth potential is attributed, at least in part, to inhibitory activity exerted by the myelin-associated molecule Nogo-A on growth-associated gene expression and axon plasticity of adult Purkinje cells. Indeed, the regenerative potential of these neurons declines during postnatal development in parallel with myelin deposition. In addition, myelin-associated factors regulate the development of the Purkinje axon intracortical plexus, contributing to the shaping of mature cerebellar connectivity. The action of environmental inhibitory factors can be partially counteracted by overexpression of neuronal growth-associated molecules. In fact, following axotomy, transgenic Purkinje cells that overexpress GAP-43 are able to sprout their axons into white matter and show a pronounced tendency to lose myelin. Altogether, these results suggest that the control exerted by myelin on Purkinje axon plasticity plays an important role in preventing aberrant growth and in stabilizing intracortical connectivity. Such a constitutive regulation may eventually hamper compensatory mechanisms set up by the same neurons in response to injury and thus determine their weak regenerative abilities.
Collapse
Affiliation(s)
- Sara Gianola
- Rita Levi Montalcini Centre for Brain Repair, Department of Neuroscience, University of Turin, Italy.
| | | |
Collapse
|
23
|
Yang Y, Xie Y, Chai H, Fan M, Liu S, Liu H, Bruce I, Wu W. Microarray analysis of gene expression patterns in adult spinal motoneurons after different types of axonal injuries. Brain Res 2006; 1075:1-12. [PMID: 16460709 DOI: 10.1016/j.brainres.2005.12.060] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2005] [Revised: 10/05/2005] [Accepted: 12/04/2005] [Indexed: 10/25/2022]
Abstract
Three experimental models of axonal injuries in adult rat spinal motoneurons were established to investigate changes of gene expression in response to such injuries. We took advantage of cDNA microarray analysis to determine the differential expression of genes in injured motoneurons following distal axotomy or root avulsion in the absence or presence of BDNF. The major finding was that, in response to proximal axonal injury (avulsion), expression of genes that are known to facilitate neuronal survival and axonal regeneration (e.g., IGFRII, PI3K, IGFBP-6, GSTs, GalR2) were down-regulated; but following treatment with BDNF they were up-regulated. In addition, the expression of genes known to be involved in apoptosis and DNA damage (e.g., ANX5, TS, ALR) were down-regulated in BDNF-treated animals with avulsion. Furthermore, many functional families of genes previously shown to play roles in the pathophysiology of axonal injury, including SNAP-25A, SV2B, Ras-related ras3a/4b, ERK1/2, 14-3-3 proteins, proteasome proteins, oncogenes, GAP-43, and NMDAR1, were altered after either distal axotomy or avulsion injury. Some of the changes in gene expression, including Lim-2, FRAG1, GlaR2, GSTs, ALR, TS, ANX3/5, and nhe1/2, are first reported here in injured motoneurons. The differential expression of genes identified by the expression arrays was confirmed by gene-specific RT-PCR for eight genes (GAP-43, IGFR II, Lim-2, MIF, NDAP1, TS, PCC3, and FRAG1) and by in situ hybridization for Lim-2. These results suggest that abnormal regulation of particular biochemical pathways may induce motoneuron death after ventral root avulsion in adult animals. This study presents an approach for selecting specific genes and their products that may be involved in motoneuron degeneration following axonal injuries.
Collapse
Affiliation(s)
- Yi Yang
- Department of Anatomy, Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Pierucci A, de Oliveira ALR. Increased sensory neuron apoptotic death 2 weeks after peripheral axotomy in C57BL/6J mice compared to A/J mice. Neurosci Lett 2005; 396:127-31. [PMID: 16359790 DOI: 10.1016/j.neulet.2005.11.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2005] [Revised: 11/05/2005] [Accepted: 11/09/2005] [Indexed: 11/21/2022]
Abstract
Peripheral nerve transection results in a disconnection of the neuron from its target. As a result, a series of metabolic changes occur in the cell body that may cause neuronal death, mainly by apoptotic mechanisms. Although neurons from neonatal animals are the most susceptible, peripheral, lesion-induced, neuronal loss also occurs in adults, and is particularly evident in mouse sensory neurons. However, differences in genetic background cause particular isogenic strains of mice to react unevenly to peripheral nerve lesion. In this work, we investigated the occurrence of apoptosis as well as the ultrastructural changes in the dorsal root ganglion sensory neurons and satellite cells of C57BL/6J and A/J mice 2 weeks after ipsilateral sciatic nerve transection at the mid-thigh level. C57BL/6J mice displayed a stronger sensory neuron chromatolytic reaction that resulted in an increased loss of neurons when compared with isogenic A/J mice (p<0.01). Additionally, most of the degenerating neurons displayed the classic features of apoptosis. These findings reinforced previous data obtained by the terminal-deoxynucleotidyl transferase nick-end labeling (TUNEL) technique.
Collapse
Affiliation(s)
- Amauri Pierucci
- Departamento de Anatomia, Instituto de Biologia, Universidade Estadual de Campinas, Cidade Universitária Zeferino Vaz s/n, Distrito de Barão Geraldo, CEP 13084-971, Campinas, SP, Brazil
| | | |
Collapse
|
25
|
Marler KJM, Kozma R, Ahmed S, Dong JM, Hall C, Lim L. Outgrowth of neurites from NIE-115 neuroblastoma cells is prevented on repulsive substrates through the action of PAK. Mol Cell Biol 2005; 25:5226-41. [PMID: 15923637 PMCID: PMC1140584 DOI: 10.1128/mcb.25.12.5226-5241.2005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In the central nervous system (CNS), damaged axons are inhibited from regeneration by glial scars, where secreted chondroitin sulfate proteoglycan (CSPG) and tenascin repulse outgrowth of neurites, the forerunners of axons and dendrites. During differentiation, these molecules are thought to form boundaries for guiding neurons to their correct targets. In neuroblastoma NIE-115 cells, outgrowth of neurites on laminin could be induced by serum starvation or inhibition of RhoA by Clostridium botulinum C3 toxin. The outgrowing neurites avoided crossing onto the repulsive substrate CSPG or tenascin. This avoidance response was partially overcome on expression of membrane-targeted and kinase-inactive forms of PAK. In these cells, the endogenous PAK isoforms colocalized with actin in distinctive sites, alphaPAK in the cell center as small clusters and along the neurite shaft and betaPAK and gammaPAK in areas with membrane ruffles and filopodia, respectively. When isoform-specific N-terminal PAK sequences were introduced to interfere with PAK function, substantially more neurites crossed onto CSPG when cells contained a gammaPAK-derived peptide but not the corresponding alphaPAK- or betaPAK-derived peptide. Thus, while neurite outgrowth can be promoted by RhoA inhibition, overcoming the accompanying repulsive guidance response will require modulation of PAK activity. These results have therapeutic implications for CNS repair processes.
Collapse
Affiliation(s)
- Katharine J M Marler
- Department of Molecular Neuroscience, Institute of Neurology, University College London, 1 Wakefield St., London WC1N 1PJ, United Kingdom
| | | | | | | | | | | |
Collapse
|
26
|
Wang WZ, Emes RD, Christoffers K, Verrall J, Blackshaw SE. Hirudo medicinalis: a platform for investigating genes in neural repair. Cell Mol Neurobiol 2005; 25:427-40. [PMID: 16047550 DOI: 10.1007/s10571-005-3151-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
We have used the nervous system of the medicinal leech as a preparation to study the molecular basis of neural repair. The leech central nervous system, unlike mammalian CNS, can regenerate to restore function, and contains identified nerve cells of known function and connectivity. We have constructed subtractive cDNA probes from whole and regenerating ganglia of the ventral nerve cord and have used these to screen a serotonergic Retzius neuron library. This identifies genes that are regulated as a result of axotomy, and are expressed by the Retzius cell. This approach identifies many genes, both novel and known. Many of the known genes identified have homologues in vertebrates, including man. For example, genes encoding thioredoxin (TRX), Rough Endoplasmic Reticulum Protein 1 (RER-1) and ATP synthase are upregulated at 24 h postinjury in leech nerve cord. To investigate the functional role of regulated genes in neuron regrowth we are using microinjection of antisense oligonucleotides in combination with horseradish peroxidase to knock down expression of a chosen gene and to assess regeneration in single neurons in 3-D ganglion culture. As an example of this approach we describe experiments to microinject antisense oligonucleotide to a leech isoform of the structural protein, Protein 4.1. Our approach thus identifies genes regulated at different times after injury that may underpin the intrinsic ability of leech neurons to survive damage, to initiate regrowth programs and to remake functional connections. It enables us to determine the time course of gene expression in the regenerating nerve cord, and to study the effects of gene knockdown in identified neurons regenerating in defined conditions in culture.
Collapse
Affiliation(s)
- W Z Wang
- Department of Human Anatomy & Genetics, South Parks Road, University of Oxford OX1 3QX, UK
| | | | | | | | | |
Collapse
|
27
|
Abstract
A century ago, Ramon y Cajal described the generalized response of regenerating peripheral axons to their environment. By using mice that express fluorescent proteins in their axons, we are now able to quantify the response of individual axons to nerve transection and repair. Sciatic nerves from nonexpressing mice were grafted into those expressing a yellow variant of green fluorescent protein, then examined at 5, 7, or 10 days after repair. Regeneration was found to be a staggered process, with only 25% of axons crossing the repair in the first week. In the setting of Wallerian degeneration, this stagger will expose growth cones to an evolving menu of molecular cues upon which to base pathway decisions. Many axons arborize, allowing them to interact simultaneously with several pathways. Arborization could serve as the anatomical substrate for specificity generation through collateral pruning. Axons often travel laterally across the face of the distal stump before choosing a pathway. As a result, the average unbranched axon has access to over 100 distal Schwann cell tubes. This extensive access, however, does not ensure correct matching of axon and end organ, suggesting that pathway choice is made on the basis of factors other than end organ identity. These observations explain the failure of refined surgical techniques to restore normal function after nerve injury. The apparent wandering of axons across the repair defies surgical control and mandates a biological approach to reuniting severed axons with appropriate distal pathways.
Collapse
Affiliation(s)
- Christian Witzel
- The Department of Orthopaedic Surgery, Johns Hopkins Medical Institutions, Baltimore, Maryland 21287, USA
| | | | | |
Collapse
|
28
|
Kerschensteiner M, Bareyre FM, Buddeberg BS, Merkler D, Stadelmann C, Brück W, Misgeld T, Schwab ME. Remodeling of axonal connections contributes to recovery in an animal model of multiple sclerosis. ACTA ACUST UNITED AC 2004; 200:1027-38. [PMID: 15492125 PMCID: PMC2211840 DOI: 10.1084/jem.20040452] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
In multiple sclerosis (MS), inflammation in the central nervous system (CNS) leads to damage of axons and myelin. Early during the clinical course, patients can compensate this damage, but little is known about the changes that underlie this improvement of neurological function. To study axonal changes that may contribute to recovery, we made use of an animal model of MS, which allows us to target inflammatory lesions to the corticospinal tract (CST), a major descending motor pathway. We demonstrate that axons remodel at multiple levels in response to a single neuroinflammatory lesion as follows: (a) surrounding the lesion, local interneurons show regenerative sprouting; (b) above the lesion, descending CST axons extend new collaterals that establish a "detour" circuit to the lumbar target area, whereas below the lesion, spared CST axons increase their terminal branching; and (c) in the motor cortex, the distribution of projection neurons is remodeled, and new neurons are recruited to the cortical motor pool. Behavioral tests directly show the importance of these changes for recovery. This paper provides evidence for a highly plastic response of the motor system to a single neuroinflammatory lesion. This framework will help to understand the endogenous repair capacity of the CNS and to develop therapeutic strategies to support it.
Collapse
Affiliation(s)
- Martin Kerschensteiner
- Department of Neuromorphology, Brain Research Institute, University of Zurich, Switzerland.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Mandolesi G, Madeddu F, Bozzi Y, Maffei L, Ratto GM. Acute physiological response of mammalian central neurons to axotomy: ionic regulation and electrical activity. FASEB J 2004; 18:1934-6. [PMID: 15451889 DOI: 10.1096/fj.04-1805fje] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The transection of the axon of central neurons has dramatic consequences on the damaged cells and nerves. Injury activates molecular programs leading to a complex repertoire of responses that, depending on the cellular context, include activation of sprouting, axonal degeneration, and cell death. Although the cellular mechanisms started at the time of lesion are likely to shape the changes affecting injured cells, the acute physiological reaction to trauma of mammalian central neurons is not completely understood yet. To characterize the physiology of the acute response to axonal transection, we have developed a model of in vitro axotomy of neurons cultured from the rodent cortex. Imaging showed that axotomy caused an increase of calcium in the soma and axon. Propagation of the response to the soma required the activation of voltage-dependent sodium channels, since it was blocked by tetrodotoxin. The electrophysiological response to axotomy was recorded in patched neurons kept in the current clamp configuration: injury was followed by vigorous spiking activity that caused a sodium load and the activation of transient calcium currents that were opened by each action potential. The decrease of the electrochemical gradient of sodium caused inversion of the Na-Ca exchanger that provided an additional mean of entry for calcium. Finally, we determined that inhibition of the physiological response to axotomy hindered the regeneration of a new neurite. These data provide elements of the framework required to link the axotomy itself to the downstream molecular machinery that contributes to the determination of the long-term fate of injured neurons and axons.
Collapse
|
30
|
Carulli D, Buffo A, Strata P. Reparative mechanisms in the cerebellar cortex. Prog Neurobiol 2004; 72:373-98. [PMID: 15177783 DOI: 10.1016/j.pneurobio.2004.03.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2003] [Accepted: 03/17/2004] [Indexed: 11/28/2022]
Abstract
In the adult brain, different neuronal populations display different degrees of plasticity. Here, we describe the highly different plastic properties of inferior olivary neurones and Purkinje cells. Olivary neurones show a basal expression of growth-associated proteins, such as GAP-43 and Krox24/EGR-1, and remarkable remodelling capabilities of their terminal arbour. They also regenerate their transected neurites into growth-permissive territories and may reinnervate the lost target. Sprouting and regrowing olivary axons are able to follow specific positional information cues to establish new connections according to the original projection map. In addition, they set a strong cell body reaction to injury, which in specific olivary subsets is regulated by inhibitory target-derived cues. In contrast, Purkinje cells do not have a constitutive level of growth-associated genes, and show little cell body reaction, no axonal regeneration after axotomy, and weak sprouting capabilities. Block of myelin-derived signals allows terminal arbour remodelling, but not regeneration, while selective over-expression of GAP-43 induces axonal sprouting along the axonal surface and at the level of the lesion. We suggest that the high constitutive intrinsic plasticity of the inferior olive neurones allows their terminal arbour to sustain the activity-dependent ongoing competition with the parallel fibres in order to maintain the post-synaptic territory, and possibly underlies mechanisms of learning and memory. Such a plasticity is used also as a reparative mechanism following axotomy. In contrast, in Purkinje cells, poor intrinsic regenerative capabilities and myelin-derived signals stabilise the mature connectivity and prevent axonal regeneration after lesion.
Collapse
Affiliation(s)
- Daniela Carulli
- Department of Neuroscience, Rita Levi Montalcini Center for Brain Repair, University of Turin, C.so Raffaello 30, 10125 Turin, Italy
| | | | | |
Collapse
|
31
|
Abstract
Following injury to the CNS, severed axons undergo a phase of abortive sprouting in the vicinity of the wound, but do not spontaneously re-grow or regenerate. From a long history of attempts to stimulate regeneraion, a major strategy that has been developed clinically is the implantation of tissue into denervated target regions. Unfortunately trials have so far not borne out the promise that this would prove a useful therapy for disorders such as Parkinson's disease. Many strategies have also been developed to stimulate the regeneration of axons across sites of injury, particularly in the spinal cord. Animal data have demonstrated that some of these approaches hold promise and that the spinal cord has a remarkable degree of intrinsic plasticity. Attempts are now being made to utilize experimental techniques in spinal patients.
Collapse
Affiliation(s)
- Peter E Batchelor
- Departments of Medicine and Neurology, University of Melbourne, Austin and Repatriation Medical Centre, Vic. 3084, Heidelberg, Australia
| | | |
Collapse
|
32
|
Zheng B, Ho C, Li S, Keirstead H, Steward O, Tessier-Lavigne M. Lack of enhanced spinal regeneration in Nogo-deficient mice. Neuron 2003; 38:213-24. [PMID: 12718856 DOI: 10.1016/s0896-6273(03)00225-3] [Citation(s) in RCA: 302] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The failure of regeneration of severed axons in the adult mammalian central nervous system is thought to be due partly to the presence of endogenous inhibitors of axon regeneration. The nogo gene encodes three proteins (Nogo-A, -B, and -C) that have been proposed to contribute to this inhibition. To determine whether deletion of nogo enhances regenerative ability, we generated two lines of mutant mice, one lacking Nogo-A and -B but not -C (Nogo-A/B mutant), and one deficient in all three isoforms (Nogo-A/B/C mutant). Although Nogo-A/B-deficient myelin has reduced inhibitory activity in a neurite outgrowth assay in vitro, tracing of corticospinal tract fibers after dorsal hemisection of the spinal cord did not reveal an obvious increase in regeneration or sprouting of these fibers in either mouse line, suggesting that elimination of Nogo alone is not sufficient to induce extensive axon regeneration.
Collapse
Affiliation(s)
- Binhai Zheng
- Department of Biological Sciences, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | | | | | | | | | | |
Collapse
|
33
|
Liu Q, Londraville RL, Azodi E, Babb SG, Chiappini-Williamson C, Marrs JA, Raymond PA. Up-regulation of cadherin-2 and cadherin-4 in regenerating visual structures of adult zebrafish. Exp Neurol 2002; 177:396-406. [PMID: 12429186 DOI: 10.1006/exnr.2002.8008] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cadherins are homophilic cell adhesion molecules that control development of a variety of tissues and maintenance of adult structures. In this study, we examined expression of zebrafish cadherin-2 (Cdh2, N-cadherin) and cadherin-4 (Cdh4, R-cadherin) in the visual system of adult zebrafish after eye or optic nerve lesions using immunocytochemistry and immunoblotting. Both Cdh2 and Cdh4 immunoreactivities were specifically up-regulated in regenerating retina and/or the optic pathway. Furthermore, temporal expression patterns of these two cadherins were distinct during the regeneration of the injured tissues. Cadherins have been shown to regulate axonal outgrowth in the developing nervous system, but this is the first report, to our knowledge, of increased cadherin expression associated with axonal regeneration in the vertebrate central nervous system. Our results suggest that both Cdh2 and Cdh4 may be important for regeneration of injured retinal ganglion cell axons.
Collapse
Affiliation(s)
- Q Liu
- Department of Biology, University of Akron, Akron, Ohio 44325-3908, USA.
| | | | | | | | | | | | | |
Collapse
|
34
|
Gutiérrez-Ospina G, Gutiérrez de la Barrera A, Larriva J, Giordano M. Insulin-like growth factor I partly prevents axon elimination in the neonate rat optic nerve. Neurosci Lett 2002; 325:207-10. [PMID: 12044657 DOI: 10.1016/s0304-3940(02)00293-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Developmental neuronal death ensues after access of innervating neurons to target-derived neurotrophic factors is restricted. Recent evidence suggests, however, that growth factors such as those of the insulin family modulate neuronal death through autocrine/paracrine mechanisms. In rats, retinal ganglion neurons (RGNs) undergo massive death during early postnatal life. During this same period, the expression of various members of the insulin-like growth factor I (IGF-I) protein family is down regulated. To evaluate whether ocular IGF-I might modulate RGN death, we administered IGF-I in the posterior chamber of the eye of newborn rats. Optic nerve fiber number was estimated in control and IGF-I treated animals at postnatal day 5 when RGN death peaks. Intraocular IGF-I treatment at birth partly prevented optic nerve fiber elimination. Because the axon number in the optic nerve correlates to some extent with the RGN number, these results suggest that IGF-I may modulate RGN death in vivo through local interactions.
Collapse
Affiliation(s)
- Gabriel Gutiérrez-Ospina
- Department of Cell Biology and Physiology, Biomedical Research Institute, National University of México, Mexico D.F., 04510, Mexico.
| | | | | | | |
Collapse
|
35
|
Neumann S, Bradke F, Tessier-Lavigne M, Basbaum AI. Regeneration of sensory axons within the injured spinal cord induced by intraganglionic cAMP elevation. Neuron 2002; 34:885-93. [PMID: 12086637 DOI: 10.1016/s0896-6273(02)00702-x] [Citation(s) in RCA: 418] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The peripheral branch of primary sensory neurons regenerates after injury, but there is no regeneration when their central branch is severed by spinal cord injury. Here we show that microinjection of a membrane-permeable analog of cAMP in lumbar dorsal root ganglia markedly increases the regeneration of injured central sensory branches. The injured axons regrow into the spinal cord lesion, often traversing the injury site. This result mimics the effect of a conditioning peripheral nerve lesion. We also demonstrate that sensory neurons exposed to cAMP in vivo, when subsequently cultured in vitro, show enhanced growth of neurites and an ability to overcome inhibition by CNS myelin. Thus, stimulating cAMP signaling increases the intrinsic growth capacity of injured sensory axons. This approach may be useful in promoting regeneration after spinal cord injury.
Collapse
Affiliation(s)
- Simona Neumann
- Department of Anatomy and W.M. Keck Foundation Center for Integrative Neuroscience, University of California, San Francisco, CA 94143, USA
| | | | | | | |
Collapse
|
36
|
Abstract
Denervation of the hippocampus triggers reactive responses in neurons and glial cells in their affected strata in a temporally ordered fashion. Many of these responses have been studied extensively, focusing on the one hand on glial initiation and clearing responses during the degeneration phase and, on the other, on transneuronal reorganization and the newly adjusted physiological balance. We used the entorhinal cortex lesion (ECL) as a model system to study the cues that underlie the layer-specific sprouting response. This lesion destroys the perforant path, which is a massive excitatory projection to the dentate gyrus and hippocampus proper. In the deafferented zones of the hippocampus, sprouting of the remaining unlesioned fibers occurs, which replaces the lost afferences of the perforant path. We focus on candidate molecules which govern the layer-specific sprouting of the remaining axons and, in particular, on membrane-bound cues. The fact that layer-specific sprouting occurs even in the adult central nervous system (CNS) provides a valuable model for understanding the mechanisms of reactive neuronal growth and reorganization in the adult CNS. Isolation and analysis of the molecules involved in these mechanisms are important steps in understanding the potential and limitations of regeneration in the CNS.
Collapse
Affiliation(s)
- N E Savaskan
- The Institute of Anatomy, Department of Cell & Neurobiology, Humboldt University Medical School Charité, Berlin, Germany.
| | | |
Collapse
|
37
|
Benítez-Temiño B, De La Cruz RR, Pastor AM. Firing properties of axotomized central nervous system neurons recover after graft reinnervation. J Comp Neurol 2002; 444:324-44. [PMID: 11891646 DOI: 10.1002/cne.10147] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Axotomy produces changes in the electrical properties of neurons and in their synaptic inputs, leading to alterations in firing pattern. We have considered the possibility that these changes occur as a result of the target deprivation induced by the lesion. Thus, we have provided a novel target to axotomized central neurons by grafting embryonic tissue at the lesion site to study the target dependence of discharge characteristics. The extracellular single-unit electrical activity of abducens internuclear neurons was recorded in the alert behaving cat in control, after axotomy, and after axotomy plus the implantation of cerebellar primordium. As recently characterized (de la Cruz et al. [2000] J. Comp. Neurol. 427:391-404), firing alterations induced by axotomy included an overall decrease in firing rate and a loss of eye-related signals, i.e., eye position and velocity neuronal sensitivities, that do not resume to normality with time. The grafting of a novel target to the injured abducens internuclear neurons restored the normal firing and sensitivities as recorded in the majority of units. To study the reinnervation of the implant, we performed anterograde labeling with biocytin combined with electron microscopy visualization. Axons of abducens internuclear neurons grew into the transplant sprouting into granule cell and molecular layers, as characterized by the immunostaining for gamma-aminobutyric acid and calbindin D-28k. Ultrastructural examination of labeled axons and boutons revealed the establishment of synaptic contacts, mainly axodendritic, with different cell types of the grafted cerebellar cortex. Therefore, these data indicate that axotomized central neurons resume to normal firing after the reinnervation of a novel target.
Collapse
Affiliation(s)
- Beatriz Benítez-Temiño
- Departamento de Fisiología y Biología Animal, Universidad de Sevilla, 41012-Seville, Spain
| | | | | |
Collapse
|
38
|
Giménez y Ribotta M, Menet V, Privat A. The role of astrocytes in axonal regeneration in the mammalian CNS. PROGRESS IN BRAIN RESEARCH 2001; 132:587-610. [PMID: 11545022 DOI: 10.1016/s0079-6123(01)32105-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Affiliation(s)
- M Giménez y Ribotta
- INSERM U336, Université Montpellier II, Place E. Bataillon, B.P. 106, 34095 Montpellier, France
| | | | | |
Collapse
|
39
|
Gianola S, Rossi F. Evolution of the Purkinje cell response to injury and regenerative potential during postnatal development of the rat cerebellum. J Comp Neurol 2001; 430:101-17. [PMID: 11135248 DOI: 10.1002/1096-9861(20010129)430:1<101::aid-cne1017>3.0.co;2-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
To understand the mechanisms leading to the progressive loss of intrinsic neuronal growth properties during central nervous system development, we have investigated the evolution of the response to injury and regenerative potential of immature Purkinje cells, axotomized at different postnatal ages from postnatal day (P)3 to P12. In adult rodents, these neurons are characterised by a weak cell body response to axotomy, which is associated with a remarkable resistance to injury and a poor regenerative capability. During the first postnatal week, Purkinje cells are strongly sensitive to injury and massively degenerate within a few days. Immature Purkinje cells react to neurite transection by a strong upregulation of c-Jun, accompanied by a moderate, but consistent, expression of the growth-associated protein (GAP)-43. In contrast, nicotinamide adenine dinucleotide monophosphate (NADPH)-diaphorase reactivity, which can be activated by adult Purkinje neurons, is not modified in their juvenile counterparts. The severed Purkinje axons show a vigorous regenerative sprouting both into the lesioned cerebellar environment and into embryonic neocortical tissue transplanted into the injury site. The typical adult features of the response to injury progressively develop during the second postnatal week, when the injured neurons acquire resistance, cell body changes become milder, the regenerative potential declines, and the severed axons undergo characteristic morphological modifications, including torpedoes and the hypertrophy of recurrent collateral branches. This complete reversal of the features and the outcome of the Purkinje cell reaction to axotomy likely results from the profound changes that occur in the maturing Purkinje cells and/or in their microenvironment during this phase of cerebellar development.
Collapse
Affiliation(s)
- S Gianola
- Department of Neuroscience and Rita Levi Montalcini Centre for Brain Repair, University of Turin, I-10125 Turin, Italy
| | | |
Collapse
|
40
|
Haas CA, Hollerbach E, Deller T, Naumann T, Frotscher M. Up-regulation of growth-associated protein 43 mRNA in rat medial septum neurons axotomized by fimbria-fornix transection. Eur J Neurosci 2000; 12:4233-42. [PMID: 11122335 DOI: 10.1046/j.0953-816x.2000.01329.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Transection of septohippocampal fibres is widely used to study the response of CNS neurons to axotomy. Septohippocampal projection neurons survive axotomy and selectively up-regulate the transcription factor c-Jun. In the present study we investigated whether these cells concomitantly up-regulate the growth-associated protein-43 (GAP-43), a potential target gene of c-Jun implicated in axonal growth and regeneration. Using in situ hybridization histochemistry (ISHH) it was demonstrated that postlesional c-jun mRNA expression is accompanied by an increased expression of GAP-43 mRNA in the medial septum 3 days following fimbria-fornix transection (FFT). The increase reached a maximum at 7 days and gradually declined thereafter (17 days, 3 weeks). Retrograde prelabeling with Fluoro-Gold followed by axotomy and ISHH revealed that GAP-43 mRNA was up-regulated in septohippocampal projection neurons. Colocalization of GAP-43 mRNA and choline acetyltransferase protein showed that GAP-43 mRNA was expressed by cholinergic medial septal neurons after axotomy. Selective immunolesioning of the cholinergic component of the septohippocampal projection with 192 IgG-saporin followed by FFT demonstrated that GAP-43 mRNA was also synthesized by axotomized GABAergic neurons. These results demonstrate an up-regulation of GAP-43 mRNA in axotomized septohippocampal projection neurons independent of their transmitter phenotype which is closely correlated with c-Jun expression. Because the GAP-43 gene contains an AP-1 site, we hypothesize a c-Jun-driven up-regulation of GAP-43 in lesioned medial septal neurons that may contribute to their survival and regenerative potential following axotomy.
Collapse
Affiliation(s)
- C A Haas
- Institute of Anatomy I, University of Freiburg, PO Box 111, D-79001 Freiburg, Germany.
| | | | | | | | | |
Collapse
|
41
|
Oliveira AL, Langone F. Non-neuronal cells are not the limiting factor for the low axonal regeneration in C57BL/6J mice. Braz J Med Biol Res 2000; 33:1467-75. [PMID: 11105100 DOI: 10.1590/s0100-879x2000001200011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Peripheral axonal regeneration was investigated in adult male mice of the C57BL/6J (C), BALB/cJ (B) and A/J (A) strains and in their F1 descendants using a predegenerated nerve transplantation model. Four types of transplants were performed: 1) isotransplants between animals of the C, B and A strains; 2) donors of the C strain and recipients of the C x B and C x A breeding; 3) donors of the B strain and recipients of the C x B breeding, and 4) donors of the A strain and recipients of the C x A breeding. Donors had the left sciatic nerve transected and two weeks later a segment of the distal stump was transplanted into the recipient. Four weeks after transplantation the regenerated nerves were used to determine the total number of regenerated myelinated fibers (TMF), diameter of myelinated fibers (FD) and myelin thickness (MT). The highest TMF values were obtained in the groups where C57BL/6J mice were the donors (C to F1 (C x B) = 4658 +/- 304; C to F1 (C x A) = 3899 +/- 198). Also, A/J grafts led to a significantly higher TMF (A to F1 (C x A) = 3933 +/- 565). Additionally, isotransplant experiments showed that when the nerve is previously degenerated, C57BL/6J mice display the largest number of myelinated fibers (C to C = 3136 +/- 287; B to B = 2759 +/- 170, and A to A = 2835 +/- 239). We also observed that when C57BL/6J was the graft donor, FD was the highest and MT did not differ significantly when compared with the other groups. These morphometric results reinforce the idea that Schwann cells and the nerve environment of C57BL/6J provide enough support to the regenerative process. In this respect, the present results support the hypothesis that the non-neuronal cells, mainly Schwann cells, present in the sciatic nerve of C57BL/6J mice are not the main limiting factor responsible for low axonal regeneration.
Collapse
Affiliation(s)
- A L Oliveira
- Departamento de Anatomia, Universidade Estadual de Campinas, Campinas, SP, Brasil.
| | | |
Collapse
|
42
|
Goldman D, Ding J. Different regulatory elements are necessary for alpha1 tubulin induction during CNS development and regeneration. Neuroreport 2000; 11:3859-63. [PMID: 11117504 DOI: 10.1097/00001756-200011270-00051] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Developing and regenerating neurons induce genes whose products are necessary for axonal growth, such as that encoding alpha1 tubulin. To determine whether alpha1 tubulin gene induction uses similar mechanisms during CNS development and regeneration, we compared wild-type and mutant alpha1 tubulin promoter activity in the developing and regenerating CNS of transgenic zebrafish. Wild-type alpha1 tubulin promoter activity increased dramatically in the developing and regenerating CNS. In contrast, we generated a mutation in the alpha1 tubulin promoter that prevented its increase during development but retained regeneration-dependent induction in the adult. These results suggest that at least some of the signaling mechanisms used to activate alpha1 tubulin promoter activity during CNS regeneration are different from those used to activate this promoter during development.
Collapse
Affiliation(s)
- D Goldman
- Mental Health Research Institute and Department of Biological Chemistry, University of Michigan, Ann Arbor 48109, USA
| | | |
Collapse
|
43
|
Pastor AM, Delgado-García JM, Martínez-Guijarro FJ, López-García C, de La Cruz RR. Response of abducens internuclear neurons to axotomy in the adult cat. J Comp Neurol 2000; 427:370-90. [PMID: 11054700 DOI: 10.1002/1096-9861(20001120)427:3<370::aid-cne5>3.0.co;2-m] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The highly specific projection of abducens internuclear neurons on the medial rectus motoneurons of the oculomotor nucleus constitutes an optimal model for investigating the effects of axotomy in the central nervous system. We have analyzed the morphological changes induced by this lesion on both the cell bodies and the transected axons of abducens internuclear neurons in the adult cat. Axotomy was performed by the transection of the medial longitudinal fascicle. Cell counts of Nissl-stained material and calretinin-immunostained abducens internuclear neurons revealed no cell death by 3 months postaxotomy. Ultrastructural examination of these cells at 6, 14, 24, and 90 days postaxotomy showed normal cytological features. However, the surface membrane of axotomized neurons appeared contacted by very few synaptic boutons compared to controls. This change was quantified by measuring the percentage of synaptic coverage of the cell bodies and the linear density of boutons. Both parameters decreased significantly after axotomy, with the lowest values at 90 days postlesion ( approximately 70% reduction). We also explored axonal regrowth and the possibility of reinnervation of a new target by means of anterograde labeling with biocytin. At all time intervals analyzed, labeled axons were observed to be interrupted at the caudal limit of the lesion; in no case did they cross the scar tissue to reach the distal part of the tract. Nonetheless, a conspicuous axonal sprouting was present at the caudal aspect of the lesion site. Structures suggestive of axonal growth were found, such as large terminal clubs, from which short filopodium-like branches frequently emerged. Similar findings were obtained after parvalbumin and calretinin immunostaining. At the electron microscopy level, biocytin-labeled boutons originating from the sprouts appeared surrounded by either extracellular space, which was extremely dilated at the lesion site, or by glial processes. The great majority of labeled boutons examined were, thus, devoid of neuronal contact, indicating absence of reinnervation of a new target. Altogether, these data indicate that abducens internuclear neurons survive axotomy in the adult cat and show some form of axonal regrowth, even in the absence of target connection.
Collapse
Affiliation(s)
- A M Pastor
- Laboratorio de Neurociencia, Facultad de Biología, Universidad de Sevilla, 41012-Sevilla, Spain
| | | | | | | | | |
Collapse
|
44
|
Martin GF, Terman JR, Wang XM. Regeneration of descending spinal axons after transection of the thoracic spinal cord during early development in the North American opossum, Didelphis virginiana. Brain Res Bull 2000; 53:677-87. [PMID: 11165803 DOI: 10.1016/s0361-9230(00)00401-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Opossums are born in an immature, fetal-like state, making it possible to lesion their spinal cord early in development without intrauterine surgery. When the thoracic spinal cord of the North American opossum, Didelphis virginiana, is transected on postnatal day 5, and injections of Fast Blue (FB) are made caudal to the lesion site 30-40 days or 6 months later, neurons are labeled in all of the spinal and supraspinal areas that are labeled after comparable injections in age-matched, unlesioned controls. Double-labeling studies document that regeneration of cut axons contributes to growth of axons through the lesion site and behavioral studies show that animals lesioned on postnatal day 5 use their hindlimbs in normal appearing locomotion as adults. The critical period for developmental plasticity of descending spinal axons extends to postnatal day 26, although axons which grow through the lesion site become fewer in number and more restricted as to origin with increasing age. Animals lesioned between postnatal day 12 and 26 use the hindlimbs better than animals lesioned as adults, but hindlimb function is markedly abnormal and uncoordinated with that of the forelimbs. We conclude that restoration of anatomical continuity occurs after transection of the spinal cord in developing opossums, that descending axons grow through the lesion site, that regeneration of cut axons contributes to such growth, and that animals lesioned early enough in development have relatively normal motor function as adults.
Collapse
Affiliation(s)
- G F Martin
- Department of Anatomy and Medical Education, The Ohio State University, College of Medicine and Public Health, Columbus, OH 43210, USA
| | | | | |
Collapse
|
45
|
Abstract
Neural geometry is the major factor that determines connectivity and, possibly, functional output from a nervous system. Recently some of the proteins and pathways involved in specific modes of branch formation or maintenance, or both, have been described. To a variable extent, dendrites and axon collaterals can be viewed as dynamic structures subject to fine modulation that can result either in further growth or retraction. Each form of branching results from specific molecular mechanisms. Cell-internal, substrate-derived factors and functional activity, however, can often differ in their effect according to cell type and physiological context at the site of branch formation. Neural branching is not a linear process but an integrative one that takes place in a microenvironment where we have only a limited experimental access. To attain a coherent mechanism for this phenomenon, quantitative in situ data on the proteins involved and their interactions will be required.
Collapse
Affiliation(s)
- A Acebes
- The Instituto Cajal (CSIC), 28002, Madrid, Spain
| | | |
Collapse
|
46
|
Haas CA, Deller T, Krsnik Z, Tielsch A, Woods A, Frotscher M. Entorhinal cortex lesion does not alter reelin messenger RNA expression in the dentate gyrus of young and adult rats. Neuroscience 2000; 97:25-31. [PMID: 10877665 DOI: 10.1016/s0306-4522(00)00007-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The extracellular matrix protein reelin plays an important role in neuronal pattern formation and axonal collateralization during the development of the central nervous system. With the concept that reelin might also be important for axonal growth in the injured nervous system we investigated whether reelin is re-expressed in areas of collateral sprouting after brain injury. The expression of reelin messenger RNA was studied in the denervated fascia dentata of adult rats one, four, seven and 14 days following entorhinal cortex lesion. In adult control animals, in situ hybridization histochemistry with digoxigenin-labeled reelin riboprobes revealed reelin messenger RNA expression in neurons located in the outer molecular layer and beneath the granule cell layer of the dentate gyrus. After entorhinal cortex lesion, this expression pattern did not change during the whole post-lesional time period investigated despite a strong glial activation and reactive sprouting in the outer molecular layer of the dentate gyrus as visualized by immunohistochemistry for glial fibrillary acidic protein and acetylcholinesterase histochemistry, respectively. The expression of reelin messenger RNA was also unaffected by entorhinal cortex lesion in the dentate gyrus of young animals (postnatal day seven), where an even stronger sprouting response occurs.
Collapse
Affiliation(s)
- C A Haas
- Institute of Anatomy, University of Freiburg, P.O. Box 111, D-79001, Freiburg, Germany
| | | | | | | | | | | |
Collapse
|
47
|
Fry EJ, Saunders NR. Spinal repair in immature animals: a novel approach using the South American opossum Monodelphis domestica. Clin Exp Pharmacol Physiol 2000; 27:542-7. [PMID: 10874515 DOI: 10.1046/j.1440-1681.2000.03296.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
1. The adult mammalian central nervous system (CNS) is unable to regenerate following injury and repair has only been seen when implants of peripheral nervous tissue, fetal tissue or Schwann cells are used, or antibodies or trophic molecules applied. However, the immature mammalian CNS has revealed a capacity to repair without extrinsic influence. 2. The marsupial mammal provides a unique opportunity to access the immature CNS without invasive in utero surgery. In particular, the South American opossum Monodelphis domestica is an ideal animal for spinal cord injury studies examining the ability of the immature CNS to repair after injury. 3. The Monodelphis spinal cord may be examined for its response to injury either as an in vitro or in vivo system and, therefore, is a flexible model, allowing many different questions to be addressed by the most suitable approach. 4. The immature Monodelphis CNS was able to support fibre growth that reappeared 4 days after a crush at P3-P8 in vitro. Conduction was also restored at this time, accompanied by synaptic connections. 5. A cut lesion performed in vivo on Monodelphis spinal cords at P7 took longer to repair, with fibres reappearing across the injury site 2 weeks after the lesion; greater disruption to structure was noted both during early stages of repair and in adulthood. 6. Neural pathway tracing with dextran amine from the lumbar cord to the brain in adult Monodelphis, which received spinal lesions at P7, revealed a similar distribution of labelled cells in brainstem and mid-brain nuclei to that of control animals. 7. Studies of the locomotor behaviour of adult Monodelphis that had received either a cut or crush lesion at P7-P8 showed remarkably similar abilities to control animals when performing complex tasks. 8. The results of spinal cord injury studies with the immature Monodelphis CNS may help in the development of treatments for spinal injury patients.
Collapse
Affiliation(s)
- E J Fry
- Department of Anatomy and Physiology, University of Tasmania, Hobart, Australia.
| | | |
Collapse
|
48
|
Fugleholm K, Schmalbruch H, Krarup C. Post reinnervation maturation of myelinated nerve fibers in the cat tibial nerve: chronic electrophysiological and morphometric studies. J Peripher Nerv Syst 2000; 5:82-95. [PMID: 10905467 DOI: 10.1046/j.1529-8027.2000.00008.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The extent to which the long-term recovery of nerve fibers differs according to the cause of Wallerian degeneration is not clear, although outgrowth of axons is better after lesions with continuity of basal lamina of the Schwann cell tubes (nerve crush) compared with lesions with interruption of basal lamina (nerve section). Post-reinnervation maturation of myelinated nerve fibers of the cat tibial nerve was followed in chronic electrophysiologic studies after crushing, sectioning, and section+freeze lesions, and compared with morphometric analysis of the same nerves. The amplitudes of the compound nerve action potentials (CNAPs) recovered to a much lesser extent after sectioning than after crushing the nerve. This difference could be related to a smaller number of large fibers, a greater degree of sprouting after sectioning than after crushing, or less synchronization of conduction in regenerated fibers. In comparison, the compound muscle action potentials (CMAPs) recovered to a greater extent than the CNAP after sectioning and section+freeze, though not to the same degree or as fast as after crushing. The difference between the recovery of the CNAP and the CMAP could be due to better regeneration of motor fibers, to differences in the size of motor units or to a better summation of motor unit action potentials. The maximal conduction velocities (CV) in mixed nerve and in motor fibers increased faster after crushing than after sectioning and section+freeze to 60%-70% of control values. The diameters of the largest myelinated fibers increased in all lesions to about 80% of controls. The relation between fiber diameter and CV was influenced by remodeling of myelin during maturation. Hence, long-term functional recovery is influenced by the nature of the nerve lesion, and a smaller proportion of fibers recovered functionally after nerve section than after crush.
Collapse
Affiliation(s)
- K Fugleholm
- Institute of Medical Physiology, Panum Institute, University of Copenhagen, Denmark
| | | | | |
Collapse
|
49
|
Abstract
The interplay between growing axons and the extracellular substrate is pivotal for directing axonal outgrowth during development and regeneration. Here we show an important role for the neuronal cell adhesion molecule alpha7beta1 integrin during peripheral nerve regeneration. Axotomy led to a strong increase of this integrin on regenerating motor and sensory neurons, but not on the normally nonregenerating CNS neurons. alpha7 and beta1 subunits were present on the axons and their growth cones in the regenerating facial nerve. Transgenic deletion of the alpha7 subunit caused a significant reduction of axonal elongation. The associated delay in the reinnervation of the whiskerpad, a peripheral target of the facial motor neurons, points to an important role for this integrin in the successful execution of axonal regeneration.
Collapse
|
50
|
Che YH, Tamatani M, Yamashita T, Gomi F, Ogawa S, Tohyama M. Changes in mRNA of protein inhibitor of neuronal nitric oxide synthase following facial nerve transection. J Chem Neuroanat 2000; 17:199-206. [PMID: 10697246 DOI: 10.1016/s0891-0618(99)00046-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Protein inhibitor of neuronal nitric oxide synthase (PIN) is reported as the protein inhibiting neuronal nitric oxide synthase (nNOS) activity by preventing dimerization of nNOS. It was also reported that PIN inhibits the activity of all nitric oxide synthase (NOS) isozymes. We examined the effects of facial nerve transection on PIN mRNA and NOS expression by in situ hybridization for PIN mRNA and nicotinamide adenine dinucleotide phosphate diaphorase (NADPH-d) staining. PIN mRNA was initially expressed and transiently increased from 3 to 5 days and returned to the basal level at 7 days after axotomy in the motoneurons of the facial nucleus. NADPH-d-positive motoneurons were found from 7 days post-operation in the facial nucleus. These results suggest that PIN may interact with NOS from 7 days post-operation.
Collapse
Affiliation(s)
- Y H Che
- Department of Anatomy and Neuroscience, Osaka University Medical School, Japan.
| | | | | | | | | | | |
Collapse
|