1
|
Ryu Y, Wague A, Liu X, Feeley BT, Ferguson AR, Morioka K. Cellular signaling pathways in the nervous system activated by various mechanical and electromagnetic stimuli. Front Mol Neurosci 2024; 17:1427070. [PMID: 39430293 PMCID: PMC11486767 DOI: 10.3389/fnmol.2024.1427070] [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: 05/02/2024] [Accepted: 09/25/2024] [Indexed: 10/22/2024] Open
Abstract
Mechanical stimuli, such as stretch, shear stress, or compression, activate a range of biomolecular responses through cellular mechanotransduction. In the nervous system, studies on mechanical stress have highlighted key pathophysiological mechanisms underlying traumatic injury and neurodegenerative diseases. However, the biomolecular pathways triggered by mechanical stimuli in the nervous system has not been fully explored, especially compared to other body systems. This gap in knowledge may be due to the wide variety of methods and definitions used in research. Additionally, as mechanical stimulation techniques such as ultrasound and electromagnetic stimulation are increasingly utilized in psychological and neurorehabilitation treatments, it is vital to understand the underlying biological mechanisms in order to develop accurate pathophysiological models and enhance therapeutic interventions. This review aims to summarize the cellular signaling pathways activated by various mechanical and electromagnetic stimuli with a particular focus on the mammalian nervous system. Furthermore, we briefly discuss potential cellular mechanosensors involved in these processes.
Collapse
Affiliation(s)
- Youngjae Ryu
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Aboubacar Wague
- Department of Veterans Affairs, San Francisco Veterans Affairs Medical Center, San Francisco, CA, United States
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Xuhui Liu
- Department of Veterans Affairs, San Francisco Veterans Affairs Medical Center, San Francisco, CA, United States
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Brian T. Feeley
- Department of Veterans Affairs, San Francisco Veterans Affairs Medical Center, San Francisco, CA, United States
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Adam R. Ferguson
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
- Brain and Spinal Injury Center, Zuckerberg San Francisco General Hospital and Trauma Center, San Francisco, CA, United States
- San Francisco Veterans Affairs Healthcare System, San Francisco, CA, United States
| | - Kazuhito Morioka
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, CA, United States
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
- Brain and Spinal Injury Center, Zuckerberg San Francisco General Hospital and Trauma Center, San Francisco, CA, United States
- Zuckerberg San Francisco General Hospital and Trauma CenterOrthopaedic Trauma Institute, , San Francisco, CA, United States
| |
Collapse
|
2
|
Lukomska A, Rheaume BA, Frost MP, Theune WC, Xing J, Damania A, Trakhtenberg EF. Augmenting fibronectin levels in injured adult CNS promotes axon regeneration in vivo. Exp Neurol 2024; 379:114877. [PMID: 38944331 PMCID: PMC11283980 DOI: 10.1016/j.expneurol.2024.114877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 06/06/2024] [Accepted: 06/25/2024] [Indexed: 07/01/2024]
Abstract
In an attempt to repair injured central nervous system (CNS) nerves/tracts, immune cells are recruited into the injury site, but endogenous response in adult mammals is insufficient for promoting regeneration of severed axons. Here, we found that a portion of retinal ganglion cell (RGC) CNS projection neurons that survive after optic nerve crush (ONC) injury are enriched for and upregulate fibronectin (Fn)-interacting integrins Itga5 and ItgaV, and that Fn promotes long-term survival and long-distance axon regeneration of a portion of axotomized adult RGCs in culture. We then show that, Fn is developmentally downregulated in the axonal tracts of optic nerve and spinal cord, but injury-activated macrophages/microglia upregulate Fn while axon regeneration-promoting zymosan augments their recruitment (and thereby increases Fn levels) in the injured optic nerve. Finally, we found that Fn's RGD motif, established to interact with Itga5 and ItgaV, promotes long-term survival and long-distance axon regeneration of adult RGCs after ONC in vivo, with some axons reaching the optic chiasm when co-treated with Rpl7a gene therapy. Thus, experimentally augmenting Fn levels in the injured CNS is a promising approach for therapeutic neuroprotection and axon regeneration of at least a portion of neurons.
Collapse
Affiliation(s)
- Agnieszka Lukomska
- Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA
| | - Bruce A Rheaume
- Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA
| | - Matthew P Frost
- Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA
| | - William C Theune
- Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA
| | - Jian Xing
- Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA
| | - Ashiti Damania
- Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA
| | - Ephraim F Trakhtenberg
- Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Ave., Farmington, CT 06030, USA..
| |
Collapse
|
3
|
Goncalves MB, Wu Y, Clarke E, Grist J, Moehlin J, Mendoza-Parra MA, Hobbs C, Kalindjian B, Fok H, Mander AP, Hassanin H, Bendel D, Täubel J, Mant T, Carlstedt T, Jack J, Corcoran JPT. C286, an orally available retinoic acid receptor β agonist drug, regulates multiple pathways to achieve spinal cord injury repair. Front Mol Neurosci 2024; 17:1411384. [PMID: 39228795 PMCID: PMC11368863 DOI: 10.3389/fnmol.2024.1411384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 07/29/2024] [Indexed: 09/05/2024] Open
Abstract
Retinoic acid receptor β2 (RARβ2) is an emerging therapeutic target for spinal cord injuries (SCIs) with a unique multimodal regenerative effect. We have developed a first-in-class RARβ agonist drug, C286, that modulates neuron-glial pathways to induce functional recovery in a rodent model of sensory root avulsion. Here, using genome-wide and pathway enrichment analysis of avulsed rats' spinal cords, we show that C286 also influences the extracellular milieu (ECM). Protein expression studies showed that C286 upregulates tenascin-C, integrin-α9, and osteopontin in the injured cord. Similarly, C286 remodulates these ECM molecules, hampers inflammation and prevents tissue loss in a rodent model of spinal cord contusion C286. We further demonstrate C286's efficacy in human iPSC-derived neurons, with treatment resulting in a significant increase in neurite outgrowth. Additionally, we identify a putative efficacy biomarker, S100B, which plasma levels correlated with axonal regeneration in nerve-injured rats. We also found that other clinically available retinoids, that are not RARβ specific agonists, did not lead to functional recovery in avulsed rats, demonstrating the requirement for RARβ specific pathways in regeneration. In a Phase 1 trial, the single ascending dose (SAD) cohorts showed increases in expression of RARβ2 in white blood cells correlative to increased doses and at the highest dose administered, the pharmacokinetics were similar to the rat proof of concept (POC) studies. Collectively, our data suggests that C286 signalling in neurite/axonal outgrowth is conserved between species and across nerve injuries. This warrants further clinical testing of C286 to ascertain POC in a broad spectrum of neurodegenerative conditions.
Collapse
Affiliation(s)
- Maria B. Goncalves
- Neuroscience Drug Discovery Unit, Wolfson Sensory, Pain and Regeneration Centre, King's College London, Guy's Campus, London, United Kingdom
| | - Yue Wu
- Neuroscience Drug Discovery Unit, Wolfson Sensory, Pain and Regeneration Centre, King's College London, Guy's Campus, London, United Kingdom
| | - Earl Clarke
- Neuroscience Drug Discovery Unit, Wolfson Sensory, Pain and Regeneration Centre, King's College London, Guy's Campus, London, United Kingdom
| | - John Grist
- Neuroscience Drug Discovery Unit, Wolfson Sensory, Pain and Regeneration Centre, King's College London, Guy's Campus, London, United Kingdom
| | - Julien Moehlin
- UMR 8030 Génomique Métabolique, Genoscope, Institut François Jacob, CEA, CNRS, University of Évry-val-d'Essonne, University Paris-Saclay, Évry, France
| | - Marco Antonio Mendoza-Parra
- UMR 8030 Génomique Métabolique, Genoscope, Institut François Jacob, CEA, CNRS, University of Évry-val-d'Essonne, University Paris-Saclay, Évry, France
| | - Carl Hobbs
- Neuroscience Drug Discovery Unit, Wolfson Sensory, Pain and Regeneration Centre, King's College London, Guy's Campus, London, United Kingdom
| | - Barret Kalindjian
- Neuroscience Drug Discovery Unit, Wolfson Sensory, Pain and Regeneration Centre, King's College London, Guy's Campus, London, United Kingdom
| | - Henry Fok
- NIHR Biomedical Research Centre at Guy's and St Thomas' NHS Foundation Trust and King's College London, London, United Kingdom
| | - Adrian P. Mander
- Centre for Trials Research, Cardiff University, Cardiff, United Kingdom
| | - Hana Hassanin
- Surrey Clinical Research Centre, University of Surrey, Guildford, United Kingdom
| | - Daryl Bendel
- Surrey Clinical Research Centre, University of Surrey, Guildford, United Kingdom
| | - Jörg Täubel
- Richmond Pharmacology Limited, London, United Kingdom
| | - Tim Mant
- NIHR Biomedical Research Centre at Guy's and St Thomas' NHS Foundation Trust and King's College London, London, United Kingdom
| | - Thomas Carlstedt
- Neuroscience Drug Discovery Unit, Wolfson Sensory, Pain and Regeneration Centre, King's College London, Guy's Campus, London, United Kingdom
| | - Julian Jack
- Neuroscience Drug Discovery Unit, Wolfson Sensory, Pain and Regeneration Centre, King's College London, Guy's Campus, London, United Kingdom
| | - Jonathan P. T. Corcoran
- Neuroscience Drug Discovery Unit, Wolfson Sensory, Pain and Regeneration Centre, King's College London, Guy's Campus, London, United Kingdom
| |
Collapse
|
4
|
Davis-Lunn M, Goult BT, Andrews MR. Clutching at Guidance Cues: The Integrin-FAK Axis Steers Axon Outgrowth. BIOLOGY 2023; 12:954. [PMID: 37508384 PMCID: PMC10376711 DOI: 10.3390/biology12070954] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/27/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023]
Abstract
Integrin receptors are essential contributors to neurite outgrowth and axon elongation. Activated integrins engage components of the extracellular matrix, enabling the growth cone to form point contacts, which connect the extracellular substrate to dynamic intracellular protein complexes. These adhesion complexes facilitate efficient growth cone migration and neurite extension. Major signalling pathways mediated by the adhesion complex are instigated by focal adhesion kinase (FAK), whilst axonal guidance molecules present in vivo promote growth cone turning or retraction by local modulation of FAK activity. Activation of FAK is marked by phosphorylation following integrin engagement, and this activity is tightly regulated during neurite outgrowth. FAK inhibition slows neurite outgrowth by reducing point contact turnover; however, mutant FAK constructs with enhanced activity stimulate aberrant outgrowth. Importantly, FAK is a major structural component of maturing adhesion sites, which provide the platform for actin polymerisation to drive leading edge advance. In this review, we discuss the coordinated signalling of integrin receptors and FAK, as well as their role in regulating neurite outgrowth and axon elongation. We also discuss the importance of the integrin-FAK axis in vivo, as integrin expression and activation are key determinants of successful axon regeneration following injury.
Collapse
Affiliation(s)
- Mathew Davis-Lunn
- Faculty of Environmental and Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Benjamin T Goult
- School of Biosciences, University of Kent, Canterbury CT2 7NJ, UK
| | - Melissa R Andrews
- Faculty of Environmental and Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
- Centre for Human Development, Stem Cells and Regeneration, School of Biological Sciences, University of Southampton, Southampton SO17 1BJ, UK
| |
Collapse
|
5
|
Sefiani A, Rusyn I, Geoffroy CG. Novel adult cortical neuron processing and screening method illustrates sex- and age-dependent effects of pharmaceutical compounds. Sci Rep 2022; 12:13125. [PMID: 35908049 PMCID: PMC9338961 DOI: 10.1038/s41598-022-17389-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 07/25/2022] [Indexed: 11/22/2022] Open
Abstract
Neurodegenerative diseases and neurotraumatic injuries are typically age-associated disorders that can reduce neuron survival, neurite outgrowth, and synaptic plasticity leading to loss of cognitive capacity, executive function, and motor control. In pursuit of reducing the loss of said neurological functions, novel compounds are sought that promote neuron viability, neuritogenesis, and/or synaptic plasticity. Current high content in vitro screenings typically use cells that are iPSC-derived, embryonic, or originate from post-natal tissues; however, most patients suffering from neurodegenerative diseases and neurotrauma are of middle-age and older. The chasm in maturity between the neurons used in drug screens and those in a target population is a barrier for translational success of in vitro results. It has been historically challenging to culture adult neurons let alone conduct screenings; therefore, age-appropriate drug screenings have previously not been plausible. We have modified Miltenyi's protocol to increase neuronal yield, neuron purity, and neural viability at a reduced cost to expand our capacity to screen compounds directly in primary adult neurons. To our knowledge, we developed the first morphology-based screening system using adult cortical neurons and the first to incorporate age and sex as biological variables in a screen using adult cortical neurons. By using primary adult cortical neurons from mice that were 4 to 48 weeks old for screening pharmaceutical agents, we have demonstrated age- and sex-dependent effects on neuritogenesis and neuron survival in vitro. Utilizing age- and sex-appropriate in vitro models to find novel compounds increasing neuron survival and neurite outgrowth, made possible by our modified adult neuron processing method, will greatly increase the relevance of in vitro screening for finding neuroprotective compounds.
Collapse
Affiliation(s)
- Arthur Sefiani
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University, Bryan, TX, 77807, USA
| | - Ivan Rusyn
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX, 77843, USA
| | - Cédric G Geoffroy
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University, Bryan, TX, 77807, USA.
| |
Collapse
|
6
|
Integrins protect sensory neurons in models of paclitaxel-induced peripheral sensory neuropathy. Proc Natl Acad Sci U S A 2021; 118:2006050118. [PMID: 33876743 DOI: 10.1073/pnas.2006050118] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a major side effect from cancer treatment with no known method for prevention or cure in clinics. CIPN often affects unmyelinated nociceptive sensory terminals. Despite the high prevalence, molecular and cellular mechanisms that lead to CIPN are still poorly understood. Here, we used a genetically tractable Drosophila model and primary sensory neurons isolated from adult mouse to examine the mechanisms underlying CIPN and identify protective pathways. We found that chronic treatment of Drosophila larvae with paclitaxel caused degeneration and altered the branching pattern of nociceptive neurons, and reduced thermal nociceptive responses. We further found that nociceptive neuron-specific overexpression of integrins, which are known to support neuronal maintenance in several systems, conferred protection from paclitaxel-induced cellular and behavioral phenotypes. Live imaging and superresolution approaches provide evidence that paclitaxel treatment causes cellular changes that are consistent with alterations in endosome-mediated trafficking of integrins. Paclitaxel-induced changes in recycling endosomes precede morphological degeneration of nociceptive neuron arbors, which could be prevented by integrin overexpression. We used primary dorsal root ganglia (DRG) neuron cultures to test conservation of integrin-mediated protection. We show that transduction of a human integrin β-subunit 1 also prevented degeneration following paclitaxel treatment. Furthermore, endogenous levels of surface integrins were decreased in paclitaxel-treated mouse DRG neurons, suggesting that paclitaxel disrupts recycling in vertebrate sensory neurons. Altogether, our study supports conserved mechanisms of paclitaxel-induced perturbation of integrin trafficking and a therapeutic potential of restoring neuronal interactions with the extracellular environment to antagonize paclitaxel-induced toxicity in sensory neurons.
Collapse
|
7
|
Lu S, Xiong Q, Du K, Gan X, Wang X, Yang L, Wang Y, Ge F, He S. Comparative iTRAQ proteomics revealed proteins associated with lobed fin regeneration in Bichirs. Proteome Sci 2019; 17:6. [PMID: 31832023 PMCID: PMC6869209 DOI: 10.1186/s12953-019-0153-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 10/09/2019] [Indexed: 11/23/2022] Open
Abstract
Background Polypterus senegalus can fully regenerate its pectoral lobed fins, including a complex endoskeleton, with remarkable precision. However, despite the enormous potential of this species for use in medical research, its regeneration mechanisms remain largely unknown. Methods To identify the differentially expressed proteins (DEPs) during the early stages of lobed fin regeneration in P. senegalus, we performed a differential proteomic analysis using isobaric tag for relative and absolute quantitation (iTRAQ) approach based quantitative proteome from the pectoral lobed fins at 3 time points. Furthermore, we validated the changes in protein expression with multiple-reaction monitoring (MRM) analysis. Results The experiment yielded a total of 3177 proteins and 15,091 unique peptides including 1006 non-redundant (nr) DEPs. Of these, 592 were upregulated while 349 were downregulated after lobed fin amputation when compared to the original tissue. Bioinformatics analyses showed that the DEPs were mainly associated with Ribosome and RNA transport, metabolic, ECM-receptor interaction, Golgi and endoplasmic reticulum, DNA replication, and Regulation of actin cytoskeleton. Conclusions To our knowledge, this is the first proteomic research to investigate alterations in protein levels and affected pathways in bichirs’ lobe-fin/limb regeneration. In addition, our study demonstrated a highly dynamic regulation during lobed fin regeneration in P. senegalus. These results not only provide a comprehensive dataset on differentially expressed proteins during the early stages of lobe-fin/limb regeneration but also advance our understanding of the molecular mechanisms underlying lobe-fin/limb regeneration.
Collapse
Affiliation(s)
- Suxiang Lu
- 1Key Laboratory of Aquatic Biodiversity and Conservation of the Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072 Hubei China.,2Present address: Medical College of Pingdingshan University, Pingdingshan, 467000 Henan Province China
| | - Qian Xiong
- 3Key Laboratory of Algal Biology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072 Hubei China
| | - Kang Du
- 1Key Laboratory of Aquatic Biodiversity and Conservation of the Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072 Hubei China
| | - Xiaoni Gan
- 1Key Laboratory of Aquatic Biodiversity and Conservation of the Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072 Hubei China
| | - Xuzhen Wang
- 1Key Laboratory of Aquatic Biodiversity and Conservation of the Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072 Hubei China
| | - Liandong Yang
- 1Key Laboratory of Aquatic Biodiversity and Conservation of the Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072 Hubei China
| | - Ying Wang
- 1Key Laboratory of Aquatic Biodiversity and Conservation of the Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072 Hubei China
| | - Feng Ge
- 3Key Laboratory of Algal Biology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072 Hubei China
| | - Shunping He
- 1Key Laboratory of Aquatic Biodiversity and Conservation of the Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072 Hubei China
| |
Collapse
|
8
|
Gabrych DR, Lau VZ, Niwa S, Silverman MA. Going Too Far Is the Same as Falling Short †: Kinesin-3 Family Members in Hereditary Spastic Paraplegia. Front Cell Neurosci 2019; 13:419. [PMID: 31616253 PMCID: PMC6775250 DOI: 10.3389/fncel.2019.00419] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 09/02/2019] [Indexed: 01/18/2023] Open
Abstract
Proper intracellular trafficking is essential for neuronal development and function, and when any aspect of this process is dysregulated, the resulting "transportopathy" causes neurological disorders. Hereditary spastic paraplegias (HSPs) are a family of such diseases attributed to over 80 spastic gait genes (SPG), specifically characterized by lower extremity spasticity and weakness. Multiple genes in the trafficking pathway such as those relating to microtubule structure and function and organelle biogenesis are representative disease loci. Microtubule motor proteins, or kinesins, are also causal in HSP, specifically mutations in Kinesin-I/KIF5A (SPG10) and two kinesin-3 family members; KIF1A (SPG30) and KIF1C (SPG58). KIF1A is a motor enriched in neurons, and involved in the anterograde transport of a variety of vesicles that contribute to pre- and post-synaptic assembly, autophagic processes, and neuron survival. KIF1C is ubiquitously expressed and, in addition to anterograde cargo transport, also functions in retrograde transport between the Golgi and the endoplasmic reticulum. Only a handful of KIF1C cargos have been identified; however, many have crucial roles such as neuronal differentiation, outgrowth, plasticity and survival. HSP-related kinesin-3 mutants are characterized mainly as loss-of-function resulting in deficits in motility, regulation, and cargo binding. Gain-of-function mutants are also seen, and are characterized by increased microtubule-on rates and hypermotility. Both sets of mutations ultimately result in misdelivery of critical cargos within the neuron. This likely leads to deleterious cell biological cascades that likely underlie or contribute to HSP clinical pathology and ultimately, symptomology. Due to the paucity of histopathological or cell biological data assessing perturbations in cargo localization, it has been difficult to positively link these mutations to the outcomes seen in HSPs. Ultimately, the goal of this review is to encourage future academic and clinical efforts to focus on "transportopathies" through a cargo-centric lens.
Collapse
Affiliation(s)
- Dominik R Gabrych
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, Canada
| | - Victor Z Lau
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, Canada
| | - Shinsuke Niwa
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Japan
| | - Michael A Silverman
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, Canada.,Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, BC, Canada
| |
Collapse
|
9
|
Zhao X, Li Z, Liang S, Li S, Ren J, Li B, Zhu Y, Xia M. Different epidermal growth factor receptor signaling pathways in neurons and astrocytes activated by extracellular matrix after spinal cord injury. Neurochem Int 2019; 129:104500. [PMID: 31295509 DOI: 10.1016/j.neuint.2019.104500] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 06/25/2019] [Accepted: 07/07/2019] [Indexed: 12/26/2022]
Abstract
Spinal cord injury (SCI) is a serious central nervous system (CNS) trauma that results in permanent and severe disability. The extracellular matrix (ECM) can affect the activation of extracellular signal-regulated kinase 1/2 (ERK1/2) by interacting with the ERK integrin subunits. In this study, we built a model of SCI with glial fibrillary acidic protein-green fluorescent protein (GFAP-GFP) and thymus cell antigen 1-yellow fluorescent protein-H (Thy1-YFPH) in mice that express specific transgenes in their astrocytes or neurons. Then, we collected spinal cord neurons or astrocytes by fluorescence-activated cell sorting (FACS). In this way, we investigated the SCI-induced phosphorylation of ERK1/2 and epidermal growth factor receptor (EGFR) in neurons and astrocytes, and we discovered that the SCI-induced EGFR signaling pathways differed between neurons and astrocytes. In the present study, we found that the Src-dependent phosphorylation of EGFR induced by SCI occurred only in neurons, not in astrocytes. This phenomenon may be due to the involvement of Thy-1, which promoted the binding between Src and EGFR in neurons after SCI. In addition, the expression of the integrin subunits after SCI differed between neurons and astrocytes. Our present study shows that the EGFR signaling pathway triggered by SCI in neurons differed from the EGFR signaling pathway triggered in astrocytes, a finding that may help to pave the way for clinical trials of therapies that inhibit EGFR signaling pathways after SCI.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Orthopaedics, The First Hospital of China Medical University, Shenyang, People's Republic of China; Department of Operating Room, First Affiliated Hospital, China Medical University, Shenyang, People's Republic of China
| | - Zexiong Li
- Practical Teaching Center, School of Forensic Medicine, China Medical University, Shenyang, People's Republic of China
| | - Shanshan Liang
- Practical Teaching Center, School of Forensic Medicine, China Medical University, Shenyang, People's Republic of China
| | - Shuai Li
- Practical Teaching Center, School of Forensic Medicine, China Medical University, Shenyang, People's Republic of China
| | - Jiaan Ren
- Department of Orthopaedics, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Baoman Li
- Practical Teaching Center, School of Forensic Medicine, China Medical University, Shenyang, People's Republic of China. http://
| | - Yue Zhu
- Department of Orthopaedics, The First Hospital of China Medical University, Shenyang, People's Republic of China. http://
| | - Maosheng Xia
- Department of Orthopaedics, The First Hospital of China Medical University, Shenyang, People's Republic of China.
| |
Collapse
|
10
|
Advanced glycation endproducts produced by in vitro glycation of type I collagen modulate the functional and secretory behavior of dorsal root ganglion cells cultivated in two-dimensional system. Exp Cell Res 2019; 382:111475. [PMID: 31255600 DOI: 10.1016/j.yexcr.2019.06.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 06/15/2019] [Accepted: 06/19/2019] [Indexed: 12/24/2022]
Abstract
Advanced glycation end-products (AGEs) are proteins/lipids that are glycated upon sugar exposure and are often increased during inflammatory diseases such as osteoarthritis and neurodegenerative disorders. Here, we developed an extracellular matrix (ECM) using glycated type I collagen (ECM-GC), which produced similar levels of AGEs to those detected in the sera of arthritic mice. In order to determine whether AGEs were sufficient to stimulate sensory neurons, dorsal root ganglia (DRGs) cells were cultured on ECM-GC or ECM-NC-coated plates. ECM-GC or ECM-NC were favorable for DRG cells expansion. However, ECM-GC cultivated neurons displayed thinner F-actin filaments, rounded morphology, and reduced neuron interconnection compared to ECM-NC. In addition, ECM-GC did not affect RAGE expression levels in the neurons, although induced rapid p38, MAPK and ERK activation. Finally, ECM-GC stimulated the secretion of nitrite and TNF-α by DRG cells. Taken together, our in vitro glycated ECM model suitably mimics the in vivo microenvironment of inflammatory disorders and provides new insights into the role of ECM impairment as a nociceptive stimulus.
Collapse
|
11
|
Forbes LH, Andrews MR. Grafted Human iPSC-Derived Neural Progenitor Cells Express Integrins and Extend Long-Distance Axons Within the Developing Corticospinal Tract. Front Cell Neurosci 2019; 13:26. [PMID: 30809126 PMCID: PMC6380224 DOI: 10.3389/fncel.2019.00026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 01/22/2019] [Indexed: 12/11/2022] Open
Abstract
After spinal cord injury (SCI), regeneration of adult motor axons such as axons in the corticospinal tract (CST) is severely limited. Alongside the inhibitory lesion environment, most neuronal subtypes in the mature central nervous system (CNS) are intrinsically unrepairable. With age, expression of growth-promoting proteins in neurons, such as integrins, declines. Integrin receptors allow communication between the extracellular matrix (ECM) and cell cytoskeleton and their expression in axons facilitates growth and guidance throughout the ECM. The α9β1 integrin heterodimer binds to tenascin-C (TN-C), an ECM glycoprotein expressed during development and after injury. In the mature CST however, expression of the α9 integrin subunit is downregulated, adding to the intrinsic inability of axons to regenerate. Our previous work has shown the α9 integrin subunit is not trafficked within axons of mature CST or rubrospinal tracts (RSTs). Thus, here we have utilized human induced pluripotent stem cell (iPSC)-derived neural progenitor cells (NPCs) to increase expression of α9 integrinwithin the developing rat CST. We demonstrate that human NPCs (hNPCs) express endogenous levels of both α9 and β1 integrin subunits as well as cortical neuron markers such as chicken ovalbumin upstream promoter transcription factor (COUP-TF) interacting protein 2 (Ctip2) and T-box brain 1 (Tbr1). In addition, lentivirus-mediated α9 integrin overexpression in hNPCs resulted in increased neurite outgrowth in the presence of TN-C in vitro. Following transplantation into the sensorimotor cortex of newborn rats, both wild type (WT) and α9-expressing hNPCs extend along the endogenous CST and retain expression of α9 throughout the length of the axonal compartment for up to 8 weeks following transplantation. These data highlight the growth potential of transplanted human iPSCs which may be a future target for regenerative therapies after nervous system injury.
Collapse
Affiliation(s)
- Lindsey H Forbes
- School of Medicine, University of St Andrews, St Andrews, United Kingdom
| | - Melissa R Andrews
- School of Medicine, University of St Andrews, St Andrews, United Kingdom.,Biological Sciences, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
12
|
Petrova V, Eva R. The Virtuous Cycle of Axon Growth: Axonal Transport of Growth-Promoting Machinery as an Intrinsic Determinant of Axon Regeneration. Dev Neurobiol 2018; 78:898-925. [PMID: 29989351 DOI: 10.1002/dneu.22608] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 05/25/2018] [Accepted: 05/26/2018] [Indexed: 02/02/2023]
Abstract
Injury to the brain and spinal cord has devastating consequences because adult central nervous system (CNS) axons fail to regenerate. Injury to the peripheral nervous system (PNS) has a better prognosis, because adult PNS neurons support robust axon regeneration over long distances. CNS axons have some regenerative capacity during development, but this is lost with maturity. Two reasons for the failure of CNS regeneration are extrinsic inhibitory molecules, and a weak intrinsic capacity for growth. Extrinsic inhibitory molecules have been well characterized, but less is known about the neuron-intrinsic mechanisms which prevent axon re-growth. Key signaling pathways and genetic/epigenetic factors have been identified which can enhance regenerative capacity, but the precise cellular mechanisms mediating their actions have not been characterized. Recent studies suggest that an important prerequisite for regeneration is an efficient supply of growth-promoting machinery to the axon; however, this appears to be lacking from non-regenerative axons in the adult CNS. In the first part of this review, we summarize the evidence linking axon transport to axon regeneration. We discuss the developmental decline in axon regeneration capacity in the CNS, and comment on how this is paralleled by a similar decline in the selective axonal transport of regeneration-associated receptors such as integrins and growth factor receptors. In the second part, we discuss the mechanisms regulating selective polarized transport within neurons, how these relate to the intrinsic control of axon regeneration, and whether they can be targeted to enhance regenerative capacity. © 2018 Wiley Periodicals, Inc. Develop Neurobiol 00: 000-000, 2018.
Collapse
Affiliation(s)
- Veselina Petrova
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 OPY, United Kingdom
| | - Richard Eva
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 OPY, United Kingdom
| |
Collapse
|
13
|
Glycans and glycosaminoglycans in neurobiology: key regulators of neuronal cell function and fate. Biochem J 2018; 475:2511-2545. [PMID: 30115748 DOI: 10.1042/bcj20180283] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 07/14/2018] [Accepted: 07/18/2018] [Indexed: 12/16/2022]
Abstract
The aim of the present study was to examine the roles of l-fucose and the glycosaminoglycans (GAGs) keratan sulfate (KS) and chondroitin sulfate/dermatan sulfate (CS/DS) with selected functional molecules in neural tissues. Cell surface glycans and GAGs have evolved over millions of years to become cellular mediators which regulate fundamental aspects of cellular survival. The glycocalyx, which surrounds all cells, actuates responses to growth factors, cytokines and morphogens at the cellular boundary, silencing or activating downstream signaling pathways and gene expression. In this review, we have focused on interactions mediated by l-fucose, KS and CS/DS in the central and peripheral nervous systems. Fucose makes critical contributions in the area of molecular recognition and information transfer in the blood group substances, cytotoxic immunoglobulins, cell fate-mediated Notch-1 interactions, regulation of selectin-mediated neutrophil extravasation in innate immunity and CD-34-mediated new blood vessel development, and the targeting of neuroprogenitor cells to damaged neural tissue. Fucosylated glycoproteins regulate delivery of synaptic neurotransmitters and neural function. Neural KS proteoglycans (PGs) were examined in terms of cellular regulation and their interactive properties with neuroregulatory molecules. The paradoxical properties of CS/DS isomers decorating matrix and transmembrane PGs and the positive and negative regulatory cues they provide to neurons are also discussed.
Collapse
|
14
|
Nieuwenhuis B, Haenzi B, Andrews MR, Verhaagen J, Fawcett JW. Integrins promote axonal regeneration after injury of the nervous system. Biol Rev Camb Philos Soc 2018; 93:1339-1362. [PMID: 29446228 PMCID: PMC6055631 DOI: 10.1111/brv.12398] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 12/23/2017] [Accepted: 01/11/2018] [Indexed: 12/13/2022]
Abstract
Integrins are cell surface receptors that form the link between extracellular matrix molecules of the cell environment and internal cell signalling and the cytoskeleton. They are involved in several processes, e.g. adhesion and migration during development and repair. This review focuses on the role of integrins in axonal regeneration. Integrins participate in spontaneous axonal regeneration in the peripheral nervous system through binding to various ligands that either inhibit or enhance their activation and signalling. Integrin biology is more complex in the central nervous system. Integrins receptors are transported into growing axons during development, but selective polarised transport of integrins limits the regenerative response in adult neurons. Manipulation of integrins and related molecules to control their activation state and localisation within axons is a promising route towards stimulating effective regeneration in the central nervous system.
Collapse
Affiliation(s)
- Bart Nieuwenhuis
- John van Geest Centre for Brain Repair, Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0PYU.K.
- Laboratory for Regeneration of Sensorimotor SystemsNetherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences (KNAW)1105 BAAmsterdamThe Netherlands
| | - Barbara Haenzi
- John van Geest Centre for Brain Repair, Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0PYU.K.
| | | | - Joost Verhaagen
- Laboratory for Regeneration of Sensorimotor SystemsNetherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences (KNAW)1105 BAAmsterdamThe Netherlands
- Centre for Neurogenomics and Cognitive Research, Amsterdam NeuroscienceVrije Universiteit Amsterdam1081 HVAmsterdamThe Netherlands
| | - James W. Fawcett
- John van Geest Centre for Brain Repair, Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0PYU.K.
- Centre of Reconstructive NeuroscienceInstitute of Experimental Medicine142 20Prague 4Czech Republic
| |
Collapse
|
15
|
DeVault L, Li T, Izabel S, Thompson-Peer KL, Jan LY, Jan YN. Dendrite regeneration of adult Drosophila sensory neurons diminishes with aging and is inhibited by epidermal-derived matrix metalloproteinase 2. Genes Dev 2018; 32:402-414. [PMID: 29563183 PMCID: PMC5900713 DOI: 10.1101/gad.308270.117] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 02/26/2018] [Indexed: 11/30/2022]
Abstract
DeVault et al. show that the capacity for regeneration was present in adult neurons but diminished as the animal aged. The regenerated dendrites showed preferential alignment with the extracellular matrix, and inhibition of matrix metalloproteinase 2 led to increased dendrite regeneration. Dendrites possess distinct structural and functional properties that enable neurons to receive information from the environment as well as other neurons. Despite their key role in neuronal function, current understanding of the ability of neurons to regenerate dendrites is lacking. This study characterizes the structural and functional capacity for dendrite regeneration in vivo in adult animals and examines the effect of neuronal maturation on dendrite regeneration. We focused on the class IV dendritic arborization (c4da) neuron of the Drosophila sensory system, which has a dendritic arbor that undergoes dramatic remodeling during the first 3 d of adult life and then maintains a relatively stable morphology thereafter. Using a laser severing paradigm, we monitored regeneration after acute and spatially restricted injury. We found that the capacity for regeneration was present in adult neurons but diminished as the animal aged. Regenerated dendrites recovered receptive function. Furthermore, we found that the regenerated dendrites show preferential alignment with the extracellular matrix (ECM). Finally, inhibition of ECM degradation by inhibition of matrix metalloproteinase 2 (Mmp2) to preserve the extracellular environment characteristics of young adults led to increased dendrite regeneration. These results demonstrate that dendrites retain regenerative potential throughout adulthood and that regenerative capacity decreases with aging.
Collapse
Affiliation(s)
- Laura DeVault
- Howard Hughes Medical Institute, Department of Physiology, Department of Biochemistry and Biophysics, University of California at San Francisco, San Francisco, California 94158, USA
| | - Tun Li
- Howard Hughes Medical Institute, Department of Physiology, Department of Biochemistry and Biophysics, University of California at San Francisco, San Francisco, California 94158, USA
| | - Sarah Izabel
- Howard Hughes Medical Institute, Department of Physiology, Department of Biochemistry and Biophysics, University of California at San Francisco, San Francisco, California 94158, USA
| | - Katherine L Thompson-Peer
- Howard Hughes Medical Institute, Department of Physiology, Department of Biochemistry and Biophysics, University of California at San Francisco, San Francisco, California 94158, USA
| | - Lily Yeh Jan
- Howard Hughes Medical Institute, Department of Physiology, Department of Biochemistry and Biophysics, University of California at San Francisco, San Francisco, California 94158, USA
| | - Yuh Nung Jan
- Howard Hughes Medical Institute, Department of Physiology, Department of Biochemistry and Biophysics, University of California at San Francisco, San Francisco, California 94158, USA
| |
Collapse
|
16
|
Abstract
Integrin activation is essential for creating functional transmembrane receptors capable of inducing downstream cellular effects such as cell migration, cell spreading, neurite outgrowth and axon regeneration. Integrins are bidirectional signalling molecules that mediate their effects by 'inside-out' and 'outside-in' signalling. This review will provide a detailed overview of integrin activation focusing on intracellular activation in neurons and discussing direct implications in the regulation of neurite outgrowth and axon regeneration.
Collapse
Affiliation(s)
- Menghon Cheah
- John van Geest Centre for Brain Repair, University of Cambridge, Cambridge CB2 0PY, UK.
- Centre for Developmental Neurobiology, King's College London, London SE1 1UL, UK.
| | - Melissa R Andrews
- Department of Biological Sciences, University of Southampton, Life Sciences Bldg 85, Highfield Campus, Southampton SO17 1BJ, UK.
| |
Collapse
|
17
|
Yu P, Pearson CS, Geller HM. Flexible Roles for Proteoglycan Sulfation and Receptor Signaling. Trends Neurosci 2018; 41:47-61. [PMID: 29150096 PMCID: PMC5748001 DOI: 10.1016/j.tins.2017.10.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 10/19/2017] [Accepted: 10/25/2017] [Indexed: 11/25/2022]
Abstract
Proteoglycans (PGs) in the extracellular matrix (ECM) play vital roles in axon growth and navigation, plasticity, and regeneration of injured neurons. Different classes of PGs may support or inhibit cell growth, and their functions are determined in part by highly specific structural features. Among these, the pattern of sulfation on the PG sugar chains is a paramount determinant of a diverse and flexible set of outcomes. Recent studies of PG sulfation illustrate the challenges of attributing biological actions to specific sulfation patterns, and suggest ways in which highly similar molecules may exert opposing effects on neurons. The receptors for PGs, which have yet to be fully characterized, display a similarly nuanced spectrum of effects. Different classes of PG function via overlapping families of receptors and signaling pathways. This enables them to control axon growth and guidance with remarkable specificity, but it poses challenges for determining the precise binding interactions and downstream effects of different PGs and their assorted sulfated epitopes. This review examines existing and emerging evidence for the roles of PG sulfation and receptor interactions in determining how these complex molecules influence neuronal development, growth, and function.
Collapse
Affiliation(s)
- Panpan Yu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration; Ministry of Education Joint International Research Laboratory of CNS Regeneration, Jinan University, Guangzhou 510632, China.
| | - Craig S Pearson
- Laboratory of Developmental Neurobiology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Herbert M Geller
- Laboratory of Developmental Neurobiology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
18
|
Forbes LH, Andrews MR. Restoring axonal localization and transport of transmembrane receptors to promote repair within the injured CNS: a critical step in CNS regeneration. Neural Regen Res 2017; 12:27-30. [PMID: 28250734 PMCID: PMC5319227 DOI: 10.4103/1673-5374.198968] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Each neuronal subtype is distinct in how it develops, responds to environmental cues, and whether it is capable of mounting a regenerative response following injury. Although the adult central nervous system (CNS) does not regenerate, several experimental interventions have been trialled with successful albeit limited instances of axonal repair. We highlight here some of these approaches including extracellular matrix (ECM) modification, cellular grafting, gene therapy-induced replacement of proteins, as well as application of biomaterials. We also review the recent report demonstrating the failure of axonal localization and transport of growth-promoting receptors within certain classes of mature neurons. More specifically, we discuss an inability of integrin receptors to localize within the axonal compartment of mature motor neurons such as in the corticospinal and rubrospinal tracts, whereas in immature neurons of those pathways and in mature sensory tracts such as in the optic nerve and dorsal column pathways these receptors readily localize within axons. Furthermore we assert that this failure of axonal localization contributes to the intrinsic inability of axonal regeneration. We conclude by highlighting the necessity for both combined therapies as well as a targeted approach specific to both age and neuronal subtype will be required to induce substantial CNS repair.
Collapse
Affiliation(s)
- Lindsey H Forbes
- School of Medicine, University of St. Andrews, St. Andrews, United Kingdom
| | - Melissa R Andrews
- School of Medicine, University of St. Andrews, St. Andrews, United Kingdom; Biological Sciences, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
19
|
Preferential Enhancement of Sensory and Motor Axon Regeneration by Combining Extracellular Matrix Components with Neurotrophic Factors. Int J Mol Sci 2016; 18:ijms18010065. [PMID: 28036084 PMCID: PMC5297700 DOI: 10.3390/ijms18010065] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 11/28/2016] [Accepted: 12/24/2016] [Indexed: 11/29/2022] Open
Abstract
After peripheral nerve injury, motor and sensory axons are able to regenerate but inaccuracy of target reinnervation leads to poor functional recovery. Extracellular matrix (ECM) components and neurotrophic factors (NTFs) exert their effect on different neuronal populations creating a suitable environment to promote axonal growth. Here, we assessed in vitro and in vivo the selective effects of combining different ECM components with NTFs on motor and sensory axons regeneration and target reinnervation. Organotypic cultures with collagen, laminin and nerve growth factor (NGF)/neurotrophin-3 (NT3) or collagen, fibronectin and brain-derived neurotrophic factor (BDNF) selectively enhanced sensory neurite outgrowth of DRG neurons and motor neurite outgrowth from spinal cord slices respectively. For in vivo studies, the rat sciatic nerve was transected and repaired with a silicone tube filled with a collagen and laminin matrix with NGF/NT3 encapsulated in poly(lactic-co-glycolic acid) (PLGA) microspheres (MP) (LM + MP.NGF/NT3), or a collagen and fibronectin matrix with BDNF in PLGA MPs (FN + MP.BDNF). Retrograde labeling and functional tests showed that LM + MP.NGF/NT3 increased the number of regenerated sensory neurons and improved sensory functional recovery, whereas FN + MP.BDNF preferentially increased regenerated motoneurons and enhanced motor functional recovery. Therefore, combination of ECM molecules with NTFs may be a good approach to selectively enhance motor and sensory axons regeneration and promote appropriate target reinnervation.
Collapse
|
20
|
Repić T, Madirazza K, Bektur E, Sapunar D. Characterization of dorsal root ganglion neurons cultured on silicon micro-pillar substrates. Sci Rep 2016; 6:39560. [PMID: 28008963 PMCID: PMC5180168 DOI: 10.1038/srep39560] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 11/23/2016] [Indexed: 01/08/2023] Open
Abstract
Our study focuses on characterization of dorsal root ganglion (DRG) neurons cultured on silicon micro-pillar substrates (MPS) with the ultimate goal of designing micro-electrode arrays (MEAs) for successful electrophysiological recordings of DRG neurons. Adult and neonatal DRG neurons were cultured on MPS and glass coverslips for 7 days in vitro. DRG neuronal distribution and morphometric analysis, including neurite alignment and length, was performed on MPS areas with different pillar width and spacing. We showed that MPS provide an environment for growth of adult and neonatal DRG neurons as permissive as control glass surfaces. Neonatal DRG neurons were present on MPS areas with narrow pillar spacing, while adult neurons preferred wider pillar spacing. Compared to the control glass surfaces the neonatal and adult DRG neurons in regions with narrow pillar spacing range developed a smaller number of longer neurites. In the same area, neurites were preferentially oriented along three directional axes at 30°, 90° and 150°. MPS architecture influenced growth directionality of all main DRG neuronal subtypes. We can conclude that specific micro-pillar substrate topography affects the morphology of DRG neurons. This knowledge can enable development of MEAs with precisely defined physical features for various neuroscience applications.
Collapse
Affiliation(s)
- Tihana Repić
- Laboratory for Pain Research, School of Medicine, University of Split, Croatia
| | - Katarina Madirazza
- Speech and Hearing Research Laboratory, School of Medicine, University of Split, Croatia
| | - Ezgi Bektur
- Histology and Embryology Department, School of Medicine, Eskisehir Osmangazi University, Turkey
| | - Damir Sapunar
- Laboratory for Pain Research, School of Medicine, University of Split, Croatia
| |
Collapse
|
21
|
Qu F, Lorenzo DN, King SJ, Brooks R, Bear JE, Bennett V. Ankyrin-B is a PI3P effector that promotes polarized α5β1-integrin recycling via recruiting RabGAP1L to early endosomes. eLife 2016; 5. [PMID: 27718357 PMCID: PMC5089861 DOI: 10.7554/elife.20417] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Accepted: 10/07/2016] [Indexed: 01/03/2023] Open
Abstract
Endosomal membrane trafficking requires coordination between phosphoinositide lipids, Rab GTPases, and microtubule-based motors to dynamically determine endosome identity and promote long-range organelle transport. Here we report that ankyrin-B (AnkB), through integrating all three systems, functions as a critical node in the protein circuitry underlying polarized recycling of α5β1-integrin in mouse embryonic fibroblasts, which enables persistent fibroblast migration along fibronectin gradients. AnkB associates with phosphatidylinositol 3-phosphate (PI3P)-positive organelles in fibroblasts and binds dynactin to promote their long-range motility. We demonstrate that AnkB binds to Rab GTPase Activating Protein 1-Like (RabGAP1L) and recruits it to PI3P-positive organelles, where RabGAP1L inactivates Rab22A, and promotes polarized trafficking to the leading edge of migrating fibroblasts. We further determine that α5β1-integrin depends on an AnkB/RabGAP1L complex for polarized recycling. Our results reveal AnkB as an unexpected key element in coordinating polarized transport of α5β1-integrin and likely of other specialized endocytic cargos.
Collapse
Affiliation(s)
- Fangfei Qu
- Department of Biochemistry, Duke University Medical Center, Durham, United States.,Department of Cell Biology, Duke University Medical Center, Durham, United States.,Department of Neurobiology, Duke University Medical Center, Durham, United States.,Howard Hughes Medical Institute, Duke University Medical Center, Durham, United States
| | - Damaris N Lorenzo
- Department of Biochemistry, Duke University Medical Center, Durham, United States.,Department of Cell Biology, Duke University Medical Center, Durham, United States.,Department of Neurobiology, Duke University Medical Center, Durham, United States.,Howard Hughes Medical Institute, Duke University Medical Center, Durham, United States
| | - Samantha J King
- UNC Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Durham, United States.,Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Rebecca Brooks
- UNC Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Durham, United States.,Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - James E Bear
- UNC Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Durham, United States.,Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Vann Bennett
- Department of Biochemistry, Duke University Medical Center, Durham, United States.,Department of Cell Biology, Duke University Medical Center, Durham, United States.,Department of Neurobiology, Duke University Medical Center, Durham, United States.,Howard Hughes Medical Institute, Duke University Medical Center, Durham, United States
| |
Collapse
|
22
|
Cheah M, Andrews MR. Targeting cell surface receptors for axon regeneration in the central nervous system. Neural Regen Res 2016; 11:1884-1887. [PMID: 28197173 PMCID: PMC5270415 DOI: 10.4103/1673-5374.197079] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Axon regeneration in the CNS is largely unsuccessful due to excess inhibitory extrinsic factors within lesion sites together with an intrinsic inability of neurons to regrow following injury. Recent work demonstrates that forced expression of certain neuronal transmembrane receptors can recapitulate neuronal growth resulting in successful growth within and through inhibitory lesion environments. More specifically, neuronal expression of integrin receptors such as alpha9beta1 integrin which binds the extracellular matrix glycoprotein tenascin-C, trk receptors such as trkB which binds the neurotrophic factor BDNF, and receptor PTPσ which binds chondroitin sulphate proteoglycans, have all been show to significantly enhance regeneration of injured axons. We discuss how reintroduction of these receptors in damaged neurons facilitates signalling from the internal environment of the cell with the external environment of the lesion milieu, effectively resulting in growth and repair following injury. In summary, we suggest an appropriate balance of intrinsic and extrinsic factors are required to obtain substantial axon regeneration.
Collapse
Affiliation(s)
- Menghon Cheah
- John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, United Kingdom
| | - Melissa R Andrews
- School of Medicine, University of St. Andrews, St. Andrews, United Kingdom
| |
Collapse
|
23
|
Gonzalez-Perez F, Alé A, Santos D, Barwig C, Freier T, Navarro X, Udina E. Substratum preferences of motor and sensory neurons in postnatal and adult rats. Eur J Neurosci 2015; 43:431-42. [PMID: 26332537 DOI: 10.1111/ejn.13057] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 08/25/2015] [Accepted: 08/25/2015] [Indexed: 12/18/2022]
Abstract
After peripheral nerve injuries, damaged axons can regenerate but functional recovery is limited by the specific reinnervation of targets. In this study we evaluated if motor and sensory neurites have a substrate preference for laminin and fibronectin in postnatal and adult stages. In postnatal dorsal root ganglia (DRG) explants, sensory neurons extended longer neurites on collagen matrices enriched with laminin (~50%) or fibronectin (~35%), whereas motoneurons extended longer neurites (~100%) in organotypic spinal cord slices embedded in fibronectin-enriched matrix. An increased percentage of parvalbumin-positive neurites (presumptive proprioceptive) vs. neurofilament-positive neurites was also found in DRG in fibronectin-enriched matrix. To test if the different preference of neurons for extracellular matrix components was maintained in vivo, these matrices were used to fill a chitosan guide to repair a 6-mm gap in the sciatic nerve of adult rats. However, the number of regenerating motor and sensory neurons after 1 month was similar between groups. Moreover, none of the retrotraced sensory neurons in DRG was positive for parvalbumin, suggesting that presumptive proprioceptive neurons had poor regenerative capabilities compared with other peripheral neurons. Using real-time PCR we evaluated the expression of α5β1 (receptor for fibronectin) and α7β1 integrin (receptor for laminin) in spinal cord and DRG 2 days after injury. Postnatal animals showed a higher increase of α5β1 integrin, whereas both integrins were similarly expressed in adult neurons. Therefore, we conclude that motor and sensory axons have a different substrate preference at early postnatal stages but this difference is lost in the adult.
Collapse
Affiliation(s)
- Francisco Gonzalez-Perez
- Institute of Neurosciences, Department of Cell Biology, Physiology, Immunology, Universitat Autònoma de Barcelona, CIBERNED, E-08193, Bellaterra, Spain
| | - Albert Alé
- Institute of Neurosciences, Department of Cell Biology, Physiology, Immunology, Universitat Autònoma de Barcelona, CIBERNED, E-08193, Bellaterra, Spain
| | - Daniel Santos
- Institute of Neurosciences, Department of Cell Biology, Physiology, Immunology, Universitat Autònoma de Barcelona, CIBERNED, E-08193, Bellaterra, Spain
| | | | | | - Xavier Navarro
- Institute of Neurosciences, Department of Cell Biology, Physiology, Immunology, Universitat Autònoma de Barcelona, CIBERNED, E-08193, Bellaterra, Spain
| | - Esther Udina
- Institute of Neurosciences, Department of Cell Biology, Physiology, Immunology, Universitat Autònoma de Barcelona, CIBERNED, E-08193, Bellaterra, Spain
| |
Collapse
|
24
|
Kim H, W Caspar T, Shah SB, Hsieh AH. Effects of proinflammatory cytokines on axonal outgrowth from adult rat lumbar dorsal root ganglia using a novel three-dimensional culture system. Spine J 2015; 15:1823-31. [PMID: 25797812 DOI: 10.1016/j.spinee.2015.03.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 02/11/2015] [Accepted: 03/16/2015] [Indexed: 02/03/2023]
Abstract
BACKGROUND CONTEXT Degeneration of the intervertebral disc is often associated with low back pain and increased infiltration of nerve fibers originating from dorsal root ganglia (DRG). The degenerated disc is also characterized by the presence of proinflammatory cytokines, which may influence axonal outgrowth. Toward an improved understanding of the growth of DRG neurons into compliant extracellular matrices, we developed a novel experimental system to measure axonal outgrowth of adult rat lumbar DRG neurons within three-dimensional (3D) collagen hydrogels and used this system to examine the effects of interleukin 1β (IL-1β) and tumor necrosis factor (TNF)-α treatment. PURPOSE The aim was to investigate the effects of proinflammatory cytokines on 3D neuronal growth into collagen matrices. STUDY DESIGN This was an in vitro study of neurite outgrowth from adult rat lumbar DRG into collagen gels in response to IL-1β and TNF-α. METHODS Lumbar DRG were obtained from adult Sprague Dawley rats, bisected to expose cell bodies and placed onto collagen gel constructs prepared in 24-well Transwell inserts. Dorsal root ganglia were then treated with nerve growth factor (NGF)-free Neurobasal media (negative control) or NGF-supplemented media containing 0, 1, and 10 ng/mL of IL-1β and TNF-α. After 7 days, collagen gel-DRG constructs were immunostained for phosphorylated neurofilament, an axonal marker. Simple Neurite Tracer (Fiji/ImageJ) was used to quantify 3D axonal outgrowth from confocal image stacks. Data were analyzed using one-way analysis of variance, with Tukey HSD post hoc correction at a level of p<.05. RESULTS Immunostaining showed robust axonal outgrowth into collagen gels from all NGF-treated DRG. The negative control demonstrated very few and short neurites. Tumor necrosis factor-α (1 and 10 ng/mL) significantly inhibited axonal outgrowth compared with NGF-only media (p<.026 and p<.02, respectively). After IL-1β treatment, average axon length was 10% lower at 1 ng/mL and 7.5% higher at 10 ng/mL, but these differences were not statistically significant. Among cytokine treatments, however, average axon length in the IL-1β (10 ng/mL) group was significantly higher than that in the other groups (p<.05). CONCLUSIONS A novel 3D collagen gel culture system was used to investigate factors modulating neuronal ingrowth. Our results showed that NGF was necessary to promote neurite growth into collagen gels. In the presence of proinflammatory cytokines, high concentrations of IL-1β induced significantly higher axonal outgrowth than TNF-α and low levels of IL-1β.
Collapse
Affiliation(s)
- Hyunchul Kim
- Fischell Department of Bioengineering, University of Maryland, College Park, Jeong H. Kim Engineering Building, College Park, MD 20742, USA
| | - Tyler W Caspar
- Fischell Department of Bioengineering, University of Maryland, College Park, Jeong H. Kim Engineering Building, College Park, MD 20742, USA
| | - Sameer B Shah
- Department of Orthopaedic Surgery, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Bioengineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Adam H Hsieh
- Fischell Department of Bioengineering, University of Maryland, College Park, Jeong H. Kim Engineering Building, College Park, MD 20742, USA; Department of Orthopaedics, University of Maryland, Baltimore, 22 S. Greene Street, Baltimore, MD 21201, USA.
| |
Collapse
|
25
|
Functional integration of human neural precursor cells in mouse cortex. PLoS One 2015; 10:e0120281. [PMID: 25763840 PMCID: PMC4357458 DOI: 10.1371/journal.pone.0120281] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 01/31/2015] [Indexed: 12/14/2022] Open
Abstract
This study investigates the electrophysiological properties and functional integration of different phenotypes of transplanted human neural precursor cells (hNPCs) in immunodeficient NSG mice. Postnatal day 2 mice received unilateral injections of 100,000 GFP+ hNPCs into the right parietal cortex. Eight weeks after transplantation, 1.21% of transplanted hNPCs survived. In these hNPCs, parvalbumin (PV)-, calretinin (CR)-, somatostatin (SS)-positive inhibitory interneurons and excitatory pyramidal neurons were confirmed electrophysiologically and histologically. All GFP+ hNPCs were immunoreactive with anti-human specific nuclear protein. The proportions of PV-, CR-, and SS-positive cells among GFP+ cells were 35.5%, 15.7%, and 17.1%, respectively; around 15% of GFP+ cells were identified as pyramidal neurons. Those electrophysiologically and histological identified GFP+ hNPCs were shown to fire action potentials with the appropriate firing patterns for different classes of neurons and to display spontaneous excitatory and inhibitory postsynaptic currents (sEPSCs and sIPSCs). The amplitude, frequency and kinetic properties of sEPSCs and sIPSCs in different types of hNPCs were comparable to host cells of the same type. In conclusion, GFP+ hNPCs produce neurons that are competent to integrate functionally into host neocortical neuronal networks. This provides promising data on the potential for hNPCs to serve as therapeutic agents in neurological diseases with abnormal neuronal circuitry such as epilepsy.
Collapse
|
26
|
Raposo C, Schwartz M. Glial scar and immune cell involvement in tissue remodeling and repair following acute CNS injuries. Glia 2014; 62:1895-904. [PMID: 24756949 DOI: 10.1002/glia.22676] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 03/30/2014] [Accepted: 04/01/2014] [Indexed: 12/17/2022]
Abstract
Inadequate axonal regeneration is a common phenomenon occurring following acute injury to the central nervous system (CNS), and is often associated with permanent neurological deficits. The injured axons attempting to regenerate face the inhospitable environment of the CNS scar, which can hinder axonal growth and sprouting. In addition, in response to the insult, intense activation and infiltration of immune cells take place. Both the scar tissue and immune response, which have received a bad reputation in the context of CNS repair are essential for the overall recovery from CNS injuries, but are not optimally controlled. The glial scar contributes to protection of the spared neural tissues by establishing a boundary between damaged and salvageable tissue, and by educating the immune cells to promote the healing of the CNS tissue. In turn, the immune cells, and in particular the infiltrating macrophages, exert several functions at the lesion site, including resolution of the microglial response, control of scar tissue degradation, and production of growth factors; thereby, promoting neuronal survival, axonal regeneration, and tissue remodeling. As axonal regeneration and tissue remodeling are viewed as critical steps for the overall functional recovery following CNS injury, a detailed understanding of the mechanisms underlying the timely formation and degradation of the CNS scar, and its crosstalk with the inflammatory response, are of great importance, both biologically and clinically.
Collapse
Affiliation(s)
- Catarina Raposo
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | | |
Collapse
|
27
|
Silver DJ, Silver J. Contributions of chondroitin sulfate proteoglycans to neurodevelopment, injury, and cancer. Curr Opin Neurobiol 2014; 27:171-8. [PMID: 24762654 DOI: 10.1016/j.conb.2014.03.016] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 03/25/2014] [Accepted: 03/25/2014] [Indexed: 01/09/2023]
Abstract
Chondroitin sulfate proteoglycans (CSPGs) are a diverse family of extracellular matrix (ECM) molecules that make significant contributions to the patterning and routing of migrating neural cells and extending axons. Three distinct modes of migration mediation result from the relative abundance and positioning of expressed CSPGs, the profile of CSPG receptors expressed by the motile cell types, and the overall way in which the CSPGs integrate into and stabilize the neural ECM. Here we discuss recent findings that help to clarify the molecular mechanisms that underlie these distinct migration-regulating properties as they pertain to neural development, CNS injury, and gliomagenesis.
Collapse
Affiliation(s)
- Daniel J Silver
- Human Biology Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, Mailstop C3-168, Seattle, WA 98109, United States
| | - Jerry Silver
- Department of Neurosciences, Case Western Reserve University, 2109 Adelbert Road Rm E-658, Cleveland, OH 44106, United States.
| |
Collapse
|
28
|
Butt GF, Habib A, Mahgoub K, Sofela A, Tilley M, Guo L, Cordeiro MF. Optic nerve regeneration. EXPERT REVIEW OF OPHTHALMOLOGY 2014. [DOI: 10.1586/eop.12.66] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
29
|
Berezin V, Walmod PS, Filippov M, Dityatev A. Targeting of ECM molecules and their metabolizing enzymes and receptors for the treatment of CNS diseases. PROGRESS IN BRAIN RESEARCH 2014; 214:353-88. [DOI: 10.1016/b978-0-444-63486-3.00015-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
30
|
Proregenerative properties of ECM molecules. BIOMED RESEARCH INTERNATIONAL 2013; 2013:981695. [PMID: 24195084 PMCID: PMC3782155 DOI: 10.1155/2013/981695] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 07/04/2013] [Accepted: 08/07/2013] [Indexed: 12/27/2022]
Abstract
After traumatic injuries to the nervous system, regrowing axons encounter a complex microenvironment where mechanisms that promote regeneration compete with inhibitory processes. Sprouting and axonal regrowth are key components of functional recovery but are often counteracted by inhibitory molecules. This review covers extracellular matrix molecules that support neuron axonal outgrowth.
Collapse
|
31
|
Lemons ML, Abanto ML, Dambrouskas N, Clements CC, Deloughery Z, Garozzo J, Condic ML. Integrins and cAMP mediate netrin-induced growth cone collapse. Brain Res 2013; 1537:46-58. [PMID: 24001590 DOI: 10.1016/j.brainres.2013.08.045] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Revised: 08/20/2013] [Accepted: 08/25/2013] [Indexed: 12/27/2022]
Abstract
Growth cones integrate a remarkably complex concert of chemical cues to guide axons to their appropriate destinations. Recent work suggests that integrins contribute to axon guidance by interacting with a wide range of extracellular molecules including axon guidance molecules, by mechanisms that are not fully understood. Here, we describe an interaction between integrins and netrin-1 in growth cones that contributes to growth cone collapse. Our data show that netrin-1 causes growth cone collapse in a substratum-specific manner and is integrin-dependent. Netrin-1 causes collapse of cultured chick dorsal root ganglion (DRG) growth cones extending on high levels of laminin-1 (LN) but not growth cones extending on low levels of LN or on fibronectin. Blocking integrin function significantly decreases netrin-induced growth cone collapse on high LN. Netrin-1 and integrins interact on growth cones; netrin-1 causes integrin activation, a conformational shift to a high ligand-affinity state. Netrin-1 directly binds to integrin α3 and α6 peptides, further suggesting a netrin-integrin interaction. Interestingly, our data reveal that netrin-1 increases growth cone levels of cAMP in a substratum-specific manner and that netrin-induced growth cone collapse requires increased cAMP in combination with integrin activation. Manipulations that either decrease cAMP levels or integrin activation block netrin-induced collapse. These results imply a common mechanism for growth cone collapse and novel interactions between integrins, netrin-1 and cAMP that contribute to growth cone guidance.
Collapse
Affiliation(s)
- M L Lemons
- Department of Natural Sciences, Assumption College, Worcester MA 01609, United States; Department of Neurobiology and Anatomy, University of Utah School of Medicine, 401 MREB, 20 North 1900 East, Salt Lake City, UT 84132, United States.
| | | | | | | | | | | | | |
Collapse
|
32
|
Abdullah M, O'Daly A, Vyas A, Rohde C, Brushart TM. Adult motor axons preferentially reinnervate predegenerated muscle nerve. Exp Neurol 2013; 249:1-7. [PMID: 23933577 DOI: 10.1016/j.expneurol.2013.07.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 07/22/2013] [Accepted: 07/24/2013] [Indexed: 12/17/2022]
Abstract
Preferential motor reinnervation (PMR) is the tendency for motor axons regenerating after repair of mixed nerve to reinnervate muscle nerve and/or muscle rather than cutaneous nerve or skin. PMR may occur in response to the peripheral nerve pathway alone in juvenile rats (Brushart, 1993; Redett et al., 2005), yet the ability to identify and respond to specific pathway markers is reportedly lost in adults (Uschold et al., 2007). The experiments reported here evaluate the relative roles of pathway and end organ in the genesis of PMR in adult rats. Fresh and 2-week predegenerated femoral nerve grafts were transferred in correct or reversed alignment to replace the femoral nerves of previously unoperated Lewis rats. After 8 weeks of regeneration the motoneurons projecting through the grafts to recipient femoral cutaneous and muscle branches and their adjacent end organs were identified by retrograde labeling. Motoneuron counts were subjected to Poisson regression analysis to determine the relative roles of pathway and end organ identity in generating PMR. Transfer of fresh grafts did not result in PMR, whereas substantial PMR was observed when predegenerated grafts were used. Similarly, the pathway through which motoneurons reached the muscle had a significant impact on PMR when grafts were predegenerated, but not when they were fresh. Comparison of the relative roles of pathway and end organ in generating PMR revealed that neither could be shown to be more important than the other. These experiments demonstrate unequivocally that adult muscle nerve and cutaneous nerve differ in qualities that can be detected by regenerating adult motoneurons and that can modify their subsequent behavior. They also reveal that two weeks of Wallerian degeneration modify the environment in the graft from one that provides no modality-specific cues for motor neurons to one that actively promotes PMR.
Collapse
Affiliation(s)
- M Abdullah
- Department of Orthopaedic Surgery, Johns Hopkins University, 601 N. Caroline Street, Baltimore, MD 21287, USA
| | | | | | | | | |
Collapse
|
33
|
Paxillin phosphorylation counteracts proteoglycan-mediated inhibition of axon regeneration. Exp Neurol 2013; 248:157-69. [PMID: 23797153 DOI: 10.1016/j.expneurol.2013.06.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 05/29/2013] [Accepted: 06/14/2013] [Indexed: 11/21/2022]
Abstract
In the adult central nervous system, the tips of axons severed by injury are commonly transformed into dystrophic endballs and cease migration upon encountering a rising concentration gradient of inhibitory proteoglycans. However, intracellular signaling networks mediating endball migration failure remain largely unknown. Here we show that manipulation of protein kinase A (PKA) or its downstream adhesion component paxillin can reactivate the locomotive machinery of endballs in vitro and facilitate axon growth after injury in vivo. In dissociated cultures of adult rat dorsal root ganglion neurons, PKA is activated in endballs formed on gradients of the inhibitory proteoglycan aggrecan, and pharmacological inhibition of PKA promotes axon growth on aggrecan gradients most likely through phosphorylation of paxillin at serine 301. Remarkably, pre-formed endballs on aggrecan gradients resume forward migration in response to PKA inhibition. This resumption of endball migration is associated with increased turnover of adhesive point contacts dependent upon paxillin phosphorylation. Furthermore, expression of phosphomimetic paxillin overcomes aggrecan-mediated growth arrest of endballs, and facilitates axon growth after optic nerve crush in vivo. These results point to the importance of adhesion dynamics in restoring endball migration and suggest a potential therapeutic target for axon tract repair.
Collapse
|
34
|
Ribeiro A, Balasubramanian S, Hughes D, Vargo S, Powell EM, Leach JB. β1-Integrin cytoskeletal signaling regulates sensory neuron response to matrix dimensionality. Neuroscience 2013; 248:67-78. [PMID: 23764511 DOI: 10.1016/j.neuroscience.2013.05.057] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 05/30/2013] [Indexed: 10/26/2022]
Abstract
Neuronal differentiation, pathfinding and morphology are directed by biochemical cues that in vivo are presented in a complex scaffold of extracellular matrix. This microenvironment is three-dimensional (3D) and heterogeneous. Therefore, it is not surprising that more physiologically-relevant cellular responses are found in 3D culture environments rather than on two-dimensional (2D) flat substrates. One key difference between 2D and 3D environments is the spatial arrangement of cell-matrix interactions. Integrins and other receptor proteins link the various molecules presented in the extracellular environment to intracellular signaling cascades and thus influence a number of neuronal responses including the availability and activation of integrins themselves. We have previously reported that a 3D substrate induces an important morphological transformation of embryonic mouse dorsal root ganglion (DRG) neurons. Here, we investigate the hypothesis that β1-integrin signaling via focal adhesion kinase (FAK) and the RhoGTPases Rac and Rho influences neuronal morphology in 2D vs 3D environments. We report that β1-integrin activity and FAK phosphorylation at tyrosine 397 (FAKpY397) are linked to neuronal polarization as well as neurite outgrowth and branching. Rac and Rho expression are decreased in 3D vs 2D culture but not correlated with β1-integrin function. These results suggest that proper β1-integrin activity is required for the elaboration of physiologic DRG morphology and that 3D culture provides a more appropriate milieu to the mimic in vivo scenario. We propose that neuronal morphology may be directed during development and regeneration by factors that influence how β1-integrin, FAK and RhoGTPase molecules integrate substrate signals in the 3D microenvironment.
Collapse
Affiliation(s)
- A Ribeiro
- Department of Chemical, Biochemical & Environmental Engineering, UMBC, Eng 314, 1000 Hilltop Circle, Baltimore, MD 21250, USA
| | - S Balasubramanian
- Department of Chemical, Biochemical & Environmental Engineering, UMBC, Eng 314, 1000 Hilltop Circle, Baltimore, MD 21250, USA
| | - D Hughes
- Department of Chemical, Biochemical & Environmental Engineering, UMBC, Eng 314, 1000 Hilltop Circle, Baltimore, MD 21250, USA
| | - S Vargo
- Department of Chemical, Biochemical & Environmental Engineering, UMBC, Eng 314, 1000 Hilltop Circle, Baltimore, MD 21250, USA
| | - E M Powell
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, HSF II S251, 20 Penn Street, Baltimore, MD 21201, USA; Department of Psychiatry, University of Maryland School of Medicine, HSF II S251, 20 Penn Street, Baltimore, MD 21201, USA; Department of Bioengineering, University of Maryland School of Medicine, HSF II S251, 20 Penn Street, Baltimore, MD 21201, USA
| | - J B Leach
- Department of Chemical, Biochemical & Environmental Engineering, UMBC, Eng 314, 1000 Hilltop Circle, Baltimore, MD 21250, USA.
| |
Collapse
|
35
|
Fudge NJ, Mearow KM. Extracellular matrix-associated gene expression in adult sensory neuron populations cultured on a laminin substrate. BMC Neurosci 2013; 14:15. [PMID: 23360524 PMCID: PMC3610289 DOI: 10.1186/1471-2202-14-15] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Accepted: 01/08/2013] [Indexed: 12/03/2022] Open
Abstract
Background In our previous investigations of the role of the extracellular matrix (ECM) in promoting neurite growth we have observed that a permissive laminin (LN) substrate stimulates differential growth responses in subpopulations of mature dorsal root ganglion (DRG) neurons. DRG neurons expressing Trk and p75 receptors grow neurites on a LN substrate in the absence of neurotrophins, while isolectin B4-binding neurons (IB4+) do not display significant growth under the same conditions. We set out to determine whether there was an expression signature of the LN-induced neurite growth phenotype. Using a lectin binding protocol IB4+ neurons were isolated from dissociated DRG neurons, creating two groups - IB4+ and IB4-. A small-scale microarray approach was employed to screen the expression of a panel of ECM-associated genes following dissociation (t=0) and after 24 hr culture on LN (t=24LN). This was followed by qRT-PCR and immunocytochemistry of selected genes. Results The microarray screen showed that 36 of the 144 genes on the arrays were consistently expressed by the neurons. The array analyses showed that six genes had lower expression in the IB4+ neurons compared to the IB4- cells at t=0 (CTSH, Icam1, Itgβ1, Lamb1, Plat, Spp1), and one gene was expressed at higher levels in the IB4+ cells (Plaur). qRT-PCR was carried out as an independent assessment of the array results. There were discrepancies between the two methods, with qRT-PCR confirming the differences in Lamb1, Plat and Plaur, and showing decreased expression of AdamTs1, FN, and Icam in the IB4+ cells at t=0. After 24 hr culture on LN, there were no significant differences detected by qRT-PCR between the IB4+ and IB4- cells. However, both groups showed upregulation of Itgβ1 and Plaur after 24 hr on LN, the IB4+ group also had increased Plat, and the IB4- cells showed decreased Lamb1, Icam1 and AdamTs1. Further, the array screen also detected a number of genes (not subjected to qRT-PCR) expressed similarly by both populations in relatively high levels but not detectably influenced by time in culture (Bsg, Cst3, Ctsb, Ctsd, Ctsl, Mmp14, Mmp19, Sparc. We carried out immunohistochemistry to confirm expression of proteins encoded by a number of these genes. Conclusions Our results show that 1B4+ and IB4- neurons differ in the expression of several genes that are associated with responsiveness to the ECM prior to culturing (AdamTs1, FN, Icam1, Lamb1, Plat, Plaur). The data suggest that the genes expressed at higher levels in the IB4- neurons could contribute to the initial growth response of these cells in a permissive environment and could also represent a common injury response that subsequently promotes axon regeneration. The differential expression of several extracellular matrix molecules (FN, Lamb1, Icam) may suggest that the IB4- neurons are capable of maintaining /secreting their local extracellular environment which could aid in the regenerative process. Overall, these data provide new information on potential targets that could be manipulated to enhance axonal regeneration in the mature nervous system.
Collapse
Affiliation(s)
- Neva J Fudge
- Division of BioMedical Sciences, Memorial University of Newfoundland, St, John's, NL, Canada
| | | |
Collapse
|
36
|
Abstract
After central nervous system (CNS) insults, such as spinal cord injury or traumatic brain injury, neurons encounter a complex microenvironment where mechanisms that promote regeneration compete with inhibitory processes. Sprouting and axonal re-growth are key components of functional recovery, but are often counteracted by inhibitory molecules. Several strategies are being pursued whereby these inhibitory molecules are either being neutralized with blocking antibodies, with enzymatic degradation or downstream signaling events are being interfered with. Two recent studies ( 1) (,) ( 2) show that activating integrin signaling in dorsal root ganglion (DRG) neurons renders them able to overcome inhibitory signals, and could possibly lead to new strategies to improve neuronal regeneration.
Collapse
Affiliation(s)
- Stefan Plantman
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
37
|
Gensel J, Kigerl K, Mandrekar-Colucci S, Gaudet A, Popovich P. Achieving CNS axon regeneration by manipulating convergent neuro-immune signaling. Cell Tissue Res 2012; 349:201-13. [PMID: 22592625 PMCID: PMC10881271 DOI: 10.1007/s00441-012-1425-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Accepted: 04/02/2012] [Indexed: 12/20/2022]
Abstract
After central nervous system (CNS) trauma, axons have a low capacity for regeneration. Regeneration failure is associated with a muted regenerative response of the neuron itself, combined with a growth-inhibitory and cytotoxic post-injury environment. After spinal cord injury (SCI), resident and infiltrating immune cells (especially microglia/macrophages) contribute significantly to the growth-refractory milieu near the lesion. By targeting both the regenerative potential of the axon and the cytotoxic phenotype of microglia/macrophages, we may be able to improve CNS repair after SCI. In this review, we discuss molecules shown to impact CNS repair by affecting both immune cells and neurons. Specifically, we provide examples of pattern recognition receptors, integrins, cytokines/chemokines, nuclear receptors and galectins that could improve CNS repair. In many cases, signaling by these molecules is complex and may have contradictory effects on recovery depending on the cell types involved or the model studied. Despite this caveat, deciphering convergent signaling pathways on immune cells (which affect axon growth indirectly) and neurons (direct effects on axon growth) could improve repair and recovery after SCI. Future studies must continue to consider how regenerative therapies targeting neurons impact other cells in the pathological CNS. By identifying molecules that simultaneously improve axon regenerative capacity and drive the protective, growth-promoting phenotype of immune cells, we may discover SCI therapies that act synergistically to improve CNS repair and functional recovery.
Collapse
Affiliation(s)
- J.C. Gensel
- Center for Brain and Spinal Cord Repair, Department of Neuroscience, The Ohio State University College of Medicine, Columbus, Ohio 43210
| | - K.A. Kigerl
- Center for Brain and Spinal Cord Repair, Department of Neuroscience, The Ohio State University College of Medicine, Columbus, Ohio 43210
| | - S. Mandrekar-Colucci
- Center for Brain and Spinal Cord Repair, Department of Neuroscience, The Ohio State University College of Medicine, Columbus, Ohio 43210
| | - A.D. Gaudet
- Center for Brain and Spinal Cord Repair, Department of Neuroscience, The Ohio State University College of Medicine, Columbus, Ohio 43210
| | - P.G. Popovich
- Center for Brain and Spinal Cord Repair, Department of Neuroscience, The Ohio State University College of Medicine, Columbus, Ohio 43210
| |
Collapse
|
38
|
Abstract
While ultimately, focus must be placed on experimentation using adult systems, vastly important clues to regeneration can be found in the study of the embryonic nervous system. In embryonic systems, axonal regeneration is successful before a critical period, and numerous advances have resulted from the study of isolated cells and tissues in vitro. Studies over many decades from the laboratory of Paul C. Letourneau have probed the cellular and molecular phenomena involved in axon outgrowth and guidance in the embryonic central and peripheral nervous system and have laid the framework for many current advances in regeneration research. Letourneau’s pioneering work related to growth cone behavior, guidance, and regeneration has resulted in considerable contributions toward our understanding not only of cellular mechanisms that underlie axon growth, but also of the specific areas of study that require attention to accomplish future breakthroughs. The present article summarizes some of the major contributions from Paul Letourneau and his team in the area of axonal regeneration.
Collapse
Affiliation(s)
- Diane M Snow
- Department of Anatomy and Neurobiology, Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky 40536, USA.
| |
Collapse
|
39
|
Specificity of peripheral nerve regeneration: interactions at the axon level. Prog Neurobiol 2012; 98:16-37. [PMID: 22609046 DOI: 10.1016/j.pneurobio.2012.05.005] [Citation(s) in RCA: 289] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Revised: 04/12/2012] [Accepted: 05/08/2012] [Indexed: 12/13/2022]
Abstract
Peripheral nerves injuries result in paralysis, anesthesia and lack of autonomic control of the affected body areas. After injury, axons distal to the lesion are disconnected from the neuronal body and degenerate, leading to denervation of the peripheral organs. Wallerian degeneration creates a microenvironment distal to the injury site that supports axonal regrowth, while the neuron body changes in phenotype to promote axonal regeneration. The significance of axonal regeneration is to replace the degenerated distal nerve segment, and achieve reinnervation of target organs and restitution of their functions. However, axonal regeneration does not always allows for adequate functional recovery, so that after a peripheral nerve injury, patients do not recover normal motor control and fine sensibility. The lack of specificity of nerve regeneration, in terms of motor and sensory axons regrowth, pathfinding and target reinnervation, is one the main shortcomings for recovery. Key factors for successful axonal regeneration include the intrinsic changes that neurons suffer to switch their transmitter state to a pro-regenerative state and the environment that the axons find distal to the lesion site. The molecular mechanisms implicated in axonal regeneration and pathfinding after injury are complex, and take into account the cross-talk between axons and glial cells, neurotrophic factors, extracellular matrix molecules and their receptors. The aim of this review is to look at those interactions, trying to understand if some of these molecular factors are specific for motor and sensory neuron growth, and provide the basic knowledge for potential strategies to enhance and guide axonal regeneration and reinnervation of adequate target organs.
Collapse
|
40
|
Extrinsic cellular and molecular mediators of peripheral axonal regeneration. Cell Tissue Res 2012; 349:5-14. [PMID: 22476657 DOI: 10.1007/s00441-012-1389-5] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Accepted: 02/23/2012] [Indexed: 12/11/2022]
Abstract
The ability of injured peripheral nerves to regenerate and reinnervate their original targets is a characteristic feature of the peripheral nervous system (PNS). On the other hand, neurons of the central nervous system (CNS), including retinal ganglion cell (RGC) axons, are incapable of spontaneous regeneration. In the adult PNS, axonal regeneration after injury depends on well-orchestrated cellular and molecular processes that comprise a highly reproducible series of degenerative reactions distal to the site of injury. During this fine-tuned process, named Wallerian degeneration, a remodeling of the distal nerve fragment prepares a permissive microenvironment that permits successful axonal regrowth originating from the proximal nerve fragment. Therefore, a multitude of adjusted intrinsic and extrinsic factors are important for surviving neurons, Schwann cells, macrophages and fibroblasts as well as endothelial cells in order to achieve successful regeneration. The aim of this review is to summarize relevant extrinsic cellular and molecular determinants of successful axonal regeneration in rodents that contribute to the regenerative microenvironment of the PNS.
Collapse
|
41
|
Hodgkinson GN, Tresco PA, Hlady V. The role of well-defined patterned substrata on the regeneration of DRG neuron pathfinding and integrin expression dynamics using chondroitin sulfate proteoglycans. Biomaterials 2012; 33:4288-97. [PMID: 22436802 DOI: 10.1016/j.biomaterials.2012.02.046] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Accepted: 02/25/2012] [Indexed: 12/13/2022]
Abstract
Injured neurons intrinsically adapt to and partially overcome inhibitory proteoglycan expression in the central nervous system by upregulating integrin expression. It remains unclear however, to what extent varying proteoglycan concentrations influence the strength of this response, how rapidly neurons adapt to proteoglycans, and how pathfinding dynamics are altered over time as integrin expression is modulated in response to proteoglycan signals. To investigate these quandaries, we created well-defined substrata in which postnatal DRG neuron pathfinding dynamics and growth cone integrin expression were interrogated as a function of proteoglycan substrata density. DRGs responded by upregulating integrin expression in a proteoglycan dose dependent fashion and exhibited robust outgrowth over all proteoglycan densities at initial time frames. However, after prolonged proteoglycan exposure, neurons exhibited decreasing velocities associated with increasing proteoglycan densities, while neurons growing on low proteoglycan levels exhibited robust outgrowth at all time points. Additionally, DRG outgrowth over proteoglycan density step boundaries, and a brief β1 integrin functional block proved that regeneration was integrin dependent and that DRGs exhibit delayed slowing and loss in persistence after even transient encounters with dense proteoglycan boundaries. These findings demonstrate the complexity of proteoglycan regulation on integrin expression and regenerative pathfinding.
Collapse
Affiliation(s)
- Gerald N Hodgkinson
- Department of Bioengineering, University of Utah, Salt Lake City, UT 84112, USA
| | | | | |
Collapse
|
42
|
Tonge DA, de Burgh HT, Docherty R, Humphries MJ, Craig SE, Pizzey J. Fibronectin supports neurite outgrowth and axonal regeneration of adult brain neurons in vitro. Brain Res 2012; 1453:8-16. [PMID: 22483961 PMCID: PMC3989037 DOI: 10.1016/j.brainres.2012.03.024] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Revised: 03/03/2012] [Accepted: 03/09/2012] [Indexed: 12/09/2022]
Abstract
The molecular basis of axonal regeneration of central nervous system (CNS) neurons remains to be fully elucidated. In part, this is due to the difficulty in maintaining CNS neurons in vitro. Here, we show that dissociated neurons from the cerebral cortex and hippocampus of adult mice may be maintained in culture for up to 9 days in defined medium without added growth factors. Outgrowth of neurites including axons was observed from both CNS sources and was significantly greater on plasma fibronectin than on other substrata such as laminin and merosin. Neurite outgrowth on fibronectin appears to be mediated by α5β1 integrin since a recombinant fibronectin fragment containing binding sites for this receptor was as effective as intact fibronectin in supporting neurite outgrowth. Conversely, function-blocking antibodies to α5 and β1 integrin sub-units inhibited neurite outgrowth on intact fibronectin. These results suggest that the axonal regeneration seen in in vivo studies using fibronectin-based matrices is due to the molecule itself and not a consequence of secondary events such as cellular infiltration. They also indicate the domains of fibronectin that may be responsible for eliciting this response.
Collapse
Affiliation(s)
- David A Tonge
- Wolfson-Centre for Age-Related Disease, King's College London, Hodgkin Building, Guy's Campus, London SE1 1UL, UK
| | | | | | | | | | | |
Collapse
|
43
|
Liu T, Houle JD, Xu J, Chan BP, Chew SY. Nanofibrous collagen nerve conduits for spinal cord repair. Tissue Eng Part A 2012; 18:1057-66. [PMID: 22220714 DOI: 10.1089/ten.tea.2011.0430] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Nerve regeneration in an injured spinal cord is often restricted, contributing to the devastating outcome of neurologic impairment below the site of injury. Although implantation of tissue-engineered scaffolds has evolved as a potential treatment method, the outcomes remain sub-optimal. One possible reason may be the lack of topographical signals from these constructs to provide contact guidance to invading cells or regrowing axons. Nanofibers mimic the natural extracellular matrix architecturally and may therefore promote physiologically relevant cellular phenotypes. In this study, the potential application of electrospun collagen nanofibers (diameter=208.2±90.4 nm) for spinal cord injury (SCI) treatment was evaluated in vitro and in vivo. Primary rat astrocytes and dorsal root ganglias (DRGs) were seeded on collagen-coated glass cover slips (two-dimensional [2D] substrate controls), and randomly oriented or aligned collagen fibers to evaluate scaffold topographical effects on astrocyte behavior and neurite outgrowth, respectively. When cultured on collagen nanofibers, astrocyte proliferation and expression of glial fibrillary acidic protein (GFAP) were suppressed as compared to cells on 2D controls at days 3 (p<0.05) and 7 (p<0.01). Aligned fibers resulted in elongated astrocytes (elongation factor >4, p<0.01) and directed the orientation of neurite outgrowth from DRGs along fiber axes. In the contrast, neurites emanated radially on randomly oriented collagen fibers. By forming collagen scaffolds into spiral tubular structures, we demonstrated the feasibility of using electrospun nanofibers for the treatment of acute SCI using a rat hemi-section model. At days 10 and 30 postimplantation, extensive cellular penetration into the constructs was observed regardless of fiber orientation. However, scaffolds with aligned fibers appeared more structurally intact at day 30. ED1 immunofluorescent staining revealed macrophage invasion by day 10, which decreased significantly by day 30. Neural fiber sprouting as evaluated by neurofilament staining was observed as early as day 10. In addition, GFAP immunostained astrocytes were found only at the boundary of the lesion site, and no astrocyte accumulation was observed in the implantation area at any time point. These findings indicate the feasibility of fabricating 3D spiral constructs using electrospun collagen fibers and demonstrated the potential of these scaffolds for SCI repair.
Collapse
Affiliation(s)
- Ting Liu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| | | | | | | | | |
Collapse
|
44
|
McCormick AM, Leipzig ND. Neural regenerative strategies incorporating biomolecular axon guidance signals. Ann Biomed Eng 2012; 40:578-97. [PMID: 22218702 DOI: 10.1007/s10439-011-0505-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Accepted: 12/28/2011] [Indexed: 01/19/2023]
Abstract
There are currently no acceptable cures for central nervous system injuries, and damage induced large gaps in the peripheral nervous system have been challenging to bridge to restore neural functionality. Innervation by neurons is made possible by the growth cone. This dynamic structure is unique to neurons, and can directly sense physical and chemical activity in its environment, utilizing these cues to propel axons to precisely reach their targets. Guidance can occur through chemoattractive factors such as neurotrophins and netrins, chemorepulsive agents like semaphorins and slits, or contact-mediated molecules such as ephrins and those located in the extracellular matrix. The understanding of biomolecular activity during nervous system development and injury has generated new techniques and tactics for improving and restoring function to the nervous system after injury. This review will focus on the major neuronal guidance molecules and their utility in current tissue engineering and neural regenerative strategies.
Collapse
Affiliation(s)
- Aleesha M McCormick
- Department of Chemical and Biomolecular Engineering, The University of Akron, Akron, OH 44325-3906, USA
| | | |
Collapse
|
45
|
Chew DJ, Fawcett JW, Andrews MR. The challenges of long-distance axon regeneration in the injured CNS. PROGRESS IN BRAIN RESEARCH 2012. [PMID: 23186719 DOI: 10.1016/b978-0-444-59544-7.00013-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Injury to the central nervous system (CNS) that results in long-tract axonal damage typically leads to permanent functional deficits in areas innervated at, and below, the level of the lesion. The initial ischemia, inflammation, and neurodegeneration are followed by a progressive generation of scar tissue, dieback of transected axons, and demyelination, creating an area inhibitory to regrowth and recovery. Two ways to combat this inhibition is to therapeutically target the extrinsic and intrinsic properties of the axon-scar environment. Scar tissue within and surrounding the lesion site can be broken down using an enzyme known as chondroitinase. Negative regulators of adult neuronal growth, such as Nogo, can be neutralized with antibodies. Both therapies greatly improve functional recovery in animal models. Alternatively, modifying the intrinsic growth properties of CNS neurons through gene therapy or pharmacotherapy has also shown promising axonal regeneration after injury. Despite these promising therapies, the main challenge of long-distance axon regeneration still remains; achieving a level of functional and organized connectivity below the level of the lesion that mimics the intact CNS.
Collapse
Affiliation(s)
- Daniel J Chew
- Centre for Brain Repair, University of Cambridge, Forvie Site, Robinson Way, Cambridge, UK
| | | | | |
Collapse
|
46
|
Eva R, Andrews MR, Franssen EHP, Fawcett JW. Intrinsic mechanisms regulating axon regeneration: an integrin perspective. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2012; 106:75-104. [PMID: 23211460 DOI: 10.1016/b978-0-12-407178-0.00004-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Adult central nervous system (CNS) axons fail to regenerate after injury because of inhibitory factors in the surrounding environment and a low intrinsic regenerative capacity. Axons in the adult peripheral nervous system have a higher regenerative capacity, due in part to the presence of certain integrins-receptors for the extracellular matrix. Integrins are critical for axon growth during the development of the nervous system but are absent from some adult CNS axons. Here, we discuss the intrinsic mechanisms that regulate axon regeneration and examine the role of integrins. As correct localization is paramount to integrin function, we further discuss the mechanisms that regulate integrin traffic toward the axonal growth cone.
Collapse
Affiliation(s)
- Richard Eva
- Cambridge Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | | | | | | |
Collapse
|
47
|
Wu X, Reddy DS. Integrins as receptor targets for neurological disorders. Pharmacol Ther 2011; 134:68-81. [PMID: 22233753 DOI: 10.1016/j.pharmthera.2011.12.008] [Citation(s) in RCA: 117] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Accepted: 12/15/2011] [Indexed: 12/18/2022]
Abstract
This review focuses on the neurobiology of integrins, pathophysiological roles of integrins in neuroplasticity and nervous system disorders, and therapeutic implications of integrins as potential drug targets and possible delivery pathways. Neuroplasticity is a central phenomenon in many neurological conditions such as seizures, trauma, and traumatic brain injury. During the course of many brain diseases, in addition to intracellular compartment changes, alterations in non-cell compartments such as extracellular matrix (ECM) are recognized as an essential process in forming and reorganizing neural connections. Integrins are heterodimeric transmembrane receptors that mediate cell-ECM and cell-cell adhesion events. Although the mechanisms of neuroplasticity remain unclear, it has been suggested that integrins undergo plasticity including clustering through interactions with ECM proteins, modulating ion channels, intracellular Ca(2+) and protein kinase signaling, and reorganization of cytoskeletal filaments. As cell surface receptors, integrins are central to the pathophysiology of many brain diseases, such as epilepsy, and are potential targets for the development of new drugs for neurological disorders.
Collapse
Affiliation(s)
- Xin Wu
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center College of Medicine, Bryan, TX 77807, USA
| | | |
Collapse
|
48
|
Gardiner NJ. Integrins and the extracellular matrix: Key mediators of development and regeneration of the sensory nervous system. Dev Neurobiol 2011; 71:1054-72. [DOI: 10.1002/dneu.20950] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
49
|
Hollis ER, Tuszynski MH. Neurotrophins: potential therapeutic tools for the treatment of spinal cord injury. Neurotherapeutics 2011; 8:694-703. [PMID: 21904786 PMCID: PMC3250295 DOI: 10.1007/s13311-011-0074-9] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Spinal cord injury permanently disrupts neuroanatomical circuitry and can result in severe functional deficits. These functional deficits, however, are not immutable and spontaneous recovery occurs in some patients. It is highly likely that this recovery is dependent upon spared tissue and the endogenous plasticity of the central nervous system. Neurotrophic factors are mediators of neuronal plasticity throughout development and into adulthood, affecting proliferation of neuronal precursors, neuronal survival, axonal growth, dendritic arborization and synapse formation. Neurotrophic factors are therefore excellent candidates for enhancing axonal plasticity and regeneration after spinal cord injury. Understanding growth factor effects on axonal growth and utilizing them to alter the intrinsic limitations on regenerative growth will provide potent tools for the development of translational therapeutic interventions for spinal cord injury.
Collapse
Affiliation(s)
- Edmund R. Hollis
- Neurobiology Section, Biological Sciences Division, University of California-San Diego, La Jolla, CA 92093-0366 USA
| | - Mark H. Tuszynski
- Department of Neurosciences, University of California-San Diego, La Jolla, CA 92093-0626 USA
- VA Medical Center, La Jolla, CA 92161 USA
| |
Collapse
|
50
|
New insights into the altered fibronectin matrix and extrasynaptic transmission in the aging brain. ACTA ACUST UNITED AC 2011. [DOI: 10.1016/j.jcgg.2010.12.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|