151
|
Postinjury Induction of Activated ErbB2 Selectively Hyperactivates Denervated Schwann Cells and Promotes Robust Dorsal Root Axon Regeneration. J Neurosci 2017; 37:10955-10970. [PMID: 28982707 DOI: 10.1523/jneurosci.0903-17.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 09/20/2017] [Accepted: 09/27/2017] [Indexed: 11/21/2022] Open
Abstract
Following nerve injury, denervated Schwann cells (SCs) convert to repair SCs, which enable regeneration of peripheral axons. However, the repair capacity of SCs and the regenerative capacity of peripheral axons are limited. In the present studies we examined a potential therapeutic strategy to enhance the repair capacity of SCs, and tested its efficacy in enhancing regeneration of dorsal root (DR) axons, whose regenerative capacity is particularly weak. We used male and female mice of a doxycycline-inducible transgenic line to induce expression of constitutively active ErbB2 (caErbB2) selectively in SCs after DR crush or transection. Two weeks after injury, injured DRs of induced animals contained far more SCs and SC processes. These SCs had not redifferentiated and continued to proliferate. Injured DRs of induced animals also contained far more axons that regrew along SC processes past the transection or crush site. Remarkably, SCs and axons in uninjured DRs remained quiescent, indicating that caErbB2 enhanced regeneration of injured DRs, without aberrantly activating SCs and axons in intact nerves. We also found that intraspinally expressed glial cell line-derived neurotrophic factor (GDNF), but not the removal of chondroitin sulfate proteoglycans, greatly enhanced the intraspinal migration of caErbB2-expressing SCs, enabling robust penetration of DR axons into the spinal cord. These findings indicate that SC-selective, post-injury activation of ErbB2 provides a novel strategy to powerfully enhance the repair capacity of SCs and axon regeneration, without substantial off-target damage. They also highlight that promoting directed migration of caErbB2-expressing SCs by GDNF might be useful to enable axon regrowth in a non-permissive environment.SIGNIFICANCE STATEMENT Repair of injured peripheral nerves remains a critical clinical problem. We currently lack a therapy that potently enhances axon regeneration in patients with traumatic nerve injury. It is extremely challenging to substantially increase the regenerative capacity of damaged nerves without deleterious off-target effects. It was therefore of great interest to discover that caErbB2 markedly enhances regeneration of damaged dorsal roots, while evoking little change in intact roots. To our knowledge, these findings are the first demonstration that repair capacity of denervated SCs can be efficaciously enhanced without altering innervated SCs. Our study also demonstrates that oncogenic ErbB2 signaling can be activated in SCs but not impede transdifferentiation of denervated SCs to regeneration-promoting repair SCs.
Collapse
|
152
|
Couve E, Lovera M, Suzuki K, Schmachtenberg O. Schwann Cell Phenotype Changes in Aging Human Dental Pulp. J Dent Res 2017; 97:347-355. [PMID: 28972819 DOI: 10.1177/0022034517733967] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Schwann cells are glial cells that support axonal development, maintenance, defense, and regeneration in the peripheral nervous system. There is limited knowledge regarding the organization, plasticity, and aging of Schwann cells within the dental pulp in adult permanent teeth. The present study sought to relate changes in the pattern of Schwann cell phenotypes between young and old adult teeth with neuronal, immune, and vascular components of the dental pulp. Schwann cells are shown to form a prominent glial network at the dentin-pulp interface, consisting of nonmyelinating and myelinating phenotypes, forming a multicellular neuroimmune interface in association with nerve fibers and dendritic cells. Schwann cell phenotypes are recognized by the expression of S100, glial fibrillary acidic protein (GFAP), myelin basic protein (MBP), Sox10, GAP43, and p75NTR markers. In young adult teeth, a dense population of nonmyelinating Schwann cells projects processes in close association with sensory nerve terminals through the odontoblast layer, reaching the adjacent predentin/dentin domain. While GAP43 and p75NTR are highly expressed in nonmyelinating Schwann cells from young adult teeth, the presence of these markers declines significantly in old adult teeth. Myelinated axons, identified by MBP expression, are mainly present at the Raschkow plexus and within nerve bundles in the dental pulp, but their density is significantly reduced in old adult versus young adult teeth. These data reveal age-related changes within the glial network of the dental pulp, in association with a reduction of coronal dental pulp innervation in old adult versus young adult teeth. The prominence of Schwann cells as a cellular component at the dentin-pulp interface supports the notion that their association with sensory nerve terminals and immune system components forms part of an integrated multicellular barrier for defense against pathogens and dentin repair.
Collapse
Affiliation(s)
- E Couve
- 1 Instituto de Biología, Laboratorio de Microscopía Electrónica, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - M Lovera
- 1 Instituto de Biología, Laboratorio de Microscopía Electrónica, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - K Suzuki
- 1 Instituto de Biología, Laboratorio de Microscopía Electrónica, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - O Schmachtenberg
- 2 Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
153
|
Tallon C, Farah MH. Beta secretase activity in peripheral nerve regeneration. Neural Regen Res 2017; 12:1565-1574. [PMID: 29171411 PMCID: PMC5696827 DOI: 10.4103/1673-5374.217319] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/26/2017] [Indexed: 12/13/2022] Open
Abstract
While the peripheral nervous system has the capacity to regenerate following a nerve injury, it is often at a slow rate and results in unsatisfactory recovery, leaving patients with reduced function. Many regeneration associated genes have been identified over the years, which may shed some insight into how we can manipulate this intrinsic regenerative ability to enhance repair following peripheral nerve injuries. Our lab has identified the membrane bound protease beta-site amyloid precursor protein-cleaving enzyme 1 (BACE1), or beta secretase, as a potential negative regulator of peripheral nerve regeneration. When beta secretase activity levels are abolished via a null mutation in mice, peripheral regeneration is enhanced following a sciatic nerve crush injury. Conversely, when activity levels are greatly increased by overexpressing beta secretase in mice, nerve regeneration and functional recovery are impaired after a sciatic nerve crush injury. In addition to our work, many substrates of beta secretase have been found to be involved in regulating neurite outgrowth and some have even been identified as regeneration associated genes. In this review, we set out to discuss BACE1 and its substrates with respect to axonal regeneration and speculate on the possibility of utilizing BACE1 inhibitors to enhance regeneration following acute nerve injury and potential uses in peripheral neuropathies.
Collapse
Affiliation(s)
- Carolyn Tallon
- Department of Neurology at Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Mohamed H. Farah
- Department of Neurology at Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
154
|
Arthur-Farraj PJ, Morgan CC, Adamowicz M, Gomez-Sanchez JA, Fazal SV, Beucher A, Razzaghi B, Mirsky R, Jessen KR, Aitman TJ. Changes in the Coding and Non-coding Transcriptome and DNA Methylome that Define the Schwann Cell Repair Phenotype after Nerve Injury. Cell Rep 2017; 20:2719-2734. [PMID: 28903050 PMCID: PMC5608958 DOI: 10.1016/j.celrep.2017.08.064] [Citation(s) in RCA: 152] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Revised: 06/17/2017] [Accepted: 08/18/2017] [Indexed: 12/12/2022] Open
Abstract
Repair Schwann cells play a critical role in orchestrating nerve repair after injury, but the cellular and molecular processes that generate them are poorly understood. Here, we perform a combined whole-genome, coding and non-coding RNA and CpG methylation study following nerve injury. We show that genes involved in the epithelial-mesenchymal transition are enriched in repair cells, and we identify several long non-coding RNAs in Schwann cells. We demonstrate that the AP-1 transcription factor C-JUN regulates the expression of certain micro RNAs in repair Schwann cells, in particular miR-21 and miR-34. Surprisingly, unlike during development, changes in CpG methylation are limited in injury, restricted to specific locations, such as enhancer regions of Schwann cell-specific genes (e.g., Nedd4l), and close to local enrichment of AP-1 motifs. These genetic and epigenomic changes broaden our mechanistic understanding of the formation of repair Schwann cell during peripheral nervous system tissue repair.
Collapse
Affiliation(s)
- Peter J Arthur-Farraj
- Department of Clinical Neurosciences, Addenbrooke's Hospital, University of Cambridge, Cambridge CB2 0QQ, UK; Department of Medicine, Imperial College, London W12 0NN, UK; Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK.
| | - Claire C Morgan
- Department of Medicine, Imperial College, London W12 0NN, UK
| | - Martyna Adamowicz
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH16 2XU, UK
| | - Jose A Gomez-Sanchez
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Shaline V Fazal
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Anthony Beucher
- Department of Medicine, Imperial College, London W12 0NN, UK
| | - Bonnie Razzaghi
- Department of Medicine, Imperial College, London W12 0NN, UK
| | - Rhona Mirsky
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Kristjan R Jessen
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Timothy J Aitman
- Department of Medicine, Imperial College, London W12 0NN, UK; Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH16 2XU, UK.
| |
Collapse
|
155
|
Tallon C, Rockenstein E, Masliah E, Farah MH. Increased BACE1 activity inhibits peripheral nerve regeneration after injury. Neurobiol Dis 2017; 106:147-157. [PMID: 28687442 DOI: 10.1016/j.nbd.2017.07.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 06/26/2017] [Accepted: 07/02/2017] [Indexed: 12/20/2022] Open
Abstract
Axons of the peripheral nervous system possess the capacity to regenerate following injury. Previously, we showed that genetically knocking out Beta-Site APP-Cleaving Enzyme 1 (BACE1) leads to increased nerve regeneration. Two cellular components, macrophages and neurons, contribute to enhanced nerve regeneration in BACE1 knockout mice. Here, we utilized a transgenic mouse model that overexpresses BACE1 in its neurons to investigate whether neuronal BACE1 has an inverse effect on regeneration following nerve injury. We performed a sciatic nerve crush in BACE1 transgenic mice and control wild-type littermates, and evaluated the extent of both morphological and physiological improvements over time. At the earliest time point of 3days, we observed a significant decrease in the length of axonal sprouts growing out from the crush site in BACE1 transgenic mice. At later times (10 and 15days post-crush), there were significant reductions in the number of myelinated axons in the sciatic nerve and the percentage of re-innervated neuromuscular junctions in the gastrocnemius muscle. Transgenic mice had a functional electrophysiological delay in the recovery up to 8weeks post-crush compared to controls. These results indicate that BACE1 activity levels have an inverse effect on peripheral nerve repair after injury. The results obtained in this study provide evidence that neuronal BACE1 activity levels impact peripheral nerve regeneration. This data has clinical relevance by highlighting a novel drug target to enhance peripheral nerve repair, an area which currently does not have any approved therapeutics.
Collapse
Affiliation(s)
- Carolyn Tallon
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Edward Rockenstein
- Department of Neurosciences, San Diego School of Medicine, University of California, San Diego, CA, United States
| | - Eliezer Masliah
- Department of Neurosciences, San Diego School of Medicine, University of California, San Diego, CA, United States; Department of Molecular Pathology, San Diego School of Medicine, University of California, San Diego, CA, United States
| | - Mohamed H Farah
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
156
|
Mau T, Pan HM, Childs LF. The natural history of recoverable vocal fold paralysis: Implications for kinetics of reinnervation. Laryngoscope 2017; 127:2585-2590. [PMID: 28608475 DOI: 10.1002/lary.26734] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 05/05/2017] [Accepted: 05/15/2017] [Indexed: 11/08/2022]
Abstract
OBJECTIVES/HYPOTHESIS Patients with unilateral vocal fold paralysis (UVFP) are commonly told to wait 12 months for spontaneous recovery. This study aims to 1) determine the time to vocal recovery in UVFP, 2) use that data to develop a neurophysiologically plausible model for recovery, and 3) use the model to generate meaningful predictions for patient counseling. STUDY DESIGN Case series with de novo mathematical modeling. METHODS Patients with UVFP who could pinpoint a discrete onset of vocal improvement were identified. The time-to-recovery data were modeled by assuming an "early" recovery group with neuropraxia and a "late" recovery group with more severe nerve injury. For the late group, a two-stage model was developed to explain the time to recovery: regenerating axons must cross the site of injury in stage 1 (probabilistic), followed by unimpeded regrowth to the larynx in stage 2 (deterministic). RESULTS Of 727 cases of UVFP over a 7-year period, 44 reported spontaneous recovery with a discrete onset of vocal improvement. A hybrid distribution incorporating the two stages (exponentially modified Gaussian) accurately modeled the time-to-recovery data (R2 = 0.918). The model predicts 86% of patients with recoverable UVFP will recover within 6 months, with 96% recovering within 9 months. Earlier vocal recovery is associated with recovery of vocal fold motion and younger age. CONCLUSIONS Waiting 12 months for spontaneous recovery is probably too conservative. Repair across the site of injury, and not regrowth to larynx, is likely the rate-determining step in reinnervation, consistent with other works on peripheral nerve regeneration. LEVEL OF EVIDENCE 4. Laryngoscope, 127:2585-2590, 2017.
Collapse
Affiliation(s)
- Ted Mau
- Clinical Center for Voice Care, Department of Otolaryngology-Head and Neck Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, U.S.A
| | - Hao-Min Pan
- Clinical Center for Voice Care, Department of Otolaryngology-Head and Neck Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, U.S.A
| | - Lesley F Childs
- Clinical Center for Voice Care, Department of Otolaryngology-Head and Neck Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, U.S.A
| |
Collapse
|
157
|
Nikolich-Žugich J, Davies JS. Homeostatic migration and distribution of innate immune cells in primary and secondary lymphoid organs with ageing. Clin Exp Immunol 2017; 187:337-344. [PMID: 28035684 DOI: 10.1111/cei.12920] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 12/18/2016] [Accepted: 12/21/2016] [Indexed: 01/09/2023] Open
Abstract
Ageing of the innate and adaptive immune system, collectively termed immune senescence, is a complex process. One method to understand the components of ageing involves dissociating the effects of ageing on the cells of the immune system, on the microenvironment in lymphoid organs and tissues where immune cells reside and on the circulating factors that interact with both immune cells and their microenvironment. Heterochronic parabiosis, a surgical union of two organisms of disparate ages, is ideal for this type of study, as it has the power to dissociate the age of the cell and the age of the microenvironment into which the cell resides or is migrating. So far, however, it has been used sparingly to study immune ageing. Here we review the limited literature on homeostatic innate immune cell trafficking in ageing in the absence of chronic inflammation. We also review our own recent data on trafficking of innate immune subsets between primary and secondary lymphoid organs in heterochronic parabiosis. We found no systemic bias in retention or acceptance of neutrophils, macrophages, dendritic cells or natural killer cells with ageing in primary and secondary lymphoid organs. We conclude that these four innate immune cell types migrate to and populate lymphoid organs (peripheral lymph nodes, spleen and bone marrow), regardless of their own age and of the age of lymphoid organs.
Collapse
Affiliation(s)
- J Nikolich-Žugich
- Department of Immunobiology and the Arizona Center on Aging, University of Arizona College of Medicine, Tucson, AZ, USA
| | - J S Davies
- Department of Immunobiology and the Arizona Center on Aging, University of Arizona College of Medicine, Tucson, AZ, USA
| |
Collapse
|
158
|
Ghosh S, Basak P, Dutta S, Chowdhury S, Sil PC. New insights into the ameliorative effects of ferulic acid in pathophysiological conditions. Food Chem Toxicol 2017; 103:41-55. [PMID: 28237775 DOI: 10.1016/j.fct.2017.02.028] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 02/16/2017] [Accepted: 02/20/2017] [Indexed: 12/21/2022]
Abstract
Ferulic acid, a natural phytochemical has gained importance as a potential therapeutic agent by virtue of its easy commercial availability, low cost and minimal side-effects. It is a derivative of curcumin and possesses the necessary pharmacokinetic properties to be retained in the general circulation for several hours. The therapeutic effects of ferulic acid are mediated through its antioxidant and anti-inflammatory properties. It exhibits different biological activities such as anti-inflammatory, anti-apoptotic, anti-carcinogenic, anti-diabetic, hepatoprotective, cardioprotective, neuroprotective actions, etc. The current review addresses its therapeutic effects under different pathophysiological conditions (eg. cancer, cardiomyopathy, skin disorders, brain disorders, viral infections, diabetes etc.).
Collapse
Affiliation(s)
- Sumit Ghosh
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India
| | - Priyanka Basak
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India
| | - Sayanta Dutta
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India
| | - Sayantani Chowdhury
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India
| | - Parames C Sil
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India.
| |
Collapse
|
159
|
Boerboom A, Dion V, Chariot A, Franzen R. Molecular Mechanisms Involved in Schwann Cell Plasticity. Front Mol Neurosci 2017; 10:38. [PMID: 28261057 PMCID: PMC5314106 DOI: 10.3389/fnmol.2017.00038] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 01/31/2017] [Indexed: 01/09/2023] Open
Abstract
Schwann cell incredible plasticity is a hallmark of the utmost importance following nerve damage or in demyelinating neuropathies. After injury, Schwann cells undergo dedifferentiation before redifferentiating to promote nerve regeneration and complete functional recovery. This review updates and discusses the molecular mechanisms involved in the negative regulation of myelination as well as in the reprogramming of Schwann cells taking place early following nerve lesion to support repair. Significant advance has been made on signaling pathways and molecular components that regulate SC regenerative properties. These include for instance transcriptional regulators such as c-Jun or Notch, the MAPK and the Nrg1/ErbB2/3 pathways. This comprehensive overview ends with some therapeutical applications targeting factors that control Schwann cell plasticity and highlights the need to carefully modulate and balance this capacity to drive nerve repair.
Collapse
Affiliation(s)
| | - Valérie Dion
- GIGA-Neurosciences, University of Liège Liège, Belgium
| | - Alain Chariot
- GIGA-Molecular Biology of Diseases, University of LiègeLiège, Belgium; Walloon Excellence in Lifesciences and Biotechnology (WELBIO)Wavre, Belgium
| | | |
Collapse
|
160
|
Mindos T, Dun XP, North K, Doddrell RDS, Schulz A, Edwards P, Russell J, Gray B, Roberts SL, Shivane A, Mortimer G, Pirie M, Zhang N, Pan D, Morrison H, Parkinson DB. Merlin controls the repair capacity of Schwann cells after injury by regulating Hippo/YAP activity. J Cell Biol 2017; 216:495-510. [PMID: 28137778 PMCID: PMC5294779 DOI: 10.1083/jcb.201606052] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 10/23/2016] [Accepted: 12/27/2016] [Indexed: 02/06/2023] Open
Abstract
Loss of the Merlin tumor suppressor and activation of the Hippo signaling pathway play major roles in the control of cell proliferation and tumorigenesis. We have identified completely novel roles for Merlin and the Hippo pathway effector Yes-associated protein (YAP) in the control of Schwann cell (SC) plasticity and peripheral nerve repair after injury. Injury to the peripheral nervous system (PNS) causes a dramatic shift in SC molecular phenotype and the generation of repair-competent SCs, which direct functional repair. We find that loss of Merlin in these cells causes a catastrophic failure of axonal regeneration and remyelination in the PNS. This effect is mediated by activation of YAP expression in Merlin-null SCs, and loss of YAP restores axonal regrowth and functional repair. This work identifies new mechanisms that control the regenerative potential of SCs and gives new insight into understanding the correct control of functional nerve repair in the PNS.
Collapse
Affiliation(s)
- Thomas Mindos
- Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth PL6 8BU, England, UK
| | - Xin-Peng Dun
- Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth PL6 8BU, England, UK
| | - Katherine North
- Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth PL6 8BU, England, UK
- University of Bath, Bath BA2 7AY, England, UK
| | - Robin D S Doddrell
- Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth PL6 8BU, England, UK
| | - Alexander Schulz
- Leibniz Institute for Age Research - Fritz Lipmann Institute Jena, D-07745 Jena, Germany
| | - Philip Edwards
- Department of Cellular and Anatomical Pathology, Derriford Hospital, Plymouth PL6 8DH, England, UK
| | - James Russell
- Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth PL6 8BU, England, UK
| | - Bethany Gray
- Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth PL6 8BU, England, UK
- University of Bath, Bath BA2 7AY, England, UK
| | - Sheridan L Roberts
- Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth PL6 8BU, England, UK
| | - Aditya Shivane
- Department of Cellular and Anatomical Pathology, Derriford Hospital, Plymouth PL6 8DH, England, UK
| | - Georgina Mortimer
- Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth PL6 8BU, England, UK
| | - Melissa Pirie
- Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth PL6 8BU, England, UK
| | - Nailing Zhang
- Department of Molecular Biology and Genetics, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Duojia Pan
- Department of Molecular Biology and Genetics, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Helen Morrison
- Leibniz Institute for Age Research - Fritz Lipmann Institute Jena, D-07745 Jena, Germany
| | - David B Parkinson
- Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth PL6 8BU, England, UK
| |
Collapse
|
161
|
Abstract
Axonal degeneration is a pivotal feature of many neurodegenerative conditions and substantially accounts for neurological morbidity. A widely used experimental model to study the mechanisms of axonal degeneration is Wallerian degeneration (WD), which occurs after acute axonal injury. In the peripheral nervous system (PNS), WD is characterized by swift dismantling and clearance of injured axons with their myelin sheaths. This is a prerequisite for successful axonal regeneration. In the central nervous system (CNS), WD is much slower, which significantly contributes to failed axonal regeneration. Although it is well-documented that Schwann cells (SCs) have a critical role in the regenerative potential of the PNS, to date we have only scarce knowledge as to how SCs ‘sense’ axonal injury and immediately respond to it. In this regard, it remains unknown as to whether SCs play the role of a passive bystander or an active director during the execution of the highly orchestrated disintegration program of axons. Older reports, together with more recent studies, suggest that SCs mount dynamic injury responses minutes after axonal injury, long before axonal breakdown occurs. The swift SC response to axonal injury could play either a pro-degenerative role, or alternatively a supportive role, to the integrity of distressed axons that have not yet committed to degenerate. Indeed, supporting the latter concept, recent findings in a chronic PNS neurodegeneration model indicate that deactivation of a key molecule promoting SC injury responses exacerbates axonal loss. If this holds true in a broader spectrum of conditions, it may provide the grounds for the development of new glia-centric therapeutic approaches to counteract axonal loss.
Collapse
Affiliation(s)
- Keit Men Wong
- Hunter James Kelly Research Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Elisabetta Babetto
- Hunter James Kelly Research Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA.,Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Bogdan Beirowski
- Hunter James Kelly Research Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA.,Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| |
Collapse
|
162
|
Sánchez M, Garate A, Delgado D, Padilla S. Platelet-rich plasma, an adjuvant biological therapy to assist peripheral nerve repair. Neural Regen Res 2017; 12:47-52. [PMID: 28250739 PMCID: PMC5319232 DOI: 10.4103/1673-5374.198973] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Therapies such as direct tension-free microsurgical repair or transplantation of a nerve autograft, are nowadays used to treat traumatic peripheral nerve injuries (PNI), focused on the enhancement of the intrinsic regenerative potential of injured axons. However, these therapies fail to recreate the suitable cellular and molecular microenvironment of peripheral nerve repair and in some cases, the functional recovery of nerve injuries is incomplete. Thus, new biomedical engineering strategies based on tissue engineering approaches through molecular intervention and scaffolding offer promising outcomes on the field. In this sense, evidence is accumulating in both, preclinical and clinical settings, indicating that platelet-rich plasma products, and fibrin scaffold obtained from this technology, hold an important therapeutic potential as a neuroprotective, neurogenic and neuroinflammatory therapeutic modulator system, as well as enhancing the sensory and motor functional nerve muscle unit recovery.
Collapse
Affiliation(s)
- Mikel Sánchez
- Arthroscopic Surgery Unit, Hospital Vithas San José, Vitoria-Gasteiz, Spain; Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Ane Garate
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Diego Delgado
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | | |
Collapse
|
163
|
Tedeschi A, Bradke F. Spatial and temporal arrangement of neuronal intrinsic and extrinsic mechanisms controlling axon regeneration. Curr Opin Neurobiol 2016; 42:118-127. [PMID: 28039763 DOI: 10.1016/j.conb.2016.12.005] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 12/07/2016] [Accepted: 12/08/2016] [Indexed: 01/30/2023]
Abstract
Axon regeneration and neuronal tissue repair varies across animal lineages as well as in the mammalian central and peripheral nervous systems. While the peripheral nervous system retains the ability to self-repair, the majority of axons in the adult mammalian central nervous system (CNS) fail to reactivate intrinsic growth programs after injury. Recent findings, however, suggest that long-distance axon regeneration, neuronal circuit assembly and recovery of functions in the adult mammalian CNS are possible. Here, we discuss our current knowledge of the cell signaling pathways and networks controlling axon regeneration. In addition, we outline a number of combinatorial strategies that include among others microtubule-based treatments to foster regeneration and functional connectivity after CNS trauma.
Collapse
Affiliation(s)
- Andrea Tedeschi
- German Center for Neurodegenerative Diseases (DZNE), Ludwig-Erhard-Allee 2, 53175 Bonn, Germany.
| | - Frank Bradke
- German Center for Neurodegenerative Diseases (DZNE), Ludwig-Erhard-Allee 2, 53175 Bonn, Germany.
| |
Collapse
|
164
|
Sánchez M, Anitua E, Delgado D, Sanchez P, Prado R, Orive G, Padilla S. Platelet-rich plasma, a source of autologous growth factors and biomimetic scaffold for peripheral nerve regeneration. Expert Opin Biol Ther 2016; 17:197-212. [DOI: 10.1080/14712598.2017.1259409] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Mikel Sánchez
- Arthroscopic Surgery Unit, Hospital Vithas San José, Vitoria-Gasteiz, Spain
| | - Eduardo Anitua
- BTI Biotechnology Institute, Vitoria, Spain
- Eduardo Anitua Foundation, Vitoria, Spain
| | - Diego Delgado
- Arthroscopic Surgery Unit Research, Hospital Vithas San José, Vitoria-Gasteiz, Spain
| | - Peio Sanchez
- Arthroscopic Surgery Unit Research, Hospital Vithas San José, Vitoria-Gasteiz, Spain
| | | | - Gorka Orive
- BTI Biotechnology Institute, Vitoria, Spain
- Eduardo Anitua Foundation, Vitoria, Spain
- Lab of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of The Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
- Centro de Investigación Biomédica en Red, Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Sabino Padilla
- BTI Biotechnology Institute, Vitoria, Spain
- Eduardo Anitua Foundation, Vitoria, Spain
| |
Collapse
|
165
|
The age factor in axonal repair after spinal cord injury: A focus on neuron-intrinsic mechanisms. Neurosci Lett 2016; 652:41-49. [PMID: 27818358 DOI: 10.1016/j.neulet.2016.11.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 10/26/2016] [Accepted: 11/01/2016] [Indexed: 11/27/2022]
Abstract
Age is an important consideration for recovery and repair after spinal cord injury. Spinal cord injury is increasingly affecting the middle-aged and aging populations. Despite rapid progress in research to promote axonal regeneration and repair, our understanding of how age can modulate this repair is rather limited. In this review, we discuss the literature supporting the notion of an age-dependent decline in axonal growth after central nervous system (CNS) injury. While both neuron-intrinsic and extrinsic factors are involved in the control of axon growth after injury, here we focus on possible intrinsic mechanisms for this age-dependent decline.
Collapse
|
166
|
Weng YL, Joseph J, An R, Song H, Ming GL. Epigenetic regulation of axonal regenerative capacity. Epigenomics 2016; 8:1429-1442. [PMID: 27642866 DOI: 10.2217/epi-2016-0058] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The intrinsic growth capacity of neurons in the CNS declines during neuronal maturation, while neurons in the adult PNS are capable of regeneration. Injured mature PNS neurons require activation of an array of regeneration-associated genes to regain axonal growth competence. Accumulating evidence indicates a pivotal role of epigenetic mechanisms in transcriptional reprogramming and regulation of neuronal growth ability upon injury. In this review, we summarize the latest findings implicating epigenetic mechanisms, including histone and DNA modifications, in axon regeneration and discuss differential epigenomic configurations between neurons in the adult mammalian CNS and PNS.
Collapse
Affiliation(s)
- Yi-Lan Weng
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jessica Joseph
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Graduate Program in Cellular & Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ran An
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hongjun Song
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Graduate Program in Cellular & Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,The Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Guo-Li Ming
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Graduate Program in Cellular & Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,The Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
167
|
Zhang Q, Nguyen P, Xu Q, Park W, Lee S, Furuhashi A, Le AD. Neural Progenitor-Like Cells Induced from Human Gingiva-Derived Mesenchymal Stem Cells Regulate Myelination of Schwann Cells in Rat Sciatic Nerve Regeneration. Stem Cells Transl Med 2016; 6:458-470. [PMID: 28191764 PMCID: PMC5442816 DOI: 10.5966/sctm.2016-0177] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 07/28/2016] [Indexed: 12/26/2022] Open
Abstract
Regeneration of peripheral nerve injury remains a major clinical challenge. Recently, mesenchymal stem cells (MSCs) have been considered as potential candidates for peripheral nerve regeneration; however, the underlying mechanisms remain elusive. Here, we show that human gingiva‐derived MSCs (GMSCs) could be directly induced into multipotent NPCs (iNPCs) under minimally manipulated conditions without the introduction of exogenous genes. Using a crush‐injury model of rat sciatic nerve, we demonstrate that GMSCs transplanted to the injury site could differentiate into neuronal cells, whereas iNPCs could differentiate into both neuronal and Schwann cells. After crush injury, iNPCs, compared with GMSCs, displayed superior therapeutic effects on axonal regeneration at both the injury site and the distal segment of the injured sciatic nerve. Mechanistically, transplantation of GMSCs, especially iNPCs, significantly attenuated injury‐triggered increase in the expression of c‐Jun, a transcription factor that functions as a major negative regulator of myelination and plays a central role in dedifferentiation/reprogramming of Schwann cells into a progenitor‐like state. Meanwhile, our results also demonstrate that transplantation of GMSCs and iNPCs consistently increased the expression of Krox‐20/EGR2, a transcription factor that governs the expression of myelin proteins and facilitates myelination. Altogether, our findings suggest that transplantation of GMSCs and iNPCs promotes peripheral nerve repair/regeneration, possibly by promoting remyelination of Schwann cells mediated via the regulation of the antagonistic myelination regulators, c‐Jun and Krox‐20/EGR2. Stem Cells Translational Medicine2017;6:458–470
Collapse
Affiliation(s)
- Qunzhou Zhang
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania, USA
| | - Phuong Nguyen
- Division of Plastic and Reconstructive Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Qilin Xu
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania, USA
| | - Wonse Park
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania, USA
| | - Sumin Lee
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania, USA
| | - Akihiro Furuhashi
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania, USA
| | - Anh D. Le
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania, USA
- Department of Oral and Maxillofacial Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
168
|
Aare S, Spendiff S, Vuda M, Elkrief D, Perez A, Wu Q, Mayaki D, Hussain SNA, Hettwer S, Hepple RT. Failed reinnervation in aging skeletal muscle. Skelet Muscle 2016; 6:29. [PMID: 27588166 PMCID: PMC5007704 DOI: 10.1186/s13395-016-0101-y] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 08/05/2016] [Indexed: 12/08/2023] Open
Abstract
BACKGROUND Skeletal muscle displays a marked accumulation of denervated myofibers at advanced age, which coincides with an acceleration of muscle atrophy. METHODS In this study, we evaluated the hypothesis that the accumulation of denervated myofibers in advanced age is due to failed reinnervation by examining muscle from young adult (YA) and very old (VO) rats and from a murine model of sporadic denervation secondary to neurotrypsin over-expression (Sarco mouse). RESULTS Both aging rat muscle and Sarco mouse muscle exhibited marked fiber-type grouping, consistent with repeating cycles of denervation and reinnervation, yet in VO muscle, rapsyn at the endplate increased and was associated with only a 10 % decline in acetylcholine receptor (AChR) intensity, whereas in Sarco mice, there was a decline in rapsyn and a 25 % decrease in AChR intensity. Transcripts of muscle-specific kinase (21-fold), acetylcholine receptor subunits α (68-fold), ε (threefold) and γ (47-fold), neural cell adhesion molecule (66-fold), and runt-related transcription factor 1 (33-fold) were upregulated in VO muscle of the rat, consistent with the marked persistent denervation evidenced by a large proportion of very small fibers (>20 %). In the Sarco mice, there were much smaller increases in denervation transcripts (0-3.5-fold) and accumulation of very small fibers (2-6 %) compared to the VO rat, suggesting a reduced capacity for reinnervation in aging muscle. Despite the marked persistent denervation in the VO rat muscle, transcripts of neurotrophins involved in promoting axonal sprouting following denervation exhibited no increase, and several miRNAs predicted to suppress neurotrophins were elevated in VO rat. CONCLUSIONS Our results support the hypothesis that the accumulation of denervated fibers with aging is due to failed reinnervation and that this may be affected by a limited neurotrophin response that mediates axonal sprouting following denervation.
Collapse
Affiliation(s)
- Sudhakar Aare
- Research Institute of the McGill University Health Centre, EM2.2232, RI MUHC, 1001 Decarie Blvd, Montreal, QC Canada H4A 3J1 ; McGill Research Centre for Physical Activity and Health, McGill University, Montreal, QC Canada
| | - Sally Spendiff
- Research Institute of the McGill University Health Centre, EM2.2232, RI MUHC, 1001 Decarie Blvd, Montreal, QC Canada H4A 3J1 ; McGill Research Centre for Physical Activity and Health, McGill University, Montreal, QC Canada
| | - Madhusudanarao Vuda
- Research Institute of the McGill University Health Centre, EM2.2232, RI MUHC, 1001 Decarie Blvd, Montreal, QC Canada H4A 3J1 ; McGill Research Centre for Physical Activity and Health, McGill University, Montreal, QC Canada
| | - Daren Elkrief
- Research Institute of the McGill University Health Centre, EM2.2232, RI MUHC, 1001 Decarie Blvd, Montreal, QC Canada H4A 3J1 ; McGill Research Centre for Physical Activity and Health, McGill University, Montreal, QC Canada ; Department of Kinesiology and Physical Education, McGill University, Montreal, QC Canada
| | - Anna Perez
- Research Institute of the McGill University Health Centre, EM2.2232, RI MUHC, 1001 Decarie Blvd, Montreal, QC Canada H4A 3J1
| | - Qinghua Wu
- Research Institute of the McGill University Health Centre, EM2.2232, RI MUHC, 1001 Decarie Blvd, Montreal, QC Canada H4A 3J1 ; McGill Research Centre for Physical Activity and Health, McGill University, Montreal, QC Canada
| | - Dominique Mayaki
- Research Institute of the McGill University Health Centre, EM2.2232, RI MUHC, 1001 Decarie Blvd, Montreal, QC Canada H4A 3J1
| | - Sabah N A Hussain
- Research Institute of the McGill University Health Centre, EM2.2232, RI MUHC, 1001 Decarie Blvd, Montreal, QC Canada H4A 3J1
| | | | - Russell T Hepple
- Research Institute of the McGill University Health Centre, EM2.2232, RI MUHC, 1001 Decarie Blvd, Montreal, QC Canada H4A 3J1 ; McGill Research Centre for Physical Activity and Health, McGill University, Montreal, QC Canada ; Department of Kinesiology and Physical Education, McGill University, Montreal, QC Canada
| |
Collapse
|
169
|
Ma KH, Hung HA, Svaren J. Epigenomic Regulation of Schwann Cell Reprogramming in Peripheral Nerve Injury. J Neurosci 2016; 36:9135-47. [PMID: 27581455 PMCID: PMC5005723 DOI: 10.1523/jneurosci.1370-16.2016] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 07/08/2016] [Accepted: 07/15/2016] [Indexed: 12/16/2022] Open
Abstract
UNLABELLED The rapid and dynamic transcriptional changes of Schwann cells in response to injury are critical to peripheral nerve repair, yet the epigenomic reprograming that leads to the induction of injury-activated genes has not been characterized. Polycomb Repressive Complex 2 (PRC2) catalyzes the trimethylation of lysine 27 of histone H3 (H3K27me3), which produces a transcriptionally repressive chromatin environment. We find that many promoters and/or gene bodies of injury-activated genes of mature rat nerves are occupied with H3K27me3. In contrast, the majority of distal enhancers that gain H3K27 acetylation after injury are not repressed by H3K27 methylation before injury, which is normally observed in developmentally poised enhancers. Injury induces demethylation of H3K27 in many genes, such as Sonic hedgehog (Shh), which is silenced throughout Schwann cell development before injury. In addition, experiments using a Schwann cell-specific mouse knock-out of the Eed subunit of PRC2 indicate that demethylation is a rate-limiting step in the activation of such genes. We also show that some transcription start sites of H3K27me3-repressed injury genes of uninjured nerves are bound with a mark of active promoters H3K4me3, for example, Shh and Gdnf, and the reduction of H3K27me3 results in increased trimethylation of H3K4. Our findings identify reversal of polycomb repression as a key step in gene activation after injury. SIGNIFICANCE STATEMENT Peripheral nerve regeneration after injury is dependent upon implementation of a novel genetic program in Schwann cells that supports axonal survival and regeneration. Identifying means to enhance Schwann cell reprogramming after nerve injury could be used to foster effective remyelination in the treatment of demyelinating disorders and in identifying pathways involved in regenerative process of myelination. Although recent progress has identified transcriptional determinants of successful reprogramming of the Schwann cell transcriptome after nerve injury, our results have highlighted a novel epigenomic pathway in which reversal of the Polycomb pathway of repressive histone methylation is required for activation of a significant number of injury-induced genes.
Collapse
Affiliation(s)
- Ki H Ma
- Waisman Center, Cellular and Molecular Pathology Graduate Program, and
| | - Holly A Hung
- Waisman Center, Cellular and Molecular Pathology Graduate Program, and
| | - John Svaren
- Waisman Center, Department of Comparative Biosciences, University of Wisconsin, Madison, Wisconsin 53705
| |
Collapse
|
170
|
Cattin AL, Lloyd AC. The multicellular complexity of peripheral nerve regeneration. Curr Opin Neurobiol 2016; 39:38-46. [PMID: 27128880 DOI: 10.1016/j.conb.2016.04.005] [Citation(s) in RCA: 169] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 03/31/2016] [Accepted: 04/13/2016] [Indexed: 12/20/2022]
Abstract
Peripheral nerves show a remarkable ability to regenerate following a transection injury. Downstream of the cut, the axons degenerate and so to regenerate the nerve, the severed axons need to regrow back to their targets and regain function. This requires the axons to navigate through two different environments. (1) The bridge of new tissue that forms between the two nerve stumps and (2) the distal stump of the nerve that remains associated with the target tissues. This involves distinct, complex multicellular responses that guide and sustain axonal regrowth. These processes have important implications for our understanding of the regeneration of an adult tissue and have parallels to aspects of tumour formation and spread.
Collapse
Affiliation(s)
- Anne-Laure Cattin
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Alison C Lloyd
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK; UCL Cancer Institute, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
171
|
Tata PR, Rajagopal J. Cellular plasticity: 1712 to the present day. Curr Opin Cell Biol 2016; 43:46-54. [PMID: 27485353 DOI: 10.1016/j.ceb.2016.07.005] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 06/18/2016] [Accepted: 07/07/2016] [Indexed: 12/18/2022]
Abstract
Cell identity is a fundamental feature of cells. Tissues are often organized into cellular hierarchies characterized by progressive differentiation and developmental commitment. However, it is been historically evident that the cells of many organisms of various phyla, especially in the context of injury, exhibit remarkable plasticity in terms of their ability to convert into other cell types. Recent modern studies, using genetic lineage tracing, have demonstrated that many mature functional cells retain a potential to undergo lineage reversion (dedifferentiation) or to convert into cells of other more distant lineages (transdifferentiation) following injury. Similarly, mimicking progenitor cell transdetermination, stem cells can interconvert. These forms of plasticity may be essential for organismal survival, and are likely part and parcel of regeneration.
Collapse
Affiliation(s)
- Purushothama Rao Tata
- Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA, USA; Harvard Stem Cell Institute, Cambridge, MA, USA; Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Jayaraj Rajagopal
- Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA, USA; Harvard Stem Cell Institute, Cambridge, MA, USA; Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA; Massachusetts General Hospital for Children, Pediatric Pulmonary Medicine, Boston, MA, USA; Division of Otology and Laryngology, Massachusetts Eye and Ear, Boston, MA, USA.
| |
Collapse
|
172
|
Scheib J, Höke A. Impaired regeneration in aged nerves: Clearing out the old to make way for the new. Exp Neurol 2016; 284:79-83. [PMID: 27451012 DOI: 10.1016/j.expneurol.2016.07.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 07/10/2016] [Accepted: 07/19/2016] [Indexed: 01/12/2023]
Abstract
Although many observational studies have shown that peripheral nerve regeneration is impaired with aging, underlying cellular and molecular mechanisms have remained obscure until recently. A series of recent genetic, live imaging and heterochronic parabiosis experiments are providing new insights into the underlying mechanisms of reduced regenerative capacity with aging. These studies show that Schwann cells pose a primary impediment to axon regeneration in older animals as they fail to support regenerating axons, while the contribution from macrophages remains an unresolved issue. Neurons do not appear to have an intrinsic defect of axonal elongation with aging but are impaired when they encounter an inhibitory environment, suggesting that therapeutic approaches to improve intrinsic neuronal regeneration capacity across inhibitory environments, as it is being done in central nervous system regeneration, can improve peripheral nerve regeneration as well. As in many aspects of neuroscience therapeutics development, a combinatorial approach may yield the best outcomes for nerve regeneration in aged individuals.
Collapse
Affiliation(s)
- Jami Scheib
- Departments of Neurology and Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Ahmet Höke
- Departments of Neurology and Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
173
|
|
174
|
Zhu X, Li K, Guo X, Wang J, Xiang Y. Schwann cell proliferation and differentiation that is induced by ferulic acid through MEK1/ERK1/2 signalling promotes peripheral nerve remyelination following crush injury in rats. Exp Ther Med 2016; 12:1915-1921. [PMID: 27588110 DOI: 10.3892/etm.2016.3525] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 05/10/2016] [Indexed: 11/06/2022] Open
Abstract
Schwann cell proliferation and differentiation is critical for the remyelination of injured peripheral nerves. Ferulic acid (FA) is a widely used antioxidant agent with neuroprotective properties. However, the potentially beneficial effects of FA on Schwann cells are unknown. Therefore, the present study was designed to examine the effects of FA on Schwann cell proliferation and differentiation. By using the cultured primary Schwann cells and proliferation assay, the results identified that FA was capable of increasing Schwann cell proliferation and expression of myelin-associated glycoprotein (MAG) and myelin basic protein (MBP) in vitro. It was also observed that the beneficial effect of FA treatment on Schwann cells was mainly dependent on the activation of MEK1/ERK1/2 signalling. Furthermore, FA was intraperitoneally administered to rats with sciatic nerve crush injury, and the results revealed an increase in Schwann cell proliferation and differentiation, while the MAG and MBP expression levels in sciatic nerves were markedly upregulated following FA administration. In conclusion, the current results demonstrate that Schwann cell proliferation and differentiation is induced by FA through MEK1/ERK1/2 signalling and that FA may accelerate injured peripheral nerve remyelination.
Collapse
Affiliation(s)
- Xiaoyan Zhu
- Center of Laboratory Medicine, Chengdu Military General Hospital, Chengdu, Sichuan 610083, P.R. China
| | - Kun Li
- Center of Laboratory Medicine, Chengdu Military General Hospital, Chengdu, Sichuan 610083, P.R. China
| | - Xin Guo
- Center of Laboratory Medicine, Chengdu Military General Hospital, Chengdu, Sichuan 610083, P.R. China
| | - Jian Wang
- Department of Neurology, Chengdu Military General Hospital, Chengdu, Sichuan 610083, P.R. China
| | - Yang Xiang
- Department of Neurology, Chengdu Military General Hospital, Chengdu, Sichuan 610083, P.R. China
| |
Collapse
|
175
|
Poppler LH, Ee X, Schellhardt L, Hoben GM, Pan D, Hunter DA, Yan Y, Moore AM, Snyder-Warwick AK, Stewart SA, Mackinnon SE, Wood MD. Axonal Growth Arrests After an Increased Accumulation of Schwann Cells Expressing Senescence Markers and Stromal Cells in Acellular Nerve Allografts. Tissue Eng Part A 2016; 22:949-61. [PMID: 27297909 DOI: 10.1089/ten.tea.2016.0003] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Acellular nerve allografts (ANAs) and other nerve constructs do not reliably facilitate axonal regeneration across long defects (>3 cm). Causes for this deficiency are poorly understood. In this study, we determined what cells are present within ANAs before axonal growth arrest in nerve constructs and if these cells express markers of cellular stress and senescence. Using the Thy1-GFP rat and serial imaging, we identified the time and location of axonal growth arrest in long (6 cm) ANAs. Axonal growth halted within long ANAs by 4 weeks, while axons successfully regenerated across short (3 cm) ANAs. Cellular populations and markers of senescence were determined using immunohistochemistry, histology, and senescence-associated β-galactosidase staining. Both short and long ANAs were robustly repopulated with Schwann cells (SCs) and stromal cells by 2 weeks. Schwann cells (S100β(+)) represented the majority of cells repopulating both ANAs. Overall, both ANAs demonstrated similar cellular populations with the exception of increased stromal cells (fibronectin(+)/S100β(-)/CD68(-) cells) in long ANAs. Characterization of ANAs for markers of cellular senescence revealed that long ANAs accumulated much greater levels of senescence markers and a greater percentage of Schwann cells expressing the senescence marker p16 compared to short ANAs. To establish the impact of the long ANA environment on axonal regeneration, short ANAs (2 cm) that would normally support axonal regeneration were generated from long ANAs near the time of axonal growth arrest ("stressed" ANAs). These stressed ANAs contained mainly S100β(+)/p16(+) cells and markedly reduced axonal regeneration. In additional experiments, removal of the distal portion (4 cm) of long ANAs near the time of axonal growth arrest and replacement with long isografts (4 cm) rescued axonal regeneration across the defect. Neuronal culture derived from nerve following axonal growth arrest in long ANAs revealed no deficits in axonal extension. Overall, this evidence demonstrates that long ANAs are repopulated with increased p16(+) Schwann cells and stromal cells compared to short ANAs, suggesting a role for these cells in poor axonal regeneration across nerve constructs.
Collapse
Affiliation(s)
- Louis H Poppler
- 1 Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine , St. Louis, Missouri
| | - Xueping Ee
- 1 Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine , St. Louis, Missouri
| | - Lauren Schellhardt
- 1 Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine , St. Louis, Missouri
| | - Gwendolyn M Hoben
- 1 Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine , St. Louis, Missouri
| | - Deng Pan
- 1 Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine , St. Louis, Missouri
| | - Daniel A Hunter
- 1 Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine , St. Louis, Missouri
| | - Ying Yan
- 1 Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine , St. Louis, Missouri
| | - Amy M Moore
- 1 Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine , St. Louis, Missouri
| | - Alison K Snyder-Warwick
- 1 Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine , St. Louis, Missouri
| | - Sheila A Stewart
- 2 Division of Cell Biology and Physiology, Washington University , St. Louis, Missouri
| | - Susan E Mackinnon
- 1 Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine , St. Louis, Missouri
| | - Matthew D Wood
- 1 Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine , St. Louis, Missouri
| |
Collapse
|
176
|
Maeda T, Yoshizawa S, Hirayama T, Saga T, Tateda K, Urita Y. Neurosyphilis Mimicking Ramsay Hunt Syndrome. J NIPPON MED SCH 2016; 82:254-6. [PMID: 26568393 DOI: 10.1272/jnms.82.254] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A 36-year-old man presented with facial nerve palsy, hearing loss, vertigo and headache. He was initially diagnosed with Ramsay Hunt syndrome and treated with a systemic steroid and valaciclovir; however, his symptoms deteriorated. Serum rapid plasma reagin (RPR) and treponema pallidum hemagglutination tests were positive. Cerebrospinal fluid analysis revealed an elevated white blood cell count and positive RPR, confirming the diagnosis of neurosyphilis. Penicillin G (PCG) was administered, and his facial nerve function and headache improved. However, left-side hearing loss worsened temporarily, which was assumed to be a Jarisch-Herxheimer reaction. Betamethasone was administered along with PCG, and he recovered completely.
Collapse
Affiliation(s)
- Tadashi Maeda
- Department of General Medicine and Emergency Care, Toho University School of Medicine
| | | | | | | | | | | |
Collapse
|
177
|
Scheib JL, Höke A. An attenuated immune response by Schwann cells and macrophages inhibits nerve regeneration in aged rats. Neurobiol Aging 2016; 45:1-9. [PMID: 27459920 DOI: 10.1016/j.neurobiolaging.2016.05.004] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 05/03/2016] [Accepted: 05/04/2016] [Indexed: 12/21/2022]
Abstract
Although peripheral nerves are capable of regeneration, advanced age decreases the potential for functional recovery after injury. The cellular mechanisms for this are not currently understood. Here, we performed sciatic nerve grafting with young (2 months old) and aged (18 months old) Brown-Norway male rats, in which 1 cm nerve grafts from young or aged rats were sutured into nerves of young or aged rats. Axons were allowed to regenerate until the nerve grafts and distal nerves were harvested at 1, 3, and 7 days and 2 and 6 weeks. At 6 weeks, our data suggested that young nerve grafts supported regeneration better than aged nerve grafts. In addition, myelin debris clearance was inhibited in young nerves when grafted into aged rats, but clearance was faster when aged nerves were grafted into young rats. Further analysis revealed that aged macrophages have delayed migration into injured nerve, and macrophages and Schwann cells from aged rats were less phagocytic for myelin debris in vitro. To understand these impairments, expression levels of pro- and anti-inflammatory cytokines were analyzed at 1 day after injury. Based on these levels, there was not a clear polarization to either an M1 or M2 phenotype; however, expression levels of IL-6, IL-10, CCL2 (MCP1), and Arg-1 were decreased in aged nerves. Taken together, both macrophages and Schwann cells had attenuated responses to nerve injury in aged rats, leading to inefficient clearance of debris and impaired axonal regeneration.
Collapse
Affiliation(s)
- Jami L Scheib
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Ahmet Höke
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
178
|
He Z, Jin Y. Intrinsic Control of Axon Regeneration. Neuron 2016; 90:437-51. [DOI: 10.1016/j.neuron.2016.04.022] [Citation(s) in RCA: 421] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 03/10/2016] [Accepted: 04/13/2016] [Indexed: 01/12/2023]
|
179
|
Davidge KM, Clarke HM, Borschel GH. Nerve Transfers in Birth Related Brachial Plexus Injuries: Where Do We Stand? Hand Clin 2016; 32:175-90. [PMID: 27094890 DOI: 10.1016/j.hcl.2015.12.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
This article reviews the assessment and management of obstetrical brachial plexus palsy. The potential role of distal nerve transfers in the treatment of infants with Erb's palsy is discussed. Current evidence for motor outcomes after traditional reconstruction via interpositional nerve grafting and extraplexal nerve transfers is reviewed and compared with the recent literature on intraplexal distal nerve transfers in obstetrical brachial plexus injury.
Collapse
Affiliation(s)
- Kristen M Davidge
- Division of Plastic & Reconstructive Surgery, The Hospital for Sick Children, 555 University Avenue, Toronto, Ontario M5G 1X8, Canada; Department of Surgery, University of Toronto, Toronto, Ontario, Canada.
| | - Howard M Clarke
- Division of Plastic & Reconstructive Surgery, The Hospital for Sick Children, 555 University Avenue, Toronto, Ontario M5G 1X8, Canada; Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Gregory H Borschel
- Division of Plastic & Reconstructive Surgery, The Hospital for Sick Children, 555 University Avenue, Toronto, Ontario M5G 1X8, Canada; Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
180
|
Focal release of neurotrophic factors by biodegradable microspheres enhance motor and sensory axonal regeneration in vitro and in vivo. Brain Res 2016; 1636:93-106. [DOI: 10.1016/j.brainres.2016.01.051] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 01/20/2016] [Accepted: 01/31/2016] [Indexed: 11/23/2022]
|
181
|
Geoffroy CG, Hilton BJ, Tetzlaff W, Zheng B. Evidence for an Age-Dependent Decline in Axon Regeneration in the Adult Mammalian Central Nervous System. Cell Rep 2016; 15:238-46. [PMID: 27050519 DOI: 10.1016/j.celrep.2016.03.028] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 01/28/2016] [Accepted: 03/07/2016] [Indexed: 10/22/2022] Open
Abstract
How aging impacts axon regeneration after CNS injury is not known. We assessed the impact of age on axon regeneration induced by Pten deletion in corticospinal and rubrospinal neurons, two neuronal populations with distinct innate regenerative abilities. As in young mice, Pten deletion in older mice remains effective in preventing axotomy-induced decline in neuron-intrinsic growth state, as assessed by mTOR activity, neuronal soma size, and axonal growth proximal to a spinal cord injury. However, axonal regeneration distal to injury is greatly diminished, accompanied by increased expression of astroglial and inflammatory markers at the injury site. Thus, the mammalian CNS undergoes an age-dependent decline in axon regeneration, as revealed when neuron-intrinsic growth state is elevated. These results have important implications for developing strategies to promote axonal repair after CNS injuries or diseases, which increasingly affect middle-aged to aging populations.
Collapse
Affiliation(s)
- Cédric G Geoffroy
- Department of Neurosciences, School of Medicine, University of California, San Diego, La Jolla, CA 92093-0691, USA
| | - Brett J Hilton
- International Collaboration on Repair Discoveries, The University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Department of Zoology, The University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Wolfram Tetzlaff
- International Collaboration on Repair Discoveries, The University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Department of Zoology, The University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Department of Surgery, The University of British Columbia, Vancouver, BC V6T 1Z4, Canada.
| | - Binhai Zheng
- Department of Neurosciences, School of Medicine, University of California, San Diego, La Jolla, CA 92093-0691, USA.
| |
Collapse
|
182
|
Krishnan VS, White Z, McMahon CD, Hodgetts SI, Fitzgerald M, Shavlakadze T, Harvey AR, Grounds MD. A Neurogenic Perspective of Sarcopenia: Time Course Study of Sciatic Nerves From Aging Mice. J Neuropathol Exp Neurol 2016; 75:464-78. [DOI: 10.1093/jnen/nlw019] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
|
183
|
Jessen KR, Mirsky R. The repair Schwann cell and its function in regenerating nerves. J Physiol 2016; 594:3521-31. [PMID: 26864683 PMCID: PMC4929314 DOI: 10.1113/jp270874] [Citation(s) in RCA: 798] [Impact Index Per Article: 88.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 11/28/2015] [Indexed: 01/05/2023] Open
Abstract
Nerve injury triggers the conversion of myelin and non‐myelin (Remak) Schwann cells to a cell phenotype specialized to promote repair. Distal to damage, these repair Schwann cells provide the necessary signals and spatial cues for the survival of injured neurons, axonal regeneration and target reinnervation. The conversion to repair Schwann cells involves de‐differentiation together with alternative differentiation, or activation, a combination that is typical of cell type conversions often referred to as (direct or lineage) reprogramming. Thus, injury‐induced Schwann cell reprogramming involves down‐regulation of myelin genes combined with activation of a set of repair‐supportive features, including up‐regulation of trophic factors, elevation of cytokines as part of the innate immune response, myelin clearance by activation of myelin autophagy in Schwann cells and macrophage recruitment, and the formation of regeneration tracks, Bungner's bands, for directing axons to their targets. This repair programme is controlled transcriptionally by mechanisms involving the transcription factor c‐Jun, which is rapidly up‐regulated in Schwann cells after injury. In the absence of c‐Jun, damage results in the formation of a dysfunctional repair cell, neuronal death and failure of functional recovery. c‐Jun, although not required for Schwann cell development, is therefore central to the reprogramming of myelin and non‐myelin (Remak) Schwann cells to repair cells after injury. In future, the signalling that specifies this cell requires further analysis so that pharmacological tools that boost and maintain the repair Schwann cell phenotype can be developed.
![]()
Collapse
Affiliation(s)
- K R Jessen
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - R Mirsky
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
| |
Collapse
|
184
|
Abstract
In response to peripheral nerve injury, the inflammatory response is almost entirely comprised of infiltrating macrophages. Macrophages are a highly plastic, heterogenic immune cell, playing an indispensable role in peripheral nerve injury, clearing debris and regulating the microenvironment to allow for efficient regeneration. There are several cells within the microenvironment that likely interact with macrophages to support their function – most notably the Schwann cell, the glial cell of the peripheral nervous system. Schwann cells express several ligands that are known to interact with receptors expressed by macrophages, yet the effects of Schwann cells in regulating macrophage phenotype remains largely unexplored. This review discusses macrophages in peripheral nerve injury and how Schwann cells may regulate their behavior.
Collapse
Affiliation(s)
- Jo Anne Stratton
- Department of Clinical Neurosciences, University of Calgary, Calgary, Canada; Hotchkiss Brain Institute, Calgary, Canada; Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, Canada
| | - Prajay T Shah
- Hotchkiss Brain Institute, Calgary, Canada; Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, Canada
| |
Collapse
|
185
|
Adult cell plasticity in vivo: de-differentiation and transdifferentiation are back in style. Nat Rev Mol Cell Biol 2016; 17:413-25. [PMID: 26979497 DOI: 10.1038/nrm.2016.24] [Citation(s) in RCA: 305] [Impact Index Per Article: 33.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Biologists have long been intrigued by the possibility that cells can change their identity, a phenomenon known as cellular plasticity. The discovery that terminally differentiated cells can be experimentally coaxed to become pluripotent has invigorated the field, and recent studies have demonstrated that changes in cell identity are not limited to the laboratory. Specifically, certain adult cells retain the capacity to de-differentiate or transdifferentiate under physiological conditions, as part of an organ's normal injury response. Recent studies have highlighted the extent to which cell plasticity contributes to tissue homeostasis, findings that have implications for cell-based therapy.
Collapse
|
186
|
Effects of High Glucose on Cell Viability and Differentiation in Primary Cultured Schwann Cells: Potential Role of ERK Signaling Pathway. Neurochem Res 2016; 41:1281-90. [PMID: 26915107 DOI: 10.1007/s11064-015-1824-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 12/08/2015] [Accepted: 12/30/2015] [Indexed: 12/13/2022]
Abstract
Diabetic peripheral neuropathy (DPN) is one of the most common complications of diabetes mellitus and hyperglycemia is considered to be the major factor in the development and progression of DPN. Because of the contribution of Schwann cells (SCs) to the pathology of DPN, we investigated the effects of high glucose on cell proliferation, apoptosis and differentiation in primary cultured SCs. Cell Counting Kit-8 (CCK-8) assay and Hoechst staining showed that high glucose inhibited SCs proliferation and increased apoptosis ratio in time and concentration dependent manner. Western blot and real-time quantitative PCR analysis revealed that the major myelin proteins and genes expressions including P0, MAG and Krox-20, were downregulated time dependently in SCs exposed to high glucose from 48 to 96 h. To further elucidate the underlying pathogenic mechanisms, we also explored the role of ERK signaling pathway in high glucose induced SC injury, which has been proved to drive demyelination of peripheral nerves. The western blot analysis showed that compared with control group phosphorylation level of ERK was increased by 14.3 % in SCs exposed to high glucose for 72 h (P < 0.01). Using immunocytochemistry analysis, we observed that the ERK specific inhibitor U0126 blocked the ERK activation induced by high glucose and reversed the inhibitory effect of high glucose on P0 expression. Taken together, these results suggest that high glucose can cause damage in primary cultured SCs and may exert the inhibitory effect on SC differentiation and myelination through ERK signaling activation.
Collapse
|
187
|
Birchmeier C, Bennett DLH. Neuregulin/ErbB Signaling in Developmental Myelin Formation and Nerve Repair. Curr Top Dev Biol 2016; 116:45-64. [PMID: 26970613 DOI: 10.1016/bs.ctdb.2015.11.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Myelin is essential for rapid and accurate conduction of electrical impulses by axons in the central and peripheral nervous system (PNS). Myelin is formed in the early postnatal period, and developmental myelination in the PNS depends on axonal signals provided by Nrg1/ErbB receptors. In addition, Nrg1 is required for effective nerve repair and remyelination in adulthood. We discuss here similarities and differences in Nrg1/ErbB functions in developmental myelination and remyelination after nerve injury.
Collapse
Affiliation(s)
- Carmen Birchmeier
- Developmental Biology, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.
| | - David L H Bennett
- The Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.
| |
Collapse
|
188
|
Affiliation(s)
- Ki H Ma
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA; Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - John Svaren
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA; Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA; Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
189
|
Pulley BR, Luo TD, Barnwell JC, Smith BP, Smith TL, Li Z. A chronically-denervated versus a freshly-harvested autograft for nerve repair in rats. HAND AND MICROSURGERY 2016; 5:124-129. [PMID: 30828670 DOI: 10.5455/handmicrosurg.215015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Objectives Autologous nerve grafting remains the gold standard for repair of peripheral nerve injuries. Its use, however, is limited by donor nerve availability and donor site morbidity. This is especially problematic after failure of an initial autograft that requires a repeat nerve graft, resulting in a second surgical site with associated morbidity. Based on the molecular differences in nerve degeneration in the proximal and distal segments after transection, we hypothesized that a chronically-denervated proximal stump may be viable for autologous nerve repair. Methods 20 Sprague-Dawley rats underwent right sciatic nerve excision and sural nerve transection. After 8 weeks, nerve repair was performed by harvesting the proximal segment of the sural nerve (n=10) or a fresh sural nerve (n=10) from the contralateral hind limb. Electrophysiological changes were analyzed to compare the fresh and denervated grafts. Results Electrophysiological testing demonstrated higher compound motor action potential in the denervated group compared to the fresh autograft group, however this difference was not statistically significant (p=0.117). Conclusion The proximal segment of a chronically-denervated sural nerve can be as effective as a fresh sural nerve for autologous repair of peripheral nerve injuries in a rodent model.
Collapse
Affiliation(s)
| | | | | | - Beth P Smith
- Wake Forest Baptist Medical Center, Winston-Salem, NC, USA
| | - Thomas L Smith
- Wake Forest Baptist Medical Center, Winston-Salem, NC, USA
| | - Zhongyu Li
- Wake Forest Baptist Medical Center, Winston-Salem, NC, USA
| |
Collapse
|
190
|
Role of macrophages in Wallerian degeneration and axonal regeneration after peripheral nerve injury. Acta Neuropathol 2015; 130:605-18. [PMID: 26419777 DOI: 10.1007/s00401-015-1482-4] [Citation(s) in RCA: 360] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 09/22/2015] [Accepted: 09/24/2015] [Indexed: 01/08/2023]
Abstract
The peripheral nervous system (PNS) has remarkable regenerative abilities after injury. Successful PNS regeneration relies on both injured axons and non-neuronal cells, including Schwann cells and immune cells. Macrophages are the most notable immune cells that play key roles in PNS injury and repair. Upon peripheral nerve injury, a large number of macrophages are accumulated at the injury sites, where they not only contribute to Wallerian degeneration, but also are educated by the local microenvironment and polarized to an anti-inflammatory phenotype (M2), thus contributing to axonal regeneration. Significant progress has been made in understanding how macrophages are educated and polarized in the injured microenvironment as well as how they contribute to axonal regeneration. Following the discussion on the main properties of macrophages and their phenotypes, in this review, we will summarize the current knowledge regarding the mechanisms of macrophage infiltration after PNS injury. Moreover, we will discuss the recent findings elucidating how macrophages are polarized to M2 phenotype in the injured PNS microenvironment, as well as the role and underlying mechanisms of macrophages in peripheral nerve injury, Wallerian degeneration and regeneration. Furthermore, we will highlight the potential application by targeting macrophages in treating peripheral nerve injury and peripheral neuropathies.
Collapse
|
191
|
Eggers R, Tannemaat MR, De Winter F, Malessy MJA, Verhaagen J. Clinical and neurobiological advances in promoting regeneration of the ventral root avulsion lesion. Eur J Neurosci 2015; 43:318-35. [PMID: 26415525 DOI: 10.1111/ejn.13089] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 08/31/2015] [Accepted: 09/23/2015] [Indexed: 12/27/2022]
Abstract
Root avulsions due to traction to the brachial plexus causes complete and permanent loss of function. Until fairly recent, such lesions were considered impossible to repair. Here we review clinical repair strategies and current progress in experimental ventral root avulsion lesions. The current gold standard in patients with a root avulsion is nerve transfer, whereas reimplantation of the avulsed root into the spinal cord has been performed in a limited number of cases. These neurosurgical repair strategies have significant benefit for the patient but functional recovery remains incomplete. Developing new ways to improve the functional outcome of neurosurgical repair is therefore essential. In the laboratory, the molecular and cellular changes following ventral root avulsion and the efficacy of intervention strategies have been studied at the level of spinal motoneurons, the ventral spinal root and peripheral nerve, and the skeletal muscle. We present an overview of cell-based pharmacological and neurotrophic factor treatment approaches that have been applied in combination with surgical reimplantation. These interventions all demonstrate neuroprotective effects on avulsed motoneurons, often accompanied with various degrees of axonal regeneration. However, effects on survival are usually transient and robust axon regeneration over long distances has as yet not been achieved. Key future areas of research include finding ways to further extend the post-lesion survival period of motoneurons, the identification of neuron-intrinsic factors which can promote persistent and long-distance axon regeneration, and finally prolonging the pro-regenerative state of Schwann cells in the distal nerve.
Collapse
Affiliation(s)
- Ruben Eggers
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA, Amsterdam, the Netherlands
| | - Martijn R Tannemaat
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA, Amsterdam, the Netherlands.,Department of Neurology, Leiden University Medical Center, Leiden, the Netherlands
| | - Fred De Winter
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA, Amsterdam, the Netherlands.,Department of Neurosurgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Martijn J A Malessy
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA, Amsterdam, the Netherlands.,Department of Neurosurgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Joost Verhaagen
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA, Amsterdam, the Netherlands.,Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognition research, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
192
|
Abstract
Schwann cells develop from the neural crest in a well-defined sequence of events. This involves the formation of the Schwann cell precursor and immature Schwann cells, followed by the generation of the myelin and nonmyelin (Remak) cells of mature nerves. This review describes the signals that control the embryonic phase of this process and the organogenesis of peripheral nerves. We also discuss the phenotypic plasticity retained by mature Schwann cells, and explain why this unusual feature is central to the striking regenerative potential of the peripheral nervous system (PNS).
Collapse
Affiliation(s)
- Kristján R Jessen
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, United Kingdom
| | - Rhona Mirsky
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, United Kingdom
| | - Alison C Lloyd
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, United Kingdom
| |
Collapse
|
193
|
Fingolimod induces the transition to a nerve regeneration promoting Schwann cell phenotype. Exp Neurol 2015; 271:25-35. [PMID: 25957629 DOI: 10.1016/j.expneurol.2015.05.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 04/29/2015] [Accepted: 05/01/2015] [Indexed: 11/23/2022]
Abstract
Successful regeneration of injured peripheral nerves is mainly attributed to the plastic behavior of Schwann cells. Upon loss of axons, these cells trans-differentiate into regeneration promoting repair cells which provide trophic support to regrowing axons. Among others, activation of cJun was revealed to be involved in this process, initiating the stereotypic pattern of Schwann cell phenotype alterations during Wallerian degeneration. Nevertheless, the ability of Schwann cells to adapt and therefore the nerve's potential to regenerate can be limited in particular after long term denervation or in neuropathies leading to incomplete regeneration only and thus emphasizing the need for novel therapeutic approaches. Here we stimulated primary neonatal and adult rat Schwann cells with Fingolimod/FTY720P and investigated its impact on the regeneration promoting phenotype. FTY720P activated a number of de-differentiation markers including cJun and interfered with maturation marker and myelin expression. Functionally, FTY720P treated Schwann cells upregulated growth factor expression and these cells enhanced dorsal root ganglion neurite outgrowth on inhibitory substrates. Our results therefore provide strong evidence that FTY720P application supports the generation of a repair promoting cellular phenotype and suggest that Fingolimod could be used as treatment for peripheral nerve injuries and diseases.
Collapse
|
194
|
Regeneration of sensory but not motor axons following visceral nerve injury. Exp Neurol 2015; 266:127-42. [DOI: 10.1016/j.expneurol.2015.02.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 02/13/2015] [Accepted: 02/17/2015] [Indexed: 11/18/2022]
|
195
|
Xiao Y, Faucherre A, Pola-Morell L, Heddleston JM, Liu TL, Chew TL, Sato F, Sehara-Fujisawa A, Kawakami K, López-Schier H. High-resolution live imaging reveals axon-glia interactions during peripheral nerve injury and repair in zebrafish. Dis Model Mech 2015; 8:553-64. [PMID: 26035865 PMCID: PMC4457030 DOI: 10.1242/dmm.018184] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 03/24/2015] [Indexed: 12/22/2022] Open
Abstract
Neural damage is a devastating outcome of physical trauma. The glia are one of the main effectors of neuronal repair in the nervous system, but the dynamic interactions between peripheral neurons and Schwann cells during injury and regeneration remain incompletely characterized. Here, we combine laser microsurgery, genetic analysis, high-resolution intravital imaging and lattice light-sheet microscopy to study the interaction between Schwann cells and sensory neurons in a zebrafish model of neurotrauma. We found that chronic denervation by neuronal ablation leads to Schwann-cell death, whereas acute denervation by axonal severing does not affect the overall complexity and architecture of the glia. Neuronal-circuit regeneration begins when Schwann cells extend bridging processes to close the injury gap. Regenerating axons grow faster and directionally after the physiological clearing of distal debris by the Schwann cells. This might facilitate circuit repair by ensuring that axons are guided through unoccupied spaces within bands of Büngner towards their original peripheral target. Accordingly, in the absence of Schwann cells, regenerating axons are misrouted, impairing the re-innervation of sensory organs. Our results indicate that regenerating axons use haptotaxis as a directional cue during the reconstitution of a neural circuit. These findings have implications for therapies aimed at neurorepair, which will benefit from preserving the architecture of the peripheral glia during periods of denervation. Summary: Schwann cells are important components of the peripheral glia. We use microsurgery and high-resolution live imaging to show how Schwann cells control the regeneration of a sensorineural circuit.
Collapse
Affiliation(s)
- Yan Xiao
- Research Unit Sensory Biology & Organogenesis, Helmholtz Zentrum München, 85764 Munich, Germany
| | - Adèle Faucherre
- Cell & Developmental Biology, Centre for Genomic Regulation, 08003 Barcelona, Spain
| | - Laura Pola-Morell
- Cell & Developmental Biology, Centre for Genomic Regulation, 08003 Barcelona, Spain
| | - John M Heddleston
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Tsung-Li Liu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Teng-Leong Chew
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Fuminori Sato
- Institute for Frontier Medical Sciences, Kyoto University, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Atsuko Sehara-Fujisawa
- Institute for Frontier Medical Sciences, Kyoto University, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Koichi Kawakami
- Division of Molecular and Developmental Biology, National Institute of Genetics, The Graduate University for Advanced Studies (Sokendai), 1111 Yata, Mishima, Shizuoka 411-8540, Japan Department of Genetics, The Graduate University for Advanced Studies (Sokendai), 1111 Yata, Mishima, Shizuoka 411-8540, Japan
| | - Hernán López-Schier
- Research Unit Sensory Biology & Organogenesis, Helmholtz Zentrum München, 85764 Munich, Germany Cell & Developmental Biology, Centre for Genomic Regulation, 08003 Barcelona, Spain
| |
Collapse
|
196
|
Grochmal J, Midha R. Recent advances in stem cell-mediated peripheral nerve repair. Cells Tissues Organs 2015; 200:13-22. [PMID: 25825283 DOI: 10.1159/000369450] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2014] [Indexed: 11/19/2022] Open
Abstract
A major advance in the field of peripheral nerve repair has been the advent of stem and progenitor cell use to supplement the regenerative environment in animal models of nerve injury. As Schwann cell replacements, stem cells may be even better suited to promoting regeneration in these scenarios. We review the recent literature detailing the search for the definitive Schwann cell replacement cell, including a look at genetic modification of transplanted cells for nerve injury repair.
Collapse
|
197
|
Hung HA, Sun G, Keles S, Svaren J. Dynamic regulation of Schwann cell enhancers after peripheral nerve injury. J Biol Chem 2015; 290:6937-50. [PMID: 25614629 PMCID: PMC4358118 DOI: 10.1074/jbc.m114.622878] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 12/31/2014] [Indexed: 12/20/2022] Open
Abstract
Myelination of the peripheral nervous system is required for axonal function and long term stability. After peripheral nerve injury, Schwann cells transition from axon myelination to a demyelinated state that supports neuronal survival and ultimately remyelination of axons. Reprogramming of gene expression patterns during development and injury responses is shaped by the actions of distal regulatory elements that integrate the actions of multiple transcription factors. We used ChIP-seq to measure changes in histone H3K27 acetylation, a mark of active enhancers, to identify enhancers in myelinating rat peripheral nerve and their dynamics after demyelinating nerve injury. Analysis of injury-induced enhancers identified enriched motifs for c-Jun, a transcription factor required for Schwann cells to support nerve regeneration. We identify a c-Jun-bound enhancer in the gene for Runx2, a transcription factor induced after nerve injury, and we show that Runx2 is required for activation of other induced genes. In contrast, enhancers that lose H3K27ac after nerve injury are enriched for binding sites of the Sox10 and early growth response 2 (Egr2/Krox20) transcription factors, which are critical determinants of Schwann cell differentiation. Egr2 expression is lost after nerve injury, and many Egr2-binding sites lose H3K27ac after nerve injury. However, the majority of Egr2-bound enhancers retain H3K27ac, indicating that other transcription factors maintain active enhancer status after nerve injury. The global epigenomic changes in H3K27ac deposition pinpoint dynamic changes in enhancers that mediate the effects of transcription factors that control Schwann cell myelination and peripheral nervous system responses to nerve injury.
Collapse
Affiliation(s)
- Holly A Hung
- From the Waisman Center, Cellular and Molecular Pathology Graduate Program, and
| | - Guannan Sun
- Departments of Biostatistics and Medical Informatics and
| | - Sunduz Keles
- Departments of Biostatistics and Medical Informatics and
| | - John Svaren
- From the Waisman Center, Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin 53705
| |
Collapse
|
198
|
Visualizing peripheral nerve regeneration by whole mount staining. PLoS One 2015; 10:e0119168. [PMID: 25738874 PMCID: PMC4349735 DOI: 10.1371/journal.pone.0119168] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 01/10/2015] [Indexed: 12/28/2022] Open
Abstract
Peripheral nerve trauma triggers a well characterised sequence of events both proximal and distal to the site of injury. Axons distal to the injury degenerate, Schwann cells convert to a repair supportive phenotype and macrophages enter the nerve to clear myelin and axonal debris. Following these events, axons must regrow through the distal part of the nerve, re-innervate and finally are re-myelinated by Schwann cells. For nerve crush injuries (axonotmesis), in which the integrity of the nerve is maintained, repair may be relatively effective whereas for nerve transection (neurotmesis) repair will likely be very poor as few axons may be able to cross between the two parts of the severed nerve, across the newly generated nerve bridge, to enter the distal stump and regenerate. Analysing axon growth and the cell-cell interactions that occur following both nerve crush and cut injuries has largely been carried out by staining sections of nerve tissue, but this has the obvious disadvantage that it is not possible to follow the paths of regenerating axons in three dimensions within the nerve trunk or nerve bridge. To try and solve this problem, we describe the development and use of a novel whole mount staining protocol that allows the analysis of axonal regeneration, Schwann cell-axon interaction and re-vascularisation of the repairing nerve following nerve cut and crush injuries.
Collapse
|
199
|
Morrison BM, Tsingalia A, Vidensky S, Lee Y, Jin L, Farah MH, Lengacher S, Magistretti PJ, Pellerin L, Rothstein JD. Deficiency in monocarboxylate transporter 1 (MCT1) in mice delays regeneration of peripheral nerves following sciatic nerve crush. Exp Neurol 2014; 263:325-38. [PMID: 25447940 DOI: 10.1016/j.expneurol.2014.10.018] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 10/16/2014] [Accepted: 10/22/2014] [Indexed: 12/20/2022]
Abstract
Peripheral nerve regeneration following injury occurs spontaneously, but many of the processes require metabolic energy. The mechanism of energy supply to axons has not previously been determined. In the central nervous system, monocarboxylate transporter 1 (MCT1), expressed in oligodendroglia, is critical for supplying lactate or other energy metabolites to axons. In the current study, MCT1 is shown to localize within the peripheral nervous system to perineurial cells, dorsal root ganglion neurons, and Schwann cells by MCT1 immunofluorescence in wild-type mice and tdTomato fluorescence in MCT1 BAC reporter mice. To investigate whether MCT1 is necessary for peripheral nerve regeneration, sciatic nerves of MCT1 heterozygous null mice are crushed and peripheral nerve regeneration was quantified electrophysiologically and anatomically. Compound muscle action potential (CMAP) recovery is delayed from a median of 21 days in wild-type mice to greater than 38 days in MCT1 heterozygote null mice. In fact, half of the MCT1 heterozygote null mice have no recovery of CMAP at 42 days, while all of the wild-type mice recovered. In addition, muscle fibers remain 40% more atrophic and neuromuscular junctions 40% more denervated at 42 days post-crush in the MCT1 heterozygote null mice than wild-type mice. The delay in nerve regeneration is not only in motor axons, as the number of regenerated axons in the sural sensory nerve of MCT1 heterozygote null mice at 4 weeks and tibial mixed sensory and motor nerve at 3 weeks is also significantly reduced compared to wild-type mice. This delay in regeneration may be partly due to failed Schwann cell function, as there is reduced early phagocytosis of myelin debris and remyelination of axon segments. These data for the first time demonstrate that MCT1 is critical for regeneration of both sensory and motor axons in mice following sciatic nerve crush.
Collapse
Affiliation(s)
- Brett M Morrison
- Department of Neurology, School of Medicine, The Johns Hopkins University, 855 North Wolfe Street, Baltimore, MD 21205, USA.
| | - Akivaga Tsingalia
- Department of Neurology, School of Medicine, The Johns Hopkins University, 855 North Wolfe Street, Baltimore, MD 21205, USA.
| | - Svetlana Vidensky
- Department of Neurology, School of Medicine, The Johns Hopkins University, 855 North Wolfe Street, Baltimore, MD 21205, USA; Brain Science Institute, School of Medicine, The Johns Hopkins University, 855 North Wolfe Street, Baltimore, MD 21205, USA.
| | - Youngjin Lee
- Department of Neurology, School of Medicine, The Johns Hopkins University, 855 North Wolfe Street, Baltimore, MD 21205, USA; Brain Science Institute, School of Medicine, The Johns Hopkins University, 855 North Wolfe Street, Baltimore, MD 21205, USA.
| | - Lin Jin
- Department of Neurology, School of Medicine, The Johns Hopkins University, 855 North Wolfe Street, Baltimore, MD 21205, USA; Brain Science Institute, School of Medicine, The Johns Hopkins University, 855 North Wolfe Street, Baltimore, MD 21205, USA.
| | - Mohamed H Farah
- Department of Neurology, School of Medicine, The Johns Hopkins University, 855 North Wolfe Street, Baltimore, MD 21205, USA.
| | - Sylvain Lengacher
- Laboratory of Neuroenergetics and Cellular Dynamics, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland.
| | - Pierre J Magistretti
- Division of Biological and Environmental Sciences and Engineering, KAUST, Thuwal, Saudi Arabia; Brain Mind Institute, Ecole Polytechnique Federale de Lausanne, SV2511, Station 19, CH-1015 Lausanne, Switzerland.
| | - Luc Pellerin
- Department of Fundamental Neurosciences, University of Lausanne, 7 Rue du Bugnon, 1005 Lausanne, Switzerland.
| | - Jeffrey D Rothstein
- Department of Neurology, School of Medicine, The Johns Hopkins University, 855 North Wolfe Street, Baltimore, MD 21205, USA; Brain Science Institute, School of Medicine, The Johns Hopkins University, 855 North Wolfe Street, Baltimore, MD 21205, USA.
| |
Collapse
|