1
|
Trivedi AH, Wang VZ, McClain EJ, Vyas PS, Swink IR, Snell ED, Cheng BC, DeMeo PJ. The Categorization of Perinatal Derivatives for Orthopedic Applications. Biomedicines 2024; 12:1544. [PMID: 39062117 PMCID: PMC11274709 DOI: 10.3390/biomedicines12071544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/01/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Musculoskeletal (MSK) pathology encompasses an array of conditions that can cause anything from mild discomfort to permanent injury. Their prevalence and impact on disability have sparked interest in more effective treatments, particularly within orthopedics. As a result, the human placenta has come into focus within regenerative medicine as a perinatal derivative (PnD). These biologics are sourced from components of the placenta, each possessing a unique composition of collagens, proteins, and factors believed to aid in healing and regeneration. This review aims to explore the current literature on PnD biologics and their potential benefits for treating various MSK pathologies. We delve into different types of PnDs and their healing effects on muscles, tendons, bones, cartilage, ligaments, and nerves. Our discussions highlight the crucial role of immune modulation in the healing process for each condition. PnDs have been observed to influence the balance between anti- and pro-inflammatory factors and, in some cases, act as biologic scaffolds for tissue growth. Additionally, we assess the range of PnDs available, while also addressing gaps in our understanding, particularly regarding biologic processing methods. Although certain PnD biologics have varying levels of support in orthopedic literature, further clinical investigations are necessary to fully evaluate their impact on human patients.
Collapse
Affiliation(s)
- Amol H. Trivedi
- Orthopaedic Institute, Allegheny General Hospital, Allegheny Health Network, Pittsburgh, PA 15212, USA; (A.H.T.); (V.Z.W.); (E.J.M.IV); (P.S.V.); (I.R.S.); (E.D.S.); (P.J.D.)
- Drexel University College of Medicine, Drexel University, University City Campus, Philadelphia, PA 19104, USA
| | - Vicki Z. Wang
- Orthopaedic Institute, Allegheny General Hospital, Allegheny Health Network, Pittsburgh, PA 15212, USA; (A.H.T.); (V.Z.W.); (E.J.M.IV); (P.S.V.); (I.R.S.); (E.D.S.); (P.J.D.)
| | - Edward J. McClain
- Orthopaedic Institute, Allegheny General Hospital, Allegheny Health Network, Pittsburgh, PA 15212, USA; (A.H.T.); (V.Z.W.); (E.J.M.IV); (P.S.V.); (I.R.S.); (E.D.S.); (P.J.D.)
| | - Praveer S. Vyas
- Orthopaedic Institute, Allegheny General Hospital, Allegheny Health Network, Pittsburgh, PA 15212, USA; (A.H.T.); (V.Z.W.); (E.J.M.IV); (P.S.V.); (I.R.S.); (E.D.S.); (P.J.D.)
| | - Isaac R. Swink
- Orthopaedic Institute, Allegheny General Hospital, Allegheny Health Network, Pittsburgh, PA 15212, USA; (A.H.T.); (V.Z.W.); (E.J.M.IV); (P.S.V.); (I.R.S.); (E.D.S.); (P.J.D.)
| | - Edward D. Snell
- Orthopaedic Institute, Allegheny General Hospital, Allegheny Health Network, Pittsburgh, PA 15212, USA; (A.H.T.); (V.Z.W.); (E.J.M.IV); (P.S.V.); (I.R.S.); (E.D.S.); (P.J.D.)
| | - Boyle C. Cheng
- Orthopaedic Institute, Allegheny General Hospital, Allegheny Health Network, Pittsburgh, PA 15212, USA; (A.H.T.); (V.Z.W.); (E.J.M.IV); (P.S.V.); (I.R.S.); (E.D.S.); (P.J.D.)
| | - Patrick J. DeMeo
- Orthopaedic Institute, Allegheny General Hospital, Allegheny Health Network, Pittsburgh, PA 15212, USA; (A.H.T.); (V.Z.W.); (E.J.M.IV); (P.S.V.); (I.R.S.); (E.D.S.); (P.J.D.)
| |
Collapse
|
2
|
Mutschler C, Fazal SV, Schumacher N, Loreto A, Coleman MP, Arthur-Farraj P. Schwann cells are axo-protective after injury irrespective of myelination status in mouse Schwann cell-neuron cocultures. J Cell Sci 2023; 136:jcs261557. [PMID: 37642648 PMCID: PMC10546878 DOI: 10.1242/jcs.261557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 08/18/2023] [Indexed: 08/31/2023] Open
Abstract
Myelinating Schwann cell (SC)-dorsal root ganglion (DRG) neuron cocultures are an important technique for understanding cell-cell signalling and interactions during peripheral nervous system (PNS) myelination, injury, and regeneration. Although methods using rat SCs and neurons or mouse DRG explants are commonplace, there are no established protocols for compartmentalised myelinating cocultures with dissociated mouse cells. There consequently is a need for a coculture protocol that allows separate genetic manipulation of mouse SCs or neurons, or use of cells from different transgenic animals to complement in vivo mouse experiments. However, inducing myelination of dissociated mouse SCs in culture is challenging. Here, we describe a new method to coculture dissociated mouse SCs and DRG neurons in microfluidic chambers and induce robust myelination. Cocultures can be axotomised to study injury and used for drug treatments, and cells can be lentivirally transduced for live imaging. We used this model to investigate axon degeneration after traumatic axotomy and find that SCs, irrespective of myelination status, are axo-protective. At later timepoints after injury, live imaging of cocultures shows that SCs break up, ingest and clear axonal debris.
Collapse
Affiliation(s)
- Clara Mutschler
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0PY, UK
| | - Shaline V. Fazal
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0PY, UK
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK
| | - Nathalie Schumacher
- Laboratory of Nervous System Disorders and Therapies, GIGA Neurosciences, University of Liège, 4000 Liège, Belgium
| | - Andrea Loreto
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0PY, UK
| | - Michael P. Coleman
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0PY, UK
| | - Peter Arthur-Farraj
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0PY, UK
| |
Collapse
|
3
|
Weiß EM, Geldermann M, Martini R, Klein D. Macrophages influence Schwann cell myelin autophagy after nerve injury and in a model of Charcot-Marie-Tooth disease. J Peripher Nerv Syst 2023; 28:341-350. [PMID: 37209383 DOI: 10.1111/jns.12561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/04/2023] [Accepted: 05/15/2023] [Indexed: 05/22/2023]
Abstract
BACKGROUND AND AIMS The complex cellular and molecular interactions between Schwann cells (SCs) and macrophages during Wallerian degeneration are a prerequisite to allow rapid uptake and degradation of myelin debris and axonal regeneration after peripheral nerve injury. In contrast, in non-injured nerves of Charcot-Marie-Tooth 1 neuropathies, aberrant macrophage activation by SCs carrying myelin gene defects is a disease amplifier that drives nerve damage and subsequent functional decline. Consequently, targeting nerve macrophages might be a translatable treatment strategy to mitigate disease outcome in CMT1 patients. Indeed, in previous approaches, macrophage targeting alleviated the axonopathy and promoted sprouting of damaged fibers. Surprisingly, this was still accompanied by robust myelinopathy in a model for CMT1X, suggesting additional cellular mechanisms of myelin degradation in mutant peripheral nerves. We here investigated the possibility of an increased SC-related myelin autophagy upon macrophage targeting in Cx32def mice. METHODS Combining ex vivo and in vivo approaches, macrophages were targeted by PLX5622 treatment. SC autophagy was investigated by immunohistochemical and electron microscopical techniques. RESULTS We demonstrate a robust upregulation of markers for SC autophagy after injury and in genetically-mediated neuropathy when nerve macrophages are pharmacologically depleted. Corroborating these findings, we provide ultrastructural evidence for increased SC myelin autophagy upon treatment in vivo. INTERPRETATION These findings reveal a novel communication and interaction between SCs and macrophages. This identification of alternative pathways of myelin degradation may have important implications for a better understanding of therapeutic mechanisms of pharmacological macrophage targeting in diseased peripheral nerves.
Collapse
Affiliation(s)
- Eva Maria Weiß
- Department of Neurology, Developmental Neurobiology, University Hospital Würzburg, Würzburg, Germany
| | - Miriam Geldermann
- Department of Neurology, Developmental Neurobiology, University Hospital Würzburg, Würzburg, Germany
| | - Rudolf Martini
- Department of Neurology, Developmental Neurobiology, University Hospital Würzburg, Würzburg, Germany
| | - Dennis Klein
- Department of Neurology, Developmental Neurobiology, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
4
|
Negro S, Pirazzini M, Rigoni M. Models and methods to study Schwann cells. J Anat 2022; 241:1235-1258. [PMID: 34988978 PMCID: PMC9558160 DOI: 10.1111/joa.13606] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 11/26/2021] [Accepted: 11/29/2021] [Indexed: 12/22/2022] Open
Abstract
Schwann cells (SCs) are fundamental components of the peripheral nervous system (PNS) of all vertebrates and play essential roles in development, maintenance, function, and regeneration of peripheral nerves. There are distinct populations of SCs including: (1) myelinating SCs that ensheath axons by a specialized plasma membrane, called myelin, which enhances the conduction of electric impulses; (2) non-myelinating SCs, including Remak SCs, which wrap bundles of multiple axons of small caliber, and perysinaptic SCs (PSCs), associated with motor axon terminals at the neuromuscular junction (NMJ). All types of SCs contribute to PNS regeneration through striking morphological and functional changes in response to nerve injury, are affected in peripheral neuropathies and show abnormalities and a diminished plasticity during aging. Therefore, methodological approaches to study and manipulate SCs in physiological and pathophysiological conditions are crucial to expand the present knowledge on SC biology and to devise new therapeutic strategies to counteract neurodegenerative conditions and age-derived denervation. We present here an updated overview of traditional and emerging methodologies for the study of SCs for scientists approaching this research field.
Collapse
Affiliation(s)
- Samuele Negro
- Department of Biomedical SciencesUniversity of PaduaPaduaItaly
| | - Marco Pirazzini
- Department of Biomedical SciencesUniversity of PaduaPaduaItaly
- CIR‐MyoCentro Interdipartimentale di Ricerca di MiologiaUniversity of PaduaPadovaItaly
| | - Michela Rigoni
- Department of Biomedical SciencesUniversity of PaduaPaduaItaly
- CIR‐MyoCentro Interdipartimentale di Ricerca di MiologiaUniversity of PaduaPadovaItaly
| |
Collapse
|
5
|
Dervan A, Franchi A, Almeida-Gonzalez FR, Dowling JK, Kwakyi OB, McCoy CE, O’Brien FJ, Hibbitts A. Biomaterial and Therapeutic Approaches for the Manipulation of Macrophage Phenotype in Peripheral and Central Nerve Repair. Pharmaceutics 2021; 13:2161. [PMID: 34959446 PMCID: PMC8706646 DOI: 10.3390/pharmaceutics13122161] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/23/2021] [Accepted: 11/25/2021] [Indexed: 12/18/2022] Open
Abstract
Injury to the peripheral or central nervous systems often results in extensive loss of motor and sensory function that can greatly diminish quality of life. In both cases, macrophage infiltration into the injury site plays an integral role in the host tissue inflammatory response. In particular, the temporally related transition of macrophage phenotype between the M1/M2 inflammatory/repair states is critical for successful tissue repair. In recent years, biomaterial implants have emerged as a novel approach to bridge lesion sites and provide a growth-inductive environment for regenerating axons. This has more recently seen these two areas of research increasingly intersecting in the creation of 'immune-modulatory' biomaterials. These synthetic or naturally derived materials are fabricated to drive macrophages towards a pro-repair phenotype. This review considers the macrophage-mediated inflammatory events that occur following nervous tissue injury and outlines the latest developments in biomaterial-based strategies to influence macrophage phenotype and enhance repair.
Collapse
Affiliation(s)
- Adrian Dervan
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland; (A.D.); (A.F.); (F.R.A.-G.); (F.J.O.)
- Trinity Centre for Bioengineering, Trinity College Dublin, D02 R590 Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, D02 YN77 Dublin, Ireland
| | - Antonio Franchi
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland; (A.D.); (A.F.); (F.R.A.-G.); (F.J.O.)
- Trinity Centre for Bioengineering, Trinity College Dublin, D02 R590 Dublin, Ireland
| | - Francisco R. Almeida-Gonzalez
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland; (A.D.); (A.F.); (F.R.A.-G.); (F.J.O.)
- Trinity Centre for Bioengineering, Trinity College Dublin, D02 R590 Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, D02 YN77 Dublin, Ireland
| | - Jennifer K. Dowling
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland; (J.K.D.); (O.B.K.); (C.E.M.)
- FutureNeuro SFI Research Centre, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland
| | - Ohemaa B. Kwakyi
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland; (J.K.D.); (O.B.K.); (C.E.M.)
- School of Medicine, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland
| | - Claire E. McCoy
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland; (J.K.D.); (O.B.K.); (C.E.M.)
- FutureNeuro SFI Research Centre, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland
| | - Fergal J. O’Brien
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland; (A.D.); (A.F.); (F.R.A.-G.); (F.J.O.)
- Trinity Centre for Bioengineering, Trinity College Dublin, D02 R590 Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, D02 YN77 Dublin, Ireland
| | - Alan Hibbitts
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland; (A.D.); (A.F.); (F.R.A.-G.); (F.J.O.)
- Trinity Centre for Bioengineering, Trinity College Dublin, D02 R590 Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, D02 YN77 Dublin, Ireland
| |
Collapse
|
6
|
Kasper M, Ellenbogen B, Hardy R, Cydis M, Mojica-Santiago J, Afridi A, Spearman BS, Singh I, Kuliasha CA, Atkinson E, Otto KJ, Judy JW, Rinaldi-Ramos C, Schmidt CE. Development of a magnetically aligned regenerative tissue-engineered electronic nerve interface for peripheral nerve applications. Biomaterials 2021; 279:121212. [PMID: 34717196 PMCID: PMC9036633 DOI: 10.1016/j.biomaterials.2021.121212] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 10/11/2021] [Accepted: 10/20/2021] [Indexed: 01/18/2023]
Abstract
Peripheral nerve injuries can be debilitating to motor and sensory function, with severe cases often resulting in complete limb amputation. Over the past two decades, prosthetic limb technology has rapidly advanced to provide users with crude motor control of up to 20° of freedom; however, the nerve-interfacing technology required to provide high movement selectivity has not progressed at the same rate. The work presented here focuses on the development of a magnetically aligned regenerative tissue-engineered electronic nerve interface (MARTEENI) that combines polyimide "threads" encapsulated within a magnetically aligned hydrogel scaffold. The technology exploits tissue-engineered strategies to address concerns over traditional peripheral nerve interfaces including poor axonal sampling through the nerve and rigid substrates. A magnetically templated hydrogel is used to physically support the polyimide threads while also promoting regeneration in close proximity to the electrode sites on the polyimide. This work demonstrates the utility of magnetic templating for use in tuning the mechanical properties of hydrogel scaffolds to match the stiffness of native nerve tissue while providing an aligned substrate for Schwann cell migration in vitro. MARTEENI devices were fabricated and implanted within a 5-mm-long rat sciatic-nerve transection model to assess regeneration at 6 and 12 weeks. MARTEENI devices do not disrupt tissue remodeling and show axon densities equivalent to fresh tissue controls around the polyimide substrates. Devices are observed to have attenuated foreign-body responses around the polyimide threads. It is expected that future studies with functional MARTEENI devices will be able to record and stimulate single axons with high selectivity and low stimulation regimes.
Collapse
Affiliation(s)
- Mary Kasper
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Dr. JG56, P.O. Box 116131, Gainesville, FL, 32611, USA
| | - Bret Ellenbogen
- Department of Chemistry, University of Florida, P.O. Box 117200, Gainesville, FL, 32611, USA
| | - Ryan Hardy
- Department of Materials Science and Engineering, University of Florida, 549 Gale Lemerand Dr., P.O. Box 116400, Gainesville, FL, 32611, USA
| | - Madison Cydis
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Dr. JG56, P.O. Box 116131, Gainesville, FL, 32611, USA
| | - Jorge Mojica-Santiago
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Dr. JG56, P.O. Box 116131, Gainesville, FL, 32611, USA
| | - Abdullah Afridi
- Department of Chemistry, University of Florida, P.O. Box 117200, Gainesville, FL, 32611, USA
| | - Benjamin S Spearman
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Dr. JG56, P.O. Box 116131, Gainesville, FL, 32611, USA
| | - Ishita Singh
- Department of Chemical Engineering, University of Florida, 1030 Center Dr., P.O. Box 116005, Gainesville, FL, 32611, USA
| | - Cary A Kuliasha
- Department of Electrical and Computer Engineering, University of Florida, 968 Center Dr., Gainesville, FL, 32611, USA
| | - Eric Atkinson
- Department of Neuroscience, 1149 Newell Dr. L1-100, P.O. Box 100244, University of Florida, Gainesville, FL, 32610, USA
| | - Kevin J Otto
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Dr. JG56, P.O. Box 116131, Gainesville, FL, 32611, USA; Department of Materials Science and Engineering, University of Florida, 549 Gale Lemerand Dr., P.O. Box 116400, Gainesville, FL, 32611, USA; Department of Electrical and Computer Engineering, University of Florida, 968 Center Dr., Gainesville, FL, 32611, USA; Department of Neuroscience, 1149 Newell Dr. L1-100, P.O. Box 100244, University of Florida, Gainesville, FL, 32610, USA; Department of Neurology, 1149 Newell Dr. L3-100, P.O. Box 100236, University of Florida, Gainesville, FL, 32610, USA
| | - Jack W Judy
- Department of Electrical and Computer Engineering, University of Florida, 968 Center Dr., Gainesville, FL, 32611, USA
| | - Carlos Rinaldi-Ramos
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Dr. JG56, P.O. Box 116131, Gainesville, FL, 32611, USA; Department of Chemistry, University of Florida, P.O. Box 117200, Gainesville, FL, 32611, USA
| | - Christine E Schmidt
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Dr. JG56, P.O. Box 116131, Gainesville, FL, 32611, USA.
| |
Collapse
|
7
|
Khan A, Diaz A, Brooks AE, Burks SS, Athauda G, Wood P, Lee YS, Silvera R, Donaldson M, Pressman Y, Anderson KD, Bunge MB, Pearse DD, Dietrich WD, Guest JD, Levi AD. Scalable culture techniques to generate large numbers of purified human Schwann cells for clinical trials in human spinal cord and peripheral nerve injuries. J Neurosurg Spine 2021; 36:135-144. [PMID: 34479193 DOI: 10.3171/2020.11.spine201433] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 11/24/2020] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Schwann cells (SCs) have been shown to play an essential role in axon regeneration in both peripheral nerve injuries (PNIs) and spinal cord injuries (SCIs). The transplantation of SCs as an adjunctive therapy is currently under investigation in human clinical trials due to their regenerative capacity. Therefore, a reliable method for procuring large quantities of SCs from peripheral nerves is necessary. This paper presents a well-developed, validated, and optimized manufacturing protocol for clinical-grade SCs that are compliant with Current Good Manufacturing Practices (CGMPs). METHODS The authors evaluated the SC culture manufacturing data from 18 clinical trial participants who were recruited for autologous SC transplantation due to subacute SCI (n = 7), chronic SCI (n = 8), or PNIs (n = 3). To initiate autologous SC cultures, a mean nerve length of 11.8 ± 3.7 cm was harvested either from the sural nerve alone (n = 17) or with the sciatic nerve (n = 1). The nerves were digested with enzymes and SCs were isolated and further expanded in multiple passages to meet the dose requirements for transplantation. RESULTS An average yield of 87.2 ± 89.2 million cells at P2 and 150.9 ± 129.9 million cells at P3 with high viability and purity was produced. Cell counts and rates of expansion increased with each subsequent passage from P0 to P3, with the largest rate of expansion between P2 and P3. Larger harvest nerve lengths correlated significantly with greater yields at P0 and P1 (p < 0.05). In addition, a viability and purity above 90% was sustained throughout all passages in nearly all cell products. CONCLUSIONS This study presents reliable CGMP-compliant manufacturing methods for autologous SC products that are suitable for regenerative treatment of patients with SCI, PNI, or other conditions.
Collapse
Affiliation(s)
- Aisha Khan
- 1The Miami Project to Cure Paralysis.,3Interdisciplinary Stem Cell Institute, and Departments of
| | - Anthony Diaz
- 1The Miami Project to Cure Paralysis.,2Department of Neurological Surgery
| | - Adriana E Brooks
- 1The Miami Project to Cure Paralysis.,3Interdisciplinary Stem Cell Institute, and Departments of
| | - S Shelby Burks
- 1The Miami Project to Cure Paralysis.,2Department of Neurological Surgery
| | - Gagani Athauda
- 7Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida; and
| | - Patrick Wood
- 1The Miami Project to Cure Paralysis.,2Department of Neurological Surgery
| | - Yee-Shuan Lee
- 3Interdisciplinary Stem Cell Institute, and Departments of
| | - Risset Silvera
- 1The Miami Project to Cure Paralysis.,3Interdisciplinary Stem Cell Institute, and Departments of
| | - Maxwell Donaldson
- 1The Miami Project to Cure Paralysis.,3Interdisciplinary Stem Cell Institute, and Departments of
| | | | - Kim D Anderson
- 8Department of Physical Medicine and Rehabilitation, MetroHealth Medical Center, Institute for Functional Restoration, Case Western Reserve University School, Cleveland, Ohio
| | - Mary Bartlett Bunge
- 1The Miami Project to Cure Paralysis.,2Department of Neurological Surgery.,4Cell Biology and
| | - Damien D Pearse
- 1The Miami Project to Cure Paralysis.,2Department of Neurological Surgery.,3Interdisciplinary Stem Cell Institute, and Departments of.,6Bruce W. Carter Department of Veterans Affairs, Veterans Affairs Medical Center, Miami
| | - W Dalton Dietrich
- 1The Miami Project to Cure Paralysis.,2Department of Neurological Surgery.,4Cell Biology and.,5Neurology, University of Miami Miller School of Medicine, Miami
| | - James D Guest
- 1The Miami Project to Cure Paralysis.,2Department of Neurological Surgery
| | - Allan D Levi
- 1The Miami Project to Cure Paralysis.,2Department of Neurological Surgery
| |
Collapse
|
8
|
Nazareth L, St John J, Murtaza M, Ekberg J. Phagocytosis by Peripheral Glia: Importance for Nervous System Functions and Implications in Injury and Disease. Front Cell Dev Biol 2021; 9:660259. [PMID: 33898462 PMCID: PMC8060502 DOI: 10.3389/fcell.2021.660259] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/17/2021] [Indexed: 12/30/2022] Open
Abstract
The central nervous system (CNS) has very limited capacity to regenerate after traumatic injury or disease. In contrast, the peripheral nervous system (PNS) has far greater capacity for regeneration. This difference can be partly attributed to variances in glial-mediated functions, such as axon guidance, structural support, secretion of growth factors and phagocytic activity. Due to their growth-promoting characteristic, transplantation of PNS glia has been trialed for neural repair. After peripheral nerve injuries, Schwann cells (SCs, the main PNS glia) phagocytose myelin debris and attract macrophages to the injury site to aid in debris clearance. One peripheral nerve, the olfactory nerve, is unique in that it continuously regenerates throughout life. The olfactory nerve glia, olfactory ensheathing cells (OECs), are the primary phagocytes within this nerve, continuously clearing axonal debris arising from the normal regeneration of the nerve and after injury. In contrast to SCs, OECs do not appear to attract macrophages. SCs and OECs also respond to and phagocytose bacteria, a function likely critical for tackling microbial invasion of the CNS via peripheral nerves. However, phagocytosis is not always effective; inflammation, aging and/or genetic factors may contribute to compromised phagocytic activity. Here, we highlight the diverse roles of SCs and OECs with the focus on their phagocytic activity under physiological and pathological conditions. We also explore why understanding the contribution of peripheral glia phagocytosis may provide us with translational strategies for achieving axonal regeneration of the injured nervous system and potentially for the treatment of certain neurological diseases.
Collapse
Affiliation(s)
- Lynn Nazareth
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia
| | - James St John
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia.,Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
| | - Mariyam Murtaza
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia.,Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
| | - Jenny Ekberg
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia.,Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
| |
Collapse
|
9
|
Fissel JA, Farah MH. The influence of BACE1 on macrophage recruitment and activity in the injured peripheral nerve. J Neuroinflammation 2021; 18:71. [PMID: 33722254 PMCID: PMC7962400 DOI: 10.1186/s12974-021-02121-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 02/26/2021] [Indexed: 01/13/2023] Open
Abstract
Following peripheral nerve injury, multiple cell types, including axons, Schwann cells, and macrophages, coordinate to promote nerve regeneration. However, this capacity for repair is limited, particularly in older populations, and current treatments are insufficient. A critical component of the regeneration response is the network of cell-to-cell signaling in the injured nerve microenvironment. Sheddases are expressed in the peripheral nerve and play a role in the regulation if this cell-to-cell signaling through cleavage of transmembrane proteins, enabling the regulation of multiple pathways through cis- and trans-cellular regulatory mechanisms. Enhanced axonal regeneration has been observed in mice with deletion of the sheddase beta-secretase (BACE1), a transmembrane aspartyl protease that has been studied in the context of Alzheimer’s disease. BACE1 knockout (KO) mice display enhanced macrophage recruitment and activity following nerve injury, although it is unclear whether this plays a role in driving the enhanced axonal regeneration. Further, it is unknown by what mechanism(s) BACE1 increases macrophage recruitment and activity. BACE1 has many substrates, several of which are known to have immunomodulatory activity. This review will discuss current knowledge of the role of BACE1 and other sheddases in peripheral nerve regeneration and outline known immunomodulatory BACE1 substrates and what potential roles they could play in peripheral nerve regeneration. Currently, the literature suggests that BACE1 and substrates that are expressed by neurons and Schwann cells are likely to be more important for this process than those expressed by macrophages. More broadly, BACE1 may play a role as an effector of immunomodulation beyond the peripheral nerve.
Collapse
Affiliation(s)
- John A Fissel
- Department of Neurology, Johns Hopkins University School of Medicine, The John G. Rangos Sr. Building, Room 239, 855 N. Wolfe Street, Baltimore, MD, 21205, USA
| | - Mohamed H Farah
- Department of Neurology, Johns Hopkins University School of Medicine, The John G. Rangos Sr. Building, Room 239, 855 N. Wolfe Street, Baltimore, MD, 21205, USA.
| |
Collapse
|
10
|
Lüningschrör P, Slotta C, Heimann P, Briese M, Weikert UM, Massih B, Appenzeller S, Sendtner M, Kaltschmidt C, Kaltschmidt B. Absence of Plekhg5 Results in Myelin Infoldings Corresponding to an Impaired Schwann Cell Autophagy, and a Reduced T-Cell Infiltration Into Peripheral Nerves. Front Cell Neurosci 2020; 14:185. [PMID: 32733205 PMCID: PMC7358705 DOI: 10.3389/fncel.2020.00185] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 05/28/2020] [Indexed: 12/14/2022] Open
Abstract
Inflammation and dysregulation of the immune system are hallmarks of several neurodegenerative diseases. An activated immune response is considered to be the cause of myelin breakdown in demyelinating disorders. In the peripheral nervous system (PNS), myelin can be degraded in an autophagy-dependent manner directly by Schwann cells or by macrophages, which are modulated by T-lymphocytes. Here, we show that the NF-κB activator Pleckstrin homology containing family member 5 (Plekhg5) is involved in the regulation of both Schwann cell autophagy and recruitment of T-lymphocytes in peripheral nerves during motoneuron disease. Plekhg5-deficient mice show defective axon/Schwann cell units characterized by myelin infoldings in peripheral nerves. Even at late stages, Plekhg5-deficient mice do not show any signs of demyelination and inflammation. Using RNAseq, we identified a transcriptional signature for an impaired immune response in sciatic nerves, which manifested in a reduced number of CD4+ and CD8+ T-cells. These findings identify Plekhg5 as a promising target to impede myelin breakdown in demyelinating PNS disorders.
Collapse
Affiliation(s)
- Patrick Lüningschrör
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Carsten Slotta
- Department of Cell Biology, University of Bielefeld, Bielefeld, Germany.,Molecular Neurobiology, University of Bielefeld, Bielefeld, Germany
| | - Peter Heimann
- Department of Cell Biology, University of Bielefeld, Bielefeld, Germany
| | - Michael Briese
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Ulrich M Weikert
- Department of Cell Biology, University of Bielefeld, Bielefeld, Germany
| | - Bita Massih
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Silke Appenzeller
- Core Unit Systems Medicine, University of Wuerzburg, Wuerzburg, Germany.,Comprehensive Cancer Center Mainfranken, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Michael Sendtner
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, Wuerzburg, Germany
| | | | - Barbara Kaltschmidt
- Department of Cell Biology, University of Bielefeld, Bielefeld, Germany.,Molecular Neurobiology, University of Bielefeld, Bielefeld, Germany
| |
Collapse
|
11
|
Shea GK, Tai EW, Leung KH, Mung AK, Li MT, Tsui AY, Tam AK, Shum DK, Chan Y. Juxtacrine signalling via Notch and ErbB receptors in the switch to fate commitment of bone marrow‐derived Schwann cells. Eur J Neurosci 2020; 52:3306-3321. [DOI: 10.1111/ejn.14837] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 05/03/2020] [Accepted: 05/18/2020] [Indexed: 01/09/2023]
Affiliation(s)
- Graham Ka‐Hon Shea
- Department of Orthopaedics and Traumatology Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong Hong Kong
| | - Evelyn Wing‐Yin Tai
- Department of Orthopaedics and Traumatology Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong Hong Kong
- Li Ka Shing Faculty of Medicine School of Biomedical Sciences The University of Hong Kong Hong Kong Hong Kong
| | - Katherine Ho‐Yan Leung
- Li Ka Shing Faculty of Medicine School of Biomedical Sciences The University of Hong Kong Hong Kong Hong Kong
| | - Alan Kwan‐Long Mung
- Department of Orthopaedics and Traumatology Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong Hong Kong
- Li Ka Shing Faculty of Medicine School of Biomedical Sciences The University of Hong Kong Hong Kong Hong Kong
| | - Maximilian Tak‐Sui Li
- Li Ka Shing Faculty of Medicine School of Biomedical Sciences The University of Hong Kong Hong Kong Hong Kong
- Research Centre of Heart, Brain, Hormone & Healthy Aging The University of Hong Kong Hong Kong Hong Kong
| | - Alex Yat‐Ping Tsui
- Li Ka Shing Faculty of Medicine School of Biomedical Sciences The University of Hong Kong Hong Kong Hong Kong
- Research Centre of Heart, Brain, Hormone & Healthy Aging The University of Hong Kong Hong Kong Hong Kong
| | - Anthony Kin‐Wai Tam
- Li Ka Shing Faculty of Medicine School of Biomedical Sciences The University of Hong Kong Hong Kong Hong Kong
- Research Centre of Heart, Brain, Hormone & Healthy Aging The University of Hong Kong Hong Kong Hong Kong
| | - Daisy Kwok‐Yan Shum
- Li Ka Shing Faculty of Medicine School of Biomedical Sciences The University of Hong Kong Hong Kong Hong Kong
- Research Centre of Heart, Brain, Hormone & Healthy Aging The University of Hong Kong Hong Kong Hong Kong
- State Key Laboratory of Brain and Cognitive Science The University of Hong Kong Hong Kong Hong Kong
| | - Ying‐Shing Chan
- Li Ka Shing Faculty of Medicine School of Biomedical Sciences The University of Hong Kong Hong Kong Hong Kong
- Research Centre of Heart, Brain, Hormone & Healthy Aging The University of Hong Kong Hong Kong Hong Kong
- State Key Laboratory of Brain and Cognitive Science The University of Hong Kong Hong Kong Hong Kong
| |
Collapse
|
12
|
Abstract
Toxic peripheral neuropathies are an important form of acquired polyneuropathy produced by a variety of xenobiotics and different exposure scenarios. Delineating the mechanisms of neurotoxicants and determining the degenerative biological pathways triggered by peripheral neurotoxicants will facilitate the development of sensitive and specific biochemical-based methods for identifying neurotoxicants, designing therapeutic interventions, and developing structure-activity relationships for predicting potential neurotoxicants. This review presents an overview of the general concepts of toxic peripheral neuropathies with the goal of providing insight into why certain agents target the peripheral nervous system and produce their associated lesions. Experimental data and the main hypotheses for the mechanisms of selected agents that produce neuronopathies, axonopathies, or myelinopathies including covalent or noncovalent modifications, compromised energy or protein biosynthesis, and oxidative injury and disruption of ionic gradients across membranes are presented. The relevance of signaling between the main components of peripheral nerve, that is, glia, neuronal perikaryon, and axon, as a target for neurotoxicants and the contribution of active programmed degenerative pathways to the lesions observed in toxic peripheral neuropathies is also discussed.
Collapse
|
13
|
Jung K, Park JH, Kim SY, Jeon NL, Cho SR, Hyung S. Optogenetic stimulation promotes Schwann cell proliferation, differentiation, and myelination in vitro. Sci Rep 2019; 9:3487. [PMID: 30837563 PMCID: PMC6401157 DOI: 10.1038/s41598-019-40173-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 01/24/2019] [Indexed: 11/18/2022] Open
Abstract
Schwann cells (SCs) constitute a crucial element of the peripheral nervous system, by structurally supporting the formation of myelin and conveying vital trophic factors to the nervous system. However, the functions of SCs in developmental and regenerative stages remain unclear. Here, we investigated how optogenetic stimulation (OS) of SCs regulates their development. In SC monoculture, OS substantially enhanced SC proliferation and the number of BrdU+-S100ß+-SCs over time. In addition, OS also markedly promoted the expression of both Krox20 and myelin basic protein (MBP) in SC culture medium containing dBcAMP/NRG1, which induced differentiation. We found that the effects of OS are dependent on the intracellular Ca2+ level. OS induces elevated intracellular Ca2+ levels through the T-type voltage-gated calcium channel (VGCC) and mobilization of Ca2+ from both inositol 1,4,5-trisphosphate (IP3)-sensitive stores and caffeine/ryanodine-sensitive stores. Furthermore, we confirmed that OS significantly increased expression levels of both Krox20 and MBP in SC-motor neuron (MN) coculture, which was notably prevented by pharmacological intervention with Ca2+. Taken together, our results demonstrate that OS of SCs increases the intracellular Ca2+ level and can regulate proliferation, differentiation, and myelination, suggesting that OS of SCs may offer a new approach to the treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Kyuhwan Jung
- Graduate Program of Nano Science and Technology, Graduate School of Yonsei University, Seoul, Korea
| | - Ji Hye Park
- Gradaute Program of Translational Neuroscience, Institute for Clinical Neurobiology, University of Wuerzburg, Wuerzburg, Germany
| | - Sung-Yon Kim
- Department of Biophysics and Chemical Biology, Seoul National University, Seoul, South Korea
| | - Noo Li Jeon
- Multiscale Mechanical Design School of Mechanical and Aerospace Engineering Institute of Advanced Machinery and Design, Seoul National University, Seoul, Korea. .,Institute of Bioengineering, Seoul National University, Seoul, Korea.
| | - Sung-Rae Cho
- Graduate Program of Nano Science and Technology, Graduate School of Yonsei University, Seoul, Korea. .,Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Korea.
| | - Sujin Hyung
- Multiscale Mechanical Design School of Mechanical and Aerospace Engineering Institute of Advanced Machinery and Design, Seoul National University, Seoul, Korea. .,BK21 Plus Transformative Training Program for Creative Mechanical and Aerospace Engineers, Seoul National University, Seoul, Korea. .,Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, USA.
| |
Collapse
|
14
|
Stierli S, Napoli I, White IJ, Cattin AL, Monteza Cabrejos A, Garcia Calavia N, Malong L, Ribeiro S, Nihouarn J, Williams R, Young KM, Richardson WD, Lloyd AC. The regulation of the homeostasis and regeneration of peripheral nerve is distinct from the CNS and independent of a stem cell population. Development 2018; 145:dev170316. [PMID: 30413560 PMCID: PMC6307893 DOI: 10.1242/dev.170316] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 10/30/2018] [Indexed: 12/22/2022]
Abstract
Peripheral nerves are highly regenerative, in contrast to the poor regenerative capabilities of the central nervous system (CNS). Here, we show that adult peripheral nerve is a more quiescent tissue than the CNS, yet all cell types within a peripheral nerve proliferate efficiently following injury. Moreover, whereas oligodendrocytes are produced throughout life from a precursor pool, we find that the corresponding cell of the peripheral nervous system, the myelinating Schwann cell (mSC), does not turn over in the adult. However, following injury, all mSCs can dedifferentiate to the proliferating progenitor-like Schwann cells (SCs) that orchestrate the regenerative response. Lineage analysis shows that these newly migratory, progenitor-like cells redifferentiate to form new tissue at the injury site and maintain their lineage, but can switch to become a non-myelinating SC. In contrast, increased plasticity is observed during tumourigenesis. These findings show that peripheral nerves have a distinct mechanism for maintaining homeostasis and can regenerate without the need for an additional stem cell population.This article has an associated 'The people behind the papers' interview.
Collapse
Affiliation(s)
- Salome Stierli
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Ilaria Napoli
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Ian J White
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Anne-Laure Cattin
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Anthony Monteza Cabrejos
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Noelia Garcia Calavia
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Liza Malong
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Sara Ribeiro
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Julie Nihouarn
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Richard Williams
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Kaylene M Young
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - William D Richardson
- Wolfson Institute for Biomedical Research, University College London (UCL), 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
|
15
|
Park HT, Kim JK, Tricaud N. The conceptual introduction of the “demyelinating Schwann cell” in peripheral demyelinating neuropathies. Glia 2018; 67:571-581. [DOI: 10.1002/glia.23509] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 06/25/2018] [Accepted: 06/27/2018] [Indexed: 11/07/2022]
Affiliation(s)
- Hwan Tae Park
- Department of Molecular Neuroscience; Peripheral Neuropathy Research Center, College of Medicine, Dong-A University; Busan South Korea
| | - Jong Kuk Kim
- Department of Neurology; Peripheral Neuropathy Research Center, College of Medicine, Dong-A University; Busan South Korea
| | - Nicolas Tricaud
- INSERM U1051, Institut des Neurosciences de Montpellier (INM); Université de Montpellier; Montpellier France
| |
Collapse
|
16
|
Zhao L, Yi S. Transcriptional landscape of alternative splicing during peripheral nerve injury. J Cell Physiol 2018; 234:6876-6885. [PMID: 30362529 DOI: 10.1002/jcp.27446] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 08/27/2018] [Indexed: 12/27/2022]
Abstract
Alternative splicing (AS) regulates a variety of biological activities in numerous tissues and organs, including the nervous system. However, the existence and specific roles of AS events during peripheral nerve repair and regeneration remain largely undetermined. In the current study, by mapping splice-crossing sequence reads, we identified AS events and relevant spliced genes in rat sciatic nerve stumps following sciatic nerve crush. AS-related genes at 1, 4, 7, and 14 days post nerve crush were compared with those at 0 day to discover alternatively spliced genes induced by sciatic nerve crush. These injury-induced alternatively spliced genes were then categorized to diseases and biological functions, genetic networks, and canonical signaling pathways. Bioinformatic analysis indicated that these alternatively spliced genes were mainly correlated to immune response, cellular growth, and cellular function maintenance. Our study elucidated AS events following peripheral nerve injury and might help deepen our understanding of the molecular mechanisms underlying peripheral nerve regeneration.
Collapse
Affiliation(s)
- Lili Zhao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing, China
| | - Sheng Yi
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
17
|
Ying Z, Pan C, Shao T, Liu L, Li L, Guo D, Zhang S, Yuan T, Cao R, Jiang Z, Chen S, Wang F, Wang X. Mixed Lineage Kinase Domain-like Protein MLKL Breaks Down Myelin following Nerve Injury. Mol Cell 2018; 72:457-468.e5. [PMID: 30344099 DOI: 10.1016/j.molcel.2018.09.011] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 07/09/2018] [Accepted: 09/11/2018] [Indexed: 11/26/2022]
Abstract
Successful regeneration of severed peripheral nerves requires the breakdown and subsequent clearance of myelin, tightly packed membrane sheaths of Schwann cells that protect nerve fibers and harbor nerve growth-inhibitory proteins. How Schwann cells initiate myelin breakdown in response to injury is still largely unknown. Here we report that, following sciatic nerve injury, MLKL, a pseudokinase known to rupture cell membranes during necroptotic cell death, is induced and targets the myelin sheath membrane of Schwann cells to promote myelin breakdown. The function of MLKL in disrupting myelin sheaths requires injury-induced phosphorylation of serine 441, an activation signal distinct from the necroptosis-inducing phosphorylation by RIP3 kinase. Mice with Mlkl specifically knocked out in Schwann cells showed delayed myelin sheath breakdown. Lack of MLKL reduced nerve regeneration following injury, whereas overexpression of MLKL accelerated myelin breakdown and promoted the regeneration of axons.
Collapse
Affiliation(s)
- Zhengxin Ying
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Chenjie Pan
- School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Tianyu Shao
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Liqing Liu
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Lin Li
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Dejia Guo
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Sitao Zhang
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Tianyi Yuan
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Ran Cao
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Zhaodi Jiang
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - She Chen
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Fengchao Wang
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Xiaodong Wang
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China.
| |
Collapse
|
18
|
Gribble KD, Walker LJ, Saint-Amant L, Kuwada JY, Granato M. The synaptic receptor Lrp4 promotes peripheral nerve regeneration. Nat Commun 2018; 9:2389. [PMID: 29921864 PMCID: PMC6008306 DOI: 10.1038/s41467-018-04806-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 05/21/2018] [Indexed: 01/05/2023] Open
Abstract
Early during PNS regeneration, regenerating axons emerge from the proximal nerve stump, yet whether they extend simultaneously or whether pioneering axons establish a path for follower axons remains unknown. Moreover, the molecular mechanisms underlying robust regeneration are incompletely understood. Using live imaging, we demonstrate that in zebrafish pioneering axons establish a regenerative path for follower axons. We find this process requires the synaptic receptor lrp4, and in lrp4 mutants pioneers are unaffected while follower axons frequently stall at the injury gap, providing evidence for molecular diversity between pioneering and follower axons in regeneration. We demonstrate that Lrp4 promotes regeneration through an axon extrinsic mechanism and independent of membrane anchoring and MuSK co-receptor signaling essential for synaptic development. Finally, we show that Lrp4 coordinates the realignment of denervated Schwann cells with regenerating axons, consistent with a model by which Lrp4 is repurposed to promote sustained peripheral nerve regeneration via axon-glia interactions.
Collapse
Affiliation(s)
- Katherine D Gribble
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Lauren J Walker
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Louis Saint-Amant
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - John Y Kuwada
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Michael Granato
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
19
|
Jang SY, Yoon BA, Shin YK, Yun SH, Jo YR, Choi YY, Ahn M, Shin T, Park JI, Kim JK, Park HT. Schwann cell dedifferentiation-associated demyelination leads to exocytotic myelin clearance in inflammatory segmental demyelination. Glia 2017; 65:1848-1862. [DOI: 10.1002/glia.23200] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 07/01/2017] [Accepted: 07/23/2017] [Indexed: 02/03/2023]
Affiliation(s)
- So Young Jang
- Department of Physiology, Peripheral Neuropathy Research Center, College of Medicine; Dong-A University; Busan 49201 Republic of Korea
| | - Byeol-A Yoon
- Department of Neurology, Peripheral Neuropathy Research Center, College of Medicine; Dong-A University; Busan 49201 Republic of Korea
| | - Yoon Kyung Shin
- Department of Physiology, Peripheral Neuropathy Research Center, College of Medicine; Dong-A University; Busan 49201 Republic of Korea
| | - Seoug Hoon Yun
- Department of Biochemistry, Peripheral Neuropathy Research Center, College of Medicine; Dong-A University; Busan 49201 Republic of Korea
| | - Young Rae Jo
- Department of Neurology, Peripheral Neuropathy Research Center, College of Medicine; Dong-A University; Busan 49201 Republic of Korea
| | - Yun Young Choi
- Department of Biochemistry, Peripheral Neuropathy Research Center, College of Medicine; Dong-A University; Busan 49201 Republic of Korea
| | - Meejung Ahn
- Department of Veterinary Anatomy, College of Veterinary Medicine and Veterinary Medical Research Institute; JeJu National University; Jeju 63243 Republic of Korea
| | - Taekyun Shin
- Department of Veterinary Anatomy, College of Veterinary Medicine and Veterinary Medical Research Institute; JeJu National University; Jeju 63243 Republic of Korea
| | - Joo In Park
- Department of Biochemistry, Peripheral Neuropathy Research Center, College of Medicine; Dong-A University; Busan 49201 Republic of Korea
| | - Jong Kuk Kim
- Department of Neurology, Peripheral Neuropathy Research Center, College of Medicine; Dong-A University; Busan 49201 Republic of Korea
| | - Hwan Tae Park
- Department of Physiology, Peripheral Neuropathy Research Center, College of Medicine; Dong-A University; Busan 49201 Republic of Korea
| |
Collapse
|
20
|
Hsieh CH, Rau CS, Kuo PJ, Liu SH, Wu CJ, Lu TH, Wu YC, Lin CW. Knockout of toll-like receptor impairs nerve regeneration after a crush injury. Oncotarget 2017; 8:80741-80756. [PMID: 29113341 PMCID: PMC5655236 DOI: 10.18632/oncotarget.20206] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 07/12/2017] [Indexed: 01/01/2023] Open
Abstract
Background Toll-like receptors (TLRs) are involved in the initiation of Schwann cell activation and subsequent recruitment of macrophages for clearance of degenerated myelin and neuronal debris after nerve injury. The present study was designed to investigate the regenerative outcome and expression of myelination-related factors in Tlr-knockout mice following a sciatic nerve crush injury. Materials and methods A standard sciatic nerve crush injury, induced by applying constant pressure to the nerve with a No. 5 jeweler's forceps for 30 s, was performed in C57BL/6, Tlr2−/−, Tlr3−/−, Tlr4−/−, Tlr5−/−, and Tlr7−/− mice. Quantitative histomorphometric analysis of toluidine blue-stained nerve specimens and walking track analysis were performed to evaluate nerve regeneration outcomes. PCR Arrays were used to detect the expression of neurogenesis-related genes of dorsal root ganglia as well as of myelination-related genes of the distal nerve segments. Results Worse nerve regeneration after nerve crush injury was found in all Tlr-knockout mice than in C57BL/6 mice. Delayed expression of myelin genes and a different expression pattern of myelination-related neurotrophin genes and transcription factors were found in Tlr-knockout mice in comparison to C57BL/6 mice. In these TLR-mediated pathways, insulin-like growth factor 2 and brain-derived neurotrophic factor, as well as early growth response 2 and N-myc downstream-regulated gene 1, were significantly decreased in the early and late stages, respectively, of nerve regeneration after a crush injury. Conclusions Knockout of Tlr genes decreases the expression of myelination-related factors and impairs nerve regeneration after a sciatic nerve crush injury.
Collapse
Affiliation(s)
- Ching-Hua Hsieh
- Department of Plastic and Reconstructive Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.,Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Cheng-Shyuan Rau
- Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Pao-Jen Kuo
- Department of Plastic and Reconstructive Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Shu-Hsuan Liu
- Faculty of Health Sciences, McMaster University, Hamilton, Canada
| | - Chia-Jung Wu
- Department of Plastic and Reconstructive Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Tsu-Hsiang Lu
- Department of Plastic and Reconstructive Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yi-Chan Wu
- Department of Plastic and Reconstructive Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chia-Wei Lin
- Department of Plastic and Reconstructive Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| |
Collapse
|
21
|
Bunge MB, Monje PV, Khan A, Wood PM. From transplanting Schwann cells in experimental rat spinal cord injury to their transplantation into human injured spinal cord in clinical trials. PROGRESS IN BRAIN RESEARCH 2017; 231:107-133. [PMID: 28554394 DOI: 10.1016/bs.pbr.2016.12.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Among the potential therapies designed to repair the injured spinal cord is cell transplantation, notably the use of autologous adult human Schwann cells (SCs). Here, we detail some of the critical research accomplished over the last four decades to establish a foundation that enables these cells to be tested in clinical trials. New culture systems allowed novel information to be gained about SCs, including discovering ways to stimulate their proliferation to acquire adequately large numbers for transplantation into the injured human spinal cord. Transplantation of rat SCs into rat models of spinal cord injury has demonstrated that SCs promote repair of injured spinal cord. Additional work required to gain approval from the Food and Drug Administration for the first SC trial in the Miami Project is disclosed. This trial and a second one now underway are described.
Collapse
Affiliation(s)
- Mary B Bunge
- The Miami Project to Cure Paralysis, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States; Department of Cell Biology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States; Department of Neurological Surgery, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States.
| | - Paula V Monje
- The Miami Project to Cure Paralysis, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States; Department of Neurological Surgery, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States
| | - Aisha Khan
- The Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States
| | - Patrick M Wood
- The Miami Project to Cure Paralysis, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States; Department of Neurological Surgery, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
22
|
Soto J, Monje PV. Axon contact-driven Schwann cell dedifferentiation. Glia 2017; 65:864-882. [PMID: 28233923 DOI: 10.1002/glia.23131] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 01/24/2017] [Accepted: 02/02/2017] [Indexed: 12/26/2022]
Abstract
Mature Schwann cells (SCs) retain dedifferentiation potential throughout adulthood. Still, how dedifferentiation occurs remains uncertain. Results from a variety of cell-based assays using in vitro cultured cAMP-differentiated and myelinating SCs revealed the existence of a novel dedifferentiating activity expressed on the surface of dorsal root ganglion (DRG) axons. This activity had the capacity to prevent SC differentiation and elicit dedifferentiation through direct SC-axon contact. Evidence is provided showing that a rapid loss of myelinating SC markers concomitant to proliferation occurred even in the presence of elevated cAMP, a signal that is required to drive and maintain a differentiated state. The dedifferentiating activity was a membrane-bound protein found exclusively in DRG neurons, as judged by its subcellular partitioning, sensitivity to proteolytic degradation and cell-type specificity, and remained active even after disruption of cellular organization. It differed from the membrane-anchored neuregulin-1 isoforms that are responsible for axon contact-induced SC proliferation and exerted its action independently of mitogenic signaling emanating from receptor tyrosine kinases and mitogen-activated protein kinases such as ERK and JNK. Interestingly, dedifferentiation occurred without concomitant changes in the expression of Krox-20, a transcriptional enhancer of myelination, and c-Jun, an inhibitor of myelination. In sum, our data indicated the existence of cell surface axon-derived signals that override pro-differentiating cues, drive dedifferentiation and allow SCs to proliferate in response to axonal mitogens. This axonal signal may negatively regulate myelination at the onset or reversal of the differentiated state. GLIA 2017;65:851-863.
Collapse
Affiliation(s)
- Jennifer Soto
- Miami Project to Cure Paralysis and Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, 33136
| | - Paula V Monje
- Miami Project to Cure Paralysis and Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, 33136
| |
Collapse
|
23
|
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: 118] [Impact Index Per Article: 14.8] [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
|
24
|
Weiss T, Taschner-Mandl S, Bileck A, Slany A, Kromp F, Rifatbegovic F, Frech C, Windhager R, Kitzinger H, Tzou CH, Ambros PF, Gerner C, Ambros IM. Proteomics and transcriptomics of peripheral nerve tissue and cells unravel new aspects of the human Schwann cell repair phenotype. Glia 2016; 64:2133-2153. [DOI: 10.1002/glia.23045] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 07/21/2016] [Accepted: 07/26/2016] [Indexed: 11/09/2022]
Affiliation(s)
- Tamara Weiss
- Children's Cancer Research Institute; Vienna Austria
| | | | - Andrea Bileck
- Department of Analytical Chemistry; University of Vienna; Vienna Austria
| | - Astrid Slany
- Department of Analytical Chemistry; University of Vienna; Vienna Austria
| | - Florian Kromp
- Children's Cancer Research Institute; Vienna Austria
| | | | | | - Reinhard Windhager
- Department of Orthopedic Surgery; Medical University of Vienna; Vienna Austria
| | - Hugo Kitzinger
- Department of Plastic and Reconstructive Surgery; Medical University of Vienna; Vienna Austria
| | - Chieh-Han Tzou
- Department of Plastic and Reconstructive Surgery; Medical University of Vienna; Vienna Austria
| | - Peter F. Ambros
- Children's Cancer Research Institute; Vienna Austria
- Department of Pediatrics; Medical University of Vienna; Vienna Austria
| | - Christopher Gerner
- Department of Analytical Chemistry; University of Vienna; Vienna Austria
| | | |
Collapse
|
25
|
Xu C, Bai L, Chen Y, Fan C, Hu Z, Xu H, Jiang B. Effect of mutated defensin NP-1 on sciatic nerve regeneration after transection—A pivot study. Neurosci Lett 2016; 617:283-7. [DOI: 10.1016/j.neulet.2015.11.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 10/30/2015] [Accepted: 11/02/2015] [Indexed: 12/24/2022]
|
26
|
Gomez-Sanchez JA, Carty L, Iruarrizaga-Lejarreta M, Palomo-Irigoyen M, Varela-Rey M, Griffith M, Hantke J, Macias-Camara N, Azkargorta M, Aurrekoetxea I, De Juan VG, Jefferies HBJ, Aspichueta P, Elortza F, Aransay AM, Martínez-Chantar ML, Baas F, Mato JM, Mirsky R, Woodhoo A, Jessen KR. Schwann cell autophagy, myelinophagy, initiates myelin clearance from injured nerves. J Cell Biol 2015; 210:153-68. [PMID: 26150392 PMCID: PMC4494002 DOI: 10.1083/jcb.201503019] [Citation(s) in RCA: 316] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 06/03/2015] [Indexed: 02/07/2023] Open
Abstract
Although Schwann cell myelin breakdown is the universal outcome of a remarkably wide range of conditions that cause disease or injury to peripheral nerves, the cellular and molecular mechanisms that make Schwann cell-mediated myelin digestion possible have not been established. We report that Schwann cells degrade myelin after injury by a novel form of selective autophagy, myelinophagy. Autophagy was up-regulated by myelinating Schwann cells after nerve injury, myelin debris was present in autophagosomes, and pharmacological and genetic inhibition of autophagy impaired myelin clearance. Myelinophagy was positively regulated by the Schwann cell JNK/c-Jun pathway, a central regulator of the Schwann cell reprogramming induced by nerve injury. We also present evidence that myelinophagy is defective in the injured central nervous system. These results reveal an important role for inductive autophagy during Wallerian degeneration, and point to potential mechanistic targets for accelerating myelin clearance and improving demyelinating disease.
Collapse
Affiliation(s)
- Jose A Gomez-Sanchez
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, England, UK
| | - Lucy Carty
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, England, UK
| | - Marta Iruarrizaga-Lejarreta
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 48160 Derio, Bizkaia, Spain
| | - Marta Palomo-Irigoyen
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 48160 Derio, Bizkaia, Spain
| | - Marta Varela-Rey
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 48160 Derio, Bizkaia, Spain
| | - Megan Griffith
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, England, UK
| | - Janina Hantke
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, England, UK
| | - Nuria Macias-Camara
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 48160 Derio, Bizkaia, Spain
| | - Mikel Azkargorta
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 48160 Derio, Bizkaia, Spain ProteoRed-ISCIII
| | - Igor Aurrekoetxea
- Department of Physiology, Faculty of Medicine and Dentistry, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain Biocruces Health Research Institute, 48903 Barakaldo, Spain
| | - Virginia Gutiérrez De Juan
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 48160 Derio, Bizkaia, Spain
| | - Harold B J Jefferies
- The Francis Crick Institute, Lincoln's Inn Fields Laboratory, London WC2A 3LY, England, UK
| | - Patricia Aspichueta
- Department of Physiology, Faculty of Medicine and Dentistry, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain Biocruces Health Research Institute, 48903 Barakaldo, Spain
| | - Félix Elortza
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 48160 Derio, Bizkaia, Spain ProteoRed-ISCIII
| | - Ana M Aransay
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 48160 Derio, Bizkaia, Spain
| | - María L Martínez-Chantar
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 48160 Derio, Bizkaia, Spain Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU), E-48080 Bilbao, Spain
| | - Frank Baas
- Department of Genome Analysis, Academic Medical Centre, 1105 AZ Amsterdam, Netherlands
| | - José M Mato
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 48160 Derio, Bizkaia, Spain
| | - Rhona Mirsky
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, England, UK
| | - Ashwin Woodhoo
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 48160 Derio, Bizkaia, Spain Ikerbasque, Basque Foundation for Science, 48011 Bilbao, Spain
| | - Kristján R Jessen
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, England, UK
| |
Collapse
|
27
|
Mietto BS, Mostacada K, Martinez AMB. Neurotrauma and inflammation: CNS and PNS responses. Mediators Inflamm 2015; 2015:251204. [PMID: 25918475 PMCID: PMC4397002 DOI: 10.1155/2015/251204] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 02/24/2015] [Accepted: 03/09/2015] [Indexed: 01/09/2023] Open
Abstract
Traumatic injury to the central nervous system (CNS) or the peripheral nervous system (PNS) triggers a cascade of events which culminate in a robust inflammatory reaction. The role played by inflammation in the course of degeneration and regeneration is not completely elucidated. While, in peripheral nerves, the inflammatory response is assumed to be essential for normal progression of Wallerian degeneration and regeneration, CNS trauma inflammation is often associated with poor recovery. In this review, we discuss key mechanisms that trigger the inflammatory reaction after nervous system trauma, emphasizing how inflammations in both CNS and PNS differ from each other, in terms of magnitude, cell types involved, and effector molecules. Knowledge of the precise mechanisms that elicit and maintain inflammation after CNS and PNS tissue trauma and their effect on axon degeneration and regeneration is crucial for the identification of possible pharmacological drugs that can positively affect the tissue regenerative capacity.
Collapse
Affiliation(s)
- Bruno Siqueira Mietto
- Laboratório de Neurodegeneração e Reparo, Departamento de Patologia, Faculdade de Medicina, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, 21941-550 Rio de Janeiro, RJ, Brazil
| | - Klauss Mostacada
- Laboratório de Neurodegeneração e Reparo, Departamento de Patologia, Faculdade de Medicina, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, 21941-550 Rio de Janeiro, RJ, Brazil
| | - Ana Maria Blanco Martinez
- Laboratório de Neurodegeneração e Reparo, Departamento de Patologia, Faculdade de Medicina, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, 21941-550 Rio de Janeiro, RJ, Brazil
| |
Collapse
|
28
|
Chernov AV, Dolkas J, Hoang K, Angert M, Srikrishna G, Vogl T, Baranovskaya S, Strongin AY, Shubayev VI. The calcium-binding proteins S100A8 and S100A9 initiate the early inflammatory program in injured peripheral nerves. J Biol Chem 2015; 290:11771-84. [PMID: 25792748 DOI: 10.1074/jbc.m114.622316] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Indexed: 11/06/2022] Open
Abstract
To shed light on the early immune response processes in severed peripheral nerves, we performed genome-wide transcriptional profiling and bioinformatics analyses of the proximal (P, regenerating) and distal (D, degenerating) nerve stumps on day 1 in the sciatic nerve axotomy model in rats. Multiple cell death-related pathways were activated in the degenerating D stump, whereas activation of the cytoskeletal motility and gluconeogenesis/glycolysis pathways was most prominent in the P stump of the axotomized nerve. Our bioinformatics analyses also identified the specific immunomodulatory genes of the chemokine, IL, TNF, MHC, immunoglobulin-binding Fc receptor, calcium-binding S100, matrix metalloproteinase, tissue inhibitor of metalloproteinase, and ion channel families affected in both the P and D segments. S100a8 and S100a9 were the top up-regulated genes in both the P and D segments. Stimulation of cultured Schwann cells using the purified S100A8/A9 heterodimer recapitulated activation of the myeloid cell and phagocyte chemotactic genes and pathways, which we initially observed in injured nerves. S100A8/A9 heterodimer injection into the intact nerve stimulated macrophage infiltration. We conclude that, following peripheral nerve injury, an immediate acute immune response occurs both distal and proximal to the lesion site and that the rapid transcriptional activation of the S100a8 and S100a9 genes results in S100A8/A9 hetero- and homodimers, which stimulate the release of chemokines and cytokines by activated Schwann cells and generate the initial chemotactic gradient that guides the transmigration of hematogenous immune cells into the injured nerve.
Collapse
Affiliation(s)
- Andrei V Chernov
- From the Sanford-Burnham Medical Research Institute, La Jolla, California 92037
| | - Jennifer Dolkas
- the Department of Anesthesiology, University of California, San Diego, La Jolla, California 92093, the Veterans Affairs San Diego Healthcare System, La Jolla, California 92037
| | - Khang Hoang
- the Department of Anesthesiology, University of California, San Diego, La Jolla, California 92093, the Veterans Affairs San Diego Healthcare System, La Jolla, California 92037
| | - Mila Angert
- the Department of Anesthesiology, University of California, San Diego, La Jolla, California 92093, the Veterans Affairs San Diego Healthcare System, La Jolla, California 92037
| | - Geetha Srikrishna
- From the Sanford-Burnham Medical Research Institute, La Jolla, California 92037
| | - Thomas Vogl
- the Institute of Immunology, University of Münster, D-48149 Münster, Germany, and
| | | | - Alex Y Strongin
- From the Sanford-Burnham Medical Research Institute, La Jolla, California 92037
| | - Veronica I Shubayev
- the Department of Anesthesiology, University of California, San Diego, La Jolla, California 92093, the Veterans Affairs San Diego Healthcare System, La Jolla, California 92037,
| |
Collapse
|
29
|
Abstract
This article introduces methods for the acute purification and primary culture of Schwann cells from the mouse sciatic nerve. Immunopanning can be used to isolate Schwann cells from intact nerves during early postnatal development as well as to purify Schwann cells from adult nerves following sciatic nerve injury. These methods facilitate the exploration of mouse Schwann cell biology in the healthy and injured peripheral nerve.
Collapse
Affiliation(s)
- Amanda Brosius Lutz
- Stanford University School of Medicine, Department of Neurobiology, Stanford, California 94305
| |
Collapse
|
30
|
Chen M, Zheng B. Axon plasticity in the mammalian central nervous system after injury. Trends Neurosci 2014; 37:583-93. [PMID: 25218468 DOI: 10.1016/j.tins.2014.08.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Revised: 08/19/2014] [Accepted: 08/21/2014] [Indexed: 12/31/2022]
Abstract
It is widely recognized that severed axons in the adult central nervous system (CNS) have limited capacity to regenerate. However, mounting evidence from studies of CNS injury response and repair is challenging the prevalent view that the adult mammalian CNS is incapable of structural reorganization to adapt to an altered environment. Animal studies demonstrate the potential to achieve significant anatomical repair and functional recovery following CNS injury by manipulating axon growth regulators alone or in combination with activity-dependent strategies. With a growing understanding of the cellular and molecular mechanisms regulating axon plasticity, and the availability of new experimental tools to map detour circuits of functional importance, directing circuit rewiring to promote functional recovery may be achieved.
Collapse
Affiliation(s)
- Meifan Chen
- Department of Neurosciences, University of California, San Diego, 9500 Gilman Drive, MC 0691, La Jolla, CA 92093-0691, USA
| | - Binhai Zheng
- Department of Neurosciences, University of California, San Diego, 9500 Gilman Drive, MC 0691, La Jolla, CA 92093-0691, USA.
| |
Collapse
|
31
|
Placheta E, Hendry JM, Wood MD, Lafontaine CW, Liu EH, Cecilia Alvarez Veronesi M, Frey M, Gordon T, Borschel GH. The ErbB2 inhibitor Herceptin (Trastuzumab) promotes axonal outgrowth four weeks after acute nerve transection and repair. Neurosci Lett 2014; 582:81-6. [PMID: 25220708 DOI: 10.1016/j.neulet.2014.09.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Revised: 08/28/2014] [Accepted: 09/01/2014] [Indexed: 12/30/2022]
Abstract
Accumulating evidence suggests that neuregulin, a potent Schwann cell mitogen, and its receptor, ErbB2, have an important role in regulating peripheral nerve regeneration. We hypothesized that Herceptin (Trastuzumab), a monoclonal antibody that binds ErbB2, would disrupt ErbB2 signaling, allowing us to evaluate ErbB2's importance in peripheral nerve regeneration. In this study, the extent of peripheral motor and sensory nerve regeneration and distal axonal outgrowth was analyzed two and four weeks after common peroneal (CP) nerve injury in rats. Outcomes analyzed included neuron counts after retrograde labeling, histomorphometry, and protein analysis. The data analysis revealed that there was no impact of Herceptin administration on either the numbers of motor or sensory neurons that regenerated their axons but histomorphometry revealed that Herceptin significantly increased the number of regenerated axons in the distal repaired nerve after 4 weeks. Protein analysis with Western blotting revealed no difference in either expression levels of ErbB2 or the amount of activated, phosphorylated ErbB2 in injured nerves. In conclusion, administration of the ErbB2 receptor inhibitor after nerve transection and surgical repair did not alter the number of regenerating neurons but markedly increased the number of regenerated axons per neuron in the distal nerve stump. Enhanced axon outgrowth in the presence of this ErbB2 inhibitor indicates that ErbB2 signaling may limit the numbers of axons that are emitted from each regenerating neuron.
Collapse
Affiliation(s)
- Eva Placheta
- Division of Plastic and Reconstructive Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; Division of Plastic and Reconstructive Surgery, The Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada
| | - J Michael Hendry
- Division of Plastic and Reconstructive Surgery, The Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada; Department of Surgery, University of Toronto, 149 College Street, 5th Floor, Toronto, ON M5T 1P5, Canada; Institute of Medical Science, University of Toronto, 1 King's College Circle, Room 2374, Toronto, ON M5S 1A8, Canada
| | - Matthew D Wood
- Division of Plastic and Reconstructive Surgery, The Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada
| | - Christine W Lafontaine
- Division of Plastic and Reconstructive Surgery, The Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada
| | - Edward H Liu
- Division of Plastic and Reconstructive Surgery, The Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada
| | - M Cecilia Alvarez Veronesi
- Division of Plastic and Reconstructive Surgery, The Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Rosebrugh Building, RM 407 164 College Street, Toronto, ON M5S 3G9, Canada
| | - Manfred Frey
- Division of Plastic and Reconstructive Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Tessa Gordon
- Division of Plastic and Reconstructive Surgery, The Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada
| | - Gregory H Borschel
- Division of Plastic and Reconstructive Surgery, The Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada; Department of Surgery, University of Toronto, 149 College Street, 5th Floor, Toronto, ON M5T 1P5, Canada; Institute of Medical Science, University of Toronto, 1 King's College Circle, Room 2374, Toronto, ON M5S 1A8, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Rosebrugh Building, RM 407 164 College Street, Toronto, ON M5S 3G9, Canada.
| |
Collapse
|
32
|
Bastien D, Lacroix S. Cytokine pathways regulating glial and leukocyte function after spinal cord and peripheral nerve injury. Exp Neurol 2014; 258:62-77. [PMID: 25017888 DOI: 10.1016/j.expneurol.2014.04.006] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2013] [Revised: 02/20/2014] [Accepted: 04/08/2014] [Indexed: 01/13/2023]
Abstract
Injury to the nervous system causes the almost immediate release of cytokines by glial cells and neurons. These cytokines orchestrate a complex array of responses leading to microgliosis, immune cell recruitment, astrogliosis, scarring, and the clearance of cellular debris, all steps that affect neuronal survival and repair. This review will focus on cytokines released after spinal cord and peripheral nerve injury and the primary signalling pathways triggered by these inflammatory mediators. Notably, the following cytokine families will be covered: IL-1, TNF, IL-6-like, TGF-β, and IL-10. Whether interfering with cytokine signalling could lead to novel therapies will also be discussed. Finally, the review will address whether manipulating the above-mentioned cytokine families and signalling pathways could exert distinct effects in the injured spinal cord versus peripheral nerve.
Collapse
Affiliation(s)
- Dominic Bastien
- Centre de recherche du Centre hospitalier universitaire de Québec-CHUL, Département de médecine moléculaire, Université Laval, Québec, QC, Canada
| | - Steve Lacroix
- Centre de recherche du Centre hospitalier universitaire de Québec-CHUL, Département de médecine moléculaire, Université Laval, Québec, QC, Canada..
| |
Collapse
|
33
|
Lee HJ, Shin YK, Park HT. Mitogen Activated Protein Kinase Family Proteins and c-jun Signaling in Injury-induced Schwann Cell Plasticity. Exp Neurobiol 2014; 23:130-7. [PMID: 24963277 PMCID: PMC4065826 DOI: 10.5607/en.2014.23.2.130] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 05/21/2014] [Accepted: 05/21/2014] [Indexed: 12/31/2022] Open
Abstract
Schwann cells (SCs) in the peripheral nerves myelinate axons during postnatal development to allow saltatory conduction of nerve impulses. Well-organized structures of myelin sheathes are maintained throughout life unless nerves are insulted. After peripheral nerve injury, unidentified signals from injured nerves drive SC dedifferentiation into an immature state. Dedifferentiated SCs participate in axonal regeneration by producing neurotrophic factors and removing degenerating nerve debris. In this review, we focus on the role of mitogen activated protein kinase family proteins (MAP kinases) in SC dedifferentiation. In addition, we will highlight neuregulin 1 and the transcription factor c-jun as upstream and downstream signals for MAP kinases in SC responses to nerve injury.
Collapse
Affiliation(s)
- Hye Jeong Lee
- Department of Pharmacology, Mitochondria Hub Regulation Center (MHRC), College of Medicine, Dong-A University, Busan 602-714, Korea
| | - Yoon Kyung Shin
- Department of Physiology, Mitochondria Hub Regulation Center (MHRC), College of Medicine, Dong-A University, Busan 602-714, Korea
| | - Hwan Tae Park
- Department of Physiology, Mitochondria Hub Regulation Center (MHRC), College of Medicine, Dong-A University, Busan 602-714, Korea
| |
Collapse
|
34
|
Wu SC, Rau CS, Lu TH, Wu CJ, Wu YC, Tzeng SL, Chen YC, Hsieh CH. Knockout of TLR4 and TLR2 impair the nerve regeneration by delayed demyelination but not remyelination. J Biomed Sci 2013; 20:62. [PMID: 23984978 PMCID: PMC3765918 DOI: 10.1186/1423-0127-20-62] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 08/27/2013] [Indexed: 01/09/2023] Open
Abstract
Background Knockout of either toll-like receptor 4 (TLR4) or 2 (TLR2) had been reported to delay the Wallerian degeneration after peripheral nerve injury by deterring the recruitment of the macrophages and clearance of myelin debris. However, the impact on the remyelination process is poorly understood. In this study, the effect of TLR2 and TLR4 knockout on the nerve regeneration and on the remyelination process was studied in a mouse model of sciatic nerve crush injury. Results A standard sciatic nerve crush injury by a No. 5 Jeweler forcep for consistent 30 seconds was performed in Tlr4−/− (B6.B10ScN-Tlr4lps-del/JthJ), Tlr2−/− (B6.129-Tlr2tm1Kir/J) and C57BL/6 mice. One centimeter of nerve segment distal to the crushed site was harvested for western blot analysis of the myelin structure protein myelin protein zero (Mpz) and the remyelination transcription factors Oct6 and Sox10 at day 0, 3, 7, 10, 14, 17, 21, 28. Nerve segment 5-mm distal to injured site from additional groups of mice at day 10 after crush injury were subjected to semi-thin section and toluidine blue stain for a quantitative histomorphometric analysis. With less remyelinated nerves and more nerve debris, the histomorphometric analysis revealed a worse nerve regeneration following the sciatic nerve crush injury in both Tlr4−/− and Tlr2−/− mice than the C57BL/6 mice. Although there was a delayed expression of Sox10 but not Oct6 during remyelination, with an average 4-day delay in the demyelination process, the subsequent complete formation of Mpz during remyelination was also delayed for 4 days, implying that the impaired nerve regeneration was mainly attributed to the delayed demyelination process. Conclusions Both TLR4 and TLR2 are crucial for nerve regeneration after nerve crush injury mainly by delaying the demyelination but not the remyelination process.
Collapse
Affiliation(s)
- Shao-Chun Wu
- Department of Plastic and Reconstructive Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, No, 123, Ta-Pei Road, Niao-Sung District, Kaohsiung City, 833, Taiwan.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Repair of the Peripheral Nerve-Remyelination that Works. Brain Sci 2013; 3:1182-97. [PMID: 24961524 PMCID: PMC4061866 DOI: 10.3390/brainsci3031182] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Revised: 07/07/2013] [Accepted: 07/19/2013] [Indexed: 12/15/2022] Open
Abstract
In this review we summarize the events known to occur after an injury in the peripheral nervous system. We have focused on the Schwann cells, as they are the most important cells for the repair process and facilitate axonal outgrowth. The environment created by this cell type is essential for the outcome of the repair process. The review starts with a description of the current state of knowledge about the initial events after injury, followed by Wallerian degeneration, and subsequent regeneration. The importance of surgical repair, carried out as soon as possible to increase the chances of a good outcome, is emphasized throughout the review. The review concludes by describing the target re-innervation, which today is one of the most serious problems for nerve regeneration. It is clear, compiling this data, that even though regeneration of the peripheral nervous system is possible, more research in this area is needed in order to perfect the outcome.
Collapse
|
36
|
Nerve regeneration in rat limb allografts: evaluation of acute rejection rescue. Plast Reconstr Surg 2013; 131:499e-511e. [PMID: 23542267 DOI: 10.1097/prs.0b013e31828275b7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
BACKGROUND Successful nerve regeneration is critical to the functional success of composite tissue allografts. The present study was designed to characterize the effect of acute rejection on nerve regeneration and functional recovery in the setting of orthotopic limb transplantation. METHODS A rat orthotopic limb transplantation model was used to evaluate the effects of acute rejection on nerve regeneration and motor recovery. Continuous administration of FK506 (full suppression), administration of FK506 for the first 8 of 12 weeks (late rejection), or delayed administration of FK506/dexamethasone following noticeable rejection (early rejection) was used to preclude or induce rejection following limb transplantation. Twelve weeks postoperatively, nerve regeneration was assessed by means of histomorphometric analysis of explanted sciatic nerve, and motor recovery was assessed by means of evoked muscle force measurement in extensor digitorum longus muscle. RESULTS A single episode of acute rejection that occurs immediately or late after reconstruction does not significantly alter the number of regenerating axonal fibers. Acute rejection occurring late after reconstruction adversely affects extensor digitorum longus muscle function in composite tissue allografts. CONCLUSIONS Collected data reinforce that adequate immunosuppressant administration in cases of allogeneic limb transplantation ensures levels of nerve regeneration and motor functional recovery equivalent to that of syngeneic transplants. Prompt rescue following acute rejection was further demonstrated not to significantly affect nerve regeneration and functional recovery postoperatively. However, instances of acute rejection that occur late after reconstruction affect graft function. In total, the present study begins to characterize the effect of immunosuppression regimens on nerve regeneration and motor recovery in the setting of composite tissue allografts.
Collapse
|
37
|
The characterisation of Pax3 expressant cells in adult peripheral nerve. PLoS One 2013; 8:e59184. [PMID: 23527126 PMCID: PMC3602598 DOI: 10.1371/journal.pone.0059184] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 02/12/2013] [Indexed: 12/25/2022] Open
Abstract
Pax3 has numerous integral functions in embryonic tissue morphogenesis and knowledge of its complex function in cells of adult tissue continues to unfold. Across a variety of adult tissue lineages, the role of Pax3 is principally linked to maintenance of the tissue’s resident stem/progenitor cell population. In adult peripheral nerves, Pax3 is reported to be expressed in nonmyelinating Schwann cells, however, little is known about the purpose of this expression. Based on the evidence of the role of Pax3 in other adult tissue stem and progenitor cells, it was hypothesised that the cells in adult peripheral nerve that express Pax3 may be peripheral glioblasts. Here, methods have been developed for identification and visualisation of Pax3 expressant cells in normal 60 day old mouse peripheral nerve that allowed morphological and phenotypic distinctions to be made between Pax3 expressing cells and other nonmyelinating Schwann cells. The distinctions described provide compelling support for a resident glioblast population in adult mouse peripheral nerve.
Collapse
|
38
|
Guo X, Spradling S, Stancescu M, Lambert S, Hickman JJ. Derivation of sensory neurons and neural crest stem cells from human neural progenitor hNP1. Biomaterials 2013; 34:4418-27. [PMID: 23498896 DOI: 10.1016/j.biomaterials.2013.02.061] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Accepted: 02/20/2013] [Indexed: 12/27/2022]
Abstract
Although sensory neurons constitute a critical component for the proper function of the nervous system, the in vitro differentiation of functional sensory neurons from human stem cells has not yet been reported. This study presents the differentiation of sensory neurons (SNs) from a human neural progenitor cell line, hNP1, and their functional maturation in a defined, in vitro culture system without murine cell feeder layers. The SNs were characterized by immunocytochemistry and their functional maturation was evaluated by electrophysiology. Neural crest (NC) precursors, as one of the cellular derivatives in the differentiation culture, were isolated, propagated, and tested for their ability to generate sensory neurons. The hSC-derived SNs, as well as the NC precursors provide valuable tools for developing in vitro functional systems that model sensory neuron-related neural circuits and for designing therapeutic models for related diseases.
Collapse
Affiliation(s)
- Xiufang Guo
- NanoScience Technology Center, University of Central Florida, Orlando, FL 32826, USA
| | | | | | | | | |
Collapse
|
39
|
Loesch A, Dashwood MR, Coppi AA. Immunoreactive Endothelin-1 and Endothelin A Receptor in Basilar Artery Perivascular Nerves of Young and Adult Capybaras. Cells Tissues Organs 2013; 198:47-56. [DOI: 10.1159/000348617] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2013] [Indexed: 11/19/2022] Open
|
40
|
Jedi-1 and MEGF10 signal engulfment of apoptotic neurons through the tyrosine kinase Syk. J Neurosci 2012; 32:13022-31. [PMID: 22993420 DOI: 10.1523/jneurosci.6350-11.2012] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
During the development of the peripheral nervous system there is extensive apoptosis, and these neuronal corpses need to be cleared to prevent an inflammatory response. Recently, Jedi-1 and MEGF10, both expressed in glial precursor cells, were identified in mouse as having an essential role in this phagocytosis (Wu et al., 2009); however, the mechanisms by which they promote engulfment remained unknown. Both Jedi-1 and MEGF10 are homologous to the Drosophila melanogaster receptor Draper, which mediates engulfment through activation of the tyrosine kinase Shark. Here, we identify Syk, the mammalian homolog of Shark, as a signal transducer for both Jedi-1 and MEGF10. Syk interacted with each receptor independently through the immunoreceptor tyrosine-based activation motifs (ITAMs) in their intracellular domains. The interaction was enhanced by phosphorylation of the tyrosines in the ITAMs by Src family kinases (SFKs). Jedi association with Syk and activation of the kinase was also induced by exposure to dead cells. Expression of either Jedi-1 or MEGF10 in HeLa cells facilitated engulfment of carboxylated microspheres to a similar extent, and there was no additive effect when they were coexpressed. Mutation of the ITAM tyrosines of Jedi-1 and MEGF10 prevented engulfment. The SFK inhibitor PP2 or a selective Syk inhibitor (BAY 61-3606) also blocked engulfment. Similarly, in cocultures of glial precursors and dying sensory neurons from embryonic mice, addition of PP2 or knock down of endogenous Syk decreased the phagocytosis of apoptotic neurons. These results indicate that both Jedi-1 and MEGF10 can mediate phagocytosis independently through the recruitment of Syk.
Collapse
|
41
|
Rajaram A, Chen XB, Schreyer DJ. Strategic Design and Recent Fabrication Techniques for Bioengineered Tissue Scaffolds to Improve Peripheral Nerve Regeneration. TISSUE ENGINEERING PART B-REVIEWS 2012; 18:454-67. [DOI: 10.1089/ten.teb.2012.0006] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Ajay Rajaram
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, Canada
| | - Xiong-Biao Chen
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, Canada
- Department of Mechanical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, Canada
| | - David J. Schreyer
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, Canada
- Department of Anatomy and Cell Biology, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| |
Collapse
|
42
|
Changes in CLIP3 expression after sciatic nerve injury in adult rats. J Mol Histol 2012; 43:669-79. [PMID: 23014974 DOI: 10.1007/s10735-012-9450-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Accepted: 09/14/2012] [Indexed: 02/04/2023]
Abstract
CLIP3 (cytoplasmic linker protein 3) is a 547 amino acid residue cytoplasmic protein that localises to Golgi stacks and tubulovesicular elements juxtaposed to Golgi cisternae. Composed of three Ank (ankyrin) repeats and two CAP-Gly (cytoskeleton-associated protein-glycine) domains, CLIP3 may function as a cytoplasmic linker protein that is involved in TGN-endosome dynamics. To define the expression and role of CLIP3 during peripheral nervous system degeneration and regeneration, we created an acute sciatic nerve injury (SNI) model in adult rats. Western blot analyses revealed prominent up-regulation of CLIP3 and PCNA (proliferating cell nuclear antigen) protein levels at 3 days after SNI. Immunohistochemistry displayed that the expression of CLIP3 was noticeably increased in the injured nerve. Immunofluorescence further revealed that the CLIP3 and PCNA proteins colocalised respectively with S100 in the cytoplasm of Schwann cells. The expression profile of the SC/neuron co-cultures demonstrated that CLIP3 and PCNA protein levels were markedly expressed during the early stage of myelination. These results suggest that CLIP3 is likely associated with the myelination of proliferating Schwann cells, and nerve tissue regeneration after peripheral nerve injury. CLIP3 and PCNA expression during early myelination may be related to the direct uptake and transport of lipids and cholesterol, which were derived from the degenerating myelin, by Schwann cells to prepare for the formation of myelin sheath-like structures around regenerated axons after SNI.
Collapse
|
43
|
Liu H, Shiryaev SA, Chernov AV, Kim Y, Shubayev I, Remacle AG, Baranovskaya S, Golubkov VS, Strongin AY, Shubayev VI. Immunodominant fragments of myelin basic protein initiate T cell-dependent pain. J Neuroinflammation 2012; 9:119. [PMID: 22676642 PMCID: PMC3416717 DOI: 10.1186/1742-2094-9-119] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Accepted: 04/23/2012] [Indexed: 12/11/2022] Open
Abstract
Background The myelin sheath provides electrical insulation of mechanosensory Aβ-afferent fibers. Myelin-degrading matrix metalloproteinases (MMPs) damage the myelin sheath. The resulting electrical instability of Aβ-fibers is believed to activate the nociceptive circuitry in Aβ-fibers and initiate pain from innocuous tactile stimulation (mechanical allodynia). The precise molecular mechanisms, responsible for the development of this neuropathic pain state after nerve injury (for example, chronic constriction injury, CCI), are not well understood. Methods and results Using mass spectrometry of the whole sciatic nerve proteome followed by bioinformatics analyses, we determined that the pathways, which are classified as the Infectious Disease and T-helper cell signaling, are readily activated in the nerves post-CCI. Inhibition of MMP-9/MMP-2 suppressed CCI-induced mechanical allodynia and concomitant TNF-α and IL-17A expression in nerves. MMP-9 proteolysis of myelin basic protein (MBP) generated the MBP84-104 and MBP68-86 digest peptides, which are prominent immunogenic epitopes. In agreement, the endogenous MBP69-86 epitope co-localized with MHCII and MMP-9 in Schwann cells and along the nodes of Ranvier. Administration of either the MBP84-104 or MBP68-86 peptides into the naïve nerve rapidly produced robust mechanical allodynia with a concomitant increase in T cells and MHCII-reactive cell populations at the injection site. As shown by the genome-wide expression profiling, a single intraneural MBP84-104 injection stimulated the inflammatory, immune cell trafficking, and antigen presentation pathways in the injected naïve nerves and the associated spinal cords. Both MBP84-104-induced mechanical allodynia and characteristic pathway activation were remarkably less prominent in the T cell-deficient athymic nude rats. Conclusions These data implicate MBP as a novel mediator of pain. Furthermore, the action of MMPs expressed within 1 day post-injury is critical to the generation of tactile allodynia, neuroinflammation, and the immunodominant MBP digest peptides in nerve. These MBP peptides initiate mechanical allodynia in both a T cell-dependent and -independent manner. In the course of Wallerian degeneration, the repeated exposure of the cryptic MBP epitopes, which are normally sheltered from immunosurveillance, may induce the MBP-specific T cell clones and a self-sustaining immune reaction, which may together contribute to the transition of acute pain into a chronic neuropathic pain state.
Collapse
Affiliation(s)
- Huaqing Liu
- Department of Anesthesiology, University of California, San Diego, 9500 Gilman Dr., Mail Box 0629, La Jolla, CA 92093-0629, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Lou D, Sun B, Wei H, Deng X, Chen H, Xu D, Li G, Xu H, Wang Y. Spatiotemporal Expression of Testicular Protein Kinase 1 After Rat Sciatic Nerve Injury. J Mol Neurosci 2012; 47:180-91. [DOI: 10.1007/s12031-012-9712-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Accepted: 01/20/2012] [Indexed: 11/28/2022]
|
45
|
Rotshenker S. Wallerian degeneration: the innate-immune response to traumatic nerve injury. J Neuroinflammation 2011; 8:109. [PMID: 21878125 PMCID: PMC3179447 DOI: 10.1186/1742-2094-8-109] [Citation(s) in RCA: 334] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Accepted: 08/30/2011] [Indexed: 12/23/2022] Open
Abstract
Traumatic injury to peripheral nerves results in the loss of neural functions. Recovery by regeneration depends on the cellular and molecular events of Wallerian degeneration that injury induces distal to the lesion site, the domain through which severed axons regenerate back to their target tissues. Innate-immunity is central to Wallerian degeneration since innate-immune cells, functions and molecules that are produced by immune and non-immune cells are involved. The innate-immune response helps to turn the peripheral nerve tissue into an environment that supports regeneration by removing inhibitory myelin and by upregulating neurotrophic properties. The characteristics of an efficient innate-immune response are rapid onset and conclusion, and the orchestrated interplay between Schwann cells, fibroblasts, macrophages, endothelial cells, and molecules they produce. Wallerian degeneration serves as a prelude for successful repair when these requirements are met. In contrast, functional recovery is poor when injury fails to produce the efficient innate-immune response of Wallerian degeneration.
Collapse
Affiliation(s)
- Shlomo Rotshenker
- Dept. of Medical Neurobiology, IMRIC, Hebrew University, Faculty of Medicine, Jerusalem, Israel.
| |
Collapse
|
46
|
Whitney KM, Schwartz Sterman AJ, O’Connor J, Foley GL, Garman RH. Light Microscopic Sciatic Nerve Changes in Control Beagle Dogs from Toxicity Studies. Toxicol Pathol 2011; 39:835-40. [DOI: 10.1177/0192623311413786] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although the dog is a common choice among nonrodent species in evaluation of compound safety for regulatory submission, information regarding the incidence of spontaneous or incidental microscopic changes in canine peripheral nerve is limited. A retrospective examination was performed of routine histologic preparations of sciatic nerve from eighty-one control dogs in toxicity studies ranging from ten days to three months in duration. Spontaneous background changes included digestion chambers, foci of vacuolation, nerve fibers circumscribed by proliferating Schwann cells (bands of Büngner), and small foci of myelin aggregation. The latter accounted for 91% of the microscopic changes and were noted in all sections examined. These changes were quantified, and the number per square millimeter of evaluable nerve tissue was determined for each slide. Densities of foci varied among the slides examined; no age- or sex-related trends were apparent. In addition, anatomic features of peripheral nerves including nodes of Ranvier, Schmidt-Lanterman incisures, Renaut bodies, and effects resulting from sectioning plane were noted. By demonstrating the range of effects observed within control animals, these observations provide a basis for recognition of possible compound-related effects in routine nerve preparations from dogs included in toxicity studies.
Collapse
Affiliation(s)
| | | | - Jackie O’Connor
- Global Preclinical Safety, Abbott Laboratories, Abbott Park, Illinois, USA
| | - George L. Foley
- Global Preclinical Safety, Abbott Laboratories, Abbott Park, Illinois, USA
| | - Robert H. Garman
- Consultants in Veterinary Pathology, Murrysville, Pennsylvania, USA
| |
Collapse
|
47
|
King-Robson J. Encouraging regeneration in the central nervous system: Is there a role for olfactory ensheathing cells? Neurosci Res 2011; 69:263-75. [DOI: 10.1016/j.neures.2010.12.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2010] [Revised: 12/16/2010] [Accepted: 12/17/2010] [Indexed: 10/18/2022]
|
48
|
Endogenous antibodies promote rapid myelin clearance and effective axon regeneration after nerve injury. Proc Natl Acad Sci U S A 2010; 107:11993-8. [PMID: 20547838 DOI: 10.1073/pnas.1001948107] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Degenerating myelin inhibits axon regeneration and is rapidly cleared after peripheral (PNS) but not central nervous system (CNS) injury. To better understand mechanisms underlying rapid PNS myelin clearance, we tested the potential role of the humoral immune system. Here, we show that endogenous antibodies are required for rapid and robust PNS myelin clearance and axon regeneration. B-cell knockout JHD mice display a significant delay in macrophage influx, myelin clearance, and axon regeneration. Rapid clearance of myelin debris is restored in mutant JHD mice by passive transfer of antibodies from naïve WT mice or by an anti-PNS myelin antibody, but not by delivery of nonneural antibodies. We demonstrate that degenerating nerve tissue is targeted by preexisting endogenous antibodies that control myelin clearance by promoting macrophage entrance and phagocytic activity. These results demonstrate a role for immunoglobulin (Ig) in clearing damaged self during healing and suggest that the immune-privileged status of the CNS may contribute to failure of CNS myelin clearance and axon regeneration after injury.
Collapse
|
49
|
Li FQ, Fowler KA, Neil JE, Colton CA, Vitek MP. An apolipoprotein E-mimetic stimulates axonal regeneration and remyelination after peripheral nerve injury. J Pharmacol Exp Ther 2010; 334:106-15. [PMID: 20406857 DOI: 10.1124/jpet.110.167882] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Elevated apolipoprotein E (apoE) synthesis within crushed sciatic nerves advocates that apoE could benefit axonal repair and reconstruction of axonal and myelin membranes. We created an apoE-mimetic peptide, COG112 (acetyl-RQIKIWFQNRRMKWKKCLRVRLASHLRKLRKRLL-amide), and found that postinjury treatment with COG112 significantly improved recovery of motor and sensory function following sciatic nerve crush in C57BL/6 mice. Morphometric analysis of injured sciatic nerves revealed that COG112 promoted axonal regrowth after 2 weeks of treatment. More strikingly, the thickness of myelin sheaths was increased by COG112 treatment. Consistent with these histological findings, COG112 potently elevated growth associated protein 43 (GAP-43) and peripheral myelin protein zero (P0), which are markers of axon regeneration and remyelination, respectively. Electron microscopic examination further suggested that the apoE-mimetic COG112 may increase clearance of myelin debris. Schwann cell uptake of cholesterol-containing low-density lipoprotein particles was selectively enhanced by COG112 treatment in a Schwann cell line S16. Moreover, COG112 significantly promoted axon elongation in primary dorsal root ganglion cultures from rat pups. Considering that cholesterol and lipids are needed for reconstructing myelin sheaths and axon extension, these data support a hypothesis where supplementation with exogenous apoE-mimetics such as COG112 may be a promising strategy for restoring lost functional and structural elements following nerve injury.
Collapse
Affiliation(s)
- Feng-Qiao Li
- Cognosci, Inc., Research Triangle Park, NC 27709, USA.
| | | | | | | | | |
Collapse
|
50
|
Tao T, Ji Y, Cheng C, Yang H, Liu H, Sun L, Qin Y, Yang J, Wang H, Shen A. Tumor necrosis factor-alpha inhibits Schwann cell proliferation by up-regulating Src-suppressed protein kinase C substrate expression. J Neurochem 2009; 111:647-55. [DOI: 10.1111/j.1471-4159.2009.06346.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|