1
|
Jessen KR, Mirsky R. The Role of c-Jun and Autocrine Signaling Loops in the Control of Repair Schwann Cells and Regeneration. Front Cell Neurosci 2022; 15:820216. [PMID: 35221918 PMCID: PMC8863656 DOI: 10.3389/fncel.2021.820216] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 12/30/2021] [Indexed: 12/12/2022] Open
Abstract
After nerve injury, both Schwann cells and neurons switch to pro-regenerative states. For Schwann cells, this involves reprogramming of myelin and Remak cells to repair Schwann cells that provide the signals and mechanisms needed for the survival of injured neurons, myelin clearance, axonal regeneration and target reinnervation. Because functional repair cells are essential for regeneration, it is unfortunate that their phenotype is not robust. Repair cell activation falters as animals get older and the repair phenotype fades during chronic denervation. These malfunctions are important reasons for the poor outcomes after nerve damage in humans. This review will discuss injury-induced Schwann cell reprogramming and the concept of the repair Schwann cell, and consider the molecular control of these cells with emphasis on c-Jun. This transcription factor is required for the generation of functional repair cells, and failure of c-Jun expression is implicated in repair cell failures in older animals and during chronic denervation. Elevating c-Jun expression in repair cells promotes regeneration, showing in principle that targeting repair cells is an effective way of improving nerve repair. In this context, we will outline the emerging evidence that repair cells are sustained by autocrine signaling loops, attractive targets for interventions aimed at promoting regeneration.
Collapse
Affiliation(s)
- Kristjan R. Jessen
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | | |
Collapse
|
2
|
Khodabakhsh P, Pournajaf S, Mohaghegh Shalmani L, Ahmadiani A, Dargahi L. Insulin Promotes Schwann-Like Cell Differentiation of Rat Epidermal Neural Crest Stem Cells. Mol Neurobiol 2021; 58:5327-5337. [PMID: 34297315 DOI: 10.1007/s12035-021-02423-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 05/05/2021] [Indexed: 10/20/2022]
Abstract
Schwann cells (SCs) are considered potentially attractive candidates for transplantation therapies in neurodegenerative diseases. However, problems arising from the isolation and expansion of the SCs restrict their clinical applications. Establishing an alternative Schwann-like cell type is a prerequisite. Epidermal neural crest stem cells (EPI-NCSCs) are well studied for their autologous accessibility, along with the ability to produce major neural crest derivatives and neurotrophic factors. In the current study, we explored insulin influence, a well-known growth factor, on directing EPI-NCSCs into the Schwann cell (SC) lineage. EPI-NCSCs were isolated from rat hair bulge explants. The viability of cells treated with a range of insulin concentrations (0.05-100 μg/ml) was defined by MTT assay at 24, 48, and 72 h. The gene expression profiles of neurotrophic factors (BDNF, FGF-2, and IL-6), key regulators involved in the development of SC (EGR-1, SOX-10, c-JUN, GFAP, OCT-6, EGR-2, and MBP), and oligodendrocyte (PDGFR-α and NG-2) were quantified 1 and 9 days post-treatment with 0.05 and 5 μg/ml insulin. Furthermore, the protein expression of nestin (stemness marker), SOX-10, PDGFR-α, and MBP was analyzed following the long-term insulin treatment. Insulin downregulated the early-stage SC differentiation marker (EGR-1) and increased neurotrophins (BDNF and IL-6) and pro-myelinating genes, including OCT-6, SOX-10, EGR-2, and MBP, as well as oligodendrocyte differentiation markers, upon exposure for 9 days. Insulin can promote EPI-NCSC differentiation toward SC lineage and possibly oligodendrocytes. Thus, employing insulin might enhance the EPI-NCSCs efficiency in cell transplantation strategies.
Collapse
Affiliation(s)
- Pariya Khodabakhsh
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Safura Pournajaf
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Mohaghegh Shalmani
- Pharmacology and Toxicology Department, Faculty of Pharmacy and Pharmaceutical Sciences, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Dargahi
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Jessen KR, Arthur-Farraj P. Repair Schwann cell update: Adaptive reprogramming, EMT, and stemness in regenerating nerves. Glia 2019; 67:421-437. [PMID: 30632639 DOI: 10.1002/glia.23532] [Citation(s) in RCA: 245] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/20/2018] [Accepted: 09/05/2018] [Indexed: 12/16/2022]
Abstract
Schwann cells respond to nerve injury by cellular reprogramming that generates cells specialized for promoting regeneration and repair. These repair cells clear redundant myelin, attract macrophages, support survival of damaged neurons, encourage axonal growth, and guide axons back to their targets. There are interesting parallels between this response and that found in other tissues. At the cellular level, many other tissues also react to injury by cellular reprogramming, generating cells specialized to promote tissue homeostasis and repair. And at the molecular level, a common feature possessed by Schwann cells and many other cells is the injury-induced activation of genes associated with epithelial-mesenchymal transitions and stemness, differentiation states that are linked to cellular plasticity and that help injury-induced tissue remodeling. The number of signaling systems regulating Schwann cell plasticity is rapidly increasing. Importantly, this includes mechanisms that are crucial for the generation of functional repair Schwann cells and nerve regeneration, although they have no or a minor role elsewhere in the Schwann cell lineage. This encourages the view that selective tools can be developed to control these particular cells, amplify their repair supportive functions and prevent their deterioration. In this review, we discuss the emerging similarities between the injury response seen in nerves and in other tissues and survey the transcription factors, epigenetic mechanisms, and signaling cascades that control repair Schwann cells, with emphasis on systems that selectively regulate the Schwann cell injury response.
Collapse
Affiliation(s)
- Kristjan R Jessen
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Peter Arthur-Farraj
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
4
|
Jessen KR, Mirsky R. The Success and Failure of the Schwann Cell Response to Nerve Injury. Front Cell Neurosci 2019; 13:33. [PMID: 30804758 PMCID: PMC6378273 DOI: 10.3389/fncel.2019.00033] [Citation(s) in RCA: 311] [Impact Index Per Article: 51.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 01/22/2019] [Indexed: 01/06/2023] Open
Abstract
The remarkable plasticity of Schwann cells allows them to adopt the Remak (non-myelin) and myelin phenotypes, which are specialized to meet the needs of small and large diameter axons, and differ markedly from each other. It also enables Schwann cells initially to mount a strikingly adaptive response to nerve injury and to promote regeneration by converting to a repair-promoting phenotype. These repair cells activate a sequence of supportive functions that engineer myelin clearance, prevent neuronal death, and help axon growth and guidance. Eventually, this response runs out of steam, however, because in the long run the phenotype of repair cells is unstable and their survival is compromised. The re-programming of Remak and myelin cells to repair cells, together with the injury-induced switch of peripheral neurons to a growth mode, gives peripheral nerves their strong regenerative potential. But it remains a challenge to harness this potential and devise effective treatments that maintain the initial repair capacity of peripheral nerves for the extended periods typically required for nerve repair in humans.
Collapse
Affiliation(s)
- Kristjan R. Jessen
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | | |
Collapse
|
5
|
Sulaiman W, Dreesen T, Nguyen D. Single Local Application of TGF-β Promotes a Proregenerative State Throughout a Chronically Injured Nerve. Neurosurgery 2018; 82:894-902. [PMID: 28973496 DOI: 10.1093/neuros/nyx362] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 06/06/2017] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The lack of nerve regeneration and functional recovery occurs frequently when injuries involve large nerve trunks because insufficient mature axons reach their targets in the distal stump and because of the loss of neurotrophic support, primarily from Schwann cells (SCs). OBJECTIVE To investigate whether a single application of transforming growth factor-beta (TGF-β) plus forskolin or forskolin alone can promote and support axonal regeneration through the distal nerve stump. METHODS Using a delayed repair rat model of nerve injury, we transected the tibial nerve. After 8 wk, end-to-end repair was done and the repair site was treated with saline, forskolin, or TGF- β plus forskolin. After 6 wk, nerve sections consisting of the proximal stump, distal to the site of repair, and the most distal part of the nerve stump were removed for nerve histology, axon counts, and immunohistochemistry for activated SCs (S100), macrophages (CD68), cell proliferation (Ki67), p75NGFR, and apoptosis (activated caspase-3). RESULTS TGF-β plus forskolin significantly increased the numbers of axons regenerated distal to the repair site and the most distal nerve sections. Both treatments significantly increased the numbers of axons regenerated in the most distal nerve sections compared to saline treated. Both treatments exhibited extended expression of regeneration-associated marker proteins. CONCLUSION TGF-β plus forskolin treatment of chronically injured nerve improved axonal regeneration and increased expression of regeneration-associated proteins beyond the repair site. This suggests that a single application at the site of repair has mitogenic effects that extended distally and may potentially overcome the decrease in regenerated axon over long distance.
Collapse
Affiliation(s)
- Wale Sulaiman
- Department of Neurosurgery, Back and Spine Center, Ochsner Neuroscience Institute, Ochsner Health System, and Tulane University Medical Center, New Orleans, Louisiana.,Laboratory of Neural Injury and Regeneration, Institute of Translational Research, Ochsner Medical Center, New Orleans, Louisiana
| | - Thomas Dreesen
- Laboratory of Neural Injury and Regeneration, Institute of Translational Research, Ochsner Medical Center, New Orleans, Louisiana
| | - Doan Nguyen
- Laboratory of Neural Injury and Regeneration, Institute of Translational Research, Ochsner Medical Center, New Orleans, Louisiana
| |
Collapse
|
6
|
Abstract
The difference in regenerative capacity between the PNS and the CNS is not due to an intrinsic inability of central neurons to extend fibers. Rather, it is probably related to the environment in the CNS that is either repulsive to axonal outgrowth and/or nonsupportive of axonal elongation. In contrast, the PNS both supports and allows for axonal elongation after injury. The Schwann cell, which is the glial cell of the PNS, is strictly required for peripheral regeneration. Here we discuss recent work describing the biology of Schwann cell- dependent regeneration, discuss what is known of the molecular basis of this phenomenon, and how it might apply to the damaged CNS. NEUROSCIENTIST 5:208-216, 1999
Collapse
Affiliation(s)
- David E. Weinstein
- Departments of Neuroscience and Pathology Albert Einstein College of Medicine Bronx, New York
| |
Collapse
|
7
|
Rao SNR, Pearse DD. Regulating Axonal Responses to Injury: The Intersection between Signaling Pathways Involved in Axon Myelination and The Inhibition of Axon Regeneration. Front Mol Neurosci 2016; 9:33. [PMID: 27375427 PMCID: PMC4896923 DOI: 10.3389/fnmol.2016.00033] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 05/02/2016] [Indexed: 01/06/2023] Open
Abstract
Following spinal cord injury (SCI), a multitude of intrinsic and extrinsic factors adversely affect the gene programs that govern the expression of regeneration-associated genes (RAGs) and the production of a diversity of extracellular matrix molecules (ECM). Insufficient RAG expression in the injured neuron and the presence of inhibitory ECM at the lesion, leads to structural alterations in the axon that perturb the growth machinery, or form an extraneous barrier to axonal regeneration, respectively. Here, the role of myelin, both intact and debris, in antagonizing axon regeneration has been the focus of numerous investigations. These studies have employed antagonizing antibodies and knockout animals to examine how the growth cone of the re-growing axon responds to the presence of myelin and myelin-associated inhibitors (MAIs) within the lesion environment and caudal spinal cord. However, less attention has been placed on how the myelination of the axon after SCI, whether by endogenous glia or exogenously implanted glia, may alter axon regeneration. Here, we examine the intersection between intracellular signaling pathways in neurons and glia that are involved in axon myelination and axon growth, to provide greater insight into how interrogating this complex network of molecular interactions may lead to new therapeutics targeting SCI.
Collapse
Affiliation(s)
- Sudheendra N R Rao
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine Miami, FL, USA
| | - Damien D Pearse
- The Miami Project to Cure Paralysis, University of Miami Miller School of MedicineMiami, FL, USA; The Department of Neurological Surgery, University of Miami Miller School of MedicineMiami, FL, USA; The Neuroscience Program, University of Miami Miller School of MedicineMiami, FL, USA; The Interdisciplinary Stem Cell Institute, University of Miami Miller School of MedicineMiami, FL, USA; Bruce W. Carter Department of Veterans Affairs Medical CenterMiami, FL, USA
| |
Collapse
|
8
|
Sakaue M, Sieber-Blum M. Human epidermal neural crest stem cells as a source of Schwann cells. Development 2015; 142:3188-97. [PMID: 26251357 PMCID: PMC4582175 DOI: 10.1242/dev.123034] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 05/22/2015] [Indexed: 12/16/2022]
Abstract
We show that highly pure populations of human Schwann cells can be derived rapidly and in a straightforward way, without the need for genetic manipulation, from human epidermal neural crest stem cells [hEPI-NCSC(s)] present in the bulge of hair follicles. These human Schwann cells promise to be a useful tool for cell-based therapies, disease modelling and drug discovery. Schwann cells are glia that support axons of peripheral nerves and are direct descendants of the embryonic neural crest. Peripheral nerves are damaged in various conditions, including through trauma or tumour-related surgery, and Schwann cells are required for their repair and regeneration. Schwann cells also promise to be useful for treating spinal cord injuries. Ex vivo expansion of hEPI-NCSC isolated from hair bulge explants, manipulating the WNT, sonic hedgehog and TGFβ signalling pathways, and exposure of the cells to pertinent growth factors led to the expression of the Schwann cell markers SOX10, KROX20 (EGR2), p75NTR (NGFR), MBP and S100B by day 4 in virtually all cells, and maturation was completed by 2 weeks of differentiation. Gene expression profiling demonstrated expression of transcripts for neurotrophic and angiogenic factors, as well as JUN, all of which are essential for nerve regeneration. Co-culture of hEPI-NCSC-derived human Schwann cells with rodent dorsal root ganglia showed interaction of the Schwann cells with axons, providing evidence of Schwann cell functionality. We conclude that hEPI-NCSCs are a biologically relevant source for generating large and highly pure populations of human Schwann cells. Summary: Human epidermal neural crest stem cells isolated from the bulge of hair follicles are used to derive Schwann cells that could be useful for regenerative therapies, disease modelling and drug discovery.
Collapse
Affiliation(s)
- Motoharu Sakaue
- Institute of Genetic Medicine, Newcastle University, Centre for Life, Newcastle upon Tyne NE1 3BZ, UK
| | - Maya Sieber-Blum
- Institute of Genetic Medicine, Newcastle University, Centre for Life, Newcastle upon Tyne NE1 3BZ, UK
| |
Collapse
|
9
|
Kipanyula MJ, Woodhoo A, Rahman M, Payne D, Jessen KR, Mirsky R. Calcineurin-nuclear factor of activated T cells regulation of Krox-20 expression in Schwann cells requires elevation of intracellular cyclic AMP. J Neurosci Res 2013; 91:105-15. [PMID: 23073893 PMCID: PMC5722200 DOI: 10.1002/jnr.23131] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Revised: 07/09/2012] [Accepted: 07/20/2012] [Indexed: 12/13/2022]
Abstract
The transcription factor Krox-20 (Egr2) is a master regulator of Schwann cell myelination. In mice from which calcineurin B had been excised in cells of the neural crest lineage, calcineurin-nuclear factor of activated T cells (NFAT) signaling was required for neuregulin-related Schwann cell myelination (Kao et al. [2009] Immunity 12:359-372). Whether NFAT signaling required simultaneous elevation of intracellular cAMP levels was not explored. In vivo, Krox-20 expression requires continuous axon-Schwann cell signaling that in Schwann cell cultures can be mimicked by elevation of intracellular cAMP. We have investigated the role of the calcineurin-NFAT pathway in Krox-20 induction in purified rat Schwann cell cultures. Activation of this pathway requires elevation of intracellular Ca(2+) levels. The calcium ionophore A23187 or ionomycin was used to increase intracellular Ca(2+) levels in Schwann cell cultures that had been treated with dibutyryl cAMP to induce Krox-20. Increase in Ca(2+) levels significantly potentiated Krox-20 induction, determined by Krox-20 immunolabeling of individual cells and Western blotting. Levels of the myelin proteins periaxin and P(0) were also elevated. The potentiating effect was blocked by cyclosporin A, a specific blocker of the calcineurin-NFAT pathway. We found that, in the absence of cAMP elevation, treatment with A23187 alone failed to induce Krox-20 expression, indicating that NFAT upregulation of Krox-20 requires elevation of cAMP levels in Schwann cells. P-VIVIT, another specific inhibitor of calcineurin-NFAT interaction, blocked Krox-20 induction in response to dibutyryl cAMP and ionophore. HA-NFAT1 (1-460)-GFP translocated to the nucleus on treatment with dibutyryl cAMP with or without added ionophore. NFAT isoforms 1-4 were detected in purified Schwann cells by quantitative RT-PCR.
Collapse
Affiliation(s)
- Maulilio J. Kipanyula
- Department of Veterinary Anatomy, Sokoine University of Agriculture, Chuo Kikuu, Morogoro, Tanzania
| | - Ashwin Woodhoo
- Metabolomics Unit, CICbioGune, Parque Tecnológico de Bizcaia, Derio, Bizcaia, Spain
| | - Mary Rahman
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Donna Payne
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Kristján R. Jessen
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Rhona Mirsky
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| |
Collapse
|
10
|
Doddrell RDS, Dun XP, Moate RM, Jessen KR, Mirsky R, Parkinson DB. Regulation of Schwann cell differentiation and proliferation by the Pax-3 transcription factor. Glia 2012; 60:1269-78. [PMID: 22532290 PMCID: PMC5722199 DOI: 10.1002/glia.22346] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2011] [Accepted: 03/30/2012] [Indexed: 12/13/2022]
Abstract
Pax-3 is a paired domain transcription factor that plays many roles during vertebrate development. In the Schwann cell lineage, Pax-3 is expressed at an early stage in Schwann cells precursors of the embryonic nerve, is maintained in the nonmyelinating cells of the adult nerve, and is upregulated in Schwann cells after peripheral nerve injury. Consistent with this expression pattern, Pax-3 has previously been shown to play a role in repressing the expression of the myelin basic protein gene in Schwann cells. We have studied the role of Pax-3 in Schwann cells and have found that it controls not only the regulation of cell differentiation but also the survival and proliferation of Schwann cells. Pax-3 expression blocks both the induction of Oct-6 and Krox-20 (K20) by cyclic AMP and completely inhibits the ability of K20, the physiological regulator of myelination in the peripheral nervous system, to induce myelin gene expression in Schwann cells. In contrast to other inhibitors of myelination, we find that Pax-3 represses myelin gene expression in a c-Jun-independent manner. In addition to this, we find that Pax-3 expression alone is sufficient to inhibit the induction of apoptosis by TGFβ1 in Schwann cells. Expression of Pax-3 is also sufficient to induce the proliferation of Schwann cells in the absence of added growth factors and to reverse K20-induced exit from the cell cycle. These findings indicate new roles for the Pax-3 transcription factor in controlling the differentiation and proliferation of Schwann cells during development and after peripheral nerve injury.
Collapse
Affiliation(s)
- Robin D. S. Doddrell
- Peninsula College of Medicine and Dentistry, University of Exeter, Plymouth, Devon, United Kingdom
| | - Xin-Peng Dun
- Peninsula College of Medicine and Dentistry, University of Exeter, Plymouth, Devon, United Kingdom
| | - Roy M. Moate
- School of Biomedical and Biological Sciences, University of Plymouth, Plymouth, Devon, United Kingdom
| | - Kristjan R. Jessen
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Rhona Mirsky
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - David B. Parkinson
- Peninsula College of Medicine and Dentistry, University of Exeter, Plymouth, Devon, United Kingdom
| |
Collapse
|
11
|
Ammoun S, Schmid MC, Ristic N, Zhou L, Hilton D, Ercolano E, Carroll C, Hanemann CO. The role of insulin-like growth factors signaling in merlin-deficient human schwannomas. Glia 2012; 60:1721-33. [DOI: 10.1002/glia.22391] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Revised: 06/20/2012] [Accepted: 06/22/2012] [Indexed: 01/05/2023]
|
12
|
Chen G, Zhang Z, Wei Z, Cheng Q, Li X, Li W, Duan S, Gu X. Lysosomal exocytosis in Schwann cells contributes to axon remyelination. Glia 2011; 60:295-305. [PMID: 22042600 DOI: 10.1002/glia.21263] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Accepted: 10/06/2011] [Indexed: 12/21/2022]
Abstract
Myelin biogenesis is a complex process involving coordinated exocytosis, endocytosis, mRNA transport, and cytoskeletal dynamics. Although abnormalities of myelin are common in lysosomal storage diseases, our understanding of the role of lysosomes in the formation and maintenance of myelin is still limited. Here, we show that late endosomes/lysosomes in Schwann cells contain abundant myelin protein P0, which accounts for over half the total protein of compact myelin in the peripheral nervous system and exhibit Ca(2+) -dependent exocytosis in response to various stimuli. Downregulation of Rab27a, a small GTPase required for the trafficking of the secretory lysosomes to the plasma membrane, largely blocked lysosomal exocytosis in Schwann cells and reduced the remyelination of regenerated sciatic nerve. These findings highlight a novel role for lysosomes in Schwann cells and suggest that the regulated lysosome exocytosis in Schwann cells may have important physiological and pathological significance in the peripheral nervous system.
Collapse
Affiliation(s)
- Gang Chen
- Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, 226001, Jiangsu, China
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Jungnickel J, Haastert K, Grzybek M, Thau N, Lipokatic-Takacs E, Ratzka A, Nölle A, Claus P, Grothe C. Mice lacking basic fibroblast growth factor showed faster sensory recovery. Exp Neurol 2010; 223:166-72. [DOI: 10.1016/j.expneurol.2009.06.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2009] [Revised: 05/20/2009] [Accepted: 06/02/2009] [Indexed: 01/08/2023]
|
14
|
|
15
|
Sulaiman OAR, Gordon T. Role of chronic Schwann cell denervation in poor functional recovery after nerve injuries and experimental strategies to combat it. Neurosurgery 2010; 65:A105-14. [PMID: 19927054 DOI: 10.1227/01.neu.0000358537.30354.63] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVE To present our data about the role of chronic denervation (CD) of the distal nerve stumps as compared with muscle denervation atrophy and experimental strategies to promote better functional recovery. METHODS A rat model of nerve injury and repair was used. The common peroneal branch of the sciatic nerve was subjected to 0 to 24 weeks of CD before cross-suture with the tibial motoneurons. Our outcome measures included the numbers of motoneurons that regenerated their axons and the numbers that reinnervated muscle targets (motor units). To overcome the effects of CD, we used subcutaneous injection of FK506 and in vitro reactivation of Schwann cells that had been subjected to 24 weeks of CD with transforming growth factor beta. RESULTS Numbers of regenerated motoneurons and reinnervated motor units decreased as a function of duration of CD. However, axons that regenerated through the distal nerve stumps reinnervated the muscle targets and even formed enlarged motor unit size regardless of the duration of CD. FK506 doubled the numbers of tibial motoneurons that regenerated their axons into the common peroneal nerve even after delayed repair. Reactivation of chronically denervated Schwann cells with transforming growth factor beta significantly increased their capacity to support axonal regeneration. CONCLUSION CD of the distal nerve stumps is the primary factor that results in poor axonal regeneration and subsequently poor functional recovery. Acceleration of the rate of axonal regeneration and/or reactivation of Schwann cells of the distal nerve stumps are effective experimental strategies to promote axonal regeneration and functional recovery.
Collapse
Affiliation(s)
- Olawale A R Sulaiman
- Department of Neurosurgery, Spine Center, Ochsner Clinic Foundation, New Orleans, Louisiana 70121, USA.
| | | |
Collapse
|
16
|
Monje PV, Rendon S, Athauda G, Bates M, Wood PM, Bunge MB. Non-antagonistic relationship between mitogenic factors and cAMP in adult Schwann cell re-differentiation. Glia 2009; 57:947-61. [PMID: 19053056 PMCID: PMC2829776 DOI: 10.1002/glia.20819] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The expression of myelination-associated genes (MGs) can be induced by cyclic adenosine monophosphate (cAMP) elevation in isolated Schwann cells (SCs). To further understand the effect of known SC mitogens in the regulation of SC differentiation, we studied the response of SCs isolated from adult nerves to combined cAMP, growth factors, including neuregulin, and serum. In adult SCs, the induction of MGs by cAMP coincided with the loss of genes expressed in non-myelin-forming SCs and with a change in cell morphology from a bipolar to an expanded epithelial-like shape. Prolonged treatment with high doses of cAMP-stimulating agents, as well as low cell density, was required for the induction of SC differentiation. Stimulation with serum, neuregulin alone, or other growth factors including PDGF, IGF and FGF, increased SC proliferation but did not induce the expression of MGs or the associated morphological change. Most importantly, when these factors were administered in combination with cAMP-stimulating agents, SC proliferation was synergistically increased without reducing the differentiating activity of cAMP. Even though the initiation of DNA synthesis and the induction of differentiation were mostly incompatible events in individual cells, SCs were able to differentiate under conditions that also supported active proliferation. Overall, the results indicate that in the absence of neurons, cAMP can trigger SC re-differentiation concurrently with, but independently of, growth factor signaling.
Collapse
Affiliation(s)
- Paula V Monje
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida, USA.
| | | | | | | | | | | |
Collapse
|
17
|
Woodhoo A, Alonso MBD, Droggiti A, Turmaine M, D'Antonio M, Parkinson DB, Wilton DK, Al-Shawi R, Simons P, Shen J, Guillemot F, Radtke F, Meijer D, Feltri ML, Wrabetz L, Mirsky R, Jessen KR. Notch controls embryonic Schwann cell differentiation, postnatal myelination and adult plasticity. Nat Neurosci 2009; 12:839-47. [PMID: 19525946 PMCID: PMC2782951 DOI: 10.1038/nn.2323] [Citation(s) in RCA: 258] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2009] [Accepted: 03/30/2009] [Indexed: 11/09/2022]
Abstract
Notch signaling is central to vertebrate development, and analysis of Notch has provided important insights into pathogenetic mechanisms in the CNS and many other tissues. However, surprisingly little is known about the role of Notch in the development and pathology of Schwann cells and peripheral nerves. Using transgenic mice and cell cultures, we found that Notch has complex and extensive regulatory functions in Schwann cells. Notch promoted the generation of Schwann cells from Schwann cell precursors and regulated the size of the Schwann cell pool by controlling proliferation. Notch inhibited myelination, establishing that myelination is subject to negative transcriptional regulation that opposes forward drives such as Krox20. Notably, in the adult, Notch dysregulation resulted in demyelination; this finding identifies a signaling pathway that induces myelin breakdown in vivo. These findings are relevant for understanding the molecular mechanisms that control Schwann cell plasticity and underlie nerve pathology, including demyelinating neuropathies and tumorigenesis.
Collapse
Affiliation(s)
- Ashwin Woodhoo
- Department of Cell and Developmental Biology, University College London, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Freidin M, Asche S, Bargiello TA, Bennett MVL, Abrams CK. Connexin 32 increases the proliferative response of Schwann cells to neuregulin-1 (Nrg1). Proc Natl Acad Sci U S A 2009; 106:3567-72. [PMID: 19218461 PMCID: PMC2651262 DOI: 10.1073/pnas.0813413106] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2008] [Indexed: 11/18/2022] Open
Abstract
Connexin 32 (Cx32), a gap junction protein, is found within the para-nodal region and Schmidt-Lanterman incisures of myelinating Schwann cells (SCs). In developing and regenerating peripheral nerves, pro-myelinating SCs express Cx32 mRNA and protein in conjunction with the expression of myelin specific genes. Neuregulin-1 (Nrg1), a member of the neuregulin family of growth factors, controls SC proliferation and differentiation depending on the cellular environment and the particular stage of SC maturation. Primary cultures of purified SCs from newborn mouse sciatic nerve were used to characterize both the role of Nrg1 in the expression of Cx32 and, conversely, the role of Cx32 in SC responsiveness to Nrg1. Glial growth factor 2, an isoform of Nrg1, up-regulated Cx32 in both proliferating and non-proliferating SCs. However, SCs from Cx32-KO mice exhibited a significantly smaller mitogenic response to glial growth factor 2. Electrical coupling between Cx32-KO SCs did not differ from that between WT SCs, indicating the presence of other connexins. These results suggest a link between Cx32 expression and Nrg1 regulation of SC proliferation that does not involve Cx32-mediated intercellular communication.
Collapse
Affiliation(s)
| | | | | | | | - Charles K. Abrams
- Department of Neurology and
- Department of Physiology and Pharmacology, State University of New York Downstate at Brooklyn, Brooklyn NY 10021; and
| |
Collapse
|
19
|
Lee HK, Seo IA, Suh DJ, Hong JI, Yoo YH, Park HT. Interleukin-6 is required for the early induction of glial fibrillary acidic protein in Schwann cells during Wallerian degeneration. J Neurochem 2009; 108:776-86. [DOI: 10.1111/j.1471-4159.2008.05826.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
20
|
Grothe C, Jungnickel J, Haastert K. Physiological role of basic FGF in peripheral nerve development and regeneration: potential for reconstruction approaches. FUTURE NEUROLOGY 2008. [DOI: 10.2217/14796708.3.5.605] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
According to expression studies and functional analyses in mutant mice and in rats, FGF-2 appears to be specifically involved during development of peripheral nerves and in de-/re-generating processes at the lesion site and in spinal ganglia. In the absence of FGF receptor (FGFR)3, axonal and myelin diameters of peripheral nerves are significantly reduced, suggesting that FGFR3 physiologically regulates axonal development. The normally occurring neuronal cell death in spinal ganglia after peripheral nerve axotomy does not take place in FGF-2 and FGFR3-deleted mice, respectively, suggesting that injury-induced apoptosis is mediated via FGF-2 binding to FGFR3. According to a bimodal function of FGF-2, lesion-induced neuron death in rat spinal ganglia can be prevented by application of FGF-2 to the proximal nerve stump, which could be mediated via FGFR1/2. At the lesion site, FGF-2 appears to be involved in stimulating Schwann cell proliferation, promoting neurite outgrowth, especially of sensory nerve fibers, and regulating remyelination.
Collapse
Affiliation(s)
- Claudia Grothe
- Hannover Medical School, Institute of Neuroanatomy, OE 4140, Carl-Neuberg Str. 1, D-30625, Hannover, Germany
| | - Julia Jungnickel
- Hannover Medical School, Institute of Neuroanatomy, OE 4140, Carl-Neuberg Str. 1, D-30625, Hannover, Germany
| | - Kirsten Haastert
- Hannover Medical School, Institute of Neuroanatomy, OE 4140, Carl-Neuberg Str. 1, D-30625, Hannover, Germany
| |
Collapse
|
21
|
Parkinson DB, Bhaskaran A, Arthur-Farraj P, Noon LA, Woodhoo A, Lloyd AC, Feltri ML, Wrabetz L, Behrens A, Mirsky R, Jessen KR. c-Jun is a negative regulator of myelination. J Cell Biol 2008; 181:625-37. [PMID: 18490512 PMCID: PMC2386103 DOI: 10.1083/jcb.200803013] [Citation(s) in RCA: 314] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2008] [Accepted: 04/17/2008] [Indexed: 12/29/2022] Open
Abstract
Schwann cell myelination depends on Krox-20/Egr2 and other promyelin transcription factors that are activated by axonal signals and control the generation of myelin-forming cells. Myelin-forming cells remain remarkably plastic and can revert to the immature phenotype, a process which is seen in injured nerves and demyelinating neuropathies. We report that c-Jun is an important regulator of this plasticity. At physiological levels, c-Jun inhibits myelin gene activation by Krox-20 or cyclic adenosine monophosphate. c-Jun also drives myelinating cells back to the immature state in transected nerves in vivo. Enforced c-Jun expression inhibits myelination in cocultures. Furthermore, c-Jun and Krox-20 show a cross-antagonistic functional relationship. c-Jun therefore negatively regulates the myelinating Schwann cell phenotype, representing a signal that functionally stands in opposition to the promyelin transcription factors. Negative regulation of myelination is likely to have significant implications for three areas of Schwann cell biology: the molecular analysis of plasticity, demyelinating pathologies, and the response of peripheral nerves to injury.
Collapse
Affiliation(s)
- David B Parkinson
- Department of Anatomy and Developmental Biology, University College London, London WC1E 6BT, England, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Zhu TS, Glaser M. Regulatory role of cytochrome P450scc and pregnenolone in myelination by rat Schwann cells. Mol Cell Biochem 2008; 313:79-89. [PMID: 18373277 DOI: 10.1007/s11010-008-9745-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2007] [Accepted: 03/13/2008] [Indexed: 11/26/2022]
Abstract
To investigate the production of steroid hormones by Schwann cells and to examine the regulation of steroid hormone production during myelination, cultures of rat Schwann cells were differentiated into their myelinating phenotype in the absence of neurons with dibutyryl cAMP (db-cAMP). During this process, the expression of P450scc (involved in steroid biosynthesis) was elevated at both the mRNA and protein levels as evident in RT-PCR, Western blots, and immunostaining. Labeling of the cells with [14C] acetate revealed enhanced production of pregnenolone during differentiation into the myelinating phenotype. Disruption of P450scc's activity with an inhibitor diminished the extent of differentiation into the myelinating phenotype as levels of mRNA and protein expression of myelin protein zero (P0) declined. However, the effect was reversed with the addition of pregnenolone. Furthermore, when the differentiating cultures were treated with pregnenolone, mRNA expression of P0 was upregulated, suggesting the stimulation of the differentiation process. Together, these results provide evidence for Schwann cells as a major producer of steroid hormones and pregnenolone production by P450scc as an important regulatory step during myelination.
Collapse
Affiliation(s)
- Thant S Zhu
- Department of Biochemistry, University of Illinois, 600 S. Mathews Avenue, Urbana, IL 61801, USA
| | | |
Collapse
|
23
|
Grothe C, Claus P, Haastert K, Lutwak E, Ron D. Expression and regulation of Sef, a novel signaling inhibitor of receptor tyrosine kinases-mediated signaling in the nervous system. Acta Histochem 2008; 110:155-62. [PMID: 17980404 DOI: 10.1016/j.acthis.2007.08.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2007] [Revised: 08/06/2007] [Accepted: 08/23/2007] [Indexed: 10/22/2022]
Abstract
Fibroblast growth factors (FGFs) signal via four distinct high affinity cell surface tyrosine kinase receptors, termed FGFR1-FGFR4 (FGFR-FGF-receptor). Recently, a new modulator of the FGF signaling pathway, the transmembrane protein 'similar expression to FGF genes' (Sef), has been identified in zebrafish and subsequently in mammals. Sef from mouse and human inhibits FGF mitogenic activity. In the present study, we analyzed the expression of Sef in distinct rat brain areas, in the spinal cord and in peripheral nerves and spinal ganglia using semi-quantitative RT-PCR. Furthermore, we studied the cellular expression pattern of Sef in intact spinal ganglia and sciatic nerves and, in addition, after crush lesion, using in situ hybridization and immunohistochemistry. Sef transcripts were expressed in all brain areas evaluated and in the spinal cord. A neuronal expression was found in both intact and injured spinal ganglia. Intact sciatic nerves, however, showed little or no Sef expression. Seven days after injury, high Sef expression was concentrated to the crush site, and Schwann cells seemed to be the source of Sef. The labeling pattern of up-regulated Sef was complementary to the patterns of FGF-2 and FGFR1-3, which were localized proximal and distal to the crush site. These results suggest an involvement of Sef during the nerve regeneration process, possibly by fine-tuning the effects of FGF signaling.
Collapse
|
24
|
Abstract
Whereas the central nervous system (CNS) usually cannot regenerate, peripheral nerves regenerate spontaneously after injury because of a permissive environment and activation of the intrinsic growth capacity of neurons. Functional regeneration requires axon regrowth and remyelination of the regenerated axons by Schwann cells. Multiple factors including neurotrophic factors, extracellular matrix (ECM) proteins, and hormones participate in Schwann cell dedifferentiation, proliferation, and remyelination. We describe the current understanding of peripheral axon regeneration and focus on the molecules and potential mechanisms involved in remyelination.
Collapse
Affiliation(s)
- Zu-Lin Chen
- Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY 10021, USA.
| | | | | |
Collapse
|
25
|
Pearse DD, Sanchez AR, Pereira FC, Andrade CM, Puzis R, Pressman Y, Golden K, Kitay BM, Blits B, Wood PM, Bunge MB. Transplantation of Schwann cells and/or olfactory ensheathing glia into the contused spinal cord: Survival, migration, axon association, and functional recovery. Glia 2007; 55:976-1000. [PMID: 17526000 DOI: 10.1002/glia.20490] [Citation(s) in RCA: 226] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Schwann cells (SCs) and olfactory ensheathing glia (OEG) have shown promise for spinal cord injury repair. We sought their in vivo identification following transplantation into the contused adult rat spinal cord at 1 week post-injury by: (i) DNA in situ hybridization (ISH) with a Y-chromosome specific probe to identify male transplants in female rats and (ii) lentiviral vector-mediated expression of EGFP. Survival, migration, and axon-glia association were quantified from 3 days to 9 weeks post-transplantation. At 3 weeks after transplantation into the lesion, a 60-90% loss of grafted cells was observed. OEG-only grafts survived very poorly within the lesion (<5%); injection outside the lesion led to a 60% survival rate, implying that the injury milieu was hostile to transplanted cells and or prevented their proliferation. At later times post-grafting, p75(+)/EGFP(-) cells in the lesion outnumbered EGFP(+) cells in all paradigms, evidence of significant host SC infiltration. SCs and OEG injected into the injury failed to migrate from the lesion. Injection of OEG outside of the injury resulted in their migration into the SC-injected injury site, not via normal-appearing host tissue but along the pia or via the central canal. In all paradigms, host axons were seen in association with or ensheathed by transplanted glia. Numerous myelinated axons were found within regions of grafted SCs but not OEG. The current study details the temporal survival, migration, axon association of SCs and OEG, and functional recovery after grafting into the contused spinal cord, research previously complicated due to a lack of quality, long-term markers for cell tracking in vivo.
Collapse
Affiliation(s)
- Damien D Pearse
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Grothe C, Haastert K, Jungnickel J. Physiological function and putative therapeutic impact of the FGF-2 system in peripheral nerve regeneration—Lessons from in vivo studies in mice and rats. ACTA ACUST UNITED AC 2006; 51:293-9. [PMID: 16430964 DOI: 10.1016/j.brainresrev.2005.12.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2005] [Accepted: 12/07/2005] [Indexed: 11/23/2022]
Abstract
Diffusible and substratum-bound molecules regulate development and regeneration of the peripheral nervous system. The understanding of physiological function of these factors could have an impact on the development of new therapeutic strategies to stimulate nerve regeneration across long gaps. Within the group of trophic factors, basic fibroblast growth factor (FGF-2) and its high-affinity receptors are expressed in the intact peripheral nervous system and regulated following nerve injury. After exogenous application, FGF-2 promotes neuronal survival and neurite outgrowth in vitro and in vivo. In this review, animal studies on the physiological role of the endogenous FGF-2 system and the regenerative capacity after exogenous FGF-2 administration are summarized. The concept of FGF-2 function is discussed in context with other growth factors that are also physiologically relevant in the peripheral nervous system. Studies of sciatic nerve axotomy in FGF-2- and FGF receptor (R) 3-deleted mice, respectively, strongly suggested that FGF-2 binding to FGFR3 is involved in injury-induced neuronal apoptosis. At the lesion site, inhibition of myelination and stimulation of Schwann cell proliferation by FGF-2 via FGFR1/2 is suggested from rat and mouse studies, whereas neurite formation is very likely enhanced via FGFR3 activation. Additionally to these demonstrated physiological functions of endogenous FGF-2, administration of FGF-2 isoforms in the rat model of nerve regeneration across long gaps revealed a role of the high molecular weight isoforms of FGF-2 on sensory recovery. Within the group of physiologically relevant trophic factors, the FGF-2 system seems to be crucially involved in the scenario of peripheral nerve development and regeneration.
Collapse
Affiliation(s)
- Claudia Grothe
- Hannover Medical School, Department of Neuroanatomy, Hannover, Germany.
| | | | | |
Collapse
|
27
|
D'Antonio M, Michalovich D, Paterson M, Droggiti A, Woodhoo A, Mirsky R, Jessen KR. Gene profiling and bioinformatic analysis of Schwann cell embryonic development and myelination. Glia 2006; 53:501-15. [PMID: 16369933 DOI: 10.1002/glia.20309] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
To elucidate the molecular mechanisms involved in Schwann cell development, we profiled gene expression in the developing and injured rat sciatic nerve. The genes that showed significant changes in expression in developing and dedifferentiated nerve were validated with RT-PCR, in situ hybridisation, Western blot and immunofluorescence. A comprehensive approach to annotating micro-array probes and their associated transcripts was performed using Biopendium, a database of sequence and structural annotation. This approach significantly increased the number of genes for which a functional insight could be found. The analysis implicates agrin and two members of the collapsin response-mediated protein (CRMP) family in the switch from precursors to Schwann cells, and synuclein-1 and alphaB-crystallin in peripheral nerve myelination. We also identified a group of genes typically related to chondrogenesis and cartilage/bone development, including type II collagen, that were expressed in a manner similar to that of myelin-associated genes. The comprehensive function annotation also identified, among the genes regulated during nerve development or after nerve injury, proteins belonging to high-interest families, such as cytokines and kinases, and should therefore provide a uniquely valuable resource for future research.
Collapse
Affiliation(s)
- Maurizio D'Antonio
- Department of Anatomy and Developmental Biology, University College London, London, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
28
|
ROSENBAUM THORSTEN, KIM HAESUNA, BOISSY YINGL, LING BO, RATNER NANCY. Neurofibromin, the Neurofibromatosis Type 1 Ras-GAP, Is Required for Appropriate P0Expression and Myelination. Ann N Y Acad Sci 2006; 883:203-214. [DOI: 10.1111/j.1749-6632.1999.tb08583.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
29
|
Jungnickel J, Haase K, Konitzer J, Timmer M, Grothe C. Faster nerve regeneration after sciatic nerve injury in mice over-expressing basic fibroblast growth factor. ACTA ACUST UNITED AC 2006; 66:940-8. [PMID: 16758491 DOI: 10.1002/neu.20265] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Basic fibroblast growth factor (FGF-2) is expressed in the peripheral nervous system and is up-regulated after nerve lesion. It has been demonstrated that administration of FGF-2 protects neurons from injury-induced cell death and promotes axonal regrowth. Using transgenic mice over-expressing FGF-2 (TgFGF-2), we addressed the importance of endogenously generated FGF-2 on sensory neuron loss and sciatic nerve regeneration. After sciatic nerve transection, wild-type and transgenic mice showed the same degree of cell death in L5 spinal ganglia. Also, the number of chromatolytic, eccentric, and pyknotic sensory neurons was not changed under elevated levels of FGF-2. Morphometric evaluation of intact nerves from TgFGF-2 mice revealed no difference in number and size of myelinated fibers compared to wild-type mice. One week after crush injury, the number of regenerated axons was doubled and the myelin thickness was significantly smaller in transgenic mice. After 2 and 4 weeks, morphometric analysis and functional tests revealed no differences in recovery of sensory and motor nerve fibers. To study the role of FGF-2 over-expression on Schwann cell proliferation during the early regeneration process, we used BrdU-labeling to mark dividing cells. In transgenic mice, the number of proliferating cells was significantly increased distal to the crush site compared to wild-types. We propose that endogenously synthesized FGF-2 influences early peripheral nerve regeneration by regulating Schwann cell proliferation, axonal regrowth, and remyelination.
Collapse
Affiliation(s)
- Julia Jungnickel
- Department of Neuroanatomy OE 4140, Center of Anatomy, Hannover Medical School, D-30623 Hannover, Germany.
| | | | | | | | | |
Collapse
|
30
|
Macica CM, Liang G, Lankford KL, Broadus AE. Induction of parathyroid hormone-related peptide following peripheral nerve injury: Role as a modulator of Schwann cell phenotype. Glia 2006; 53:637-48. [PMID: 16470617 DOI: 10.1002/glia.20319] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Parathyroid hormone-related peptide (PTHrP) is widely distributed in the rat nervous system, including the peripheral nervous system, where its function is unknown. PTHrP mRNA expression has recently been shown to be significantly elevated following axotomy of sympathetic ganglia, although the role of PTHrP was not investigated. The role of PTHrP in peripheral nerve injury was investigated in this study using the sciatic nerve injury model and dorsal root ganglion (DRG) explant model of nerve regeneration. We find that PTHrP is a constitutively secreted peptide of proliferating Schwann cells and that the PTHrP receptor (PTH1R) mRNA is expressed in isolated DRG and in sciatic nerve. Using the sciatic nerve injury model, we show that PTHrP is significantly upregulated in DRG and in sciatic nerve. In addition, in situ hybridization revealed significant localization of PTHrP mRNA to Schwann cells in the injured sciatic nerve. We also find that PTHrP causes a dramatic increase in the number of Schwann cells that align with and bundle regrowing axons in explants, characteristic of immature, dedifferentiated Schwann cells. In addition to stimulating migration of Schwann cells along the axonal membrane, PTHrP also stimulates migration on a type 1 collagen matrix. Furthermore, treatment of purified Schwann cell cultures with PTHrP results in the rapid phosphorylation of the cAMP response element protein, CREB. We propose that PTHrP acts by promoting the dedifferentiation of Schwann cells, a critical requirement for successful nerve regeneration and an effect consistent with known PTHrP functions in other cellular differentiation programs.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Cell Differentiation/drug effects
- Cell Differentiation/physiology
- Cell Proliferation/drug effects
- Cells, Cultured
- Cyclic AMP Response Element-Binding Protein/metabolism
- Disease Models, Animal
- Ganglia, Spinal/cytology
- Ganglia, Spinal/injuries
- Ganglia, Spinal/metabolism
- Growth Cones/metabolism
- Ligation
- Mice
- Nerve Regeneration/drug effects
- Nerve Regeneration/physiology
- Neurons, Afferent/cytology
- Neurons, Afferent/metabolism
- Parathyroid Hormone-Related Protein/metabolism
- Parathyroid Hormone-Related Protein/pharmacology
- Peripheral Nerve Injuries
- Peripheral Nerves/cytology
- Peripheral Nerves/metabolism
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptor, Parathyroid Hormone, Type 1/genetics
- Schwann Cells/cytology
- Schwann Cells/drug effects
- Schwann Cells/metabolism
- Sciatic Nerve/cytology
- Sciatic Nerve/injuries
- Sciatic Nerve/metabolism
- Sciatic Neuropathy/metabolism
- Sciatic Neuropathy/physiopathology
- Up-Regulation/drug effects
- Up-Regulation/physiology
Collapse
Affiliation(s)
- Carolyn M Macica
- Department of Internal Medicine, Division of Endocrinology, Yale University School of Medicine, New Haven, CT 06520-8020, USA.
| | | | | | | |
Collapse
|
31
|
Haastert K, Lipokatic E, Fischer M, Timmer M, Grothe C. Differentially promoted peripheral nerve regeneration by grafted Schwann cells over-expressing different FGF-2 isoforms. Neurobiol Dis 2005; 21:138-53. [PMID: 16122933 DOI: 10.1016/j.nbd.2005.06.020] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2005] [Revised: 06/27/2005] [Accepted: 06/27/2005] [Indexed: 10/25/2022] Open
Abstract
Artificial nerve grafts are needed to reconstruct massive defects in the peripheral nervous system when autologous nerve grafts are not available in sufficient amounts. Nerve grafts containing Schwann cells display a suitable substrate for long-distance regeneration. We present here a comprehensive analysis of the in vivo effects of different isoforms of fibroblast growth factor-2 (FGF-2) on peripheral nerve regeneration across long gaps. FGF-2 isoforms were provided by grafted, genetically modified Schwann cells over-expressing 18-kDa-FGF-2 and 21-/23-kDa-FGF-2, respectively. Functional tests evaluated motor and sensory recovery. Additionally, morphometrical analyses of regenerated nerves were performed 3 and 6 months after grafting. Distinct regeneration promoting effects of the different FGF-2 isoforms were found. 18-kDa-FGF-2 mediated inhibitory effects on the grade of myelination of regenerating axons, whereas 21-/23-kDa-FGF-2 mediated early recovery of sensory functions and stimulation of long-distance myelination of regenerating axons. The results contribute to the development of new therapeutic strategies in peripheral nerve repair.
Collapse
Affiliation(s)
- Kirsten Haastert
- Department of Neuroanatomy, Hannover Medical School, OE 4140, Carl-Neuberg-Str.1, 30625 Hannover, Germany.
| | | | | | | | | |
Collapse
|
32
|
Dore JJ, Crotty KL, Birren SJ. Inhibition of glial maturation by bone morphogenetic protein 2 in a neural crest-derived cell line. Dev Neurosci 2005; 27:37-48. [PMID: 15886483 DOI: 10.1159/000084531] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2004] [Accepted: 11/05/2004] [Indexed: 12/23/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) regulate developmental decisions in many neural and nonneural lineages. BMPs influence both CNS neuronal and glial development and promote neuronal differentiation in neural crest derivatives. We investigated the actions of BMP2 on glial differentiation in the peripheral nervous system using NCM1 cells, a neural crest-derived cell line with the properties of peripheral glial precursor cells. BMP2 prevented the acquisition of a mature Schwann cell-like morphology, blocking the expression of mature genes and maintaining expression of several early glial markers. We provide evidence that BMP2 activates the GFAP promoter and define signaling pathways underlying this regulation. Our results demonstrate a novel role for BMPs as inhibitors of glial differentiation in the peripheral nervous system and suggest that BMPs may regulate the developmental timing of glial maturation.
Collapse
Affiliation(s)
- Justin J Dore
- Department of Biology, Volen Center for Complex Systems, Brandeis University Waltham, Waltham, MA 02454, USA
| | | | | |
Collapse
|
33
|
Robinson M, Parsons Perez MC, Tébar L, Palmer J, Patel A, Marks D, Sheasby A, De Felipe C, Coffin R, Livesey FJ, Hunt SP. FLRT3 is expressed in sensory neurons after peripheral nerve injury and regulates neurite outgrowth. Mol Cell Neurosci 2005; 27:202-14. [PMID: 15485775 DOI: 10.1016/j.mcn.2004.06.008] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2004] [Revised: 06/10/2004] [Accepted: 06/15/2004] [Indexed: 11/23/2022] Open
Abstract
We used a molecular screen to identify genes upregulated in regenerating adult rat dorsal root ganglion cells. FLRT3 mRNA and protein characterized by a fibronectin type III domain and a leucine-rich repeat motif was upregulated in damaged sensory neurons. The protein was then transported into their peripheral and central processes where the FLRT3 protein was localized to presynaptic axon terminals. In vitro, the FLRT3 protein was expressed at the cell surface, regulated neurite outgrowth in sensory neurons, but did not exhibit homophilic binding. FLRT3 was widely expressed in the developing embryo, particularly in the central nervous system and somites. However, in the adult, we found no evidence for accumulation or reexpression of the FLRT3 protein in damaged axons of the central nervous system. We conclude that FLRT3 codes for a putative cell surface receptor implicated in both the development of the nervous system and in the regeneration of the peripheral nervous system (PNS).
Collapse
Affiliation(s)
- M Robinson
- Department of Anatomy and Developmental Biology, UCL, London, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Studzinski DM, Benjamins JA. Expression of P0 glycoprotein in CNS glia: Effects of overexpression in N20.1 cells. Glia 2005; 52:234-44. [PMID: 15968635 DOI: 10.1002/glia.20240] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
To examine effects of expression of the PNS myelin P0 glycoprotein in glial cells of CNS lineage, we transfected murine N20.1 glial cells with a rat P0 cDNA. A stably transfected cell line expressing high levels of P0 message showed P0 immunostaining, along with changes in morphology. Polymerase chain reaction (PCR) identified the predicted rat P0 sequence in the transfected N20.1 cells and further revealed low levels of mouse P0 message in the nontransfected cells and in primary mouse astrocytes. This is the first evidence of endogenous expression of message for P0 glycoprotein in CNS glia. Quantitative RT-PCR confirmed the expression of rat P0 mRNA in the transfected N20.1 cells, at levels about 400 times greater than murine P0 in nontransfected cells. A 27-kD band was detected in the transfected cells by Western blot with P0 antibody, but not in mock-transfected or nontransfected N20.1 cells. Immunocytochemistry following permeabilization showed intracellular vesicular localization of P0 in the cytoplasm and perinuclear rings in transfected cells, with a similar pattern but much lower levels in nontransfected cells. Faint surface staining for P0 protein without permeabilization was seen only on the transfected cells. A few transfected cells with membrane sheets stained more intensely for surface P0. Quantitative RT-PCR was used to determine if P0 overexpression altered expression of other myelin-related genes compared with glial fibrillary acidic protein (GFAP); the ratios of myelin basic protein (MBP)/GFAP and proteolipid protein (PLP)/GFAP were increased 2- to 3-fold in the P0-transfected cells. We conclude that P0 overexpression alters N20.1 gene expression and cell morphology, and shifts the cells from astroglial to oligodendroglial phenotype.
Collapse
|
35
|
Parkinson DB, Bhaskaran A, Droggiti A, Dickinson S, D'Antonio M, Mirsky R, Jessen KR. Krox-20 inhibits Jun-NH2-terminal kinase/c-Jun to control Schwann cell proliferation and death. ACTA ACUST UNITED AC 2004; 164:385-94. [PMID: 14757751 PMCID: PMC2172235 DOI: 10.1083/jcb.200307132] [Citation(s) in RCA: 188] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The transcription factor Krox-20 controls Schwann cell myelination. Schwann cells in Krox-20 null mice fail to myelinate, and unlike myelinating Schwann cells, continue to proliferate and are susceptible to death. We find that enforced Krox-20 expression in Schwann cells cell-autonomously inactivates the proliferative response of Schwann cells to the major axonal mitogen β–neuregulin-1 and the death response to TGFβ or serum deprivation. Even in 3T3 fibroblasts, Krox-20 not only blocks proliferation and death but also activates the myelin genes periaxin and protein zero, showing properties in common with master regulatory genes in other cell types. Significantly, a major function of Krox-20 is to suppress the c-Jun NH2-terminal protein kinase (JNK)–c-Jun pathway, activation of which is required for both proliferation and death. Thus, Krox-20 can coordinately control suppression of mitogenic and death responses. Krox-20 also up-regulates the scaffold protein JNK-interacting protein 1 (JIP-1). We propose this as a possible component of the mechanism by which Krox-20 regulates JNK activity during Schwann cell development.
Collapse
Affiliation(s)
- David B Parkinson
- Department of Anatomy and Developmental Biology, University College London, Gower Street, London, WC1E 6BT UK.
| | | | | | | | | | | | | |
Collapse
|
36
|
Atanasoski S, Notterpek L, Lee HY, Castagner F, Young P, Ehrengruber MU, Meijer D, Sommer L, Stavnezer E, Colmenares C, Suter U. The Protooncogene Ski Controls Schwann Cell Proliferation and Myelination. Neuron 2004; 43:499-511. [PMID: 15312649 DOI: 10.1016/j.neuron.2004.08.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2004] [Revised: 06/29/2004] [Accepted: 07/28/2004] [Indexed: 01/11/2023]
Abstract
Schwann cell proliferation and subsequent differentiation to nonmyelinating and myelinating cells are closely linked processes. Elucidating the molecular mechanisms that control these events is key to the understanding of nerve development, regeneration, nerve-sheath tumors, and neuropathies. We define the protooncogene Ski, an inhibitor of TGF-beta signaling, as an essential component of the machinery that controls Schwann cell proliferation and myelination. Functional Ski overexpression inhibits TGF-beta-mediated proliferation and prevents growth-arrested Schwann cells from reentering the cell cycle. Consistent with these findings, myelinating Schwann cells upregulate Ski during development and remyelination after injury. Myelination is blocked in myelin-competent cultures derived from Ski-deficient animals, and genes encoding myelin components are downregulated in Ski-deficient nerves. Conversely, overexpression of Ski in Schwann cells causes an upregulation of myelin-related genes. The myelination-regulating transcription factor Oct6 is involved in a complex modulatory relationship with Ski. We conclude that Ski is a crucial signal in Schwann cell development and myelination.
Collapse
Affiliation(s)
- Suzana Atanasoski
- Institute of Cell Biology, Department of Biology, Swiss Federal Institute of Technology, ETH-Hönggerberg, Zurich, Switzerland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Pearse DD, Pereira FC, Marcillo AE, Bates ML, Berrocal YA, Filbin MT, Bunge MB. cAMP and Schwann cells promote axonal growth and functional recovery after spinal cord injury. Nat Med 2004; 10:610-6. [PMID: 15156204 DOI: 10.1038/nm1056] [Citation(s) in RCA: 539] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2004] [Accepted: 05/10/2004] [Indexed: 12/14/2022]
Abstract
Central neurons regenerate axons if a permissive environment is provided; after spinal cord injury, however, inhibitory molecules are present that make the local environment nonpermissive. A promising new strategy for inducing neurons to overcome inhibitory signals is to activate cAMP signaling. Here we show that cAMP levels fall in the rostral spinal cord, sensorimotor cortex and brainstem after spinal cord contusion. Inhibition of cAMP hydrolysis by the phosphodiesterase IV inhibitor rolipram prevents this decrease and when combined with Schwann cell grafts promotes significant supraspinal and proprioceptive axon sparing and myelination. Furthermore, combining rolipram with an injection of db-cAMP near the graft not only prevents the drop in cAMP levels but increases them above those in uninjured controls. This further enhances axonal sparing and myelination, promotes growth of serotonergic fibers into and beyond grafts, and significantly improves locomotion. These findings show that cAMP levels are key for protection, growth and myelination of injured CNS axons in vivo and recovery of function.
Collapse
Affiliation(s)
- Damien D Pearse
- The Miami Project to Cure Paralysis, University of Miami School of Medicine, 1095 NW 14th Terrace, Miami, Florida 33136, USA.
| | | | | | | | | | | | | |
Collapse
|
38
|
Jungnickel J, Gransalke K, Timmer M, Grothe C. Fibroblast growth factor receptor 3 signaling regulates injury-related effects in the peripheral nervous system. Mol Cell Neurosci 2004; 25:21-9. [PMID: 14962737 DOI: 10.1016/j.mcn.2003.09.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2003] [Revised: 09/16/2003] [Accepted: 09/29/2003] [Indexed: 12/22/2022] Open
Abstract
Fibroblast growth factor receptor (FGFR) signaling is crucial for neural development and regeneration. Here we investigated the L5 spinal ganglion and the sciatic nerve of intact Fgfr3-deficient mice after nerve injury. Quantification of sensory neurons in the L5 spinal ganglion revealed no significant differences between wild-type and Fgfr3-deficient mice. Seven days after nerve lesion, the normally occurring neuron loss in wild-type mice was not found in Fgfr3-deficient animals, suggesting that FGFR3 signaling is involved in the cell death process. Morphometric analysis of the sciatic nerve showed similar numbers of myelinated axons, but the axonal and myelin diameter was significantly smaller in Fgfr3-deficient mice compared to the wild types. Evaluation of regenerating myelinated axons of the sciatic nerve revealed no differences between both mouse strains 7 days after crush injury. Our results suggest that FGFR3 signaling seems to be involved in processes of damage-induced neuron death and axonal development.
Collapse
MESH Headings
- Animals
- Cell Death/physiology
- Denervation
- Disease Models, Animal
- Ganglia, Spinal/metabolism
- Ganglia, Spinal/pathology
- Ganglia, Spinal/physiopathology
- Growth Cones/metabolism
- Growth Cones/ultrastructure
- Lumbar Vertebrae/metabolism
- Mice
- Mice, Knockout
- Microscopy, Electron
- Nerve Degeneration/metabolism
- Nerve Degeneration/pathology
- Nerve Degeneration/physiopathology
- Nerve Fibers, Myelinated/metabolism
- Nerve Fibers, Myelinated/pathology
- Nerve Fibers, Myelinated/ultrastructure
- Nerve Regeneration/physiology
- Neurons, Afferent/metabolism
- Neurons, Afferent/pathology
- Neurons, Afferent/ultrastructure
- Peripheral Nerve Injuries
- Peripheral Nerves/metabolism
- Peripheral Nerves/physiopathology
- Protein-Tyrosine Kinases
- Receptor, Fibroblast Growth Factor, Type 3
- Receptors, Fibroblast Growth Factor/deficiency
- Receptors, Fibroblast Growth Factor/genetics
- Receptors, Fibroblast Growth Factor/physiology
- Sciatic Nerve/metabolism
- Sciatic Nerve/pathology
- Sciatic Nerve/physiopathology
- Sciatic Neuropathy/metabolism
- Sciatic Neuropathy/pathology
- Sciatic Neuropathy/physiopathology
- Signal Transduction/physiology
Collapse
Affiliation(s)
- Julia Jungnickel
- Department of Neuroanatomy OE 4140, Center of Anatomy, Hannover Medical School, D-30623 Hannover, Germany.
| | | | | | | |
Collapse
|
39
|
Day WA, Koishi K, McLennan IS. Transforming growth factor beta 1 may regulate the stability of mature myelin sheaths. Exp Neurol 2004; 184:857-64. [PMID: 14769378 DOI: 10.1016/s0014-4886(03)00308-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2002] [Revised: 06/09/2003] [Accepted: 06/10/2003] [Indexed: 12/27/2022]
Abstract
The molecular mechanisms underlying peripheral neuropathies have only been partially elucidated. In particular, the regulatory factors that control the stability and turnover of mature myelin are largely unknown. Transforming growth factor beta 1 (TGF-beta1), and its associated receptors, are expressed by mature Schwann cells. On this basis, we postulated that TGF-beta1 may be an autocrine regulator of mature myelin. This hypothesis was tested by examining the ultrastructure of myelin in adult mice that have a null mutation of their TGF-beta1 gene. We report here that the myelin of these mice is grossly abnormal. At the nodes of Ranvier, the cytoplasmic collars of the Schwann cells were expanded and the myelin had a honeycomb appearance. Focal (tomacula-like) hypermyelin structures were observed in the internodal regions of a significant number of axons in mutant nerve, and were not observed in littermate controls. Axon diameters were within the normal range and no axonal pathology was evident in mutant nerve and macrophages were absent. Results imply that lack of TGF-beta1 may have a direct effect on Schwann cells. We suggest that TGF-beta1 may stabilise compact myelin via an autocrine mechanism.
Collapse
Affiliation(s)
- W Antony Day
- Arizona Research Laboratories, Department of Neuroscience, University of Arizona, Tucson, AZ, USA
| | | | | |
Collapse
|
40
|
Benmessahel Y, Troadec JD, Cadepond F, Guennoun R, Hales DB, Schumacher M, Groyer G. Downregulation of steroidogenic acute regulatory protein (StAR) gene expression by cyclic AMP in cultured Schwann cells. Glia 2004; 45:213-28. [PMID: 14730695 DOI: 10.1002/glia.10326] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Steroidogenic acute regulatory protein (StAR) plays a key role in the availability of cholesterol to the inner mitochondrial membrane, where the first step of steroidogenesis, its conversion to pregnenolone, takes place. Here, we demonstrate for the first time that the StAR gene is also expressed in the rat sciatic nerve and in cultured Schwann cells. The addition to the culture medium of the cAMP-elevating agent forskolin or of the cAMP analogue 8Br-cAMP produced a time-course extinction of StAR gene expression. An inverse relationship was demonstrated between StAR gene expression and the intracellular cAMP content. Accordingly, pharmacological inhibition of the activities of Schwann cell adenylyl cyclase or of phosphodiesterase IV resulted in modifications of StAR gene expression. Since StAR gene expression is stimulated by cAMP in classical steroidogenic cells, our work is the first demonstration of a negative regulation of StAR gene by cAMP.
Collapse
|
41
|
Jungnickel J, Claus P, Gransalke K, Timmer M, Grothe C. Targeted disruption of the FGF-2 gene affects the response to peripheral nerve injury. Mol Cell Neurosci 2004; 25:444-52. [PMID: 15033172 DOI: 10.1016/j.mcn.2003.11.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2003] [Revised: 11/06/2003] [Accepted: 11/18/2003] [Indexed: 01/03/2023] Open
Abstract
Basic fibroblast growth factor (FGF-2) is involved in the development, maintenance, and survival of the nervous system. To study the physiological role of endogenous FGF-2 during peripheral nerve regeneration, we analyzed sciatic nerves of FGF-2-deleted mice by using morphometric, morphological, and immunocytochemical methods. Quantification of number and size of myelinated axons in intact sciatic nerves revealed no difference between wild-type and FGF-2 knock-out (ko) animals. One week after nerve crush, FGF-2 ko mice showed about five times more regenerated myelinated axons with increased myelin and axon diameter in comparison to wild-types close to the injury site. In addition, quantitative distribution of macrophages and collapsed myelin profiles suggested faster Wallerian degeneration in FGF-2-deleted mice close to the lesion site. Our results suggest that endogenous FGF-2 is crucially involved in the early phase of peripheral nerve regeneration possibly by regulation of Schwann cell differentiation.
Collapse
Affiliation(s)
- Julia Jungnickel
- Department of Neuroanatomy OE 4140, Center of Anatomy, Hannover Medical School, D-30623 Hannover, Germany.
| | | | | | | | | |
Collapse
|
42
|
Parkinson DB, Dickinson S, Bhaskaran A, Kinsella MT, Brophy PJ, Sherman DL, Sharghi-Namini S, Duran Alonso MB, Mirsky R, Jessen KR. Regulation of the myelin gene periaxin provides evidence for Krox-20-independent myelin-related signalling in Schwann cells. Mol Cell Neurosci 2003; 23:13-27. [PMID: 12799134 DOI: 10.1016/s1044-7431(03)00024-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We investigated the role of Krox-20 (Egr2), a transcription factor that regulates myelination, in controlling the myelin-associated protein periaxin. In developing Schwann cells, periaxin immunoreactivity appeared at least 2 days before Krox-20-immunopositive nuclei. Consistent with this, in Krox-20 null mice periaxin was upregulated on schedule, albeit to a lower level. In culture Krox-20 and periaxin were upregulated by cAMP as expected for myelin genes. Only those cells with the highest periaxin levels also expressed Krox-20, while other periaxin-positive cells remained Krox-20-negative. Furthermore, cAMP elevated periaxin even in Krox-20 null cells. We also found that in culture enforced Krox-20 expression induced expression of periaxin mRNA and protein in the absence of cAMP elevating agents, and that this induction was inhibited by the co-repressor NAB2. These findings reveal a dual mechanism for periaxin regulation and suggest that the role of Krox-20 is to amplify an earlier Krox-20-independent activation of the periaxin gene. Thus the axonal signals responsible for myelination are only partially transduced in Schwann cells by mechanisms that depend on Krox-20.
Collapse
Affiliation(s)
- David B Parkinson
- Department of Anatomy and Developmental Biology, University College London, Gower Street, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Slutsky SG, Kamaraju AK, Levy AM, Chebath J, Revel M. Activation of myelin genes during transdifferentiation from melanoma to glial cell phenotype. J Biol Chem 2003; 278:8960-8. [PMID: 12643284 DOI: 10.1074/jbc.m210569200] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Induction of myelin genes occurs around birth in the last stage of Schwann cells differentiation and is reactivated in case of nerve injury. Previous studies showed that activation of the gp130 receptor system, using as ligand interleukin-6 fused to its soluble receptor (IL6RIL6), causes induction of myelin genes such as myelin basic protein (MBP) and myelin protein zero (Po) in embryonic dorsal root ganglia Schwann cells. We also reported that in murine melanoma B16/F10.9 cells, IL6RIL6 causes a shut-off of melanogenesis mediated by a down-regulation of the paired-homeodomain factor Pax3. The present work demonstrates that these IL6RIL6-treated F10.9 cells undergo transdifferentiation to a myelinating glial phenotype characterized by induction of the transcriptional activities of both Po and MBP promoters and accumulation of myelin gene products. For both Po and MBP promoters, a repression by Pax3 and stimulation by Sox10 can be demonstrated. Because after IL6RIL6-treatment, Pax3 disappears from the F10.9 cells (as it does in mature myelinating Schwann cells) whereas the level of Sox10 rather increases, we modulated the relative level of these factors and show their involvement in the induction of myelin gene expression by IL6RIL6. In addition, however, we show that a C/G-rich CACC box in the Po promoter is required for activation by IL6RIL6, as well as by ectopic Sox10, and identify a Kruppel-type zinc finger factor acting through this CACC box, which stimulates Po promoter activity.
Collapse
Affiliation(s)
- Shalom G Slutsky
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | | | | | | | | |
Collapse
|
44
|
Yfanti E, Sidera K, Margaritis LH, Patsavoudi E. The 4C5 antigen is associated with Schwann cell migration during development and regeneration of the rat peripheral nervous system. Glia 2003; 45:39-53. [PMID: 14648544 DOI: 10.1002/glia.10307] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The monoclonal antibody 4C5 recognizes a cell surface antigen of the developing central nervous system (CNS) and peripheral nervous system (PNS). In vitro antibody perturbation experiments have shown that the 4C5 antigen is involved in horizontal and vertical migration processes of granule cells during development of the rodent cerebellum. Moreover, results concerning the cellular localization and temporal expression of the 4C5 antigen during development and after injury of the rat sciatic nerve suggested that it may participate in Schwann cell migrations that occur during the above processes. To test this possibility, we examined the effects of our function-blocking antibody on Schwann cell migration in three in vitro bioassays: in tissue cultures from developing sciatic nerve, in dorsal root ganglion cultures on cryostat sections of normal or denervated adult sciatic nerve, and in pure Schwann cell cultures. The results showed that the presence of monoclonal antibody 4C5 in all the above culture systems strongly inhibited Schwann cell migration, indicating that the 4C5 antigen participates in migration processes that take place during development and regeneration of the peripheral nervous system. Moreover, staining of migrating Schwann cells in the presence of monoclonal antibody 4C5 with rhodamine-phalloidin showed that 4C5 antigen activity is associated with actin cytoskeletal organization of these cells, and more specifically with lamellipodia formation.
Collapse
Affiliation(s)
- Eleni Yfanti
- Department of Biochemistry, Hellenic Pasteur Institute, Athens, Greece
| | | | | | | |
Collapse
|
45
|
Awatramani R, Shumas S, Kamholz J, Scherer SS. TGFbeta1 modulates the phenotype of Schwann cells at the transcriptional level. Mol Cell Neurosci 2002; 19:307-19. [PMID: 11906205 DOI: 10.1006/mcne.2001.1094] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We have examined the effects of transforming growth factor beta1 (TGFbeta1) on gene expression in cultured rat Schwann cells (SCs). TGFbeta1 decreased the steady-state mRNA levels of several genes that are expressed by myelinating SCs but had varied effects on the mRNA levels of NCAM, L1, GAP-43, and p75-genes that are expressed by denervated and nonmyelinating SCs. TGFbeta1 antagonized the effects of forskolin on the mRNA levels of the transcription factors Oct-6/tst-1/SCIP and Krox20. Transcriptional run-off analysis demonstrated that the effects of TGFbeta1 on gene expression occur at least in part at the level of transcription. Thus, TGFbeta1 suppresses the expression of genes that characterize the different phenotypes of SCs, and these changes occur at least in part at a transcriptional level.
Collapse
Affiliation(s)
- Rajeshwar Awatramani
- Department of Neurology, Wayne State University, Elliman Building 3206, 421 East Canfield, Detroit, Michigan 48201, USA
| | | | | | | |
Collapse
|
46
|
Sulaiman OAR, Gordon T. Transforming growth factor-beta and forskolin attenuate the adverse effects of long-term Schwann cell denervation on peripheral nerve regeneration in vivo. Glia 2002; 37:206-18. [PMID: 11857679 DOI: 10.1002/glia.10022] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Transforming growth factor-beta (TGF-beta) plays a central role in the regulation of Schwann cell (SC) proliferation and differentiation and is essential for the neurotrophic effects of several neurotrophic factors (reviewed by Unsicker and Krieglstein, 2000; Unsicker and Strelau, 2000). However, its role in peripheral nerve regeneration in vivo is not yet understood. Our studies were carried out to characterize (1) the effects of duration of regeneration, and chronic SC denervation on the number of tibial (TIB) motor neurons that regenerated axons over a fixed distance (25 mm into distal common peroneal [CP] nerve stumps), and (2) the effect of in vitro incubation of 6-month chronically denervated sciatic nerve explants with TGF-beta and forskolin on their capacity to support axonal regeneration in vivo. TIB--CP cross-suture in Silastic tubing was used, and regeneration into 0-24-week chronically denervated CP stumps was allowed for either 1.5 or 3 months. Chronically denervated rat sciatic nerve explants (3 x 3 mm(2)) were incubated in vitro with either DMEM and 15% fetal calf serum (D-15) plus TGF-beta/forskolin or D-15 alone for 48 h and placed into a 10-mm Silastic tube that bridged the proximal and distal nerve stumps of a freshly cut TIB nerve. The number of tibial motor neurons that regenerated axons through the explants and 25 mm into the distal nerve stump after 6 months, and TIB regeneration into the CP nerve stumps, were assessed using retrograde tracers, fluorogold, or fluororuby. We found that all tibial motor neurons regenerate their axons 25 mm into 0-4-week denervated CP nerve stumps after a regeneration period of 3 months. Reducing regeneration time to 1.5 months and chronic denervation, reduced the number of motor neurons that regenerated axons over 25 mm. Exposure of 6-month denervated nerve explants to TGF-beta/forskolin increased the number of motor neurons that regenerated through them from 258 +/-13; mean +/- SE to 442 +/- 22. Hence, acute treatment of atrophic SC with TGF-beta can reactivate the growth-permissive SC phenotype to support axonal regeneration.
Collapse
Affiliation(s)
- Olawale A R Sulaiman
- Department of Pharmacology, Division of Neuroscience, University of Alberta, Edmonton, Alberta, Canada
| | | |
Collapse
|
47
|
Tabernero A, Velasco A, Granda B, Lavado EM, Medina JM. Transcytosis of albumin in astrocytes activates the sterol regulatory element-binding protein-1, which promotes the synthesis of the neurotrophic factor oleic acid. J Biol Chem 2002; 277:4240-6. [PMID: 11724788 DOI: 10.1074/jbc.m108760200] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We have recently reported that albumin, a serum protein present in the developing brain, stimulates the synthesis of oleic acid by astrocytes, which promotes neuronal differentiation. In this work, we gain insight into the mechanism by which albumin induces the synthesis of this neurotrophic factor. Our results show that astrocytes internalize albumin in vesicle-like structures by receptor-mediated endocytosis. Albumin uptake was followed by transcytosis, including passage through the endoplasmic reticulum, which was required to induce the synthesis of oleic acid. Oleic acid synthesis is feedback-regulated by the sterol regulatory element-binding protein-1, which induces the transcription of stearoyl-CoA 9-desaturase, the key rate-limiting enzyme for oleic acid synthesis. In our research, the presence of albumin activated the sterol regulatory element-binding protein-1 and increased stearoyl-CoA 9-desaturase mRNA. Moreover, when the activity of sterol regulatory element-binding protein-1 was inhibited by overexpression of a truncated form of this protein, albumin did not affect stearoyl-CoA 9-desaturase mRNA, indicating that the effect of albumin is mediated by this transcription factor. The effect of albumin was abolished when traffic to the endoplasmic reticulum was prevented or when albumin was accompanied with oleic acid. In conclusion, our results suggest that the transcytosis of albumin includes passage through the endoplasmic reticulum, where oleic acid is sequestrated, initiating the signal cascade leading to an increase in its own synthesis.
Collapse
Affiliation(s)
- Arantxa Tabernero
- Departamento de Bioquimica y Biologia Molecular, Facultad de Farmacia, Universidad de Salamanca, 37007 Salamanca, Spain
| | | | | | | | | |
Collapse
|
48
|
Hansen MR, Vijapurkar U, Koland JG, Green SH. Reciprocal signaling between spiral ganglion neurons and Schwann cells involves neuregulin and neurotrophins. Hear Res 2001; 161:87-98. [PMID: 11744285 DOI: 10.1016/s0378-5955(01)00360-4] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
To investigate the role of neuron-glial cell interactions in the auditory nerve, we asked whether spiral ganglion neurons (SGNs) express neuregulin and whether neuregulin regulates proliferation and/or neurotrophin expression in spiral ganglion Schwann cells (SGSCs). Using immunocytochemistry, we found that type I and type II SGNs express neuregulin in vivo and in vitro. Cultured SGSCs express the neuregulin receptors ErbB2 and ErbB3, but not ErbB4. Neuregulin activates ErbB2 and ErbB3 in cultured SGSCs, evidenced by increased tyrosine phosphorylation of the receptors following neuregulin treatment. Neuregulin treatment increased the proliferation rate of cultured SGSCs by 2.5-fold. Fibroblast growth factor-2 (FGF-2) and transforming growth factor beta (TGF-beta) also increased SGSC proliferation. The mitogenic effect of neuregulin and FGF-2 was blocked by inhibition of mitogen-activated protein kinase signaling but not by inhibition of phosphatidylinositol-3'-OH kinase. Using RT-PCR, we found that cultured SGSCs express neurotrophins, including brain-derived neurotrophic factor and neurotrophin-3 (NT-3), raising the possibility that SGSCs contribute to the trophic support of SGNs. Treatment with neither neuregulin nor TGF-beta increased neurotrophin expression in cultured SGSCs, as had been observed in developing sympathetic ganglia, but appeared to negatively regulate NT-3 expression. Thus, neuregulin and neurotrophins may mediate reciprocal neuron-glial interactions in the auditory nerve.
Collapse
Affiliation(s)
- M R Hansen
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, IA 52242, USA
| | | | | | | |
Collapse
|
49
|
Tabernero A, Lavado EM, Granda B, Velasco A, Medina JM. Neuronal differentiation is triggered by oleic acid synthesized and released by astrocytes. J Neurochem 2001; 79:606-16. [PMID: 11701764 DOI: 10.1046/j.1471-4159.2001.00598.x] [Citation(s) in RCA: 93] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Unlike in the adult brain, the newborn brain specifically takes up serum albumin during the postnatal period, coinciding with the stage of maximal brain development. Here we report that albumin stimulates oleic acid synthesis by astrocytes from the main metabolic substrates available during brain development. Oleic acid released by astrocytes is used by neurons for the synthesis of phospholipids and is specifically incorporated into growth cones. Oleic acid promotes axonal growth, neuronal clustering, and expression of the axonal growth-associated protein-43, GAP-43; all these observations indicating neuronal differentiation. The effect of oleic acid on GAP-43 synthesis is brought about by the activation of protein kinase C, since it was prevented by inhibitors of this kinase, such as H-7, polymyxin or sphingosine. The expression of GAP-43 was significantly increased in neurons co-cultured with astrocytes by the presence of albumin indicating that neuronal differentiation takes place in the presence of oleic acid synthesized and released by astrocytes in situ. In conclusion, during brain development the presence of albumin could play an important role by triggering the synthesis and release of oleic acid by astrocytes, which induces neuronal differentiation.
Collapse
Affiliation(s)
- A Tabernero
- Unidad de Investigación, Hospital Universitario de Salamanca, Spain
| | | | | | | | | |
Collapse
|
50
|
Scarlato M, Xu T, Bannerman P, Beesley J, Reddy UR, Rostami A, Scherer SS, Pleasure D. Axon-Schwann cell interactions regulate the expression of fibroblast growth factor-5 (FGF-5). J Neurosci Res 2001; 66:16-22. [PMID: 11598998 DOI: 10.1002/jnr.1193] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
We screened for genes whose expression is significantly up- or downregulated during Wallerian degeneration in adult rat sciatic nerve with cDNA arrays. Fibroblast growth factor-5 (FGF-5) mRNA seemed to be induced. This was confirmed by northern blotting and in situ hybridization, as well as Western blotting for FGF-5 in axotomized nerve. Axon-Schwann cell interactions decreased the steady-state level of FGF-5 mRNA in regenerating sciatic nerves, and forskolin diminished its expression in cultured Schwann cells. We conclude that denervated Schwann cells synthesize FGF-5, which is a secreted, neuronotrophic member of the FGF family.
Collapse
Affiliation(s)
- M Scarlato
- Neurology Research, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | | | |
Collapse
|