1
|
Li C, Rassekh N, O'Daly A, Kebaisch F, Wolinsky R, Vyas A, Skolasky R, Hoke A, Brushart T. Preferential motor reinnervation is modulated by both repair site and distal nerve environments. Exp Neurol 2025; 385:115066. [PMID: 39579958 PMCID: PMC11783203 DOI: 10.1016/j.expneurol.2024.115066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 11/02/2024] [Accepted: 11/14/2024] [Indexed: 11/25/2024]
Abstract
To restore function after nerve injury, axons must regenerate from the injury site to the periphery, then reinnervate appropriate end organs when they arrive. Only 10 % of adults who suffer nerve injury will regain normal function, often because axons regenerate to functionally inappropriate targets (Brushart, 2011). The peripheral destination of these axons is largely determined by the pathways they enter at the site of nerve repair. To improve clinical outcomes, it is thus critical to improve the accuracy of axon pathfinding. In rodents, motor axons regenerating in mixed nerve preferentially reinnervate pathways leading to muscle, a process termed preferential motor reinnervation (PMR). Previous experiments have shown that PMR can be enhanced by predegenerating nerve grafts to enhance growth factor production and remove inhibitory factors (Abdullah et al., 2013). The current experiments explore the relative contributions of motor pathways, sensory pathways, and the repair environment to this enhancement. Sensory and/or motor pathways within rat femoral nerve grafts were predegenerated for 3 weeks to optimize growth factor production (Brushart et al., 2013) or for 12 weeks to deplete it. Optimizing the environment within previously motor Schwann cell tubes promoted PMR, regardless of whether adjacent sensory pathways were optimized or chronically denervated. However, this positive effect was abolished when sensory pathways were undergoing acute Wallerian degeneration immediately after nerve repair. The repair environment thus precluded motor axon pathfinding in spite of an optimized distal motor pathway. When sensory pathways were optimized and motor pathways were chronically denervated, not only was PMR abolished, but motoneurons failed to respond to the greater volume of growth factors in the sensory nerve. Small sensory neurons, however, selectively reinnervated cutaneous nerve under these conditions. These experiments thus strengthen the concept that, in adult rats, sensory and motor pathways have unique identities capable of influencing both sensory and motor axon regeneration. Furthermore, they demonstrate that, in the rat, delaying nerve repair for 3 weeks to enhance growth factor production and clear the products of acute Wallerian degeneration can enhance regeneration specificity without the need for exogenous treatments.
Collapse
Affiliation(s)
- C Li
- Department of Orthopaedic Surgery, Johns Hopkins University, 601 N Caroline St, Baltimore, MD 21287, United States of America
| | - N Rassekh
- Department of Orthopaedic Surgery, Johns Hopkins University, 601 N Caroline St, Baltimore, MD 21287, United States of America
| | - A O'Daly
- Department of Orthopaedic Surgery, Johns Hopkins University, 601 N Caroline St, Baltimore, MD 21287, United States of America
| | - F Kebaisch
- Department of Orthopaedic Surgery, Johns Hopkins University, 601 N Caroline St, Baltimore, MD 21287, United States of America
| | - R Wolinsky
- Department of Orthopaedic Surgery, Johns Hopkins University, 601 N Caroline St, Baltimore, MD 21287, United States of America
| | - A Vyas
- Department of Orthopaedic Surgery, Johns Hopkins University, 601 N Caroline St, Baltimore, MD 21287, United States of America
| | - R Skolasky
- Department of Orthopaedic Surgery, Johns Hopkins University, 601 N Caroline St, Baltimore, MD 21287, United States of America
| | - A Hoke
- Department of Neurology, Johns Hopkins University, 855 N Wolfe St, Baltimore, MD 21287, United States of America; The Solomon H Snyder Department of Neuroscience, Johns Hopkins University, 725 N Wolfe St, Baltimore, MD 21205, United States of America
| | - T Brushart
- Department of Orthopaedic Surgery, Johns Hopkins University, 601 N Caroline St, Baltimore, MD 21287, United States of America; Department of Neurology, Johns Hopkins University, 855 N Wolfe St, Baltimore, MD 21287, United States of America.
| |
Collapse
|
2
|
Romero-Ortega M. Peripheral Nerves, Anatomy and Physiology of. ENCYCLOPEDIA OF COMPUTATIONAL NEUROSCIENCE 2022:2715-2719. [DOI: 10.1007/978-1-0716-1006-0_214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
3
|
Wang K, Qin B. [Research progress of peripheral nerve mismatch regeneration]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2021; 35:387-391. [PMID: 33719250 DOI: 10.7507/1002-1892.202008085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Objective To review the research progress of peripheral nerve mismatch regeneration, and to provide reference for its related basic research and clinical treatment. Methods The pathophysiology of peripheral nerve after injury, several main factors affecting the mismatch regeneration of peripheral nerve, and the fate of axon after mismatch regeneration were summarized by referring to the relevant literature at home and abroad in recent years. Results Distal pathways and target organs can selectively affect the mismatch regeneration of peripheral nerves; different phenotypes of Schwann cells have different effects on the mismatch regeneration of peripheral nerves; studying the mechanism of action of exosomes from different Schwann cells on different types of axons can provide a new direction for solving the mismatch regeneration of peripheral nerves. Conclusion Peripheral nerve mismatch regeneration is affected by various factors. However, the specific mechanism and characteristics of these factors remain to be further studied.
Collapse
Affiliation(s)
- Kunliang Wang
- Department of Microsurgery, Orthopaedic Trauma and Hand Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou Guangdong, 510080, P.R.China
| | - Bengang Qin
- Department of Microsurgery, Orthopaedic Trauma and Hand Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou Guangdong, 510080, P.R.China
| |
Collapse
|
4
|
Suzuki H, Araki K, Matsui T, Tanaka Y, Uno K, Tomifuji M, Yamashita T, Satoh Y, Kobayashi Y, Shiotani A. TrkA inhibitor promotes motor functional regeneration of recurrent laryngeal nerve by suppression of sensory nerve regeneration. Sci Rep 2020; 10:16892. [PMID: 33037246 PMCID: PMC7547101 DOI: 10.1038/s41598-020-72288-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 08/19/2020] [Indexed: 11/19/2022] Open
Abstract
Recurrent laryngeal nerve (RLN) injury, in which hoarseness and dysphagia arise as a result of impaired vocal fold movement, is a serious complication. Misdirected regeneration is an issue for functional regeneration. In this study, we demonstrated the effect of TrkA inhibitors, which blocks the NGF-TrkA pathway that acts on the sensory/automatic nerves thus preventing misdirected regeneration among motor and sensory nerves, and thereby promoting the regeneration of motor neurons to achieve functional recovery. RLN axotomy rat models were used in this study, in which cut ends of the nerve were bridged with polyglycolic acid-collagen tube with and without TrkA inhibitor (TrkAi) infiltration. Our study revealed significant improvement in motor nerve fiber regeneration and function, in assessment of vocal fold movement, myelinated nerve regeneration, compound muscle action potential, and prevention of laryngeal muscle atrophy. Retrograde labeling demonstrated fewer labeled neurons in the vagus ganglion, which confirmed reduced misdirected regeneration among motor and sensory fibers, and a change in distribution of the labeled neurons in the nucleus ambiguus. Our study demonstrated that TrkAi have a strong potential for clinical application in the treatment of RLN injury.
Collapse
Affiliation(s)
- Hiroshi Suzuki
- Department of Otolaryngology-Head and Neck Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan.,Department of Otolaryngology, Self-Defense Forces Central Hospital, Tokyo, Japan
| | - Koji Araki
- Department of Otolaryngology-Head and Neck Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan.
| | - Toshiyasu Matsui
- Department of Anatomy and Neurobiology, National Defense Medical College, Tokorozawa, Japan.,Laboratory of Veterinary Anatomy, Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Japan
| | - Yuya Tanaka
- Department of Otolaryngology-Head and Neck Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Kosuke Uno
- Department of Otolaryngology-Head and Neck Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Masayuki Tomifuji
- Department of Otolaryngology-Head and Neck Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Taku Yamashita
- Department of Otolaryngology-Head and Neck Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan.,Department of Otolaryngology-Head and Neck Surgery, Kitasato University School of Medicine, Sagamihara, Japan
| | - Yasushi Satoh
- Department of Biochemistry, National Defense Medical College, Tokorozawa, Japan
| | - Yasushi Kobayashi
- Department of Anatomy and Neurobiology, National Defense Medical College, Tokorozawa, Japan
| | - Akihiro Shiotani
- Department of Otolaryngology-Head and Neck Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| |
Collapse
|
5
|
Inositol 1,4,5-Trisphosphate Receptor Type 3 Regulates Neuronal Growth Cone Sensitivity to Guidance Signals. iScience 2020; 23:100963. [PMID: 32199289 PMCID: PMC7082556 DOI: 10.1016/j.isci.2020.100963] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 02/10/2020] [Accepted: 02/29/2020] [Indexed: 11/22/2022] Open
Abstract
During neurodevelopment, the growth cone deciphers directional information from extracellular guidance cues presented as shallow concentration gradients via signal amplification. However, it remains unclear how the growth cone controls this amplification process during its navigation through an environment in which basal cue concentrations vary widely. Here, we identified inositol 1,4,5-trisphosphate (IP3) receptor type 3 as a regulator of axonal sensitivity to guidance cues in vitro and in vivo. Growth cones lacking the type 3 subunit are hypersensitive to nerve growth factor (NGF), an IP3-dependent attractive cue, and incapable of turning toward normal concentration ranges of NGF to which wild-type growth cones respond. This is due to globally, but not asymmetrically, activated Ca2+ signaling in the hypersensitive growth cones. Remarkably, lower NGF concentrations can polarize growth cones for turning if IP3 receptor type 3 is deficient. These data suggest a subtype-specific IP3 receptor function in sensitivity adjustment during axon navigation. IP3 receptor type 3 (IP3R3) controls axonal sensitivity to IP3-based guidance cues IP3R3−/− growth cones are not attracted to NGF due to global Ca2+ responses Lower NGF concentrations can polarize IP3R3−/− growth cones for attractive turning NGF knockdown in vivo can revert abnormal trajectory of IP3R3−/− axons
Collapse
|
6
|
Endo T, Kadoya K, Kawamura D, Iwasaki N. Evidence for cell-contact factor involvement in neurite outgrowth of dorsal root ganglion neurons stimulated by Schwann cells. Exp Physiol 2019; 104:1447-1454. [PMID: 31294871 DOI: 10.1113/ep087634] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 07/08/2019] [Indexed: 12/14/2022]
Abstract
NEW FINDINGS What is the central question of this study? Although the factors secreted from Schwann cells that promote axonal growth in the peripheral nervous system have been well studied, the effect of cell-contact factors on Schwann cells remains to be determined. What is the main finding and its importance? This study demonstrates that Schwann cells stimulate neurite outgrowth by direct contact with neurites and by secreting factors. Notably, the effect of cell-contact factors in neurite outgrowth is comparable to that of secreted factors, indicating that the identification of cell surface molecules on Schwann cells that promote neurite outgrowth could lead to development of a new therapy for peripheral nervous system injury. ABSTRACT Schwann cells (SCs) play a variety of roles in the regeneration process after injury to the peripheral nervous system. The factors secreted from SCs that promote axonal growth have been well studied. However, the involvement of cell-contact factors on SCs remains to be determined. Here, we demonstrate a significant contribution of a cell-contact mechanism in the effect of SCs on promotion of neuronal outgrowth. Neurite outgrowth of adult sensory neurons from dorsal root ganglia was quantified during co-culture with adult SCs. Direct contact of SCs with neurons was eliminated by culturing SCs on an insert placed in the same well; this resulted in a 51% reduction in the length of neurite outgrowth. In addition, when dorsal root ganglion neurons were cultured on sparsely seeded SCs, neurons that made contact with SCs on their neurites had 118% longer neurites than neurons that lacked contacts with SCs. Collectively, these findings provide evidence that SCs stimulate neurite outgrowth via direct contact with neurites in addition to secreting factors. The identification of cell surface molecules on SCs that promote neurite outgrowth could lead to development of a new therapy for peripheral nervous system injury.
Collapse
Affiliation(s)
- Takeshi Endo
- Department of Orthopaedic Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Ken Kadoya
- Department of Orthopaedic Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Daisuke Kawamura
- Department of Orthopaedic Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Norimasa Iwasaki
- Department of Orthopaedic Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| |
Collapse
|
7
|
Dang Z, Avolio E, Albertario A, Sala-Newby GB, Thomas AC, Wang N, Emanueli C, Madeddu P. Nerve growth factor gene therapy improves bone marrow sensory innervation and nociceptor-mediated stem cell release in a mouse model of type 1 diabetes with limb ischaemia. Diabetologia 2019; 62:1297-1311. [PMID: 31016359 PMCID: PMC6560027 DOI: 10.1007/s00125-019-4860-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 03/04/2019] [Indexed: 12/16/2022]
Abstract
AIMS/HYPOTHESIS Sensory neuropathy is common in people with diabetes; neuropathy can also affect the bone marrow of individuals with type 2 diabetes. However, no information exists on the state of bone marrow sensory innervation in type 1 diabetes. Sensory neurons are trophically dependent on nerve growth factor (NGF) for their survival. The aim of this investigation was twofold: (1) to determine if sensory neuropathy affects the bone marrow in a mouse model of type 1 diabetes, with consequences for stem cell liberation after tissue injury; and (2) to verify if a single systemic injection of the NGF gene exerts long-term beneficial effects on these phenomena. METHODS A mouse model of type 1 diabetes was generated in CD1 mice by administration of streptozotocin; vehicle was administered to non-diabetic control animals. Diabetic animals were randomised to receive systemic gene therapy with either human NGF or β-galactosidase. After 13 weeks, limb ischaemia was induced in both groups to study the recovery post injury. When the animals were killed, samples of tissue and peripheral blood were taken to assess stem cell mobilisation and homing, levels of substance P and muscle vascularisation. An in vitro cellular model was adopted to verify signalling downstream to human NGF and related neurotrophic or pro-apoptotic effects. Normally distributed variables were compared between groups using the unpaired Student's t test and non-normally distributed variables were assessed by the Wilcoxon-Mann-Whitney test. The Fisher's exact test was employed for categorical variables. RESULTS Immunohistochemistry indicated a 3.3-fold reduction in the number of substance P-positive nociceptive fibres in the bone marrow of type 1 diabetic mice (p < 0.001 vs non-diabetic). Moreover, diabetes abrogated the creation of a neurokinin gradient which, in non-diabetic mice, favoured the mobilisation and homing of bone-marrow-derived stem cells expressing the substance P receptor neurokinin 1 receptor (NK1R). Pre-emptive gene therapy with NGF prevented bone marrow denervation, contrasting with the inhibitory effect of diabetes on the mobilisation of NK1R-expressing stem cells, and restored blood flow recovery from limb ischaemia. In vitro hNGF induced neurite outgrowth and exerted anti-apoptotic actions on rat PC12 cells exposed to high glucose via activation of the canonical neurotrophic tyrosine kinase receptor type 1 (TrkA) signalling pathway. CONCLUSIONS/INTERPRETATION This study shows, for the first time, the occurrence of sensory neuropathy in the bone marrow of type 1 diabetic mice, which translates into an altered modulation of substance P and depressed release of substance P-responsive stem cells following ischaemia. NGF therapy improves bone marrow sensory innervation, with benefits for healing on the occurrence of peripheral ischaemia. Nociceptors may represent a new target for the treatment of ischaemic complications in diabetes.
Collapse
Affiliation(s)
- Zexu Dang
- Experimental Cardiovascular Medicine, Faculty of Translational Health Sciences, Bristol Medical School, University of Bristol, Upper Maudlin Street, Bristol, BS2 8HW, UK
| | - Elisa Avolio
- Experimental Cardiovascular Medicine, Faculty of Translational Health Sciences, Bristol Medical School, University of Bristol, Upper Maudlin Street, Bristol, BS2 8HW, UK
| | - Ambra Albertario
- Experimental Cardiovascular Medicine, Faculty of Translational Health Sciences, Bristol Medical School, University of Bristol, Upper Maudlin Street, Bristol, BS2 8HW, UK
| | - Graciela B Sala-Newby
- Experimental Cardiovascular Medicine, Faculty of Translational Health Sciences, Bristol Medical School, University of Bristol, Upper Maudlin Street, Bristol, BS2 8HW, UK
| | - Anita C Thomas
- Experimental Cardiovascular Medicine, Faculty of Translational Health Sciences, Bristol Medical School, University of Bristol, Upper Maudlin Street, Bristol, BS2 8HW, UK
| | - Nianhong Wang
- Experimental Cardiovascular Medicine, Faculty of Translational Health Sciences, Bristol Medical School, University of Bristol, Upper Maudlin Street, Bristol, BS2 8HW, UK
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Pudong, Shanghai, China
| | - Costanza Emanueli
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Paolo Madeddu
- Experimental Cardiovascular Medicine, Faculty of Translational Health Sciences, Bristol Medical School, University of Bristol, Upper Maudlin Street, Bristol, BS2 8HW, UK.
| |
Collapse
|
8
|
In vitro efficacy of a gene-activated nerve guidance conduit incorporating non-viral PEI-pDNA nanoparticles carrying genes encoding for NGF, GDNF and c-Jun. Acta Biomater 2018; 75:115-128. [PMID: 29885855 DOI: 10.1016/j.actbio.2018.06.014] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 06/04/2018] [Accepted: 06/06/2018] [Indexed: 01/09/2023]
Abstract
Despite the success of tissue engineered nerve guidance conduits (NGCs) for the treatment of small peripheral nerve injuries, autografts remain the clinical gold standard for larger injuries. The delivery of neurotrophic factors from conduits might enhance repair for more effective treatment of larger injuries but the efficacy of such systems is dependent on a safe, effective platform for controlled and localised therapeutic delivery. Gene therapy might offer an innovative approach to control the timing, release and level of neurotrophic factor production by directing cells to transiently sustain therapeutic protein production in situ. In this study, a gene-activated NGC was developed by incorporating non-viral polyethyleneimine-plasmid DNA (PEI-pDNA) nanoparticles (N/P 7 ratio, 2 μg dose) with the pDNA encoding for nerve growth factor (NGF), glial derived neurotrophic factor (GDNF) or the transcription factor c-Jun. The physicochemical properties of PEI-pDNA nanoparticles, morphology, size and charge, were shown to be suitable for gene delivery and demonstrated high Schwann cell transfection efficiency (60 ± 13%) in vitro. While all three genes showed therapeutic potential in terms of enhancing neurotrophic cytokine production while promoting neurite outgrowth, delivery of the gene encoding for c-Jun showed the greatest capacity to enhance regenerative cellular processes in vitro. Ultimately, this gene-activated NGC construct was shown to be capable of transfecting both Schwann cells (S42 cells) and neuronal cells (PC12 and dorsal root ganglia) in vitro, demonstrating potential for future therapeutic applications in vivo. STATEMENT OF SIGNIFICANCE The basic requirements of biomaterial-based nerve guidance conduits have now been well established and include being able to bridge a nerve injury to support macroscopic guidance between nerve stumps, while being strong enough to withstand longitudinal tension and circumferential compression, in addition to being mechanically sound to facilitate surgical handling and implantation. While meeting these criteria, conduits are still limited to the treatment of small defects clinically and might benefit from additional biochemical stimuli to enhance repair for the effective treatment of larger injuries. In this study, a gene activated conduit was successfully developed by incorporating non-viral nanoparticles capable of efficient Schwann cell and neuronal cell transfection with therapeutic genes in vitro, which showed potential to enhance repair in future applications particularly when taking advantage of the transcription factor c-Jun. This innovative approach may provide an alternative to conduits used as platforms for the delivery neurotrophic factors or genetically modified cells (viral gene therapy), and a potential solution for the unmet clinical need to repair large peripheral nerve injury effectively.
Collapse
|
9
|
Nascimento AI, Mar FM, Sousa MM. The intriguing nature of dorsal root ganglion neurons: Linking structure with polarity and function. Prog Neurobiol 2018; 168:86-103. [PMID: 29729299 DOI: 10.1016/j.pneurobio.2018.05.002] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 04/26/2018] [Accepted: 05/01/2018] [Indexed: 11/26/2022]
Abstract
Dorsal root ganglion (DRG) neurons are the first neurons of the sensory pathway. They are activated by a variety of sensory stimuli that are then transmitted to the central nervous system. An important feature of DRG neurons is their unique morphology where a single process -the stem axon- bifurcates into a peripheral and a central axonal branch, with different functions and cellular properties. Distinctive structural aspects of the two DRG neuron branches may have important implications for their function in health and disease. However, the link between DRG axonal branch structure, polarity and function has been largely neglected in the field, and relevant information is rather scattered across the literature. In particular, ultrastructural differences between the two axonal branches are likely to account for the higher transport and regenerative ability of the peripheral DRG neuron axon when compared to the central one. Nevertheless, the cell intrinsic factors contributing to this central-peripheral asymmetry are still unknown. Here we critically review the factors that may underlie the functional asymmetry between the peripheral and central DRG axonal branches. Also, we discuss the hypothesis that DRG neurons may assemble a structure resembling the axon initial segment that may be responsible, at least in part, for their polarity and electrophysiological features. Ultimately, we suggest that the clarification of the axonal ultrastructure of DRG neurons using state-of-the-art techniques will be crucial to understand the physiology of this peculiar cell type.
Collapse
Affiliation(s)
- Ana Isabel Nascimento
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular-IBMC and Instituto de Inovação e Investigação em Saúde, University of Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; Instituto de Ciências Biomédicas Abel Salazar-ICBAS, Rua Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Fernando Milhazes Mar
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular-IBMC and Instituto de Inovação e Investigação em Saúde, University of Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Mónica Mendes Sousa
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular-IBMC and Instituto de Inovação e Investigação em Saúde, University of Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal.
| |
Collapse
|
10
|
Anand S, Desai V, Alsmadi N, Kanneganti A, Nguyen DHT, Tran M, Patil L, Vasudevan S, Xu C, Hong Y, Cheng J, Keefer E, Romero-Ortega MI. Asymmetric Sensory-Motor Regeneration of Transected Peripheral Nerves Using Molecular Guidance Cues. Sci Rep 2017; 7:14323. [PMID: 29085079 PMCID: PMC5662603 DOI: 10.1038/s41598-017-14331-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 10/06/2017] [Indexed: 11/22/2022] Open
Abstract
Neural interfaces are designed to decode motor intent and evoke sensory precepts in amputees. In peripheral nerves, recording movement intent is challenging because motor axons are only a small fraction compared to sensory fibers and are heterogeneously mixed particularly at proximal levels. We previously reported that pain and myelinated axons regenerating through a Y-shaped nerve guide with sealed ends, can be modulated by luminar release of nerve growth factor (NGF) and neurotrophin-3 (NT-3), respectively. Here, we evaluate the differential potency of NGF, glial cell line-derived neurotrophic factor (GDNF), brain-derived neurotrophic factor (BDNF), pleiotrophin (PTN), and NT-3 in asymmetrically guiding the regeneration of sensory and motor neurons. We report that, in the absence of distal target organs, molecular guidance cues can mediate the growth of electrically conductive fascicles with normal microanatomy. Compared to Y-tube compartments with bovine serum albumin (BSA), GDNF and NGF increased the motor and sensory axon content, respectively. In addition, the sensory to motor ratio was significantly increased by PTN (12.7:1) when compared to a BDNF + GDNF choice. The differential content of motor and sensory axons modulated by selective guidance cues may provide a strategy to better define axon types in peripheral nerve interfaces.
Collapse
Affiliation(s)
- Sanjay Anand
- Department of Bioengineering, University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX, 75080, USA
| | - Vidhi Desai
- Department of Bioengineering, University of Texas at Arlington, 500 UTA Blvd, Arlington, Texas, 76010, USA
| | - Nesreen Alsmadi
- Department of Bioengineering, University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX, 75080, USA
| | - Aswini Kanneganti
- Department of Bioengineering, University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX, 75080, USA
| | - Dianna Huyen-Tram Nguyen
- Department of Bioengineering, University of Texas at Arlington, 500 UTA Blvd, Arlington, Texas, 76010, USA
| | - Martin Tran
- Department of Bioengineering, University of Texas at Arlington, 500 UTA Blvd, Arlington, Texas, 76010, USA
| | - Lokesh Patil
- Department of Bioengineering, University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX, 75080, USA
| | - Srikanth Vasudevan
- Division of Biomedical Physics, Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Cancan Xu
- Department of Bioengineering, University of Texas at Arlington, 500 UTA Blvd, Arlington, Texas, 76010, USA
| | - Yi Hong
- Department of Bioengineering, University of Texas at Arlington, 500 UTA Blvd, Arlington, Texas, 76010, USA
| | - Jonathan Cheng
- Department of Plastic Surgery, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Edward Keefer
- Nerves Incorporated, P.O. Box 141295, Dallas, TX 75214, USA
| | - Mario I Romero-Ortega
- Department of Bioengineering, University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX, 75080, USA.
| |
Collapse
|
11
|
Robinson GA, Madison RD. Polyethylene glycol fusion repair prevents reinnervation accuracy in rat peripheral nerve. J Neurosci Res 2016; 94:636-44. [PMID: 26994857 DOI: 10.1002/jnr.23734] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 02/08/2016] [Accepted: 02/28/2016] [Indexed: 11/12/2022]
Abstract
Functional recovery following a peripheral nerve injury is made easier when regenerating axons correctly reinnervate their original targets. Polyethylene glycol (PEG) has recently been used in attempts to fuse severed peripheral axons during suture-based repair, but an analysis of target selectivity following such repair has not been undertaken. The rat femoral nerve (in which muscle and cutaneous pathways comingle proximally but segregate distally into separate terminal nerve branches) is a convenient in vivo model for assessing motor neuron regeneration accuracy. The present study uses retrograde labeling of motor neurons to compare reinnervation accuracy after suture-based nerve repair with and without PEG fusion. The results show that adding PEG to the suture repair site blocked the preference of motor neurons to reinnervate correctly the distal terminal nerve branch to muscle that was seen with suture repair. Retrograde transport and diffusion studies also determined that PEG fusion allowed passage of probes across the repair site, as has previously been seen, but did not result in motor neuron labeling in the spinal cord. The results suggest that PEG fusion disrupts the beneficial trophic influence of muscle on motor neuron reinnervation accuracy normally seen after suture repair and that such fusion-based approaches may be best suited to nerve injuries in which accurate target reinnervation at the terminal nerve branch level is not a priority. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Grant A Robinson
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | - Roger D Madison
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina.,Biological Laboratory Research and Development Service, Veterans Affairs Medical Center, Durham, North Carolina
| |
Collapse
|
12
|
Goganau I, Blesch A. Gene Therapy for Spinal Cord Injury. Transl Neurosci 2016. [DOI: 10.1007/978-1-4899-7654-3_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
13
|
Liu GY, Jin Y, Zhang Q, Li R. Peripheral nerve repair: a hot spot analysis on treatment methods from 2010 to 2014. Neural Regen Res 2015. [PMID: 26199620 PMCID: PMC4498365 DOI: 10.4103/1673-5374.158368] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Therapeutic strategies for neurological deficits and for promoting nerve regeneration after peripheral nerve injuries have received much focus in clinical research. Advances in basic research in recent years have increased our understanding of the anatomy of peripheral nerves and the importance of the microenvironment. Various new intervention methods have been developed, but with varying effectiveness. In the present study, we selected 911 papers on different repair methods for peripheral nerve injury from the Web of Science and indexed in the Science Citation Index from 2010 to 2014. We quantitatively examine new repair methods and strategies using bibliometrics, and we discuss the present state of knowledge and the problems and prospects of various repair methods, including nerve transfer, neural transplantation, tissue engineering and genetic engineering. Our findings should help in the study and development of repair methods for peripheral nerve injury.
Collapse
Affiliation(s)
- Guang-Yao Liu
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | - Yan Jin
- Department of Ophthalmology, the Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Qiao Zhang
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | - Rui Li
- Hand & Foot Surgery and Reparative & Reconstruction Surgery Center, the Second Hospital of Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
14
|
Hoyng SA, de Winter F, Tannemaat MR, Blits B, Malessy MJA, Verhaagen J. Gene therapy and peripheral nerve repair: a perspective. Front Mol Neurosci 2015; 8:32. [PMID: 26236188 PMCID: PMC4502351 DOI: 10.3389/fnmol.2015.00032] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 07/01/2015] [Indexed: 12/19/2022] Open
Abstract
Clinical phase I/II studies have demonstrated the safety of gene therapy for a variety of central nervous system disorders, including Canavan's, Parkinson's (PD) and Alzheimer's disease (AD), retinal diseases and pain. The majority of gene therapy studies in the CNS have used adeno-associated viral vectors (AAV) and the first AAV-based therapeutic, a vector encoding lipoprotein lipase, is now marketed in Europe under the name Glybera. These remarkable advances may become relevant to translational research on gene therapy to promote peripheral nervous system (PNS) repair. This short review first summarizes the results of gene therapy in animal models for peripheral nerve repair. Secondly, we identify key areas of future research in the domain of PNS-gene therapy. Finally, a perspective is provided on the path to clinical translation of PNS-gene therapy for traumatic nerve injuries. In the latter section we discuss the route and mode of delivery of the vector to human patients, the efficacy and safety of the vector, and the choice of the patient population for a first possible proof-of-concept clinical study.
Collapse
Affiliation(s)
- Stefan A Hoyng
- Department of Neuroregeneration, Netherlands Institute for Neuroscience Amsterdam, Netherlands ; Department of Neurosurgery, Leiden University Medical Center Leiden, Netherlands
| | - Fred de Winter
- Department of Neuroregeneration, Netherlands Institute for Neuroscience Amsterdam, Netherlands ; Department of Neurosurgery, Leiden University Medical Center Leiden, Netherlands
| | - Martijn R Tannemaat
- Department of Neuroregeneration, Netherlands Institute for Neuroscience Amsterdam, Netherlands ; Department of Neurology, Leiden University Medical Center Leiden, Netherlands
| | | | - Martijn J A Malessy
- Department of Neuroregeneration, Netherlands Institute for Neuroscience Amsterdam, Netherlands ; Department of Neurosurgery, Leiden University Medical Center Leiden, Netherlands
| | - Joost Verhaagen
- Department of Neuroregeneration, Netherlands Institute for Neuroscience Amsterdam, Netherlands ; Center for Neurogenomics and Cognition Research, Neuroscience Campus Amsterdam Amsterdam, Netherlands
| |
Collapse
|
15
|
Alsmadi NZ, Patil LS, Hor EM, Lofti P, Razal JM, Chuong CJ, Wallace GG, Romero-Ortega MI. Coiled polymeric growth factor gradients for multi-luminal neural chemotaxis. Brain Res 2015; 1619:72-83. [PMID: 25801117 DOI: 10.1016/j.brainres.2015.01.055] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 01/14/2015] [Accepted: 01/27/2015] [Indexed: 11/24/2022]
Abstract
In the injured adult nervous system, re-establishment of growth-promoting molecular gradients is known to entice and guide nerve repair. However, incorporation of three-dimensional chemotactic gradients in nerve repair scaffolds, particularly in those with multi-luminal architectures, remains extremely challenging. We developed a method that establishes highly tunable three-dimensional molecular gradients in multi-luminal nerve guides by anchoring growth-factor releasing coiled polymeric fibers onto the walls of collagen-filled hydrogel microchannels. Differential pitch in the coiling of neurotrophin-eluting fibers generated sustained chemotactic gradients that appropriately induced the differentiation of Pheochromocytoma (PC12) cells into neural-like cells along an increasing concentration of nerve growth factor (NGF). Computer modeling estimated the stability of the molecular gradient within the luminal collagen, which we confirmed by observing the significant effects of neurotrophin gradients on axonal growth from dorsal root ganglia (DRG). Neurons growing in microchannels exposed to a NGF gradient showed a 60% increase in axonal length compared to those treated with a linear growth factor concentration. In addition, a two-fold increment in the linearity of axonal growth within the microchannels was observed and confirmed by a significant reduction in the turning angle ratios of individual axons. These data demonstrate the ability of growth factor-loaded polymeric coiled fibers to establish three-dimensional chemotactic gradients to promote and direct nerve regeneration in the nervous system and provides a unique platform for molecularly guided tissue repair.
Collapse
Affiliation(s)
- Nesreen Zoghoul Alsmadi
- Department of Bioengineering, University of Texas at Dallas, 800 W. Campbell Rd., EC39, Richardson, 75080 TX, USA
| | - Lokesh S Patil
- Department of Bioengineering, University of Texas Arlington, Arlington, TX, USA
| | - Elijah M Hor
- Department of Bioengineering, University of Texas Arlington, Arlington, TX, USA
| | - Parisa Lofti
- Department of Bioengineering, University of Texas Arlington, Arlington, TX, USA
| | - Joselito M Razal
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, Innovation Campus, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Cheng-Jen Chuong
- Department of Bioengineering, University of Texas Arlington, Arlington, TX, USA
| | - Gordon G Wallace
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, Innovation Campus, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Mario I Romero-Ortega
- Department of Bioengineering, University of Texas at Dallas, 800 W. Campbell Rd., EC39, Richardson, 75080 TX, USA.
| |
Collapse
|
16
|
Hoyng SA, De Winter F, Gnavi S, de Boer R, Boon LI, Korvers LM, Tannemaat MR, Malessy MJ, Verhaagen J. A comparative morphological, electrophysiological and functional analysis of axon regeneration through peripheral nerve autografts genetically modified to overexpress BDNF, CNTF, GDNF, NGF, NT3 or VEGF. Exp Neurol 2014; 261:578-93. [DOI: 10.1016/j.expneurol.2014.08.002] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 08/05/2014] [Indexed: 01/21/2023]
|
17
|
Allodi I, Mecollari V, González-Pérez F, Eggers R, Hoyng S, Verhaagen J, Navarro X, Udina E. Schwann cells transduced with a lentiviral vector encoding Fgf-2 promote motor neuron regeneration following sciatic nerve injury. Glia 2014; 62:1736-46. [PMID: 24989458 DOI: 10.1002/glia.22712] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 06/12/2014] [Accepted: 06/16/2014] [Indexed: 01/07/2023]
Abstract
Fibroblast growth factor 2 (FGF-2) is a trophic factor expressed by glial cells and different neuronal populations. Addition of FGF-2 to spinal cord and dorsal root ganglia (DRG) explants demonstrated that FGF-2 specifically increases motor neuron axonal growth. To further explore the potential capability of FGF-2 to promote axon regeneration, we produced a lentiviral vector (LV) to overexpress FGF-2 (LV-FGF2) in the injured rat peripheral nerve. Cultured Schwann cells transduced with FGF-2 and added to collagen matrix embedding spinal cord or DRG explants significantly increased motor but not sensory neurite outgrowth. LV-FGF2 was as effective as direct addition of the trophic factor to promote motor axon growth in vitro. Direct injection of LV-FGF2 into the rat sciatic nerve resulted in increased expression of FGF-2, which was localized in the basal lamina of Schwann cells. To investigate the in vivo effect of FGF-2 overexpression on axonal regeneration after nerve injury, Schwann cells transduced with LV-FGF2 were grafted in a silicone tube used to repair the resected rat sciatic nerve. Electrophysiological tests conducted for up to 2 months after injury revealed accelerated and more marked reinnervation of hindlimb muscles in the animals treated with LV-FGF2, with an increase in the number of motor and sensory neurons that reached the distal tibial nerve at the end of follow-up.
Collapse
Affiliation(s)
- Ilary Allodi
- Institute of Neurosciences and Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Bellaterra, Spain
| | | | | | | | | | | | | | | |
Collapse
|
18
|
M F G, M M, S H, Khan WS. Peripheral nerve injury: principles for repair and regeneration. Open Orthop J 2014; 8:199-203. [PMID: 25067975 PMCID: PMC4110386 DOI: 10.2174/1874325001408010199] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2013] [Revised: 02/01/2014] [Accepted: 04/02/2014] [Indexed: 01/24/2023] Open
Abstract
Peripheral Nerve Injuries are one of the most common causes of hand dysfunction caused by upper limb trauma but still current management has remained suboptimal. This review aims to explain the traditional view of pathophysiology of nerve repair and also describe why surgical management is still inadequate in using the new biological research that has documented the changes that occur after the nerve injury, which, could cause suboptimal clinical outcomes. Subsequently presentation and diagnosis will be described for peripheral nerve injuries. When traditional surgical repair using end-to-end anastomosis is not adequate nerve conduits are required with the gold standard being the autologous nerve. Due to associated donor site morbidity and poor functional outcome documented with autologous nerve repair several new advancements for alternatives to bridge the gap are being investigated. We will summarise the new and future advancements of non-biological and biological replacements as well as gene therapy, which are being considered as the alternatives for peripheral nerve repair.
Collapse
Affiliation(s)
- Griffin M F
- Plastic Surgery Department, Good Hope Hospital, West Midlands, B75 7RR, UK
| | - Malahias M
- Plastic Surgery Department, Good Hope Hospital, West Midlands, B75 7RR, UK
| | - Hindocha S
- Plastic Surgery Department, Whiston Hospital, Liverpool, L35 5DR, UK
| | - Wasim S Khan
- University College London Institute of Orthopaedics & Musculoskeletal Sciences, Royal National Orthopaedic Hospital, Stanmore, London, HA7 4LP, UK
| |
Collapse
|
19
|
Nerve growth factor improves functional recovery by inhibiting endoplasmic reticulum stress-induced neuronal apoptosis in rats with spinal cord injury. J Transl Med 2014; 12:130. [PMID: 24884850 PMCID: PMC4039547 DOI: 10.1186/1479-5876-12-130] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 03/14/2014] [Indexed: 12/03/2022] Open
Abstract
Background Endoplasmic reticulum (ER) stress-induced apoptosis plays a major role in various diseases, including spinal cord injury (SCI). Nerve growth factor (NGF) show neuroprotective effect and improve the recovery of SCI, but the relations of ER stress-induced apoptosis and the NGF therapeutic effect in SCI still unclear. Methods Young adult female Sprague-Dawley rats’s vertebral column was exposed and a laminectomy was done at T9 vertebrae and moderate contusion injuries were performed using a vascular clip. NGF stock solution was diluted with 0.9% NaCl and administered intravenously at a dose of 20 μg/kg/day after SCI and then once per day until they were executed. Subsequently, the rats were executed at 1d, 3 d, 7d and 14d. The locomotor activities of SCI model rats were tested by the 21-point Basso-Beattie-Bresnahan (BBB) locomotion scale, inclined plane test and footprint analysis. In addition, Western blot analysis was performed to identify the expression of ER-stress related proteins including CHOP, GRP78 and caspase-12 both in vivo and in vitro. The level of cell apoptosis was determined by TUNEL in vivo and Flow cytometry in vitro. Relative downstream signals Akt/GSK-3β and ERK1/2were also analyzed with or without inhibitors in vitro. Results Our results demonstrated that ER stress-induced apoptosis was involved in the injury of SCI model rats. NGF administration improved the motor function recovery and increased the neurons survival in the spinal cord lesions of the model rats. NGF decreases neuron apoptosis which measured by TUNEL and inhibits the activation of caspase-3 cascade. The ER stress-induced apoptosis response proteins CHOP, GRP78 and caspase-12 are inhibited by NGF treatment. Meanwhile, NGF administration also increased expression of growth-associated protein 43 (GAP43). The administration of NGF activated downstream signals Akt/GSK-3β and ERK1/2 in ER stress cell model in vitro. Conclusion The neuroprotective role of NGF in the recovery of SCI is related to the inhibition of ER stress-induced cell death via the activation of downstream signals, also suggested a new trend of NGF translational drug development in the central neural system injuries which involved in the regulation of chronic ER stress.
Collapse
|
20
|
Richner M, Ulrichsen M, Elmegaard SL, Dieu R, Pallesen LT, Vaegter CB. Peripheral nerve injury modulates neurotrophin signaling in the peripheral and central nervous system. Mol Neurobiol 2014; 50:945-70. [PMID: 24752592 DOI: 10.1007/s12035-014-8706-9] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 04/01/2014] [Indexed: 12/21/2022]
Abstract
Peripheral nerve injury disrupts the normal functions of sensory and motor neurons by damaging the integrity of axons and Schwann cells. In contrast to the central nervous system, the peripheral nervous system possesses a considerable capacity for regrowth, but regeneration is far from complete and functional recovery rarely returns to pre-injury levels. During development, the peripheral nervous system strongly depends upon trophic stimulation for neuronal differentiation, growth and maturation. The perhaps most important group of trophic substances in this context is the neurotrophins (NGF, BDNF, NT-3 and NT-4/5), which signal in a complex spatial and timely manner via the two structurally unrelated p75(NTR) and tropomyosin receptor kinase (TrkA, Trk-B and Trk-C) receptors. Damage to the adult peripheral nerves induces cellular mechanisms resembling those active during development, resulting in a rapid and robust increase in the synthesis of neurotrophins in neurons and Schwann cells, guiding and supporting regeneration. Furthermore, the injury induces neurotrophin-mediated changes in the dorsal root ganglia and in the spinal cord, which affect the modulation of afferent sensory signaling and eventually may contribute to the development of neuropathic pain. The focus of this review is on the expression patterns of neurotrophins and their receptors in neurons and glial cells of the peripheral nervous system and the spinal cord. Furthermore, injury-induced changes of expression patterns and the functional consequences in relation to axonal growth and remyelination as well as to neuropathic pain development will be reviewed.
Collapse
Affiliation(s)
- Mette Richner
- Danish Research Institute of Translational Neuroscience DANDRITE, Nordic EMBL Partnership, and Lundbeck Foundation Research Center MIND, Department of Biomedicine, Aarhus University, Ole Worms Allé 3, 8000, Aarhus C, Denmark
| | | | | | | | | | | |
Collapse
|
21
|
Liu Y, Keefe K, Tang X, Lin S, Smith GM. Use of self-complementary adeno-associated virus serotype 2 as a tracer for labeling axons: implications for axon regeneration. PLoS One 2014; 9:e87447. [PMID: 24498323 PMCID: PMC3911946 DOI: 10.1371/journal.pone.0087447] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 12/26/2013] [Indexed: 01/21/2023] Open
Abstract
Various types of tracers are available for use in axon regeneration, but they require an extra operational tracer injection, time-consuming immunohistochemical analysis and cause non-specific labeling. Considerable efforts over the past years have explored other methodologies, especially the use of viral vectors, to investigate axon regeneration after injury. Recent studies have demonstrated that self-complementary Adeno-Associated Virus (scAAV) induced a high transduction efficiency and faster expression of transgenes. Here, we describe for the first time the use of scAAV2-GFP to label long-projection axons in the corticospinal tract (CST), rubrospinal tract (RST) and the central axons of dorsal root ganglion (DRG) in the normal and lesioned animal models. We found that scAAV2-GFP could efficiently transduce neurons in the sensorimotor cortex, red nucleus and DRG. Strong GFP expression could be transported anterogradely along the axon to label the numerous axon fibers from CST, RST and central axons of DRG separately. Comparison of the scAAV2 vector with single-stranded (ss) AAV2 vector in co-labeled sections showed that the scAAV2 vector induced a faster and stronger transgene expression than the ssAAV2 vector in DRG neurons and their axons. In both spinal cord lesion and dorsal root crush injury models, scAAV-GFP could efficiently label the lesioned and regenerated axons around the lesion cavity and the dorsal root entry zone (DREZ) respectively. Further, scAAV2-GFP vector could be combined with traditional tracer to specifically label sensory and motor axons after spinal cord lesion. Thus, we show that using scAAV2-GFP as a tracer is a more effective and efficient way to study axon regeneration following injury.
Collapse
Affiliation(s)
- Yingpeng Liu
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Kathy Keefe
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Xiaoqing Tang
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Shen Lin
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - George M. Smith
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
22
|
de Ruiter GCW, Spinner RJ, Verhaagen J, Malessy MJA. Misdirection and guidance of regenerating axons after experimental nerve injury and repair. J Neurosurg 2014; 120:493-501. [DOI: 10.3171/2013.8.jns122300] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Misdirection of regenerating axons is one of the factors that can explain the limited results often found after nerve injury and repair. In the repair of mixed nerves innervating different distal targets (skin and muscle), misdirection may, for example, lead to motor axons projecting toward skin, and vice versa—that is, sensory axons projecting toward muscle. In the repair of motor nerves innervating different distal targets, misdirection may result in reinnervation of the wrong target muscle, which might function antagonistically. In sensory nerve repair, misdirection might give an increased perceptual territory. After median nerve repair, for example, this might lead to a dysfunctional hand.
Different factors may be involved in the misdirection of regenerating axons, and there may be various mechanisms that can later correct for misdirection. In this review the authors discuss these different factors and mechanisms that act along the pathway of the regenerating axon. The authors review recently developed evaluation methods that can be used to investigate the accuracy of regeneration after nerve injury and repair (including the use of transgenic fluorescent mice, retrograde tracing techniques, and motion analysis). In addition, the authors discuss new strategies that can improve in vivo guidance of regenerating axons (including physical guidance with multichannel nerve tubes and biological guidance accomplished using gene therapy).
Collapse
Affiliation(s)
| | | | - Joost Verhaagen
- 3Department of Neuroregeneration, Netherlands Institute for Neuroscience, Amsterdam
- 4Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognition Research, Vrije Universiteit Amsterdam, The Netherlands; and
| | - Martijn J. A. Malessy
- 1Department of Neurosurgery, Leiden University Medical Center, Leiden
- 3Department of Neuroregeneration, Netherlands Institute for Neuroscience, Amsterdam
| |
Collapse
|
23
|
Romero-Ortega M. Peripheral Nerves, Anatomy and Physiology of. ENCYCLOPEDIA OF COMPUTATIONAL NEUROSCIENCE 2014:1-5. [DOI: 10.1007/978-1-4614-7320-6_214-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 03/06/2014] [Indexed: 09/02/2023]
|
24
|
Doxycycline-regulated GDNF expression promotes axonal regeneration and functional recovery in transected peripheral nerve. J Control Release 2013; 172:841-51. [PMID: 24140746 DOI: 10.1016/j.jconrel.2013.10.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 09/25/2013] [Accepted: 10/04/2013] [Indexed: 12/27/2022]
Abstract
Increased production of neurotrophic factors (NTFs) is one of the key responses seen following peripheral nerve injury, making them an attractive choice for pro-regenerative gene therapies. However, the downside of over-expression of certain NTFs, including glial cell line-derived neurotrophic factor (GDNF), was earlier found to be the trapping and misdirection of regenerating axons, the so-called 'candy-store' effect. We report a proof-of-principle study on the application of conditional GDNF expression system in injured peripheral nerve. We engineered Schwann cells (SCs) using dendrimers or lentiviral transduction with the vector providing doxycycline-regulated GDNF expression. Injection of GDNF-modified cells into the injured peripheral nerve followed by time-restricted administration of doxycycline demonstrated that GDNF expression in SCs can also be controlled locally in the peripheral nerves of the experimental animals. Cell-based GDNF therapy was shown to increase the extent of axonal regeneration, while controlled deactivation of GDNF effectively prevented trapping of regenerating axons in GDNF-enriched areas, and was associated with improved functional recovery.
Collapse
|
25
|
Godinho MJ, Teh L, Pollett MA, Goodman D, Hodgetts SI, Sweetman I, Walters M, Verhaagen J, Plant GW, Harvey AR. Immunohistochemical, ultrastructural and functional analysis of axonal regeneration through peripheral nerve grafts containing Schwann cells expressing BDNF, CNTF or NT3. PLoS One 2013; 8:e69987. [PMID: 23950907 PMCID: PMC3739754 DOI: 10.1371/journal.pone.0069987] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 06/14/2013] [Indexed: 01/13/2023] Open
Abstract
We used morphological, immunohistochemical and functional assessments to determine the impact of genetically-modified peripheral nerve (PN) grafts on axonal regeneration after injury. Grafts were assembled from acellular nerve sheaths repopulated ex vivo with Schwann cells (SCs) modified to express brain-derived neurotrophic factor (BDNF), a secretable form of ciliary neurotrophic factor (CNTF), or neurotrophin-3 (NT3). Grafts were used to repair unilateral 1 cm defects in rat peroneal nerves and 10 weeks later outcomes were compared to normal nerves and various controls: autografts, acellular grafts and grafts with unmodified SCs. The number of regenerated βIII-Tubulin positive axons was similar in all grafts with the exception of CNTF, which contained the fewest immunostained axons. There were significantly lower fiber counts in acellular, untransduced SC and NT3 groups using a PanNF antibody, suggesting a paucity of large caliber axons. In addition, NT3 grafts contained the greatest number of sensory fibres, identified with either IB4 or CGRP markers. Examination of semi- and ultra-thin sections revealed heterogeneous graft morphologies, particularly in BDNF and NT3 grafts in which the fascicular organization was pronounced. Unmyelinated axons were loosely organized in numerous Remak bundles in NT3 grafts, while the BDNF graft group displayed the lowest ratio of umyelinated to myelinated axons. Gait analysis revealed that stance width was increased in rats with CNTF and NT3 grafts, and step length involving the injured left hindlimb was significantly greater in NT3 grafted rats, suggesting enhanced sensory sensitivity in these animals. In summary, the selective expression of BDNF, CNTF or NT3 by genetically modified SCs had differential effects on PN graft morphology, the number and type of regenerating axons, myelination, and locomotor function.
Collapse
Affiliation(s)
- Maria João Godinho
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, Western Australia, Australia
| | - Lip Teh
- Cranio-Maxillo-Facial Unit, Princess Margaret Hospital for Children, Perth, Western Australia, Australia
| | - Margaret A. Pollett
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, Western Australia, Australia
| | - Douglas Goodman
- School of Veterinary and Biomedical Sciences, Murdoch University, Murdoch, Western Australia, Australia
| | - Stuart I. Hodgetts
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, Western Australia, Australia
| | - Iain Sweetman
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, Western Australia, Australia
| | - Mark Walters
- Cranio-Maxillo-Facial Unit, Princess Margaret Hospital for Children, Perth, Western Australia, Australia
| | - Joost Verhaagen
- Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Giles W. Plant
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, Western Australia, Australia
| | - Alan R. Harvey
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, Western Australia, Australia
- * E-mail:
| |
Collapse
|
26
|
Lankford KL, Arroyo EJ, Liu CN, Somps CJ, Zorbas MA, Shelton DL, Evans MG, Hurst SI, Kocsis JD. Sciatic nerve regeneration is not inhibited by anti-NGF antibody treatment in the adult rat. Neuroscience 2013; 241:157-69. [PMID: 23531437 DOI: 10.1016/j.neuroscience.2013.03.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Revised: 03/11/2013] [Accepted: 03/12/2013] [Indexed: 12/31/2022]
Abstract
Elevated nerve growth factor (NGF) is believed to play a role in many types of pain. An NGF-blocking antibody (muMab 911) has been shown to reduce pain and hyperalgesia in pain models, suggesting a novel therapeutic approach for pain management. Since NGF also plays important roles in peripheral nervous system development and sensory nerve outgrowth, we asked whether anti-NGF antibodies would adversely impact peripheral nerve regeneration. Adult rats underwent a unilateral sciatic nerve crush to transect axons and were subcutaneously dosed weekly for 8weeks with muMab 911 or vehicle beginning 1day prior to injury. Plasma levels of muMab 911 were assessed from blood samples and foot print analysis was used to assess functional recovery. At 8-weeks post-nerve injury, sciatic nerves were prepared for light and electron microscopy. In a separate group, Fluro-Gold was injected subcutaneously at the ankle prior to perfusion, and counts and sizes of retrogradely labeled and unlabeled dorsal root ganglion neurons were obtained. There was no difference in the time course of gait recovery in antibody-treated and vehicle-treated animals. The number of myelinated and nonmyelinated axons was the same in the muMab 911-treated crushed nerves and intact nerves, consistent with observed complete recovery. Treatment with muMab 911 did however result in a small decrease in average cell body size on both the intact and injured sides. These results indicate that muMab 911 did not impair functional recovery or nerve regeneration after nerve injury in adult rats.
Collapse
Affiliation(s)
- K L Lankford
- Department of Neurology, Yale University School of Medicine and Center for Neuroscience & Regeneration Research, VA CT Healthcare System, West Haven, CT 06516, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Cassereau J, Nicolas G, Lonchampt P, Pinier M, Barthelaix A, Eyer J, Letournel F. Axonal regeneration is compromised in NFH-LacZ transgenic mice but not in NFH-GFP mice. Neuroscience 2013; 228:101-8. [DOI: 10.1016/j.neuroscience.2012.10.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Revised: 10/04/2012] [Accepted: 10/04/2012] [Indexed: 11/26/2022]
|
28
|
Ferreira LMR, Floriddia EM, Quadrato G, Di Giovanni S. Neural Regeneration: Lessons from Regenerating and Non-regenerating Systems. Mol Neurobiol 2012; 46:227-41. [DOI: 10.1007/s12035-012-8290-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2011] [Accepted: 06/07/2012] [Indexed: 12/22/2022]
|
29
|
Early regenerative effects of NGF-transduced Schwann cells in peripheral nerve repair. Mol Cell Neurosci 2012; 50:103-12. [PMID: 22735691 DOI: 10.1016/j.mcn.2012.04.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Revised: 03/27/2012] [Accepted: 04/06/2012] [Indexed: 01/04/2023] Open
Abstract
Peripheral nerve injury leads to a rapid and robust increase in the synthesis of neurotrophins which guide and support regenerating axons. To further optimize neurotrophin supply at the earliest stages of regeneration, we over-expressed NGF in Schwann cells (SCs) by transducing these cells with a lentiviral vector encoding NGF (NGF-SCs). Transplantation of NGF-SCs in a rat sciatic nerve transection/repair model led to significant increase of NGF levels 2weeks after injury and correspondingly to substantial improvement in axonal regeneration. Numbers of NF200, ChAT and CGRP-positive axon profiles, as well as the gastrocnemius muscle weights, were significantly higher in the NGF-Schwann cell group compared to the animals that received control SCs transduced with a lentiviral vector encoding GFP (GFP-SCs). Comparison with other models of NGF application signifies the important role of this neurotrophin during the early stages of regeneration, and supports the importance of developing combined gene and cell therapy for peripheral nerve repair.
Collapse
|
30
|
McCall J, Weidner N, Blesch A. Neurotrophic factors in combinatorial approaches for spinal cord regeneration. Cell Tissue Res 2012; 349:27-37. [PMID: 22526621 DOI: 10.1007/s00441-012-1388-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Accepted: 02/23/2012] [Indexed: 01/09/2023]
Abstract
Axonal regeneration is inhibited by a plethora of different mechanisms in the adult central nervous system (CNS). While neurotrophic factors have been shown to stimulate axonal growth in numerous animal models of nervous system injury, a lack of suitable growth substrates, an insufficient activation of neuron-intrinsic regenerative programs, and extracellular inhibitors of regeneration limit the efficacy of neurotrophic factor delivery for anatomical and functional recovery after spinal cord injury. Thus, growth-stimulating factors will likely have to be combined with other treatment approaches to tap into the full potential of growth factor therapy for axonal regeneration. In addition, the temporal and spatial distribution of growth factors have to be tightly controlled to achieve biologically active concentrations, to allow for the chemotropic guidance of axons, and to prevent adverse effects related to the widespread distribution of neurotrophic factors. Here, we will review the rationale for combinatorial treatments in axonal regeneration and summarize some recent progress in promoting axonal regeneration in the injured CNS using such approaches.
Collapse
Affiliation(s)
- Julianne McCall
- Spinal Cord Injury Center, Heidelberg University Hospital, Schlierbacher Landstrasse 200 a, 69118 Heidelberg, Germany
| | | | | |
Collapse
|
31
|
Li BH, Kim SM, Yoo SB, Kim MJ, Jahng JW, Lee JH. Recombinant human nerve growth factor (rhNGF-β) gene transfer promotes regeneration of crush-injured mental nerve in rats. Oral Surg Oral Med Oral Pathol Oral Radiol 2012; 113:e26-34. [DOI: 10.1016/j.tripleo.2011.07.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Revised: 06/22/2011] [Accepted: 07/11/2011] [Indexed: 10/14/2022]
|
32
|
Hoyng SA, Tannemaat MR, De Winter F, Verhaagen J, Malessy MJA. Nerve surgery and gene therapy: a neurobiological and clinical perspective. J Hand Surg Eur Vol 2011; 36:735-46. [PMID: 21914696 DOI: 10.1177/1753193411420348] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Despite major microsurgical improvements the clinical outcome of peripheral nerve surgery is still regarded as suboptimal. Over the past decade several innovative techniques have been developed to extend the armamentarium of the nerve surgeon. This review evaluates the potential of gene therapy in the context of peripheral nerve repair. First the main challenges impeding peripheral nerve regeneration are presented. This is followed by a short introduction to gene therapy and an overview of its most important advantages over the classical delivery of therapeutic proteins. Next, this review focuses on the most promising viral vectors capable of targeting the peripheral nervous system and their first application in animal models. In addition, the challenges of translating these experimental results to the clinic, the limitations of current vectors and the further developments needed, are discussed. Finally, four strategies are presented on how gene therapy could help patients that have to undergo reconstructive nerve surgery in the future.
Collapse
Affiliation(s)
- S A Hoyng
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, Royal Academy of Arts and Sciences, Amsterdam, The Netherlands.
| | | | | | | | | |
Collapse
|
33
|
Lotfi P, Garde K, Chouhan AK, Bengali E, Romero-Ortega MI. Modality-specific axonal regeneration: toward selective regenerative neural interfaces. FRONTIERS IN NEUROENGINEERING 2011; 4:11. [PMID: 22016734 PMCID: PMC3191531 DOI: 10.3389/fneng.2011.00011] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Accepted: 09/26/2011] [Indexed: 01/08/2023]
Abstract
Regenerative peripheral nerve interfaces have been proposed as viable alternatives for the natural control of robotic prosthetic devices. However, sensory and motor axons at the neural interface are of mixed sub-modality types, which difficult the specific recording from motor axons and the eliciting of precise sensory modalities through selective stimulation. Here we evaluated the possibility of using type specific neurotrophins to preferentially entice the regeneration of defined axonal populations from transected peripheral nerves into separate compartments. Segregation of mixed sensory fibers from dorsal root ganglion neurons was evaluated in vitro by compartmentalized diffusion delivery of nerve growth factor (NGF) and neurotrophin-3 (NT-3), to preferentially entice the growth of TrkA+ nociceptive and TrkC+ proprioceptive subsets of sensory neurons, respectively. The average axon length in the NGF channel increased 2.5-fold compared to that in saline or NT-3, whereas the number of branches increased threefold in the NT-3 channels. These results were confirmed using a 3D “Y”-shaped in vitro assay showing that the arm containing NGF was able to entice a fivefold increase in axonal length of unbranched fibers. To address if such segregation can be enticed in vivo, a “Y”-shaped tubing was used to allow regeneration of the transected adult rat sciatic nerve into separate compartments filled with either NFG or NT-3. A significant increase in the number of CGRP+ pain fibers were attracted toward the sural nerve, while N-52+ large-diameter axons were observed in the tibial and NT-3 compartments. This study demonstrates the guided enrichment of sensory axons in specific regenerative chambers, and supports the notion that neurotrophic factors can be used to segregate sensory and perhaps motor axons in separate peripheral interfaces.
Collapse
Affiliation(s)
- Parisa Lotfi
- Department of Bioengineering, University of Texas at Arlington Arlington, TX, USA
| | | | | | | | | |
Collapse
|
34
|
Epidermal neural crest stem cell (EPI-NCSC)--mediated recovery of sensory function in a mouse model of spinal cord injury. Stem Cell Rev Rep 2010; 6:186-98. [PMID: 20414748 PMCID: PMC2887506 DOI: 10.1007/s12015-010-9152-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Here we show that epidermal neural crest stem cell (EPI-NCSC) transplants in the contused spinal cord caused a 24% improvement in sensory connectivity and a substantial recovery of touch perception. Furthermore we present a novel method for the ex vivo expansion of EPI-NCSC into millions of stem cells that takes advantage of the migratory ability of neural crest stem cells and is based on a new culture medium and the use of microcarriers. Functional improvement was shown by two independent methods, spinal somatosensory evoked potentials (SpSEP) and the Semmes-Weinstein touch test. Subsets of transplanted cells differentiated into myelinating oligodendrocytes. Unilateral injections of EPI-NCSC into the lesion of midline contused mouse spinal cords elicited bilateral improvements. Intraspinal EPI-NCSC did not migrate laterally in the spinal cord or invade the spinal roots and dorsal root ganglia, thus implicating diffusible factors. EPI-NCSC expressed neurotrophic factors, angiogenic factors, and metalloproteases. The strength of EPI-NCSC thus is that they can exert a combination of pertinent functions in the contused spinal cord, including cell replacement, neuroprotection, angiogenesis and modulation of scar formation. EPI-NCSC are uniquely qualified for cell-based therapy in spinal cord injury, as neural crest cells and neural tube stem cells share a higher order stem cell and are thus ontologically closely related.
Collapse
|
35
|
Smith GM, Onifer SM. Construction of pathways to promote axon growth within the adult central nervous system. Brain Res Bull 2010; 84:300-5. [PMID: 20554000 DOI: 10.1016/j.brainresbull.2010.05.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2010] [Revised: 05/25/2010] [Accepted: 05/31/2010] [Indexed: 12/12/2022]
Abstract
Inducing significant axon growth or regeneration after spinal cord injury has been difficult, primarily due to the poor growth supportive environment and low intrinsic growth ability of neurons within the CNS. Neurotrophins alone have been shown to readily induce regeneration of sensory axons after dorsal root lesions, however if neurotrophin gradients are expressed within the spinal cord these axons fail to terminate within appropriate target regions. Under such conditions, addition of a "stop" signal reduces growth into deeper dorsal laminae to support more specific targeting. Such neurotrophin gradients alone lose their effectiveness when lesions are within the spinal cord, requiring a combined treatment regime. Construction of pathways using combined treatments support good regeneration when they increase the intrinsic growth properties of neurons, provide a bridge across the lesion site, and supply a growth supportive substrate to induce axon growth out of the bridge and back into the host. Neurotrophin gradients distal to the bridge greatly enhance axon outgrowth. In disorders where neuronal circuits are lost, construction of preformed growth supportive pathways sustain long distance axon growth from a neuronal transplant to distal target locations.
Collapse
Affiliation(s)
- George M Smith
- Department of Physiology, Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, 40536, USA.
| | | |
Collapse
|