1
|
Pottorf TS, Rotterman TM, McCallum WM, Haley-Johnson ZA, Alvarez FJ. The Role of Microglia in Neuroinflammation of the Spinal Cord after Peripheral Nerve Injury. Cells 2022; 11:cells11132083. [PMID: 35805167 PMCID: PMC9265514 DOI: 10.3390/cells11132083] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 06/26/2022] [Accepted: 06/27/2022] [Indexed: 12/12/2022] Open
Abstract
Peripheral nerve injuries induce a pronounced immune reaction within the spinal cord, largely governed by microglia activation in both the dorsal and ventral horns. The mechanisms of activation and response of microglia are diverse depending on the location within the spinal cord, type, severity, and proximity of injury, as well as the age and species of the organism. Thanks to recent advancements in neuro-immune research techniques, such as single-cell transcriptomics, novel genetic mouse models, and live imaging, a vast amount of literature has come to light regarding the mechanisms of microglial activation and alluding to the function of microgliosis around injured motoneurons and sensory afferents. Herein, we provide a comparative analysis of the dorsal and ventral horns in relation to mechanisms of microglia activation (CSF1, DAP12, CCR2, Fractalkine signaling, Toll-like receptors, and purinergic signaling), and functionality in neuroprotection, degeneration, regeneration, synaptic plasticity, and spinal circuit reorganization following peripheral nerve injury. This review aims to shed new light on unsettled controversies regarding the diversity of spinal microglial-neuronal interactions following injury.
Collapse
Affiliation(s)
- Tana S. Pottorf
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA; (T.S.P.); (W.M.M.); (Z.A.H.-J.)
| | - Travis M. Rotterman
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30318, USA;
| | - William M. McCallum
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA; (T.S.P.); (W.M.M.); (Z.A.H.-J.)
| | - Zoë A. Haley-Johnson
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA; (T.S.P.); (W.M.M.); (Z.A.H.-J.)
| | - Francisco J. Alvarez
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA; (T.S.P.); (W.M.M.); (Z.A.H.-J.)
- Correspondence:
| |
Collapse
|
2
|
Arthur-Farraj P, Coleman MP. Lessons from Injury: How Nerve Injury Studies Reveal Basic Biological Mechanisms and Therapeutic Opportunities for Peripheral Nerve Diseases. Neurotherapeutics 2021; 18:2200-2221. [PMID: 34595734 PMCID: PMC8804151 DOI: 10.1007/s13311-021-01125-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2021] [Indexed: 12/25/2022] Open
Abstract
Since Waller and Cajal in the nineteenth and early twentieth centuries, laboratory traumatic peripheral nerve injury studies have provided great insight into cellular and molecular mechanisms governing axon degeneration and the responses of Schwann cells, the major glial cell type of peripheral nerves. It is now evident that pathways underlying injury-induced axon degeneration and the Schwann cell injury-specific state, the repair Schwann cell, are relevant to many inherited and acquired disorders of peripheral nerves. This review provides a timely update on the molecular understanding of axon degeneration and formation of the repair Schwann cell. We discuss how nicotinamide mononucleotide adenylyltransferase 2 (NMNAT2) and sterile alpha TIR motif containing protein 1 (SARM1) are required for axon survival and degeneration, respectively, how transcription factor c-JUN is essential for the Schwann cell response to nerve injury and what each tells us about disease mechanisms and potential therapies. Human genetic association with NMNAT2 and SARM1 strongly suggests aberrant activation of programmed axon death in polyneuropathies and motor neuron disorders, respectively, and animal studies suggest wider involvement including in chemotherapy-induced and diabetic neuropathies. In repair Schwann cells, cJUN is aberrantly expressed in a wide variety of human acquired and inherited neuropathies. Animal models suggest it limits axon loss in both genetic and traumatic neuropathies, whereas in contrast, Schwann cell secreted Neuregulin-1 type 1 drives onion bulb pathology in CMT1A. Finally, we discuss opportunities for drug-based and gene therapies to prevent axon loss or manipulate the repair Schwann cell state to treat acquired and inherited neuropathies and neuronopathies.
Collapse
Affiliation(s)
- Peter Arthur-Farraj
- Department of Clinical Neurosciences, John Van Geest Centre for Brain Repair, University of Cambridge, Robinson Way, Cambridge, CB2 0PY, UK.
| | - Michael P Coleman
- Department of Clinical Neurosciences, John Van Geest Centre for Brain Repair, University of Cambridge, Robinson Way, Cambridge, CB2 0PY, UK.
| |
Collapse
|
3
|
Nashine S. Potential Therapeutic Candidates for Age-Related Macular Degeneration (AMD). Cells 2021; 10:cells10092483. [PMID: 34572131 PMCID: PMC8464988 DOI: 10.3390/cells10092483] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/02/2021] [Accepted: 09/09/2021] [Indexed: 12/27/2022] Open
Abstract
Aging contributes to the risk of development of ocular diseases including, but not limited to, Age-related Macular Degeneration (AMD) that is a leading cause of blindness in the United States as well as worldwide. Retinal aging, that contributes to AMD pathogenesis, is characterized by accumulation of drusen deposits, alteration in the composition of Bruch’s membrane and extracellular matrix, vascular inflammation and dysregulation, mitochondrial dysfunction, and accumulation of reactive oxygen species (ROS), and subsequent retinal pigment epithelium (RPE) cell senescence. Since there are limited options available for the prophylaxis and treatment of AMD, new therapeutic interventions are constantly being looked into to identify new therapeutic targets for AMD. This review article discusses the potential candidates for AMD therapy and their known mechanisms of cytoprotection in AMD. These target therapeutic candidates include APE/REF-1, MRZ-99030, Ciliary NeuroTrophic Factor (CNTF), RAP1 GTPase, Celecoxib, and SS-31/Elamipretide.
Collapse
Affiliation(s)
- Sonali Nashine
- Department of Ophthalmology, University of California Irvine, Irvine, CA 92697, USA
| |
Collapse
|
4
|
Manthou ME, Gencheva D, Sinis N, Rink S, Papamitsou T, Abdulla D, Bendella H, Sarikcioglu L, Angelov DN. Facial Nerve Repair by Muscle-Vein Conduit in Rats: Functional Recovery and Muscle Reinnervation. Tissue Eng Part A 2020; 27:351-361. [PMID: 32731808 DOI: 10.1089/ten.tea.2020.0045] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The facial nerve is the most frequently damaged nerve in head and neck traumata. Repair of interrupted nerves is generally reinforced by fine microsurgical techniques; nevertheless, regaining all functions is the exception rather than the rule. The so-called "postparalytic syndrome," which includes synkinesia and altered blink reflexes, follows nerve injury. The purpose of this study was to examine if nerve-gap repair using an autologous vein filled with skeletal muscle would improve axonal regeneration, reduce neuromuscular junction polyinnervation, and improve the recovery of whisking in rats with transected and sutured right buccal branches of the facial nerve. Vibrissal motor performance was studied with the use of a video motion analysis. Immunofluorescence was used to visualize and analyze target muscle reinnervation. The results taken together indicate a positive effect of muscle-vein-combined conduit (MVCC) on the improvement of the whisking function after reparation of the facial nerve in rats. The findings support the recent suggestion that a venal graft with implantation of a trophic source, such as autologous denervated skeletal muscle, may promote the monoinnervation degree and ameliorate coordinated function of the corresponding muscles.
Collapse
Affiliation(s)
- Maria Eleni Manthou
- Department of Histology and Embryology, Medical Faculty, School of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece.,Department of Anatomy I, University of Cologne, Cologne, Germany
| | - Dilyana Gencheva
- Department of Anatomy I, University of Cologne, Cologne, Germany
| | - Nektarios Sinis
- Privatklinik für Plastische- und Ästhetische Chirurgie, Berlin Wilmersdorf, Germany
| | - Svenja Rink
- Department of Prosthetic Dentistry, School of Dental and Oral Medicine, University of Cologne, Cologne, Germany
| | - Theodora Papamitsou
- Department of Histology and Embryology, Medical Faculty, School of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Diana Abdulla
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Habib Bendella
- Department of Neurosurgery, University of Witten/Herdecke, Cologne Merheim Medical Center (CMMC), Cologne, Germany
| | | | | |
Collapse
|
5
|
Houshyar S, Bhattacharyya A, Shanks R. Peripheral Nerve Conduit: Materials and Structures. ACS Chem Neurosci 2019; 10:3349-3365. [PMID: 31273975 DOI: 10.1021/acschemneuro.9b00203] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Peripheral nerve injuries (PNIs) are the most common injury types to affect the nervous system. Restoration of nerve function after PNI is a challenging medical issue. Extended gaps in transected peripheral nerves are only repaired using autologous nerve grafting. This technique, however, in which nerve tissue is harvested from a donor site and grafted onto a recipient site in the same body, has many limitations and disadvantages. Recent studies have revealed artificial nerve conduits as a promising alternative technique to substitute autologous nerves. This Review summarizes different types of artificial nerve grafts used to repair peripheral nerve injuries. These include synthetic and natural polymers with biological factors. Then, desirable properties of nerve guides are discussed based on their functionality and effectiveness. In the final part of this Review, fabrication methods and commercially available nerve guides are described.
Collapse
Affiliation(s)
- Shadi Houshyar
- School of Engineering, RMIT University, Melbourne, Victoria 3000, Australia
| | - Amitava Bhattacharyya
- Nanoscience and Technology, Department of Electronics and Communication, PSG College of Technology, Coimbatore − 641004, India
| | - Robert Shanks
- School of Science, RMIT University, Melbourne, Victoria 3000, Australia
| |
Collapse
|
6
|
Gallaher ZR, Steward O. Modest enhancement of sensory axon regeneration in the sciatic nerve with conditional co-deletion of PTEN and SOCS3 in the dorsal root ganglia of adult mice. Exp Neurol 2018; 303:120-133. [PMID: 29458059 DOI: 10.1016/j.expneurol.2018.02.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 01/09/2018] [Accepted: 02/15/2018] [Indexed: 11/25/2022]
Abstract
Axons within the peripheral nervous system are capable of regeneration, but full functional recovery is rare. Recent work has shown that conditional deletion of two key signaling inhibitors of the PI3K and Jak/Stat pathways-phosphatase and tensin homolog (PTEN) and suppressor of cytokine signaling-3 (SOCS3), respectively-promotes regeneration of normally non-regenerative central nervous system axons. Moreover, in studies of optic nerve regeneration, co-deletion of both PTEN and SOCS3 has an even greater effect. Here, we test the hypotheses (1) that PTEN deletion enhances axon regeneration following sciatic nerve crush and (2) that PTEN/SOCS3 co-deletion further promotes regeneration. PTENfl/fl and PTEN/SOCS3fl/fl mice received direct injections of AAV-Cre into the fourth and fifth lumbar dorsal root ganglia (DRG) two weeks prior to sciatic nerve crush. Western blot analysis of whole cell lysates from DRG using phospho-specific antibodies revealed that PTEN deletion did not enhance or prolong PI3K signaling following sciatic nerve crush. However, PTEN/SOCS3 co-deletion activated PI3K for at least 7 days post-injury in contrast to controls, where activation peaked at 3 days. Quantification of SCG10-expressing regenerating sensory axons in the sciatic nerve after crush injury revealed longer distance regeneration at 3 days post-injury with both PTEN and PTEN/SOCS3 co-deletion. Additionally, analysis of noxious thermosensation and mechanosensation with PTEN/SOCS3 co-deletion revealed enhanced sensation at 14 and 21 days after crush, respectively, after which all treatment groups reached the same functional plateau. These findings indicate that co-deletion of PTEN and SOCS3 results in modest but measureable enhancement of early regeneration of DRG axons following crush injury.
Collapse
Affiliation(s)
- Zachary R Gallaher
- Reeve-Irvine Research Center, Department of Anatomy and Neurobiology, School of Medicine, University of California Irvine, Irvine, CA 92697, USA.
| | - Oswald Steward
- Reeve-Irvine Research Center, Department of Anatomy and Neurobiology, School of Medicine, University of California Irvine, Irvine, CA 92697, USA; Department of Neurobiology and Behavior, Department of Neurosurgery, University of California Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
7
|
Grosheva M, Nohroudi K, Schwarz A, Rink S, Bendella H, Sarikcioglu L, Klimaschewski L, Gordon T, Angelov DN. Comparison of trophic factors' expression between paralyzed and recovering muscles after facial nerve injury. A quantitative analysis in time course. Exp Neurol 2016; 279:137-148. [PMID: 26940083 DOI: 10.1016/j.expneurol.2016.02.020] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 02/07/2016] [Accepted: 02/26/2016] [Indexed: 01/08/2023]
Abstract
After peripheral nerve injury, recovery of motor performance negatively correlates with the poly-innervation of neuromuscular junctions (NMJ) due to excessive sprouting of the terminal Schwann cells. Denervated muscles produce short-range diffusible sprouting stimuli, of which some are neurotrophic factors. Based on recent data that vibrissal whisking is restored perfectly during facial nerve regeneration in blind rats from the Sprague Dawley (SD)/RCS strain, we compared the expression of brain derived neurotrophic factor (BDNF), fibroblast growth factor-2 (FGF2), insulin growth factors 1 and 2 (IGF1, IGF2) and nerve growth factor (NGF) between SD/RCS and SD-rats with normal vision but poor recovery of whisking function after facial nerve injury. To establish which trophic factors might be responsible for proper NMJ-reinnervation, the transected facial nerve was surgically repaired (facial-facial anastomosis, FFA) for subsequent analysis of mRNA and proteins expressed in the levator labii superioris muscle. A complicated time course of expression included (1) a late rise in BDNF protein that followed earlier elevated gene expression, (2) an early increase in FGF2 and IGF2 protein after 2 days with sustained gene expression, (3) reduced IGF1 protein at 28 days coincident with decline of raised mRNA levels to baseline, and (4) reduced NGF protein between 2 and 14 days with maintained gene expression found in blind rats but not the rats with normal vision. These findings suggest that recovery of motor function after peripheral nerve injury is due, at least in part, to a complex regulation of lesion-associated neurotrophic factors and cytokines in denervated muscles. The increase of FGF-2 protein and concomittant decrease of NGF (with no significant changes in BDNF or IGF levels) during the first week following FFA in SD/RCS blind rats possibly prevents the distal branching of regenerating axons resulting in reduced poly-innervation of motor endplates.
Collapse
Affiliation(s)
- Maria Grosheva
- Department of Oto-Rhino-Laryngology, University of Cologne, Germany
| | | | - Alisa Schwarz
- Department of Anatomy I, University of Cologne, Germany
| | - Svenja Rink
- Department of Anatomy I, University of Cologne, Germany
| | - Habib Bendella
- Department of Neurosurgery, Hospital Merheim, University of Witten-Herdecke, Cologne, Germany
| | | | - Lars Klimaschewski
- Division of Neuroanatomy Innsbruck Medical University, 6020 Innsbruck, Austria
| | - Tessa Gordon
- Department of Surgery,The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | | |
Collapse
|
8
|
DeFrancesco-Lisowitz A, Lindborg JA, Niemi JP, Zigmond RE. The neuroimmunology of degeneration and regeneration in the peripheral nervous system. Neuroscience 2015; 302:174-203. [PMID: 25242643 PMCID: PMC4366367 DOI: 10.1016/j.neuroscience.2014.09.027] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 09/08/2014] [Accepted: 09/10/2014] [Indexed: 12/25/2022]
Abstract
Peripheral nerves regenerate following injury due to the effective activation of the intrinsic growth capacity of the neurons and the formation of a permissive pathway for outgrowth due to Wallerian degeneration (WD). WD and subsequent regeneration are significantly influenced by various immune cells and the cytokines they secrete. Although macrophages have long been known to play a vital role in the degenerative process, recent work has pointed to their importance in influencing the regenerative capacity of peripheral neurons. In this review, we focus on the various immune cells, cytokines, and chemokines that make regeneration possible in the peripheral nervous system, with specific attention placed on the role macrophages play in this process.
Collapse
Affiliation(s)
| | - J A Lindborg
- Department of Neurosciences, Case Western Reserve University, Cleveland OH 44106-4975
| | - J P Niemi
- Department of Neurosciences, Case Western Reserve University, Cleveland OH 44106-4975
| | - R E Zigmond
- Department of Neurosciences, Case Western Reserve University, Cleveland OH 44106-4975
| |
Collapse
|
9
|
Li B, Qiu T, Iyer KS, Yan Q, Yin Y, Xie L, Wang X, Li S. PRGD/PDLLA conduit potentiates rat sciatic nerve regeneration and the underlying molecular mechanism. Biomaterials 2015; 55:44-53. [PMID: 25934451 DOI: 10.1016/j.biomaterials.2015.03.028] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 03/09/2015] [Accepted: 03/15/2015] [Indexed: 02/01/2023]
Abstract
Peripheral nerve injury requires optimal conditions in both macro-environment and micro-environment for reestablishment. Though various strategies have been carried out to improve the macro-environment, the underlying molecular mechanism of axon regeneration in the micro-environment provided by nerve conduit remains unclear. In this study, the rat sciatic nerve of 10 mm defect was made and bridged by PRGD/PDLLA nerve conduit. We investigated the process of nerve regeneration using histological, functional and real time PCR analyses after implantation from 7 to 35 days. Our data demonstrated that the ciliary neurotrophic factor highly expressed and up-regulated the downstream signaling pathways, in the case of activated signals, the expressions of axon sprout relative proteins, such as tubulin and growth-associated protein-43, were strongly augmented. Taken together, these data suggest a possible mechanism of axon regeneration promoted by PRGD/PDLLA conduit, which created a micro-environment for enhancement of diffusion of neurotrophic factors secreted by the injured nerve stumps, and activation of molecular signal transduction involved in growth cone, to potentiate the nerve recovery.
Collapse
Affiliation(s)
- Binbin Li
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, PR China; Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan 430070, PR China
| | - Tong Qiu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, PR China; Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan 430070, PR China.
| | - K Swaminathan Iyer
- School of Chemistry and Biochemistry, The University of Western Australia, Crawley WA 6009, Australia
| | - Qiongjiao Yan
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, PR China; Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan 430070, PR China
| | - Yixia Yin
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, PR China; Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan 430070, PR China
| | - Lijuan Xie
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, PR China; Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan 430070, PR China
| | - Xinyu Wang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, PR China; Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan 430070, PR China
| | - Shipu Li
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, PR China; Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan 430070, PR China.
| |
Collapse
|
10
|
Hong AR, Hong SM, Shin YA. Effects of resistance training on muscle strength, endurance, and motor unit according to ciliary neurotrophic factor polymorphism in male college students. J Sports Sci Med 2014; 13:680-688. [PMID: 25177199 PMCID: PMC4126309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 06/16/2014] [Indexed: 06/03/2023]
Abstract
Changes in muscle mass and strength across the adult age span are variable and related to the ciliary neurotrophic factor (CNTF) genotype. In particular, a single CNTF haplotype (1357 G→A) is important for neuronal and muscular developments and may be associated with muscle strength response to resistance training. We examined whether CNTF genotype differentially influences the effect of resistance training on neuromuscular improvement in male college students. Resistance training of the upper extremities comprised 3 sets at 75%-85% intensity per 1 repetition maximum, 3 times a week, for a total of 8 weeks. We measured isokinetic muscle function of the elbow joint with regard to strength (60°/s) and endurance (180°/s) by using an isokinetic dynamometer. The biceps brachii (BB) and brachioradialis muscles were studied using surface electromyography with spike-triggered averaging to assess surface-detected motor unit potential (SMUP) area. After resistance training, the SMUP of the BB increased significantly at 60°/s (p < 0.05), but no difference in the CNTF genotype was observed. The SMUP of the BB at 180°/s increased significantly in the GG/AA genotype group compared with that in the GA genotype group (p < 0.05). The average power of the elbow flexor at 180°/s increased significantly after resistance training (p < 0.05), but again, no difference in the CNTF genotype was observed. Thus, improvements in muscle strength and endurance may have resulted directly from resistance training rather than from genetic factors related to nerves in muscle tissue. Key PointsResistance training improves muscle strength and endurance in young men.This improvement in muscular strength and endurance is irrespective of CNTF genotypes.
Collapse
Affiliation(s)
- Ae-Rim Hong
- Department of Exercise Prescription & Rehabilitation, College of Sports Science, Dankook University , Anseo-dong, Cheonan-si, Chungnam Republic of Korea
| | - Sang-Min Hong
- Department of Physical Education, College of Education, Dongguk University, Pildong-ro-1-gil , Jung-gu, Seoul, Republic of Korea
| | - Yun-A Shin
- Department of Exercise Prescription & Rehabilitation, College of Sports Science, Dankook University , Anseo-dong, Cheonan-si, Chungnam Republic of Korea
| |
Collapse
|
11
|
Yao D, Li M, Shen D, Ding F, Lu S, Zhao Q, Gu X. Expression changes and bioinformatic analysis of Wallerian degeneration after sciatic nerve injury in rat. Neurosci Bull 2013; 29:321-32. [PMID: 23700281 PMCID: PMC5561847 DOI: 10.1007/s12264-013-1340-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Accepted: 09/21/2012] [Indexed: 12/22/2022] Open
Abstract
Wallerian degeneration (WD) remains an important research topic. Many genes are differentially expressed during the process of WD, but the precise mechanisms responsible for these differentiations are not completely understood. In this study, we used microarrays to analyze the expression changes of the distal nerve stump at 0, 1, 4, 7, 14, 21 and 28 days after sciatic nerve injury in rats. The data revealed 6 076 differentially-expressed genes, with 23 types of expression, specifically enriched in genes associated with nerve development and axonogenesis, cytokine biosynthesis, cell differentiation, cytokine/chemokine production, neuron differentiation, cytokinesis, phosphorylation and axon regeneration. Kyoto Encyclopedia of Genes and Genomes pathway analysis gave findings related mainly to the MAPK signaling pathway, the Jak-STAT signaling pathway, the cell cycle, cytokine-cytokine receptor interaction, the p53 signaling pathway and the Wnt signaling pathway. Some key factors were NGF, MAG, CNTF, CTNNA2, p53, JAK2, PLCB1, STAT3, BDNF, PRKC, collagen II, FGF, THBS4, TNC and c-Src, which were further validated by real-time quantitative PCR, Western blot, and immunohistochemistry. Our findings contribute to a better understanding of the functional analysis of differentially-expressed genes in WD and may shed light on the molecular mechanisms of nerve degeneration and regeneration.
Collapse
Affiliation(s)
- Dengbing Yao
- Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, 226019 China
- School of Life Sciences, Nantong University, Nantong, 226019 China
| | - Meiyuan Li
- Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, 226019 China
| | - Dingding Shen
- Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, 226019 China
| | - Fei Ding
- Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, 226019 China
| | - Shibi Lu
- Key Laboratory of the People’s Liberation Army, Institute of Orthopaedics, Chinese PLA General Hospital, Beijing, 100853 China
| | - Qing Zhao
- Key Laboratory of the People’s Liberation Army, Institute of Orthopaedics, Chinese PLA General Hospital, Beijing, 100853 China
| | - Xiaosong Gu
- Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, 226019 China
| |
Collapse
|
12
|
Neurotrophic molecules in the treatment of neurodegenerative disease with focus on the retina: status and perspectives. Cell Tissue Res 2013; 353:205-18. [PMID: 23463189 DOI: 10.1007/s00441-013-1585-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Accepted: 02/07/2013] [Indexed: 01/19/2023]
Abstract
Neurotrophic factors are operationally defined as molecules that promote the survival and differentiation of neurons. Chemically, they belong to divergent classes of molecules but most of the classic neurotrophic factors are proteins. Together with stem cells, viral vectors and genetically engineered cells, they constitute important tools in neuroprotective and regenerative neurobiology. Protein neurotrophic molecules signal through receptors located on the cell membrane. Their downstream signaling exploits pathways that are often common to chemically different factors and frequently target a relatively restricted set of transcription factors, RNA interference and diverse molecular machinery involved in the life vs. death decisions of neurons. Application of neurotrophic factors with the aim of curing or, at least, improving the outcome of neurodegenerative diseases requires (1) profound knowledge of the complex molecular pathology of the disease, (2) the development of animal models as closely as possible resembling the human disease, (3) the identification of target cells to be addressed, (4) intense efforts in chemical engineering to ensure the stability of molecules or to design carriers and small analogs with the ability to cross the blood-brain barrier and (5) scrutinity with regard to possible side effects. Last, but not least, engineering efforts to optimize administration, e.g., by designing the right canulae and infusion devices, are important for the successful translation of preclinical advances into clinical benefit. This article presents selected examples of neurotrophic factors that are currently being tested in animal models or developed for transfer to the clinic, with a major focus on factors with the potential of becoming applicable in various forms of retinal degeneration.
Collapse
|
13
|
Selvaraj BT, Frank N, Bender FLP, Asan E, Sendtner M. Local axonal function of STAT3 rescues axon degeneration in the pmn model of motoneuron disease. ACTA ACUST UNITED AC 2013; 199:437-51. [PMID: 23109669 PMCID: PMC3483126 DOI: 10.1083/jcb.201203109] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Axonal maintenance, plasticity, and regeneration are influenced by signals from neighboring cells, in particular Schwann cells of the peripheral nervous system. Schwann cells produce neurotrophic factors, but the mechanisms by which ciliary neurotrophic factor (CNTF) and other neurotrophic molecules modify the axonal cytoskeleton are not well understood. In this paper, we show that activated signal transducer and activator of transcription-3 (STAT3), an intracellular mediator of the effects of CNTF and other neurotrophic cytokines, acts locally in axons of motoneurons to modify the tubulin cytoskeleton. Specifically, we show that activated STAT3 interacted with stathmin and inhibited its microtubule-destabilizing activity. Thus, ectopic CNTF-mediated activation of STAT3 restored axon elongation and maintenance in motoneurons from progressive motor neuronopathy mutant mice, a mouse model of motoneuron disease. This mechanism could also be relevant for other neurodegenerative diseases and provide a target for new therapies for axonal degeneration.
Collapse
|
14
|
Role of inflammation and cytokines in peripheral nerve regeneration. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2013; 108:173-206. [PMID: 24083435 DOI: 10.1016/b978-0-12-410499-0.00007-1] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This chapter provides a review of immune reactions involved in classic as well as alternative methods of peripheral nerve regeneration, and mainly with a view to understanding their beneficial effects. Axonal degeneration distal to nerve damage triggers a cascade of inflammatory events alongside injured nerve fibers known as Wallerian degeneration (WD). The early inflammatory reactions of WD comprise the complement system, arachidonic acid metabolites, and inflammatory mediators that are related to myelin fragmentation and activation of Schwann cells. Fine-tuned upregulation of the cytokine/chemokine network by Schwann cells activates resident and hematogenous macrophages to complete the clearance of axonal and myelin debris and stimulate regrowth of axonal sprouts. In addition to local effects, immune reactions of neuronal bodies and glial cells are also implicated in the survival and conditioning of neurons to regenerate severed nerves. Understanding of the cellular and molecular interactions between the immune system and peripheral nerve injury opens new possibilities for targeting inflammatory mediators to improve functional reinnervation.
Collapse
|
15
|
Patodia S, Raivich G. Downstream effector molecules in successful peripheral nerve regeneration. Cell Tissue Res 2012; 349:15-26. [PMID: 22580509 DOI: 10.1007/s00441-012-1416-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Accepted: 03/19/2012] [Indexed: 12/16/2022]
Abstract
The robust axon regeneration that occurs following peripheral nerve injury is driven by transcriptional activation of the regeneration program and by the expression of a wide range of downstream effector molecules from neuropeptides and neurotrophic factors to adhesion molecules and cytoskeletal adaptor proteins. These regeneration-associated effector molecules regulate the actin-tubulin machinery of growth-cones, integrate intracellular signalling and stimulatory and inhibitory signals from the local environment and translate them into axon elongation. In addition to the neuronally derived molecules, an important transcriptional component is found in locally activated Schwann cells and macrophages, which release a number of cytokines, growth factors and neurotrophins that support neuronal survival and axonal regeneration and that might provide directional guidance cues towards appropriate peripheral targets. This review aims to provide a comprehensive up-to-date account of the transcriptional regulation and functional role of these effector molecules and of the information that they can give us with regard to the organisation of the regeneration program.
Collapse
Affiliation(s)
- Smriti Patodia
- Centre for Perinatal Brain Protection and Repair, University College London, Chenies Mews 86-96, London, WC1E 6HX, UK
| | | |
Collapse
|
16
|
Jablonka S, Holtmann B, Sendtner M, Metzger F. Therapeutic effects of PEGylated insulin-like growth factor I in the pmn mouse model of motoneuron disease. Exp Neurol 2011; 232:261-9. [DOI: 10.1016/j.expneurol.2011.09.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Revised: 08/31/2011] [Accepted: 09/09/2011] [Indexed: 02/08/2023]
|
17
|
Wilkinson AE, McCormick AM, Leipzig ND. Central Nervous System Tissue Engineering: Current Considerations and Strategies. ACTA ACUST UNITED AC 2011. [DOI: 10.2200/s00390ed1v01y201111tis008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
18
|
Abstract
After axotomy of embryonic hippocampal neurons in vitro, some of the axotomized axons lose their identity, and new axons arise and grow. This axotomy-induced axonogenesis requires importin, suggesting that some injury-induced signals are transported via axons to elicit axonogenesis after axotomy. In this study, we show that STAT3 is activated in response to axotomy. Because STAT3 was co-immunoprecipitated with importin β in the axotomized neurons, we suggest that STAT3 is retrogradely transported as molecular cargo of importin α/β heterodimers. Indeed, inhibition of importin α binding with STAT3 resulted in the attenuation of axonogenesis. Silencing STAT3 blocked the axonogenesis, demonstrating that STAT3 is necessary for axotomy-induced axonogenesis. Furthermore, the overexpression of STAT3 enhanced axotomy-induced axonogenesis. Taken together, these results demonstrate that activation and retrograde transport of STAT3 in injured axons have key roles in the axotomy-induced axonogenesis of hippocampal neurons.
Collapse
|
19
|
Raivich G. Transcribing the path to neurological recovery-From early signals through transcription factors to downstream effectors of successful regeneration. Ann Anat 2011; 193:248-58. [PMID: 21501955 DOI: 10.1016/j.aanat.2011.01.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Revised: 01/16/2011] [Accepted: 01/19/2011] [Indexed: 11/29/2022]
Abstract
The peripheral nervous system is known to regenerate comparatively well and this ability is mirrored in the de novo expression or upregulation of a wide variety of molecules involved in axonal outgrowth starting with transcription factors, but also including growth-stimulating substances, guidance and cell adhesion molecules, intracellular signaling enzymes and proteins involved in regulating cell-surface cytoskeletal interactions. Recent studies using pharmacological agents, and global as well as neuron-selective gene inactivation techniques have shed light on those endogenous molecules that play a non-redundant role in mediating regenerative axonal outgrowth in vivo. The aim of the current review is to sketch the sequence of molecular events from early sensors of injury to transcription factors to downstream effectors that cooperate in successful regeneration and functional recovery.
Collapse
Affiliation(s)
- Gennadij Raivich
- Perinatal Brain Repair Group, Department of Obstetrics and Gynaecology, University College London, 86-96 Chenies Mews, London, UK.
| |
Collapse
|
20
|
Girolami EI, Bouhy D, Haber M, Johnson H, David S. Differential expression and potential role of SOCS1 and SOCS3 in Wallerian degeneration in injured peripheral nerve. Exp Neurol 2010; 223:173-82. [PMID: 19576891 PMCID: PMC2849922 DOI: 10.1016/j.expneurol.2009.06.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2009] [Revised: 06/23/2009] [Accepted: 06/24/2009] [Indexed: 01/27/2023]
Abstract
Pro-inflammatory chemokines and cytokines play an important role in Wallerian degeneration (WD) after peripheral nerve injury. These pro-inflammatory signals are "turned-off" in a timely manner to ensure that the inflammatory response in the injured nerve is limited. The factors that regulate the turning-off of the pro-inflammatory state are not fully understood. The suppressors of cytokine signaling (SOCS) proteins are potential candidates that could limit the inflammatory response by acting to regulate cytokine signaling at the intracellular level. In this work we show that the expression SOCS1 and SOCS3 proteins differ from each other during WD in the mouse sciatic nerve after cut/ligation and crush injuries. SOCS1 is mainly expressed by macrophages and its expression is inversely correlated with phosphorylation of JAK2 and STAT3 signaling proteins and the expression of pro-inflammatory cytokines IL-1beta and TNFalpha. In addition, treatment of cut/ligated nerves, which express lower levels of SOCS1 as compared to crush injury, with a SOCS1 mimetic peptide leads to a decrease in macrophage numbers at 14 days post-injury and reduces IL-1beta mRNA expression 1 day post-injury. In contrast, SOCS3 expression is restricted mainly to Schwann cells and is negatively correlated with the expression of IL-6 and LIF. These data suggest that SOCS1 and SOCS3 may play different roles in WD and provide a better understanding of some of the potential regulatory mechanisms that may control inflammation and regeneration in the injured peripheral nerve.
Collapse
Affiliation(s)
- Elizabeth I. Girolami
- Centre for Research in Neuroscience, The Research Institute of the McGill University Health Centre, Livingston Hall, Room L7−210, 1650 Cedar Ave., Montreal, Quebec, Canada H3G 1A4
| | - Delphine Bouhy
- Centre for Research in Neuroscience, The Research Institute of the McGill University Health Centre, Livingston Hall, Room L7−210, 1650 Cedar Ave., Montreal, Quebec, Canada H3G 1A4
| | - Michael Haber
- Centre for Research in Neuroscience, The Research Institute of the McGill University Health Centre, Livingston Hall, Room L7−210, 1650 Cedar Ave., Montreal, Quebec, Canada H3G 1A4
| | - Howard Johnson
- Department of Microbiology and Cell Science, University of Florida, PO Box 110700, Building 981, Room 1052, Gainesville, FL 32611, USA
| | - Samuel David
- Centre for Research in Neuroscience, The Research Institute of the McGill University Health Centre, Livingston Hall, Room L7−210, 1650 Cedar Ave., Montreal, Quebec, Canada H3G 1A4
| |
Collapse
|
21
|
Simon CM, Jablonka S, Ruiz R, Tabares L, Sendtner M. Ciliary neurotrophic factor-induced sprouting preserves motor function in a mouse model of mild spinal muscular atrophy. Hum Mol Genet 2009; 19:973-86. [PMID: 20022887 DOI: 10.1093/hmg/ddp562] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Proximal spinal muscular atrophy (SMA) is caused by homozygous loss or mutation of the SMN1 gene on human chromosome 5. Depending on the levels of SMN protein produced from a second SMN gene (SMN2), different forms of the disease are distinguished. In patients with milder forms of the disease, type III or type IV SMA that normally reach adulthood, enlargement of motor units is regularly observed. However, the underlying mechanisms are not understood. Smn(+/-) mice, a mouse model of type III/IV SMA, reveal progressive loss of motor neurons and denervation of motor endplates starting at 4 weeks of age. Loss of spinal motor neurons between 1 month and 12 months reaches 40%, whereas muscle strength is not reduced. In these animals, amplitude of single motor unit action potentials in the gastrocnemic muscle is increased more than 2-fold. Confocal analysis reveals pronounced sprouting of innervating motor axons. As ciliary neurotrophic factor (CNTF) is highly expressed in Schwann cells, we investigated its role for a compensatory sprouting response and maintenance of muscle strength in this mouse model. Genetic ablation of CNTF results in reduced sprouting and decline of muscle strength in Smn(+/-) mice. These findings indicate that CNTF is necessary for a sprouting response and thus enhances the size of motor units in skeletal muscles of Smn(+/-) mice. This compensatory mechanism could guide the way to new therapies for this motor neuron disease.
Collapse
Affiliation(s)
- Christian M Simon
- Institute for Clinical Neurobiology, Josef-Schneider-Str. 11, 97080 Wuerzburg, Germany
| | | | | | | | | |
Collapse
|
22
|
Andreassen CS, Jakobsen J, Flyvbjerg A, Andersen H. Expression of neurotrophic factors in diabetic muscle--relation to neuropathy and muscle strength. Brain 2009; 132:2724-33. [PMID: 19696031 DOI: 10.1093/brain/awp208] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Diabetic polyneuropathy can lead to atrophy and weakness of distally located striated muscles due to denervation. Lack of neurotrophic support is believed to contribute to the development of diabetic neuropathy. In this study, we measured the expression of nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), neurotrophin 3 (NT-3), neurotrophin 4 (NT-4) and ciliary neurotrophic factor (CNTF) in muscle biopsies taken from the gastrocnemic and deltoid muscles in 42 diabetic patients and 20 healthy control subjects. To express the distal neuropathic gradient and to reduce interindividual variation, a distal/proximal ratio between expression levels in the gastrocnemic and deltoid muscles was calculated for all neurotrophic factors. Neuropathic status was determined by clinical examination, electrophysiological studies and quantitative sensory examination in diabetic patients, and muscle strength at both the shoulder and ankle was assessed by isokinetic dynamometry. Distal/proximal ratios for NT-3 were lower in diabetic patients [median (range) 110.7 (39.8-546.8)] than in controls [157.6 (63.3-385.4); (P < 0.05)], and in neuropathic diabetic patients [107.1 (39.8-326.0)] versus patients without neuropathy [134.5 (46.6-546.8); (P < 0.005)]. Further, ratios for NT-3 were related to muscle strength (r(s) = 0.41, P < 0.01) and showed a tendency towards a negative relationship to the combined score of all measures of neuropathy [Neuropathy rank-sum score (NRSS)] (r(s) = -0.27, P = 0.09). Similar trends were observed for ratios for NT-4. Ratios for NGF (r(s) = -0.32, P < 0.05) and BDNF (r(s) = -0.32, P < 0.05) were related to NRSS, but not to muscle strength. Ratios for CNTF were higher in diabetic patients [64.6 (23.7-258.7)] compared with controls [50.2 (27.2-186.4); (P < 0.05)], but showed no relationship to neither NRSS nor muscle strength. Our results show that the expression of NT-3 is reduced in striated muscles in diabetic patients and is related to muscle weakness and neuropathy. We suggest that lack of NT-3 contributes to insufficient re-innervation leading to the loss of muscle strength in diabetic neuropathy.
Collapse
Affiliation(s)
- C S Andreassen
- Clinical Neurology Research Group, Department of Neurology, Noerrebrogade 44, Aarhus University Hospital, Aarhus C., Denmark.
| | | | | | | |
Collapse
|
23
|
Abstract
Clinical trials in amyotrophic lateral sclerosis have significantly evolved over the last decade. New outcome measures have been developed that have reduced the sample size requirement as compared with survival studies. There has been increasing recognition that dose-ranging studies are crucial to full evaluation of experimental agents. While the requirements of late stage trials have not changed, many new designs have been suggested for earlier phase development. While no design achieves the perfect balance of sensitivity and efficiency, clinical trialists continue to work toward the goals of smaller and shorter trials so that more compounds can be studied concurrently.
Collapse
|
24
|
Neuroprotective properties of ciliary neurotrophic factor for cultured adult rat dorsal root ganglion neurons. Histochem Cell Biol 2008; 130:669-79. [PMID: 18679704 DOI: 10.1007/s00418-008-0484-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2008] [Indexed: 12/15/2022]
Abstract
We observed that recombinant ciliary neurotrophic factor (CNTF) enhanced survival and neurite outgrowth of cultured adult rat dorsal root ganglion (DRG) neurons. Among other neurotrophic factors (NGF and GDNF) and interleukin (IL)-6 cytokine members [IL-6, LIF, cardiotrophin-1, and oncostatin M (OSM)] at the same concentration (50 ng/ml), CNTF, as well as LIF and OSM, displayed high efficacy for the promotion of the number of viable neurons and neurite-bearing cells. CNTF enhanced the number of neurite-bearing cells in both small neurons (soma diameter <30 microm) and large neurons (soma diameter > or =30 microm), whereas NGF and GDNF promoted that in only small neurons. Western blot analysis revealed that CNTF induced phosphorylation of STAT3, Akt, and ERK1/2 in the neurons. Furthermore, the neurite outgrowth-promoting activity of CNTF was diminished by co-treatment with Janus kinase (JAK) 2 inhibitor, AG490; STAT3 inhibitor, STA-21; phosphatidyl inositol-3'-phosphate-kinase (PI3K) inhibitor, LY294002; and mitogen-activated protein kinase kinase (MEK) inhibitor, PD98059, in a concentration-dependent manner. Its survival-promoting activity was also affected by AG490, STA-21, and LY294002 at higher concentrations, but not by PD98059. These findings suggest the involvement of JAK2/STAT3, PI3K/Akt, and MEK/ERK signaling pathways in CNTF-induced neurite outgrowth, where the former two pathways are thought to play major roles in mediating the survival response of neurons to CNTF.
Collapse
|
25
|
Kanungo AK, Hao Z, Elia AJ, Mak TW, Henderson JT. Inhibition of Apoptosome Activation Protects Injured Motor Neurons from Cell Death. J Biol Chem 2008; 283:22105-12. [DOI: 10.1074/jbc.m800988200] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
|
26
|
An Analysis of Time-Dependent Changes of Neurotrophic Factors (BDNF, CNTF) in Traumatic Facial Nerve Injury of a Nerve-Cut and Nerve-Crush Model in Rats. Otol Neurotol 2008; 29:392-6. [DOI: 10.1097/mao.0b013e318161ab3e] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
27
|
Park OH, Lee KJ, Rhyu IJ, Geum D, Kim H, Buss R, Oppenheim RW, Sun W. Bax-dependent and -independent death of motoneurons after facial nerve injury in adult mice. Eur J Neurosci 2007; 26:1421-32. [PMID: 17822434 DOI: 10.1111/j.1460-9568.2007.05787.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Nerve injury-induced neuronal death may occur after accidental trauma or nerve inflammation. Although the response to facial root avulsion has been examined in rodent models, there are conflicting results as to whether motoneuron (MN) death is mediated by apoptosis or necrosis. We examined the response of MNs and proximal nerves after facial nerve avulsion in adult mice. Following facial nerve avulsion in 4-5-week-old mice, we observed a progressive reduction of MNs such that by 4 weeks less than 10% of avulsed MNs remained compared with the control side. The profile of MN degeneration was distinct from axotomy-induced responses. For example, the onset of MN death was more rapid, and the extent of MN loss was greater compared with axotomy. Furthermore, the degeneration of oligodendrocytes and the activation of microglia were increased in the proximal nerve after avulsion. Ultrastructural observations suggested that root avulsion mainly induces non-apoptotic neuronal death, although a small subset of neurons appeared to die via apoptosis. To evaluate the contribution of apoptotic death, we evaluated MN responses in Bax-knockout (KO) mice in which neurons are rescued from apoptotic death. Surprisingly, although the majority of Bax-KO mice exhibited only a moderate MN loss after avulsion, a subset of Bax-KO mice (25%) exhibited extensive MN death and injury-induced changes in the nerve that were indistinguishable from events in wild-type littermates. These results suggest that both Bax-dependent and -independent forms of cell death are evoked by root avulsion, and that programmed cell death may be involved in triggering a robust necrotic response.
Collapse
Affiliation(s)
- Ok-hee Park
- Department of Anatomy, College of Medicine, Korea University, 126-1 Anam-Dong, Sungbuk-Gu, Seoul, Korea 136-705
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Goss JR. The therapeutic potential of gene transfer for the treatment of peripheral neuropathies. Expert Rev Mol Med 2007; 9:1-20. [PMID: 17367556 DOI: 10.1017/s1462399407000270] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Peripheral neuropathy is a common medical problem with numerous aetiologies. Unfortunately, for the majority of cases there is no available medical solution for the underlying cause, and the only option is to try to treat the resulting symptoms. Treatment options exist when neuropathy results in positive symptoms such as pain, but there is a significant lack of treatments for negative symptoms such as numbness and weakness. Systemic application of growth factor peptides has shown promise in protecting nerves from neuropathic insults in preclinical animal studies, but translation into human trials has been problematic and disappointing. Significant advancements have been made in the past few years in utilising gene therapy approaches to treat peripheral neuropathy by expressing neuroprotective gene products either systemically or in specific nervous tissues. For example, plasmids expressing vascular endothelial growth factor injected into muscle, or herpes-simplex-virus-based vectors expressing neurotrophin gene products delivered to dorsal root ganglion neurons, have been used to protect peripheral nerve function in animal models of diabetes-associated peripheral neuropathy. Many published studies support the feasibility of this approach, although several questions still need to be addressed as gene therapy to treat peripheral neuropathy moves out of the laboratory and into the clinic.
Collapse
Affiliation(s)
- James R Goss
- Molecular Genetics and Biochemistry, Center for Biotechnology and Bioengineering, University of Pittsburgh, 300 Technology Drive, Rm 208, Pittsburgh, PA 15219, USA.
| |
Collapse
|
29
|
Raivich G, Makwana M. The making of successful axonal regeneration: Genes, molecules and signal transduction pathways. ACTA ACUST UNITED AC 2007; 53:287-311. [PMID: 17079020 DOI: 10.1016/j.brainresrev.2006.09.005] [Citation(s) in RCA: 133] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2006] [Revised: 09/12/2006] [Accepted: 09/18/2006] [Indexed: 12/16/2022]
Abstract
Unlike its central counterpart, the peripheral nervous system is well known for its comparatively good potential for regeneration following nerve fiber injury. This ability is mirrored by the de novo expression or upregulation of a wide variety of molecules including transcription factors, growth-stimulating substances, cell adhesion molecules, intracellular signaling enzymes and proteins involved in regulating cell-surface cytoskeletal interactions, that promote neurite outgrowth in cultured neurons. However, their role in vivo is less known. Recent studies using neutralizing antibodies, gene inactivation and overexpression techniques have started to shed light on those endogenous molecules that play a key role in axonal outgrowth and the process of successful functional repair in the injured nervous system. The aim of the current review is to provide a summary on this rapidly growing field and the experimental techniques used to define the specific effects of candidate signaling molecules on axonal regeneration in vivo.
Collapse
Affiliation(s)
- Gennadij Raivich
- Perinatal Brain Repair Group, Department of Obstetrics and Gynaecology, University College London, 86-96 Chenies Mews, London, UK.
| | | |
Collapse
|
30
|
Ito Y, Wiese S, Funk N, Chittka A, Rossoll W, Bömmel H, Watabe K, Wegner M, Sendtner M. Sox10 regulates ciliary neurotrophic factor gene expression in Schwann cells. Proc Natl Acad Sci U S A 2006; 103:7871-6. [PMID: 16684879 PMCID: PMC1472537 DOI: 10.1073/pnas.0602332103] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Ciliary neurotrophic factor (Cntf) plays an essential role in postnatal maintenance of spinal motoneurons. Whereas the expression of this neurotrophic factor is low during embryonic development, it is highly up-regulated after birth in myelinating Schwann cells of rodents. To characterize the underlying transcriptional mechanisms, we have analyzed and compared the effects of various glial transcription factors. In contrast to Pit-1, Oct-1, Unc-86 homology region (POU) domain class 3, transcription factor 1 (Oct6/SCIP/Tst-1) and paired box gene 3 (Pax3), SRY-box-containing gene 10 (Sox10) induces Cntf expression in Schwann cells. Subsequent promoter analysis using luciferase reporter gene and EMSA identified the corresponding response elements within the Cntf promoter. Overexpression of Sox10 in primary sciatic nerve Schwann cells leads to a >100-fold up-regulation of Cntf protein, and suppression of Sox10 by RNA interference in the spontaneously immortalized Schwann cell line 32 reduces Cntf expression by >80%. Mice with heterozygous inactivation of the Sox10 gene show significantly reduced Cntf protein levels in sciatic nerves, indicating that Sox10 is necessary and sufficient for regulating Cntf expression in the peripheral nervous system.
Collapse
Affiliation(s)
- Yasuhiro Ito
- *Institute for Clinical Neurobiology, University of Wuerzburg, D-97080 Wuerzburg, Germany
| | - Stefan Wiese
- *Institute for Clinical Neurobiology, University of Wuerzburg, D-97080 Wuerzburg, Germany
| | - Natalja Funk
- *Institute for Clinical Neurobiology, University of Wuerzburg, D-97080 Wuerzburg, Germany
| | - Alexandra Chittka
- *Institute for Clinical Neurobiology, University of Wuerzburg, D-97080 Wuerzburg, Germany
| | - Wilfried Rossoll
- *Institute for Clinical Neurobiology, University of Wuerzburg, D-97080 Wuerzburg, Germany
| | - Heike Bömmel
- *Institute for Clinical Neurobiology, University of Wuerzburg, D-97080 Wuerzburg, Germany
| | - Kazuhiko Watabe
- Department of Molecular Neuropathology, Tokyo Metropolitan Institute for Neuroscience, 2-6 Musashidai, Fuchu-shi, Tokyo 183-8526, Japan; and
| | - Michael Wegner
- Institute of Biochemistry, Erlangen University, D-91054 Erlangen, Germany
| | - Michael Sendtner
- *Institute for Clinical Neurobiology, University of Wuerzburg, D-97080 Wuerzburg, Germany
- To whom correspondence should be addressed at:
Institute for Clinical Neurobiology, Josef-Schneider-Strasse 11, University of Wuerzburg, D-97080 Wuerzburg, Germany. E-mail:
| |
Collapse
|
31
|
Naumann T, Schnell O, Zhi Q, Kirsch M, Schubert KO, Sendtner M, Hofmann HD. Endogenous ciliary neurotrophic factor protects GABAergic, but not cholinergic, septohippocampal neurons following fimbria-fornix transection. Brain Pathol 2006; 13:309-21. [PMID: 12946020 PMCID: PMC8095902 DOI: 10.1111/j.1750-3639.2003.tb00030.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Application of neurotrophic proteins including ciliary neurotrophic factor (CNTF) and leukemia inhibitory factor (LIF), members of the family of gp130-associated cytokines, can rescue CNS neurons from injury-induced degeneration. However, it is not clear so far if these effects reflect a physiological function of the endogenous cytokines. Using fimbria-fornix transection as a model, we examined whether responses of GABAergic and cholinergic septohippocampal neurons to axotomy are altered in mice lacking CNTF. In addition, we studied the cellular expression of CNTF, LIF and related cytokine receptor components in the septal complex following lesion. Degeneration of septohippocampal GABAergic neurons in the medial septum as indicated by the loss of parvalbumin-immunoreactive neurons was accelerated and permanently enhanced in CNTF(-/-) mice as compared to wild-type animals. Unexpectedly, the number of axotomized cholinergic MS neurons was significantly higher in CNTF-deficient mice during the first 2 weeks postlesion. Both in wild-type and in CNTF(-/-) mutants, expression of mRNA for the CNTF-specific alpha-subunit of the cytokine receptor complex was specifically upregulated in axotomized GABAergic septal neurons, whereas enhanced expression of the LIF-binding beta-subunit was specifically observed in axotomized cholinergic neurons. Following lesion, CNTF expression in wild-type mice was induced in activated astrocytes surrounding the axotomized neurons and at the lesion site. Expression of LIF mRNA was localized in the GABAergic and cholinergic septohippocampal neurons. These results strongly indicate that endogenous CNTF, supplied by reactive glia cells, acts as a neuroprotective factor for axotomized CNS neurons. In the septum, endogenous CNTF specifically supports lesioned GABAergic projection neurons, whereas LIF may play a similar role for the cholinergic counterparts.
Collapse
|
32
|
Abstract
Neurotrophic factors are proteins which promote the survival of specific neuronal populations. Many have other physiological effects on neurons such as inducing morphological differentiation, enhancing nerve regeneration, stimulating neurotransmitter expression, and otherwise altering the physiological characteristics of neurons. These properties suggest that neurotrophic factors are highly promising as potential therapeutic agents for neurological disease. Neurotrophic factors will most likely be applied to the peripheral nervous system initially, since there are fewer problems for large proteins to gain access to peripheral neurons. Many of the most intensively studied factors are active in the peripheral nervous system. These include the neurotrophins (nerve growth factor, brain derived neurotrophic factor, neurotrophin-3, neurotrophin-4/5), the insulin like growth factors, ciliary neurotrophic factor, and glial cell derived neurotrophic factor and its related proteins. The biology of these factors and their receptors in the peripheral nervous system is reviewed here. We also review data suggesting that abnormal availability of some factors may contribute towards the pathogenesis of certain types of peripheral neuropathy. Finally, the pre-clinical data suggesting that individual factors might be effective in treating neuropathy is reviewed, along with data relating to possible side effects of neurotrophic factor therapy. Several factors have already entered clinical trials with variable success. The data from these trials is reviewed as well.
Collapse
Affiliation(s)
- S C Apfel
- Dept. of Neurology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
33
|
Kamholz J, Awatramani R, Menichella D, Jiang H, Xu W, Shy M. Regulation of Myelin-Specific Gene Expression: Relevance to CMT1. Ann N Y Acad Sci 2006; 883:91-108. [PMID: 29086995 DOI: 10.1111/j.1749-6632.1999.tb08572.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Schwann cells, the myelinating cells of the peripheral nervous system, are derived from the neural crest. Once neural crest cells are committed to the Schwann cell fate, they can take on one of two phenotypes to become myelinating or nonmyelinating Schwann cells, a decision that is determined by interactions with axons. The critical step in the differentiation of myelinating Schwann cells is the establishment of a one-to-one relationship with axons, the so-called "promyelinating" stage of Schwann cell development. The transition from the promyelinating to the myelinating stage of development is then accompanied by a number of significant changes in the pattern of gene expression, including the activation of a set of genes encoding myelin structural proteins and lipid biosynthetic enzymes, and the inactivation of a set of genes expressed only in immature or nonmyelinating Schwann cells. These changes are regulated mainly at the transcriptional level and also require continuous interaction between Schwann cells and their axons. Two transcription factors, Krox 20 (EGR2) and Oct 6 (SCIP/Tst1), are necessary for the transition from the promyelinating to the myelinating stage of Schwann cell development. Krox 20, expressed in myelinating but not promyelinating Schwann cells, is absolutely required for this transition, and myelination cannot occur in its absence. Oct 6, expressed mainly in promyelinating Schwann cells and then downregulated before myelination, is necessary for the correct timing of this transition, since myelination is delayed in its absence. Neither Krox 20 nor Oct 6, however, is required for the initial activation of myelin gene expression. Although the mechanisms of Krox 20 and Oct 6 action during myelination are not known, mutation in Krox 20 has been shown to cause CMT1, further implicating this protein in the pathogenesis of this disease. Identifying the molecular mechanisms of Krox 20 and Oct 6 action will thus be important both for understanding myelination and for designing future treatments for CMT1. Point mutations in the genes encoding the myelin proteins PMP22 and P0 cause CMT1A without a gene duplication and CMT1B, respectively. Although the clinical and pathological phenotypes of CMT1A and CMT1B are similar, their molecular pathogenesis is quite different. Point mutations in PMP22 alter the trafficking of the protein, so that it accumulates in the endoplasmic reticulum (ER) and intermediate compartment (IC). Mutant PMP22 also sequesters its normal counterpart in the ER, further reducing the amount of PMP22 available for myelin synthesis at the membrane, and accounting, at least in part, for its severe effect on myelination. Mutant PMP22 probably also activates an ER-to-nucleus signal transduction pathway associated with misfolded proteins, which may account for the decrease of myelin gene expression in Schwann cells in Trembler mutant mice. In contrast, absence of expression of the homotypic adhesion molecule, P0, in mice in which the gene has been inactivated, produces a unique pattern of Schwann cell gene expression, demonstrating that P0 plays a regulatory as well as a structural role in myelination. Whether this role is direct, through a P0-mediated adhesion pathway, or indirect, through adhesion pathways mediated by cadherins or integrins, however, remains to be determined. The molecular mechanisms underlying dysmyelination in CMT1 are thus complex, with pleitropic effects on Schwann cell physiology that are determined both by the type of mutation and the protein mutated. Identifying these molecular mechanisms, however, are important both for understanding myelination and for designing future treatments for CMT1. Although demyelination is the hallmark of CMT1, the clinical signs and symptoms of this disease are probably produced by axonal degeneration, not demyelination. Interestingly, a number of recent studies have demonstrated that Schwann cells from Trembler mice or patients with CMT1A can induce local axonal abnormalities, including decreased axonal transport, and altered neurofilament phosphorylation. These data thus suggest that disability of patients with CMT1 is caused by abnormal Schwann cell-axonal interactions. Efforts both to understand the effects of myelinating Schwann cells on their axons and to prevent axonal degeneration or promote axonal regeneration are thus central for the future development of a rational molecular therapy for CMT1.
Collapse
Affiliation(s)
- John Kamholz
- Department of Neurology, Wayne State University School of Medicine, Detroit, Michigan 48201, USAGraduate Program in Molecular Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USAInstitute of Neurology, University of Milan, IRCCS, Ospedale Maggiore, Policlinico, Milan, ItalyCenter for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan 48201, USA
| | - Raj Awatramani
- Department of Neurology, Wayne State University School of Medicine, Detroit, Michigan 48201, USAGraduate Program in Molecular Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USAInstitute of Neurology, University of Milan, IRCCS, Ospedale Maggiore, Policlinico, Milan, ItalyCenter for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan 48201, USA
| | - Daniela Menichella
- Department of Neurology, Wayne State University School of Medicine, Detroit, Michigan 48201, USAGraduate Program in Molecular Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USAInstitute of Neurology, University of Milan, IRCCS, Ospedale Maggiore, Policlinico, Milan, ItalyCenter for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan 48201, USA
| | - Huiyuan Jiang
- Department of Neurology, Wayne State University School of Medicine, Detroit, Michigan 48201, USAGraduate Program in Molecular Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USAInstitute of Neurology, University of Milan, IRCCS, Ospedale Maggiore, Policlinico, Milan, ItalyCenter for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan 48201, USA
| | - Wenbo Xu
- Department of Neurology, Wayne State University School of Medicine, Detroit, Michigan 48201, USAGraduate Program in Molecular Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USAInstitute of Neurology, University of Milan, IRCCS, Ospedale Maggiore, Policlinico, Milan, ItalyCenter for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan 48201, USA
| | - Michael Shy
- Department of Neurology, Wayne State University School of Medicine, Detroit, Michigan 48201, USAGraduate Program in Molecular Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USAInstitute of Neurology, University of Milan, IRCCS, Ospedale Maggiore, Policlinico, Milan, ItalyCenter for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan 48201, USA
| |
Collapse
|
34
|
Osamura N, Ikeda K, Ito T, Higashida H, Tomita K, Yokoyama S. Induction of interleukin-6 in dorsal root ganglion neurons after gradual elongation of rat sciatic nerve. Exp Neurol 2005; 195:61-70. [PMID: 15913608 DOI: 10.1016/j.expneurol.2005.03.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2005] [Revised: 03/25/2005] [Accepted: 03/30/2005] [Indexed: 11/22/2022]
Abstract
In the reconstruction of a segmental defect in injured peripheral nerves, gradual nerve elongation has become an important alternative to nerve grafting. To clarify biochemical responses in peripheral sensory neurons after nerve elongation, we examined the expression of cytokines and neurotrophins related to nerve regeneration. We first established rat elongation models by lengthening left femurs up to 20.0 mm at the rate of 1.0, 2.0, or 20.0 mm/day. In toluidine blue staining, the acutely elongated, 20-mm/day group showed nuclear eccentricity in the nerve cell body in L5 dorsal root ganglion (DRG) and axonal degeneration in the sciatic nerves; in contrast, the gradually elongated, 1- and 2-mm/day groups remained intact, indicating adaptation. Reverse transcription-polymerase chain reaction analysis revealed that interleukin-6 (IL-6) mRNA was induced in ipsilateral L4-6 DRG in an elongation rate-dependent manner. In contrast, none of the elongated groups exhibited a significant change in mRNA levels for interleukin-1beta, tumor necrosis factor-alpha, nerve growth factor, brain-derived neurotrophic factor, neurotropnin-3, and neurotrophin-4/5. Levels of IL-6 mRNA in all the elongated groups reached the maximum level at day 4 after 20-mm lengthening, while the axotomized group showed a decrease from the maximum level at day 1. Induction of IL-6 mRNA was also detected in the contralateral L4-6 DRG of all the elongated groups, but not detected in the axotomized group. In histochemical analysis, IL-6-immunoreactivity was predominant in neurofilament-positive, medium to large DRG neurons. Application of IL-6 to cultured Schwann cells increased mRNA for peripheral myelin protein 22 (PMP22), a major myelin component. These results suggest that IL-6 plays a key role in biochemical responses in peripheral sensory neurons after gradual nerve elongation.
Collapse
Affiliation(s)
- Naoki Osamura
- Department of Orthopaedic Surgery, School of Medicine, Kanazawa University, 13-1 Takaramachi, Kanazawa 920-8641, Japan.
| | | | | | | | | | | |
Collapse
|
35
|
Abstract
Peripheral nerve injury is normally followed by a robust regenerative response. Here we describe the early changes associated with injury from the initial rise in intracellular calcium and the subsequent activation of transcription factors and cytokines leading to an inflammatory reaction, and the expression of growth factors, cytokines, neuropeptides, and other secreted molecules involved in cell-to-cell communication promoting regeneration and neurite outgrowth. The aim of this review is to summarize the molecular mechanisms that play a part in executing successful regeneration.
Collapse
Affiliation(s)
- Milan Makwana
- Centre for Perinatal Brain Protection & Repair, Department of Obstetrics and Gynaecology, University College London, UK
| | | |
Collapse
|
36
|
Nishimune H, Bernreuther C, Carroll P, Chen S, Schachner M, Henderson CE. Neural adhesion molecules L1 and CHL1 are survival factors for motoneurons. J Neurosci Res 2005; 80:593-9. [PMID: 15880726 DOI: 10.1002/jnr.20517] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Many neurotrophic factors with survival activity for motoneurons in vivo were first identified using cultures of purified embryonic motoneurons. The L1 neural cell adhesion molecule has multiple roles in brain development. We showed by in situ hybridization and RT-PCR that L1 mRNA was expressed at significant levels in motoneurons of embryonic and postnatal spinal cord. We therefore cultured purified motoneurons from E14 rat embryos in the absence of trophic factors but with L1-Fc and CHL1-Fc fusion proteins. L1-Fc prevented the death of approximately half of the motoneurons that were saved by BDNF in a dose-dependent manner (EC50 = 10 pM). CHL1-Fc saved the same number of motoneurons as did L1-Fc, whereas P0-Fc had little neurotrophic activity at the same concentrations. Survival induced by L1 and CHL1 was completely inhibited by 20 microM LY294002 and PD98059, indicating that both MEK and PI3K pathways are required for signaling by these molecules. L1 can signal in other cell types through the FGF receptor FGFR1. In cultures of motoneurons, effects of suboptimal concentrations of L1 and suboptimal concentrations of FGF-2 were additive, but the effects of optimal concentrations of FGF-2 (50 ng/ml) were not further increased in the presence of L1-Fc. Thus, in this system, too, FGF and L1 may use similar signaling pathways.
Collapse
Affiliation(s)
- H Nishimune
- INSERM UMR623, IBDM (CNRS-INSERM-Univ. Mediterranee), Marseille, France
| | | | | | | | | | | |
Collapse
|
37
|
Malik MA, Blusztajn JK, Greenwood CE. Nutrients as trophic factors in neurons and the central nervous system: role of retinoic acid. J Nutr Biochem 2005; 11:2-13. [PMID: 15539337 DOI: 10.1016/s0955-2863(99)00066-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/1999] [Accepted: 09/28/1999] [Indexed: 12/21/2022]
Abstract
In multicellular organisms, death, survival, proliferation, and differentiation of a given cell depend on signals produced by neighboring and/or distant cells, resulting in the coordinated development and function of the various tissues. In the nervous system, control of cell survival and differentiation is achieved through the action of a distinct group of polypeptides collectively known as neurotrophic factors. Recent findings support the view that trophic factors also are involved in the response of the nervous system to acute injury. By contrast, nutrients are not traditionally viewed as potential trophic factors; however, there is increasing evidence that at least some influence neuronal differentiation. During development the brain is responsive to variations in nutrient supply, and this increased sensitivity or vulnerability of the brain to nutrient supply may reappear during neuronal repair, a period during which a rapid membrane resynthesis and reestablishment of synthetic pathways occur. To further evaluate the potential of specific nutrients to act as pharmacologic agents in the repair of injured neurons, the effects of retinoic acid, an active metabolite of vitamin A, and its role as a trophic factor are discussed. This literature review is intended to provide background information regarding the effect of retinoic acid on the cholinergic phenotype and the differentiation of these neurons and to explain how it may promote neuronal repair and survival following injury.
Collapse
Affiliation(s)
- M A Malik
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | | |
Collapse
|
38
|
van Adel BA, Arnold JM, Phipps J, Doering LC, Ball AK. Ciliary neurotrophic factor protects retinal ganglion cells from axotomy-induced apoptosis via modulation of retinal gliain vivo. ACTA ACUST UNITED AC 2005; 63:215-34. [PMID: 15719421 DOI: 10.1002/neu.20117] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Adenoviral-mediated transfer of ciliary neurotrophic factor (CNTF) to the retina rescued retinal ganglion cells (RGCs) from axotomy-induced apoptosis, presumably via activation of the high affinity CNTF receptor alpha (CNTFRalpha) expressed on RGCs. CNTF can also activate astrocytes, via its low affinity leukemia inhibitory receptor beta expressed on mature astrocytes, suggesting that CNTF may also protect injured neurons indirectly by modulating glia. Adenoviral-mediated overexpression of CNTF in normal and axotomized rat retinas was examined to determine if it could increase the expression of several glial markers previously demonstrated to have a neuroprotective function in the injured brain and retina. Using Western blotting, the expression of glial fibrillary acid protein (GFAP), glutamate/aspartate transporter-1 (GLAST-1), glutamine synthetase (GS), and connexin 43 (Cx43) was examined 7 days after intravitreal injections of Ad.CNTF or control Ad.LacZ. Compared to controls, intravitreal injection of Ad.CNTF led to significant changes in the expression of CNTFRalpha, pSTAT(3), GFAP, GLAST, GS, and Cx43 in normal and axotomized retinas. Taken together, these results suggest that the neuroprotective effects of CNTF may result from a shift of retinal glia cells to a more neuroprotective phenotype. Moreover, the modulation of astrocytes may buffer high concentrations of glutamate that have been shown to contribute to the death of RGCs after optic nerve transection.
Collapse
Affiliation(s)
- B A van Adel
- Department of Pathology and Molecular Medicine, Faculty of Health Sciences, HSC-1R1, McMaster University, Hamilton, Canada
| | | | | | | | | |
Collapse
|
39
|
Dorval-Coiffec I, Delcros JG, Hakovirta H, Toppari J, Jégou B, Piquet-Pellorce C. Identification of the leukemia inhibitory factor cell targets within the rat testis. Biol Reprod 2004; 72:602-11. [PMID: 15537862 DOI: 10.1095/biolreprod.104.034892] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Leukemia inhibitory factor (LIF), a pleiotropic cytokine, is expressed in the rat testis and produced predominantly by peritubular myoid cells. The aims of this study were to characterize the testicular cell targets of LIF and to identify the role of LIF in the testis. The LIF receptor (LIF-R)/gp190 transcript was detected by reverse transcription-polymerase chain reaction (RT-PCR) in the rat testis from Day 13.5 postcoitum until adulthood. Seven highly purified testicular cell populations, representative of the major testicular constituents, were studied at transcriptional and protein levels by, respectively, RT-PCR and flow cytometry with biotinylated-LIF. Spermatogonia and, to a lesser extent, the somatic cells, exhibited specific LIF-binding sites. These results were strengthened by in situ analysis, showing predominant LIF-R immunoreactivity in spermatogonia at all ages studied. In addition to the 190-kDa LIF-R, Western blot analysis revealed the presence of a 50- to 60-kDa C-terminal gp190 isoform. This truncated form, which is unable to bind LIF, was the only form expressed in meiotic germ cells, suggesting an original down-regulation process of LIF-R expression during spermatogenesis. Finally, we showed that LIF increased [3H]-thymidine incorporation in spermatogonia in microdissected, cultured seminiferous tubules. Taken together, our results strongly suggest that LIF has a role in the regulation of the spermatogonial cell compartment.
Collapse
Affiliation(s)
- Isabelle Dorval-Coiffec
- INSERM U.625, Groupe d'Etude de la Reproduction chez l'homme et les mammifères, Université de Rennes I, 35042 Rennes cedex, France
| | | | | | | | | | | |
Collapse
|
40
|
Lee N, Neitzel KL, Devlin BK, MacLennan AJ. STAT3 phosphorylation in injured axons before sensory and motor neuron nuclei: potential role for STAT3 as a retrograde signaling transcription factor. J Comp Neurol 2004; 474:535-45. [PMID: 15174071 DOI: 10.1002/cne.20140] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
STAT3 is a latent transcription factor that is activated by plasma membrane growth factor receptor complexes. Conditional gene disruption data indicate that it contributes to the survival of cranial motor neurons after peripheral nerve lesion. In agreement, levels of activated STAT3 (Tyr705-phosphorylated STAT3) have been shown to increase in the nuclei of adult cranial motor neurons during their regeneration after the same injury. The data presented here demonstrate that STAT3 is similarly but not identically affected in sciatic motor neurons after sciatic nerve injury. In addition, we find that sensory neuron nuclei also display an analogous increase in activated STAT3, thereby supporting a role for STAT3 in the survival and regeneration of these cells. Most interesting, the present data indicate that peripheral nerve lesion leads to a very rapid activation of STAT3 in axons at the lesion site. This response increases during the first 24 hours after injury and extends back to the motor and sensory neurons such that phospho-STAT3-immunoreactive axons are first detected in the dorsal root ganglia and ventral spinal cord at the same postlesion time intervals at which the activated STAT3 is first detected in the neuronal nuclei. Together these data raise the possibility that axonal STAT3, activated at the injury site, acts as a retrograde signaling transcription factor, which promotes the survival and regeneration of both sensory and motor neurons.
Collapse
Affiliation(s)
- Nancy Lee
- Department of Molecular and Cellular Physiology, University of Cincinnati, Cincinnati, Ohio 45267-0576, USA
| | | | | | | |
Collapse
|
41
|
Abstract
Experimental models such as the facial nerve axotomy paradigm in rodents allow the systematic and detailed study of the response of neurones and their microenvironment to various types of challenges. Well-studied experimental examples include peripheral nerve trauma, the retrograde axonal transport of neurotoxins and locally enhanced inflammation following the induction of experimental autoimmune encephalomyelitis in combination with axotomy. These studies have led to novel insights into the regeneration programme of the motoneurone, the role of microglia and astrocytes in synaptic plasticity and the biology of glial cells. Importantly, many of the findings obtained have proven to be valid in other functional systems and even across species barriers. In particular, microglial expression of major histocompatibility complex molecules has been found to occur in response to various types of neuronal damage and is now regarded as a characteristic component of "glial inflammation". It is found in the context of numerous neurodegenerative disorders including Parkinson's and Alzheimer's disease. The detachment of afferent axonal endings from the surface membrane of regenerating motoneurones and their subsequent displacement by microglia ("synaptic stripping") and long-lasting insulation by astrocytes have also been confirmed in humans. The medical implications of these findings are significant. Also, the facial nerve system of rats and mice has become the best studied and most widely used test system for the evaluation of neurotrophic factors.
Collapse
Affiliation(s)
- Linda B Moran
- Department of Neuropathology, Division of Neuroscience and Psychological Medicine, Faculty of Medicine, Imperial College London, Charing Cross Campus, Fulham Palace Road, London W6 8RF, UK
| | | |
Collapse
|
42
|
Ji JZ, Elyaman W, Yip HK, Lee VWH, Yick LW, Hugon J, So KF. CNTF promotes survival of retinal ganglion cells after induction of ocular hypertension in rats: the possible involvement of STAT3 pathway. Eur J Neurosci 2004; 19:265-72. [PMID: 14725620 DOI: 10.1111/j.0953-816x.2003.03107.x] [Citation(s) in RCA: 145] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We examined the neuroprotective effect of ciliary neurotrophic factor (CNTF) on retinal ganglion cells (RGCs) in a rat glaucoma model with increased intraocular pressure (IOP) and studied the CNTF-mediated activation of Janus kinase/signal transducer and activator of transcription (JAK-STAT) pathway. Elevated IOP was induced by laser photocoagulation of the episcleral and limbal veins. The survival of RGCs was studied using Fluoro-Gold labelled in ocular hypertensive eyes with or without CNTF intravitreal injection. Immunochemical staining and immunoblot analysis for CNTF and phosphorylated STAT3 (pSTAT3) were performed. There was a significant and progressive loss of RGCs in the retinas following the induction of elevated IOP. A single intravitreal injection of 2 microg in 2 microL CNTF significantly protected RGCs up to 4 weeks. pSTAT3 was only transiently expressed in ocular hypertensive eyes. However, in eyes treated with CNTF, pSTAT3 was observed up to 2 weeks after the induction of elevated IOP. In ocular hypertensive eyes, CNTF-positive cells were found in the inner nuclear layer (INL), and there was a transient increase in the pSTAT3 cells in the ganglion cell layer and INL. Immunoblots showed that STAT3 was transiently phosphorylated after IOP increase, but with an injection of CNTF, pSTAT3 protein was observed up to 2 weeks after hypertensive glaucoma induction. Laser-induced chronic ocular hypertension in rats resulted in the death of RGCs and a transient activation of STAT3 in the retina. Intravitreal injection of CNTF showed a significant protection of RGCs, and the JAK-STAT signalling could be one of the important pathways that underlie the mechanism of CNTF neuroprotection in this rat glaucoma model.
Collapse
Affiliation(s)
- Jian-Zhong Ji
- Department of Anatomy, The University of Hong Kong, Hong Kong, China
| | | | | | | | | | | | | |
Collapse
|
43
|
Alberch J, Pérez-Navarro E, Canals JM. Neurotrophic factors in Huntington's disease. PROGRESS IN BRAIN RESEARCH 2004; 146:195-229. [PMID: 14699966 DOI: 10.1016/s0079-6123(03)46014-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Huntington's disease is a neurodegenerative disorder characterized by the selective loss of striatal neurons and, to a lesser extent, cortical neurons. The neurodegenerative process is caused by the mutation of huntingtin gene. Recent studies have established a link between mutant huntingtin, excitotoxicity and neurotrophic factors. Neurotrophic factors prevent cell death in degenerative processes but they can also enhance growth and function of neurons that are affected in Huntington's disease. The endogenous regulation of the expression of neurotrophic factors and their receptors in the striatum and its connections can be important to protect striatal cells and maintains basal ganglia connectivity. The administration of exogenous neurotrophic factors, in animal models of Huntington's disease, has been used to characterize the trophic requirements of striatal and cortical neurons. Neurotrophins, glial cell line-derived neurotrophic factor family members and ciliary neurotrophic factor have shown a potent neuroprotective effects on different neuronal populations of the striatum. Furthermore, they are also useful to maintain the integrity of the corticostriatal pathway. Thus, these neurotrophic factors may be suitable for the development of a neuroprotective therapy for neurodegenerative disorders of the basal ganglia.
Collapse
Affiliation(s)
- Jordi Alberch
- Department of Cell Biology and Pathology, Medical School, IDIBAPS, University of Barcelona, Casanova 143, E-08036 Barcelona, Spain.
| | | | | |
Collapse
|
44
|
Bongioanni P, Reali C, Sogos V. Ciliary neurotrophic factor (CNTF) for amyotrophic lateral sclerosis/motor neuron disease. Cochrane Database Syst Rev 2004; 2004:CD004302. [PMID: 15266526 PMCID: PMC8078584 DOI: 10.1002/14651858.cd004302.pub2] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND Amyotrophic lateral sclerosis, also known as motor neuron disease, is a fatal neuromuscular disease characterized by progressive muscle weakness resulting in paralysis, which might be treated with ciliary neurotrophic factor. OBJECTIVES The objective of this review was to examine the efficacy of ciliary neutrophic factor in amyotrophic lateral sclerosis. SEARCH STRATEGY We searched the Cochrane Neuromuscular Disease Group trials register (searched June 2003) for randomized trials, MEDLINE (from January 1966 to October 2003) and EMBASE (from January 1980 to October 2003), checked the reference lists of papers identified and contacted the authors of studies identified to get additional unpublished results. SELECTION CRITERIA We considered the following selection criteria: Types of studies: randomized controlled clinical trials; TYPES OF PARTICIPANTS adults with a diagnosis of either probable or definite amyotrophic lateral sclerosis according to the El Escorial criteria; Types of interventions: treatment with ciliary neurotrophic factor for at least six months, in a placebo-controlled randomized format; Types of outcome measures Primary: survival; Secondary: muscle strength, respiratory function, changes in bulbar functions, changes in quality of life, proportion of patients with adverse side effects (such as cough, asthenia, nausea, anorexia, weight loss and increased salivation). DATA COLLECTION AND ANALYSIS We identified two randomized trials. The data were extracted and examined independently by the reviewers. Some missing data were obtained from investigators. MAIN RESULTS Two trials, with a total population of 1,300 amyotrophic lateral sclerosis patients treated with subcutaneous injections of recombinant human ciliary neurotrophic factor, were examined in this review. The methodological quality of these trials was considered adequate. No significant difference was observed between ciliary neurotrophic factor and placebo groups for survival, the primary outcome measure. The relative risk was 1.07 (95% CI 0.81 to 1.41). No significant differences between the groups were observed for most of the secondary outcomes. However, a significant increase of the incidence of several adverse events was noted in groups treated with higher doses of CNTF. REVIEWERS' CONCLUSIONS Ciliary neurotrophic factor treatment has no effect on amyotrophic lateral sclerosis progression. At high concentration, several side effects were observed. A combination of ciliary neurotrophic factor with other neurotrophic factors (as suggested by results on animal models), and more efficient delivery methods should be tested.
Collapse
Affiliation(s)
- P Bongioanni
- Department of Neuroscience, University of Pisa, Via Paradisa, 2, Pisa, Italy, 56100.
| | | | | |
Collapse
|
45
|
Sun W, Oppenheim RW. Response of motoneurons to neonatal sciatic nerve axotomy in Bax-knockout mice. Mol Cell Neurosci 2003; 24:875-86. [PMID: 14697655 DOI: 10.1016/s1044-7431(03)00219-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Neonatal motoneurons (MNs) die rapidly after axotomy, a response that is mediated by the pro-apoptotic gene Bax and is followed by a mitochondria-mediated apoptotic cascade. Although motoneurons in neonatal Bax-deficient mice fail to degenerate following axotomy, it has not been previously examined whether the rescued MNs can regenerate following injury. We report here that although spinal MNs in Bax-knockout (Bax-KO) mice survive indefinitely, they undergo severe atrophy by 14 days after axotomy. By 1 month following axotomy, MN regeneration was observed and cellular atrophy was partially reversed. Interestingly, we observed that all MNs, including those previously rescued from normal developmental cell death in the embryo by Bax deletion, exhibit a regenerative response to peripheral nerve injury. The regenerative response may be mediated by specific trophic factors because the expression of glial cell line-derived neurotrophic factor (GDNF) was greatly increased in the proximal stump of injured nerves and application of a GDNF-blocking antibody greatly reduced regeneration/regrowth of rescued MNs in Bax-KO mice. These results indicate that MNs rescued from developmental or injury-induced cell death by Bax deletion have the potential to regenerate or regrow in response to nerve-derived signals following neonatal axotomy.
Collapse
Affiliation(s)
- Woong Sun
- Department of Neurobiology and Anatomy and Neuroscience Program, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | | |
Collapse
|
46
|
Kinameri E, Matsuoka I. Autocrine action of BMP2 regulates expression of GDNF-mRNA in sciatic Schwann cells. ACTA ACUST UNITED AC 2003; 117:221-7. [PMID: 14559157 DOI: 10.1016/s0169-328x(03)00326-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Schwann cell is a cell type that forms myelin sheath and provides trophic supports for neuronal cells by producing neurotrophic factors in both normal and traumatic situations. It was recently reported that after lesion of sciatic nerve, mRNA for glial cell line-derived neurotrophic factor (GDNF) is induced in nonneuronal cells in the nerve. However, the mechanism regulating GDNF-mRNA has remained largely unknown. In the present study, we searched for factors regulating the GDNF-mRNA expression in Schwann cells. First, we found that after transfer into explant culture as an in vitro lesion model, sciatic nerve segments began to express mRNA for bone morphogenetic protein-2 (BMP2) concomitantly with the induction of GDNF-mRNA. Treatment of the Schwann cells isolated from the sciatic nerve with combination of BMP2 and retinoic acid (RA) dramatically induced GDNF-mRNA, while BMP2 or RA alone had no effect. Furthermore, ionomycin, a calcium ionophore, which had even stronger activity on the induction of GDNF-mRNA also induced also BMP2-mRNA in cultured Schwann cells. Effects of inhibitors of intracellular signaling pathways such as protein kinase C inhibitor and MAPKK inhibitor suggested that the molecular mechanism of the induction of GDNF-mRNA is distinct from that of BMP2-mRNA. These results suggest that the Schwann cell-produced BMP2 plays an important role in the induction of GDNF after nerve injury in an autocrine fashion.
Collapse
Affiliation(s)
- Emi Kinameri
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-12-Nishi-6, Kita-Ku, Sapporo 060-0812, Japan
| | | |
Collapse
|
47
|
Rusnak M, House SB, Gainer H. Long-term effects of ciliary neurotrophic factor on the survival of vasopressin magnocellular neurones in the rat supraoptic nucleus in vitro. J Neuroendocrinol 2003; 15:933-9. [PMID: 12969237 DOI: 10.1046/j.1365-2826.2003.01080.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The use of hypothalamic organotypic cultures for the long-term study of mechanisms in magnocellular neurones (MCNs) of the hypothalamic-neurohypophysial system has been limited by the relatively poor maintenance of the vasopressin MCNs in vitro. Recent studies have shown that addition of ciliary neurotrophic factor (CNTF) to the media significantly reduced the apoptosis of both oxytocin and vasopressin MCNs. Here, we studied various temporal factors in the CNTF treatment that can influence the efficacy of MCN survival. Immunohistochemistry was used to identify and count surviving vasopressin and oxytocin MCNs in the supraoptic nucleus (SON) in hypothalamic slices cultured in the presence of CNTF (10 ng/ml media) for various time intervals, and in situ hybridization for vasopressin mRNA was used to evaluate the vasopressin mRNA gene expression in the SON under the same conditions. The presence of CNTF in the medium for 10 days produced a maximal increase in the survival of vasopressin MCNs (by 11-fold) and in the survival of oxytocin-MCNs (by approximately four-fold) over controls. These effects persisted for an additional 7-10 days even in the absence of CNTF. The ability of CNTF to increase survival of the MCNs or increase vasopressin mRNA levels in the SON required that the CNTF be present during the initial 7-10 days of culture. CNTF failed to rescue vasopressin or oxytocin MCNs when added to the media only for the last 7 days of a total of 14 days in vitro. Similar results were observed when SON vasopressin mRNA levels were measured. These results indicate that the presence of CNTF is required at the outset to rescue the vasopressin and oxytocin MCN from axotomy induced apoptosis, and that, after 10 days in CNTF, the MCNs no longer require the CNTF for survival.
Collapse
Affiliation(s)
- M Rusnak
- Laboratory of Neurochemistry, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
48
|
Kirsch M, Terheggen U, Hofmann HD. Ciliary neurotrophic factor is an early lesion-induced retrograde signal for axotomized facial motoneurons. Mol Cell Neurosci 2003; 24:130-8. [PMID: 14550774 DOI: 10.1016/s1044-7431(03)00130-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
To investigate the involvement of ciliary neurotropic factor (CNTF) in the postlesional response of motoneurons, we studied the activation of STAT3 signaling, the main signal transduction pathway of CNTF-like cytokines, in the facial nucleus of wildtype and CNTF-deficient mice following peripheral nerve transection. As shown by immunocytochemistry and immunoblot analysis, phosphorylation and nuclear translocation of STAT3 was maximally induced within 12 h postlesion in motoneurons of the ipsilateral facial nucleus of wildtype mice and is maintained for at least 3 days. In CNTF(-/-) mouse mutants, activation of STAT3 signaling was delayed by 10-12 h. Application of CNTF to the transected nerve restored rapid STAT3 activation in CNTF-deficient animals, whereas application of colchicine suppressed STAT3 signaling in wildtype mice for at least 24 h. These results identify CNTF as an early retrograde signal in axotomized facial motoneurons by showing that CNTF released at the lesion site is responsible for the initial induction of STAT3 signaling. Other cytokines like leukemia inhibitory factor obviously become active at later time points.
Collapse
Affiliation(s)
- Matthias Kirsch
- Institute of Anatomy, University of Freiburg, P.O. Box 111, D-79001 Freiburg, Germany
| | | | | |
Collapse
|
49
|
Gatzinsky KP, Holtmann B, Daraie B, Berthold CH, Sendtner M. Early onset of degenerative changes at nodes of Ranvier in alpha-motor axons of Cntf null (-/-) mutant mice. Glia 2003; 42:340-9. [PMID: 12730954 DOI: 10.1002/glia.10221] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The nodes of Ranvier are sites of specific interaction between Schwann cells and axons. Besides their crucial role in transmission of action potentials, the nodes of Ranvier and in particular the paranodal axon-Schwann cell networks (ASNs) are thought to function as local centers in large motor axons for removal, degradation, and disposal of organelles. In order to test whether ciliary neurotrophic factor (CNTF), which is present at high levels in the Schwann cell cytoplasm, is involved in the maintenance of these structures, we have examined lumbar ventral root nerve fibers of alpha-motor neurons by electron microscopy in 3- and 9-month-old Cntf null ((-/-)) mutant mice. Nerve fibers and nodes of Ranvier in 3-month-old Cntf(-/-) mutants appeared morphologically normal, except that ASNs were more voluminous in the mutants than in wild-type control animals at this age. In 9-month-old Cntf(-/-) animals, morphological changes, such as reduction in nerve fiber and axon diameter, myelin sheath disruption, and loss of ASNs at nodes of Ranvier, were observed. These findings suggest that endogenous CNTF, in addition to its role in promoting motor neuron survival and regeneration, is needed for long-term maintenance of alpha-motor nerve fibers. The premature loss of paranodal ASNs in animals lacking CNTF, which seems to be a defect related to a disturbed interaction in the nodal region between the axon and its myelinating Schwann cells, could impede the maintenance of a normal milieu in the motor axon, preceding more general neuronal damage.
Collapse
Affiliation(s)
- Kliment P Gatzinsky
- Department of Anatomy and Cell Biology, University of Göteborg, Göteborg, Sweden.
| | | | | | | | | |
Collapse
|
50
|
van Adel BA, Kostic C, Déglon N, Ball AK, Arsenijevic Y. Delivery of ciliary neurotrophic factor via lentiviral-mediated transfer protects axotomized retinal ganglion cells for an extended period of time. Hum Gene Ther 2003; 14:103-15. [PMID: 12614562 DOI: 10.1089/104303403321070801] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Ciliary neurotrophic factor (CNTF) has recently been demonstrated to be one of the most promising neurotrophic factors to improve both the survival and regeneration of injured retinal ganglion cells (RGCs). In the present study, we used optic nerve transection as an in vivo model to evaluate the effectiveness of a self-inactivating, replication-deficient lentiviral-mediated transfer of human ciliary neurotrophic factor (SIN-PGK-CNTF) on the survival of axotomized adult rat RGCs. Counts of dextran-fluorescein isothiocyanate conjugated (D-FITC)-retrogradely labeled RGCs revealed that the percentage of RGCs was drastically reduced (<90% cell death) 21 days after optic nerve transection. Retinal sections stained with X-gal revealed that intravitreal injection of the control LacZ-expressing lentiviral vector (LV-LacZ) resulted in the transduction of RGCs and retinal pigment epithelium (RPE) cells. A single intravitreal injection of LV-CNTF at the time of axotomy significantly enhanced RGC survival at 14 and 21 days postaxotomy compared to controls. These results demonstrate for the first time that rapid and prolonged delivery of CNTF using lentiviral-mediated gene transfer to the retina is an effective treatment for rescuing axotomized RGCs for an extended period of time. These results suggest that early and continuous administration of CNTF could serve as a potential treatment for retinal disorders involving optic neuropathy and RGC injury such as in glaucoma.
Collapse
Affiliation(s)
- Brian A van Adel
- Department of Pathology and Molecular Medicine, Faculty of Health Sciences, McMaster University, Hamilton, L8N 3ZS, Canada
| | | | | | | | | |
Collapse
|