1
|
Welch HF, Sankaranarayanan I, Hong VM, Mazhar K, Kolber BJ, Price TJ, Thorn CA. Lateralized nodose ganglia gene expression implicates cholecystokinin receptors in interoceptive reward signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.01.621506. [PMID: 39554115 PMCID: PMC11565973 DOI: 10.1101/2024.11.01.621506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
The vagus nerves are important carriers of sensory information from the viscera to the central nervous system. Emerging evidence suggests that sensory signaling through the right, but not the left, vagus nerve evokes striatal dopamine release and reinforces appetitive behaviors. However, the extent to which differential gene expression within vagal sensory neurons contributes to this asymmetric reward-related signaling remains unknown. Here, we use single-cell RNA sequencing to identify genes that are differentially expressed between the left and right nodose ganglia (NG) to identify candidate genes likely to contribute to vagus-mediated reward signaling. We find that a group of neurons expressing Chrna3 (nicotinic acetylcholine receptor subunit 3) and Cckar (cholecystokinin A receptor) is preferentially expressed in the right NG of both rats and mice. This result suggests that differential expression of gut-innervating nutrient sensors in NG neurons may contribute to asymmetric encoding of interoceptive rewards by the vagus nerves.
Collapse
|
2
|
Takeya H, Itai S, Kimura H, Kurashina Y, Amemiya T, Nagoshi N, Iwamoto T, Sato K, Shibata S, Matsumoto M, Onoe H, Nakamura M. Schwann cell-encapsulated chitosan-collagen hydrogel nerve conduit promotes peripheral nerve regeneration in rodent sciatic nerve defect models. Sci Rep 2023; 13:11932. [PMID: 37488180 PMCID: PMC10366170 DOI: 10.1038/s41598-023-39141-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 07/20/2023] [Indexed: 07/26/2023] Open
Abstract
Chitosan has various tissue regeneration effects. This study was designed to investigate the nerve regeneration effect of Schwann cell (SC)-encapsulated chitosan-collagen hydrogel nerve conduit (CCN) transplanted into a rat model of sciatic nerve defect. We prepared a CCN consisting of an outer layer of chitosan hydrogel and an inner layer of collagen hydrogel to encapsulate the intended cells. Rats with a 10-mm sciatic nerve defect were treated with SCs encapsulated in CCN (CCN+), CCN without SCs (CCN-), SC-encapsulated silicone tube (silicone+), and autologous nerve transplanting (auto). Behavioral and histological analyses indicated that motor functional recovery, axonal regrowth, and myelination of the CCN+ group were superior to those of the CCN- and silicone+ groups. Meanwhile, the CCN- and silicone+ groups showed no significant differences in the recovery of motor function and nerve histological restoration. In conclusion, SC-encapsulated CCN has a synergistic effect on peripheral nerve regeneration, especially axonal regrowth and remyelination of host SCs. In the early phase after transplantation, SC-encapsulated CCNs have a positive effect on recovery. Therefore, using SC-encapsulated CCNs may be a promising approach for massive peripheral nerve defects.
Collapse
Affiliation(s)
- Hiroaki Takeya
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Shun Itai
- Department of Mechanical Engineering, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-Ku, Yokohama-Shi, Kanagawa, 223-8522, Japan
- Division of Medical Science, Graduate School of Biomedical Engineering, Tohoku University, 1-1 Seiryomachi, Aoba-Ku, Sendai, Miyagi, 980-8574, Japan
| | - Hiroo Kimura
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi Shinjuku-Ku, Tokyo, 160-8582, Japan.
| | - Yuta Kurashina
- Division of Advanced Mechanical Systems Engineering, Institute of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei-Shi, Tokyo, 184-8588, Japan
| | - Tsuyoshi Amemiya
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Narihito Nagoshi
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Takuji Iwamoto
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Kazuki Sato
- Institute for Integrated Sports Medicine, Keio University School of Medicine, 35 Shinanomachi Shinjuku-Ku, Tokyo, Japan
| | - Shinsuke Shibata
- Division of Microscopic Anatomy, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-Dori, Chuo-Ku, Niigata, 951-8510, Japan
| | - Morio Matsumoto
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Hiroaki Onoe
- Department of Mechanical Engineering, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-Ku, Yokohama-Shi, Kanagawa, 223-8522, Japan
| | - Masaya Nakamura
- Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi Shinjuku-Ku, Tokyo, 160-8582, Japan
| |
Collapse
|
3
|
Sanchez D, Ganfornina MD. The Lipocalin Apolipoprotein D Functional Portrait: A Systematic Review. Front Physiol 2021; 12:738991. [PMID: 34690812 PMCID: PMC8530192 DOI: 10.3389/fphys.2021.738991] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 08/30/2021] [Indexed: 12/18/2022] Open
Abstract
Apolipoprotein D is a chordate gene early originated in the Lipocalin protein family. Among other features, regulation of its expression in a wide variety of disease conditions in humans, as apparently unrelated as neurodegeneration or breast cancer, have called for attention on this gene. Also, its presence in different tissues, from blood to brain, and different subcellular locations, from HDL lipoparticles to the interior of lysosomes or the surface of extracellular vesicles, poses an interesting challenge in deciphering its physiological function: Is ApoD a moonlighting protein, serving different roles in different cellular compartments, tissues, or organisms? Or does it have a unique biochemical mechanism of action that accounts for such apparently diverse roles in different physiological situations? To answer these questions, we have performed a systematic review of all primary publications where ApoD properties have been investigated in chordates. We conclude that ApoD ligand binding in the Lipocalin pocket, combined with an antioxidant activity performed at the rim of the pocket are properties sufficient to explain ApoD association with different lipid-based structures, where its physiological function is better described as lipid-management than by long-range lipid-transport. Controlling the redox state of these lipid structures in particular subcellular locations or extracellular structures, ApoD is able to modulate an enormous array of apparently diverse processes in the organism, both in health and disease. The new picture emerging from these data should help to put the physiological role of ApoD in new contexts and to inspire well-focused future research.
Collapse
Affiliation(s)
- Diego Sanchez
- Instituto de Biologia y Genetica Molecular, Unidad de Excelencia, Universidad de Valladolid-Consejo Superior de Investigaciones Cientificas, Valladolid, Spain
| | - Maria D Ganfornina
- Instituto de Biologia y Genetica Molecular, Unidad de Excelencia, Universidad de Valladolid-Consejo Superior de Investigaciones Cientificas, Valladolid, Spain
| |
Collapse
|
4
|
Inactivation of vimentin in satellite glial cells affects dorsal root ganglion intermediate filament expression and neuronal axon growth in vitro. Mol Cell Neurosci 2021; 115:103659. [PMID: 34400333 DOI: 10.1016/j.mcn.2021.103659] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 08/05/2021] [Accepted: 08/10/2021] [Indexed: 11/20/2022] Open
Abstract
Peripheral nerve trauma and regeneration are complex events, and little is known concerning how occurrences in the distal stump affect the cell body's response to injury. Intermediate filament (IF) proteins underpin cellular architecture and take part in nerve cell proliferation, differentiation and axon regeneration, but their role in these processes is not yet fully understood. The present study aimed to investigate the regulation and interrelationship of major neural IFs in adult dorsal root ganglion (DRG) neurons and satellite glial cells (SGCs) following sciatic nerve injury. We demonstrated that the expression of neural IFs in DRG neurons and SGCs after axotomy depends on vimentin activity. In intact DRGs, synemin M and peripherin proteins are detected in small neurons while neurofilament L (NFL) and synemin L characterize large neurons. Both neuronal populations are surrounded by vimentin positive- and glial fibrillary acidic protein (GFAP)-negative SGCs. In response to axotomy, synemin M and peripherin were upregulated in large wild-type DRG neurons and, to a lesser extent, in vim-/- and synm-/- DRG neurons, suggesting the role for these IFs in axon regeneration. However, an increase in the number of NFL-positive small neurons was observed in vim-/- mice, accompanied by a decrease of peripherin-positive small neurons. These findings suggest that vimentin is required for injury-induced neuronal IF remodeling. We further show that vimentin is also indispensable for nerve injury-induced GFAP upregulation in perineuronal SGCs and that inactivation of vimentin and synemin appears to accelerate the rate of DRG neurite regeneration at early stages in vitro.
Collapse
|
5
|
Bott CJ, Winckler B. Intermediate filaments in developing neurons: Beyond structure. Cytoskeleton (Hoboken) 2020; 77:110-128. [PMID: 31970897 DOI: 10.1002/cm.21597] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/06/2020] [Accepted: 01/08/2020] [Indexed: 12/20/2022]
Abstract
Neuronal development relies on a highly choreographed progression of dynamic cellular processes by which newborn neurons migrate, extend axons and dendrites, innervate their targets, and make functional synapses. Many of these dynamic processes require coordinated changes in morphology, powered by the cell's cytoskeleton. Intermediate filaments (IFs) are the third major cytoskeletal elements in vertebrate cells, but are rarely considered when it comes to understanding axon and dendrite growth, pathfinding and synapse formation. In this review, we first introduce the many new and exciting concepts of IF function, discovered mostly in non-neuronal cells. These roles include dynamic rearrangements, crosstalk with microtubules and actin filaments, mechano-sensing and -transduction, and regulation of signaling cascades. We then discuss the understudied roles of neuronally expressed IFs, with a particular focus on IFs expressed during development, such as nestin, vimentin and α-internexin. Lastly, we illustrate how signaling modulation by the unconventional IF nestin shapes neuronal morphogenesis in unexpected and novel ways. Even though the first IF knockout mice were made over 20 years ago, the study of the cell biological functions of IFs in the brain still has much room for exciting new discoveries.
Collapse
Affiliation(s)
- Christopher J Bott
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia
| | - Bettina Winckler
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
6
|
Hsu CG, Talukder MAH, Yue L, Turpin LC, Noble M, Elfar JC. Human equivalent dose of oral 4-aminopyridine differentiates nerve crush injury from transection injury and improves post-injury function in mice. Neural Regen Res 2020; 15:2098-2107. [PMID: 32394968 PMCID: PMC7716044 DOI: 10.4103/1673-5374.280319] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
4-Aminopyridine (4-AP), an FDA-approved drug for the symptomatic treatment of multiple sclerosis, is used to improve neuromuscular function in patients with diverse demyelinating disorders. We recently demonstrated that local, transdermal or injectable forms of 4-AP improve myelination, nerve conduction velocity, muscle atrophy, and motor function after traumatic peripheral nerve injury in mice. While oral 4-AP is most commonly used in the clinic, it is unknown whether human equivalent oral doses of 4-AP have effects on traumatic peripheral nerve injury differentiation, myelination, muscle atrophy, functional recovery, and post-injury inflammatory processes in animals. Mice with sciatic nerve crush or denervation injury received oral or intraperitoneal 4-AP (10 μg) or vehicle alone and were examined for pharmacokinetics, motor function, muscle mass, intrinsic muscle force, nerve morphological and gene expression profiles. 4-AP showed linear pharmacokinetics and the maximum plasma 4-AP concentrations were proportional to 4-AP dose. Acute single dose of oral 4-AP administration induced a rapid transient improvement in motor function that was different in traumatic peripheral nerve injury with or without nerve continuity, chronic daily oral 4-AP treatment significantly enhanced post crush injury motor function recovery and this effect was associated with improved myelination, muscle mass, and ex vivo muscle force. Polymerase chain reaction array analysis with crushed nerve revealed significant alterations in gene involved in axonal inflammation and regeneration. These findings provide convincing evidence that regardless of the route of administration, 4-AP can acutely differentiate traumatic peripheral nerve injury with or without nerve continuity and can enhance in vivo functional recovery with better preservation of myelin sheaths, muscle mass, and muscle force. The animal experiments were approved by the University Committee on Animal Research (UCAR) at the University of Rochester (UCAR-2009-019) on March 31, 2017.
Collapse
Affiliation(s)
- Chia George Hsu
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - M A Hassan Talukder
- Center for Orthopaedic Research and Translational Science, Penn State Hershey College of Medicine, Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Li Yue
- Department of Orthopedics, The Warren Alpert Medical School of Brown University/Rhode Island Hospital, Providence, RI, USA
| | - Loel C Turpin
- Department of Neuroscience, The University of Rochester Medical Center, Rochester, NY, USA
| | - Mark Noble
- Department of Biomedical Genetics, The University of Rochester Medical Center, Rochester, NY, USA
| | - John C Elfar
- Center for Orthopaedic Research and Translational Science, Penn State Hershey College of Medicine, Milton S. Hershey Medical Center, Hershey, PA, USA
| |
Collapse
|
7
|
Kumar A, Gupta S, Sharma P, Prasad R, Pal A. In silico method for identification of novel copper and iron metabolism proteins in various neurodegenerative disorders. Neurotoxicology 2019; 73:50-57. [DOI: 10.1016/j.neuro.2019.02.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/19/2019] [Accepted: 02/27/2019] [Indexed: 12/15/2022]
|
8
|
Nardo G, Trolese MC, Verderio M, Mariani A, de Paola M, Riva N, Dina G, Panini N, Erba E, Quattrini A, Bendotti C. Counteracting roles of MHCI and CD8 + T cells in the peripheral and central nervous system of ALS SOD1 G93A mice. Mol Neurodegener 2018; 13:42. [PMID: 30092791 PMCID: PMC6085701 DOI: 10.1186/s13024-018-0271-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 07/02/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The major histocompatibility complex I (MHCI) is a key molecule for the interaction of mononucleated cells with CD8+T lymphocytes. We previously showed that MHCI is upregulated in the spinal cord microglia and motor axons of transgenic SOD1G93A mice. METHODS To assess the role of MHCI in the disease, we examined transgenic SOD1G93A mice crossbred with β2 microglobulin-deficient mice, which express little if any MHCI on the cell surface and are defective for CD8+ T cells. RESULTS The lack of MHCI and CD8+ T cells in the sciatic nerve affects the motor axon stability, anticipating the muscle atrophy and the disease onset. In contrast, MHCI depletion in resident microglia and the lack of CD8+ T cell infiltration in the spinal cord protect the cervical motor neurons delaying the paralysis of forelimbs and prolonging the survival of SOD1G93A mice. CONCLUSIONS We provided straightforward evidence for a dual role of MHCI in the peripheral nervous system (PNS) compared to the CNS, pointing out regional and temporal differences in the clinical responses of ALS mice. These findings offer a possible explanation for the failure of systemic immunomodulatory treatments and suggest new potential strategies to prevent the progression of ALS.
Collapse
Affiliation(s)
- Giovanni Nardo
- Laboratory of Molecular Neurobiology, Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa 19, 20156, Milan, Italy.
| | - Maria Chiara Trolese
- Laboratory of Molecular Neurobiology, Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa 19, 20156, Milan, Italy
| | - Mattia Verderio
- Laboratory of Molecular Neurobiology, Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa 19, 20156, Milan, Italy
| | - Alessandro Mariani
- Laboratory of Analytical Biochemistry, Department of Environmental Health Sciences, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa 19, 20156, Milan, Italy
| | - Massimiliano de Paola
- Laboratory of Analytical Biochemistry, Department of Environmental Health Sciences, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa 19, 20156, Milan, Italy
| | - Nilo Riva
- Neuropathology Unit, Department of Neurology, INSPE- San Raffaele Scientific Institute, Dibit II, Via Olgettina 48, 20132, Milan, Italy
| | - Giorgia Dina
- Neuropathology Unit, Department of Neurology, INSPE- San Raffaele Scientific Institute, Dibit II, Via Olgettina 48, 20132, Milan, Italy
| | - Nicolò Panini
- Laboratory of Cancer Pharmacology Department of Oncology, Flow Cytometry Unit, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, via La Masa 19, 20156, Milan, Italy
| | - Eugenio Erba
- Laboratory of Cancer Pharmacology Department of Oncology, Flow Cytometry Unit, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, via La Masa 19, 20156, Milan, Italy
| | - Angelo Quattrini
- Neuropathology Unit, Department of Neurology, INSPE- San Raffaele Scientific Institute, Dibit II, Via Olgettina 48, 20132, Milan, Italy
| | - Caterina Bendotti
- Laboratory of Molecular Neurobiology, Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa 19, 20156, Milan, Italy
| |
Collapse
|
9
|
Nabeka H, Saito S, Li X, Shimokawa T, Khan MSI, Yamamiya K, Kawabe S, Doihara T, Hamada F, Kobayashi N, Matsuda S. Interneurons secrete prosaposin, a neurotrophic factor, to attenuate kainic acid-induced neurotoxicity. IBRO Rep 2017; 3:17-32. [PMID: 30135939 PMCID: PMC6084830 DOI: 10.1016/j.ibror.2017.07.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 07/16/2017] [Accepted: 07/21/2017] [Indexed: 12/30/2022] Open
Abstract
PS increased mainly in the axons of PV positive interneurons after kainic acid (KA) injection. Electron microscopy revealed PS containing vesicles in PV positive axons. PS is secreted with secretogranin from synapses. The increased PS in the interneurons was due to increases in PS + 0, as in the choroid plexus. Interneurons produce and secrete intact PS around the hippocampal pyramidal neurons to protect them from KA neurotoxicity.
Prosaposin (PS) is a secretory neurotrophic factor, as well as a regulator of lysosomal enzymes. We previously reported the up-regulation of PS and the possibility of its axonal transport by GABAergic interneurons after exocitotoxicity induced by kainic acid (KA), a glutamate analog. In the present study, we performed double immunostaining with PS and three calcium binding protein markers: parvalbumin (PV), calbindin, and calretinin, for the subpopulation of GABAergic interneurons, and clarified that the increased PS around the hippocampal pyramidal neurons after KA injection existed mainly in the axons of PV positive interneurons. Electron microscopy revealed PS containing vesicles in the PV positive axon. Double immunostaining with PS and secretogranin or synapsin suggested that PS is secreted with secretogranin from synapses. Based on the results from in situ hybridization with two alternative splicing forms of PS mRNA, the increase of PS in the interneurons was due to the increase of PS + 0 (mRNA without 9-base insertion) as in the choroid plexus, but not PS + 9 (mRNA with 9-base insertion). These results were similar to those from the choroid plexus, which secretes an intact form PS + 0 to the cerebrospinal fluid. Neurons, especially PV positive GABAergic interneurons, produce and secrete the intact form of PS around hippocampal pyramidal neurons to protect them against KA neurotoxicity.
Collapse
Affiliation(s)
- Hiroaki Nabeka
- Department of Anatomy and Embryology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Shouichiro Saito
- Laboratory of Veterinary Anatomy, Faculty of Applied Biological Sciences, Gifu University, Yanagido, Gifu, Japan
| | - Xuan Li
- Department of Anatomy and Embryology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Tetsuya Shimokawa
- Department of Anatomy and Embryology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Md Sakirul Islam Khan
- Department of Anatomy and Embryology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Kimiko Yamamiya
- Department of Anatomy and Embryology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | | | - Takuya Doihara
- Department of Anatomy and Embryology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Fumihiko Hamada
- Department of Human Anatomy, Oita University Fuculty of Medicine, Yufu, Oita, Japan
| | - Naoto Kobayashi
- Medical Education Center, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Seiji Matsuda
- Department of Anatomy and Embryology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| |
Collapse
|
10
|
Lim TKY, Anderson KM, Hari P, Di Falco M, Reihsen TE, Wilcox GL, Belani KG, LaBoissiere S, Pinto MR, Beebe DS, Kehl LJ, Stone LS. Evidence for a Role of Nerve Injury in Painful Intervertebral Disc Degeneration: A Cross-Sectional Proteomic Analysis of Human Cerebrospinal Fluid. THE JOURNAL OF PAIN 2017; 18:1253-1269. [PMID: 28652204 DOI: 10.1016/j.jpain.2017.06.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 06/08/2017] [Accepted: 06/08/2017] [Indexed: 12/18/2022]
Abstract
Intervertebral disc degeneration (DD) is a cause of low back pain (LBP) in some individuals. However, although >30% of adults have DD, LBP only develops in a subset of individuals. To gain insight into the mechanisms underlying nonpainful versus painful DD, human cerebrospinal fluid (CSF) was examined using differential expression shotgun proteomic techniques comparing healthy control participants, subjects with nonpainful DD, and patients with painful DD scheduled for spinal fusion surgery. Eighty-eight proteins were detected, 27 of which were differentially expressed. Proteins associated with DD tended to be related to inflammation (eg, cystatin C) regardless of pain status. In contrast, most differentially expressed proteins in DD-associated chronic LBP patients were linked to nerve injury (eg, hemopexin). Cystatin C and hemopexin were selected for further examination using enzyme-linked immunosorbent assay in a larger cohort. While cystatin C correlated with DD severity but not pain or disability, hemopexin correlated with pain intensity, physical disability, and DD severity. This study shows that CSF can be used to study mechanisms underlying painful DD in humans, and suggests that while painful DD is associated with nerve injury, inflammation itself is not sufficient to develop LBP. PERSPECTIVE CSF was examined for differential protein expression in healthy control participants, pain-free adults with asymptomatic intervertebral DD, and LBP patients with painful intervertebral DD. While DD was related to inflammation regardless of pain status, painful degeneration was associated with markers linked to nerve injury.
Collapse
Affiliation(s)
- Tony K Y Lim
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada; Alan Edwards Centre for Research on Pain, McGill University, Montreal, Quebec, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada; Department of Neurology and Neurosurgery, Montreal, McGill University, Quebec, Canada
| | - Kathleen M Anderson
- Program in Physical Therapy, Department of Physical Medicine and Rehabilitation, University of Minnesota, Minneapolis, Minnesota
| | - Pawan Hari
- Department of Epidemiology, University of Minnesota, Minneapolis, Minnesota
| | - Marcos Di Falco
- Genome Quebec, McGill University Innovation Centre, Montreal, Quebec, Canada
| | - Troy E Reihsen
- Department of Anesthesiology, University of Minnesota, Minneapolis, Minnesota
| | - George L Wilcox
- Department of Anesthesiology, University of Minnesota, Minneapolis, Minnesota; Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota
| | - Kumar G Belani
- Department of Anesthesiology, University of Minnesota, Minneapolis, Minnesota
| | - Sylvie LaBoissiere
- Genome Quebec, McGill University Innovation Centre, Montreal, Quebec, Canada
| | | | - David S Beebe
- Department of Anesthesiology, University of Minnesota, Minneapolis, Minnesota
| | - Lois J Kehl
- Department of Anesthesiology, University of Minnesota, Minneapolis, Minnesota; Minnesota Head & Neck Pain Clinic, St. Paul, Minnesota
| | - Laura S Stone
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada; Alan Edwards Centre for Research on Pain, McGill University, Montreal, Quebec, Canada; Faculty of Dentistry, McGill University, Montreal, Quebec, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
11
|
Neurotoxic Doses of Chronic Methamphetamine Trigger Retrotransposition of the Identifier Element in Rat Dorsal Dentate Gyrus. Genes (Basel) 2017; 8:genes8030096. [PMID: 28272323 PMCID: PMC5368700 DOI: 10.3390/genes8030096] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 02/27/2017] [Indexed: 12/16/2022] Open
Abstract
Short interspersed elements (SINEs) are typically silenced by DNA hypermethylation in somatic cells, but can retrotranspose in proliferating cells during adult neurogenesis. Hypomethylation caused by disease pathology or genotoxic stress leads to genomic instability of SINEs. The goal of the present investigation was to determine whether neurotoxic doses of binge or chronic methamphetamine (METH) trigger retrotransposition of the identifier (ID) element, a member of the rat SINE family, in the dentate gyrus genomic DNA. Adult male Sprague-Dawley rats were treated with saline or high doses of binge or chronic METH and sacrificed at three different time points thereafter. DNA methylation analysis, immunohistochemistry and next-generation sequencing (NGS) were performed on the dorsal dentate gyrus samples. Binge METH triggered hypomethylation, while chronic METH triggered hypermethylation of the CpG-2 site. Both METH regimens were associated with increased intensities in poly(A)-binding protein 1 (PABP1, a SINE regulatory protein)-like immunohistochemical staining in the dentate gyrus. The amplification of several ID element sequences was significantly higher in the chronic METH group than in the control group a week after METH, and they mapped to genes coding for proteins regulating cell growth and proliferation, transcription, protein function as well as for a variety of transporters. The results suggest that chronic METH induces ID element retrotransposition in the dorsal dentate gyrus and may affect hippocampal neurogenesis.
Collapse
|
12
|
Abstract
Injury of peripheral nerve in mammals leads to a complex but stereotypical pattern of histological events that comprise a highly reproducible sequence of degenerative reactions (Wallerian degeneration) succeeded by regenerative responses. These reactions are based on a corresponding sequence of cellular and mo lecular interactions that, in turn, reflect the differential expression of specific genes with functions in nerve degeneration and repair. We report on more than 60 genes and their products that show a specific pattern of regulation following peripheral nerve lesion. The group of regulated genes encoding, e.g., transcription factors, growth factors and their receptors, cytokines, neuropeptides, myelin proteins and lipid carriers, and cytoskeletal proteins as well as extracellular matrix and cell adhesion molecules. We describe and compare the distinct time-courses and cellular origin of expression and further discuss established or putative mo lecular interrelationships and functions with respect to the contribution of these genes/gene products to the molecular regeneration program of the PNS. NEUROSCIENTIST 3:112-122, 1997
Collapse
Affiliation(s)
- Clemens Gillen
- Molecular Neurobiology Laboratory, Department of Neurology, University of Düsseldorf Düsseldorf
| | - Christian Korfhage
- Molecular Neurobiology Laboratory, Department of Neurology, University of Düsseldorf Düsseldorf
| | - Hans Werner Müller
- Molecular Neurobiology Laboratory, Department of Neurology, University of Düsseldorf Düsseldorf
| |
Collapse
|
13
|
Trevisan G, Benemei S, Materazzi S, De Logu F, De Siena G, Fusi C, Fortes Rossato M, Coppi E, Marone IM, Ferreira J, Geppetti P, Nassini R. TRPA1 mediates trigeminal neuropathic pain in mice downstream of monocytes/macrophages and oxidative stress. Brain 2016; 139:1361-77. [PMID: 26984186 DOI: 10.1093/brain/aww038] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 01/24/2016] [Indexed: 02/04/2023] Open
Abstract
Despite intense investigation, the mechanisms of the different forms of trigeminal neuropathic pain remain substantially unidentified. The transient receptor potential ankyrin 1 channel (encoded by TRPA1) has been reported to contribute to allodynia or hyperalgesia in some neuropathic pain models, including those produced by sciatic nerve constriction. However, the role of TRPA1 and the processes that cause trigeminal pain-like behaviours from nerve insult are poorly understood. The role of TRPA1, monocytes and macrophages, and oxidative stress in pain-like behaviour evoked by the constriction of the infraorbital nerve in mice were explored. C57BL/6 and wild-type (Trpa1(+/+)) mice that underwent constriction of the infraorbital nerve exhibited prolonged (20 days) non-evoked nociceptive behaviour and mechanical, cold and chemical hypersensitivity in comparison to sham-operated mice (P < 0.05-P < 0.001). Both genetic deletion of Trpa1 (Trpa1(-/-)) and pharmacological blockade (HC-030031 and A-967079) abrogated pain-like behaviours (both P < 0.001), which were abated by the antioxidant, α-lipoic acid, and the nicotinamide adenine dinucleotide phosphate oxidase inhibitor, apocynin (both P < 0.001). Nociception and hypersensitivity evoked by constriction of the infraorbital nerve was associated with intra- and perineural monocytic and macrophagic invasion and increased levels of oxidative stress by-products (hydrogen peroxide and 4-hydroxynonenal). Attenuation of monocyte/macrophage increase by systemic treatment with an antibody against the monocyte chemoattractant chemokine (C-C motif) ligand 2 (CCL2) or the macrophage-depleting agent, clodronate (both P < 0.05), was associated with reduced hydrogen peroxide and 4-hydroxynonenal perineural levels and pain-like behaviours (all P < 0.01), which were abated by perineural administration of HC-030031, α-lipoic acid or the anti-CCL2 antibody (all P < 0.001). The present findings propose that, in the constriction of the infraorbital nerve model of trigeminal neuropathic pain, pain-like behaviours are entirely mediated by the TRPA1 channel, targeted by increased oxidative stress by-products released from monocytes and macrophages clumping at the site of nerve injury.
Collapse
Affiliation(s)
- Gabriela Trevisan
- Laboratory of Cellular and Molecular Biology, Graduate Program in Health Sciences, University of the Extreme South of Santa Catarina (UNESC), Criciúma 88806-000, SC, Brazil
| | - Silvia Benemei
- Department of Health Sciences, Clinical Pharmacology and Oncology Unit, University of Florence, Florence 50139, Italy
| | - Serena Materazzi
- Department of Health Sciences, Clinical Pharmacology and Oncology Unit, University of Florence, Florence 50139, Italy
| | - Francesco De Logu
- Department of Health Sciences, Clinical Pharmacology and Oncology Unit, University of Florence, Florence 50139, Italy
| | - Gaetano De Siena
- Department of Health Sciences, Clinical Pharmacology and Oncology Unit, University of Florence, Florence 50139, Italy
| | - Camilla Fusi
- Department of Health Sciences, Clinical Pharmacology and Oncology Unit, University of Florence, Florence 50139, Italy
| | - Mateus Fortes Rossato
- Department of Pharmacology, Federal University of Santa Catarina (UFSC), Florianópolis 88040-900, SC, Brazil
| | - Elisabetta Coppi
- Department of Health Sciences, Clinical Pharmacology and Oncology Unit, University of Florence, Florence 50139, Italy
| | - Ilaria Maddalena Marone
- Department of Health Sciences, Clinical Pharmacology and Oncology Unit, University of Florence, Florence 50139, Italy
| | - Juliano Ferreira
- Department of Health Sciences, Clinical Pharmacology and Oncology Unit, University of Florence, Florence 50139, Italy Department of Pharmacology, Federal University of Santa Catarina (UFSC), Florianópolis 88040-900, SC, Brazil
| | - Pierangelo Geppetti
- Department of Health Sciences, Clinical Pharmacology and Oncology Unit, University of Florence, Florence 50139, Italy
| | - Romina Nassini
- Department of Health Sciences, Clinical Pharmacology and Oncology Unit, University of Florence, Florence 50139, Italy
| |
Collapse
|
14
|
Sharma R, Lahiri R, Scollard DM, Pena M, Williams DL, Adams LB, Figarola J, Truman RW. The armadillo: a model for the neuropathy of leprosy and potentially other neurodegenerative diseases. Dis Model Mech 2012; 6:19-24. [PMID: 23223615 PMCID: PMC3529335 DOI: 10.1242/dmm.010215] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Leprosy (also known as Hansen’s disease) is an infectious peripheral neurological disorder caused by Mycobacterium leprae that even today leaves millions of individuals worldwide with life-long disabilities. The specific mechanisms by which this bacterium induces nerve injury remain largely unknown, mainly owing to ethical and practical limitations in obtaining affected human nerve samples. In addition to humans, nine-banded armadillos (Dasypus novemcinctus) are the only other natural host of M. leprae, and they develop a systemically disseminated disease with extensive neurological involvement. M. leprae is an obligate intracellular parasite that cannot be cultivated in vitro. Because of the heavy burdens of bacilli they harbor, nine-banded armadillos have become the organism of choice for propagating large quantities of M. leprae, and they are now advancing as models of leprosy pathogenesis and nerve damage. Although armadillos are exotic laboratory animals, the recently completed whole genome sequence for this animal is enabling researchers to undertake more sophisticated molecular studies and to develop armadillo-specific reagents. These advances will facilitate the use of armadillos in piloting new therapies and diagnostic regimens, and will provide new insights into the oldest known infectious neurodegenerative disorder.
Collapse
Affiliation(s)
- Rahul Sharma
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Coste O, Möser CV, Sisignano M, Kynast KL, Minden A, Geisslinger G, Niederberger E. The p21-activated kinase PAK 5 is involved in formalin-induced nociception through regulation of MAP-kinase signaling and formalin-specific receptors. Behav Brain Res 2012; 234:121-8. [PMID: 22732262 DOI: 10.1016/j.bbr.2012.06.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Revised: 06/11/2012] [Accepted: 06/15/2012] [Indexed: 10/28/2022]
Abstract
p21-activated kinases (PAKs) are involved in signal cascades relevant for nociceptive processing and neuropathic pain. Particularly, the recently described group B PAKs 4, 5 and 6 regulate MAP-kinases and the rearrangement of the actin cytoskeleton, both of which have been linked to pain processing. However, a specific role of these PAKs in nociception has not yet been demonstrated. We found PAK 4, 5 and 6 expression in pain-relevant tissues in peripheral and CNS. Since viable knock-out mice only exist for the PAK isoform 5, we further assessed the impact of this PAK on acute and chronic pain using different behavioral models in mice. PAK 5 knock-out mice showed normal acute nociception and did not differ from wild type mice in their neuropathic pain behavior. However, the nociceptive response in formalin-induced paw inflammation was significantly reduced in knock-out mice associated with inhibition of MAP-kinase activation and a decreased number of formalin-induced c-Fos positive neurons in the spinal cord. Furthermore, in isolated neurons, we found a significantly reduced calcium response after stimulation of TRPA1-channels in PAK 5(-/-)- compared to PAK 5(+/+)-cells. Our results indicate that PAK 5 is involved in formalin-induced inflammatory nociception through regulation of MAPK-induced c-Fos-activation and formalin-specific TRP-channels.
Collapse
Affiliation(s)
- Ovidiu Coste
- Pharmazentrum frankfurt/ZAFES, Klinikum der Johann Wolfgang Goethe-Universität, Frankfurt, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany.
| | | | | | | | | | | | | |
Collapse
|
16
|
Cavalcanti F, Kidd T, Patitucci A, Valentino P, Bono F, Nisticò R, Quattrone A. An axon regeneration signature in a Charcot-Marie-Tooth disease type 2 patient. J Neurogenet 2011; 23:324-8. [PMID: 19657941 DOI: 10.1080/01677060802447585] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Charcot-Marie-Tooth disease (CMT) is one of the most common inherited peripheral neuropathies. The underlying mutations in demyelinating forms tend to affect genes expressed in Schwann cells (CMT types 1, 3, and 4), while axonal forms of the disease usually have their origins in genes expressed in the affected neurons (CMT type 2). Repeated rounds of nerve degeneration and regeneration characterize CMT2, but evidence for regeneration has not been demonstrated at a molecular level. Subtractive hybridization was performed on sural nerve biopsies from a patient presenting an axonal form of CMT and an unaffected sibling, which revealed an overexpression of genes associated with the regeneration of axons, including PMP22, SPARC/osteonectin, CD9, CD44, EEF1A1, and gamma-actin. These results suggest that axonal degeneration elicits a regeneration transcriptional response in the surrounding Schwann cells. This response contrasts with other neurodegenerative diseases, in which programmed cell death or an inappropriate immune response are activated. Additionally, Lamin A/C, which is mutated in CMT2B1, was overexpressed in the patient, suggesting that CMT-causing genes may interact in a regulatory network.
Collapse
Affiliation(s)
- Francesca Cavalcanti
- Institute of Neurological Sciences, National Research Council, Mangone, Cosenza, Italy
| | | | | | | | | | | | | |
Collapse
|
17
|
Kruse F, Bosse F, Vogelaar CF, Brazda N, Küry P, Gasis M, Müller HW. Cortical gene expression in spinal cord injury and repair: insight into the functional complexity of the neural regeneration program. Front Mol Neurosci 2011; 4:26. [PMID: 21994489 PMCID: PMC3182759 DOI: 10.3389/fnmol.2011.00026] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 09/05/2011] [Indexed: 11/29/2022] Open
Abstract
Traumatic spinal cord injury (SCI) results in the formation of a fibrous scar acting as a growth barrier for regenerating axons at the lesion site. We have previously shown (Klapka et al., 2005) that transient suppression of the inhibitory lesion scar in rat spinal cord leads to long distance axon regeneration, retrograde rescue of axotomized cortical motoneurons, and improvement of locomotor function. Here we applied a systemic approach to investigate for the first time specific and dynamic alterations in the cortical gene expression profile following both thoracic SCI and regeneration-promoting anti-scarring treatment (AST). In order to monitor cortical gene expression we carried out microarray analyses using total RNA isolated from layer V/VI of rat sensorimotor cortex at 1–60 days post-operation (dpo). We demonstrate that cortical neurons respond to injury by massive changes in gene expression, starting as early as 1 dpo. AST, in turn, results in profound modifications of the lesion-induced expression profile. The treatment attenuates SCI-triggered transcriptional changes of genes related to inhibition of axon growth and impairment of cell survival, while upregulating the expression of genes associated with axon outgrowth, cell protection, and neural development. Thus, AST not only modifies the local environment impeding spinal cord regeneration by reduction of fibrous scarring in the injured spinal cord, but, in addition, strikingly changes the intrinsic capacity of cortical pyramidal neurons toward enhanced cell maintenance and axonal regeneration.
Collapse
Affiliation(s)
- Fabian Kruse
- Molecular Neurobiology Laboratory, Department of Neurology, Heinrich-Heine-University Düsseldorf, Germany
| | | | | | | | | | | | | |
Collapse
|
18
|
Fang M, He X, Zhang W, Zhou X, Nie Q, Zhang X. Transcript variants, expression, and polymorphisms of the pig prosaposin gene. DNA Cell Biol 2011; 30:481-9. [PMID: 21563967 DOI: 10.1089/dna.2011.1225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Prosaposin (PASP) is a sphingolipid hydrolysis protein that plays roles in both the nervous and reproduction systems. In this study, we cloned the pig PASP gene and studied its genomic organization, polymorphism, and expression pattern. Two PASP transcripts, TV1 (HQ245644) and TV2 (HQ245646), were identified in pig. TV1 was the complete transcript that encoded 527 amino acids, whereas TV2 was 9 bp shorter due to an exon 8 deletion. The pig PASP gene spanned over 34 kb in length on chromosome 14 (SSC14), and consisted of 15 exons and 14 introns. The pig PASP gene (TV1 and TV2) expressed predominantly in the cerebellum, lymphnode, pituitary, abdominal fat, hypothalamus, and cerebrum in both females and males. PASP TV1 expressed mainly in teh cerebrum, cerebellum, hypothalamus, pituitary, heart, subcutaneous fat, and foreleg muscle, while TV2 was expressed in the liver, spleen, lung, kidney, and lymphnode. In foreleg muscle, the predominant transcript was TV2 in males and TV1 in females. Some potential transcriptional elements were predicted in 5' flanking region (~3000 bp) of the PASP gene, and they were TATA boxes, RORE, Sp1, SRY, oct-1, Cdx A, and cap. Additionally, we identified 68 single-nucleotide polymorphisms and 9 indels in the pig PASP gene, and three single-nucleotide polymorphisms (C77932320T or L15F; C77928094T or P191L; A77917401G or K522R) were nonsynonymous substitutions. These results provide useful information for future functional investigations of the pig PASP gene.
Collapse
Affiliation(s)
- Meixia Fang
- Department of Laboratory Animal Science, Medical College of Jinan University, Guangzhou, China
| | | | | | | | | | | |
Collapse
|
19
|
Ma CHE, Bampton ETW, Evans MJ, Taylor JSH. Synergistic effects of osteonectin and brain-derived neurotrophic factor on axotomized retinal ganglion cells neurite outgrowth via the mitogen-activated protein kinase-extracellular signal-regulated kinase 1/2 pathways. Neuroscience 2010; 165:463-74. [PMID: 19837135 DOI: 10.1016/j.neuroscience.2009.10.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2009] [Revised: 10/06/2009] [Accepted: 10/13/2009] [Indexed: 11/19/2022]
Abstract
Our previous study identified osteonectin (ON) in a screen of factors made by Schwann cells (SCs) which promoted peripheral and central neurons survival and neuritogenesis, however, the mechanisms of ON promoting effects are largely unknown. In the present study, we investigated the effects of ON-deficient SC-conditioned medium (SCCM) and molecular mechanisms of ON, in regulating retinal ganglion cells (RGCs) survival and neurite outgrowth. Neonatal rat RGCs and SCs were purified by immunopanning technique. RGC survival and neuritogenesis reduced significantly when treated with either ON-null mice SCCM or ON-immunodepleted (IP) SCCM (P<0.05). In contrast to wild type SCCM, in the presence of a tyrosine kinase receptor (Trk) inhibitor (K252a), ON-null mice SCCM-induced neuritogenesis were further reduced by 24%. The Trk-mediated signaling pathways became more sensitive to K252a inhibition in the absence of ON. We also showed the synergistic effects of ON and brain-derived neurotrophic factor (BDNF) in promoting RGCs growth and the involvement of ON in two major neurotrophin-mediated signaling pathways, PI-3K-Akt and MAPK-Erk1/2. ON alone activated Akt phosphorylation and increased survival. Blockage of TrkB signalling pathway by TrkB-Fc chimera (BDNF scavenger) or K252a in ON-treated cultures reduced Akt-P level significantly. This suggests that ON induces BDNF synthesis and secretion from RGCs. The enhancement of neuritogenesis and Erk1/2 phosphorylation by ON in BDNF-treated cultures further demonstrate the signaling pathways responsible for the synergistic effect of ON on BDNF-induced neurite outgrowth. To the best of our knowledge, this is the first report showing the synergistic effects of ON on classical neurotrophins which participate in the same signalling pathways in regulating RGC neurite outgrowth.
Collapse
Affiliation(s)
- C H E Ma
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.
| | | | | | | |
Collapse
|
20
|
Amiri S, Movahedin M, Mowla SJ, Hajebrahimi Z, Tavallaei M. Differential gene expression and alternative splicing of survivin following mouse sciatic nerve injury. Spinal Cord 2009; 47:739-44. [DOI: 10.1038/sc.2009.26] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
21
|
Starkey ML, Davies M, Yip PK, Carter LM, Wong DJN, McMahon SB, Bradbury EJ. Expression of the regeneration-associated protein SPRR1A in primary sensory neurons and spinal cord of the adult mouse following peripheral and central injury. J Comp Neurol 2009; 513:51-68. [PMID: 19107756 DOI: 10.1002/cne.21944] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Small proline-rich repeat protein 1A (SPRR1A) is expressed in dorsal root ganglion (DRG) neurons following peripheral nerve injury but it is not known whether SPRR1A is differentially expressed following injury to peripheral versus central DRG projections and a detailed characterization of expression in sensory neuron subpopulations and spinal cord has not been performed. Here we use immunocytochemical techniques to characterize SPRR1A expression following sciatic nerve, dorsal root, and dorsal column injury in adult mice. SPRR1A was not detected in naïve spinal cord, DRG, or peripheral nerves and there was minimal expression following injury to the centrally projecting branches of DRG neurons. However, following peripheral (sciatic) nerve injury, intense SPRR1A immunoreactivity was observed in the dorsal horn and motoneurons of the spinal cord, in L4/5 DRG neurons, and in the injured nerve. A time-course study comparing expression following sciatic nerve crush and transection revealed maximum SPRR1A levels at day 7 in both models. However, while SPRR1A was downregulated to baseline by 30 days postlesion following crush injury, it remained elevated 30 days after transection. Cell-size and double-labeling studies revealed that SPRR1A was expressed by DRG cells of all sizes and colocalized with classical markers of DRG subpopulations and their primary afferent terminals. High coexpression of SPRR1A with activating transcription factor-3 and growth-associated protein-43 was observed, indicating that it is expressed by injured and regenerating neurons. This study supports the hypothesis that SPRR1A is a regeneration-associated gene and that SPRR1A provides a valuable marker to assess the regenerative potential of injured neurons.
Collapse
Affiliation(s)
- Michelle L Starkey
- Neurorestoration Group, Wolfson Centre for Age-Related Diseases, Wolfson Wing, King's College London, London Bridge, London.
| | | | | | | | | | | | | |
Collapse
|
22
|
Impaired prosaposin secretion during nerve regeneration in diabetic rats and protection of nerve regeneration by a prosaposin-derived peptide. J Neuropathol Exp Neurol 2008; 67:702-10. [PMID: 18596543 DOI: 10.1097/nen.0b013e31817e23f4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Prosaposin is both a precursor of sphingolipid activator proteins and a secreted neurotrophic and myelinotrophic factor. Because peripheral nerve regeneration is impaired in diabetes mellitus, we measured prosaposin protein levels from control and streptozotocin-diabetic rats by collecting endoneurial fluid secreted into a bridging tube connecting the ends of transected sciatic nerve. Prosaposin protein levels were significantly reduced in endoneurial fluid from diabetic rats and increased in the proximal nerve stump compared to controls. To investigate whether a prosaposin-derived peptide could improve nerve regeneration, rats were treated with prosaptide TX14(A) after sciatic nerve crush. In control rats, TX14(A) was without effect in the uninjured nerve but shortened toe spread recovery time after nerve crush. In diabetic rats, efficacy of prosaptide TX14(A) was confirmed by correction of thermal hypoalgesia, formalin-evoked hyperalgesia, and conduction slowing in the uninjured nerve. The peptide also prevented diabetes-induced abnormalities in nerve regeneration distance and mean axonal diameter of regenerated axons, whereas delayed recovery of toe spread was not improved. Muscle denervation atrophy was attenuated by TX14(A) in both control and diabetic rats. These results suggest that reduced prosaposin secretion after nerve injury may contribute to impaired regeneration rates in diabetic rats, and that prosaptide TX14(A) can improve aspects of nerve regeneration.
Collapse
|
23
|
The secretion and maturation of prosaposin and procathepsin D are blocked in embryonic neural progenitor cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2008; 1783:1480-9. [DOI: 10.1016/j.bbamcr.2008.01.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2007] [Accepted: 01/30/2008] [Indexed: 01/14/2023]
|
24
|
Roglio I, Bianchi R, Gotti S, Scurati S, Giatti S, Pesaresi M, Caruso D, Panzica G, Melcangi R. Neuroprotective effects of dihydroprogesterone and progesterone in an experimental model of nerve crush injury. Neuroscience 2008; 155:673-85. [DOI: 10.1016/j.neuroscience.2008.06.034] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2008] [Revised: 05/29/2008] [Accepted: 06/17/2008] [Indexed: 11/25/2022]
|
25
|
Ochiai T, Takenaka Y, Kuramoto Y, Kasuya M, Fukuda K, Kimura M, Shimeno H, Misasi R, Hiraiwa M, Soeda S. Molecular mechanism for neuro-protective effect of prosaposin against oxidative stress: its regulation of dimeric transcription factor formation. Biochim Biophys Acta Gen Subj 2008; 1780:1441-7. [PMID: 18706485 DOI: 10.1016/j.bbagen.2008.07.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2008] [Revised: 07/16/2008] [Accepted: 07/18/2008] [Indexed: 12/09/2022]
Abstract
Prosaposin triggers G-protein-coupled receptor (GPCR)-mediated protein kinase B (Akt)/extracellular signal-regulated kinase (ERK) phosphorylation cascades to exert its neurotrophic and myelinotrophic activity capable of preventing neural cell death and promoting neural proliferation and glial differentiation. In the present study, we investigated the down-stream neurotrophic signaling mechanism of prosaposin by which rat pheochromocytoma (PC-12) cells are protected from cell death induced by oxidative stress. When PC-12 cells were exposed to H2O2, the cells underwent abrupt shrinkage followed by apoptosis. Prosaposin treatment at as low as 1 nM protected PC-12 cells from cell death by the oxidative stress with the activation of an ERK phosphorylation cascade. Simultaneously, prosaposin blocked the oxidative stress induced-Akt phosphorylation that acts on the down-stream of caspase-3 activation. A MEK inhibitor, PD98059, or a phosphatidylinositol 3-kinase (PI3K) inhibitor, LY294002, abolished the survival effect of prosaposin on the oxidative stress-induced cell death. Furthermore, prosaposin blocked the oxidative stress-induced phosphorylations of c-Jun N-terminal kinase (JNK) and p38 stress-activated protein kinase. We further investigated the effect of prosaposin treatment on the phosphorylation of activating protein-1 (AP-1) complex components, c-Jun and activating transcription factor (ATF)-3. Western blot analysis demonstrated that prosaposin treatment at 100 ng/ml decreased the levels of c-Jun and ATF-3 induced by H2O2 stimulation. Our results suggest that prosaposin aids survival of PC-12 cells from oxidative stress not only by reducing the phosphorylation levels of JNK and p38, but also by regulating the c-Jun/AP-1 pathway.
Collapse
Affiliation(s)
- Takashi Ochiai
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Toth C, Shim SY, Wang J, Jiang Y, Neumayer G, Belzil C, Liu WQ, Martinez J, Zochodne D, Nguyen MD. Ndel1 promotes axon regeneration via intermediate filaments. PLoS One 2008; 3:e2014. [PMID: 18431495 PMCID: PMC2291557 DOI: 10.1371/journal.pone.0002014] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2007] [Accepted: 03/13/2008] [Indexed: 01/27/2023] Open
Abstract
Failure of axons to regenerate following acute or chronic neuronal injury is attributed to both the inhibitory glial environment and deficient intrinsic ability to re-grow. However, the underlying mechanisms of the latter remain unclear. In this study, we have investigated the role of the mammalian homologue of aspergillus nidulans NudE, Ndel1, emergently viewed as an integrator of the cytoskeleton, in axon regeneration. Ndel1 was synthesized de novo and upregulated in crushed and transected sciatic nerve axons, and, upon injury, was strongly associated with neuronal form of the intermediate filament (IF) Vimentin while dissociating from the mature neuronal IF (Neurofilament) light chain NF-L. Consistent with a role for Ndel1 in the conditioning lesion-induced neurite outgrowth of Dorsal Root Ganglion (DRG) neurons, the long lasting in vivo formation of the neuronal Ndel1/Vimentin complex was associated with robust axon regeneration. Furthermore, local silencing of Ndel1 in transected axons by siRNA severely reduced the extent of regeneration in vivo. Thus, Ndel1 promotes axonal regeneration; activating this endogenous repair mechanism may enhance neuroregeneration during acute and chronic axonal degeneration.
Collapse
Affiliation(s)
- Cory Toth
- Department of Clinical Neurosciences, University of Calgary, Hotchkiss Brain Institute, Calgary, Canada
| | - Su Yeon Shim
- Department of Clinical Neurosciences, University of Calgary, Hotchkiss Brain Institute, Calgary, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Hotchkiss Brain Institute, Calgary, Canada
- Department of Biochemistry and Molecular Biology, University of Calgary, Hotchkiss Brain Institute, Calgary, Canada
| | - Jian Wang
- Department of Clinical Neurosciences, University of Calgary, Hotchkiss Brain Institute, Calgary, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Hotchkiss Brain Institute, Calgary, Canada
- Department of Biochemistry and Molecular Biology, University of Calgary, Hotchkiss Brain Institute, Calgary, Canada
| | - Yulan Jiang
- Department of Biochemistry and Molecular Biology, University of Calgary, Hotchkiss Brain Institute, Calgary, Canada
| | - Gernot Neumayer
- Department of Clinical Neurosciences, University of Calgary, Hotchkiss Brain Institute, Calgary, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Hotchkiss Brain Institute, Calgary, Canada
- Department of Biochemistry and Molecular Biology, University of Calgary, Hotchkiss Brain Institute, Calgary, Canada
| | - Camille Belzil
- Department of Clinical Neurosciences, University of Calgary, Hotchkiss Brain Institute, Calgary, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Hotchkiss Brain Institute, Calgary, Canada
- Department of Biochemistry and Molecular Biology, University of Calgary, Hotchkiss Brain Institute, Calgary, Canada
| | - Wei-Qiao Liu
- Department of Clinical Neurosciences, University of Calgary, Hotchkiss Brain Institute, Calgary, Canada
| | - Jose Martinez
- Department of Clinical Neurosciences, University of Calgary, Hotchkiss Brain Institute, Calgary, Canada
| | - Douglas Zochodne
- Department of Clinical Neurosciences, University of Calgary, Hotchkiss Brain Institute, Calgary, Canada
| | - Minh Dang Nguyen
- Department of Clinical Neurosciences, University of Calgary, Hotchkiss Brain Institute, Calgary, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Hotchkiss Brain Institute, Calgary, Canada
- Department of Biochemistry and Molecular Biology, University of Calgary, Hotchkiss Brain Institute, Calgary, Canada
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
27
|
Expression patterns in alternative splicing forms of prosaposin mRNA in the rat facial nerve nucleus after facial nerve transection. Neurosci Res 2007; 60:82-94. [PMID: 18022721 DOI: 10.1016/j.neures.2007.09.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2006] [Revised: 08/20/2007] [Accepted: 09/26/2007] [Indexed: 11/24/2022]
Abstract
Prosaposin acts as a neurotrophic factor, in addition to its role as the precursor protein for saposins A, B, C, and D, which are activators for specific sphingolipid hydrolases in lysosomes. In rats, the prosaposin gene generates two alternative splicing forms of mRNA: Pro+9 containing a 9-base insertion and Pro+0 without. The expression of these mRNAs changes after brain injury. We examined the expression patterns of the alternative splicing forms of prosaposin mRNA in the rat facial nerve nucleus for 52 days following facial nerve transection. Pro+0 mRNA increased within 3 days of transection, peaked after 5-10 days, and remained significantly elevated for 21 days. In contrast, the expression of Pro+9 mRNA was constant throughout the regenerative period. Prosaposin mRNA expression increased not only in facial motoneurons, but also in microglia during facial nerve regeneration. Our findings indicate that the saposin B domain of prosaposin, which is the domain affected by alternative splicing, plays an important role in both neurons and microglia during neuroregeneration.
Collapse
|
28
|
Mühlfriedel S, Kirsch F, Gruss P, Chowdhury K, Stoykova A. Novel genes differentially expressed in cortical regions during late neurogenesis. Eur J Neurosci 2007; 26:33-50. [PMID: 17614941 DOI: 10.1111/j.1460-9568.2007.05639.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Differential gene expression across the embryonic cerebral cortex is assumed to play a role in the subdivision of the cortex into distinct areas with specific morphology, physiology and function. In a search for genes that may be involved in the cortical regionalization during late neurogenesis in mouse, we performed an extensive in-situ expression analysis at embryonic day (E)16 and E18. The examined candidate genes were selected beforehand by a microarray screen by virtue of their preferential expression in the anlagen of the motor, somatosensory, visual and cingulate cortices or hippocampus. We present new information about graded or regionally enriched expression of 25 genes (nine of which are novel genes) across the mouse embryonic cortex, in progenitor cells as well as in the cortical plate. The established differential expression of most of these genes is persistent at both stages studied, suggesting that their expression is regulated by an intrinsic programme. For some of the genes, the concept of intrinsic regulation is further substantiated by the high similarity of the reported expression patterns at E16 and E18 and published data from earlier stages. Few genes with robust expression in the E16 caudal cortex showed a more restricted pattern at E18, possibly because of their response to extrinsic cues. In addition, several genes appeared to be suitable novel markers for amygdalar and diencephalic nuclei. Taken together, our findings reveal novel molecular partitions of the late mouse cortex that are in accordance with the model of a leading role of intrinsic mechanisms in cortical arealization.
Collapse
Affiliation(s)
- Sven Mühlfriedel
- Department of Molecular Cell Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen, Germany
| | | | | | | | | |
Collapse
|
29
|
Triolo D, Dina G, Lorenzetti I, Malaguti M, Morana P, Del Carro U, Comi G, Messing A, Quattrini A, Previtali SC. Loss of glial fibrillary acidic protein (GFAP) impairs Schwann cell proliferation and delays nerve regeneration after damage. J Cell Sci 2006; 119:3981-93. [PMID: 16988027 DOI: 10.1242/jcs.03168] [Citation(s) in RCA: 152] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Axonal loss causes disabling and permanent deficits in many peripheral neuropathies, and may result from inefficient nerve regeneration due to a defective relationship between Schwann cells, axons and the extracellular matrix. These interactions are mediated by surface receptors and transduced by cytoskeletal molecules. We investigated whether peripheral nerve regeneration is perturbed in mice that lack glial fibrillary acidic protein (GFAP), a Schwann-cell-specific cytoskeleton constituent upregulated after damage. Peripheral nerves develop and function normally in GFAP-null mice. However, axonal regeneration after damage was delayed. Mutant Schwann cells maintained the ability to dedifferentiate but showed defective proliferation, a key event for successful nerve regeneration. We also showed that GFAP and the other Schwann-cell-intermediate filament vimentin physically interact in two distinct signaling pathways involved in proliferation and nerve regeneration. GFAP binds integrin αvβ8, which initiates mitotic signals soon after damage by interacting with fibrin. Consistently, ERK phosphorylation was reduced in crushed GFAP-null nerves. Vimentin instead binds integrin α5β1, which regulates proliferation and differentiation later in regeneration, and may compensate for the absence of GFAP in mutant mice. GFAP might contribute to form macro-complexes to initiate mitogenic and differentiating signaling for efficient nerve regeneration.
Collapse
Affiliation(s)
- Daniela Triolo
- Neuropathology Unit, San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Wintz H, Yoo LJ, Loguinov A, Wu YY, Steevens JA, Holland RD, Beger RD, Perkins EJ, Hughes O, Vulpe CD. Gene Expression Profiles in Fathead Minnow Exposed to 2,4-DNT: Correlation with Toxicity in Mammals. Toxicol Sci 2006; 94:71-82. [PMID: 16917068 DOI: 10.1093/toxsci/kfl080] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Toxicogenomics, the genome-wide analysis of gene expression to study the effect of toxicants, has great potential for use in environmental toxicology. Applied to standard test organisms, it has possible applications in aquatic toxicology as a sensitive monitoring tool to detect the presence of contaminants while providing information on the mechanisms of action of these pollutants. We describe the use of a complementary DNA (cDNA) microarray of the fathead minnow (Pimephales promelas) a standard sentinel organism in aquatic toxicology, to better understand the mechanisms of toxicity of 2,4-dinitrotoluene (2,4-DNT) which is released in the environment through military and industrial use. We have constructed a fathead minnow microarray containing 5000 randomly picked anonymous cDNAs from a whole fish cDNA library. Expression profiles were analyzed in fish exposed to 2,4-DNT for 10 days at three concentrations (11, 22, and 44 microM, respectively) below the measured median lethal concentration (58 microM). Sequence analysis of cDNAs corresponding to differentially expressed genes affected by exposure revealed that lipid metabolism and oxygen transport genes were prominently affected in a dose-specific manner. We measured liver lipids and demonstrate that lipid metabolism is indeed perturbed following exposure. These observations correlate well with available toxicological data on 2,4-DNT. We present possible modes of action of 2,4-DNT toxicity and suggest that fathead minnow cDNA microarrays can be useful to identify mechanisms of toxicity in fish and as a predictive tool for toxicity in mammals.
Collapse
Affiliation(s)
- Henri Wintz
- Department of Nutritional Sciences and Toxicology, Morgan Hall and Berkeley Institute of the Environment, University of California, Berkeley, California 94720, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Jolivalt CG, Ramos KM, Herbetsson K, Esch FS, Calcutt NA. Therapeutic efficacy of prosaposin-derived peptide on different models of allodynia. Pain 2006; 121:14-21. [PMID: 16480831 DOI: 10.1016/j.pain.2005.11.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2005] [Revised: 11/08/2005] [Accepted: 11/21/2005] [Indexed: 10/25/2022]
Abstract
We have previously demonstrated that the prosaposin-derived 14-mer peptide TX14(A) prevents structural and functional abnormalities associated with peripheral neuropathy in diabetic rats. Unusually, this neuroprotective peptide also exhibited acute anti-hyperalgesic properties in the same model, suggesting a dual action of TX14(A) that could allow therapeutic targeting of both degenerative neuropathy and neuropathic pain. In the present study, we have extended investigation of the anti-allodynic properties of TX14(A) to a range of models in which allodynia is induced using metabolic, physical, neurotoxic or chemical/inflammatory damage to the peripheral nerve. Single systemic doses of TX14(A) rapidly alleviated tactile allodynia in rats in which nerve injury was induced by diabetes, sciatic nerve hemiligation, systemic paclitaxel treatment or paw formalin injection. Further, TX14(A) pre-treatment prevented onset of allodynia in the paclitaxel and formalin injection models. These results indicate that TX14(A) has anti-allodynic properties in diverse models of neuropathic pain and support further exploration of its potential as a therapeutic agent for a wide range of peripheral neuropathies and neuropathic pain states.
Collapse
|
32
|
Li-Korotky HS, Hebda PA, Kelly LA, Lo CY, Dohar JE. Identification of a pre-mRNA splicing factor, arginine/serine-rich 3 (Sfrs3), and its co-expression with fibronectin in fetal and postnatal rabbit airway mucosal and skin wounds. Biochim Biophys Acta Mol Basis Dis 2006; 1762:34-45. [PMID: 16168628 DOI: 10.1016/j.bbadis.2005.08.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2004] [Revised: 06/28/2005] [Accepted: 08/09/2005] [Indexed: 10/25/2022]
Abstract
Fibronectin (FN) is a multi-functional, adhesion protein and involved in multi-steps of the wound healing process. Strong evidence suggests that FN protein diversity is controlled by alternative RNA splicing; a coordinated transcription and RNA processing that is development-, age-, and tissue/cell type-regulated. We previously demonstrated that fetal rabbit airway mucosal healing is regenerative and scarless. Expression, regulation, and biological function of the FN gene and various spliced forms in this model are unknown. Airway and skin incisional wounds were made in fetal (gestation days 21-23), weanling (4-6 weeks) and adult (>6 months) rabbits. Non-wounded and wounded tissues were collected at 12 h (all age groups), 24 h and 48 h (weanling only) post-wounding. Expression profiles were obtained using mRNA differential display and cDNAs of interest were cloned, sequenced and validated by real-time PCR. Here, we report two rabbit cDNAs that showed similar expression patterns after wounding. One encodes a rabbit fibronectin gene, Fn1, and another shares a high sequence homology to a human pre-mRNA splicing factor, arginine/serine-rich 3 (Sfrs3), coding for a RNA binding protein, SRp20. Both Fn1 and Sfrs3 mRNAs were suppressed in fetal wounds but induced in postnatal wounds 12 h post-wounding. The increased levels of both Fn1 and Sfrs3 transcripts were sustained up to 48 h in weanling airway mucosal wounds. The augmentations of the two genes in postnatal airway mucosal wounds were more prominent than that in skin wounds, indicating that the involvement of Sfrs3 and Fn1 genes in postnatal airway mucosal wounds is tissue-specific. Literature provides evidence that SRp20 is indeed involved in the alternative splicing of FN and that the embryonic FN variants reappear during adult wound healing. A connection between the enhanced molecular activity of Sfrs3 and the regulation of the FN gene expression through alternative splicing during the early events of postnatal airway mucosal wound repair was proposed.
Collapse
Affiliation(s)
- Ha-Sheng Li-Korotky
- Division of Pediatric Otolaryngology, Children's Hospital of Pittsburgh, Pittsburgh, PA 15213, USA.
| | | | | | | | | |
Collapse
|
33
|
Desouches C, Alluin O, Mutaftschiev N, Dousset E, Magalon G, Boucraut J, Feron F, Decherchi P. La réparation nerveuse périphérique : 30 siècles de recherche. Rev Neurol (Paris) 2005; 161:1045-59. [PMID: 16288170 DOI: 10.1016/s0035-3787(05)85172-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Nerve injury compromises sensory and motor functions. Techniques of peripheral nerve repair are based on our knowledge regarding regeneration. Microsurgical techniques introduced in the late 1950s and widely developed for the past 20 years have improved repairs. However, functional recovery following a peripheral mixed nerve injury is still incomplete. STATE OF ART Good motor and sensory function after nerve injury depends on the reinnervation of the motor end plates and sensory receptors. Nerve regeneration does not begin if the cell body has not survived the initial injury or if it is unable to initiate regeneration. The regenerated axons must reach and reinnervate the appropriate target end-organs in a timely fashion. Recovery of motor function requires a critical number of motor axons reinnervating the muscle fibers. Sensory recovery is possible if the delay in reinnervation is short. Many additional factors influence the success of nerve repair or reconstruction. The timing of the repair, the level of injury, the extent of the zone of injury, the technical skill of the surgeon, and the method of repair and reconstruction contribute to the functional outcome after nerve injury. CONCLUSION This review presents the recent advances in understanding of neural regeneration and their application to the management of primary repairs and nerve gaps.
Collapse
Affiliation(s)
- C Desouches
- Service de Chirurgie de la Main, Chirurgie Plastique et Réparatrice des Membres, Assistance Publique, Hôpitaux de Marseille, Hôpital de la Conception, Marseille
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Bampton ETW, Ma CH, Tolkovsky AM, Taylor JSH. Osteonectin is a Schwann cell-secreted factor that promotes retinal ganglion cell survival and process outgrowth. Eur J Neurosci 2005; 21:2611-23. [PMID: 15926910 DOI: 10.1111/j.1460-9568.2005.04128.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
We have investigated the factors made by Schwann cells (SCs) that stimulate survival and neurite outgrowth from postnatal rat retinal ganglion cells (RGCs). These effects are preserved under K252a blockade of the Trk family of neurotrophin receptors and are not fully mimicked by the action of a number of known trophic factors. To identify novel factors responsible for this regenerative activity, we have used a radiolabelling assay. Proteins made by SCs were labelled radioactively and then fed to purified RGCs. The proteins taken up by the RGCs were then isolated and further characterized. Using this assay we have identified a major 40 kDa factor taken up by RGCs, which was microsequenced and shown to be the matricellular protein osteonectin (ON). Using an in vitro assay of purified RGCs we show that ON promotes both survival and neurite outgrowth. We conclude that ON has a potential new role in promoting CNS repair.
Collapse
Affiliation(s)
- Edward T W Bampton
- Department of Human Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | | | | | | |
Collapse
|
35
|
Yuan Y, Zhang P, Yang Y, Wang X, Gu X. The interaction of Schwann cells with chitosan membranes and fibers in vitro. Biomaterials 2004; 25:4273-8. [PMID: 15046917 DOI: 10.1016/j.biomaterials.2003.11.029] [Citation(s) in RCA: 178] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2003] [Accepted: 11/11/2003] [Indexed: 11/29/2022]
Abstract
The bridging of nerve gaps is still one of the major problems in peripheral nerve regeneration. A promising alternative for the repair of peripheral nerve injuries is the bioartificial nerve graft, comprised of a biomaterial pre-seeded with Schwann cells (SCs), which is an effective substrate for enhancing nerve regeneration. Interaction between cultured SCs and biomaterials is of importance. For the purposes of this study, culture systems of normal SCs were used. The biocompatibility of chitosan, including chitosan membranes and chitosan fibers, was evaluated in vitro. The growth of SCs was observed by light and scanning electron microscopy at regular intervals. SCs were identified by immunocytochemical staining and the viability of SCs was measured by MTT assay. The experimental results indicated that SCs could grow onto chitosan materials with two different shapes: spherical and long olivary. They contacted with the extensions. The long olivary cells inclined to encircle chitosan fibers up. It was also found that the cells on the chitosan fibers migrated faster than those on the chitosan membranes. There was a good biological compatibility between chitosan and SCs. Compared with the chitosan membranes, SCs migrated more easily onto the stereoframe of chitosan fibers. These studies contribute information necessary to enhancing our understanding of biocompatibility of chitosan.
Collapse
Affiliation(s)
- Ying Yuan
- The Jiangsu Key Laboratory of Neuroregeneration, Nantong Medical College, Nantong, Jiangsu 226001, PR China
| | | | | | | | | |
Collapse
|
36
|
Ara J, Bannerman P, Hahn A, Ramirez S, Pleasure D. Modulation of sciatic nerve expression of class 3 semaphorins by nerve injury. Neurochem Res 2004; 29:1153-9. [PMID: 15176472 DOI: 10.1023/b:nere.0000023602.72354.82] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Beginning with the unexpected finding by cDNA array analysis that neuropilin-2 is induced in sciatic nerve distal to a transection, we document, for the first time, up-regulation in the axotomized adult peripheral nervous system of class 3 semaphorins and their receptors, which are known to play prominent roles in axonal guidance during neural development. Previously, we described the use of cDNA arrays to screen for novel peripheral nervous system axotomy-induced candidate neurotrophic proteins. A novel finding of that prior study was substantial induction of neuropilin 2 (NP2) mRNA in the axotomized nerve segments. Following up on that initial observation, we have now used real-time quantitative reverse transcription-polymerase chain reaction to demonstrate induction of genes encoding neuropilin 1 (NP1), which, like NP2, serves as a coreceptor for members of the class 3 semaphorin family of axonal guidance molecules and of five of the six known class 3 semaphorins (Sema3A, Sema3B, Sema3C, Sema3E, and Sema3F, but not Sema3D) in crushed or transected sciatic nerves.
Collapse
Affiliation(s)
- Jahan Ara
- Division of Neurology, Joseph Stokes, Jr. Research Institute, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | |
Collapse
|
37
|
Bareyre FM, Schwab ME. Inflammation, degeneration and regeneration in the injured spinal cord: insights from DNA microarrays. Trends Neurosci 2003; 26:555-63. [PMID: 14522149 DOI: 10.1016/j.tins.2003.08.004] [Citation(s) in RCA: 218] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
GeneChip microarrays have recently been introduced to the field of neurobiology to identify and monitor the expression levels of thousands of genes simultaneously. This powerful technique is now used for studying the pathophysiology of CNS injuries including spinal cord lesions. Early stages after injury are characterized by the strong upregulation of genes involved in transcription and inflammation and a general downregulation of structural proteins and proteins involved in neurotransmission. Later, an increase in the expression of growth factors, axonal guidance factors, extracellular matrix molecules and angiogenic factors reflects the attempts for repair, while upregulation of stress genes and proteases and downregulation of cytoskeletal and synaptic mRNA reflect the struggle of the tissue to survive. DNA microarrays have the potential to aid discovery of new targets for neuroprotective or restorative therapeutic approaches
Collapse
Affiliation(s)
- Florence M Bareyre
- Brain Research Institute, University of Zürich, Winterthurerstrasse 190, 8057 Zürich, Switzerland.
| | | |
Collapse
|
38
|
Hiraiwa M, Liu J, Lu AG, Wang CY, Misasi R, Yamauchi T, Hozumi I, Inuzuka T, O'Brien JS. Regulation of gene expression in response to brain injury: enhanced expression and alternative splicing of rat prosaposin (SGP-1) mRNA in injured brain. J Neurotrauma 2003; 20:755-65. [PMID: 12965054 DOI: 10.1089/089771503767869980] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Prosaposin, the precursor of saposins or saps, is an injury-repair protein that acts on both neurons and glia. Previous studies identified the prosaposin gene as one of differentially expressed genes following nerve injury. In the present study, we investigated expression of prosaposin mRNA in injured brain utilizing rat models of focal cerebral ischemia and cortical stab wound in order to explore the significance of prosaposin in nerve injury. In ischemic brain, the level of prosaposin mRNA was elevated greater than 400% over controls within 5 days after ischemic insults. Importantly, this induction was accompanied by a 9-base splicing consistent with the alternative Exon-8 splicing of human prosaposin mRNA. In normal brain, two prosaposin mRNA species with and without the 9-base insertion were expressed at a ratio of 85:15; however, this equilibrium reverted to 5:95 following ischemic injury. Similar inductions were observed in stab wound brains. Immunohistochemical staining and in situ hybridization demonstrated an enhanced signal distribution of prosaposin mRNA and injury-induced prosaposin protein around the lesion. The data suggest the expression and processing of prosaposin mRNA may be crucially regulated not only for cerebral homeostasis but also during nerve regenerative and degenerative processes.
Collapse
Affiliation(s)
- Masao Hiraiwa
- Department of Neurosciences, School of Medicine, Center for Molecular Genetics, University of California at San Diego, La Jolla, California 92093-0634, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Schweitzer J, Becker T, Becker CG, Schachner M. Expression of protein zero is increased in lesioned axon pathways in the central nervous system of adult zebrafish. Glia 2003; 41:301-17. [PMID: 12528184 DOI: 10.1002/glia.10192] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The immunoglobulin superfamily molecule protein zero (P0) is important for myelin formation and may also play a role in adult axon regeneration, since it promotes neurite outgrowth in vitro. Moreover, it is expressed in the regenerating central nervous system (CNS) of fish, but not in the nonregenerating CNS of mammals. We identified a P0 homolog in zebrafish. Cell type-specific expression of P0 begins in the ventromedial hindbrain and the optic chiasm at 3-5 days of development. Later (at 4 weeks) expression has spread throughout the optic system and spinal cord. This is consistent with a role for P0 in CNS myelination during development. In the adult CNS, glial cells constitutively express P0 mRNA. After an optic nerve crush, expression is increased within 2 days in the entire optic pathway. Expression peaks at 1 to 2 months and remains elevated for at least 6 months postlesion. After enucleation, P0 mRNA expression is also upregulated but fails to reach the high levels observed in crush-lesioned animals at 4 weeks postlesion. Spinal cord transection leads to increased expression of P0 mRNA in the spinal cord caudal to the lesion site. The glial upregulation of P0 mRNA expression after a lesion of the adult zebrafish CNS suggests roles for P0 in promoting axon regeneration and remyelination after injury.
Collapse
Affiliation(s)
- Jörn Schweitzer
- Zentrum für Molekulare Neurobiologie, Universität Hamburg, Hamburg, Germany
| | | | | | | |
Collapse
|
40
|
Setton-Avruj CP, Aquino JB, Goedelman CJ, Soto EF, Villar MJ. P0 and myelin basic protein-like immunoreactivities following ligation of the sciatic nerve in the rat. Neurochem Res 2002; 27:1293-303. [PMID: 12512935 DOI: 10.1023/a:1021663414122] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
In this work we analyzed variations in the expression of MBPs and P0 in ligated sciatic nerves of young and adult rats at 3, 7, and 14 days postligation (PL), by immunohistochemistry and SDS-PAGE of isolated myelin. A protein redistribution was seen in the distal stump of ligated nerves with the appearance of immunoreactive clusters. Using the KS400 image analyzer, immunostained area values were obtained from the different nerves dissected. In adult rats, there was an increase of the immunostained area for MBP from 3 to 7 days PL, coincident with a reorganization of the marker in clusters, followed by a marked decrease at 14 days. P0 immunolabeling gave similar results without, however, a decrease of the immunostained area at the longer survival time tested. Young animals showed an acceleration in the process of protein redistribution and digestion within ligated nerves, which followed a similar pattern as that of adult animals. Analysis by electrophoresis showed a marked decrease in P0 and MBP at 7 days PL in young rats and 14 days PL in adult rats. The functional significance of protein clustering within myelin in injured nerves deserves further analysis.
Collapse
Affiliation(s)
- C Patricia Setton-Avruj
- Department of Biochemistry, Faculty of Pharmacy and Biochemistry, University of Buenos Aires, IQUIFIB-CONICET, Junín 956, 1113, Argentina
| | | | | | | | | |
Collapse
|
41
|
Mammalian achaete scute homolog 2 is expressed in the adult sciatic nerve and regulates the expression of Krox24, Mob-1, CXCR4, and p57kip2 in Schwann cells. J Neurosci 2002. [PMID: 12196582 DOI: 10.1523/jneurosci.22-17-07586.2002] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The molecular control mechanisms and regulatory molecules involved in nerve repair are not yet well known. Schwann cells have been attributed an important role in peripheral nerve regeneration; therefore, attention has been drawn to regulatory factors expressed by these glial cells. Here, we demonstrate that Mash2, a basic helix-loop-helix (bHLH) transcription factor previously shown to be crucial for placenta development, is expressed by Schwann cells of adult peripheral nerves. We observed that this gene is downregulated after nerve lesion and, using cDNA array hybridization technology, we could demonstrate that Mash2 is a regulator of Krox24, Mob-1, and CXCR4 expression in cultured Schwann cells. In addition, we provide strong evidence that Mash2 is a negative regulator of Schwann cell proliferation. Mash2 represents a first candidate for the missing class B bHLH proteins in peripheral nerves.
Collapse
|
42
|
Kim DS, Lee SJ, Park SY, Yoo HJ, Kim SH, Kim KJ, Cho HJ. Differentially expressed genes in rat dorsal root ganglia following peripheral nerve injury. Neuroreport 2001; 12:3401-5. [PMID: 11711894 DOI: 10.1097/00001756-200110290-00050] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Ordered differential display PCR was used to identify differentially expressed genes in rat dorsal root ganglia at 7 days following chronic constriction injury (CCI) of the sciatic nerve. Fourteen differentially displayed cDNA bands were isolated, cloned and verified by RT-PCR. The four mRNAs were increased, which included mRNAs encoding heat shock protein 27, fatty acid binding protein, apolipoprotein D and one novel gene. Six down-regulated clones were microtubule-associated protein 1B, protein tyrosine phosphatase alpha, Kv1.2 channel, myelin protein SR13, medium-sized neurofilament protein, and one novel gene. Our results show that many differentially regulated genes after CCI may play a role in nerve degeneration and/or regeneration and provide a molecular framework for understanding the peripheral mechanism underlying neuropathic pain.
Collapse
Affiliation(s)
- D S Kim
- Department of Anatomy, School of Medicine, Kyungpook National University, 2-101, Dongin Dong, Taegu, 700-422, Korea
| | | | | | | | | | | | | |
Collapse
|
43
|
Fan M, Mi R, Yew DT, Chan WY. Analysis of gene expression following sciatic nerve crush and spinal cord hemisection in the mouse by microarray expression profiling. Cell Mol Neurobiol 2001; 21:497-508. [PMID: 11860187 PMCID: PMC11533824 DOI: 10.1023/a:1013867306555] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
1. The responses of periphery (PNS) and central nervous systems (CNS) towards nerve injury are different: while injured mammalian periphery nerons can successfully undergo regeneration, axons in the central nervous system are usually not able to regenerate. 2. In the present study, the genes which were differentially expressed in the PNS and CNS following nerve injury were identified and compared by microarray profiling techniques. 3. Sciatic nerve crush and hemisection of the spinal cord of adult mice were used as the models for nerve injury in PNS and CNS respectivey. 4. It was found that of all the genes examined, 14% (80/588) showed changes in expression following either PNS or CNS injury, and only 3% (18/588) showed changes in both types of injuries. 5. Among all the differentially expressed genes, only 8% (6/80) exhibited similar changes in gene expression (either up- or down-regulation) following injury in both PNS and CNS nerve injuries. 6. Our results indicated that microarray expression profiling is an efficient and useful method to identify genes that are involved in the regeneration process following nerve injuries, and several genes which are differentially expressed in the PNS and/or CNS following nerve injuries were identified in the present study.
Collapse
Affiliation(s)
- M Fan
- Institute of Basic Medical Science, Academy of Military Science, Beijing, People's Republic of China
| | | | | | | |
Collapse
|
44
|
Mizisin AP, Steinhardt RC, O'Brien JS, Calcutt NA. TX14(A), a prosaposin-derived peptide, reverses established nerve disorders in streptozotocin-diabetic rats and prevents them in galactose-fed rats. J Neuropathol Exp Neurol 2001; 60:953-60. [PMID: 11589426 DOI: 10.1093/jnen/60.10.953] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Recently, TX14(A), a prosaposin-derived neurotrophic peptide, was shown to prevent both large and small fiber deficits in streptozotocin diabetes. Here, the efficacy of TX14(A) in reversing established nerve conduction disorders in streptozotocin diabetes, a model of insulin deficiency, and preventing them in galactose feeding, an insulin-replete model of polyol pathway flux, was investigated. Following streptozotocin injection (50 mg/kg ip), TX14(A) treatment (1 mg/kg ip thrice weekly) was initiated in half of the animals. After 8 wk, treatment was begun in half of the untreated animals and discontinued in half of the treated animals, and the experiment continued for 6 wk. TX14(A) reversed established motor and sensory nerve conduction deficits in streptozotocin-diabetic rats and the impact of previous treatment was still evident 3 wk after withdrawal. With the onset of 40% galactose feeding, the same dose of TX14(A) was given to half of the control and half of the galactose-fed animals for 16 wk. TX14(A) was without effect in control animals but it attenuated motor and sensory nerve conduction deficits in galactose-fed rats, an effect associated with amelioration of axonal dwindling in the sciatic nerve. These observations extend the therapeutic utility of TX14(A) and highlight its potential in treating established diabetic neuropathy.
Collapse
Affiliation(s)
- A P Mizisin
- Department of Pathology, School of Medicine, University of California, San Diego, La Jolla, 92093-0612, USA
| | | | | | | |
Collapse
|
45
|
Nagarajan R, Svaren J, Le N, Araki T, Watson M, Milbrandt J. EGR2 mutations in inherited neuropathies dominant-negatively inhibit myelin gene expression. Neuron 2001; 30:355-68. [PMID: 11394999 DOI: 10.1016/s0896-6273(01)00282-3] [Citation(s) in RCA: 189] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The identification of EGR2 mutations in patients with neuropathies and the phenotype Egr2/Krox20(-/-) have demonstrated that the Egr2 transcription factor is critical for peripheral nerve myelination. However, the mechanism by which these mutations cause disease remains unclear, as most patients present with disease in the heterozygous state, whereas Egr2(+/-) mice are phenotypically normal. To understand the effect of aberrant Egr2 activity on Schwann cell gene expression, we performed microarray expression profiling to identify genes regulated by Egr2 in Schwann cells. These include genes encoding myelin proteins and enzymes required for synthesis of normal myelin lipids. Using these newly identified targets, we have shown that neuropathy-associated EGR2 mutants dominant-negatively inhibit wild-type Egr2-mediated expression of essential myelin genes to levels sufficiently low to result in the abnormal myelination observed in these patients.
Collapse
Affiliation(s)
- R Nagarajan
- Department of Pathology, Washington University School of Medicine, 660 South Euclid Avenue, Box 8118, St. Louis, MO 63110, USA
| | | | | | | | | | | |
Collapse
|
46
|
Bosse F, Petzold G, Greiner-Petter R, Pippirs U, Gillen C, Müller HW. Cellular localization of the disintegrin CRII-7/rMDC15 mRNA in rat PNS and CNS and regulated expression in postnatal development and after nerve injury. Glia 2000; 32:313-27. [PMID: 11102971 DOI: 10.1002/1098-1136(200012)32:3<313::aid-glia100>3.0.co;2-g] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Disintegrins perform putative functions in cell adhesion, signaling and fusion. We have isolated a 2815-bp rat cDNA (CRII-7) representing a transcript that is differentially expressed during sciatic nerve regeneration. Nucleotide sequence comparison indicates that CRII-7 is the rat homologue to the recently cloned cDNAs MDC15 (ADAM 15) and metargidin (hMDC15) of mouse and human, respectively. The CRII-7 cDNA (rMDC15) encodes a membrane-anchored glycoprotein of approximately 85 kDa containing a disintegrin and a metalloprotease domain. Cellular metalloprotease disintegrins are a family of proteins (ADAMs or MDC proteins) with important roles, e.g., in cell-cell interactions during fertilization, muscle and nerve development, or tumor necrosis factor-alpha (TNF-alpha) cleavage. Northern blot analysis demonstrated a predominant expression of CRII-7/rMDC15 in the nervous system (PNS and CNS) and lung. Analysis of the CRII-7/rMDC15 transcript levels following peripheral nerve lesions demonstrated regulated mRNA expression during Wallerian degeneration and nerve regeneration. The steady-state levels of CRII-7/rMDC15 transcripts markedly increased within the first day after lesion and then steadily decreased for at least 4 weeks. CRII-7/rMDC15 mRNA expression was further examined during postnatal development and maturation of rat sciatic nerve and brain, as well as in cultured Schwann cells, meningeal fibroblasts, and astrocytes. In situ hybridization on paraffin sections showed the cellular localization of CRII-7/rMDC15 mRNA in Schwann cells and endothelial cells of peripheral nerve and in various neuronal populations in brain and spinal cord.
Collapse
Affiliation(s)
- F Bosse
- Molecular Neurobiology Laboratory, Department of Neurology, Heinrich-Heine University of Düsseldorf, Düsseldorf, Germany.
| | | | | | | | | | | |
Collapse
|
47
|
Chan JR, Rodriguez-Waitkus PM, Ng BK, Liang P, Glaser M. Progesterone synthesized by Schwann cells during myelin formation regulates neuronal gene expression. Mol Biol Cell 2000; 11:2283-95. [PMID: 10888668 PMCID: PMC14919 DOI: 10.1091/mbc.11.7.2283] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Previously, progesterone was found to regulate the initiation and biosynthetic rate of myelin synthesis in Schwann cell/neuronal cocultures. The mRNA for cytochrome P450scc (converts cholesterol to pregnenolone), 3beta-hydroxysteroid dehydrogenase (3beta-HSD, converts pregnenolone to progesterone), and the progesterone receptor were found to be markedly induced during active myelin synthesis. However, the cells in the cocultures responsible for these changes were not identified. In this study, in situ hybridization was used to determine the localization of the enzymes responsible for steroid biosynthesis. The mRNA for cytochrome P450scc and 3beta-HSD were detected only in actively myelinating cocultures and were localized exclusively in the Schwann cells. Using immunocytochemistry, with minimal staining of the Schwann cells, we found the progesterone receptor in the dorsal root ganglia (DRG) neurons. The progesterone receptor in the neurons translocated into the nuclei of these cells when progesterone was added to neuronal cultures or during myelin synthesis in the cocultures. Additionally, a marked induction of the progesterone receptor was found in neuronal cultures after the addition of progesterone. The induction of various genes in the neurons was also investigated using mRNA differential display PCR in an attempt to elucidate the mechanism of steroid action on myelin synthesis. Two novel genes were induced in neuronal cultures by progesterone. These genes, along with the progesterone receptor, were also induced in cocultures during myelin synthesis, and their induction was blocked by RU-486 (a progesterone receptor antagonist). These genes were not induced in Schwann cells cultured alone after the addition of progesterone. These results suggest that progesterone is synthesized in Schwann cells and that it can indirectly regulate myelin formation by activating transcription via the classical steroid receptor in the DRG neurons.
Collapse
Affiliation(s)
- J R Chan
- Department of Biochemistry and Neuroscience Program, University of Illinois, Urbana, Illinois 61801, USA
| | | | | | | | | |
Collapse
|
48
|
Gleichmann M, Gillen C, Czardybon M, Bosse F, Greiner-Petter R, Auer J, Müller HW. Cloning and characterization of SDF-1gamma, a novel SDF-1 chemokine transcript with developmentally regulated expression in the nervous system. Eur J Neurosci 2000; 12:1857-66. [PMID: 10886327 DOI: 10.1046/j.1460-9568.2000.00048.x] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The cytokines SDF-1alpha and -1beta are two alternatively spliced variants of the CXC (alpha) chemokines that are highly conserved among species. SDF-1alpha was shown to function as a B-cell maturation factor, a ligand for the CXCR4 (LESTR/fusin) chemokine receptor, thereby inhibiting replication of T cell-tropic HIV-1 strains and inducing cell death in human neuronal cell lines. In this report the cloning of the rat SDF-1beta cDNA and a new SDF-1 isoform, SDF-1gamma, are presented. Using Northern blot analysis, the expression pattern of both isoforms was studied in different tissues and it is shown that during postnatal development of the central and peripheral nervous system SDF-1beta- and SDF-1gamma-mRNA expression is inversely regulated. Whilst SDF-1beta-mRNA is the predominant isoform in embryonic and early postnatal nerve tissue, SDF-1gamma-mRNA is expressed at higher levels in adulthood. After peripheral nerve lesion a transient increase in SDF-1beta-mRNA expression is observed. As revealed by in situ hybridization, neurons and Schwann cells are the main cellular sources of both SDF-1beta and SDF-1gamma mRNAs in the nervous system. Computer-assisted analysis revealed that both transcripts encode secreted peptides with putative proteolytic cleavage sites which might generate novel neuropeptides.
Collapse
Affiliation(s)
- M Gleichmann
- Molecular Neurobiology Laboratory, Department of Neurology, andBiomedical Research Center, University of Düsseldorf, Moorenstrasse 5, D-40225 Düsseldorf, Germany
| | | | | | | | | | | | | |
Collapse
|
49
|
Hiebert GW, Dyer JK, Tetzlaff W, Steeves JD. Immunological myelin disruption does not alter expression of regeneration-associated genes in intact or axotomized rubrospinal neurons. Exp Neurol 2000; 163:149-56. [PMID: 10785453 DOI: 10.1006/exnr.2000.7351] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The inability of axotomized neurons to regenerate within the CNS has been partially attributed to a number of inhibitory factors associated with CNS myelin that are extrinsic to the severed neurons. However, some neurons are capable of limited regeneration after injury and this ability has been shown to correlate with the expression of certain regeneration-associated genes (RAGs) intrinsic to injured neurons. It has therefore been postulated that neutralization of inhibitory factors, as well as the induction of an appropriate neuronal cell body response, would facilitate improved regrowth of injured CNS axons. In previous studies we have shown that immunological removal of myelin from the spinal cord facilitates axonal regeneration by rubrospinal neurons, as indicated by retrograde transport of a fluorescent dye placed distal to the site of injury. Here, we investigated whether the immunological focal removal of spinal cord myelin, following a thoracic spinal cord injury, concomitantly stimulated an increase in the expression of RAGs in rubrospinal neurons. In situ hybridization for Talpha-1 tubulin and GAP-43 at days 7, 14, and 21 revealed no significant increase in gene expression in rubrospinal neurons following immunological demyelination. The ability of various neuronal populations to sprout or slowly regrow without expressing the previously characterized cell body response is reviewed. We conclude that the recently demonstrated regeneration of rubrospinal tract, after immunologically directed spinal cord demyelination, is the result of either axonal sprouting or slow axonal regrowth without the increased expression of RAGs characteristic for fast axon regeneration.
Collapse
Affiliation(s)
- G W Hiebert
- CORD (Collaboration On Repair Discoveries), c/o Biosciences Building, Department of Zoology, University of British Columbia, 6270 University Boulevard, Vancouver, British Columbia, V6T 1Z4, Canada
| | | | | | | |
Collapse
|
50
|
Jetten AM, Suter U. The peripheral myelin protein 22 and epithelial membrane protein family. PROGRESS IN NUCLEIC ACID RESEARCH AND MOLECULAR BIOLOGY 2000; 64:97-129. [PMID: 10697408 DOI: 10.1016/s0079-6603(00)64003-5] [Citation(s) in RCA: 85] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The peripheral myelin protein 22 (PMP22) and the epithelial membrane proteins (EMP-1, -2, and -3) comprise a subfamily of small hydrophobic membrane proteins. The putative four-transmembrane domain structure as well as the genomic structure are highly conserved among family members. PMP22 and EMPs are expressed in many tissues, and functions in cell growth, differentiation, and apoptosis have been reported. EMP-1 is highly up-regulated during squamous differentiation and in certain tumors, and a role in tumorigenesis has been proposed. PMP22 is most highly expressed in peripheral nerves, where it is localized in the compact portion of myelin. It plays a crucial role in normal physiological and pathological processes in the peripheral nervous system. Progress in molecular genetics has revealed that genetic alterations in the PMP22 gene, including duplications, deletions, and point mutations, are responsible for several forms of hereditary peripheral neuropathies, including Charcot-Marie-Tooth disease type 1A (CMT1A), Dejerine-Sottas syndrome (DDS), and hereditary neuropathy with liability to pressure palsies (HNPP). The natural mouse mutants Trembler and Trembler-J contain a missense mutation in different hydrophobic domains of PMP22, resulting in demyelination and Schwann cell proliferation. Transgenic mice carrying many copies of the PMP22 gene and PMP22-null mice display a variety of defects in the initial steps of myelination and/or maintenance of myelination, whereas no pathological alterations are detected in other tissues normally expressing PMP22. Further characterization of the interactions of PMP22 and EMPs with other proteins as well as their regulation will provide additional insight into their normal physiological function and their roles in disease and possibly will result in the development of therapeutic tools.
Collapse
Affiliation(s)
- A M Jetten
- Cell Biology Section, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
| | | |
Collapse
|