1
|
Tian R, Zhou Y, Ren Y, Zhang Y, Tang W. Wallerian degeneration: From mechanism to disease to imaging. Heliyon 2025; 11:e40729. [PMID: 39811315 PMCID: PMC11730939 DOI: 10.1016/j.heliyon.2024.e40729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 08/12/2024] [Accepted: 11/25/2024] [Indexed: 01/16/2025] Open
Abstract
Wallerian degeneration (WD) was first discovered by Augustus Waller in 1850 in a transection of the glossopharyngeal and hypoglossal nerves in frogs. Initial studies suggested that the formation mechanism of WD is related to the nutrition of neuronal cell bodies to axons. However, with the wide application of transgenic mice in experiments, the latest studies have found that the mechanism of WD is related to axonal degeneration, myelin clearance and extracellular matrix. This review summarizes the discovery and research progress of WD and discusses the mechanism of WD from the perspective of molecular biology. In addition, this review combines the etiology, symptoms and imaging results of WD patients, and analyzes the clinical and imaging characteristics of WD, to provide the best perspective for future clinical research.
Collapse
Affiliation(s)
- Ruiqi Tian
- Department of Neurology, Xinhua Hospital Affiliated with Dalian University, Dalian, Liaoning Province, China
| | - Yingying Zhou
- Department of Neurology, Xinhua Hospital Affiliated with Dalian University, Dalian, Liaoning Province, China
| | - Yuan Ren
- Department of Neurology, Xinhua Hospital Affiliated with Dalian University, Dalian, Liaoning Province, China
| | - Yisen Zhang
- Department of Neurology, Xinhua Hospital Affiliated with Dalian University, Dalian, Liaoning Province, China
| | - Wei Tang
- Department of Neurology, Xinhua Hospital Affiliated with Dalian University, Dalian, Liaoning Province, China
| |
Collapse
|
2
|
Abstract
Significant advances have been made in recent years in identifying the genetic components of Wallerian degeneration, the process that brings the progressive destruction and removal of injured axons. It has now been accepted that Wallerian degeneration is an active and dynamic cellular process that is well regulated at molecular and cellular levels. In this review, we describe our current understanding of Wallerian degeneration, focusing on the molecular players and mechanisms that mediate the injury response, activate the degenerative program, transduce the death signal, execute the destruction order, and finally, clear away the debris. By highlighting the starring roles and sketching out the molecular script of Wallerian degeneration, we hope to provide a useful framework to understand Wallerian and Wallerian-like degeneration and to lay a foundation for developing new therapeutic strategies to treat axon degeneration in neural injury as well as in neurodegenerative disease. Expected final online publication date for the Annual Review of Genetics, Volume 55 is November 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Kai Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201203, China; , , .,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mingsheng Jiang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201203, China; , , .,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yanshan Fang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201203, China; , , .,University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
3
|
Mailhot B, Christin M, Tessandier N, Sotoudeh C, Bretheau F, Turmel R, Pellerin È, Wang F, Bories C, Joly-Beauparlant C, De Koninck Y, Droit A, Cicchetti F, Scherrer G, Boilard E, Sharif-Naeini R, Lacroix S. Neuronal interleukin-1 receptors mediate pain in chronic inflammatory diseases. J Exp Med 2021; 217:151879. [PMID: 32573694 PMCID: PMC7478735 DOI: 10.1084/jem.20191430] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 03/03/2020] [Accepted: 05/13/2020] [Indexed: 12/20/2022] Open
Abstract
Chronic pain is a major comorbidity of chronic inflammatory diseases. Here, we report that the cytokine IL-1β, which is abundantly produced during multiple sclerosis (MS), arthritis (RA), and osteoarthritis (OA) both in humans and in animal models, drives pain associated with these diseases. We found that the type 1 IL-1 receptor (IL-1R1) is highly expressed in the mouse and human by a subpopulation of TRPV1+ dorsal root ganglion neurons specialized in detecting painful stimuli, termed nociceptors. Strikingly, deletion of the Il1r1 gene specifically in TRPV1+ nociceptors prevented the development of mechanical allodynia without affecting clinical signs and disease progression in mice with experimental autoimmune encephalomyelitis and K/BxN serum transfer–induced RA. Conditional restoration of IL-1R1 expression in nociceptors of IL-1R1–knockout mice induced pain behavior but did not affect joint damage in monosodium iodoacetate–induced OA. Collectively, these data reveal that neuronal IL-1R1 signaling mediates pain, uncovering the potential benefit of anti–IL-1 therapies for pain management in patients with chronic inflammatory diseases.
Collapse
Affiliation(s)
- Benoit Mailhot
- Axe Neurosciences du Centre de recherche du CHU de Québec-Université Laval et Département de médecine moléculaire de l'Université Laval, Québec, Canada
| | - Marine Christin
- Department of Physiology and Cell Information Systems Group, McGill University, Montreal, Canada
| | - Nicolas Tessandier
- Axe Maladies infectieuses et immunitaires du Centre de recherche du CHU de Québec-Université Laval et Département de microbiologie-infectiologie et d'immunologie de l'Université Laval, Québec, Canada
| | - Chaudy Sotoudeh
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Palo Alto, CA
| | - Floriane Bretheau
- Axe Neurosciences du Centre de recherche du CHU de Québec-Université Laval et Département de médecine moléculaire de l'Université Laval, Québec, Canada
| | - Roxanne Turmel
- Axe Neurosciences du Centre de recherche du CHU de Québec-Université Laval et Département de médecine moléculaire de l'Université Laval, Québec, Canada
| | - Ève Pellerin
- Axe Neurosciences du Centre de recherche du CHU de Québec-Université Laval et Département de médecine moléculaire de l'Université Laval, Québec, Canada
| | - Feng Wang
- Centre de recherche CERVO, Québec, Canada
| | | | - Charles Joly-Beauparlant
- Axe Endocrinologie-néphrologie du Centre de recherche du CHU de Québec-Université Laval et Département de médecine moléculaire de l'Université Laval, Québec, Canada
| | | | - Arnaud Droit
- Axe Endocrinologie-néphrologie du Centre de recherche du CHU de Québec-Université Laval et Département de médecine moléculaire de l'Université Laval, Québec, Canada
| | - Francesca Cicchetti
- Axe Neurosciences du Centre de recherche du CHU de Québec-Université Laval et Département de psychiatrie et de neurosciences de l'Université Laval, Québec, Canada
| | - Grégory Scherrer
- Department of Cell Biology and Physiology, University of North Carolina Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC.,New York Stem Cell Foundation - Robertson Investigator, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Eric Boilard
- Axe Maladies infectieuses et immunitaires du Centre de recherche du CHU de Québec-Université Laval et Département de microbiologie-infectiologie et d'immunologie de l'Université Laval, Québec, Canada
| | - Reza Sharif-Naeini
- Department of Physiology and Cell Information Systems Group, McGill University, Montreal, Canada
| | - Steve Lacroix
- Axe Neurosciences du Centre de recherche du CHU de Québec-Université Laval et Département de médecine moléculaire de l'Université Laval, Québec, Canada
| |
Collapse
|
4
|
Pieper AA, McKnight SL. Benefits of Enhancing Nicotinamide Adenine Dinucleotide Levels in Damaged or Diseased Nerve Cells. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2019; 83:207-217. [PMID: 30787047 DOI: 10.1101/sqb.2018.83.037622] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Three unbiased lines of research have commonly pointed to the benefits of enhanced levels of nicotinamide adenine dinucleotide (NAD+) to diseased or damaged neurons. Mice carrying a triplication of the gene encoding the culminating enzyme in NAD+ salvage from nicotinamide, NMNAT, are protected from a variety of insults to axons. Protection from Wallerian degeneration of axons is also observed in flies and mice bearing inactivating mutations in the SARM1 gene. Functional studies of the SARM1 gene product have revealed the presence of an enzymatic activity directed toward the hydrolysis of NAD+ Finally, an unbiased drug screen performed in living mice led to the discovery of a neuroprotective chemical designated P7C3. Biochemical studies of the P7C3 chemical show that it can enhance recovery of NAD+ from nicotinamide by activating NAMPT, the first enzyme in the salvage pathway. In combination, these three unrelated research endeavors offer evidence of the benefits of enhanced NAD+ levels to damaged neurons.
Collapse
Affiliation(s)
- Andrew A Pieper
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio 44106, USA
- Department of Psychiatry, Case Western Reserve University, Cleveland, Ohio 44106, USA
- Geriatric Research Education and Clinical Centers, Louis Stokes Cleveland VAMC, Cleveland, Ohio 44106, USA
| | - Steven L McKnight
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| |
Collapse
|
5
|
Turkiew E, Falconer D, Reed N, Höke A. Deletion of Sarm1 gene is neuroprotective in two models of peripheral neuropathy. J Peripher Nerv Syst 2018; 22:162-171. [PMID: 28485482 DOI: 10.1111/jns.12219] [Citation(s) in RCA: 137] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 05/04/2017] [Accepted: 05/05/2017] [Indexed: 12/13/2022]
Abstract
Distal axon degeneration seen in many peripheral neuropathies is likely to share common molecular mechanisms with Wallerian degeneration. Although several studies in mouse models of peripheral neuropathy showed prevention of axon degeneration in the slow Wallerian degeneration (Wlds) mouse, the role of a recently identified player in Wallerian degeneration, Sarm1, has not been explored extensively. In this study, we show that mice lacking the Sarm1 gene are resistant to distal axonal degeneration in a model of chemotherapy induced peripheral neuropathy caused by paclitaxel and a model of high fat diet induced putative metabolic neuropathy. This study extends the role of Sarm1 to axon degeneration seen in peripheral neuropathies and identifies it as a likely target for therapeutic development.
Collapse
Affiliation(s)
- Elliot Turkiew
- Department of Neurology and Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Debbie Falconer
- Department of Neurology and Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Nicole Reed
- Department of Neurology and Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Ahmet Höke
- Department of Neurology and Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
6
|
Farley JE, Burdett TC, Barria R, Neukomm LJ, Kenna KP, Landers JE, Freeman MR. Transcription factor Pebbled/RREB1 regulates injury-induced axon degeneration. Proc Natl Acad Sci U S A 2018; 115:1358-1363. [PMID: 29295933 PMCID: PMC5819420 DOI: 10.1073/pnas.1715837115] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Genetic studies of Wallerian degeneration have led to the identification of signaling molecules (e.g., dSarm/Sarm1, Axundead, and Highwire) that function locally in axons to drive degeneration. Here we identify a role for the Drosophila C2H2 zinc finger transcription factor Pebbled [Peb, Ras-responsive element binding protein 1 (RREB1) in mammals] in axon death. Loss of Peb in Drosophila glutamatergic sensory neurons results in either complete preservation of severed axons, or an axon death phenotype where axons fragment into large, continuous segments, rather than completely disintegrate. Peb is expressed in developing and mature sensory neurons, suggesting it is required to establish or maintain their competence to undergo axon death. peb mutant phenotypes can be rescued by human RREB1, and they exhibit dominant genetic interactions with dsarm mutants, linking peb/RREB1 to the axon death signaling cascade. Surprisingly, Peb is only able to fully block axon death signaling in glutamatergic, but not cholinergic sensory neurons, arguing for genetic diversity in axon death signaling programs in different neuronal subtypes. Our findings identify a transcription factor that regulates axon death signaling, and peb mutant phenotypes of partial fragmentation reveal a genetically accessible step in axon death signaling.
Collapse
Affiliation(s)
- Jonathan E Farley
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01655
| | - Thomas C Burdett
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01655
| | - Romina Barria
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01655
| | - Lukas J Neukomm
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01655
| | - Kevin P Kenna
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655
| | - John E Landers
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655
| | - Marc R Freeman
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01655;
| |
Collapse
|
7
|
Singh S, Dallenga T, Winkler A, Roemer S, Maruschak B, Siebert H, Brück W, Stadelmann C. Relationship of acute axonal damage, Wallerian degeneration, and clinical disability in multiple sclerosis. J Neuroinflammation 2017; 14:57. [PMID: 28302146 PMCID: PMC5356322 DOI: 10.1186/s12974-017-0831-8] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 03/06/2017] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Axonal damage and loss substantially contribute to the incremental accumulation of clinical disability in progressive multiple sclerosis. Here, we assessed the amount of Wallerian degeneration in brain tissue of multiple sclerosis patients in relation to demyelinating lesion activity and asked whether a transient blockade of Wallerian degeneration decreases axonal loss and clinical disability in a mouse model of inflammatory demyelination. METHODS Wallerian degeneration and acute axonal damage were determined immunohistochemically in the periplaque white matter of multiple sclerosis patients with early actively demyelinating lesions, chronic active lesions, and inactive lesions. Furthermore, we studied the effects of Wallerian degeneration blockage on clinical severity, inflammatory pathology, acute axonal damage, and long-term axonal loss in experimental autoimmune encephalomyelitis using Wallerian degeneration slow (Wld S ) mutant mice. RESULTS The highest numbers of axons undergoing Wallerian degeneration were found in the perilesional white matter of multiple sclerosis patients early in the disease course and with actively demyelinating lesions. Furthermore, Wallerian degeneration was more abundant in patients harboring chronic active as compared to chronic inactive lesions. No co-localization of neuropeptide Y-Y1 receptor, a bona fide immunohistochemical marker of Wallerian degeneration, with amyloid precursor protein, frequently used as an indicator of acute axonal transport disturbance, was observed in human and mouse tissue, indicating distinct axon-degenerative processes. Experimentally, a delay of Wallerian degeneration, as observed in Wld S mice, did not result in a reduction of clinical disability or acute axonal damage in experimental autoimmune encephalomyelitis, further supporting that acute axonal damage as reflected by axonal transport disturbances does not share common molecular mechanisms with Wallerian degeneration. Furthermore, delaying Wallerian degeneration did not result in a net rescue of axons in late lesion stages of experimental autoimmune encephalomyelitis. CONCLUSIONS Our data indicate that in multiple sclerosis, ongoing demyelination in focal lesions is associated with axonal degeneration in the perilesional white matter, supporting a role for focal pathology in diffuse white matter damage. Also, our results suggest that interfering with Wallerian degeneration in inflammatory demyelination does not suffice to prevent acute axonal damage and finally axonal loss.
Collapse
Affiliation(s)
- Shailender Singh
- Institute of Neuropathology, University Medical Center, Göttingen, Germany
| | - Tobias Dallenga
- Institute of Neuropathology, University Medical Center, Göttingen, Germany.,Cellular Microbiology, Research Center Borstel, Borstel, Germany
| | - Anne Winkler
- Institute of Neuropathology, University Medical Center, Göttingen, Germany
| | - Shanu Roemer
- Institute of Neuropathology, University Medical Center, Göttingen, Germany.,Department of Neurology, Rigshospitalet, Copenhagen, Denmark
| | - Brigitte Maruschak
- Institute of Neuropathology, University Medical Center, Göttingen, Germany
| | - Heike Siebert
- Institute of Neuropathology, University Medical Center, Göttingen, Germany
| | - Wolfgang Brück
- Institute of Neuropathology, University Medical Center, Göttingen, Germany
| | | |
Collapse
|
8
|
Abstract
Abstract
Axonal loss is an important process both during development and diseases of the nervous system. While the molecular mechanisms that mediate axonal loss are largely elusive, modern imaging technology affords an increasingly clear view of the cellular processes that allow nerve cells to shed individiual axon branches or even dismantle entire parts of their axonal projections. The present review discusses the characteristics of post-traumatic Wallerian degeneration, the process of axonal loss currently best understood. Subsequently, the properties of a number of recently discovered axonal loss phenomena are described. These phenomena explain some of the axonal loss that occurs locally after axon transection, during neuro-inflammatory insults, and as part of normal neurodevelopment.
Collapse
|
9
|
Bittner GD, Schallert T, Peduzzi JD. Degeneration, Trophic Interactions, and Repair of Severed Axons: A Reconsideration of Some Common Assumptions. Neuroscientist 2016. [DOI: 10.1177/107385840000600207] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We suggest that several interrelated properties of severed axons (degeneration, trophic dependencies, initial repair, and eventual repair) differ in important ways from commonly held assumptions about those properties. Specifically, (1) axotomy does not necessarily produce rapid degeneration of distal axonal segments because (2) the trophic maintenance of nerve axons does not necessarily depend entirely on proteins transported from the perikaryon—but instead axonal proteins can be trophically maintained by slowing their degradation and/or by acquiring new proteins via axonal synthesis or transfer from adjacent cells (e.g., glia). (3) The initial repair of severed distal or proximal segments occurs by barriers (seals) formed amid accumulations of vesicles and/or myelin delaminations induced by calcium influx at cut axonal ends—rather than by collapse and fusion of cut axolemmal leaflets. (4) The eventual repair of severed mammalian CNS axons does not necessarily have to occur by neuritic outgrowths, which slowly extend from cut proximal ends to possibly reestablish lost functions weeks to years after axotomy—but instead complete repair can be induced within minutes by polyethylene glycol to rejoin (fuse) the cut ends of surviving proximal and distal stumps. Strategies to repair CNS lesions based on fusion techniques combined with rehabilitative training and induced axonal outgrowth may soon provide therapies that can at least partially restore lost CNS functions.
Collapse
Affiliation(s)
- George D. Bittner
- School of Biological Sciences (Neurobiology Section) and Institute of Neuroscience, The University of Texas at Austin, Austin, Texas
| | - Timothy Schallert
- School of Biological Sciences (Neurobiology Section) and Institute of Neuroscience, Department of Pyschology, The University of Texas at Austin, Austin, Texas
| | - Jean D. Peduzzi
- School of Optometry, Department of Physiological Optics, Injury Control and Vision Science Research Centers, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
10
|
Axon degeneration: context defines distinct pathways. Curr Opin Neurobiol 2016; 39:108-15. [PMID: 27197022 DOI: 10.1016/j.conb.2016.05.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 05/02/2016] [Accepted: 05/03/2016] [Indexed: 12/29/2022]
Abstract
Axon degeneration is an essential part of development, plasticity, and injury response and has been primarily studied in mammalian models in three contexts: 1) Axotomy-induced Wallerian degeneration, 2) Apoptosis-induced axon degeneration (axon apoptosis), and 3) Axon pruning. These three contexts dictate engagement of distinct pathways for axon degeneration. Recent advances have identified the importance of SARM1, NMNATs, NAD+ depletion, and MAPK signaling in axotomy-induced Wallerian degeneration. Interestingly, apoptosis-induced axon degeneration and axon pruning have many shared mechanisms both in signaling (e.g. DLK, JNKs, GSK3α/β) and execution (e.g. Puma, Bax, caspase-9, caspase-3). However, the specific mechanisms by which caspases are activated during apoptosis versus pruning appear distinct, with apoptosis requiring Apaf-1 but not caspase-6 while pruning requires caspase-6 but not Apaf-1.
Collapse
|
11
|
DeFrancesco-Lisowitz A, Lindborg JA, Niemi JP, Zigmond RE. The neuroimmunology of degeneration and regeneration in the peripheral nervous system. Neuroscience 2015; 302:174-203. [PMID: 25242643 PMCID: PMC4366367 DOI: 10.1016/j.neuroscience.2014.09.027] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 09/08/2014] [Accepted: 09/10/2014] [Indexed: 12/25/2022]
Abstract
Peripheral nerves regenerate following injury due to the effective activation of the intrinsic growth capacity of the neurons and the formation of a permissive pathway for outgrowth due to Wallerian degeneration (WD). WD and subsequent regeneration are significantly influenced by various immune cells and the cytokines they secrete. Although macrophages have long been known to play a vital role in the degenerative process, recent work has pointed to their importance in influencing the regenerative capacity of peripheral neurons. In this review, we focus on the various immune cells, cytokines, and chemokines that make regeneration possible in the peripheral nervous system, with specific attention placed on the role macrophages play in this process.
Collapse
Affiliation(s)
| | - J A Lindborg
- Department of Neurosciences, Case Western Reserve University, Cleveland OH 44106-4975
| | - J P Niemi
- Department of Neurosciences, Case Western Reserve University, Cleveland OH 44106-4975
| | - R E Zigmond
- Department of Neurosciences, Case Western Reserve University, Cleveland OH 44106-4975
| |
Collapse
|
12
|
Ryu J, Horkayne-Szakaly I, Xu L, Pletnikova O, Leri F, Eberhart C, Troncoso JC, Koliatsos VE. The problem of axonal injury in the brains of veterans with histories of blast exposure. Acta Neuropathol Commun 2014; 2:153. [PMID: 25422066 PMCID: PMC4260204 DOI: 10.1186/s40478-014-0153-3] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Accepted: 10/11/2014] [Indexed: 11/18/2022] Open
Abstract
Introduction Blast injury to brain, a hundred-year old problem with poorly characterized neuropathology, has resurfaced as health concern in recent deployments in Iraq and Afghanistan. To characterize the neuropathology of blast injury, we examined the brains of veterans for the presence of amyloid precursor protein (APP)-positive axonal swellings typical of diffuse axonal injury (DAI) and compared them to healthy controls as well as controls with opiate overdose, anoxic-ischemic encephalopathy, and non-blast TBI (falls and motor vehicle crashes). Results In cases with blast history, we found APP (+) axonal abnormalities in several brain sites, especially the medial dorsal frontal white matter. In white matter, these abnormalities were featured primarily by clusters of axonal spheroids or varicosities in a honeycomb pattern with perivascular distribution. Axonal abnormalities colocalized with IBA1 (+) reactive microglia and had an appearance that was distinct from classical DAI encountered in TBI due to motor vehicle crashes. Opiate overdose cases also showed APP (+) axonal abnormalities, but the intensity of these lesions was lower compared to cases with blast histories and there was no clear association of such lesions with microglial activation. Conclusions Our findings demonstrate that many cases with history of blast exposure are featured by APP (+) axonopathy that may be related to blast exposure, but an important role for opiate overdose, antemortem anoxia, and concurrent blunt TBI events in war theater or elsewhere cannot be discounted. Electronic supplementary material The online version of this article (doi:10.1186/s40478-014-0153-3) contains supplementary material, which is available to authorized users.
Collapse
|
13
|
Collyer E, Catenaccio A, Lemaitre D, Diaz P, Valenzuela V, Bronfman F, Court FA. Sprouting of axonal collaterals after spinal cord injury is prevented by delayed axonal degeneration. Exp Neurol 2014; 261:451-61. [PMID: 25079366 DOI: 10.1016/j.expneurol.2014.07.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 07/02/2014] [Accepted: 07/20/2014] [Indexed: 01/24/2023]
Abstract
After an incomplete spinal cord injury (SCI), partial recovery of locomotion is accomplished with time. Previous studies have established a functional link between extension of axon collaterals from spared spinal tracts and locomotor recovery after SCI, but the tissular signals triggering collateral sprouting have not been identified. Here, we investigated whether axonal degeneration after SCI contributes to the sprouting of collaterals from axons spared after injury. To this end, we evaluated collateral sprouting from BDA-labeled uninjured corticospinal axons after spinal cord hemisection (SCI(H)) in wild type (WT) mouse and Wld(S) mouse strains, which shows a significant delay in Wallerian degeneration after injury. After SCI(H), spared fibers of WT mice extend collateral sprouts to both intact and denervated sides of the spinal cord distant from the injury site. On the contrary, in the Wld(S) mice collateral sprouting from spared fibers was greatly reduced after SCI(H). Consistent with a role for collateral sprouting in functional recovery after SCI, locomotor recovery after SCI(H) was impaired in Wld(S) mice compared to WT animals. In conclusion, our results identify axonal degeneration as one of the triggers for collateral sprouting from the contralesional uninjured fibers after an SCI(H). These results open the path for identifying molecular signals associated with tissular changes after SCI that promotes collateral sprouting and functional recovery.
Collapse
Affiliation(s)
- E Collyer
- Millenium Nucleus for Regenerative Biology, Faculty of Biology, Pontificia Universidad Catolica de Chile, Santiago, Chile.
| | - A Catenaccio
- Millenium Nucleus for Regenerative Biology, Faculty of Biology, Pontificia Universidad Catolica de Chile, Santiago, Chile.
| | - D Lemaitre
- Millenium Nucleus for Regenerative Biology, Faculty of Biology, Pontificia Universidad Catolica de Chile, Santiago, Chile.
| | - P Diaz
- Millenium Nucleus for Regenerative Biology, Faculty of Biology, Pontificia Universidad Catolica de Chile, Santiago, Chile.
| | - V Valenzuela
- Millenium Nucleus for Regenerative Biology, Faculty of Biology, Pontificia Universidad Catolica de Chile, Santiago, Chile.
| | - F Bronfman
- Millenium Nucleus for Regenerative Biology, Faculty of Biology, Pontificia Universidad Catolica de Chile, Santiago, Chile.
| | - F A Court
- Millenium Nucleus for Regenerative Biology, Faculty of Biology, Pontificia Universidad Catolica de Chile, Santiago, Chile; Neurounion Biomedical Foundation, Santiago, Chile.
| |
Collapse
|
14
|
Zhu S, Yang Y, Hu J, Qian L, Jiang Y, Li X, Yang Q, Bai H, Chen Q. Wld(S) ameliorates renal injury in a type 1 diabetic mouse model. Am J Physiol Renal Physiol 2014; 306:F1348-56. [PMID: 24598800 DOI: 10.1152/ajprenal.00418.2013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Diabetic nephropathy (DN) is the leading cause of end-stage kidney disease worldwide. The purpose of this study is to investigate whether the Wld(S) (slow Wallerian degeneration; also known as Wld) gene plays a renoprotective role during the progression of DN. Diabetes was induced in 8-wk-old male wild-type (WT) and C57BL/Wld(S) mice by streptozotocin (STZ) injection. Blood and urinary variables including blood glucose, glycated hemoglobin (GHb), insulin, urea nitrogen, and albumin/creatinine ratio were assessed 4, 7, and 14 wk after STZ injection. Periodic acid-Schiff staining, Masson staining, and silver staining were performed for renal pathological analyses. In addition, the renal ultrastructure was observed by electron microscope. The activities of p38 and ERK signaling in renal cortical tissues were evaluated by Western blotting. NAD(+)/NADH ratio and NADPH oxidase activity were also measured. Moreover, the expressions of TNF-α, IL-1, and IL-6 were examined. We provide experimental evidence demonstrating that the Wld(S) gene is expressed in kidney cells and protects against the early stage of diabetes-induced renal dysfunction and extracellular matrix accumulation through delaying the reduction of the NAD(+)/NADH ratio, inhibiting the activation of p38 and ERK signaling, and suppressing oxidative stress as evidenced by the decreased NADPH oxidase activity and lower expression of TNF-α, IL-1, and IL-6.
Collapse
Affiliation(s)
- Shuaishuai Zhu
- Atherosclerosis Research Centre, Nanjing Medical University, Nanjing, People's Republic of China
| | - Yelin Yang
- Atherosclerosis Research Centre, Nanjing Medical University, Nanjing, People's Republic of China
| | - Jin Hu
- Atherosclerosis Research Centre, Nanjing Medical University, Nanjing, People's Republic of China
| | - Lingling Qian
- Atherosclerosis Research Centre, Nanjing Medical University, Nanjing, People's Republic of China
| | - Yuchen Jiang
- Atherosclerosis Research Centre, Nanjing Medical University, Nanjing, People's Republic of China
| | - Xiaoyu Li
- Atherosclerosis Research Centre, Nanjing Medical University, Nanjing, People's Republic of China
| | - Qing Yang
- Atherosclerosis Research Centre, Nanjing Medical University, Nanjing, People's Republic of China
| | - Hui Bai
- Atherosclerosis Research Centre, Nanjing Medical University, Nanjing, People's Republic of China
| | - Qi Chen
- Atherosclerosis Research Centre, Nanjing Medical University, Nanjing, People's Republic of China
| |
Collapse
|
15
|
Beirowski B. Concepts for regulation of axon integrity by enwrapping glia. Front Cell Neurosci 2013; 7:256. [PMID: 24391540 PMCID: PMC3867696 DOI: 10.3389/fncel.2013.00256] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 11/25/2013] [Indexed: 12/16/2022] Open
Abstract
Long axons and their enwrapping glia (EG; Schwann cells (SCs) and oligodendrocytes (OLGs)) form a unique compound structure that serves as conduit for transport of electric and chemical information in the nervous system. The peculiar cytoarchitecture over an enormous length as well as its substantial energetic requirements make this conduit particularly susceptible to detrimental alterations. Degeneration of long axons independent of neuronal cell bodies is observed comparatively early in a range of neurodegenerative conditions as a consequence of abnormalities in SCs and OLGs . This leads to the most relevant disease symptoms and highlights the critical role that these glia have for axon integrity, but the underlying mechanisms remain elusive. The quest to understand why and how axons degenerate is now a crucial frontier in disease-oriented research. This challenge is most likely to lead to significant progress if the inextricable link between axons and their flanking glia in pathological situations is recognized. In this review I compile recent advances in our understanding of the molecular programs governing axon degeneration, and mechanisms of EG’s non-cell autonomous impact on axon-integrity. A particular focus is placed on emerging evidence suggesting that EG nurture long axons by virtue of their intimate association, release of trophic substances, and neurometabolic coupling. The correction of defects in these functions has the potential to stabilize axons in a variety of neuronal diseases in the peripheral nervous system and central nervous system (PNS and CNS).
Collapse
Affiliation(s)
- Bogdan Beirowski
- Department of Genetics, Washington University School of Medicine Saint Louis, MO, USA
| |
Collapse
|
16
|
Abstract
Rodent models of nerve injury have increased our understanding of peripheral nerve regeneration, but clinical applications have been scarce, partly because such models do not adequately recapitulate the situation in humans. In human injuries, axons are often required to extend over much longer distances than in mice, and injury leaves distal nerve fibres and target tissues without axonal contact for extended amounts of time. Distal Schwann cells undergo atrophy owing to the lack of contact with proximal neurons, which results in reduced expression of neurotrophic growth factors, changes in the extracellular matrix and loss of Schwann cell basal lamina, all of which hamper axonal extension. Furthermore, atrophy and denervation-related changes in target tissues make good functional recovery difficult to achieve even when axons regenerate all the way to the target tissue. To improve functional outcomes in humans, strategies to increase the speed of axonal growth, maintain Schwann cells in a healthy, repair-capable state and keep target tissues receptive to reinnervation are needed. Use of rodent models of chronic denervation will facilitate our understanding of the molecular mechanisms of peripheral nerve regeneration and create the potential to test therapeutic advances.
Collapse
|
17
|
Howell GR, Soto I, Libby RT, John SWM. Intrinsic axonal degeneration pathways are critical for glaucomatous damage. Exp Neurol 2013; 246:54-61. [PMID: 22285251 PMCID: PMC3831512 DOI: 10.1016/j.expneurol.2012.01.014] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Revised: 11/15/2011] [Accepted: 01/10/2012] [Indexed: 12/13/2022]
Abstract
Glaucoma is a neurodegenerative disease affecting 70million people worldwide. For some time, analysis of human glaucoma and animal models suggested that RGC axonal injury in the optic nerve head (where RGC axons exit the eye) is an important early event in glaucomatous neurodegeneration. During the last decade advances in molecular biology and genome manipulation have allowed this hypothesis to be tested more critically, at least in animal models. Data indicate that RGC axon degeneration precedes soma death. Preventing soma death using mouse models that are mutant for BAX, a proapoptotic gene, is not sufficient to prevent the degeneration of RGC axons. This indicates that different degeneration processes occur in different compartments of the RGC during glaucoma. Furthermore, the Wallerian degeneration slow allele (Wld(s)) slows or prevents RGC axon degeneration in rodent models of glaucoma. These experiments and many others, now strongly support the hypothesis that axon degeneration is a critical pathological event in glaucomatous neurodegeneration. However, the events that lead from a glaucomatous insult (e.g. elevated intraocular pressure) to axon damage in glaucoma are not well defined. For developing new therapies, it will be necessary to clearly define and order the molecular events that lead from glaucomatous insults to axon degeneration.
Collapse
Affiliation(s)
- Gareth R Howell
- The Howard Hughes Medical Institute, The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA.
| | | | | | | |
Collapse
|
18
|
Axonal degeneration in the peripheral nervous system: Implications for the pathogenesis of amyotrophic lateral sclerosis. Exp Neurol 2013; 246:6-13. [DOI: 10.1016/j.expneurol.2013.05.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Revised: 04/22/2013] [Accepted: 05/02/2013] [Indexed: 12/13/2022]
|
19
|
Spatiotemporal resolution of BDNF neuroprotection against glutamate excitotoxicity in cultured hippocampal neurons. Neuroscience 2013; 237:66-86. [PMID: 23384605 DOI: 10.1016/j.neuroscience.2013.01.054] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 01/28/2013] [Indexed: 02/02/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) protects hippocampal neurons from glutamate excitotoxicity as determined by analysis of chromatin condensation, through activation of extracellular signal-regulated kinase (ERK) and phosphatidylinositol 3-kinase (PI3-K) signaling pathways. However, it is still unknown whether BDNF also prevents the degeneration of axons and dendrites, and the functional demise of synapses, which would be required to preserve neuronal activity. Herein, we have studied the time-dependent changes in several neurobiological markers, and the regulation of proteolytic mechanisms in cultured rat hippocampal neurons, through quantitative western blot and immunocytochemistry. Calpain activation peaked immediately after the neurodegenerative input, followed by a transient increase in ubiquitin-conjugated proteins and increased abundance of cleaved-caspase-3. Proteasome and calpain inhibition did not reproduce the protective effect of BDNF and caspase inhibition in preventing chromatin condensation. However, proteasome and calpain inhibition did protect the neuronal markers for dendrites (MAP-2), axons (Neurofilament-H) and the vesicular glutamate transporters (VGLUT1-2), whereas caspase inhibition was unable to mimic the protective effect of BDNF on neurites and synaptic markers. BDNF partially prevented the downregulation of synaptic activity measured by the KCl-evoked glutamate release using a Förster (Fluorescence) resonance energy transfer (FRET) glutamate nanosensor. These results translate a time-dependent activation of proteases and spatial segregation of these mechanisms, where calpain activation is followed by proteasome deregulation, from neuronal processes to the soma, and finally by caspase activation in the cell body. Moreover, PI3-K and PLCγ small molecule inhibitors significantly blocked the protective action of BDNF, suggesting an activity-dependent mechanism of neuroprotection. Ultimately, we hypothesize that neuronal repair after a degenerative insult is initiated at the synaptic level.
Collapse
|
20
|
Zhu Y, Zhang L, Sasaki Y, Milbrandt J, Gidday JM. Protection of mouse retinal ganglion cell axons and soma from glaucomatous and ischemic injury by cytoplasmic overexpression of Nmnat1. Invest Ophthalmol Vis Sci 2013; 54:25-36. [PMID: 23211826 DOI: 10.1167/iovs.12-10861] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
PURPOSE The Wlds mutation affords protection of retinal ganglion cell (RGC) axons in retinal ischemia and in inducible and hereditary preclinical models of glaucoma. We undertook the present study to determine whether the Nmnat1 portion of the chimeric protein provides axonal and somatic protection of RGCs in models of ischemia and glaucoma, particularly when localized to nonnuclear regions of the cell. METHODS The survival and integrity of RGC axons and soma from transgenic mice with confirmed cytoplasmic overexpression of Nmnat1 in retina and optic nerve (cytNmnat1-Tg mice) were examined in the retina and postlaminar optic nerve 4 days following acute retinal ischemia, and 3 weeks following the chronic elevation of intraocular pressure. RESULTS Ischemia- and glaucoma-induced disruptions of proximal segments of RGC axons that comprise the nerve fiber layer in wild-type mice were both robustly abrogated in cytNmnat1-Tg mice. More distal portions of RGC axons within the optic nerve were also protected from glaucomatous disruption in the transgenic mice. In both disease models, Nmnat1 overexpression in extranuclear locations significantly enhanced the survival of RGC soma. CONCLUSIONS Overexpression of Nmnat1 in the cytoplasm and axons of RGCs robustly protected against both ischemic and glaucomatous loss of RGC axonal integrity, as well as loss of RGC soma. These findings reflect the more pan-cellular protection of CNS neurons that is realized by cytoplasmic Nmnat1 expression, and thus provide a therapeutic strategy for protecting against retinal neurodegeneration, and perhaps other CNS neurodegenerative diseases as well.
Collapse
Affiliation(s)
- Yanli Zhu
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | |
Collapse
|
21
|
Abstract
The fundamental roles of Schwann cells during peripheral nerve formation and regeneration have been recognized for more than 100 years, but the cellular and molecular mechanisms that integrate Schwann cell and axonal functions continue to be elucidated. Derived from the embryonic neural crest, Schwann cells differentiate into myelinating cells or bundle multiple unmyelinated axons into Remak fibers. Axons dictate which differentiation path Schwann cells follow, and recent studies have established that axonal neuregulin1 signaling via ErbB2/B3 receptors on Schwann cells is essential for Schwann cell myelination. Extracellular matrix production and interactions mediated by specific integrin and dystroglycan complexes are also critical requisites for Schwann cell-axon interactions. Myelination entails expansion and specialization of the Schwann cell plasma membrane over millimeter distances. Many of the myelin-specific proteins have been identified, and transgenic manipulation of myelin genes have provided novel insights into myelin protein function, including maintenance of axonal integrity and survival. Cellular events that facilitate myelination, including microtubule-based protein and mRNA targeting, and actin based locomotion, have also begun to be understood. Arguably, the most remarkable facet of Schwann cell biology, however, is their vigorous response to axonal damage. Degradation of myelin, dedifferentiation, division, production of axonotrophic factors, and remyelination all underpin the substantial regenerative capacity of the Schwann cells and peripheral nerves. Many of these properties are not shared by CNS fibers, which are myelinated by oligodendrocytes. Dissecting the molecular mechanisms responsible for the complex biology of Schwann cells continues to have practical benefits in identifying novel therapeutic targets not only for Schwann cell-specific diseases but other disorders in which axons degenerate.
Collapse
Affiliation(s)
- Grahame J Kidd
- Department of Neuroscience, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.
| | | | | |
Collapse
|
22
|
Zeinab RA, Wu H, Sergi C, Leng R. UBE4B: a promising regulatory molecule in neuronal death and survival. Int J Mol Sci 2012; 13:16865-79. [PMID: 23222733 PMCID: PMC3546727 DOI: 10.3390/ijms131216865] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2012] [Revised: 11/22/2012] [Accepted: 11/27/2012] [Indexed: 12/13/2022] Open
Abstract
Neuronal survival and death of neurons are considered a fundamental mechanism in the regulation of the nervous system during early development of the system and in adulthood. Defects in this mechanism are highly problematic and are associated with many neurodegenerative diseases. Because neuronal programmed death is apoptotic in nature, indicating that apoptosis is a key regulatory process, the p53 family members (p53, p73, p63) act as checkpoints in neurons due to their role in apoptosis. The complexity of this system is due to the existence of different naturally occurring isoforms that have different functions from the wild types (WT), varying from apoptotic to anti-apoptotic effects. In this review, we focus on the role of UBE4B (known as Ube4b or Ufd2a in mouse), an E3/E4 ligase that triggers substrate polyubiquitination, as a master regulatory ligase associated with the p53 family WT proteins and isoforms in regulating neuronal survival. UBE4B is also associated with other pathways independent of the p53 family, such as polyglutamine aggregation and Wallerian degeneration, both of which are critical in neurodegenerative diseases. Many of the hypotheses presented here are gateways to understanding the programmed death/survival of neurons regulated by UBE4B in normal physiology, and a means of introducing potential therapeutic approaches with implications in treating several neurodegenerative diseases.
Collapse
Affiliation(s)
- Rami Abou Zeinab
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2S2, Canada.
| | | | | | | |
Collapse
|
23
|
Hicks AN, Lorenzetti D, Gilley J, Lu B, Andersson KE, Miligan C, Overbeek PA, Oppenheim R, Bishop CE. Nicotinamide mononucleotide adenylyltransferase 2 (Nmnat2) regulates axon integrity in the mouse embryo. PLoS One 2012; 7:e47869. [PMID: 23082226 PMCID: PMC3474723 DOI: 10.1371/journal.pone.0047869] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Accepted: 09/24/2012] [Indexed: 12/04/2022] Open
Abstract
Using transposon-mediated gene-trap mutagenesis, we have generated a novel mouse mutant termed Blad (Bloated Bladder). Homozygous mutant mice die perinatally showing a greatly distended bladder, underdeveloped diaphragm and a reduction in total skeletal muscle mass. Wild type and heterozygote mice appear normal. Using PCR, we identified a transposon insertion site in the first intron of Nmnat2 (Nicotinamide mononucleotide adenyltransferase 2). Nmnat2 is expressed predominantly in the brain and nervous system and has been linked to the survival of axons. Expression of this gene is undetectable in Nmnat2blad/blad mutants. Examination of the brains of E18.5 Nmnat2blad/blad mutant embryos did not reveal any obvious morphological changes. In contrast, E18.5 Nmnat2blad/blad homozygotes showed an approximate 60% reduction of spinal motoneurons in the lumbar region and a more than 80% reduction in the sensory neurons of the dorsal root ganglion (DRG). In addition, facial motoneuron numbers were severely reduced, and there was virtually a complete absence of axons in the hind limb. Our observations suggest that during embryogenesis, Nmnat2 plays an important role in axonal growth or maintenance. It appears that in the absence of Nmnat2, major target organs and tissues (e.g., muscle) are not functionally innervated resulting in perinatal lethality. In addition, neither Nmnat1 nor 3 can compensate for the loss of Nmnat2. Whilst there have been recent suggestions that Nmnat2 may be an endogenous modulator of axon integrity, this work represents the first in vivo study demonstrating that Nmnat2 is involved in axon development or survival in a mammal.
Collapse
Affiliation(s)
- Amy N Hicks
- Wake Forest Institute for Regenerative Medicine, Wake Forest University, Winston Salem, North Carolina, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Chen M, Maloney JA, Kallop DY, Atwal JK, Tam SJ, Baer K, Kissel H, Kaminker JS, Lewcock JW, Weimer RM, Watts RJ. Spatially coordinated kinase signaling regulates local axon degeneration. J Neurosci 2012; 32:13439-53. [PMID: 23015435 PMCID: PMC6621382 DOI: 10.1523/jneurosci.2039-12.2012] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Revised: 07/06/2012] [Accepted: 07/28/2012] [Indexed: 01/02/2023] Open
Abstract
In addition to being a hallmark of neurodegenerative disease, axon degeneration is used during development of the nervous system to prune unwanted connections. In development, axon degeneration is tightly regulated both temporally and spatially. Here, we provide evidence that degeneration cues are transduced through various kinase pathways functioning in spatially distinct compartments to regulate axon degeneration. Intriguingly, glycogen synthase kinase-3 (GSK3) acts centrally, likely modulating gene expression in the cell body to regulate distally restricted axon degeneration. Through a combination of genetic and pharmacological manipulations, including the generation of an analog-sensitive kinase allele mutant mouse for GSK3β, we show that the β isoform of GSK3, not the α isoform, is essential for developmental axon pruning in vitro and in vivo. Additionally, we identify the dleu2/mir15a/16-1 cluster, previously characterized as a regulator of B-cell proliferation, and the transcription factor tbx6, as likely downstream effectors of GSK3β in axon degeneration.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Axons/metabolism
- Cells, Cultured
- Electroporation
- Embryo, Mammalian
- Enzyme Inhibitors/pharmacology
- Female
- Ganglia, Spinal/cytology
- Gene Expression Profiling/methods
- Gene Expression Regulation, Developmental/drug effects
- Gene Expression Regulation, Developmental/genetics
- Gene Expression Regulation, Developmental/physiology
- Genotype
- Glycogen Synthase Kinase 3/genetics
- Glycogen Synthase Kinase 3/metabolism
- Glycogen Synthase Kinase 3 beta
- Green Fluorescent Proteins/genetics
- Hippocampus/cytology
- Humans
- Immunoprecipitation
- Luminescent Proteins/genetics
- Luminescent Proteins/metabolism
- MAP Kinase Signaling System/drug effects
- MAP Kinase Signaling System/genetics
- MAP Kinase Signaling System/physiology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Mutation/genetics
- Nerve Degeneration/drug therapy
- Nerve Degeneration/enzymology
- Nerve Degeneration/pathology
- Nerve Degeneration/prevention & control
- Nerve Growth Factor/deficiency
- Nerve Tissue Proteins/metabolism
- Neurons/pathology
- Oligonucleotide Array Sequence Analysis
- Organ Culture Techniques
- Phosphorylation/physiology
- Phosphotransferases/metabolism
- RNA, Small Interfering/administration & dosage
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Retinal Ganglion Cells/metabolism
- Signal Transduction/drug effects
- Signal Transduction/physiology
- Transfection
- Red Fluorescent Protein
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Joshua S. Kaminker
- Bioinformatics, Genentech, Inc., South San Francisco, California 94080, and
| | | | | | | |
Collapse
|
25
|
Fang Y, Bonini NM. Axon degeneration and regeneration: insights from Drosophila models of nerve injury. Annu Rev Cell Dev Biol 2012; 28:575-97. [PMID: 22831639 DOI: 10.1146/annurev-cellbio-101011-155836] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Axon degeneration is the pivotal pathological event of acute traumatic neural injury as well as many chronic neurodegenerative diseases. It is an active cellular program and yet molecularly distinct from cell death. Much effort is devoted toward understanding the nature of axon degeneration and promoting axon regeneration. However, the fundamental mechanisms of self-destruction of damaged axons remain unclear, and there are still few treatments for traumatic brain injury (TBI) or spinal cord injury (SCI). Genetically approachable model organisms such as Drosophila melanogaster, the fruit fly, have proven exceptionally successful in modeling human neurodegenerative diseases. More recently, this success has been extended into the field of acute axon injury and regeneration. In this review, we discuss recent findings, focusing on how these models hold promise for accelerating mechanistic insight into axon injury and identifying potential therapeutic targets for TBI and SCI.
Collapse
Affiliation(s)
- Yanshan Fang
- Howard Hughes Medical Institute and Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA.
| | | |
Collapse
|
26
|
Osterloh JM, Yang J, Rooney TM, Fox AN, Adalbert R, Powell EH, Sheehan AE, Avery MA, Hackett R, Logan MA, MacDonald JM, Ziegenfuss JS, Milde S, Hou YJ, Nathan C, Ding A, Brown RH, Conforti L, Coleman M, Tessier-Lavigne M, Züchner S, Freeman MR. dSarm/Sarm1 is required for activation of an injury-induced axon death pathway. Science 2012; 337:481-4. [PMID: 22678360 DOI: 10.1126/science.1223899] [Citation(s) in RCA: 492] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Axonal and synaptic degeneration is a hallmark of peripheral neuropathy, brain injury, and neurodegenerative disease. Axonal degeneration has been proposed to be mediated by an active autodestruction program, akin to apoptotic cell death; however, loss-of-function mutations capable of potently blocking axon self-destruction have not been described. Here, we show that loss of the Drosophila Toll receptor adaptor dSarm (sterile α/Armadillo/Toll-Interleukin receptor homology domain protein) cell-autonomously suppresses Wallerian degeneration for weeks after axotomy. Severed mouse Sarm1 null axons exhibit remarkable long-term survival both in vivo and in vitro, indicating that Sarm1 prodegenerative signaling is conserved in mammals. Our results provide direct evidence that axons actively promote their own destruction after injury and identify dSarm/Sarm1 as a member of an ancient axon death signaling pathway.
Collapse
Affiliation(s)
- Jeannette M Osterloh
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Avery MA, Rooney TM, Pandya JD, Wishart TM, Gillingwater TH, Geddes JW, Sullivan P, Freeman MR. WldS prevents axon degeneration through increased mitochondrial flux and enhanced mitochondrial Ca2+ buffering. Curr Biol 2012; 22:596-600. [PMID: 22425157 PMCID: PMC4175988 DOI: 10.1016/j.cub.2012.02.043] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Revised: 12/29/2011] [Accepted: 02/22/2012] [Indexed: 01/09/2023]
Abstract
Wld(S) (slow Wallerian degeneration) is a remarkable protein that can suppress Wallerian degeneration of axons and synapses, but how it exerts this effect remains unclear. Here, using Drosophila and mouse models, we identify mitochondria as a key site of action for Wld(S) neuroprotective function. Targeting the NAD(+) biosynthetic enzyme Nmnat to mitochondria was sufficient to fully phenocopy Wld(S), and Wld(S) was specifically localized to mitochondria in synaptic preparations from mouse brain. Axotomy of live wild-type axons induced a dramatic spike in axoplasmic Ca(2+) and termination of mitochondrial movement-Wld(S) potently suppressed both of these events. Surprisingly, Wld(S) also promoted increased basal mitochondrial motility in axons before injury, and genetically suppressing mitochondrial motility in vivo dramatically reduced the protective effect of Wld(S). Intriguingly, purified mitochondria from Wld(S) mice exhibited enhanced Ca(2+) buffering capacity. We propose that the enhanced Ca(2+) buffering capacity of Wld(S+) mitochondria leads to increased mitochondrial motility, suppression of axotomy-induced Ca(2+) elevation in axons, and thereby suppression of Wallerian degeneration.
Collapse
Affiliation(s)
- Michelle A. Avery
- Department of Neurobiology, University of Massachusetts Medical School
| | - Timothy M. Rooney
- Department of Neurobiology, University of Massachusetts Medical School
| | - Jignesh D. Pandya
- Spinal Cord and Brain Injury Research Center, Department of Anatomy and Neurobiology, University of Kentucky, Lexington, KY 40536-0509, USA
| | - Thomas M. Wishart
- Euan MacDonald Centre for Motor Neurone Disease Research & Centre for Integrative Physiology, University of Edinburgh, Edinburgh, EH8 9XD, United Kingdom
| | - Thomas H. Gillingwater
- Euan MacDonald Centre for Motor Neurone Disease Research & Centre for Integrative Physiology, University of Edinburgh, Edinburgh, EH8 9XD, United Kingdom
| | - James W. Geddes
- Spinal Cord and Brain Injury Research Center, Department of Anatomy and Neurobiology, University of Kentucky, Lexington, KY 40536-0509, USA
| | - Patrick Sullivan
- Spinal Cord and Brain Injury Research Center, Department of Anatomy and Neurobiology, University of Kentucky, Lexington, KY 40536-0509, USA
| | - Marc R. Freeman
- Department of Neurobiology, University of Massachusetts Medical School
| |
Collapse
|
28
|
Wang JT, Medress ZA, Barres BA. Axon degeneration: molecular mechanisms of a self-destruction pathway. ACTA ACUST UNITED AC 2012; 196:7-18. [PMID: 22232700 PMCID: PMC3255986 DOI: 10.1083/jcb.201108111] [Citation(s) in RCA: 318] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Axon degeneration is a characteristic event in many neurodegenerative conditions including stroke, glaucoma, and motor neuropathies. However, the molecular pathways that regulate this process remain unclear. Axon loss in chronic neurodegenerative diseases share many morphological features with those in acute injuries, and expression of the Wallerian degeneration slow (WldS) transgene delays nerve degeneration in both events, indicating a common mechanism of axonal self-destruction in traumatic injuries and degenerative diseases. A proposed model of axon degeneration is that nerve insults lead to impaired delivery or expression of a local axonal survival factor, which results in increased intra-axonal calcium levels and calcium-dependent cytoskeletal breakdown.
Collapse
Affiliation(s)
- Jack T Wang
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | | | | |
Collapse
|
29
|
Lingor P, Koch JC, Tönges L, Bähr M. Axonal degeneration as a therapeutic target in the CNS. Cell Tissue Res 2012; 349:289-311. [PMID: 22392734 PMCID: PMC3375418 DOI: 10.1007/s00441-012-1362-3] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Accepted: 02/02/2012] [Indexed: 12/15/2022]
Abstract
Degeneration of the axon is an important step in the pathomechanism of traumatic, inflammatory and degenerative neurological diseases. Increasing evidence suggests that axonal degeneration occurs early in the course of these diseases and therefore represents a promising target for future therapeutic strategies. We review the evidence for axonal destruction from pathological findings and animal models with particular emphasis on neurodegenerative and neurotraumatic disorders. We discuss the basic morphological and temporal modalities of axonal degeneration (acute, chronic and focal axonal degeneration and Wallerian degeneration). Based on the mechanistic concepts, we then delineate in detail the major molecular mechanisms that underlie the degenerative cascade, such as calcium influx, axonal transport, protein aggregation and autophagy. We finally concentrate on putative therapeutic targets based on the mechanistic prerequisites.
Collapse
Affiliation(s)
- Paul Lingor
- Department of Neurology, University Medicine Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany.
| | | | | | | |
Collapse
|
30
|
Wilkinson AE, McCormick AM, Leipzig ND. Central Nervous System Tissue Engineering: Current Considerations and Strategies. ACTA ACUST UNITED AC 2011. [DOI: 10.2200/s00390ed1v01y201111tis008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
31
|
Zhu SS, Ren Y, Zhang M, Cao JQ, Yang Q, Li XY, Bai H, Jiang L, Jiang Q, He ZG, Chen Q. Wld(S) protects against peripheral neuropathy and retinopathy in an experimental model of diabetes in mice. Diabetologia 2011; 54:2440-50. [PMID: 21739347 DOI: 10.1007/s00125-011-2226-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2011] [Accepted: 05/31/2011] [Indexed: 10/18/2022]
Abstract
AIMS/HYPOTHESIS We aimed to evaluate the effect of the mutant Wld(S) (slow Wallerian degeneration; also known as Wld) gene in experimental diabetes on early experimental peripheral diabetic neuropathy and diabetic retinopathy. METHODS The experiments were performed in four groups of mice: wild-type (WT), streptozotocin (STZ)-induced diabetic WT, C57BL/Wld(S) and STZ-induced diabetic C57BL/Wld(S). In each group, intraperitoneal glucose and insulin tolerance tests were performed; blood glucose, glycated haemoglobin and serum insulin were monitored. These mice were also subjected to the following behavioural tests: grasping test, hot-plate test and von Frey aesthesiometer test. For some animals, sciatic-tibial motor nerve conduction velocity, tail sensory nerve conduction velocity and eye pattern electroretinogram were measured. At the end of the experiments, islets were isolated to detect glucose-stimulated insulin secretion, ATP content and extent of apoptosis. The NAD/NADH ratio in islets and retinas was evaluated. Surviving retinal ganglion cells were estimated by immunohistochemistry. RESULTS We found that the Wld(S) gene is expressed in islets and protects beta cells against multiple low doses of STZ by increasing the NAD/NADH ratio, maintaining the ATP concentration, and reducing apoptosis. Consistently, significantly higher insulin concentrations, lower blood glucose concentrations, and better glucose tolerance were observed in Wld(S) mice compared with WT mice after STZ treatment. Furthermore, Wld(S) alleviated abnormal sensory responses, nerve conduction, retina dysfunction and reduction of surviving retinal ganglion cells in STZ-induced diabetic models. CONCLUSIONS/INTERPRETATION We provide the first evidence that expression of the Wld(S) gene decreases beta cell destruction and preserves islet function in STZ-induced diabetes, thus revealing a novel protective strategy for diabetic models.
Collapse
Affiliation(s)
- S S Zhu
- Atherosclerosis Research Centre, Nanjing Medical University, 140 Hanzhong Road, Nanjing 210029, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Gaudet AD, Popovich PG, Ramer MS. Wallerian degeneration: gaining perspective on inflammatory events after peripheral nerve injury. J Neuroinflammation 2011; 8:110. [PMID: 21878126 PMCID: PMC3180276 DOI: 10.1186/1742-2094-8-110] [Citation(s) in RCA: 606] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Accepted: 08/30/2011] [Indexed: 01/15/2023] Open
Abstract
In this review, we first provide a brief historical perspective, discussing how peripheral nerve injury (PNI) may have caused World War I. We then consider the initiation, progression, and resolution of the cellular inflammatory response after PNI, before comparing the PNI inflammatory response with that induced by spinal cord injury (SCI).In contrast with central nervous system (CNS) axons, those in the periphery have the remarkable ability to regenerate after injury. Nevertheless, peripheral nervous system (PNS) axon regrowth is hampered by nerve gaps created by injury. In addition, the growth-supportive milieu of PNS axons is not sustained over time, precluding long-distance regeneration. Therefore, studying PNI could be instructive for both improving PNS regeneration and recovery after CNS injury. In addition to requiring a robust regenerative response from the injured neuron itself, successful axon regeneration is dependent on the coordinated efforts of non-neuronal cells which release extracellular matrix molecules, cytokines, and growth factors that support axon regrowth. The inflammatory response is initiated by axonal disintegration in the distal nerve stump: this causes blood-nerve barrier permeabilization and activates nearby Schwann cells and resident macrophages via receptors sensitive to tissue damage. Denervated Schwann cells respond to injury by shedding myelin, proliferating, phagocytosing debris, and releasing cytokines that recruit blood-borne monocytes/macrophages. Macrophages take over the bulk of phagocytosis within days of PNI, before exiting the nerve by the circulation once remyelination has occurred. The efficacy of the PNS inflammatory response (although transient) stands in stark contrast with that of the CNS, where the response of nearby cells is associated with inhibitory scar formation, quiescence, and degeneration/apoptosis. Rather than efficiently removing debris before resolving the inflammatory response as in other tissues, macrophages infiltrating the CNS exacerbate cell death and damage by releasing toxic pro-inflammatory mediators over an extended period of time. Future research will help determine how to manipulate PNS and CNS inflammatory responses in order to improve tissue repair and functional recovery.
Collapse
Affiliation(s)
- Andrew D Gaudet
- Department of Neuroscience and Center for Brain and Spinal Cord Repair, College of Medicine, The Ohio State University, 770 Biomedical Research Tower, 460 West 12th Ave, Columbus, OH, 43210, USA
- International Collaboration On Repair Discoveries (ICORD), Vancouver Coastal Health Research Institute, and Department of Zoology, University of British Columbia, 818 West 10th Ave, Vancouver, BC, V5T 1M9, Canada
| | - Phillip G Popovich
- Department of Neuroscience and Center for Brain and Spinal Cord Repair, College of Medicine, The Ohio State University, 770 Biomedical Research Tower, 460 West 12th Ave, Columbus, OH, 43210, USA
| | - Matt S Ramer
- International Collaboration On Repair Discoveries (ICORD), Vancouver Coastal Health Research Institute, and Department of Zoology, University of British Columbia, 818 West 10th Ave, Vancouver, BC, V5T 1M9, Canada
| |
Collapse
|
33
|
Kristian T, Balan I, Schuh R, Onken M. Mitochondrial dysfunction and nicotinamide dinucleotide catabolism as mechanisms of cell death and promising targets for neuroprotection. J Neurosci Res 2011; 89:1946-55. [PMID: 21488086 DOI: 10.1002/jnr.22626] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2010] [Revised: 01/07/2011] [Accepted: 01/17/2011] [Indexed: 11/12/2022]
Abstract
Both acute and chronic neurodegenerative diseases are frequently associated with mitochondrial dysfunction as an essential component of mechanisms leading to brain damage. Although loss of mitochondrial functions resulting from prolonged activation of the mitochondrial permeability transition (MPT) pore has been shown to play a significant role in perturbation of cellular bioenergetics and in cell death, the detailed mechanisms are still elusive. Enzymatic reactions linked to glycolysis, the tricarboxylic acid cycle, and mitochondrial respiration are dependent on the reduced or oxidized form of nicotinamide dinucleotide [NAD(H)] as a cofactor. Loss of mitochondrial NAD(+) resulting from MPT pore opening, although transient, allows detrimental depletion of mitochondrial and cellular NAD(+) pools by activated NAD(+) glycohydrolases. Poly(ADP-ribose) polymerase (PARP) is considered to be a major NAD(+) degrading enzyme, particularly under conditions of extensive DNA damage. We propose that CD38, a main cellular NAD(+) level regulator, can significantly contribute to NAD(+) catabolism. We discuss NAD(+) catabolic and NAD(+) synthesis pathways and their role in different strategies to prevent cellular NAD(+) degradation in brain, particularly following an ischemic insult. These therapeutic approaches are based on utilizing endogenous intermediates of NAD(+) metabolism that feed into the NAD(+) salvage pathway and also inhibit CD38 activity.
Collapse
Affiliation(s)
- Tibor Kristian
- Department of Anesthesiology, Center for Shock, Trauma and Anesthesiology Research, School of Medicine, University of Maryland Baltimore, Baltimore, Maryland 21201, USA.
| | | | | | | |
Collapse
|
34
|
Abstract
Traditionally, researchers have believed that axons are highly dependent on their cell bodies for long-term survival. However, recent studies point to the existence of axon-autonomous mechanism(s) that regulate rapid axon degeneration after axotomy. Here, we review the cellular and molecular events that underlie this process, termed Wallerian degeneration. We describe the biphasic nature of axon degeneration after axotomy and our current understanding of how Wld(S)--an extraordinary protein formed by fusing a Ube4b sequence to Nmnat1--acts to protect severed axons. Interestingly, the neuroprotective effects of Wld(S) span all species tested, which suggests that there is an ancient, Wld(S)-sensitive axon destruction program. Recent studies with Wld(S) also reveal that Wallerian degeneration is genetically related to several dying back axonopathies, thus arguing that Wallerian degeneration can serve as a useful model to understand, and potentially treat, axon degeneration in diverse traumatic or disease contexts.
Collapse
Affiliation(s)
- Michael P Coleman
- Laboratory of Molecular Signaling, The Babraham Institute, Cambridge CB223AT, United Kingdom
| | | |
Collapse
|
35
|
Kilinc D, Peyrin JM, Soubeyre V, Magnifico S, Saias L, Viovy JL, Brugg B. Wallerian-like degeneration of central neurons after synchronized and geometrically registered mass axotomy in a three-compartmental microfluidic chip. Neurotox Res 2011; 19:149-61. [PMID: 20162389 PMCID: PMC3006648 DOI: 10.1007/s12640-010-9152-8] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2009] [Revised: 10/15/2009] [Accepted: 01/12/2010] [Indexed: 01/09/2023]
Abstract
Degeneration of central axons may occur following injury or due to various diseases and it involves complex molecular mechanisms that need to be elucidated. Existing in vitro axotomy models are difficult to perform, and they provide limited information on the localization of events along the axon. We present here a novel experimental model system, based on microfluidic isolation, which consists of three distinct compartments, interconnected by parallel microchannels allowing axon outgrowth. Neurons cultured in one compartment successfully elongated their axons to cross a short central compartment and invade the outermost compartment. This design provides an interesting model system for studying axonal degeneration and death mechanisms, with a previously impossible spatial and temporal control on specific molecular pathways. We provide a proof-of-concept of the system by reporting its application to a well-characterized experimental paradigm, axotomy-induced Wallerian degeneration in primary central neurons. Using this model, we applied localized central axotomy by a brief, isolated flux of detergent. We report that mouse embryonic cortical neurons exhibit rapid Wallerian-like distal degeneration but no somatic death following central axotomy. Distal axons show progressive degeneration leading to axonal beading and cytoskeletal fragmentation within a few hours after axotomy. Degeneration is asynchronous, reminiscent of in vivo Wallerian degeneration. Axonal cytoskeletal fragmentation is significantly delayed with nicotinamide adenine dinucleotide pretreatment, but it does not change when distal calpain or caspase activity is inhibited. These findings, consistent with previous experiments in vivo, confirm the power and biological relevance of this microfluidic architecture.
Collapse
Affiliation(s)
- Devrim Kilinc
- Laboratoire de Neurobiologie des Processus Adaptatifs, UMR 7102 CNRS/UPMC, Univ. P. et M. Curie, Bat B, 6eme Etage, Case courrier 12, 9 Quai St. Bernard, 75252 Paris, France
| | - Jean-Michel Peyrin
- Laboratoire de Neurobiologie des Processus Adaptatifs, UMR 7102 CNRS/UPMC, Univ. P. et M. Curie, Bat B, 6eme Etage, Case courrier 12, 9 Quai St. Bernard, 75252 Paris, France
| | - Vanessa Soubeyre
- Laboratoire de Neurobiologie des Processus Adaptatifs, UMR 7102 CNRS/UPMC, Univ. P. et M. Curie, Bat B, 6eme Etage, Case courrier 12, 9 Quai St. Bernard, 75252 Paris, France
| | - Sébastien Magnifico
- Laboratoire de Neurobiologie des Processus Adaptatifs, UMR 7102 CNRS/UPMC, Univ. P. et M. Curie, Bat B, 6eme Etage, Case courrier 12, 9 Quai St. Bernard, 75252 Paris, France
| | - Laure Saias
- Laboratoire Physicochimie-Curie, UMR 168 Institut Curie/CNRS/UPMC, Paris, France
| | - Jean-Louis Viovy
- Laboratoire Physicochimie-Curie, UMR 168 Institut Curie/CNRS/UPMC, Paris, France
| | - Bernard Brugg
- Laboratoire de Neurobiologie des Processus Adaptatifs, UMR 7102 CNRS/UPMC, Univ. P. et M. Curie, Bat B, 6eme Etage, Case courrier 12, 9 Quai St. Bernard, 75252 Paris, France
| |
Collapse
|
36
|
Barrette B, Calvo E, Vallières N, Lacroix S. Transcriptional profiling of the injured sciatic nerve of mice carrying the Wld(S) mutant gene: identification of genes involved in neuroprotection, neuroinflammation, and nerve regeneration. Brain Behav Immun 2010; 24:1254-67. [PMID: 20688153 DOI: 10.1016/j.bbi.2010.07.249] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2010] [Revised: 07/29/2010] [Accepted: 07/29/2010] [Indexed: 12/21/2022] Open
Abstract
Wallerian degeneration (WD) involves the fragmentation of axonal segments disconnected from their cell bodies, segmentation of the myelin sheath, and removal of debris by Schwann cells and immune cells. The removal and downregulation of myelin-associated inhibitors of axonal regeneration and synthesis of growth factors by these two cell types are critical responses to successful nerve repair. Here, we analyzed the transcriptome of the sciatic nerve of mice carrying the Wallerian degeneration slow (Wld(S)) mutant gene, a gene that confers axonal protection in the distal stump after injury, therefore causing significant delays in WD, neuroinflammation, and axonal regeneration. Of the thousands of genes analyzed by microarray, 719 transcripts were differentially expressed between Wld(S) and wild-type (wt) mice. Notably, the Nmnat1, a transcript contained within the sequence of the Wld(S) gene, was upregulated by five to eightfold in the sciatic nerve of naive Wld(S) mice compared with wt. The injured sciatic nerve of wt could be further distinguished from the one of Wld(S) mice by the preferential upregulation of genes involved in axonal processes and plasticity (Chl1, Epha5, Gadd45b, Jun, Nav2, Nptx1, Nrcam, Ntm, Sema4f), inflammation and immunity (Arg1, Lgals3, Megf10, Panx1), growth factors/cytokines and their receptors (Clcf1, Fgf5, Gdnf, Gfrα1, Il7r, Lif, Ngfr/p75(NTR), Shh), and cell adhesion and extracellular matrix (Adam8, Gpc1, Mmp9, Tnc). These results will help understand how the nervous and immune systems interact to modulate nerve repair, and identify the molecules that drive these responses.
Collapse
Affiliation(s)
- Benoit Barrette
- Laboratory of Endocrinology and Genomics, Department of Molecular Medicine, Université Laval, CHUL Research Center, Québec, Canada
| | | | | | | |
Collapse
|
37
|
Feng Y, Yan T, Zheng J, Ge X, Mu Y, Zhang Y, Wu D, Du JL, Zhai Q. Overexpression of Wldsor Nmnat2 in mauthner cells by single-cell electroporation delays axon degeneration in live zebrafish. J Neurosci Res 2010; 88:3319-27. [DOI: 10.1002/jnr.22498] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
38
|
Axonal degeneration is regulated by the apoptotic machinery or a NAD+-sensitive pathway in insects and mammals. J Neurosci 2010; 30:6375-86. [PMID: 20445064 DOI: 10.1523/jneurosci.0922-10.2010] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Selective degeneration of neuronal projections and neurite pruning are critical for establishment and maintenance of functional neural circuits in both insects and mammals. However, the molecular mechanisms that govern developmental neurite pruning versus injury-induced neurite degeneration are still mostly unclear. Here, we show that the effector caspases 6 and 3 are both expressed within axons and that, on trophic deprivation, they exhibit distinct modes of activation. Surprisingly, inhibition of caspases is not sufficient for axonal protection and a parallel modulation of a NAD(+)-sensitive pathway is required. The proapoptotic protein BAX is a key element in both pathways as its genetic ablation protected sensory axons against developmental degeneration both in vitro and in vivo. Last, we demonstrate that both pathways are also involved in developmental dendritic pruning in Drosophila. More specifically, the mouse Wld(S) (Wallerian degeneration slow) protein, which is mainly composed of the full-length sequence of the NAD(+) biosynthetic Nmnat1 enzyme, can suppress dendritic pruning in C4da (class IV dendritic arborization) sensory neurons in parallel to the fly effector caspases. These findings indicate that two distinct autodestruction pathways act separately or in concert to regulate developmental neurite pruning.
Collapse
|
39
|
Feng Y, Yan T, He Z, Zhai Q. Wld(S), Nmnats and axon degeneration--progress in the past two decades. Protein Cell 2010; 1:237-45. [PMID: 21203970 DOI: 10.1007/s13238-010-0021-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2009] [Accepted: 12/21/2009] [Indexed: 01/06/2023] Open
Abstract
A chimeric protein called Wallerian degeneration slow (Wld(S)) was first discovered in a spontaneous mutant strain of mice that exhibited delayed Wallerian degeneration. This provides a useful tool in elucidating the mechanisms of axon degeneration. Over-expression of Wld(S) attenuates the axon degeneration that is associated with several neurodegenerative disease models, suggesting a new logic for developing a potential protective strategy. At molecular level, although Wld(S) is a fusion protein, the nicotinamide mononucleotide adenylyl transferase 1 (Nmnat1) is required and sufficient for the protective effects of Wld(S), indicating a critical role of NAD biosynthesis and perhaps energy metabolism in axon degeneration. These findings challenge the proposed model in which axon degeneration is operated by an active programmed process and thus may have important implication in understanding the mechanisms of neurodegeneration. In this review, we will summarize these recent findings and discuss their relevance to the mechanisms of axon degeneration.
Collapse
Affiliation(s)
- Yan Feng
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Shanghai 200031, China
| | | | | | | |
Collapse
|
40
|
Wallerian degeneration and axonal regeneration after sciatic nerve crush are altered in ICAM-1-deficient mice. Cell Tissue Res 2009; 338:19-28. [DOI: 10.1007/s00441-009-0837-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2009] [Accepted: 06/26/2009] [Indexed: 01/09/2023]
|
41
|
Non-nuclear Wld(S) determines its neuroprotective efficacy for axons and synapses in vivo. J Neurosci 2009; 29:653-68. [PMID: 19158292 DOI: 10.1523/jneurosci.3814-08.2009] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Axon degeneration contributes widely to neurodegenerative disease but its regulation is poorly understood. The Wallerian degeneration slow (Wld(S)) protein protects axons dose-dependently in many circumstances but is paradoxically abundant in nuclei. To test the hypothesis that Wld(S) acts within nuclei in vivo, we redistributed it from nucleus to cytoplasm in transgenic mice. Surprisingly, instead of weakening the phenotype as expected, extranuclear Wld(S) significantly enhanced structural and functional preservation of transected distal axons and their synapses. In contrast to native Wld(S) mutants, distal axon stumps remained continuous and ultrastructurally intact up to 7 weeks after injury and motor nerve terminals were robustly preserved even in older mice, remaining functional for 6 d. Moreover, we detect extranuclear Wld(S) for the first time in vivo, and higher axoplasmic levels in transgenic mice with Wld(S) redistribution. Cytoplasmic Wld(S) fractionated predominantly with mitochondria and microsomes. We conclude that Wld(S) can act in one or more non-nuclear compartments to protect axons and synapses, and that molecular changes can enhance its therapeutic potential.
Collapse
|
42
|
Abstract
The CNS contains relatively few unmyelinated nerve fibers, and thus benefits from the advantages that are conferred by myelination, including faster conduction velocities, lower energy consumption for impulse transmission, and greater stability of point-to-point connectivity. In the PNS many fibers or regions of fibers the Schwann do not form myelin. Examples include C fibers nociceptors, postganglionic sympathetic fibers, and the Schwann cells associated with motor nerve terminals at neuromuscular junctions. These examples retain a degree of plasticity and a capacity to sprout collaterally that is unusual in myelinated fibers. Nonmyelin-forming Schwann cells, including those associated with uninjured fibers, have the capacity to act as the "first responders" to injury or disease in their neighborhoods.
Collapse
Affiliation(s)
- John W Griffin
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| | | |
Collapse
|
43
|
Avery MA, Sheehan AE, Kerr KS, Wang J, Freeman MR. Wld S requires Nmnat1 enzymatic activity and N16-VCP interactions to suppress Wallerian degeneration. ACTA ACUST UNITED AC 2009; 184:501-13. [PMID: 19237597 PMCID: PMC2654119 DOI: 10.1083/jcb.200808042] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Slow Wallerian degeneration (Wld(S)) encodes a chimeric Ube4b/nicotinamide mononucleotide adenylyl transferase 1 (Nmnat1) fusion protein that potently suppresses Wallerian degeneration, but the mechanistic action of Wld(S) remains controversial. In this study, we characterize Wld(S)-mediated axon protection in vivo using Drosophila melanogaster. We show that Nmnat1 can protect severed axons from autodestruction but at levels significantly lower than Wld(S), and enzyme-dead versions of Nmnat1 and Wld(S) exhibit severely reduced axon-protective function. Interestingly, a 16-amino acid N-terminal domain of Wld(S) (termed N16) accounts for the differences in axon-sparing activity between Wld(S) and Nmnat1, and N16-dependent enhancement of Nmnat1-protective activity in Wld(S) requires the N16-binding protein valosin-containing protein (VCP)/TER94. Thus, Wld(S)-mediated suppression of Wallerian degeneration results from VCP-N16 interactions and Nmnat1 activity converging in vivo. Surprisingly, mouse Nmnat3, a mitochondrial Nmnat enzyme that localizes to the cytoplasm in Drosophila cells, protects severed axons at levels indistinguishable from Wld(S). Thus, nuclear Nmnat activity does not appear to be essential for Wld(S)-like axon protection.
Collapse
Affiliation(s)
- Michelle A Avery
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | | | | | | |
Collapse
|
44
|
Bogan KL, Brenner C. Nicotinic acid, nicotinamide, and nicotinamide riboside: a molecular evaluation of NAD+ precursor vitamins in human nutrition. Annu Rev Nutr 2008; 28:115-30. [PMID: 18429699 DOI: 10.1146/annurev.nutr.28.061807.155443] [Citation(s) in RCA: 513] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Although baseline requirements for nicotinamide adenine dinucleotide (NAD+) synthesis can be met either with dietary tryptophan or with less than 20 mg of daily niacin, which consists of nicotinic acid and/or nicotinamide, there is growing evidence that substantially greater rates of NAD+ synthesis may be beneficial to protect against neurological degeneration, Candida glabrata infection, and possibly to enhance reverse cholesterol transport. The distinct and tissue-specific biosynthetic and/or ligand activities of tryptophan, nicotinic acid, nicotinamide, and the newly identified NAD+ precursor, nicotinamide riboside, reviewed herein, are responsible for vitamin-specific effects and side effects. Because current data suggest that nicotinamide riboside may be the only vitamin precursor that supports neuronal NAD+ synthesis, we present prospects for human nicotinamide riboside supplementation and propose areas for future research.
Collapse
Affiliation(s)
- Katrina L Bogan
- Department of Genetics and the Norris Cotton Cancer Center, Dartmouth Medical School, Lebanon, New Hampshire 03756, USA.
| | | |
Collapse
|
45
|
Giuditta A, Tai Chun J, Eyman M, Cefaliello C, Bruno AP, Crispino M. Local Gene Expression in Axons and Nerve Endings: The Glia-Neuron Unit. Physiol Rev 2008; 88:515-55. [DOI: 10.1152/physrev.00051.2006] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Neurons have complex and often extensively elongated processes. This unique cell morphology raises the problem of how remote neuronal territories are replenished with proteins. For a long time, axonal and presynaptic proteins were thought to be exclusively synthesized in the cell body, which delivered them to peripheral sites by axoplasmic transport. Despite this early belief, protein has been shown to be synthesized in axons and nerve terminals, substantially alleviating the trophic burden of the perikaryon. This observation raised the question of the cellular origin of the peripheral RNAs involved in protein synthesis. The synthesis of these RNAs was initially attributed to the neuron soma almost by default. However, experimental data and theoretical considerations support the alternative view that axonal and presynaptic RNAs are also transcribed in the flanking glial cells and transferred to the axon domain of mature neurons. Altogether, these data suggest that axons and nerve terminals are served by a distinct gene expression system largely independent of the neuron cell body. Such a local system would allow the neuron periphery to respond promptly to environmental stimuli. This view has the theoretical merit of extending to axons and nerve terminals the marginalized concept of a glial supply of RNA (and protein) to the neuron cell body. Most long-term plastic changes requiring de novo gene expression occur in these domains, notably in presynaptic endings, despite their intrinsic lack of transcriptional capacity. This review enlightens novel perspectives on the biology and pathobiology of the neuron by critically reviewing these issues.
Collapse
|
46
|
Howell GR, Libby RT, Jakobs TC, Smith RS, Phalan FC, Barter JW, Barbay JM, Marchant JK, Mahesh N, Porciatti V, Whitmore AV, Masland RH, John SWM. Axons of retinal ganglion cells are insulted in the optic nerve early in DBA/2J glaucoma. J Cell Biol 2007; 179:1523-37. [PMID: 18158332 PMCID: PMC2373494 DOI: 10.1083/jcb.200706181] [Citation(s) in RCA: 476] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2007] [Accepted: 11/19/2007] [Indexed: 01/21/2023] Open
Abstract
Here, we use a mouse model (DBA/2J) to readdress the location of insult(s) to retinal ganglion cells (RGCs) in glaucoma. We localize an early sign of axon damage to an astrocyte-rich region of the optic nerve just posterior to the retina, analogous to the lamina cribrosa. In this region, a network of astrocytes associates intimately with RGC axons. Using BAX-deficient DBA/2J mice, which retain all of their RGCs, we provide experimental evidence for an insult within or very close to the lamina in the optic nerve. We show that proximal axon segments attached to their cell bodies survive to the proximity of the lamina. In contrast, axon segments in the lamina and behind the eye degenerate. Finally, the Wld(s) allele, which is known to protect against insults to axons, strongly protects against DBA/2J glaucoma and preserves RGC activity as measured by pattern electroretinography. These experiments provide strong evidence for a local insult to axons in the optic nerve.
Collapse
|
47
|
Touma E, Kato S, Fukui K, Koike T. Calpain-mediated cleavage of collapsin response mediator protein(CRMP)-2 during neurite degeneration in mice. Eur J Neurosci 2007; 26:3368-81. [PMID: 18052987 DOI: 10.1111/j.1460-9568.2007.05943.x] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Axon or dendrite degeneration involves activation of the ubiquitin-proteasome system, failure to maintain neuritic ATP levels, microtubule fragmentation and a mitochondrial permeability transition that occur independently of the somal death programs. To gain further insight into the neurite degeneration mechanims we have compared two-dimensional gel electrophoresis patterns of neurite proteins from suprior cervical ganglia during degeneration caused by nerve growth factor (NGF) deprivation. We show here that collapsin response mediator protein (CRMP)-2 and CMRP-4 protein patterns were altered during beading formation, an early hallmark of neurite degeneration, prior to neurite fragmentation, the final stage of degeneration. Western blotting using a monoclonal antibody against CRMP-2 shows that the native form (64 kDa) was cleaved to generate a truncated form (58 kDa). No cleavage of CRMP-2 or -4 occurred in NGF-deprived neurites from Wld(s) (Wallerian degeneration slow) mutant mice in which neurite degeneration is markedly delayed. Using different protease inhibitors, purified calpain 1 protein and calpain 1-specific siRNA, we have demonstrated that CRMP-2 is a substrate for calpain 1. Indeed, caplain activity was activated at an early phase of neuronal degeneration in cerebellar granule neurons, and down-regulation of caplain 1 expression suppressed CRMP-2 cleavage. Furthermore, this cleavage occurred after vinblastine treatment or in vitro Wallerian degeneration, suggesting that it represents a common step in the process of dying neurites. CRMP-2 and -4 play a pivotal role in axonal growth and transport, and the C-terminus region of CRMP-2 is essential for its binding to kinesin-1. Hence, this cleavage will render them dysfunctional and subject to autophagic processing associated with beading formation, as evidenced by the finding that the truncated form was localized in the beadings.
Collapse
Affiliation(s)
- Ekatherina Touma
- Molecular Neurobiology Laboratory, Graduate School of Life Science, Hokkaido University, Sapporo 060-0810, Japan
| | | | | | | |
Collapse
|
48
|
Axon & dendrite degeneration: its mechanisms and protective experimental paradigms. Neurochem Int 2007; 52:751-60. [PMID: 18029056 DOI: 10.1016/j.neuint.2007.09.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2007] [Revised: 09/01/2007] [Accepted: 09/07/2007] [Indexed: 12/13/2022]
Abstract
Accumulating evidence suggests that axon and dendrite (or neurite) degeneration both in vivo and in vitro requires self-destructive programs independent of cell death programs to segregate neurite degeneration from cell soma demise. This review will deal with the mechanisms of neurite degeneration caused by several experimental paradigms including trophic factor deprivation and Wallerian degeneration as well as those under pathological conditions. The involvement of autophagy and mitochondrial dysfunction is emphasized in these mechanisms. The mechanisms through which protective agents including the Wld(s) protein rescue neurites from degeneration or fail to do so will be discussed.
Collapse
|
49
|
Saxena S, Caroni P. Mechanisms of axon degeneration: from development to disease. Prog Neurobiol 2007; 83:174-91. [PMID: 17822833 DOI: 10.1016/j.pneurobio.2007.07.007] [Citation(s) in RCA: 194] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2007] [Revised: 03/31/2007] [Accepted: 07/20/2007] [Indexed: 01/15/2023]
Abstract
Axon degeneration is an active, tightly controlled and versatile process of axon segment self-destruction. Although not involving cell death, it resembles apoptosis in its logics. It involves three distinct steps: induction of competence in specific neurons, triggering of degeneration at defined axon segments of competent neurons, and rapid fragmentation and removal of the segments. The mechanisms that initiate degeneration are specific to individual settings, but the final pathway of pruning is shared; it involves microtubule disassembly, axon swellings, axon fragmentation, and removal of the remnants by locally recruited phagocytes. The tight regulatory properties of axon degeneration distinguish it from passive loss phenomena, and confer significance to processes that involve it. Axon degeneration has prominent roles in development, upon lesions and in disease. In development, it couples the progressive specification of neurons and circuits to the removal of defined axon branches. Competence might involve transcriptional switches, and local triggering can involve axon guidance molecules and synaptic activity patterns. Lesion-induced Wallerian degeneration is inhibited in the presence of Wld(S) fusion protein in neurons; it involves early local, and later, distal degeneration. It has recently become clear that like in other settings, axon degeneration in disease is a rapid and specific process, which should not be confused with a variety of disease-related pathologies. Elucidating the specific mechanisms that initiate axon degeneration should open up new avenues to investigate principles of circuit assembly and plasticity, to uncover mechanisms of disease progression, and to identify ways of protecting synapses and axons in disease.
Collapse
Affiliation(s)
- Smita Saxena
- Friedrich Miescher Institute, Maulbeerstrasse 66, CH-4058 Basel, Switzerland
| | | |
Collapse
|
50
|
Abstract
Wallerian degeneration (WD) is the set of molecular and cellular events by which degenerating axons and myelin are cleared after injury. Why WD is rapid and robust in the PNS but slow and incomplete in the CNS is a longstanding mystery. Here we review current work on the mechanisms of WD with an emphasis on deciphering this mystery and on understanding whether slow WD in the CNS could account for the failure of CNS axons to regenerate.
Collapse
Affiliation(s)
- Mauricio E Vargas
- Department of Neurobiology, Stanford University School of Medicine, Stanford, California 94305, USA.
| | | |
Collapse
|