1
|
Sarkar A, Kumari N, Mukherjee P. The curious case of SARM1: Dr. Jekyll and Mr. Hyde in cell death and immunity? FEBS J 2023; 290:340-358. [PMID: 34710262 DOI: 10.1111/febs.16256] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/21/2021] [Accepted: 10/27/2021] [Indexed: 02/06/2023]
Abstract
Sterile alpha and toll/interleukin-1 receptor motif-containing protein 1 (SARM1) was first identified as a novel ortholog of Drosophila protein CG7915 and was subsequently placed as the fifth member of the human TIR-containing adaptor protein. SARM1 holds a unique position in this family where, unlike other members, it downregulates NFκB activity in response to immunogenic stimulation, interacts with another member of the family, TRIF, to negatively regulate its function, and it also mediates cell death responses. Over the past decade, SARM1 has emerged as one of the primary mediators of programmed axonal degeneration and this robust regulation of axonal degeneration-especially in models of peripheral neuropathy and traumatic injury-makes it an attractive target for therapeutic intervention. The TIR domain of SARM1 possesses an intrinsic NADase activity resulting in cellular energy deficits within the axons, a striking deviation from its other family members of human TLR adaptors. Interestingly, the TIR NADase activity, as seen in SARM1, is also observed in several prokaryotic TIR-containing proteins where they are involved in immune evasion once within the host. Although the immune function of SARM1 is yet to be conclusively discerned, this closeness in function with the prokaryotic TIR-domain containing proteins, places it at an interesting juncture of evolution raising questions about its origin and function in cell death and immunity. In this review, we discuss how a conserved immune adaptor protein like SARM1 switches to a pro-neurodegenerative function and the evolutionarily significance of the process.
Collapse
Affiliation(s)
- Ankita Sarkar
- School of Biotechnology, Presidency University, Kolkata, West Bengal, India
| | - Nripa Kumari
- School of Biotechnology, Presidency University, Kolkata, West Bengal, India
| | - Piyali Mukherjee
- School of Biotechnology, Presidency University, Kolkata, West Bengal, India
| |
Collapse
|
2
|
Ward JM, Vogel P, Sundberg JP. Brain and spinal cord lesions in 28 inbred strains of aging mice. Vet Pathol 2022; 59:1047-1055. [PMID: 36062914 DOI: 10.1177/03009858221120009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Brain and spinal cord histopathology findings in male and female 20-month-old mice in a large-scale aging study of 28 inbred Jackson Laboratory mouse strains from 7 genetic families are described. Brain sections from selected strains at 12 and 24 months of age or older were also reviewed. Common lesions include axonal dystrophy in the gracile and/or cuneate nucleus in the sensory tract of the dorsal medulla and in the spinal cord in all strains. Hirano-like bodies were seen in 24/28 strains, and mineralization was observed in the thalamus of 9/28 strains. Less common lesions were also seen in the cerebellum, cerebral cortex, and other brain areas. No brain or spinal cord tumors were found. Evidence of an impairment of the ubiquitin-proteasome system (UPS) and/or suspected autophagy was manifested as medullary axonal dystrophy with intra-axonal granular eosinophilic bodies and LC3B immunohistochemistry in most strains. RIIIS/J, the most severely affected strain, showed moderate axonal dystrophy at 12 months, which progressed to severe lesions at 20 months. Comparative pathology in various species is discussed.
Collapse
Affiliation(s)
- Jerrold M Ward
- The Jackson Laboratory, Bar Harbor, ME.,Global Vet Pathology, Montgomery Village, MD
| | - Peter Vogel
- St. Jude Children's Research Hospital, Memphis, TN
| | | |
Collapse
|
3
|
Cao Y, Wang Y, Yang J. NAD +-dependent mechanism of pathological axon degeneration. CELL INSIGHT 2022; 1:100019. [PMID: 37193131 PMCID: PMC10120281 DOI: 10.1016/j.cellin.2022.100019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 05/18/2023]
Abstract
Pathological axon degeneration is broadly observed in neurodegenerative diseases. This unique process of axonal pathology could directly interfere with the normal functions of neurocircuitries and contribute to the onset of clinical symptoms in patients. It has been increasingly recognized that functional preservation of axonal structures is an indispensable part of therapeutic strategies for treating neurological disorders. In the past decades, the research field has witnessed significant breakthroughs in understanding the stereotyped self-destruction of axons upon neurodegenerative insults, which is distinct from all the known types of programmed cell death. In particular, the novel NAD+-dependent mechanism involving the WLDs, NMNAT2, and SARM1 proteins has emerged. This review summarizes the landmark discoveries elucidating the molecular pathway of pathological axon degeneration and highlights the evolving concept that neurodegeneration would be intrinsically linked to NAD+ and energy metabolism.
Collapse
Affiliation(s)
- Ying Cao
- Center for Life Sciences, Peking University, Beijing, 100871, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Yi Wang
- Center for Life Sciences, Peking University, Beijing, 100871, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Jing Yang
- Center for Life Sciences, Peking University, Beijing, 100871, China
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, 100871, China
- IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
- Chinese Institute for Brain Research, Beijing, 102206, China
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, 518055, China
| |
Collapse
|
4
|
Li S, Shi S, Luo B, Xia F, Ha Y, Merkley KH, Motamedi M, Zhang W, Liu H. Tauopathy induces degeneration and impairs regeneration of sensory nerves in the cornea. Exp Eye Res 2021; 215:108900. [PMID: 34929160 DOI: 10.1016/j.exer.2021.108900] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/21/2021] [Accepted: 12/13/2021] [Indexed: 12/26/2022]
Abstract
The cornea is transparent and innervated by a dense collection of sensory nerves originating from the ocular branch of the trigeminal nerve. This study was designed to comprehensively analyze alterations of corneal sub-basal nerve plexus in a mouse model of tauopathy (P301L transgenic mice) to test the possibility of using corneal nerves as a biomarker for tauopathy. Corneal sensitivity, thickness and epithelial wound healing were measured non-invasively by aeshesiometer, optical coherence tomography and fluorescein staining, respectively. Tau, corneal nerves and immune cells were examined by immunohistochemistry or Western blot. At the early stage of tauopathy, although corneal sensitivity, thickness and nerve fiber density were not greatly altered, corneal nerve abnormalities were observed in the peripheral region of young P301L mice. With aging, the density of abnormal nerves increased, while corneal sensitivity, epithelial thickness, nerve fiber density and length decreased in middle-aged P301L mice compared with WT mice. After corneal epithelial injury in young mice, no difference in reepithelialization was observed between two groups of mice, however, the regeneration of corneal nerves in P301L mice lagged behind WT mice, which was reflected by delayed recovery of corneal sensitivity, decreased corneal nerve density and length and density of CD45+ dendriform cells in P301L mice. In conclusion, our data provide compelling evidence that corneal nerves were changed in a mouse model of tauopathy in an age-dependent manner. Moreover, tau overexpression impairs corneal nerve regeneration. These results suggest that cornea may serve as a promising ocular site for the early diagnosis of tauopathy.
Collapse
Affiliation(s)
- Shengguo Li
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China; Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Shuizhen Shi
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Ban Luo
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Fan Xia
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Yonju Ha
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Kevin H Merkley
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Massoud Motamedi
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Wenbo Zhang
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA; Departments of Neuroscience, Cell Biology & Anatomy, University of Texas Medical Branch, Galveston, TX, USA.
| | - Hua Liu
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
5
|
Arthur-Farraj P, Coleman MP. Lessons from Injury: How Nerve Injury Studies Reveal Basic Biological Mechanisms and Therapeutic Opportunities for Peripheral Nerve Diseases. Neurotherapeutics 2021; 18:2200-2221. [PMID: 34595734 PMCID: PMC8804151 DOI: 10.1007/s13311-021-01125-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2021] [Indexed: 12/25/2022] Open
Abstract
Since Waller and Cajal in the nineteenth and early twentieth centuries, laboratory traumatic peripheral nerve injury studies have provided great insight into cellular and molecular mechanisms governing axon degeneration and the responses of Schwann cells, the major glial cell type of peripheral nerves. It is now evident that pathways underlying injury-induced axon degeneration and the Schwann cell injury-specific state, the repair Schwann cell, are relevant to many inherited and acquired disorders of peripheral nerves. This review provides a timely update on the molecular understanding of axon degeneration and formation of the repair Schwann cell. We discuss how nicotinamide mononucleotide adenylyltransferase 2 (NMNAT2) and sterile alpha TIR motif containing protein 1 (SARM1) are required for axon survival and degeneration, respectively, how transcription factor c-JUN is essential for the Schwann cell response to nerve injury and what each tells us about disease mechanisms and potential therapies. Human genetic association with NMNAT2 and SARM1 strongly suggests aberrant activation of programmed axon death in polyneuropathies and motor neuron disorders, respectively, and animal studies suggest wider involvement including in chemotherapy-induced and diabetic neuropathies. In repair Schwann cells, cJUN is aberrantly expressed in a wide variety of human acquired and inherited neuropathies. Animal models suggest it limits axon loss in both genetic and traumatic neuropathies, whereas in contrast, Schwann cell secreted Neuregulin-1 type 1 drives onion bulb pathology in CMT1A. Finally, we discuss opportunities for drug-based and gene therapies to prevent axon loss or manipulate the repair Schwann cell state to treat acquired and inherited neuropathies and neuronopathies.
Collapse
Affiliation(s)
- Peter Arthur-Farraj
- Department of Clinical Neurosciences, John Van Geest Centre for Brain Repair, University of Cambridge, Robinson Way, Cambridge, CB2 0PY, UK.
| | - Michael P Coleman
- Department of Clinical Neurosciences, John Van Geest Centre for Brain Repair, University of Cambridge, Robinson Way, Cambridge, CB2 0PY, UK.
| |
Collapse
|
6
|
Peters OM, Weiss A, Metterville J, Song L, Logan R, Smith GA, Schwarzschild MA, Mueller C, Brown RH, Freeman M. Genetic diversity of axon degenerative mechanisms in models of Parkinson's disease. Neurobiol Dis 2021; 155:105368. [PMID: 33892050 PMCID: PMC8292971 DOI: 10.1016/j.nbd.2021.105368] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 04/01/2021] [Accepted: 04/18/2021] [Indexed: 12/26/2022] Open
Abstract
Parkinson's disease (PD) is the most common form of neurodegenerative movement disorder, associated with profound loss of dopaminergic neurons from the basal ganglia. Though loss of dopaminergic neuron cell bodies from the substantia nigra pars compacta is a well-studied feature, atrophy and loss of their axons within the nigrostriatal tract is also emerging as an early event in disease progression. Genes that drive the Wallerian degeneration, like Sterile alpha and toll/interleukin-1 receptor motif containing (Sarm1), are excellent candidates for driving this axon degeneration, given similarities in the morphology of axon degeneration after axotomy and in PD. In the present study we assessed whether Sarm1 contributes to loss of dopaminergic projections in mouse models of PD. In Sarm1 deficient mice, we observed a significant delay in the degeneration of severed dopaminergic axons distal to a 6-OHDA lesion of the medial forebrain bundle (MFB) in the nigrostriatal tract, and an accompanying rescue of morphological, biochemical and behavioural phenotypes. However, we observed no difference compared to controls when striatal terminals were lesioned with 6-OHDA to induce a dying back form of neurodegeneration. Likewise, when PD phenotypes were induced using AAV-induced alpha-synuclein overexpression, we observed similar modest loss of dopaminergic terminals in Sarm1 knockouts and controls. Our data argues that axon degeneration after MFB lesion is Sarm1-dependent, but that other models for PD do not require Sarm1, or that Sarm1 acts with other redundant genetic pathways. This work adds to a growing body of evidence indicating Sarm1 contributes to some, but not all types of neurodegeneration, and supports the notion that while axon degeneration in many context appears morphologically similar, a diversity of axon degeneration programs exist.
Collapse
Affiliation(s)
- Owen M Peters
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01655, USA; Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| | - Alexandra Weiss
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Jake Metterville
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Lina Song
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Robert Logan
- Molecular Neurobiology Laboratory, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA; Eastern Nazarene College, Quincy, MA 02170, USA
| | - Gaynor A Smith
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Michael A Schwarzschild
- Molecular Neurobiology Laboratory, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Christian Mueller
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Robert H Brown
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Marc Freeman
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| |
Collapse
|
7
|
Kikuchi T. Circular breakdown of neural networks due to loss of deubiquitinating enzyme (UCH-L1) in gracile axonal dystrophy (<i>gad</i>) mouse. AIMS MOLECULAR SCIENCE 2021. [DOI: 10.3934/molsci.2021024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
<abstract>
<p>Gracile axonal dystrophy (gad) mouse shows tremor, ataxia and muscular atrophy of hind limbs from about 80-days of age. These clinical features become progressively severe to death. Pathological examination reveals that main and early axonal degeneration exists in a long ascending nervous tract in dorsal column of the spinal cord: gracile nucleus and fascicules. Similar lesions are seen in axonal terminals of peripheral sensory (muscle spindles) and motor endplates. Most striking features of axonal dystrophy are “dying-back” axonal degeneration with partial swellings (“spheroids” in matured type) which come to be most frequently in gracile nucleus, followed by in order of gracile fasciculus of cervical, thoracic and lumber cord levels. Immunocytochemical increase of glial fibrillary acidic protein (GFAP) and substance P (SP) is seen in reactive astrocytes and degenerating axons. Likewise, amyloid precursor protein (APP) and amyloid β-protein (AβP) activity become positive in axons and astrocytes along ascending tract. Moreover, ubiquitin-positive dot-like structures accumulate in gracile nucleus, spinocerebellar tract, and cerebellum in <italic>gad</italic> mice after 9<sup>th</sup>-week old. Ubiquitinated structures are localized in spheroids with a larger diameter than normal. The <italic>gad</italic> mutation is caused by an in-frame deletion including exon 7 and 8 of <italic>UCH-L1</italic> gene, encoding the ubiquitin c-terminal hydrolase (UCH) isozyme (UCH-L1) selectively expressed in nervous system and testis/ovary. The <italic>gad</italic> allele encodes a truncated UCH-L1 lacking a segment of 42 amino acids containing catalytic site. The evaluation as mouse models for Parkinson's and Alzheimer's diseases and the collapse of synapse-axon circulation around central nervous system from peripheral nervous system are discussed.</p>
</abstract>
Collapse
|
8
|
Yong Y, Gamage K, Cushman C, Spano A, Deppmann C. Regulation of degenerative spheroids after injury. Sci Rep 2020; 10:15472. [PMID: 32963272 PMCID: PMC7508847 DOI: 10.1038/s41598-020-71906-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 08/05/2020] [Indexed: 12/26/2022] Open
Abstract
Neuronal injury leads to rapid, programmed disintegration of axons distal to the site of lesion. Much like other forms of axon degeneration (e.g. developmental pruning, toxic insult from neurodegenerative disorder), Wallerian degeneration associated with injury is preceded by spheroid formation along axons. The mechanisms by which injury leads to formation of spheroids and whether these spheroids have a functional role in degeneration remain elusive. Here, using neonatal mouse primary sympathetic neurons, we investigate the roles of players previously implicated in the progression of Wallerian degeneration in injury-induced spheroid formation. We find that intra-axonal calcium flux is accompanied by actin-Rho dependent growth of calcium rich axonal spheroids that eventually rupture, releasing material to the extracellular space prior to catastrophic axon degeneration. Importantly, after injury, Sarm1-/- and DR6-/-, but not Wlds (excess NAD+) neurons, are capable of forming spheroids that eventually rupture, releasing their contents to the extracellular space to promote degeneration. Supplementation of exogenous NAD+ or expressing WLDs suppresses Rho-dependent spheroid formation and degeneration in response to injury. Moreover, injured or trophically deprived Sarm1-/- and DR6-/-, but not Wlds neurons, are resistant to degeneration induced by conditioned media collected from wild-type axons after spheroid rupture. Taken together, these findings place Rho-actin and NAD+ upstream of spheroid formation and may suggest that other mediators of degeneration, such as DR6 and SARM1, mediate post-spheroid rupture events that lead to catastrophic axon disassembly.
Collapse
Affiliation(s)
- Yu Yong
- Department of Biology, University of Virginia, Charlottesville, VA, 22904-4328, USA
| | - Kanchana Gamage
- Amgen, Massachusetts and Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Courtny Cushman
- Department of Neuroscience and Biomedical Engineering, University of Virginia, Charlottesville, VA, 22904-4328, USA
| | - Anthony Spano
- Department of Biology, University of Virginia, Charlottesville, VA, 22904-4328, USA
| | - Christopher Deppmann
- Department of Biology, University of Virginia, Charlottesville, VA, 22904-4328, USA.
- Department of Neuroscience and Biomedical Engineering, University of Virginia, Charlottesville, VA, 22904-4328, USA.
- Department of Cell Biology, University of Virginia, Charlottesville, VA, 22904-4328, USA.
| |
Collapse
|
9
|
Cimaglia G, Votruba M, Morgan JE, André H, Williams PA. Potential Therapeutic Benefit of NAD + Supplementation for Glaucoma and Age-Related Macular Degeneration. Nutrients 2020; 12:nu12092871. [PMID: 32961812 PMCID: PMC7551676 DOI: 10.3390/nu12092871] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/14/2020] [Accepted: 09/17/2020] [Indexed: 12/13/2022] Open
Abstract
Glaucoma and age-related macular degeneration are leading causes of irreversible blindness worldwide with significant health and societal burdens. To date, no clinical cures are available and treatments target only the manageable symptoms and risk factors (but do not remediate the underlying pathology of the disease). Both diseases are neurodegenerative in their pathology of the retina and as such many of the events that trigger cell dysfunction, degeneration, and eventual loss are due to mitochondrial dysfunction, inflammation, and oxidative stress. Here, we critically review how a decreased bioavailability of nicotinamide adenine dinucleotide (NAD; a crucial metabolite in healthy and disease states) may underpin many of these aberrant mechanisms. We propose how exogenous sources of NAD may become a therapeutic standard for the treatment of these conditions.
Collapse
Affiliation(s)
- Gloria Cimaglia
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, 112 82 Stockholm, Sweden;
- School of Optometry and Vision Sciences, Cardiff University, Cardiff CF24 4HQ, Wales, UK; (M.V.); (J.E.M.)
| | - Marcela Votruba
- School of Optometry and Vision Sciences, Cardiff University, Cardiff CF24 4HQ, Wales, UK; (M.V.); (J.E.M.)
- Cardiff Eye Unit, University Hospital Wales, Cardiff CF14 4XW, Wales, UK
| | - James E. Morgan
- School of Optometry and Vision Sciences, Cardiff University, Cardiff CF24 4HQ, Wales, UK; (M.V.); (J.E.M.)
- School of Medicine, Cardiff University, Cardiff CF14 4YS, Wales, UK
| | - Helder André
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, 112 82 Stockholm, Sweden;
- Correspondence: (H.A.); (P.A.W.)
| | - Pete A. Williams
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, 112 82 Stockholm, Sweden;
- Correspondence: (H.A.); (P.A.W.)
| |
Collapse
|
10
|
Abstract
PURPOSE OF REVIEW Diffuse or traumatic axonal injury is one of the principal pathologies encountered in traumatic brain injury (TBI) and the resulting axonal loss, disconnection, and brain atrophy contribute significantly to clinical morbidity and disability. The seminal discovery of the slow Wallerian degeneration mice (Wld) in which transected axons do not degenerate but survive and function independently for weeks has transformed concepts on axonal biology and raised hopes that axonopathies may be amenable to specific therapeutic interventions. Here we review mechanisms of axonal degeneration and also describe how these mechanisms may inform biological therapies of traumatic axonopathy in the context of TBI. RECENT FINDINGS In the last decade, SARM1 [sterile a and Toll/interleukin-1 receptor (TIR) motif containing 1] and the DLK (dual leucine zipper bearing kinase) and LZK (leucine zipper kinase) MAPK (mitogen-activated protein kinases) cascade have been established as the key drivers of Wallerian degeneration, a complex program of axonal self-destruction which is activated by a wide range of injurious insults, including insults that may otherwise leave axons structurally robust and potentially salvageable. Detailed studies on animal models and postmortem human brains indicate that this type of partial disruption is the main initial pathology in traumatic axonopathy. At the same time, the molecular dissection of Wallerian degeneration has revealed that the decision that commits axons to degeneration is temporally separated from the time of injury, a window that allows potentially effective pharmacological interventions. SUMMARY Molecular signals initiating and triggering Wallerian degeneration appear to be playing an important role in traumatic axonopathy and recent advances in understanding their nature and significance is opening up new therapeutic opportunities for TBI.
Collapse
|
11
|
Kline RA, Dissanayake KN, Hurtado ML, Martínez NW, Ahl A, Mole AJ, Lamont DJ, Court FA, Ribchester RR, Wishart TM, Murray LM. Altered mitochondrial bioenergetics are responsible for the delay in Wallerian degeneration observed in neonatal mice. Neurobiol Dis 2019; 130:104496. [PMID: 31176719 PMCID: PMC6704473 DOI: 10.1016/j.nbd.2019.104496] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 04/26/2019] [Accepted: 06/05/2019] [Indexed: 01/10/2023] Open
Abstract
Neurodegenerative and neuromuscular disorders can manifest throughout the lifespan of an individual, from infant to elderly individuals. Axonal and synaptic degeneration are early and critical elements of nearly all human neurodegenerative diseases and neural injury, however the molecular mechanisms which regulate this process are yet to be fully elucidated. Furthermore, how the molecular mechanisms governing degeneration are impacted by the age of the individual is poorly understood. Interestingly, in mice which are under 3 weeks of age, the degeneration of axons and synapses following hypoxic or traumatic injury is significantly slower. This process, known as Wallerian degeneration (WD), is a molecularly and morphologically distinct subtype of neurodegeneration by which axons and synapses undergo distinct fragmentation and death following a range of stimuli. In this study, we first use an ex-vivo model of axon injury to confirm the significant delay in WD in neonatal mice. We apply tandem mass-tagging quantitative proteomics to profile both nerve and muscle between P12 and P24 inclusive. Application of unbiased in silico workflows to relevant protein identifications highlights a steady elevation in oxidative phosphorylation cascades corresponding to the accelerated degeneration rate. We demonstrate that inhibition of Complex I prevents the axotomy-induced rise in reactive oxygen species and protects axons following injury. Furthermore, we reveal that pharmacological activation of oxidative phosphorylation significantly accelerates degeneration at the neuromuscular junction in neonatal mice. In summary, we reveal dramatic changes in the neuromuscular proteome during post-natal maturation of the neuromuscular system, and demonstrate that endogenous dynamics in mitochondrial bioenergetics during this time window have a functional impact upon regulating the stability of the neuromuscular system.
Collapse
Affiliation(s)
- Rachel A Kline
- Centre for Discovery Brain Science, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK; Euan McDonald Centre for Motor Neuron Disease Research, University of Edinburgh, UK; The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, EH25 9RG, UK
| | - Kosala N Dissanayake
- Euan McDonald Centre for Motor Neuron Disease Research, University of Edinburgh, UK; Centre for Cognitive and Neural Systems, University of Edinburgh, 1 George Square, Edinburgh EH8 9JZ, UK
| | - Maica Llavero Hurtado
- Euan McDonald Centre for Motor Neuron Disease Research, University of Edinburgh, UK; The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, EH25 9RG, UK
| | - Nicolás W Martínez
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
| | - Alexander Ahl
- Centre for Discovery Brain Science, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
| | - Alannah J Mole
- Centre for Discovery Brain Science, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK; Euan McDonald Centre for Motor Neuron Disease Research, University of Edinburgh, UK
| | - Douglas J Lamont
- Fingerprints Proteomics Facility, Dundee University, Dundee DD1 4HN, United Kingdom
| | - Felipe A Court
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile; Geroscience Center for Brain Health and Metabolism, Santiago, Chile; The Buck Institute for Research on Aging, Novato, CA, United States
| | - Richard R Ribchester
- Euan McDonald Centre for Motor Neuron Disease Research, University of Edinburgh, UK; Centre for Cognitive and Neural Systems, University of Edinburgh, 1 George Square, Edinburgh EH8 9JZ, UK
| | - Thomas M Wishart
- Euan McDonald Centre for Motor Neuron Disease Research, University of Edinburgh, UK; The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, EH25 9RG, UK
| | - Lyndsay M Murray
- Centre for Discovery Brain Science, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK; Euan McDonald Centre for Motor Neuron Disease Research, University of Edinburgh, UK.
| |
Collapse
|
12
|
Peters OM, Lewis EA, Osterloh JM, Weiss A, Salameh JS, Metterville J, Brown RH, Freeman MR. Loss of Sarm1 does not suppress motor neuron degeneration in the SOD1G93A mouse model of amyotrophic lateral sclerosis. Hum Mol Genet 2019; 27:3761-3771. [PMID: 30010873 PMCID: PMC6196650 DOI: 10.1093/hmg/ddy260] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 07/09/2018] [Indexed: 12/14/2022] Open
Abstract
Axon degeneration occurs in all neurodegenerative diseases, but the molecular pathways regulating axon destruction during neurodegeneration are poorly understood. Sterile Alpha and TIR Motif Containing 1 (Sarm1) is an essential component of the prodegenerative pathway driving axon degeneration after axotomy and represents an appealing target for therapeutic intervention in neurological conditions involving axon loss. Amyotrophic lateral sclerosis (ALS) is characterized by rapid, progressive motor neuron degeneration and muscle atrophy, causing paralysis and death. Patient tissue and animal models of ALS show destruction of upper and lower motor neuron cell bodies and loss of their associated axons. Here, we investigate whether loss of Sarm1 can mitigate motor neuron degeneration in the SOD1G93A mouse model of ALS. We found no change in survival, behavioral, electrophysiogical or histopathological outcomes in SOD1G93A mice null for Sarm1. Blocking Sarm1-mediated axon destruction alone is therefore not sufficient to suppress SOD1G93A-induced neurodegeneration. Our data suggest the molecular pathways driving axon loss in ALS may be Sarm1-independent or involve genetic pathways that act in a redundant fashion with Sarm1.
Collapse
Affiliation(s)
- Owen M Peters
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA, USA.,Department of Neurology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Elizabeth A Lewis
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Jeannette M Osterloh
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Alexandra Weiss
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Johnny S Salameh
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Jake Metterville
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Robert H Brown
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Marc R Freeman
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
13
|
Sasaki Y. Metabolic aspects of neuronal degeneration: From a NAD + point of view. Neurosci Res 2018; 139:9-20. [PMID: 30006197 DOI: 10.1016/j.neures.2018.07.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/21/2018] [Accepted: 06/22/2018] [Indexed: 12/14/2022]
Abstract
Cellular metabolism maintains the life of cells, allowing energy production required for building cellular constituents and maintaining homeostasis under constantly changing external environments. Neuronal cells maintain their structure and function for the entire life of organisms and the loss of neurons, with limited neurogenesis in adults, directly causes loss of complexity in the neuronal networks. The nervous system organizes the neurons by placing cell bodies containing nuclei of similar types of neurons in discrete regions. Accordingly, axons must travel great distances to connect different types of neurons and peripheral organs. The enormous surface area of neurons makes them high-energy demanding to keep their membrane potential. Distal axon survival is dependent on axonal transport that is another energy demanding process. All of these factors make metabolic stress a potential risk factor for neuronal death and neuronal degeneration often associated with metabolic diseases. This review discusses recent findings on metabolic dysregulations under neuronal degeneration and pathways protecting neurons in these conditions.
Collapse
Affiliation(s)
- Yo Sasaki
- Department of Genetics, Washington University in St. Louis, Couch Biomedical Research Building, 4515 McKinley Ave., Saint Louis, MO, 63110, United States
| |
Collapse
|
14
|
Sarm1 Deletion, but Not Wld S, Confers Lifelong Rescue in a Mouse Model of Severe Axonopathy. Cell Rep 2018; 21:10-16. [PMID: 28978465 PMCID: PMC5640801 DOI: 10.1016/j.celrep.2017.09.027] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 08/25/2017] [Accepted: 09/07/2017] [Indexed: 12/17/2022] Open
Abstract
Studies with the WldS mutant mouse have shown that axon and synapse pathology in several models of neurodegenerative diseases are mechanistically related to injury-induced axon degeneration (Wallerian degeneration). Crucially, an absence of SARM1 delays Wallerian degeneration as robustly as WldS, but their relative capacities to confer long-term protection against related, non-injury axonopathy and/or synaptopathy have not been directly compared. While Sarm1 deletion or WldS can rescue perinatal lethality and widespread Wallerian-like axonopathy in young NMNAT2-deficient mice, we report that an absence of SARM1 enables these mice to survive into old age with no overt phenotype, whereas those rescued by WldS invariantly develop a progressive neuromuscular defect in their hindlimbs from around 3 months of age. We therefore propose Sarm1 deletion as a more reliable tool than WldS for investigating Wallerian-like mechanisms in disease models and suggest that SARM1 blockade may have greater therapeutic potential than WLDS-related strategies. Rescue of an axonopathy model by Sarm1 deletion or WldS compared in an aging study Young adult NMNAT2-deficient mice rescued by WldS develop a hindlimb motor defect NMNAT2-deficient mice rescued by Sarm1 deletion are overtly normal up to 24 months SARM1 depletion/inhibition may have analytical and therapeutic advantages over WLDS
Collapse
|
15
|
Ubiquitin C-terminal hydrolase L1 (UCH-L1): structure, distribution and roles in brain function and dysfunction. Biochem J 2017; 473:2453-62. [PMID: 27515257 PMCID: PMC4980807 DOI: 10.1042/bcj20160082] [Citation(s) in RCA: 177] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2016] [Accepted: 04/29/2016] [Indexed: 12/13/2022]
Abstract
Ubiquitin C-terminal hydrolase L1 (UCH-L1) is an extremely abundant protein in the brain where, remarkably, it is estimated to make up 1–5% of total neuronal protein. Although it comprises only 223 amino acids it has one of the most complicated 3D knotted structures yet discovered. Beyond its expression in neurons UCH-L1 has only very limited expression in other healthy tissues but it is highly expressed in several forms of cancer. Although UCH-L1 is classed as a deubiquitinating enzyme (DUB) the direct functions of UCH-L1 remain enigmatic and a wide array of alternative functions has been proposed. UCH-L1 is not essential for neuronal development but it is absolutely required for the maintenance of axonal integrity and UCH-L1 dysfunction is implicated in neurodegenerative disease. Here we review the properties of UCH-L1, and how understanding its complex structure can provide new insights into its roles in neuronal function and pathology.
Collapse
|
16
|
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
|
17
|
Tagliaferro P, Burke RE. Retrograde Axonal Degeneration in Parkinson Disease. JOURNAL OF PARKINSONS DISEASE 2017; 6:1-15. [PMID: 27003783 PMCID: PMC4927911 DOI: 10.3233/jpd-150769] [Citation(s) in RCA: 181] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In spite of tremendous research efforts we have not yet achieved two of our principal therapeutic goals in the treatment of Parkinson’s disease (PD), to prevent its onward progression and to provide restoration of systems that have already been damaged by the time of diagnosis. There are many possible reasons for our inability to make progress. One possibility is that our efforts thus far may not have been directed towards the appropriate cellular compartments. Up until now research has been largely focused on the loss of neurons in the disease. Thus, neuroprotection approaches have been largely aimed at blocking mechanisms that lead to destruction of the neuronal cell body. Attempts to provide neurorestoration have been almost entirely focused on replacement of neurons. We herein review the evidence that the axonal component of diseased neuronal systems merit more of our attention. Evidence from imaging studies, from postmortem neurochemical studies, and from genetic animal models suggests that the axons of the dopaminergic system are involved predominantly and early in PD. Since the mechanisms of axonal destruction are distinct from those of neuron cell body degeneration, a focus on axonal neurobiology will offer new opportunities for preventing their degeneration. At present these mechanisms remain largely obscure. However, defining them is likely to offer new opportunities for neuroprotection. In relation to neurorestoration, while it has been classically believed that neurons of the adult central nervous system are incapable of new axon growth, recent evidence shows that this is not true for the dopaminergic projection. In conclusion, the neurobiology of axons is likely to offer many new approaches to protective and restorative therapeutics.
Collapse
Affiliation(s)
| | - Robert E Burke
- Department of Neurology, Columbia University Medical Center, New York, NY, USA.,Departments of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
18
|
Gerdts J, Summers DW, Milbrandt J, DiAntonio A. Axon Self-Destruction: New Links among SARM1, MAPKs, and NAD+ Metabolism. Neuron 2016; 89:449-60. [PMID: 26844829 PMCID: PMC4742785 DOI: 10.1016/j.neuron.2015.12.023] [Citation(s) in RCA: 251] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Wallerian axon degeneration is a form of programmed subcellular death that promotes axon breakdown in disease and injury. Active degeneration requires SARM1 and MAP kinases, including DLK, while the NAD+ synthetic enzyme NMNAT2 prevents degeneration. New studies reveal that these pathways cooperate in a locally mediated axon destruction program, with NAD+ metabolism playing a central role. Here, we review the biology of Wallerian-type axon degeneration and discuss the most recent findings, with special emphasis on critical signaling events and their potential as therapeutic targets for axonopathy.
Collapse
Affiliation(s)
- Josiah Gerdts
- Department of Genetics, Washington University School of Medicine in St. Louis, 660 Euclid Avenue, St. Louis, MO 63110, USA
| | - Daniel W Summers
- Department of Genetics, Washington University School of Medicine in St. Louis, 660 Euclid Avenue, St. Louis, MO 63110, USA; Department of Developmental Biology, Washington University School of Medicine in St. Louis, 660 Euclid Avenue, St. Louis, MO 63110, USA
| | - Jeffrey Milbrandt
- Department of Genetics, Washington University School of Medicine in St. Louis, 660 Euclid Avenue, St. Louis, MO 63110, USA; Hope Center for Neurological Disorders, Washington University School of Medicine in St. Louis, 660 Euclid Avenue, St. Louis, MO 63110, USA
| | - Aaron DiAntonio
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, 660 Euclid Avenue, St. Louis, MO 63110, USA; Hope Center for Neurological Disorders, Washington University School of Medicine in St. Louis, 660 Euclid Avenue, St. Louis, MO 63110, USA.
| |
Collapse
|
19
|
Liebert AD, Chow RT, Bicknell BT, Varigos E. Neuroprotective Effects Against POCD by Photobiomodulation: Evidence from Assembly/Disassembly of the Cytoskeleton. J Exp Neurosci 2016; 10:1-19. [PMID: 26848276 PMCID: PMC4737522 DOI: 10.4137/jen.s33444] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 12/09/2015] [Accepted: 12/15/2015] [Indexed: 02/07/2023] Open
Abstract
Postoperative cognitive dysfunction (POCD) is a decline in memory following anaesthesia and surgery in elderly patients. While often reversible, it consumes medical resources, compromises patient well-being, and possibly accelerates progression into Alzheimer's disease. Anesthetics have been implicated in POCD, as has neuroinflammation, as indicated by cytokine inflammatory markers. Photobiomodulation (PBM) is an effective treatment for a number of conditions, including inflammation. PBM also has a direct effect on microtubule disassembly in neurons with the formation of small, reversible varicosities, which cause neural blockade and alleviation of pain symptoms. This mimics endogenously formed varicosities that are neuroprotective against damage, toxins, and the formation of larger, destructive varicosities and focal swellings. It is proposed that PBM may be effective as a preconditioning treatment against POCD; similar to the PBM treatment, protective and abscopal effects that have been demonstrated in experimental models of macular degeneration, neurological, and cardiac conditions.
Collapse
Affiliation(s)
| | - Roberta T. Chow
- Brain and Mind Institute, University of Sydney, Sydney, NSW, Australia
| | | | | |
Collapse
|
20
|
Morales I, Sanchez A, Rodriguez-Sabate C, Rodriguez M. The degeneration of dopaminergic synapses in Parkinson's disease: A selective animal model. Behav Brain Res 2015; 289:19-28. [PMID: 25907749 DOI: 10.1016/j.bbr.2015.04.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 04/09/2015] [Accepted: 04/11/2015] [Indexed: 12/21/2022]
Abstract
Available evidence increasingly suggests that the degeneration of dopamine neurons in Parkinson's disease starts in the striatal axons and synaptic terminals. A selective procedure is described here to study the mechanisms involved in the striatal denervation of dopaminergic terminals. This procedure can also be used to analyze mechanisms involved in the dopaminergic re-innervation of the striatum, and the role of astrocytes and microglia in both processes. Adult Sprague-Dawley rats were injected in the lateral ventricles with increasing doses of 6-hydroxydopamine (12-50 μg), which generated a dose-dependent loss of dopaminergic synapses and axons in the striatum, followed by an axonal sprouting (weeks later) and by a progressive recovery of striatal dopaminergic synapses (months later). Both the degeneration and regeneration of the dopaminergic terminals were accompanied by astrogliosis. Because the experimental manipulations did not induce unspecific damage in the striatal tissue, this method could be particularly suitable to study the basic mechanisms involved in the distal degeneration and regeneration of dopaminergic nigrostriatal neurons, and the possible role of astrocytes and microglia in the dynamics of both processes.
Collapse
Affiliation(s)
- Ingrid Morales
- Laboratory of Neurobiology and Experimental Neurology, Department of Physiology Faculty of Medicine, University of La Laguna, La Laguna, Tenerife, Canary Islands, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Alberto Sanchez
- Laboratory of Neurobiology and Experimental Neurology, Department of Physiology Faculty of Medicine, University of La Laguna, La Laguna, Tenerife, Canary Islands, Spain
| | - Clara Rodriguez-Sabate
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Manuel Rodriguez
- Laboratory of Neurobiology and Experimental Neurology, Department of Physiology Faculty of Medicine, University of La Laguna, La Laguna, Tenerife, Canary Islands, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| |
Collapse
|
21
|
Wen J, Sun D, Tan J, Young W. A consistent, quantifiable, and graded rat lumbosacral spinal cord injury model. J Neurotrauma 2015; 32:875-92. [PMID: 25313633 PMCID: PMC4492780 DOI: 10.1089/neu.2013.3321] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The purpose of this study is to develop a rat lumbosacral spinal cord injury (SCI) model that causes consistent motoneuronal loss and behavior deficits. Most SCI models focus on the thoracic or cervical spinal cord. Lumbosacral SCI accounts for about one third of human SCI but no standardized lumbosacral model is available for evaluating therapies. Twenty-six adult female Sprague-Dawley rats were randomized to three groups: sham (n=9), 25 mm (n=8), and 50 mm (n=9). Sham rats had laminectomy only, while 25 mm and 50 mm rats were injured by dropping a 10 g rod from a height of 25 mm or 50 mm, respectively, onto the L4-5 spinal cord at the T13/L1 vertebral junction. We measured footprint length (FL), toe spreading (TS), intermediate toe spreading (ITS), and sciatic function index (SFI) from walking footprints, and static toe spreading (STS), static intermediate toe spreading (SITS), and static sciatic index (SSI) from standing footprints. At six weeks, we assessed neuronal and white matter loss, quantified axons, diameter, and myelin thickness in the peroneal and tibial nerves, and measured cross-sectional areas of tibialis anterior and gastrocnemius muscle fibers. The result shows that peroneal and tibial motoneurons were respectively distributed in 4.71 mm and 5.01 mm columns in the spinal cord. Dropping a 10-g weight from 25 mm or 50 mm caused 1.5 mm or 3.75 mm gaps in peroneal and tibial motoneuronal columns, respectively, and increased spinal cord white matter loss. Fifty millimeter contusions significantly increased FL and reduced TS, ITS, STS, SITS, SFI, and SSI more than 25 mm contusions, and resulted in smaller axon and myelinated axon diameters in tibial and peroneal nerves and greater atrophy of gastrocnemius and anterior tibialis muscles, than 25 mm contusions. This model of lumbosacral SCI produces consistent and graded loss of white matter, motoneuronal loss, peripheral nerve axonal changes, and anterior tibialis and gastrocnemius muscles atrophy in rats.
Collapse
Affiliation(s)
- Junxiang Wen
- 1 Department of Cell Biology and Neuroscience, Rutgers, the State University of New Jersey , Piscataway, New Jersey.,2 Department of Orthopaedics, Tongji University School of Medicine , Shanghai, China
| | - Dongming Sun
- 1 Department of Cell Biology and Neuroscience, Rutgers, the State University of New Jersey , Piscataway, New Jersey
| | - Jun Tan
- 2 Department of Orthopaedics, Tongji University School of Medicine , Shanghai, China
| | - Wise Young
- 1 Department of Cell Biology and Neuroscience, Rutgers, the State University of New Jersey , Piscataway, New Jersey
| |
Collapse
|
22
|
Wallerian degeneration: an emerging axon death pathway linking injury and disease. Nat Rev Neurosci 2014; 15:394-409. [DOI: 10.1038/nrn3680] [Citation(s) in RCA: 387] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
23
|
Axonal transport plays a crucial role in mediating the axon-protective effects of NmNAT. Neurobiol Dis 2014; 68:78-90. [PMID: 24787896 DOI: 10.1016/j.nbd.2014.04.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 04/06/2014] [Accepted: 04/21/2014] [Indexed: 12/29/2022] Open
Abstract
Deficits in axonal transport are thought to contribute to the pathology of many neurodegenerative diseases. Expressing the slow Wallerian degeneration protein (Wld(S)) or related nicotinamide mononucleotide adenyltransferases (NmNATs) protects axons against damage from a broad range of insults, but the ability of these proteins to protect against inhibition of axonal transport has received little attention. We set out to determine whether these proteins can protect the axons of cultured hippocampal neurons from damage due to hydrogen peroxide or oxygen-glucose deprivation (OGD) and, in particular, whether they can reduce the damage that these agents cause to the axonal transport machinery. Exposure to these insults inhibited the axonal transport of both mitochondria and of the vesicles that carry axonal membrane proteins; this inhibition occurred hours before the first signs of axonal degeneration. Expressing a cytoplasmically targeted version of NmNAT1 (cytNmNAT1) protected the axons against both insults. It also reduced the inhibition of transport when cells were exposed to hydrogen peroxide and enhanced the recovery of transport following both insults. The protective effects of cytNmNAT1 depend on mitochondrial transport. When mitochondrial transport was inhibited, cytNmNAT1 was unable to protect axons against either insult. The protective effects of mitochondrially targeted NmNAT also were blocked by inhibiting mitochondrial transport. These results establish that NmNAT robustly protects the axonal transport system following exposure to OGD and reactive oxygen species and may offer similar protection in other disease models. Understanding how NmNAT protects the axonal transport system may lead to new strategies for neuroprotection in neurodegenerative diseases.
Collapse
|
24
|
Ferraz Franco C, Santos R, Varela Coelho A. Proteolytic events are relevant cellular responses during nervous system regeneration of the starfish Marthasterias glacialis. J Proteomics 2014; 99:1-25. [DOI: 10.1016/j.jprot.2013.12.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 12/03/2013] [Accepted: 12/09/2013] [Indexed: 01/12/2023]
|
25
|
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
|
26
|
King AE, Southam KA, Dittmann J, Vickers JC. Excitotoxin-induced caspase-3 activation and microtubule disintegration in axons is inhibited by taxol. Acta Neuropathol Commun 2013; 1:59. [PMID: 24252213 PMCID: PMC3893530 DOI: 10.1186/2051-5960-1-59] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 09/01/2013] [Indexed: 11/29/2022] Open
Abstract
Background Axon degeneration, a key pathological event in many neurodegenerative diseases and injury, can be induced by somatodendritic excitotoxin exposure. It is currently unclear, however, whether excitotoxin-induced axon degeneration is mechanistically similar to Wallerian degeneration, which occurs following axon transection, but does not involve axonal caspase activation. Results We have used mouse primary cortical neurons at 9 days in vitro, in a compartmented culture model that allows separation of the axon from the soma, to examine the pathological cascade of excitotoxin-induced axon degeneration. Excitotoxicity induced by chronic exposure to kainic acid, resulted in axonal fragmentation, which was associated with activation of caspase-3 in the axonal compartment. To examine the role of microtubules in these events, the microtubule-stabilizing agent, taxol, was added to either the axonal or somatodendritic compartment. Our results demonstrated that microtubule stabilization of axons resulted in a significant reduction in the number of fragmented axons following excitotoxin exposure. Interestingly, taxol exposure to either the somatodendritic or axonal compartment resulted in reduced caspase-3 activation in axons, suggesting that caspase activation is a downstream event of microtubule destabilization and involves signalling from the cell soma. Conclusion These data suggest that excitotoxin-induced axon degeneration shows some mechanistic differences to Wallerian degeneration, and that microtubule stabilization may assist in protecting nerve cells from excitotoxic effects.
Collapse
|
27
|
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]
|
28
|
Milde S, Gilley J, Coleman MP. Subcellular localization determines the stability and axon protective capacity of axon survival factor Nmnat2. PLoS Biol 2013; 11:e1001539. [PMID: 23610559 PMCID: PMC3627647 DOI: 10.1371/journal.pbio.1001539] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 03/06/2013] [Indexed: 11/19/2022] Open
Abstract
Modulation of the subcellular localization of the endogenous axon survival factor Nmnat2 boosts its axon protective capacity, suggesting a novel approach to delaying axon degeneration in neurodegenerative disease. Axons require a constant supply of the labile axon survival factor Nmnat2 from their cell bodies to avoid spontaneous axon degeneration. Here we investigate the mechanism of fast axonal transport of Nmnat2 and its site of action for axon maintenance. Using dual-colour live-cell imaging of axonal transport in SCG primary culture neurons, we find that Nmnat2 is bidirectionally trafficked in axons together with markers of the trans-Golgi network and synaptic vesicles. In contrast, there is little co-migration with mitochondria, lysosomes, and active zone precursor vesicles. Residues encoded by the small, centrally located exon 6 are necessary and sufficient for stable membrane association and vesicular axonal transport of Nmnat2. Within this sequence, a double cysteine palmitoylation motif shared with GAP43 and surrounding basic residues are all required for efficient palmitoylation and stable association with axonal transport vesicles. Interestingly, however, disrupting this membrane association increases the ability of axonally localized Nmnat2 to preserve transected neurites in primary culture, while re-targeting the strongly protective cytosolic mutants back to membranes abolishes this increase. Larger deletions within the central domain including exon 6 further enhance Nmnat2 axon protective capacity to levels that exceed that of the slow Wallerian degeneration protein, WldS. The mechanism underlying the increase in axon protection appears to involve an increased half-life of the cytosolic forms, suggesting a role for palmitoylation and membrane attachment in Nmnat2 turnover. We conclude that Nmnat2 activity supports axon survival through a site of action distinct from Nmnat2 transport vesicles and that protein stability, a key determinant of axon protection, is enhanced by mutations that disrupt palmitoylation and dissociate Nmnat2 from these vesicles. Neurons are polarized cells that rely on bidirectional transport to deliver thousands of cargos between the cell body and the most distal ends of their axons. One cargo that is of particular importance is the NAD-synthesising enzyme Nmnat2. This surprisingly unstable protein is produced in the cell body and its constant supply into axons is required to keep them alive. If this supply is interrupted, Nmnat2 levels in the distal axon drop below a critical threshold, leading to axon degeneration. The rapid turnover of Nmnat2 contributes critically to the time course of axon degeneration. If its half-life could be extended, axons may be able to survive transient interruptions of its supply. In this study, we find that disruption of Nmnat2 localization to axonal transport vesicles increases both its half-life and its capacity to protect injured neurites. Specifically, association of Nmnat2 with transport vesicles reduces it stability by making it vulnerable to ubiquitination and proteasome-mediated degradation. These findings suggest that modulation of the subcellular localization of Nmnat2 on transport vesicles could serve as a potential avenue for therapeutic treatment of axon degeneration.
Collapse
Affiliation(s)
- Stefan Milde
- The Babraham Institute, Cambridge, United Kingdom
| | | | | |
Collapse
|
29
|
Höke A, Simpson DM, Freeman R. Challenges in developing novel therapies for peripheral neuropathies: a summary of The Foundation for Peripheral Neuropathy Scientific Symposium 2012. J Peripher Nerv Syst 2013; 18:1-6. [PMID: 23521637 DOI: 10.1111/jns5.12000] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
| | | | - Roy Freeman
- Department of Neurology, Harvard Medical School, Beth Israel Deaconess Medical Center; Boston MA USA
| |
Collapse
|
30
|
Rallis A, Lu B, Ng J. Molecular chaperones protect against JNK- and Nmnat-regulated axon degeneration in Drosophila. J Cell Sci 2012; 126:838-49. [PMID: 23264732 DOI: 10.1242/jcs.117259] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Axon degeneration is observed at the early stages of many neurodegenerative conditions and this often leads to subsequent neuronal loss. We previously showed that inactivating the c-Jun N-terminal kinase (JNK) pathway leads to axon degeneration in Drosophila mushroom body (MB) neurons. To understand this process, we screened candidate suppressor genes and found that the Wallerian degeneration slow (Wld(S)) protein blocked JNK axonal degeneration. Although the nicotinamide mononucleotide adenylyltransferase (Nmnat1) portion of Wld(S) is required, we found that its nicotinamide adenine dinucleotide (NAD(+)) enzyme activity and the Wld(S) N-terminus (N70) are dispensable, unlike axotomy models of neurodegeneration. We suggest that Wld(S)-Nmnat protects against axonal degeneration through chaperone activity. Furthermore, ectopically expressed heat shock proteins (Hsp26 and Hsp70) also protected against JNK and Nmnat degeneration phenotypes. These results suggest that molecular chaperones are key in JNK- and Nmnat-regulated axonal protective functions.
Collapse
Affiliation(s)
- Andrew Rallis
- MRC Centre for Developmental Neurobiology, King's College London, Guy's Campus, London SE1 1UL, UK.
| | | | | |
Collapse
|
31
|
Zhu X, Libby RT, de Vries WN, Smith RS, Wright DL, Bronson RT, Seburn KL, John SWM. Mutations in a P-type ATPase gene cause axonal degeneration. PLoS Genet 2012; 8:e1002853. [PMID: 22912588 PMCID: PMC3415440 DOI: 10.1371/journal.pgen.1002853] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 06/07/2012] [Indexed: 01/13/2023] Open
Abstract
Neuronal loss and axonal degeneration are important pathological features of many neurodegenerative diseases. The molecular mechanisms underlying the majority of axonal degeneration conditions remain unknown. To better understand axonal degeneration, we studied a mouse mutant wabbler-lethal (wl). Wabbler-lethal (wl) mutant mice develop progressive ataxia with pronounced neurodegeneration in the central and peripheral nervous system. Previous studies have led to a debate as to whether myelinopathy or axonopathy is the primary cause of neurodegeneration observed in wl mice. Here we provide clear evidence that wabbler-lethal mutants develop an axonopathy, and that this axonopathy is modulated by Wlds and Bax mutations. In addition, we have identified the gene harboring the disease-causing mutations as Atp8a2. We studied three wl alleles and found that all result from mutations in the Atp8a2 gene. Our analysis shows that ATP8A2 possesses phosphatidylserine translocase activity and is involved in localization of phosphatidylserine to the inner leaflet of the plasma membrane. Atp8a2 is widely expressed in the brain, spinal cord, and retina. We assessed two of the mutant alleles of Atp8a2 and found they are both nonfunctional for the phosphatidylserine translocase activity. Thus, our data demonstrate for the first time that mutation of a mammalian phosphatidylserine translocase causes axon degeneration and neurodegenerative disease. Axonal degeneration is an important pathological feature of many neurodegenerative diseases, such as Alzheimer disease, Parkinson's disease, and amyotrophic lateral sclerosis. In most of these disease conditions, molecular mechanisms of axonal degeneration remain largely unknown. Spontaneous mouse mutants are important in human disease studies. Identification of a disease-causing gene in mice can lead to the identification of the human ortholog as the disease gene in humans. This approach has the power to identify unexpected genes and pathways involved in disease. Our study centered on wabbler lethal (wl) mutant mice, which display axonal degeneration in both the central and peripheral nervous systems. We identified the disease-causing gene in mice with different wl mutations. The mutations are in Atp8a2, a gene encoding a phosphatidylserine translocase. This protein functions to keep phosphatidylserine enriched to the inner leaflet of the plasma membrane. Our study demonstrates a new role for phospholipid asymmetry in maintaining axon health, and it also reveals a novel function for phosphatidyleserine translocase in neurodegenerative diseases.
Collapse
Affiliation(s)
- Xianjun Zhu
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
- The Howard Hughes Medical Institute, Bar Harbor, Maine, United States of America
| | - Richard T. Libby
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Wilhelmine N. de Vries
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
- The Howard Hughes Medical Institute, Bar Harbor, Maine, United States of America
| | - Richard S. Smith
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
- The Howard Hughes Medical Institute, Bar Harbor, Maine, United States of America
| | - Dana L. Wright
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | | | - Kevin L. Seburn
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Simon W. M. John
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
- The Howard Hughes Medical Institute, Bar Harbor, Maine, United States of America
- Department of Ophthalmology, Tufts University of Medicine, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
32
|
Carpenter S, Soares H, Brandão O, Souto Moura C, Castro L, Rodrigues E, Cunha AL, Bartosch C. A novel type of familial proximal axonal dystrophy: three cases and a review of the axonal dystrophies. Eur J Paediatr Neurol 2012; 16:292-300. [PMID: 21925911 DOI: 10.1016/j.ejpn.2011.08.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Revised: 08/24/2011] [Accepted: 08/28/2011] [Indexed: 01/09/2023]
Abstract
Three related infants of Roma ancestry, two of them siblings, showed hypotonia, predominantly axial, from birth, difficulty swallowing, myoclonic seizures, and respiratory difficulty. Dysmorphic features, principally micrognathia were present. EEGs showed focal epileptiform abnormalities. All three died in their 5th month from respiratory insufficiency complicated by pneumonia. Autopsy showed small brains without malformation. Microscopy revealed numerous axonal spheroids involving particularly the brain stem and spinal cord, with especial prominence in the middle cerebellar peduncle, the anterior part of the thalamic reticular nuclei, and the anterior horns and columns of the spinal cord. Spheroids that appeared to be on axons of lower motor neurons were especially large. No spheroids were seen in peripheral nerves; electron microscopy did not show spheroids in skin. By electron microscopy spheroids contained neurofilaments, sparse mitochondria, and electron dense granules. The material did not allow identification of microtubules. Closely packed vesicles excluded neurofilamanets from the center of many spheroids, especially in the middle cerebellar peduncle. Sprouting of axons from the surface of many spheroids was seen. This disease is distinct from the well described type of infantile neuroaxonal dystrophy (Seitelberger's disease) in view of the distribution of spheroids, presence of spheroids on proximal rather than distal parts of axons, sparing of the peripheral nerves, lack of staining for synuclein, presence of sprouting, and lack of membranous profiles in the spheroids. A review of reported types of axonal dystrophy has not shown identical cases.
Collapse
Affiliation(s)
- Stirling Carpenter
- Department of Anatomic Pathology, Hospital São Joâo, Alameda Professor Hernani Monteiro, Porto 4200, Portugal.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
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
|
34
|
Antenor-Dorsey JAV, O'Malley KL. WldS but not Nmnat1 protects dopaminergic neurites from MPP+ neurotoxicity. Mol Neurodegener 2012; 7:5. [PMID: 22315973 PMCID: PMC3322348 DOI: 10.1186/1750-1326-7-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Accepted: 02/08/2012] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND The WldS mouse mutant ("Wallerian degeneration-slow") delays axonal degeneration in a variety of disorders including in vivo models of Parkinson's disease. The mechanisms underlying WldS -mediated axonal protection are unclear, although many studies have attributed WldS neuroprotection to the NAD+-synthesizing Nmnat1 portion of the fusion protein. Here, we used dissociated dopaminergic cultures to test the hypothesis that catalytically active Nmnat1 protects dopaminergic neurons from toxin-mediated axonal injury. RESULTS Using mutant mice and lentiviral transduction of dopaminergic neurons, the present findings demonstrate that WldS but not Nmnat1, Nmnat3, or cytoplasmically-targeted Nmnat1 protects dopamine axons from the parkinsonian mimetic N-methyl-4-phenylpyridinium (MPP+). Moreover, NAD+ synthesis is not required since enzymatically-inactive WldS still protects. In addition, NAD+ by itself is axonally protective and together with WldS is additive in the MPP+ model. CONCLUSIONS Our data suggest that NAD+ and WldS act through separate and possibly parallel mechanisms to protect dopamine axons. As MPP+ is thought to impair mitochondrial function, these results suggest that WldS might be involved in preserving mitochondrial health or maintaining cellular metabolism.
Collapse
Affiliation(s)
- Jo Ann V Antenor-Dorsey
- Department of Anatomy and Neurobiology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | | |
Collapse
|
35
|
Kessell AE, Finnie JW, Blumbergs PC, Manavis J, Jerrett IV. Neuroaxonal dystrophy in Australian Merino lambs. J Comp Pathol 2011; 147:62-72. [PMID: 22055697 DOI: 10.1016/j.jcpa.2011.09.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2011] [Revised: 09/12/2011] [Accepted: 09/24/2011] [Indexed: 01/16/2023]
Abstract
Neuroaxonal dystrophy (NAD) is a morphological abnormality in man and animals that is characterized by the occurrence of numerous axonal swellings (spheroids) in the nervous system. NAD has been described in Suffolk lambs in the USA, Merino lambs in Australia and several breeds of sheep in New Zealand. This paper describes the clinicopathological changes of only the second occurrence of NAD reported in Merino lambs. There were some features (myelin loss, gliosis and visual impairment) in these Australian cases that have not been reported previously in ovine NAD. Application of immunohistochemical markers of axonal transport suggested that disruption of this transport mechanism contributed to spheroid development.
Collapse
Affiliation(s)
- A E Kessell
- School of Animal and Veterinary Science, Charles Sturt University, Wagga Wagga, New South Wales, Australia
| | | | | | | | | |
Collapse
|
36
|
Ljungberg MC, Ali YO, Zhu J, Wu CS, Oka K, Zhai RG, Lu HC. CREB-activity and nmnat2 transcription are down-regulated prior to neurodegeneration, while NMNAT2 over-expression is neuroprotective, in a mouse model of human tauopathy. Hum Mol Genet 2011; 21:251-67. [PMID: 22027994 DOI: 10.1093/hmg/ddr492] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Tauopathies, characterized by neurofibrillary tangles (NFTs) of phosphorylated tau proteins, are a group of neurodegenerative diseases, including frontotemporal dementia and both sporadic and familial Alzheimer's disease. Forebrain-specific over-expression of human tau(P301L), a mutation associated with frontotemporal dementia with parkinsonism linked to chromosome 17, in rTg4510 mice results in the formation of NFTs, learning and memory impairment and massive neuronal death. Here, we show that the mRNA and protein levels of NMNAT2 (nicotinamide mononucleotide adenylyltransferase 2), a recently identified survival factor for maintaining neuronal health in peripheral nerves, are reduced in rTg4510 mice prior to the onset of neurodegeneration or cognitive deficits. Two functional cAMP-response elements (CREs) were identified in the nmnat2 promoter region. Both the total amount of phospho-CRE binding protein (CREB) and the pCREB bound to nmnat2 CRE sites in the cortex and the hippocampus of rTg4510 mice are significantly reduced, suggesting that NMNAT2 is a direct target of CREB under physiological conditions and that tau(P301L) overexpression down-regulates CREB-mediated transcription. We found that over-expressing NMNAT2 or its homolog NMNAT1, but not NMNAT3, in rTg4510 hippocampi from 6 weeks of age using recombinant adeno-associated viral vectors significantly reduced neurodegeneration caused by tau(P301L) over-expression at 5 months of age. In summary, our studies strongly support a protective role of NMNAT2 in the mammalian central nervous system. Decreased endogenous NMNAT2 function caused by reduced CREB signaling during pathological insults may be one of underlying mechanisms for neuronal death in tauopathies.
Collapse
Affiliation(s)
- M Cecilia Ljungberg
- The Cain Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Differential protection of neuromuscular sensory and motor axons and their endings in Wld(S) mutant mice. Neuroscience 2011; 200:142-58. [PMID: 22062136 DOI: 10.1016/j.neuroscience.2011.10.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Revised: 10/10/2011] [Accepted: 10/12/2011] [Indexed: 11/21/2022]
Abstract
Orthograde Wallerian degeneration normally brings about fragmentation of peripheral nerve axons and their sensory or motor endings within 24-48 h in mice. However, neuronal expression of the chimaeric, Wld(S) gene mutation extends survival of functioning axons and their distal endings for up to 3 weeks after nerve section. Here we studied the pattern and rate of degeneration of sensory axons and their annulospiral endings in deep lumbrical muscles of Wld(S) mice, and compared these with motor axons and their terminals, using neurone-specific transgenic expression of the fluorescent proteins yellow fluorescent protein (YFP) or cyan fluorescent protein (CFP) as morphological reporters. Surprisingly, sensory endings were preserved for up to 20 days, at least twice as long as the most resilient motor nerve terminals. Protection of sensory endings and axons was also much less sensitive to Wld(S) gene-copy number or age than motor axons and their endings. Protection of γ-motor axons and their terminals innervating the juxtaequatorial and polar regions of the spindles was less than sensory axons but greater than α-motor axons. The differences between sensory and motor axon protection persisted in electrically silent, organotypic nerve-explant cultures suggesting that residual axonal activity does not contribute to the sensory-motor axon differences in vivo. Quantitative, Wld(S)-specific immunostaining of dorsal root ganglion (DRG) neurones and motor neurones in homozygous Wld(S) mice suggested that the nuclei of large DRG neurones contain about 2.4 times as much Wld(S) protein as motor neurones. By contrast, nuclear fluorescence of DRG neurones in homozygotes was only 1.5 times brighter than in heterozygotes stained under identical conditions. Thus, differences in axonal or synaptic protection within the same Wld(S) mouse may most simply be explained by differences in expression level of Wld(S) protein between neurones. Mimicry of Wld(S)-induced protection may also have applications in treatment of neurotoxicity or peripheral neuropathies in which the integrity of sensory endings may be especially implicated.
Collapse
|
38
|
Lichtenbergová L, Lassmann H, Jones MK, Kolářová L, Horák P. Trichobilharzia regenti: host immune response in the pathogenesis of neuroinfection in mice. Exp Parasitol 2011; 128:328-35. [PMID: 21554878 DOI: 10.1016/j.exppara.2011.04.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Revised: 04/18/2011] [Accepted: 04/26/2011] [Indexed: 10/18/2022]
Abstract
Besides their natural bird hosts, Trichobilharzia regenti cercariae are able to penetrate skin of mammals, including humans. Experimental infections of mice showed that schistosomula of this species are able to avoid the immune response in skin of their non-specific mammalian host and escape the skin to migrate to the CNS. Schistosomula do not mature in mammals, but can survive in nervous tissue for several days post infection. Neuroinfections of specific bird hosts as well as accidental mammalian hosts can lead to neuromotor effects, for example, leg paralysis and thus this parasite serves as a model of parasite invasion of the CNS. Here, we show by histological and immunohistochemical investigation of CNS invasion of immunocompetent (BALB/c) and immunodeficient (SCID) mice by T. regenti schistosomula that the presence of parasites in the nervous tissue initiated an influx of immune cells, activation of microglia, astrocytes and development of inflammatory lesions. Schistosomula elimination in the tissue depended on the host immune status. In the absence of CD3+ T-cells in immunodeficient SCID mice, parasite destruction was slower than that in immunocompetent BALB/c mice. Axon injury and subsequent secondary demyelination in the CNS were associated with mechanical damage due to migration of schistosomula through the nervous tissue, and not by host immune processes. Immunoreactivity of the parasite intestinal content for specific antigens of oligodendrocytes/myelin and neurofilaments showed for the first time that schistosomula ingest the nervous tissue components during their migration.
Collapse
Affiliation(s)
- Lucie Lichtenbergová
- Department of Parasitology, Faculty of Science, Charles University in Prague, Viničná 7, 128 44 Prague 2, Czech Republic.
| | | | | | | | | |
Collapse
|
39
|
Comley LH, Wishart TM, Baxter B, Murray LM, Nimmo A, Thomson D, Parson SH, Gillingwater TH. Induction of cell stress in neurons from transgenic mice expressing yellow fluorescent protein: implications for neurodegeneration research. PLoS One 2011; 6:e17639. [PMID: 21408118 PMCID: PMC3050905 DOI: 10.1371/journal.pone.0017639] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Accepted: 02/04/2011] [Indexed: 11/30/2022] Open
Abstract
Background Mice expressing fluorescent proteins in neurons are one of the most powerful tools in modern neuroscience research and are increasingly being used for in vivo studies of neurodegeneration. However, these mice are often used under the assumption that the fluorescent proteins present are biologically inert. Methodology/Principal Findings Here, we show that thy1-driven expression of yellow fluorescent protein (YFP) in neurons triggers multiple cell stress responses at both the mRNA and protein levels in vivo. The presence of YFP in neurons also subtly altered neuronal morphology and modified the time-course of dying-back neurodegeneration in experimental axonopathy, but not in Wallerian degeneration triggered by nerve injury. Conclusions/Significance We conclude that fluorescent protein expressed in thy1-YFP mice is not biologically inert, modifies molecular and cellular characteristics of neurons in vivo, and has diverse and unpredictable effects on neurodegeneration pathways.
Collapse
Affiliation(s)
- Laura H. Comley
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, Lothian, United Kingdom
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, Lothian, United Kingdom
| | - Thomas M. Wishart
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, Lothian, United Kingdom
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, Lothian, United Kingdom
| | - Becki Baxter
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, Lothian, United Kingdom
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, Lothian, United Kingdom
| | - Lyndsay M. Murray
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, Lothian, United Kingdom
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, Lothian, United Kingdom
| | - Ailish Nimmo
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, Lothian, United Kingdom
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, Lothian, United Kingdom
| | - Derek Thomson
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, Lothian, United Kingdom
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, Lothian, United Kingdom
| | - Simon H. Parson
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, Lothian, United Kingdom
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, Lothian, United Kingdom
| | - Thomas H. Gillingwater
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, Lothian, United Kingdom
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, Lothian, United Kingdom
- * E-mail:
| |
Collapse
|
40
|
Frisardi G, Chessa G, Barone S, Paoli A, Razionale A, Frisardi F. Integration of 3D anatomical data obtained by CT imaging and 3D optical scanning for computer aided implant surgery. BMC Med Imaging 2011; 11:5. [PMID: 21338504 PMCID: PMC3047291 DOI: 10.1186/1471-2342-11-5] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2010] [Accepted: 02/21/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND A precise placement of dental implants is a crucial step to optimize both prosthetic aspects and functional constraints. In this context, the use of virtual guiding systems has been recognized as a fundamental tool to control the ideal implant position. In particular, complex periodontal surgeries can be performed using preoperative planning based on CT data. The critical point of the procedure relies on the lack of accuracy in transferring CT planning information to surgical field through custom-made stereo-lithographic surgical guides. METHODS In this work, a novel methodology is proposed for monitoring loss of accuracy in transferring CT dental information into periodontal surgical field. The methodology is based on integrating 3D data of anatomical (impression and cast) and preoperative (radiographic template) models, obtained by both CT and optical scanning processes. RESULTS A clinical case, relative to a fully edentulous jaw patient, has been used as test case to assess the accuracy of the various steps concurring in manufacturing surgical guides. In particular, a surgical guide has been designed to place implants in the bone structure of the patient. The analysis of the results has allowed the clinician to monitor all the errors, which have been occurring step by step manufacturing the physical templates. CONCLUSIONS The use of an optical scanner, which has a higher resolution and accuracy than CT scanning, has demonstrated to be a valid support to control the precision of the various physical models adopted and to point out possible error sources. A case study regarding a fully edentulous patient has confirmed the feasibility of the proposed methodology.
Collapse
|
41
|
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
|
42
|
Barrientos SA, Martinez NW, Yoo S, Jara JS, Zamorano S, Hetz C, Twiss JL, Alvarez J, Court FA. Axonal degeneration is mediated by the mitochondrial permeability transition pore. J Neurosci 2011; 31:966-78. [PMID: 21248121 PMCID: PMC3245862 DOI: 10.1523/jneurosci.4065-10.2011] [Citation(s) in RCA: 168] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Revised: 10/27/2010] [Accepted: 11/01/2010] [Indexed: 01/23/2023] Open
Abstract
Axonal degeneration is an active process that has been associated with neurodegenerative conditions triggered by mechanical, metabolic, infectious, toxic, hereditary and inflammatory stimuli. This degenerative process can cause permanent loss of function, so it represents a focus for neuroprotective strategies. Several signaling pathways are implicated in axonal degeneration, but identification of an integrative mechanism for this self-destructive process has remained elusive. Here, we show that rapid axonal degeneration triggered by distinct mechanical and toxic insults is dependent on the activation of the mitochondrial permeability transition pore (mPTP). Both pharmacological and genetic targeting of cyclophilin D, a functional component of the mPTP, protects severed axons and vincristine-treated neurons from axonal degeneration in ex vivo and in vitro mouse and rat model systems. These effects were observed in axons from both the peripheral and central nervous system. Our results suggest that the mPTP is a key effector of axonal degeneration, upon which several independent signaling pathways converge. Since axonal and synapse degeneration are increasingly considered early pathological events in neurodegeneration, our work identifies a potential target for therapeutic intervention in a wide variety of conditions that lead to loss of axons and subsequent functional impairment.
Collapse
Affiliation(s)
- Sebastian A. Barrientos
- Department of Physiology, Faculty of Biology, Catholic University of Chile, Santiago 8331150, Chile
| | - Nicolas W. Martinez
- Department of Physiology, Faculty of Biology, Catholic University of Chile, Santiago 8331150, Chile
| | - Soonmoon Yoo
- Nemours Biomedical Research Institute, Alfred I. duPont Hospital for Children, Wilmington, Delaware 19716
| | - Juan S. Jara
- Department of Physiology, Faculty of Biology, Catholic University of Chile, Santiago 8331150, Chile
| | - Sebastian Zamorano
- Institute of Biomedical Sciences, Center for Molecular Studies of the Cell and Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - Claudio Hetz
- Institute of Biomedical Sciences, Center for Molecular Studies of the Cell and Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
- Department of Immunology and Infectious Diseases, Harvard University School of Public Health, Boston, Massachusetts 02115
- NeuroUnion Biomedical Foundation, Santiago 7630614, Chile, and
| | - Jeffery L. Twiss
- Department of Biology, Drexel University, Philadelphia, Pennsylvania 19104
| | - Jaime Alvarez
- Department of Physiology, Faculty of Biology, Catholic University of Chile, Santiago 8331150, Chile
| | - Felipe A. Court
- Department of Physiology, Faculty of Biology, Catholic University of Chile, Santiago 8331150, Chile
- NeuroUnion Biomedical Foundation, Santiago 7630614, Chile, and
| |
Collapse
|
43
|
An assessment of mechanisms underlying peripheral axonal degeneration caused by aminoacyl-tRNA synthetase mutations. Mol Cell Neurosci 2010; 46:432-43. [PMID: 21115117 DOI: 10.1016/j.mcn.2010.11.006] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2010] [Revised: 10/26/2010] [Accepted: 11/16/2010] [Indexed: 01/09/2023] Open
Abstract
Mutations in glycyl-, tyrosyl-, and alanyl-tRNA synthetases (GARS, YARS and AARS respectively) cause autosomal dominant Charcot-Marie-Tooth disease, and mutations in Gars cause a similar peripheral neuropathy in mice. Aminoacyl-tRNA synthetases (ARSs) charge amino acids onto their cognate tRNAs during translation; however, the pathological mechanism(s) of ARS mutations remains unclear. To address this, we tested possible mechanisms using mouse models. First, amino acid mischarging was discounted by examining the recessive "sticky" mutation in alanyl-tRNA synthetase (Aars(sti)), which causes cerebellar neurodegeneration through a failure to efficiently correct mischarging of tRNA(Ala). Aars(sti/sti) mice do not have peripheral neuropathy, and they share no phenotypic features with the Gars mutant mice. Next, we determined that the Wallerian Degeneration Slow (Wlds) mutation did not alter the Gars phenotype. Therefore, no evidence for misfolding of GARS itself or other proteins was found. Similarly, there were no indications of general insufficiencies in protein synthesis caused by Gars mutations based on yeast complementation assays. Mutant GARS localized differently than wild type GARS in transfected cells, but a similar distribution was not observed in motor neurons derived from wild type mouse ES cells, and there was no evidence for abnormal GARS distribution in mouse tissue. Both GARS and YARS proteins were present in sciatic axons and Schwann cells from Gars mutant and control mice, consistent with a direct role for tRNA synthetases in peripheral nerves. Unless defects in translation are in some way restricted to peripheral axons, as suggested by the axonal localization of GARS and YARS, we conclude that mutations in tRNA synthetases are not causing peripheral neuropathy through amino acid mischarging or through a defect in their known function in translation.
Collapse
|
44
|
Abstract
Axon and synapse degeneration are common components of many neurodegenerative diseases, and their rescue is essential for effective neuroprotection. The chimeric Wallerian degeneration slow protein (Wld(S)) protects axons dose dependently, but its mechanism is still elusive. We recently showed that Wld(S) acts at a non-nuclear location and is present in axons. This and other recent reports support a model in which Wld(S) protects by extranuclear redistribution of its nuclear NMNAT1 portion. However, it remains unclear whether cytoplasmic NMNAT1 acts locally in axons and synapses or at a non-nuclear site within cell bodies. The potency of axon protection by non-nuclear NMNAT1 relative to Wld(S) also needs to be established in vivo. Because the N-terminal portion of Wld(S) (N70) localized to axons, we hypothesized that it mediates the trafficking of the NMNAT1 portion. To test this, we substituted N70 with an axonal targeting peptide derived from amyloid precursor protein, and fused this to NMNAT1 with disrupted nuclear targeting. In transgenic mice, this transformed NMNAT1 from a molecule unable to inhibit Wallerian degeneration, even at high expression levels, into a protein more potent than Wld(S), able to preserve injured axons for several weeks at undetectable expression levels. Preventing NMNAT1 axonal delivery abolished its protective effect. Axonally targeted NMNAT1 localized to vesicular structures, colocalizing with extranuclear Wld(S), and was cotransported at least partially with mitochondria. We conclude that axonal targeting of NMNAT activity is both necessary and sufficient to delay Wallerian degeneration, and that promoting axonal and synaptic delivery greatly enhances the effectiveness.
Collapse
|
45
|
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]
|
46
|
Cheng HC, Ulane CM, Burke RE. Clinical progression in Parkinson disease and the neurobiology of axons. Ann Neurol 2010; 67:715-25. [PMID: 20517933 DOI: 10.1002/ana.21995] [Citation(s) in RCA: 704] [Impact Index Per Article: 46.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Despite tremendous growth in recent years in our knowledge of the molecular basis of Parkinson disease (PD) and the molecular pathways of cell injury and death, we remain without therapies that forestall disease progression. Although there are many possible explanations for this lack of success, one is that experimental therapeutics to date have not adequately focused on an important component of the disease process, that of axon degeneration. It remains unknown what neuronal compartment, either the soma or the axon, is involved at disease onset, although some have proposed that it is the axons and their terminals that take the initial brunt of injury. Nevertheless, this concept has not been formally incorporated into many of the current theories of disease pathogenesis, and it has not achieved a wide consensus. More importantly, in view of growing evidence that the molecular mechanisms of axon degeneration are separate and distinct from the canonical pathways of programmed cell death that mediate soma destruction, the possibility of early involvement of axons in PD has not been adequately emphasized as a rationale to explore the neurobiology of axons for novel therapeutic targets. We propose that ongoing degeneration of axons, not cell bodies, is the primary determinant of clinically apparent progression of disease, and that future experimental therapeutics intended to forestall disease progression will benefit from a new focus on the distinct mechanisms of axon degeneration.
Collapse
Affiliation(s)
- Hsiao-Chun Cheng
- Department of Neurology, Columbia University Medical Center, New York, NY 10032, USA
| | | | | |
Collapse
|
47
|
Sun F, Cavalli V. Neuroproteomics approaches to decipher neuronal regeneration and degeneration. Mol Cell Proteomics 2010; 9:963-75. [PMID: 20019051 PMCID: PMC2871427 DOI: 10.1074/mcp.r900003-mcp200] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Revised: 12/14/2009] [Indexed: 01/02/2023] Open
Abstract
Given the complexity of brain and nerve tissues, systematic approaches are essential to understand normal physiological conditions and functional alterations in neurological diseases. Mass spectrometry-based proteomics is increasingly used in neurosciences to determine both basic and clinical differential protein expression, protein-protein interactions, and post-translational modifications. Proteomics approaches are especially useful to understand the mechanisms of nerve regeneration and degeneration because changes in axons following injury or in disease states often occur without the contribution of transcriptional events in the cell body. Indeed, the current understanding of axonal function in health and disease emphasizes the role of proteolysis, local axonal protein synthesis, and a broad range of post-translational modifications. Deciphering how axons regenerate and degenerate has thus become a postgenomics problem, which depends in part on proteomics approaches. This review focuses on recent proteomics approaches designed to uncover the mechanisms and molecules involved in neuronal regeneration and degeneration. It emerges that the principal degenerative mechanisms converge to oxidative stress, dysfunctions of axonal transport, mitochondria, chaperones, and the ubiquitin-proteasome systems. The mechanisms regulating nerve regeneration also impinge on axonal transport, cytoskeleton, and chaperones in addition to changes in signaling pathways. We also discuss the major challenges to proteomics work in the nervous system given the complex organization of the brain and nerve tissue at the anatomical, cellular, and subcellular levels.
Collapse
Affiliation(s)
- Faneng Sun
- From the Department of Anatomy and Neurobiology, Washington University, St. Louis, Missouri 63110
| | - Valeria Cavalli
- From the Department of Anatomy and Neurobiology, Washington University, St. Louis, Missouri 63110
| |
Collapse
|
48
|
Mechanisms of Axonal Spheroid Formation in Central Nervous System Wallerian Degeneration. J Neuropathol Exp Neurol 2010; 69:455-72. [DOI: 10.1097/nen.0b013e3181da84db] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
49
|
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
|
50
|
Cheng HC, Burke RE. The Wld(S) mutation delays anterograde, but not retrograde, axonal degeneration of the dopaminergic nigro-striatal pathway in vivo. J Neurochem 2010; 113:683-91. [PMID: 20132467 DOI: 10.1111/j.1471-4159.2010.06632.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
For many neurodegenerative disorders, such as Parkinson's disease, there is evidence that the disease first affects axons and terminals of neurons that are selectively vulnerable. This would suggest that it may be possible to forestall progression by targeting the cellular mechanisms of axon degeneration. While it is now clear that these mechanisms are distinct from the pathways of programmed cell death, they are less well known. Compelling evidence of the distinctiveness of these mechanisms has derived from studies of the Wld(S) mutation, which confers resistance to axon degeneration. Little is known about how this mutation affects degeneration in dopaminergic axons, those that are affected in Parkinson's disease. We have characterized the Wld(S) phenotype in these axons in four models of injury: two that utilize the neurotoxin 6-hydroxydopamine or axotomy to induce anterograde degeneration, and two that use these methods to induce retrograde degeneration. For both 6-hydroxydopamine and axotomy, Wld(S) provides protection from anterograde, but not retrograde degeneration. This protection is observed as preserved immunostaining for tyrosine hydroxylase in axons and striatum, and by structural integrity visualized by GFP in tyrosine hydroxylase-GFP mice. Therefore, Wld(S) offers axon protection, but it reveals fundamentally different processes underlying antero- and retrograde degeneration in this system.
Collapse
Affiliation(s)
- Hsiao-Chun Cheng
- Department of Neurology, The College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | | |
Collapse
|