Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 169
Filter
1.
Annu Rev Genet ; 55: 93-113, 2021 11 23.
Article in English | MEDLINE | ID: mdl-34351802

ABSTRACT

Significant advances have been made in recent years in identifying the genetic components of Wallerian degeneration, the process that brings the progressive destruction and removal of injured axons. It has now been accepted that Wallerian degeneration is an active and dynamic cellular process that is well regulated at molecular and cellular levels. In this review, we describe our current understanding of Wallerian degeneration, focusing on the molecular players and mechanisms that mediate the injury response, activate the degenerative program, transduce the death signal, execute the destruction order, and finally, clear away the debris. By highlighting the starring roles and sketching out the molecular script of Wallerian degeneration, we hope to provide a useful framework to understand Wallerian and Wallerian-like degeneration and to lay a foundation for developing new therapeutic strategies to treat axon degeneration in neural injury as well as in neurodegenerative disease.


Subject(s)
Neurodegenerative Diseases , Wallerian Degeneration , Axons/pathology , Axons/physiology , Humans , Neurodegenerative Diseases/pathology , Wallerian Degeneration/genetics , Wallerian Degeneration/pathology
2.
Cell Death Dis ; 12(1): 104, 2021 01 20.
Article in English | MEDLINE | ID: mdl-33473103

ABSTRACT

While the consequences of nuclear DNA damage have been well studied, the exact consequences of acute and selective mitochondrial DNA (mtDNA) damage are less understood. DNA damaging chemotherapeutic drugs are known to activate p53-dependent apoptosis in response to sustained nuclear DNA damage. While it is recognized that whole-cell exposure to these drugs also damages mtDNA, the specific contribution of mtDNA damage to cellular degeneration is less clear. To examine this, we induced selective mtDNA damage in neuronal axons using microfluidic chambers that allow for the spatial and fluidic isolation of neuronal cell bodies (containing nucleus and mitochondria) from the axons (containing mitochondria). Exposure of the DNA damaging drug cisplatin selectively to only the axons induced mtDNA damage in axonal mitochondria, without nuclear damage. We found that this resulted in the selective degeneration of only the targeted axons that were exposed to DNA damage, where ROS was induced but mitochondria were not permeabilized. mtDNA damage-induced axon degeneration was not mediated by any of the three known axon degeneration pathways: apoptosis, axon pruning, and Wallerian degeneration, as Bax-deficiency, or Casp3-deficiency, or Sarm1-deficiency failed to protect the degenerating axons. Strikingly, p53, which is essential for degeneration after nuclear DNA damage, was also not required for degeneration induced with mtDNA damage. This was most evident when the p53-deficient neurons were globally exposed to cisplatin. While the cell bodies of p53-deficient neurons were protected from degeneration in this context, the axons farthest from the cell bodies still underwent degeneration. These results highlight how whole cell exposure to DNA damage activates two pathways of degeneration; a faster, p53-dependent apoptotic degeneration that is triggered in the cell bodies with nuclear DNA damage, and a slower, p53-independent degeneration that is induced with mtDNA damage.


Subject(s)
DNA Damage , DNA, Mitochondrial/metabolism , Neurons/metabolism , Tumor Suppressor Protein p53/metabolism , Wallerian Degeneration/metabolism , Cell Nucleus/genetics , Cell Nucleus/metabolism , DNA, Mitochondrial/genetics , Humans , Wallerian Degeneration/genetics
3.
Sci Rep ; 10(1): 21889, 2020 12 14.
Article in English | MEDLINE | ID: mdl-33318563

ABSTRACT

Cisplatin is a commonly used chemotherapy agent with significant dose-limiting neurotoxicity resulting in peripheral neuropathy. Although it is postulated that formation of DNA-platinum adducts is responsible for both its cytotoxicity in cancer cells and side effects in neurons, downstream mechanisms that lead to distal axonal degeneration are unknown. Here we show that activation of calpains is required for both neurotoxicity and formation of DNA-platinum adduct formation in neurons but not in cancer cells. Furthermore, we show that neurotoxicity of cisplatin requires activation of Sarm1, a key regulator of Wallerian degeneration, as mice lacking the Sarm1 gene do not develop peripheral neuropathy as evaluated by both behavioral or pathological measures. These findings indicate that Sarm1 and/or specific calpain inhibitors could be developed to prevent cisplatin induced peripheral neuropathy.


Subject(s)
Armadillo Domain Proteins/metabolism , Calpain/metabolism , Cisplatin/adverse effects , Cytoskeletal Proteins/metabolism , Neurotoxicity Syndromes/metabolism , Animals , Armadillo Domain Proteins/genetics , Calpain/genetics , Cells, Cultured , Cisplatin/pharmacology , Cytoskeletal Proteins/genetics , Enzyme Activation/drug effects , Enzyme Activation/genetics , Mice , Mice, Knockout , Neurotoxicity Syndromes/genetics , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/genetics , Peripheral Nervous System Diseases/metabolism , Rats , Rats, Sprague-Dawley , Wallerian Degeneration/chemically induced , Wallerian Degeneration/genetics , Wallerian Degeneration/metabolism
4.
Mol Neurobiol ; 57(10): 4232-4249, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32696431

ABSTRACT

Peripheral nerve injuries result in motor and sensory dysfunction which can be recovered by compensatory or regenerative processes. In situations where axonal regeneration of injured neurons is hampered, compensation by collateral sprouting from uninjured neurons contributes to target reinnervation and functional recovery. Interestingly, this process of collateral sprouting from uninjured neurons has been associated with the activation of growth-associated programs triggered by Wallerian degeneration. Nevertheless, the molecular alterations at the transcriptomic level associated with these compensatory growth mechanisms remain to be fully elucidated. We generated a surgical model of partial sciatic nerve injury in mice to mechanistically study degeneration-induced collateral sprouting from spared fibers in the peripheral nervous system. Using next-generation sequencing and Ingenuity Pathway Analysis, we described the sprouting-associated transcriptome of uninjured sensory neurons and compare it with the activated by regenerating neurons. In vitro approaches were used to functionally assess sprouting gene candidates in the mechanisms of axonal growth. Using a novel animal model, we provide the first description of the sprouting transcriptome observed in uninjured sensory neurons after nerve injury. This collateral sprouting-associated transcriptome differs from that seen in regenerating neurons, suggesting a molecular program distinct from axonal growth. We further demonstrate that genetic upregulation of novel sprouting-associated genes activates a specific growth program in vitro, leading to increased neuronal branching. These results contribute to our understanding of the molecular mechanisms associated with collateral sprouting in vivo. The data provided here will therefore be instrumental in developing therapeutic strategies aimed at promoting functional recovery after injury to the nervous system.


Subject(s)
Gene Expression Profiling , Neurogenesis/genetics , Peripheral Nerves/physiology , Sensory Receptor Cells/physiology , Transcriptome/genetics , Animals , Cell Proliferation , Female , Ganglia, Spinal/pathology , Gene Expression Regulation , Lumbar Vertebrae/pathology , Mice, Inbred C57BL , Myelin Sheath/metabolism , Peripheral Nerve Injuries/genetics , Peripheral Nerve Injuries/pathology , Peripheral Nerves/ultrastructure , Sciatic Nerve/metabolism , Sciatic Nerve/pathology , Sensory Receptor Cells/ultrastructure , Wallerian Degeneration/genetics , Wallerian Degeneration/pathology
5.
Commun Biol ; 3(1): 49, 2020 01 30.
Article in English | MEDLINE | ID: mdl-32001778

ABSTRACT

Protecting the nervous system from chronic effects of physical and chemical stress is a pressing clinical challenge. The obligate pro-degenerative protein Sarm1 is essential for Wallerian axon degeneration. Thus, blocking Sarm1 function is emerging as a promising neuroprotective strategy with therapeutic relevance. Yet, the conditions that will most benefit from inhibiting Sarm1 remain undefined. Here we combine genome engineering, pharmacology and high-resolution intravital videmicroscopy in zebrafish to show that genetic elimination of Sarm1 increases Schwann-cell resistance to toxicity by diverse chemotherapeutic agents after axonal injury. Synthetic degradation of Sarm1-deficient axons reversed this effect, suggesting that glioprotection is a non-autonomous effect of delayed axon degeneration. Moreover, loss of Sarm1 does not affect macrophage recruitment to nerve-wound microenvironment, injury resolution, or neural-circuit repair. These findings anticipate that interventions aimed at inhibiting Sarm1 can counter heightened glial vulnerability to chemical stressors and may be an effective strategy to reduce chronic consequences of neurotrauma.


Subject(s)
Antineoplastic Agents/adverse effects , Armadillo Domain Proteins/deficiency , Axons/metabolism , Schwann Cells/drug effects , Schwann Cells/metabolism , Wallerian Degeneration/genetics , Animals , Animals, Genetically Modified , Armadillo Domain Proteins/genetics , Axons/pathology , Fluorescent Antibody Technique , Genetic Loci , Mutagenesis , Phenotype , Zebrafish
6.
Exp Neurol ; 320: 112961, 2019 10.
Article in English | MEDLINE | ID: mdl-31136762

ABSTRACT

The three nicotinamide mononucleotide adenylyltransferase (NMNAT) family members synthesize the electron carrier nicotinamide adenine dinucleotide (NAD+) and are essential for cellular metabolism. In mammalian axons, NMNAT activity appears to be required for axon survival and is predominantly provided by NMNAT2. NMNAT2 has recently been shown to also function as a chaperone to aid in the refolding of misfolded proteins. Nmnat2 deficiency in mice, or in its ortholog dNmnat in Drosophila, results in axon outgrowth and survival defects. Peripheral nerve axons in NMNAT2-deficient mice fail to extend and innervate targets, and skeletal muscle is severely underdeveloped. In addition, removing NMNAT2 from established axons initiates axon death by Wallerian degeneration. We report here on two stillborn siblings with fetal akinesia deformation sequence (FADS), severely reduced skeletal muscle mass and hydrops fetalis. Clinical exome sequencing identified compound heterozygous NMNAT2 variant alleles in both cases. Both protein variants are incapable of supporting axon survival in mouse primary neuron cultures when overexpressed. In vitro assays demonstrate altered protein stability and/or defects in NAD+ synthesis and chaperone functions. Thus, both patient NMNAT2 alleles are null or severely hypo-morphic. These data indicate a previously unknown role for NMNAT2 in human neurological development and provide the first direct molecular evidence to support the involvement of Wallerian degeneration in a human axonal disorder. SIGNIFICANCE: Nicotinamide Mononucleotide Adenylyltransferase 2 (NMNAT2) both synthesizes the electron carrier Nicotinamide Adenine Dinucleotide (NAD+) and acts a protein chaperone. NMNAT2 has emerged as a major neuron survival factor. Overexpression of NMNAT2 protects neurons from Wallerian degeneration after injury and declining levels of NMNAT2 have been implicated in neurodegeneration. While the role of NMNAT2 in neurodegeneration has been extensively studied, the role of NMNAT2 in human development remains unclear. In this work, we present the first human variants in NMNAT2 identified in two fetuses with severe skeletal muscle hypoplasia and fetal akinesia. Functional studies in vitro showed that the mutations impair both NMNAT2 NAD+ synthase and chaperone functions. This work identifies the critical role of NMNAT2 in human development.


Subject(s)
Arthrogryposis/genetics , Neurogenesis/genetics , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Wallerian Degeneration/genetics , Animals , Fetus , Humans , Mice , Mutation , Stillbirth
7.
Exp Neurol ; 320: 112958, 2019 10.
Article in English | MEDLINE | ID: mdl-31132363

ABSTRACT

We identified a homozygous missense mutation in the gene encoding NAD synthesizing enzyme NMNAT2 in two siblings with childhood onset polyneuropathy with erythromelalgia. No additional homozygotes for this rare allele, which leads to amino acid substitution T94M, were present among the unaffected relatives tested or in the 60,000 exomes of the ExAC database. For axons to survive, axonal NMNAT2 activity has to be maintained above a threshold level but the T94M mutation confers a partial loss of function both in the ability of NMNAT2 to support axon survival and in its enzymatic properties. Electrophysiological tests and histological analysis of sural nerve biopsies in the patients were consistent with loss of distal sensory and motor axons. Thus, it is likely that NMNAT2 mutation causes this pain and axon loss phenotype making this the first disorder associated with mutation of a key regulator of Wallerian-like axon degeneration in humans. This supports indications from numerous animal studies that the Wallerian degeneration pathway is important in human disease and raises important questions about which other human phenotypes could be linked to this gene.


Subject(s)
Erythromelalgia/genetics , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Polyneuropathies/genetics , Female , Homozygote , Humans , Mutation, Missense , Pedigree , Siblings , Wallerian Degeneration/genetics
8.
Biochem Biophys Res Commun ; 514(4): 1037-1039, 2019 07 05.
Article in English | MEDLINE | ID: mdl-31097218

ABSTRACT

Axonal degeneration occurs in patients with various neurological diseases and traumatic nerve injuries, and Wallerian degeneration is a phenomenon in the prototypical axonal degradation that is observed after injury. Collapsin response mediator protein 2 (CRMP2) is phosphorylated by glycogen synthase kinase 3ß (GSK3ß), and it is involved in Wallerian degeneration after optic nerve injury. We previously developed a CRMP2 knock-in (CRMP2 KI) mouse line, in which CRMP2 phosphorylation by GSK3ß is inhibited; however, Wallerian degeneration in CRMP2 KI mice has not yet been examined. In this study, we examined whether Wallerian degeneration of the optic nerve is suppressed in CRMP2 KI mice. Using one eye removal model, we compared Wallerian degeneration of the optic nerve based on histological and biochemical analyses. Our experimental results indicated that the genetic inhibition of CRMP2 phosphorylation delays Wallerian degeneration after optic nerve injury.


Subject(s)
Intercellular Signaling Peptides and Proteins/genetics , Nerve Tissue Proteins/genetics , Optic Nerve Injuries/genetics , Wallerian Degeneration/genetics , Animals , Disease Models, Animal , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Phosphorylation/drug effects , Phosphorylation/genetics , Semaphorin-3A/pharmacology
9.
Mol Cell Biochem ; 453(1-2): 187-196, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30191480

ABSTRACT

NLRX1, the mitochondrial NOD-like receptor (NLR), modulates apoptosis in response to both intrinsic and extrinsic cues. Insights into the mechanism of how NLRX1 influences apoptosis remain to be determined. Here, we demonstrate that NLRX1 associates with SARM1, a protein with a toll/interleukin-1 receptor (TIR)-containing domain also found in adaptor proteins downstream of toll-like receptors, such as MyD88. While a direct role of SARM1 in innate immunity is unclear, the protein plays essential roles in Wallerian degeneration (WD), a type of neuronal catabolism occurring following axonal severing or damage. In non-neuronal cells, we found that endogenous SARM1 was equally distributed in the cytosol and the mitochondrial matrix, where association with NLRX1 occurred. In these cells, the apoptotic role of NLRX1 was fully dependent on SARM1, indicating that SARM1 was downstream of NLRX1 in apoptosis regulation. In primary murine neurons, however, Wallerian degeneration induced by vinblastine or NGF deprivation occurred in SARM1- yet NLRX1-independent manner, suggesting that WD requires the cytosolic pool of SARM1 or that NLRX1 levels in neurons are too low to contribute to WD regulation. Together, these results shed new light into the mechanisms through which NLRX1 controls apoptosis and provides evidence of a new link between NLR and TIR-containing proteins.


Subject(s)
Apoptosis , Armadillo Domain Proteins/immunology , Axons/immunology , Cytoskeletal Proteins/immunology , Immunity, Innate , Mitochondria/immunology , Mitochondrial Proteins/immunology , Animals , Armadillo Domain Proteins/genetics , Axons/pathology , Cytoskeletal Proteins/genetics , HEK293 Cells , Humans , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Proteins/genetics , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Vinblastine/adverse effects , Vinblastine/pharmacology , Wallerian Degeneration/chemically induced , Wallerian Degeneration/genetics , Wallerian Degeneration/immunology , Wallerian Degeneration/pathology
10.
Cell Death Dis ; 9(11): 1116, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30389906

ABSTRACT

Apoptotic cells expose Phosphatidylserine (PS), that serves as an "eat me" signal for engulfing cells. Previous studies have shown that PS also marks degenerating axonsduring developmental pruning or in response to insults (Wallerian degeneration), but the pathways that control PS exposure on degenerating axons are largely unknown. Here, we used a series of in vitro assays to systematically explore the regulation of PS exposure during axonal degeneration. Our results show that PS exposure is regulated by the upstream activators of axonal pruning and Wallerian degeneration. However, our investigation of signaling further downstream revealed divergence between axon degeneration and PS exposure. Importantly, elevation of the axonal energetic status hindered PS exposure, while inhibition of mitochondrial activity caused PS exposure, without degeneration. Overall, our results suggest that the levels of PS on the outer axonal membrane can be dissociated from the degeneration process and that the axonal energetic status plays a key role in the regulation of PS exposure.


Subject(s)
Ganglia, Spinal/drug effects , Neuronal Plasticity/drug effects , Phosphatidylserines/pharmacology , Sensory Receptor Cells/drug effects , Wallerian Degeneration/metabolism , Adenosine Triphosphate/biosynthesis , Animals , Apoptosis/drug effects , Apoptosis/genetics , Armadillo Domain Proteins/deficiency , Armadillo Domain Proteins/genetics , Axotomy , Biomarkers/metabolism , Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/genetics , Embryo, Mammalian , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Gene Expression , Mice , Mice, Knockout , Microfluidic Analytical Techniques , Nerve Growth Factor/pharmacology , Neuronal Plasticity/genetics , Phosphatidylserines/metabolism , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/pathology , Tissue Culture Techniques , Vincristine/pharmacology , Wallerian Degeneration/genetics , bcl-2-Associated X Protein/deficiency , bcl-2-Associated X Protein/genetics
11.
Sci Rep ; 8(1): 11179, 2018 07 25.
Article in English | MEDLINE | ID: mdl-30046125

ABSTRACT

Peripheral nerve injury can result in the decreased quality of life and bring us economic burden on society and individuals. Wallerian degeneration (WD) is critical for nerve degeneration and regeneration, but the mechanisms of WD are still elusive. Here, we report the effect of Toll-like receptor 4 (TLR4) on cultured Schwann cells (SCs) in vitro. The data showed that TLR4 expression was up-regulated after sciatic nerve injury of rat. TLR4 was expressed in cultured SCs. Enhanced or silenced expression of TLR4 affected SC proliferation, migration, apoptosis and relative gene expression. Furthermore, altered expression of TLR4 resulted in expression changes in c-Jun, ERK and catenin but not AKT and c-Fos pathways in SCs. These results suggested that TLR4 may be an important effective target in peripheral nerve degeneration and/or regeneration during WD in future investigations.


Subject(s)
Peripheral Nerve Injuries/genetics , Sciatic Neuropathy/genetics , Toll-Like Receptor 4/genetics , Wallerian Degeneration/genetics , Animals , Apoptosis/genetics , Cell Movement/genetics , Cells, Cultured , Gene Expression Regulation/genetics , Humans , Nerve Regeneration/genetics , Peripheral Nerve Injuries/physiopathology , Rats , Schwann Cells/metabolism , Schwann Cells/pathology , Sciatic Nerve/metabolism , Sciatic Nerve/physiopathology , Sciatic Neuropathy/physiopathology , Signal Transduction/genetics , Wallerian Degeneration/physiopathology
12.
Transgenic Res ; 27(2): 135-153, 2018 04.
Article in English | MEDLINE | ID: mdl-29453733

ABSTRACT

Bone marrow mononuclear cells (BMMC) constitute a heterogeneous population with potential to promote tissue regeneration. For this reason, this cell fraction has recently become a therapeutic alternative to mesenchymal stem cells, as culture is not required and phenotypic transformations can be hence avoided. In this work, and in order to attain long-lasting cell labeling and study longer survival times, we used BMMC isolated from adult transgenic rats expressing GFP to reproduce our wild type model and evaluate their remyelination ability in a reversible model of Wallerian degeneration. RT-PCR and flow cytometry analysis confirmed that cells isolated from the transgenic strain exhibited similar expression levels of markers specific to multipotent progenitors (CD34, CD90 and CD105) and Schwann cells (MPZ, MBP, S100ß and p75NTR) compared to wild type BMMC. BMMC expressing GFP retained their migration capacity, arriving exclusively at the injured nerve. Most importantly, and as detected through long-lasting cell tracking, some of these BMMC settled in the demyelinated area, mingled with endogenous cells, underwent phenotypic changes and colocalized with Schwann cell markers MBP and S100ß. Also worth highlighting, transgenic BMMC replicated wild type BMMC effects in terms of MBP organization and levels. On the basis of these findings, BMMC isolated from transgenic animals constitute a useful tool to evaluate their role in peripheral nervous system demyelination-remyelination and the underlying mechanisms.


Subject(s)
Bone Marrow Transplantation , Cell Tracking/methods , Green Fluorescent Proteins/genetics , Remyelination/genetics , Animals , Animals, Genetically Modified , Bone Marrow Cells/ultrastructure , Cell Lineage/genetics , Flow Cytometry , Gene Expression Regulation/genetics , Humans , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Rats , Schwann Cells/metabolism , Schwann Cells/ultrastructure , Transgenes/genetics , Wallerian Degeneration/genetics , Wallerian Degeneration/pathology
13.
Proc Natl Acad Sci U S A ; 115(6): 1358-1363, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29295933

ABSTRACT

Genetic studies of Wallerian degeneration have led to the identification of signaling molecules (e.g., dSarm/Sarm1, Axundead, and Highwire) that function locally in axons to drive degeneration. Here we identify a role for the Drosophila C2H2 zinc finger transcription factor Pebbled [Peb, Ras-responsive element binding protein 1 (RREB1) in mammals] in axon death. Loss of Peb in Drosophila glutamatergic sensory neurons results in either complete preservation of severed axons, or an axon death phenotype where axons fragment into large, continuous segments, rather than completely disintegrate. Peb is expressed in developing and mature sensory neurons, suggesting it is required to establish or maintain their competence to undergo axon death. peb mutant phenotypes can be rescued by human RREB1, and they exhibit dominant genetic interactions with dsarm mutants, linking peb/RREB1 to the axon death signaling cascade. Surprisingly, Peb is only able to fully block axon death signaling in glutamatergic, but not cholinergic sensory neurons, arguing for genetic diversity in axon death signaling programs in different neuronal subtypes. Our findings identify a transcription factor that regulates axon death signaling, and peb mutant phenotypes of partial fragmentation reveal a genetically accessible step in axon death signaling.


Subject(s)
Axons/pathology , Drosophila Proteins/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Wallerian Degeneration/pathology , Animals , Animals, Genetically Modified , Armadillo Domain Proteins/genetics , Armadillo Domain Proteins/metabolism , Axons/metabolism , Cholinergic Neurons/pathology , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics , Wallerian Degeneration/genetics , Wallerian Degeneration/metabolism , Wings, Animal/innervation , Wings, Animal/metabolism , Zinc Fingers/genetics
14.
J Neurosci ; 38(6): 1351-1365, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29279307

ABSTRACT

Injury to the peripheral axons of sensory neurons strongly enhances the regeneration of their central axons in the spinal cord. It remains unclear on what molecules that initiate such conditioning effect. Because ATP is released extracellularly by nerve and other tissue injury, we hypothesize that injection of ATP into a peripheral nerve might mimic the stimulatory effect of nerve injury on the regenerative state of the primary sensory neurons. We found that a single injection of 6 µl of 150 µm ATP into female rat sciatic nerve quadrupled the number of axons growing into a lesion epicenter in spinal cord after a concomitant dorsal column transection. A second boost ATP injection 1 week after the first one markedly reinforced the stimulatory effect of a single injection. Single ATP injection increased expression of phospho-STAT3 and GAP43, two markers of regenerative activity, in sensory neurons. Double ATP injections sustained the activation of phospho-STAT3 and GAP43, which may account for the marked axonal growth across the lesion epicenter. Similar studies performed on P2X7 or P2Y2 receptor knock-out mice indicate P2Y2 receptors are involved in the activation of STAT3 after ATP injection or conditioning lesion, whereas P2X7 receptors are not. Injection of ATP at 150 µm caused little Wallerian degeneration and behavioral tests showed no significant long-term adverse effects on sciatic nerve functions. The results in this study reveal possible mechanisms underlying the stimulation of regenerative programs and suggest a practical strategy for stimulating axonal regeneration following spinal cord injury.SIGNIFICANCE STATEMENT Injury of peripheral axons of sensory neurons has been known to strongly enhance the regeneration of their central axons in the spinal cord. In this study, we found that injection of ATP into a peripheral nerve can mimic the effect of peripheral nerve injury and significantly increase the number of sensory axons growing across lesion epicenter in the spinal cord. ATP injection increased expression of several markers for regenerative activity in sensory neurons, including phospho-STAT3 and GAP43. ATP injection did not cause significant long-term adverse effects on the functions of the injected nerve. These results may lead to clinically applicable strategies for enhancing neuronal responses that support regeneration of injured axons.


Subject(s)
Adenosine Triphosphate/pharmacology , Axons/drug effects , Nerve Regeneration/drug effects , Neurons/drug effects , Sensory Receptor Cells/drug effects , Spinal Cord/drug effects , Adenosine Triphosphate/administration & dosage , Animals , Behavior, Animal , Female , GAP-43 Protein/biosynthesis , GAP-43 Protein/genetics , Injections , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Peripheral Nerve Injuries/genetics , Peripheral Nerve Injuries/pathology , Rats , Receptors, Purinergic P2X7/genetics , Receptors, Purinergic P2Y2/genetics , STAT3 Transcription Factor/biosynthesis , STAT3 Transcription Factor/genetics , Sciatic Nerve , Spinal Cord Injuries/pathology , Wallerian Degeneration/genetics , Wallerian Degeneration/physiopathology
15.
Cell Rep ; 21(1): 10-16, 2017 Oct 03.
Article in English | MEDLINE | ID: mdl-28978465

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.


Subject(s)
Armadillo Domain Proteins/genetics , Cytoskeletal Proteins/genetics , Genes, Lethal , Muscular Atrophy/genetics , Nerve Tissue Proteins/genetics , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Wallerian Degeneration/genetics , Animals , Armadillo Domain Proteins/antagonists & inhibitors , Armadillo Domain Proteins/deficiency , Axons/metabolism , Axons/pathology , Cytoskeletal Proteins/antagonists & inhibitors , Cytoskeletal Proteins/deficiency , Disease Models, Animal , Female , Gene Deletion , Gene Expression Regulation , Hindlimb/innervation , Hindlimb/metabolism , Hindlimb/pathology , Humans , Locomotion , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Muscular Atrophy/prevention & control , Nerve Tissue Proteins/deficiency , Nicotinamide-Nucleotide Adenylyltransferase/deficiency , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Time Factors , Wallerian Degeneration/metabolism , Wallerian Degeneration/pathology , Wallerian Degeneration/prevention & control
16.
Neuron ; 95(1): 78-91.e5, 2017 Jul 05.
Article in English | MEDLINE | ID: mdl-28683272

ABSTRACT

Axon degeneration is a hallmark of neurodegenerative disease and neural injury. Axotomy activates an intrinsic pro-degenerative axon death signaling cascade involving loss of the NAD+ biosynthetic enzyme Nmnat/Nmnat2 in axons, activation of dSarm/Sarm1, and subsequent Sarm-dependent depletion of NAD+. Here we identify Axundead (Axed) as a mediator of axon death. axed mutants suppress axon death in several types of axons for the lifespan of the fly and block the pro-degenerative effects of activated dSarm in vivo. Neurodegeneration induced by loss of the sole fly Nmnat ortholog is also fully blocked by axed, but not dsarm, mutants. Thus, pro-degenerative pathways activated by dSarm signaling or Nmnat elimination ultimately converge on Axed. Remarkably, severed axons morphologically preserved by axon death pathway mutations remain integrated in circuits and able to elicit complex behaviors after stimulation, indicating that blockade of axon death signaling results in long-term functional preservation of axons.


Subject(s)
Armadillo Domain Proteins/genetics , Axons/metabolism , Cytoskeletal Proteins/genetics , Drosophila Proteins/genetics , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Wallerian Degeneration/genetics , Animals , Animals, Genetically Modified , Armadillo Domain Proteins/metabolism , Arthropod Antennae/injuries , Arthropod Antennae/innervation , Axotomy , Behavior, Animal , Blotting, Western , Cell Line , Cytoskeletal Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster , Grooming , Immunity, Active , NAD/metabolism , Neurons/metabolism , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Optogenetics , Wallerian Degeneration/metabolism , Wings, Animal/injuries , Wings, Animal/innervation
17.
Mol Genet Genomics ; 292(4): 909-922, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28447196

ABSTRACT

Many forms of neurodegenerative disease are characterized by Wallerian degeneration, an active program of axonal destruction. Recently, the important player which enacts Wallerian degeneration was discovered, the multidomain protein SARM1. Since the SARM1 protein has classically been thought of as an innate immune molecule, its role in Wallerian degeneration has raised questions on the evolutionary forces acting on it. Here, we synthesize a picture of SARM1's evolution through various organisms by examining the molecular and genetic changes of SARM1 and the genes around it. Using proteins that possess domains homologous to SARM1, we established distances and Ka/Ks values through 5671 pairwise species-species comparisons. We demonstrate that SARM1 diverged across species in a pattern similar to other SAM domain-containing proteins. This is surprising, because it was expected that SARM1 would behave more like its TIR domain relatives. Going along with this divorce from TIR, we also noted that SARM1's TIR is under stronger purifying selection than the rest of the TIR domain-containing proteins (remaining highly conserved). In addition, SARM1's synteny analysis reveals that the surrounding gene cluster is highly conserved, functioning as a potential nexus of gene functionality across species. Taken together, SARM1 demonstrates a unique evolutionary pattern, separate from the TIR domain protein family.


Subject(s)
Armadillo Domain Proteins/genetics , Cytoskeletal Proteins/genetics , Eagles/genetics , Esocidae/genetics , Horses/genetics , Wallerian Degeneration/genetics , Amino Acid Sequence/genetics , Animals , Axons/pathology , Base Composition/genetics , Biological Evolution , Databases, Genetic
18.
Curr Biol ; 27(6): 784-794, 2017 Mar 20.
Article in English | MEDLINE | ID: mdl-28262487

ABSTRACT

Axons require the axonal NAD-synthesizing enzyme NMNAT2 to survive. Injury or genetically induced depletion of NMNAT2 triggers axonal degeneration or defective axon growth. We have previously proposed that axonal NMNAT2 primarily promotes axon survival by maintaining low levels of its substrate NMN rather than generating NAD; however, this is still debated. NMN deamidase, a bacterial enzyme, shares NMN-consuming activity with NMNAT2, but not NAD-synthesizing activity, and it delays axon degeneration in primary neuronal cultures. Here we show that NMN deamidase can also delay axon degeneration in zebrafish larvae and in transgenic mice. Like overexpressed NMNATs, NMN deamidase reduces NMN accumulation in injured mouse sciatic nerves and preserves some axons for up to three weeks, even when expressed at a low level. Remarkably, NMN deamidase also rescues axonal outgrowth and perinatal lethality in a dose-dependent manner in mice lacking NMNAT2. These data further support a pro-degenerative effect of accumulating NMN in axons in vivo. The NMN deamidase mouse will be an important tool to further probe the mechanisms underlying Wallerian degeneration and its prevention.


Subject(s)
Amidohydrolases/genetics , Axons/pathology , Nerve Degeneration/genetics , Nicotinamide-Nucleotide Adenylyltransferase/deficiency , Wallerian Degeneration/genetics , Amidohydrolases/metabolism , Animals , Mice , Mice, Transgenic , Nerve Degeneration/metabolism , Wallerian Degeneration/metabolism
19.
Curr Biol ; 27(6): 890-896, 2017 Mar 20.
Article in English | MEDLINE | ID: mdl-28285993

ABSTRACT

Axon degeneration during development is required to sculpt a functional nervous system and is also a hallmark of pathological insult, such as injury [1, 2]. Despite similar morphological characteristics, very little overlap in molecular mechanisms has been reported between pathological and developmental degeneration [3-5]. In the peripheral nervous system (PNS), developmental axon pruning relies on receptor-mediated extrinsic degeneration mechanisms to determine which axons are maintained or degenerated [5-7]. Receptors have not been implicated in Wallerian axon degeneration; instead, axon autonomous, intrinsic mechanisms are thought to be the primary driver for this type of axon disintegration [8-10]. Here we survey the role of neuronally expressed, paralogous tumor necrosis factor receptor super family (TNFRSF) members in Wallerian degeneration. We find that an orphan receptor, death receptor 6 (DR6), is required to drive axon degeneration after axotomy in sympathetic and sensory neurons cultured in microfluidic devices. We sought to validate these in vitro findings in vivo using a transected sciatic nerve model. Consistent with the in vitro findings, DR6-/- animals displayed preserved axons up to 4 weeks after injury. In contrast to phenotypes observed in Wlds and Sarm1-/- mice, preserved axons in DR6-/- animals display profound myelin remodeling. This indicates that deterioration of axons and myelin after axotomy are mechanistically distinct processes. Finally, we find that JNK signaling after injury requires DR6, suggesting a link between this novel extrinsic pathway and the axon autonomous, intrinsic pathways that have become established for Wallerian degeneration.


Subject(s)
Axons/pathology , Myelin Sheath/pathology , Receptors, Tumor Necrosis Factor/genetics , Wallerian Degeneration/genetics , Animals , Axotomy , Mice , Receptors, Tumor Necrosis Factor/metabolism , Wallerian Degeneration/pathology
20.
PLoS Genet ; 12(12): e1006503, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27923046

ABSTRACT

Axon injury can lead to several cell survival responses including increased stability and axon regeneration. Using an accessible Drosophila model system, we investigated the regulation of injury responses and their relationship. Axon injury stabilizes the rest of the cell, including the entire dendrite arbor. After axon injury we found mitochondrial fission in dendrites was upregulated, and that reducing fission increased stabilization or neuroprotection (NP). Thus axon injury seems to both turn on NP, but also dampen it by activating mitochondrial fission. We also identified caspases as negative regulators of axon injury-mediated NP, so mitochondrial fission could control NP through caspase activation. In addition to negative regulators of NP, we found that nicotinamide mononucleotide adenylyltransferase (Nmnat) is absolutely required for this type of NP. Increased microtubule dynamics, which has previously been associated with NP, required Nmnat. Indeed Nmnat overexpression was sufficient to induce NP and increase microtubule dynamics in the absence of axon injury. DLK, JNK and fos were also required for NP. Because NP occurs before axon regeneration, and NP seems to be actively downregulated, we tested whether excessive NP might inhibit regeneration. Indeed both Nmnat overexpression and caspase reduction reduced regeneration. In addition, overexpression of fos or JNK extended the timecourse of NP and dampened regeneration in a Nmnat-dependent manner. These data suggest that NP and regeneration are conflicting responses to axon injury, and that therapeutic strategies that boost NP may reduce regeneration.


Subject(s)
Axons/metabolism , Drosophila melanogaster/genetics , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Wallerian Degeneration/genetics , Animals , Axons/pathology , Caspases/biosynthesis , Caspases/genetics , Dendrites/metabolism , Dendrites/pathology , Drosophila Proteins/biosynthesis , Drosophila Proteins/genetics , Drosophila melanogaster/growth & development , Humans , MAP Kinase Kinase 4/biosynthesis , MAP Kinase Kinase 4/genetics , Microtubules/genetics , Microtubules/pathology , Mitochondrial Dynamics/genetics , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/metabolism , Nicotinamide-Nucleotide Adenylyltransferase/antagonists & inhibitors , Nicotinamide-Nucleotide Adenylyltransferase/biosynthesis , RNA, Small Interfering/genetics , Wallerian Degeneration/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...