Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
J Anat ; 241(5): 1211-1218, 2022 11.
Article in English | MEDLINE | ID: mdl-35728923

ABSTRACT

Neurological disorders are prevalent in horses, but their study is challenging due to anatomic constraints and the large body size; very few host-specific in vitro models have been established to study these types of diseases, particularly from adult donor tissue. Here we report the generation of primary neuronal dorsal root ganglia (DRG) cultures from adult horses: the mixed, dissociated cultures, containing neurons and glial cells, remained viable for at least 90 days. Similar to DRG neurons in vivo, cultured neurons varied in size, and they developed long neurites. The mitochondrial movement was detected in cultured cells and was significantly slower in glial cells compared to DRG-derived neurons. In addition, mitochondria were more elongated in glial cells than those in neurons. Our culture model will be a useful tool to study the contribution of axonal transport defects to specific neurodegenerative diseases in horses as well as comparative studies aimed at evaluating species-specific differences in axonal transport and survival.


Subject(s)
Axonal Transport , Ganglia, Spinal , Animals , Cells, Cultured , Horses , Neurites/physiology , Neurons
2.
Bioorg Med Chem Lett ; 26(12): 2920-2926, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27158141

ABSTRACT

NAMPT may represent a novel target for drug discovery in various therapeutic areas, including oncology and inflammation. Additionally, recent work has suggested that targeting NAMPT has potential in treating axon degeneration. In this work, publicly available X-ray co-crystal structures of NAMPT and the structures of two known NAMPT inhibitors were used as the basis for a structure- and ligand-based virtual screening campaign. From this, two novel series of NAMPT inhibitors were identified, one of which showed a statistically significant protective effect when tested in a cellular model of axon degeneration.


Subject(s)
Antineoplastic Agents/pharmacology , Axons/drug effects , Cytokines/antagonists & inhibitors , Drug Discovery , Enzyme Inhibitors/pharmacology , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Axons/metabolism , Axons/pathology , Caco-2 Cells , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallography, X-Ray , Cytochrome P-450 Enzyme System/metabolism , Cytokines/metabolism , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Mice , Mice, Nude , Models, Molecular , Molecular Structure , Nicotinamide Phosphoribosyltransferase/metabolism , Structure-Activity Relationship
3.
J Neurosci ; 33(33): 13410-24, 2013 Aug 14.
Article in English | MEDLINE | ID: mdl-23946398

ABSTRACT

NMNAT2 is an NAD(+)-synthesizing enzyme with an essential axon maintenance role in primary culture neurons. We have generated an Nmnat2 gene trap mouse to examine the role of NMNAT2 in vivo. Homozygotes die perinatally with a severe peripheral nerve/axon defect and truncated axons in the optic nerve and other CNS regions. The cause appears to be limited axon extension, rather than dying-back degeneration of existing axons, which was previously proposed for the NMNAT2-deficient Blad mutant mouse. Neurite outgrowth in both PNS and CNS neuronal cultures consistently stalls at 1-2 mm, similar to the length of truncated axons in the embryos. Crucially, this suggests an essential role for NMNAT2 during axon growth. In addition, we show that the Wallerian degeneration slow protein (Wld(S)), a more stable, aberrant NMNAT that can substitute the axon maintenance function of NMNAT2 in primary cultures, can also correct developmental defects associated with NMNAT2 deficiency. This is dose-dependent, with extension of life span to at least 3 months by homozygous levels of Wld(S) the most obvious manifestation. Finally, we propose that endogenous mechanisms also compensate for otherwise limiting levels of NMNAT2. This could explain our finding that conditional silencing of a single Nmnat2 allele triggers substantial degeneration of established neurites, whereas similar, or greater, reduction of NMNAT2 in constitutively depleted neurons is compatible with normal axon growth and survival. A requirement for NMNAT2 for both axon growth and maintenance suggests that reduced levels could impair axon regeneration as well as axon survival in aging and disease.


Subject(s)
Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Neurites/pathology , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Animals , Axons , Brain/metabolism , Brain/pathology , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Gene Knockdown Techniques , Immunoblotting , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Nerve Degeneration/genetics , Neurites/metabolism , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord/metabolism , Spinal Cord/pathology , Spinal Nerves/metabolism , Spinal Nerves/pathology , Superior Cervical Ganglion/metabolism , Superior Cervical Ganglion/pathology , Transplantation Chimera
4.
Nat Commun ; 14(1): 6911, 2023 10 30.
Article in English | MEDLINE | ID: mdl-37903797

ABSTRACT

Axon degeneration and functional decline in myelin diseases are often attributed to loss of myelin but their relation is not fully understood. Perturbed myelinating glia can instigate chronic neuroinflammation and contribute to demyelination and axonal damage. Here we study mice with distinct defects in the proteolipid protein 1 gene that develop axonal damage which is driven by cytotoxic T cells targeting myelinating oligodendrocytes. We show that persistent ensheathment with perturbed myelin poses a risk for axon degeneration, neuron loss, and behavioral decline. We demonstrate that CD8+ T cell-driven axonal damage is less likely to progress towards degeneration when axons are efficiently demyelinated by activated microglia. Mechanistically, we show that cytotoxic T cell effector molecules induce cytoskeletal alterations within myelinating glia and aberrant actomyosin constriction of axons at paranodal domains. Our study identifies detrimental axon-glia-immune interactions which promote neurodegeneration and possible therapeutic targets for disorders associated with myelin defects and neuroinflammation.


Subject(s)
Demyelinating Diseases , Microglia , Animals , Mice , Axons/metabolism , CD8-Positive T-Lymphocytes , Demyelinating Diseases/metabolism , Myelin Sheath/metabolism , Neuroinflammatory Diseases
5.
Methods Mol Biol ; 2431: 73-93, 2022.
Article in English | MEDLINE | ID: mdl-35412272

ABSTRACT

Neurones are highly polarized cells with extensive axonal projections that rely on transport of proteins, RNAs, and organelles in a bidirectional manner to remain healthy. This process, known as axonal transport, can be imaged in real time through epifluorescent imaging of fluorescently labeled proteins, organelles, and other cargoes. While this is most conveniently done in primary neuronal cultures, it is more physiologically relevant when carried out in the context of a developed nerve containing both axons and glia. Here we outline how to image axonal transport ex vivo in sciatic and optic nerves, and the fimbria of the fornix. These methods could be altered to image other fluorescently labeled molecules, as well as different mechanisms of intracellular transport.


Subject(s)
Axonal Transport , Axons , Axonal Transport/physiology , Axons/metabolism , Neurons , Optic Nerve/physiology , Peripheral Nerves/metabolism , Sciatic Nerve
6.
Biochem Soc Trans ; 39(4): 933-8, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21787326

ABSTRACT

Considering the many differences between mice and humans, it is perhaps surprising how well mice model late-onset human neurodegenerative disease. Models of Alzheimer's disease, frontotemporal dementia, Parkinson's disease and Huntington's disease show some striking similarities to the corresponding human pathologies in terms of axonal transport disruption, protein aggregation, synapse loss and some behavioural phenotypes. However, there are also major differences. To extrapolate from mouse models to human disease, we need to understand how these differences relate to intrinsic limitations of the mouse system and to the effects of transgene overexpression. In the present paper, we use examples from an amyloid-overexpression model and a mutant-tau-knockin model to illustrate what we learn from each type of approach and what the limitations are. Finally, we discuss the further contributions that knockin and similar approaches can make to understanding pathogenesis and how best to model disorders of aging in a short-lived mammal.


Subject(s)
Disease Models, Animal , Neurodegenerative Diseases/genetics , Animals , Axons/metabolism , Axons/pathology , Humans , Mice , Mice, Transgenic , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Plaque, Amyloid/metabolism , Protein Isoforms , tau Proteins/genetics , tau Proteins/metabolism
7.
Exp Neurol ; 338: 113607, 2021 04.
Article in English | MEDLINE | ID: mdl-33460644

ABSTRACT

Chemotherapy-induced peripheral neuropathy (CIPN) is a common dose-limiting side effect of cancer treatment, often associated with degeneration of sensory axons or their terminal regions. Presence of the slow Wallerian degeneration protein (WLDS), or genetic deletion of sterile alpha and TIR motif containing protein 1 (SARM1), which strongly protect axons from degeneration after injury or axonal transport block, alleviate pain in several CIPN models. However, oxaliplatin can cause an acute pain response, suggesting a different mechanism of pain generation. Here, we tested whether the presence of WLDS or absence of SARM1 protects against acute oxaliplatin-induced pain in mice after a single oxaliplatin injection. In BL/6 and WldS mice, oxaliplatin induced significant mechanical and cold hypersensitivities which were absent in Sarm1-/- mice. Despite the presence of hypersensitivity there was no significant loss of intraepidermal nerve fibers (IENFs) in the footpads of any mice after oxaliplatin treatment, suggesting that early stages of pain hypersensitivity could be independent of axon degeneration. To identify other changes that could underlie the pain response, RNA sequencing was carried out in DRGs from treated and control mice of each genotype. Sarm1-/- mice had fewer gene expression changes than either BL/6 or WldS mice. This is consistent with the pain measurements in demonstrating that Sarm1-/- DRGs remain relatively unchanged after oxaliplatin treatment, unlike those in BL/6 and WldS mice. Changes in levels of four transcripts - Alas2, Hba-a1, Hba-a2, and Tfrc - correlated with oxaliplatin-induced pain, or absence thereof, across the three genotypes. Our findings suggest that targeting SARM1 could be a viable therapeutic approach to prevent oxaliplatin-induced acute neuropathic pain.


Subject(s)
Antineoplastic Agents/toxicity , Armadillo Domain Proteins/metabolism , Cytoskeletal Proteins/metabolism , Neuralgia/chemically induced , Neuralgia/metabolism , Oxaliplatin/toxicity , Animals , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
8.
Brain ; 132(Pt 2): 402-16, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19059977

ABSTRACT

Synapse loss precedes cell death in Alzheimer's disease, but the timing of axon degeneration relative to these events, and the causal relationships remain unclear. Axons become so severely dystrophic near amyloid plaques that their interruption, causing permanent loss of function, extensive synapse loss, and potentially cell death appears imminent. However, it remains unclear whether axons are truly interrupted at plaques and whether cell bodies fail to support their axons and dendrites. We traced TgCRND8 mouse axons longitudinally through, distal to, and proximal from dystrophic regions. The corresponding neurons not only survived but remained morphologically unaltered, indicating absence of axonal damage signalling or a failure to respond to it. Axons, no matter how dystrophic, remained continuous and initially morphologically normal outside the plaque region, reflecting support by metabolically active cell bodies and continued axonal transport. Immunochemical and ultrastructural studies showed dystrophic axons were tightly associated with disruption of presynaptic transmission machinery, suggesting local functional impairment. Thus, we rule out long-range degeneration axons or dendrites as major contributors to early synapse loss in this model, raising the prospect of a therapeutic window for functional rescue of individual neurons lasting months or even years after their axons become highly dystrophic. We propose that multi-focal pathology has an important role in the human disease in bringing about the switch from local, and potentially recoverable, synapse loss into permanent loss of neuronal processes and eventually their cell bodies.


Subject(s)
Alzheimer Disease/pathology , Axons/pathology , Nerve Degeneration , Neurons/pathology , Plaque, Amyloid/pathology , Animals , Breeding , Cell Survival , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron , Models, Animal , Synaptic Transmission
9.
ACS Chem Neurosci ; 11(3): 258-267, 2020 02 05.
Article in English | MEDLINE | ID: mdl-31845794

ABSTRACT

Disruption of axonal transport causes a number of rare, inherited axonopathies and is heavily implicated in a wide range of more common neurodegenerative disorders, many of them age-related. Acetylation of α-tubulin is one important regulatory mechanism, influencing microtubule stability and motor protein attachment. Of several strategies so far used to enhance axonal transport, increasing microtubule acetylation through inhibition of the deacetylase enzyme histone deacetylase 6 (HDAC6) has been one of the most effective. Several inhibitors have been developed and tested in animal and cellular models, but better drug candidates are still needed. Here we report the development and characterization of two highly potent HDAC6 inhibitors, which show low toxicity, promising pharmacokinetic properties, and enhance microtubule acetylation in the nanomolar range. We demonstrate their capacity to rescue axonal transport of mitochondria in a primary neuronal culture model of the inherited axonopathy Charcot-Marie-Tooth Type 2F, caused by a dominantly acting mutation in heat shock protein beta 1.


Subject(s)
Histone Deacetylase 6/antagonists & inhibitors , Mitochondria/metabolism , Neurons/drug effects , Tubulin/drug effects , Acetylation/drug effects , Animals , Charcot-Marie-Tooth Disease/enzymology , Disease Models, Animal , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Mice, Inbred C57BL , Microtubules/metabolism , Mitochondria/drug effects , Neurons/metabolism , Tubulin/metabolism
10.
JCI Insight ; 5(7)2020 04 09.
Article in English | MEDLINE | ID: mdl-32271161

ABSTRACT

Although human endogenous retroviruses (HERVs) represent a substantial proportion of the human genome and some HERVs, such as HERV-K(HML-2), are reported to be involved in neurological disorders, little is known about their biological function. We report that RNA from an HERV-K(HML-2) envelope gene region binds to and activates human Toll-like receptor (TLR) 8, as well as murine Tlr7, expressed in neurons and microglia, thereby causing neurodegeneration. HERV-K(HML-2) RNA introduced into the cerebrospinal fluid (CSF) of either C57BL/6 wild-type mice or APPPS1 mice, a mouse model for Alzheimer's disease (AD), resulted in neurodegeneration and microglia accumulation. Tlr7-deficient mice were protected against neurodegenerative effects but were resensitized toward HERV-K(HML-2) RNA when neurons ectopically expressed murine Tlr7 or human TLR8. Transcriptome data sets of human AD brain samples revealed a distinct correlation of upregulated HERV-K(HML-2) and TLR8 RNA expression. HERV-K(HML-2) RNA was detectable more frequently in CSF from individuals with AD compared with controls. Our data establish HERV-K(HML-2) RNA as an endogenous ligand for species-specific TLRs 7/8 and imply a functional contribution of human endogenous retroviral transcripts to neurodegenerative processes, such as AD.


Subject(s)
Alzheimer Disease , Endogenous Retroviruses , Membrane Glycoproteins , RNA, Viral , Toll-Like Receptor 7 , Toll-Like Receptor 8 , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Disease Models, Animal , Endogenous Retroviruses/genetics , Endogenous Retroviruses/metabolism , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , RNA, Viral/genetics , RNA, Viral/metabolism , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 8/genetics , Toll-Like Receptor 8/metabolism
11.
Trends Neurosci ; 30(4): 159-66, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17339056

ABSTRACT

Neurodegenerative disorders involve death of cell bodies, axons, dendrites and synapses, but it is surprisingly difficult to determine the spatiotemporal sequence of events and the causal relationships among these events. Neuronal compartments often crucially depend upon one another for survival, and molecular defects in one compartment can trigger cellular degeneration in distant parts of the neuron. Here, we consider the novel approaches used to understand these biologically complex and technically challenging questions in amyotrophic lateral sclerosis, spinal muscular atrophy, glaucoma, Alzheimer's disease, Parkinson's disease and polyglutamine disorders. We conclude that there is partial understanding of what degenerates first and why, but that controversy remains the rule not the exception. Finally, we highlight strategies for resolving these fundamental issues.


Subject(s)
Apoptosis/physiology , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/physiopathology , Neurons/physiology , Animals , Disease Progression , Humans
12.
Mol Cell Neurosci ; 38(3): 325-40, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18468455

ABSTRACT

Wallerian degeneration slow (Wld(S)) mice express a chimeric protein that delays axonal degeneration. The N-terminal domain (N70), which is essential for axonal protection in vivo, binds valosin-containing protein (VCP) and targets both Wld(S) and VCP to discrete nuclear foci. We characterized the formation, composition and localization of these potentially important foci. Missense mutations show that the N-terminal sixteen residues (N16) of Wld(S) are essential for both VCP binding and targeting Wld(S) to nuclear foci. Removing N16 abolishes foci, and VCP binding sequences from ataxin-3 or HrdI restore them. In vitro, these puncta co-localize with proteasome subunits. In vivo, Wld(S) assumes a range of nuclear distribution patterns, including puncta, and its neuronal expression and intranuclear distribution is region-specific and varies between spontaneous and transgenic Wld(S) models. We conclude that VCP influences Wld(S) intracellular distribution, and thus potentially its function, by binding within the N70 domain required for axon protection.


Subject(s)
Adenosine Triphosphatases/metabolism , Cell Cycle Proteins/metabolism , Intracellular Fluid/metabolism , Nerve Tissue Proteins/metabolism , Adenosine Triphosphatases/analysis , Adenosine Triphosphatases/genetics , Animals , Animals, Genetically Modified , Brain Chemistry/genetics , Cell Cycle Proteins/analysis , Cell Cycle Proteins/genetics , Cells, Cultured , Cytoplasm/chemistry , Cytoplasm/metabolism , HeLa Cells , Humans , Intracellular Fluid/chemistry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation, Missense , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/genetics , PC12 Cells , Protein Binding/physiology , Rats , Valosin Containing Protein , Wallerian Degeneration/genetics , Wallerian Degeneration/metabolism
13.
Mol Biol Cell ; 17(3): 1075-84, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16371511

ABSTRACT

Slow Wallerian degeneration (Wld(S)) mutant mice express a chimeric nuclear protein that protects sick or injured axons from degeneration. The C-terminal region, derived from NAD(+) synthesizing enzyme Nmnat1, is reported to confer neuroprotection in vitro. However, an additional role for the N-terminal 70 amino acids (N70), derived from multiubiquitination factor Ube4b, has not been excluded. In wild-type Ube4b, N70 is part of a sequence essential for ubiquitination activity but its role is not understood. We report direct binding of N70 to valosin-containing protein (VCP; p97/Cdc48), a protein with diverse cellular roles including a pivotal role in the ubiquitin proteasome system. Interaction with Wld(S) targets VCP to discrete intranuclear foci where ubiquitin epitopes can also accumulate. Wld(S) lacking its N-terminal 16 amino acids (N16) neither binds nor redistributes VCP, but continues to accumulate in intranuclear foci, targeting its intrinsic NAD(+) synthesis activity to these same foci. Wild-type Ube4b also requires N16 to bind VCP, despite a more C-terminal binding site in invertebrate orthologues. We conclude that N-terminal sequences of Wld(S) protein influence the intranuclear location of both ubiquitin proteasome and NAD(+) synthesis machinery and that an evolutionary recent sequence mediates binding of mammalian Ube4b to VCP.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Nucleus/metabolism , Nerve Tissue Proteins/metabolism , Adenosine Triphosphatases , Amino Acid Sequence , Animals , COS Cells , Cell Cycle Proteins/chemistry , Cells, Cultured , Chlorocebus aethiops , Evolution, Molecular , HeLa Cells , Humans , Intranuclear Space/metabolism , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Nerve Tissue Proteins/chemistry , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Protein Binding , Protein Transport , Rats , Recombinant Fusion Proteins/metabolism , Ubiquitin/metabolism , Valosin Containing Protein
14.
Trends Mol Med ; 13(4): 135-42, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17344096

ABSTRACT

Mutations in amyloid precursor protein (APP), tau and apolipoprotein E4 (ApoE4) lead to Alzheimer's disease (AD) or related pathologies. Pathogenesis and interactions between these pathways have been studied in mouse models. Here, we highlight the fact that axons are important sites of cellular pathology in each pathway and propose that pathway convergence at the molecular level might occur in axons. Recent developments suggest that axonal transport of APP influences beta-amyloid deposition and that tau regulates axonal transport. ApoE4 influences both axonal tau phosphorylation and amyloid-induced neurite pathology. Thus, a better understanding of axonal events in AD might help connect the pathogenic mechanisms of beta-amyloid, ApoE4 and tau, indicating the most important steps for therapeutic targeting.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Apolipoprotein E4/metabolism , tau Proteins/metabolism , Alzheimer Disease/etiology , Amyloid beta-Peptides/genetics , Animals , Apolipoprotein E4/genetics , Axonal Transport , Axons/metabolism , Axons/pathology , Disease Models, Animal , Humans , Mice , Models, Biological , tau Proteins/genetics
15.
Neurobiol Aging ; 68: 68-75, 2018 08.
Article in English | MEDLINE | ID: mdl-29729423

ABSTRACT

In Alzheimer's disease, many indicators point to a central role for poor axonal transport, but the potential for stimulating axonal transport to alleviate the disease remains largely untested. Previously, we reported enhanced anterograde axonal transport of mitochondria in 8- to 11-month-old MAPTP301L knockin mice, a genetic model of frontotemporal dementia with parkinsonism-17T. In this study, we further characterized the axonal transport of mitochondria in younger MAPTP301L mice crossed with the familial Alzheimer's disease model, TgCRND8, aiming to test whether boosting axonal transport in young TgCRND8 mice can alleviate axonal swelling. We successfully replicated the enhancement of anterograde axonal transport in young MAPTP301L/P301L knockin animals. Surprisingly, we found that in the presence of the amyloid precursor protein mutations, MAPTP301L/P3101L impaired anterograde axonal transport. The numbers of plaque-associated axonal swellings or amyloid plaques in TgCRND8 brains were unaltered. These findings suggest that amyloid-ß promotes an action of mutant tau that impairs axonal transport. As amyloid-ß levels increase with age even without amyloid precursor protein mutation, we suggest that this rise could contribute to age-related decline in frontotemporal dementia.


Subject(s)
Aging/genetics , Aging/physiology , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/genetics , Axonal Transport/genetics , Frontotemporal Dementia/etiology , Frontotemporal Dementia/genetics , Genetic Association Studies , Genetic Variation , Mutation , tau Proteins/genetics , Aging/metabolism , Alzheimer Disease/etiology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Axonal Transport/physiology , Brain/metabolism , Brain/pathology , Disease Models, Animal , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/pathology , Male , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/genetics , Mitochondria/metabolism , Plaque, Amyloid/metabolism , tau Proteins/metabolism
16.
Nat Neurosci ; 21(4): 552-563, 2018 04.
Article in English | MEDLINE | ID: mdl-29556029

ABSTRACT

Amyotrophic lateral sclerosis-frontotemporal dementia (ALS-FTD) constitutes a devastating disease spectrum characterized by 43-kDa TAR DNA-binding protein (TDP-43) pathology. Understanding how TDP-43 contributes to neurodegeneration will help direct therapeutic efforts. Here we have created a TDP-43 knock-in mouse with a human-equivalent mutation in the endogenous mouse Tardbp gene. TDP-43Q331K mice demonstrate cognitive dysfunction and a paucity of parvalbumin interneurons. Critically, TDP-43 autoregulation is perturbed, leading to a gain of TDP-43 function and altered splicing of Mapt, another pivotal dementia-associated gene. Furthermore, a new approach to stratify transcriptomic data by phenotype in differentially affected mutant mice revealed 471 changes linked with improved behavior. These changes included downregulation of two known modifiers of neurodegeneration, Atxn2 and Arid4a, and upregulation of myelination and translation genes. With one base change in murine Tardbp, this study identifies TDP-43 misregulation as a pathogenic mechanism that may underpin ALS-FTD and exploits phenotypic heterogeneity to yield candidate suppressors of neurodegenerative disease.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/physiopathology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dementia/genetics , Dementia/physiopathology , Gene Expression Regulation/genetics , Mutation/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Brain/metabolism , Brain/pathology , Choice Behavior/physiology , Cognition Disorders/etiology , Cognition Disorders/genetics , Conditioning, Operant/physiology , Dementia/pathology , Disease Models, Animal , Female , Male , Memory Disorders/genetics , Memory Disorders/pathology , Memory Disorders/physiopathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/genetics , Neuromuscular Junction/pathology , Neuromuscular Junction/physiopathology , Psychomotor Performance/physiology , Reaction Time/genetics
17.
Nat Neurosci ; 21(8): 1138, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29872124

ABSTRACT

In the version of this article initially published, the footnote number 17 was missing from the author list for the two authors who contributed equally. Also, the authors have added a middle initial for author Justin R. Fallon and an acknowledgement to the Babraham Institute Imaging Facility and Sequencing Core Facility. The errors have been corrected in the HTML and PDF versions of the article.

18.
Brain ; 128(Pt 2): 405-16, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15644421

ABSTRACT

Axonal dystrophy is the hallmark of axon pathology in many neurodegenerative disorders of the CNS, including Alzheimer's disease, Parkinson's disease and stroke. Axons can also form larger swellings, or spheroids, as in multiple sclerosis and traumatic brain injury. Some spheroids are terminal endbulbs of axon stumps, but swellings may also occur on unbroken axons and their role in axon loss remains uncertain. Similarly, it is not known whether spheroids and axonal dystrophy in so many different CNS disorders arise by a common mechanism. These surprising gaps in current knowledge result largely from the lack of experimental methods to manipulate axon pathology. The slow Wallerian degeneration gene, Wld(S), delays Wallerian degeneration after injury, and also delays 'dying-back' in peripheral nervous system disorders, revealing a mechanistic link between two forms of axon degeneration traditionally considered distinct. We now report that Wld(S) also inhibits axonal spheroid pathology in gracile axonal dystrophy (gad) mice. Both gracile nucleus (P < 0.001) and cervical gracile fascicle (P = 0.001) contained significantly fewer spheroids in gad/Wld(S) mice, and secondary signs of axon pathology such as myelin loss were also reduced. Motor nerve terminals at neuromuscular junctions continued to degenerate in gad/Wld(S) mice, consistent with previous observations that Wld(S) has a weaker effect on synapses than on axons, and probably contributing to the fact that Wld(S) did not alleviate gad symptoms. Wld(S) acts downstream of the initial pathogenic events to block gad pathology, suggesting that its effect on axonal swelling need not be specific to this disease. We conclude that axon degeneration mechanisms are more closely related than previously thought and that a link exists in gad between spheroid pathology and Wallerian degeneration that could hold for other disorders.


Subject(s)
Axons , Neurodegenerative Diseases/genetics , Wallerian Degeneration/genetics , Animals , Axons/metabolism , Axons/pathology , Medulla Oblongata/pathology , Mice , Mice, Inbred C57BL , Mice, Neurologic Mutants , Myelin Sheath/metabolism , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neuromuscular Junction/pathology , Phenotype , Spinal Cord/pathology , Ubiquitin/metabolism , Wallerian Degeneration/metabolism , Wallerian Degeneration/pathology
19.
BMC Neurosci ; 6: 6, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15686598

ABSTRACT

BACKGROUND: The progressive nature of Wallerian degeneration has long been controversial. Conflicting reports that distal stumps of injured axons degenerate anterogradely, retrogradely, or simultaneously are based on statistical observations at discontinuous locations within the nerve, without observing any single axon at two distant points. As axon degeneration is asynchronous, there are clear advantages to longitudinal studies of individual degenerating axons. We recently validated the study of Wallerian degeneration using yellow fluorescent protein (YFP) in a small, representative population of axons, which greatly improves longitudinal imaging. Here, we apply this method to study the progressive nature of Wallerian degeneration in both wild-type and slow Wallerian degeneration (WldS) mutant mice. RESULTS: In wild-type nerves, we directly observed partially fragmented axons (average 5.3%) among a majority of fully intact or degenerated axons 37-42 h after transection and 40-44 h after crush injury. Axons exist in this state only transiently, probably for less than one hour. Surprisingly, axons degenerated anterogradely after transection but retrogradely after a crush, but in both cases a sharp boundary separated intact and fragmented regions of individual axons, indicating that Wallerian degeneration progresses as a wave sequentially affecting adjacent regions of the axon. In contrast, most or all WldS axons were partially fragmented 15-25 days after nerve lesion, WldS axons degenerated anterogradely independent of lesion type, and signs of degeneration increased gradually along the nerve instead of abruptly. Furthermore, the first signs of degeneration were short constrictions, not complete breaks. CONCLUSIONS: We conclude that Wallerian degeneration progresses rapidly along individual wild-type axons after a heterogeneous latent phase. The speed of progression and its ability to travel in either direction challenges earlier models in which clearance of trophic or regulatory factors by axonal transport triggers degeneration. WldS axons, once they finally degenerate, do so by a fundamentally different mechanism, indicated by differences in the rate, direction and abruptness of progression, and by different early morphological signs of degeneration. These observations suggest that WldS axons undergo a slow anterograde decay as axonal components are gradually depleted, and do not simply follow the degeneration pathway of wild-type axons at a slower rate.


Subject(s)
Axons/pathology , Axons/physiology , Nerve Tissue Proteins/genetics , Wallerian Degeneration/genetics , Wallerian Degeneration/pathology , Animals , Axonal Transport/genetics , Disease Progression , Mice , Mice, Transgenic , Nerve Crush/methods
20.
J Neurol Sci ; 233(1-2): 133-8, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15899498

ABSTRACT

Valuable clues about how axons degenerate in MS can be gained from axon pathology in other disorders and experimental models. We discuss the similarities in mechanism and morphology of axon pathology in diverse circumstances revealed using mutant mice. The slow Wallerian degeneration mutation, Wld(S), delays three types of axon degeneration previously considered distinct: Wallerian degeneration of injured axons, 'dying-back' of axons in peripheral nervous system disease, and axonal spheroid pathology in gracile axonal dystrophy (gad) mice. Therefore, axon degeneration mechanisms are more uniform than previously thought and, in gad at least, axonal swelling is either related to or a consequence of Wallerian degeneration. Both axonal swelling and the accumulation of amyloid precursor protein through impaired axonal transport are common to MS, gad, and many other CNS disorders, indicating a degree of shared mechanism. YFP-H transgenic mice express YFP in a representative subset of neurons enabling unprecedented imaging of axon morphology and pathology over considerable longitudinal distances. Using this method, we have observed unbroken axons with multiple constrictions and dilatations in VEGF(delta/delta) mice, a model of amyotrophic lateral sclerosis (ALS). Similar morphologies have been described in MS, stroke, and other disorders, again suggesting a uniformity of axon degeneration mechanisms.


Subject(s)
Mice, Inbred C57BL , Mice, Neurologic Mutants/physiology , Multiple Sclerosis/therapy , Wallerian Degeneration/prevention & control , Animals , Axons/pathology , Disease Models, Animal , Humans , Mice , Recombinant Fusion Proteins/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism , Wallerian Degeneration/etiology , Wallerian Degeneration/genetics
SELECTION OF CITATIONS
SEARCH DETAIL