Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Brain ; 144(10): 3226-3238, 2021 11 29.
Article in English | MEDLINE | ID: mdl-33964142

ABSTRACT

Axonal degeneration is an early and ongoing event that causes disability and disease progression in many neurodegenerative disorders of the peripheral and central nervous systems. Chemotherapy-induced peripheral neuropathy (CIPN) is a major cause of morbidity and the main cause of dose reductions and discontinuations in cancer treatment. Preclinical evidence indicates that activation of the Wallerian-like degeneration pathway driven by sterile alpha and TIR motif containing 1 (SARM1) is responsible for axonopathy in CIPN. SARM1 is the central driver of an evolutionarily conserved programme of axonal degeneration downstream of chemical, inflammatory, mechanical or metabolic insults to the axon. SARM1 contains an intrinsic NADase enzymatic activity essential for its pro-degenerative functions, making it a compelling therapeutic target to treat neurodegeneration characterized by axonopathies of the peripheral and central nervous systems. Small molecule SARM1 inhibitors have the potential to prevent axonal degeneration in peripheral and central axonopathies and to provide a transformational disease-modifying treatment for these disorders. Using a biochemical assay for SARM1 NADase we identified a novel series of potent and selective irreversible isothiazole inhibitors of SARM1 enzymatic activity that protected rodent and human axons in vitro. In sciatic nerve axotomy, we observed that these irreversible SARM1 inhibitors decreased a rise in nerve cADPR and plasma neurofilament light chain released from injured sciatic nerves in vivo. In a mouse paclitaxel model of CIPN we determined that Sarm1 knockout mice prevented loss of axonal function, assessed by sensory nerve action potential amplitudes of the tail nerve, in a gene-dosage-dependent manner. In that CIPN model, the irreversible SARM1 inhibitors prevented loss of intraepidermal nerve fibres induced by paclitaxel and provided partial protection of axonal function assessed by sensory nerve action potential amplitude and mechanical allodynia.


Subject(s)
Armadillo Domain Proteins/antagonists & inhibitors , Axons/drug effects , Cytoskeletal Proteins/antagonists & inhibitors , Paclitaxel/toxicity , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/drug therapy , Thiazoles/therapeutic use , Animals , Antineoplastic Agents, Phytogenic/toxicity , Armadillo Domain Proteins/deficiency , Armadillo Domain Proteins/genetics , Axons/metabolism , Cells, Cultured , Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/genetics , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Mice , Mice, Knockout , Peripheral Nervous System Diseases/genetics , Peripheral Nervous System Diseases/metabolism , Thiazoles/pharmacology
2.
Acta Neuropathol Commun ; 9(1): 89, 2021 05 17.
Article in English | MEDLINE | ID: mdl-34001261

ABSTRACT

Traumatic brain injury (TBI) causes chronic symptoms and increased risk of neurodegeneration. Axons in white matter tracts, such as the corpus callosum (CC), are critical components of neural circuits and particularly vulnerable to TBI. Treatments are needed to protect axons from traumatic injury and mitigate post-traumatic neurodegeneration. SARM1 protein is a central driver of axon degeneration through a conserved molecular pathway. Sarm1-/- mice with knockout (KO) of the Sarm1 gene enable genetic proof-of-concept testing of the SARM1 pathway as a therapeutic target. We evaluated Sarm1 deletion effects after TBI using a concussive model that causes traumatic axonal injury and progresses to CC atrophy at 10 weeks, indicating post-traumatic neurodegeneration. Sarm1 wild-type (WT) mice developed significant CC atrophy that was reduced in Sarm1 KO mice. Ultrastructural classification of pathology of individual axons, using electron microscopy, demonstrated that Sarm1 KO preserved more intact axons and reduced damaged or demyelinated axons. Longitudinal MRI studies in live mice identified significantly reduced CC volume after TBI in Sarm1 WT mice that was attenuated in Sarm1 KO mice. MR diffusion tensor imaging detected reduced fractional anisotropy in both genotypes while axial diffusivity remained higher in Sarm1 KO mice. Immunohistochemistry revealed significant attenuation of CC atrophy, myelin loss, and neuroinflammation in Sarm1 KO mice after TBI. Functionally, Sarm1 KO mice exhibited beneficial effects in motor learning and sleep behavior. Based on these findings, Sarm1 inactivation can protect axons and white matter tracts to improve translational outcomes associated with CC atrophy and post-traumatic neurodegeneration.


Subject(s)
Armadillo Domain Proteins/deficiency , Axons/metabolism , Brain Injuries, Traumatic/diagnostic imaging , Brain Injuries, Traumatic/metabolism , Cytoskeletal Proteins/deficiency , Diffusion Tensor Imaging/methods , Gene Silencing/physiology , Animals , Armadillo Domain Proteins/genetics , Axons/pathology , Brain Injuries, Traumatic/genetics , Brain Injuries, Traumatic/pathology , Cytoskeletal Proteins/genetics , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Degeneration/diagnostic imaging , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Treatment Outcome
3.
Protein Cell ; 12(8): 621-638, 2021 08.
Article in English | MEDLINE | ID: mdl-33871822

ABSTRACT

Axonal degeneration is one of the key features of neurodegenerative disorders. In the canonical view, axonal degeneration destructs neural connections and promotes detrimental disease defects. Here, we assessed the enteric nervous system (ENS) of the mouse, non-human primate, and human by advanced 3D imaging. We observed the profound neurodegeneration of catecholaminergic axons in human colons with ulcerative colitis, and similarly, in mouse colons during acute dextran sulfate sodium-induced colitis. However, we unexpectedly revealed that blockage of such axonal degeneration by the Sarm1 deletion in mice exacerbated the colitis condition. In contrast, pharmacologic ablation or chemogenetic inhibition of catecholaminergic axons suppressed the colon inflammation. We further showed that the catecholaminergic neurotransmitter norepinephrine exerted a pro-inflammatory function by enhancing the expression of IL-17 cytokines. Together, this study demonstrated that Sarm1-mediated neurodegeneration within the ENS mitigated local inflammation of the colon, uncovering a previously-unrecognized beneficial role of axonal degeneration in this disease context.


Subject(s)
Armadillo Domain Proteins/genetics , Colitis, Ulcerative/genetics , Cytoskeletal Proteins/genetics , Enteric Nervous System/metabolism , Neurodegenerative Diseases/genetics , Animals , Armadillo Domain Proteins/deficiency , Catecholamines/metabolism , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/diagnostic imaging , Colitis, Ulcerative/metabolism , Colon/diagnostic imaging , Colon/metabolism , Colon/pathology , Cytoskeletal Proteins/deficiency , Dextran Sulfate/administration & dosage , Disease Models, Animal , Enteric Nervous System/diagnostic imaging , Enteric Nervous System/pathology , Gene Expression Regulation , Humans , Imaging, Three-Dimensional , Interleukin-17/genetics , Interleukin-17/metabolism , Macaca mulatta , Male , Mice , Mice, Knockout , Neurodegenerative Diseases/chemically induced , Neurodegenerative Diseases/diagnostic imaging , Neurodegenerative Diseases/metabolism , Neurons/metabolism , Neurons/pathology , Norepinephrine/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
4.
Neurobiol Dis ; 155: 105368, 2021 07.
Article in English | MEDLINE | ID: mdl-33892050

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.


Subject(s)
Armadillo Domain Proteins/genetics , Axons/pathology , Cytoskeletal Proteins/genetics , Genetic Variation/physiology , Parkinsonian Disorders/genetics , Parkinsonian Disorders/pathology , Animals , Armadillo Domain Proteins/deficiency , Axons/metabolism , Cytoskeletal Proteins/deficiency , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nerve Degeneration/chemically induced , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Oxidopamine/toxicity , Parkinsonian Disorders/chemically induced
5.
Exp Neurol ; 339: 113636, 2021 05.
Article in English | MEDLINE | ID: mdl-33548217

ABSTRACT

Axonal degeneration contributes to the pathogenesis of many neurodegenerative disorders, motivating efforts to dissect the mechanism of pathological axon loss in order to develop therapies for axonal preservation. SARM1 is a particularly attractive therapeutic target, as it is an inducible NAD+ cleaving enzyme that is required for axon loss in multiple mouse models of traumatic and degenerative neurological disease. However, it is essential to establish whether SARM1 triggers axon degeneration in human neurons before proceeding with the development of SARM1-directed therapeutics. Here we combine genome engineering with the production of human stem cell-derived neurons to test the role of human SARM1 in traumatic and neurotoxic axon degeneration. We have generated two independent SARM1 knockout human iPSC lines that do not express SARM1 protein upon differentiation into neurons. We have developed a modified sensory neuron differentiation protocol that generates human sensory neurons with high yield and purity. We find that SARM1 is required for axon degeneration in response to both physical trauma and in a cellular model of chemotherapy-induced peripheral neuropathy. Finally, we identify cADPR as a biomarker of SARM1 enzyme activity in both healthy and injured human sensory neurons. These findings are consistent with prior molecular and cellular studies in mouse neurons, and highlight the therapeutic potential of SARM1 inhibition for the prevention and treatment of human neurological disease.


Subject(s)
Armadillo Domain Proteins/deficiency , Armadillo Domain Proteins/genetics , Axons/metabolism , Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/genetics , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Sensory Receptor Cells/metabolism , Base Sequence , Cell Line , Gene Knockout Techniques/methods , Humans , Induced Pluripotent Stem Cells/metabolism
6.
Methods Mol Biol ; 2143: 145-157, 2020.
Article in English | MEDLINE | ID: mdl-32524478

ABSTRACT

We describe here an organotypic culture system we have used to investigate mechanisms that maintain structure and function of axon terminals at the neuromuscular junction (NMJ). We developed this by taking advantage of the slow Wallerian degeneration phenotype in mutant Wlds mice, using these to compare preservation of NMJs with degeneration in nerve-muscle preparations from wild-type mice. We take hind limb tibial nerve/flexor digitorum brevis and lumbrical muscles and incubate them in mammalian physiological saline at 32 °C for 24-48 h. Integrity of NMJs can then be compared using a combination of electrophysiological and morphological techniques. We illustrate our method with data showing synaptic preservation ex vivo in nerve-muscle explants from Sarm-1 null-mutant mice. The ex vivo assays of NMJ integrity we describe here may therefore be useful for detailed investigation of synaptic maintenance and degeneration.


Subject(s)
Neuromuscular Junction/physiology , Organ Culture Techniques/methods , Wallerian Degeneration/physiopathology , Animals , Armadillo Domain Proteins/deficiency , Axons/physiology , Cytoskeletal Proteins/deficiency , Dissection/methods , Electrophysiology/methods , Female , Immunohistochemistry/methods , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Muscle, Skeletal , Neuromuscular Junction/ultrastructure , Organ Culture Techniques/instrumentation , Synapses/ultrastructure , Tibial Nerve
7.
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
9.
Exp Neurol ; 321: 113040, 2019 11.
Article in English | MEDLINE | ID: mdl-31445042

ABSTRACT

Traumatic brain injury (TBI) often damages axons in white matter tracts and causes corpus callosum (CC) atrophy in chronic TBI patients. Injured axons encounter irreversible damage if transected, or alternatively may maintain continuity and subsequently either recover or degenerate. Secondary mechanisms can cause further axon damage, myelin pathology, and neuroinflammation. Molecular mechanisms regulating the progression of white matter pathology indicate potential therapeutic targets. SARM1 is essential for execution of the conserved axon death pathway. We examined white matter pathology following mild TBI with CC traumatic axonal injury in mice with Sarm1 gene deletion (Sarm1-/-). High resolution ultrastructural analysis at 3 days post-TBI revealed dramatically reduced axon damage in Sarm1-/- mice, as compared to Sarm1+/+ wild-type controls. Sarm1 deletion produced larger axons with thinner myelin, and attenuated TBI induced demyelination, i.e. myelin loss along apparently intact axons. At 6 weeks post-TBI, Sarm1-/- mice had less demyelination and thinner myelin than Sarm1+/+ mice, but axonal protection was no longer observed. We next used Thy1-YFP crosses to assess Sarm1 involvement in white matter neurodegeneration and neuroinflammation at 8 weeks post-TBI, when significant CC atrophy indicates chronic pathology. Thy1-YFP expression demonstrated continued CC axon damage yet absence of overt cortical pathology. Importantly, significant CC atrophy in Thy1-YFP/Sarm1+/+ mice was associated with reduced neurofilament immunolabeling of axons. Both effects were attenuated in Thy1-YFP/Sarm1-/- mice. Surprisingly, Thy1-YFP/Sarm1-/- mice had increased CC astrogliosis. This study demonstrates that Sarm1 inactivation reduces demyelination, and white matter atrophy after TBI, while the post-injury stage impacts when axon protection is effective.


Subject(s)
Armadillo Domain Proteins/deficiency , Brain Injuries, Traumatic/pathology , Cytoskeletal Proteins/deficiency , Demyelinating Diseases/pathology , White Matter/pathology , Animals , Atrophy/metabolism , Atrophy/pathology , Axons/metabolism , Axons/pathology , Brain Injuries, Traumatic/metabolism , Demyelinating Diseases/metabolism , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Knockout , White Matter/metabolism
10.
J Exp Med ; 216(4): 743-756, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30842236

ABSTRACT

SARM1 (sterile α and HEAT/armadillo motif-containing protein) is a member of the MyD88 (myeloid differentiation primary response gene 88) family, which mediates innate immune responses. Because inactivation of SARM1 prevents various forms of axonal degeneration, we tested whether it might protect against prion-induced neurotoxicity. Instead, we found that SARM1 deficiency exacerbates the progression of prion pathogenesis. This deleterious effect was not due to SARM1-dependent modulation of prion-induced neuroinflammation, since microglial activation, astrogliosis, and brain cytokine profiles were not altered by SARM1 deficiency. Whole-transcriptome analyses indicated that SARM1 deficiency led to strong, selective overexpression of the pro-apoptotic gene XAF1 (X-linked inhibitor of apoptosis-associated factor 1). Consequently, the activity of pro-apoptotic caspases and neuronal death were enhanced in prion-infected SARM1 -/- mice. These results point to an unexpected function of SARM1 as a regulator of prion-induced neurodegeneration and suggest that XAF1 might constitute a therapeutic target in prion disease.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins/genetics , Apoptosis/genetics , Armadillo Domain Proteins/deficiency , Armadillo Domain Proteins/metabolism , Axons/metabolism , Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/metabolism , Scrapie/metabolism , Up-Regulation/genetics , Animals , Armadillo Domain Proteins/genetics , Brain/metabolism , Cytoskeletal Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Degeneration , Protein Isoforms/metabolism , Scrapie/pathology
11.
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
12.
Cell Rep ; 23(3): 716-724, 2018 Apr 17.
Article in English | MEDLINE | ID: mdl-29669278

ABSTRACT

Traumatic injuries can trigger inflammatory reactions, leading to profound neuropathological consequences. However, the immune capacity of neurons, distinct from that of immune cells or glial cells, in response to traumatic insults remains to be fully characterized. In this study, we demonstrate that neurons can detect, cell autonomously, distant axonal damage, resulting in rapid production of a specific collection of cytokines and chemokines. This neuronal immune response appears spatially and temporally separated from injury-induced axon degeneration. We then identify through the genetic screen that this immune response is regulated by TIR-domain adaptor Sarm1/Myd88-5. We further show that Sarm1 functions through the downstream Jnk-c-Jun signal, and blockage of this Sarm1-Jnk-c-Jun pathway effectively abolishes the recruitment of immune cells to injury-afflicted neural tissues. We therefore uncover the key function of the Sarm1 signaling pathway, independent of its known role in axon degeneration, in the neuronal intrinsic immune response to traumatic axonal injuries.


Subject(s)
Armadillo Domain Proteins/genetics , Axons/metabolism , Cytoskeletal Proteins/genetics , Myeloid Differentiation Factor 88/metabolism , Sciatic Nerve/injuries , Animals , Armadillo Domain Proteins/deficiency , Chemokines/genetics , Chemokines/metabolism , Cytokines/genetics , Cytokines/metabolism , Cytoskeletal Proteins/deficiency , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/antagonists & inhibitors , Myeloid Differentiation Factor 88/genetics , Neurons/cytology , Neurons/immunology , Neurons/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction
13.
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
14.
J Peripher Nerv Syst ; 22(3): 162-171, 2017 09.
Article in English | MEDLINE | ID: mdl-28485482

ABSTRACT

Distal axon degeneration seen in many peripheral neuropathies is likely to share common molecular mechanisms with Wallerian degeneration. Although several studies in mouse models of peripheral neuropathy showed prevention of axon degeneration in the slow Wallerian degeneration (Wlds) mouse, the role of a recently identified player in Wallerian degeneration, Sarm1, has not been explored extensively. In this study, we show that mice lacking the Sarm1 gene are resistant to distal axonal degeneration in a model of chemotherapy induced peripheral neuropathy caused by paclitaxel and a model of high fat diet induced putative metabolic neuropathy. This study extends the role of Sarm1 to axon degeneration seen in peripheral neuropathies and identifies it as a likely target for therapeutic development.


Subject(s)
Armadillo Domain Proteins/deficiency , Cytoskeletal Proteins/deficiency , Diet, High-Fat/adverse effects , Peripheral Nervous System Diseases/genetics , Peripheral Nervous System Diseases/prevention & control , Action Potentials/genetics , Analysis of Variance , Animals , Antineoplastic Agents, Phytogenic/toxicity , Armadillo Domain Proteins/genetics , Cytoskeletal Proteins/genetics , Disease Models, Animal , Hyperalgesia/etiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Conduction/genetics , Paclitaxel/toxicity , Pain Threshold/physiology , Peripheral Nervous System Diseases/chemically induced , Reaction Time/genetics , Sural Nerve/pathology
15.
Brain ; 139(Pt 4): 1094-105, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26912636

ABSTRACT

Axonal degeneration is a critical, early event in many acute and chronic neurological disorders. It has been consistently observed after traumatic brain injury, but whether axon degeneration is a driver of traumatic brain injury remains unclear. Molecular pathways underlying the pathology of traumatic brain injury have not been defined, and there is no efficacious treatment for traumatic brain injury. Here we show that mice lacking the mouse Toll receptor adaptor Sarm1 (sterile α/Armadillo/Toll-Interleukin receptor homology domain protein) gene, a key mediator of Wallerian degeneration, demonstrate multiple improved traumatic brain injury-associated phenotypes after injury in a closed-head mild traumatic brain injury model. Sarm1(-/-) mice developed fewer ß-amyloid precursor protein aggregates in axons of the corpus callosum after traumatic brain injury as compared to Sarm1(+/+) mice. Furthermore, mice lacking Sarm1 had reduced plasma concentrations of the phophorylated axonal neurofilament subunit H, indicating that axonal integrity is maintained after traumatic brain injury. Strikingly, whereas wild-type mice exibited a number of behavioural deficits after traumatic brain injury, we observed a strong, early preservation of neurological function in Sarm1(-/-) animals. Finally, using in vivo proton magnetic resonance spectroscopy we found tissue signatures consistent with substantially preserved neuronal energy metabolism in Sarm1(-/-) mice compared to controls immediately following traumatic brain injury. Our results indicate that the SARM1-mediated prodegenerative pathway promotes pathogenesis in traumatic brain injury and suggest that anti-SARM1 therapeutics are a viable approach for preserving neurological function after traumatic brain injury.


Subject(s)
Armadillo Domain Proteins/deficiency , Axons/metabolism , Axons/pathology , Brain Injuries/metabolism , Brain Injuries/pathology , Cytoskeletal Proteins/deficiency , Recovery of Function/physiology , Amyloid beta-Peptides/metabolism , Animals , Corpus Callosum/metabolism , Corpus Callosum/pathology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Wallerian Degeneration/metabolism , Wallerian Degeneration/pathology
16.
Cell Rep ; 13(11): 2539-2552, 2015 Dec 22.
Article in English | MEDLINE | ID: mdl-26686637

ABSTRACT

Axon injury leads to rapid depletion of NAD-biosynthetic enzyme NMNAT2 and high levels of its substrate, NMN. We proposed a key role for NMN in Wallerian degeneration but downstream events and their relationship to other mediators remain unclear. Here, we show, in vitro and in vivo, that axotomy leads to a late increase in intra-axonal Ca(2+), abolished by pharmacological or genetic reduction of NMN levels. NMN requires the pro-degenerative protein SARM1 to stimulate Ca(2+) influx and axon degeneration. While inhibition of NMN synthesis and SARM1 deletion block Ca(2+) rise and preserve axonal integrity, they fail to prevent early mitochondrial dynamic changes. Furthermore, depolarizing mitochondria does not alter the rate of Wallerian degeneration. These data reveal that NMN and SARM1 act in a common pathway culminating in intra-axonal Ca(2+) increase and fragmentation and dissociate mitochondrial dysfunctions from this pathway, elucidating which steps may be most effective as targets for therapy.


Subject(s)
Armadillo Domain Proteins/genetics , Calcium/metabolism , Cytoskeletal Proteins/genetics , Mitochondria/metabolism , Nicotinamide Mononucleotide/metabolism , Acrylamides/pharmacology , Amidohydrolases/metabolism , Animals , Armadillo Domain Proteins/deficiency , Axons/drug effects , Axons/metabolism , Cytoskeletal Proteins/deficiency , Ions/chemistry , Ions/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Nicotinamide Mononucleotide/pharmacology , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Piperidines/pharmacology , Wallerian Degeneration/pathology
17.
J Cell Biol ; 193(4): 769-84, 2011 May 16.
Article in English | MEDLINE | ID: mdl-21555464

ABSTRACT

Dendritic arborization is a critical neuronal differentiation process. Here, we demonstrate that syndecan-2 (Sdc2), a synaptic heparan sulfate proteoglycan that triggers dendritic filopodia and spine formation, regulates dendritic arborization in cultured hippocampal neurons. This process is controlled by sterile α and TIR motif-containing 1 protein (Sarm1), a negative regulator of Toll-like receptor 3 (TLR3) in innate immunity signaling. We show that Sarm1 interacts with and receives signal from Sdc2 and controls dendritic arborization through the MKK4-JNK pathway. In Sarm1 knockdown mice, dendritic arbors of neurons were less complex than those of wild-type littermates. In addition to acting downstream of Sdc2, Sarm1 is expressed earlier than Sdc2, which suggests that it has multiple roles in neuronal morphogenesis. Specifically, it is required for proper initiation and elongation of dendrites, axonal outgrowth, and neuronal polarization. These functions likely involve Sarm1-mediated regulation of microtubule stability, as Sarm1 influenced tubulin acetylation. This study thus reveals the molecular mechanism underlying the action of Sarm1 in neuronal morphogenesis.


Subject(s)
Armadillo Domain Proteins/metabolism , Cell Shape , Cytoskeletal Proteins/metabolism , Hippocampus/metabolism , Immunity, Innate , Neurons/metabolism , Syndecan-2/metabolism , Animals , Armadillo Domain Proteins/deficiency , Armadillo Domain Proteins/genetics , COS Cells , Chlorocebus aethiops , Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/genetics , Dendrites/metabolism , Fluorescent Antibody Technique , HEK293 Cells , Hippocampus/embryology , Hippocampus/immunology , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase 4/metabolism , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Microscopy, Video , Microtubules/metabolism , Morphogenesis , Neurons/immunology , RNA Interference , Recombinant Fusion Proteins/metabolism , Syndecan-2/genetics , Time Factors , Transfection
18.
Development ; 138(10): 2099-109, 2011 May.
Article in English | MEDLINE | ID: mdl-21521738

ABSTRACT

Defects in the development or maintenance of tubule diameter correlate with polycystic kidney disease. Here, we report that absence of the cadherin regulator p120 catenin (p120ctn) from the renal mesenchyme prior to tubule formation leads to decreased cadherin levels with abnormal morphologies of early tubule structures and developing glomeruli. In addition, mutant mice develop cystic kidney disease, with markedly increased tubule diameter and cellular proliferation, and detached luminal cells only in proximal tubules. The p120ctn homolog Arvcf is specifically absent from embryonic proximal tubules, consistent with the specificity of the proximal tubular phenotype. p120ctn knockdown in renal epithelial cells in 3D culture results in a similar cystic phenotype with reduced levels of E-cadherin and active RhoA. We find that E-cadherin knockdown, but not RhoA inhibition, phenocopies p120ctn knockdown. Taken together, our data show that p120ctn is required for early tubule and glomerular morphogenesis, as well as control of luminal diameter, probably through regulation of cadherins.


Subject(s)
Catenins/metabolism , Kidney Glomerulus/embryology , Kidney Glomerulus/metabolism , Kidney Tubules/embryology , Kidney Tubules/metabolism , Animals , Armadillo Domain Proteins/deficiency , Armadillo Domain Proteins/genetics , Armadillo Domain Proteins/metabolism , Base Sequence , Cadherins/deficiency , Cadherins/genetics , Cadherins/metabolism , Catenins/deficiency , Catenins/genetics , Cell Adhesion Molecules/deficiency , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Line , Cell Polarity , Cell Proliferation , Cytoskeleton/metabolism , Dogs , Female , Gene Knockdown Techniques , Kidney Diseases, Cystic/embryology , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/metabolism , Male , Mice , Mice, Knockout , Models, Biological , Morphogenesis , Nephrons/embryology , Nephrons/metabolism , Phenotype , Phosphoproteins/deficiency , Phosphoproteins/genetics , Phosphoproteins/metabolism , Pregnancy , RNA, Small Interfering/genetics , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein , Delta Catenin
19.
J Virol ; 83(18): 9329-38, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19587044

ABSTRACT

Sterile alpha and HEAT/Armadillo motif (SARM) is a highly conserved Toll/interleukin-1 receptor (TIR)-containing adaptor protein that is believed to negatively regulate signaling of the pathogen recognition receptors Toll-like receptor 3 (TLR3) and TLR4. To test its physiological function in the context of a microbial infection, we generated SARM(-/-) mice and evaluated the impact of this deficiency on the pathogenesis of West Nile virus (WNV), a neurotropic flavivirus that requires TLR signaling to restrict infection. Although SARM was preferentially expressed in cells of the central nervous system (CNS), studies with primary macrophages, neurons, or astrocytes showed no difference in viral growth kinetics. In contrast, viral replication was increased specifically in the brainstem of SARM(-/-) mice, and this was associated with enhanced mortality after inoculation with a virulent WNV strain. A deficiency of SARM was also linked to reduced levels of tumor necrosis factor alpha (TNF-alpha), decreased microglia activation, and increased neuronal death in the brainstem after WNV infection. Thus, SARM appears to be unique among the TIR adaptor molecules, since it functions to restrict viral infection and neuronal injury in a brain region-specific manner, possibly by modulating the activation of resident CNS inflammatory cells.


Subject(s)
Armadillo Domain Proteins/physiology , Brain Stem/pathology , Cytoskeletal Proteins/physiology , Microglia/physiology , Tumor Necrosis Factor-alpha/biosynthesis , West Nile virus/pathogenicity , Animals , Armadillo Domain Proteins/analysis , Armadillo Domain Proteins/deficiency , Brain Stem/virology , Cell Death , Cytoskeletal Proteins/analysis , Cytoskeletal Proteins/deficiency , Mice , Mice, Knockout , Neurons/pathology , Neurons/virology , Tissue Distribution , Toll-Like Receptors
SELECTION OF CITATIONS
SEARCH DETAIL
...