Genetic Inactivation of SARM1 Axon Degeneration Pathway Improves

Total Page:16

File Type:pdf, Size:1020Kb

Genetic Inactivation of SARM1 Axon Degeneration Pathway Improves Bradshaw Jr. et al. acta neuropathol commun (2021) 9:89 https://doi.org/10.1186/s40478-021-01193-8 RESEARCH Open Access Genetic inactivation of SARM1 axon degeneration pathway improves outcome trajectory after experimental traumatic brain injury based on pathological, radiological, and functional measures Donald V. Bradshaw Jr.1,2, Andrew K. Knutsen3,4, Alexandru Korotcov3,4, Genevieve M. Sullivan2, Kryslaine L. Radomski2,4, Bernard J. Dardzinski3,4,6, Xiaomei Zi2, Dennis P. McDaniel5 and Regina C. Armstrong1,2,4* Abstract Traumatic brain injury (TBI) causes chronic symptoms and increased risk of neurodegeneration. Axons in white mat- ter 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− therapeu- tic target. We evaluated Sarm1 deletion efects 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 signifcant CC atrophy that was reduced in Sarm1 KO mice. Ultrastructural classifcation 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 identifed signifcantly reduced CC volume after TBI in Sarm1 WT mice that was attenuated in Sarm1 KO mice. MR difusion tensor imaging detected reduced fractional anisotropy in both genotypes while axial difusivity remained higher in Sarm1 KO mice. Immunohistochem- istry revealed signifcant attenuation of CC atrophy, myelin loss, and neuroinfammation in Sarm1 KO mice after TBI. Functionally, Sarm1 KO mice exhibited benefcial efects in motor learning and sleep behavior. Based on these fnd- ings, Sarm1 inactivation can protect axons and white matter tracts to improve translational outcomes associated with CC atrophy and post-traumatic neurodegeneration. Keywords: Chronic traumatic brain injury, SARM1, Sterile alpha and Toll, interleukin-1 receptor containing motif 1, White matter, Corpus callosum atrophy, Magnetic resonance imaging Background Traumatic brain injury (TBI) results in long term dis- *Correspondence: [email protected] ability in more severe cases and can cause persistent 2 Department of Anatomy, Physiology and Genetics, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, symptoms even in patients who receive a “mild” diag- 4301 Jones Bridge Rd., Bethesda, MD 20814, USA nosis [42, 67, 72]. TBI may also lead to post-traumatic Full list of author information is available at the end of the article neurodegeneration and increase the risk for co-morbid © The Author(s) 2021. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http:// creat iveco mmons. org/ licen ses/ by/4. 0/. The Creative Commons Public Domain Dedication waiver (http:// creat iveco mmons. org/ publi cdoma in/ zero/1. 0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data. Bradshaw Jr. et al. acta neuropathol commun (2021) 9:89 Page 2 of 19 neurodegenerative diseases, such as Alzheimer’s dis- in cultured human sensory neurons and in live mice ease [16, 19, 31, 73]. In patients with moderate-severe reduced axon damage in models of peripheral neuropa- TBI, difuse axonal injury has been shown to predict thy induced by trauma or chemotherapy [14, 24, 26, 70]. the extent of post-traumatic neurodegeneration, based In multiple models of TBI, Sarm1 knockout (KO) mice on MRI volumetric and difusion tensor imaging (DTI) exhibited signifcantly reduced axon damage at acute and data [30]. Te strongest relationship was found in central late phase time points [35, 52, 58, 96]. Long-term studies white matter tracts, including the corpus callosum (CC). of TBI in mice can model post-traumatic neurodegenera- Te CC is one of the main structures exhibiting atrophy tion that includes atrophy of the CC [47, 53, 64]. Initial across patients with complicated mild to severe TBI [16, results from our prior studies provided the frst evidence 31]. Furthermore, DTI tractography demonstrated dis- that Sarm1 deletion may reduce both late stage axon rupted fber tract continuity in anterior CC regions after damage and CC atrophy [52]. concussions while broad areas of disrupted tracts were We now focus on CC atrophy as an important trans- found throughout the CC in patients with MRI fndings lational outcome measure of post-traumatic neurode- of difuse axonal injury [38]. Ex vivo MRI of a patient generation. We evaluate the efect of Sarm1 deletion who died 26 days after TBI demonstrated that white mat- on axon damage, demyelination, and neuroinfamma- ter tract disruptions detected by DTI correlated with tion on the progression of CC atrophy after closed head neuropathological identifcation of axonal injury [69]. TBI. We use longitudinal MRI to detect changes in CC Long axons in white matter tracts are particularly vul- volume and white matter integrity in live mice, which nerable to damage in all forms of closed head TBI [11, 40, provides a highly translational outcome measure of pro- 57, 78]. At an early stage of damage, axons can exhibit longed Sarm1 inactivation. To demonstrate whether the focal swellings yet longitudinal in vitro and in vivo imag- pathological efects of Sarm1 deletion have a meaning- ing has shown the potential to recover [32, 37, 68]. Once ful impact on complex functions, we assess motor skill mechanical or molecular processes fragment the axon, learning and sleep behavior. Tis experimental design then the distal axon irreversibly degrades by Wallerian provides a rigorous and translationally relevant screen to degeneration [32, 37, 54, 56, 68]. Te majority of axon evaluate the impact of Sarm1 deletion on long-term out- damage is due to secondary mechanisms of damage that come measures after TBI. can cause axons to initiate Wallerian degeneration for weeks or more after the initial TBI event [13, 54, 56]. Materials and methods Irreversible axonal injury leads to disconnection between Mice brain regions and disruption of neural circuits that may All mice were treated in accordance with guidelines of contribute to diverse TBI symptoms [33, 59]. To improve the Uniformed Services University of the Health Sci- outcomes for patients after TBI, research is needed to ences and the National Institutes of Health Guide for identify approaches to protect against axon degeneration the Care and Use of Laboratory Animals. Mice were and, further, to determine whether acute axon protection socially housed with 2–5 mice per 35 cm × 16.5 cm can reduce post-traumatic neurodegeneration. × 18 cm cages containing enrichment objects. Mice Wallerian degeneration is an “active program of axon were maintained on a standard 12 h cycle of daytime self-destruction” [76]. After injury or mitochondrial dys- light (6:00–18:00). Sarm1−/− knockout (KO) mice, function, the SARM1 (sterile alpha and Toll/interleukin-1 B6.129 × 1-Sarm1tm1Aidi/J (RRID:IMSR_JAX:018069) receptor motif-containing 1) protein executes a highly were originally obtained from Te Jackson Laboratory. conserved molecular axon degeneration pathway [27, 48, Sarm1 KO mice were generated by targeted mutation 70, 84]. Injury releases SARM1 from an auto-inhibited that replaced exons 3 through 6 with a neo cassette in state that is found in healthy axons [22, 76, 80]. Active the reverse orientation and absence of SARM1 protein in SARM1 is a glycohydrolase which depletes nicotinamide the brain was confrmed by western blot [43]. Sarm1 KO adenine dinucleotide (NAD +) that is critical for energy mice were backcrossed to C57BL/6 mice for 15 genera- stores in axons [18, 23, 26]. Loss of sufcient NAD + leads tions before arrival at Te Jackson Laboratory and then to a cascade of axon destruction in which ionic imbalance crossed at least once to C57BL/6 J mice. For our studies, leads to cytoskeletal and structural breakdown within the Sarm1 KO mice were crossed to C57BL/6 J mice and then axon [25, 26, 85]. heterozygotes were bred to generate littermates used in Deletion of the Sarm1 gene provides a genetic proof- experiments. Sarm1 is highly expressed in the brain; lack of-concept to examine the efects of long-term inac- of detectable SARM1 protein in Sarm1 KO mice does not tivation of this axon degeneration pathway in chronic produce gross or microscopic brain pathology [28, 43]. TBI and to evaluate the relationship of axonal injury to Te colony was maintained at Charles River Laboratories post-traumatic neurodegeneration. Knockout of Sarm1 (Wilmington, MA) where ear biopsies were genotyped Bradshaw Jr. et al. acta neuropathol commun (2021) 9:89 Page 3 of 19 using a 3-primer allele specifc PCR assay that targets the immersed in 2% osmium tetroxide (OsO4; Electron mutated Sarm1 region. Experimental mice were accli- Microscopy Sciences; Cat #19100) infltrated with Spurr mated for 3 days after shipment and prior to the start of epoxy resin (Electron Microscopy Sciences; Cat #14300), experiments.
Recommended publications
  • Synthesis and Consumption: a Focus on the PARP Family
    Downloaded from genesdev.cshlp.org on September 30, 2021 - Published by Cold Spring Harbor Laboratory Press SPECIAL SECTION: REVIEW Interplay between compartmentalized NAD+ synthesis and consumption: a focus on the PARP family Michael S. Cohen Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, Oregon 97210, USA Nicotinamide adenine dinucleotide (NAD+) is an essential erate a polymer of ADP-ribose (ADPr), a process known as cofactor for redox enzymes, but also moonlights as a sub- poly-ADP-ribosylation or PARylation (more on this below) strate for signaling enzymes. When used as a substrate by (Fig. 1; Chambon et al. 1963, 1966; Fujimura et al. 1967a,b). signaling enzymes, it is consumed, necessitating the recy- Unlike NAD+-mediated redox reactions, this glycosidic cling of NAD+ consumption products (i.e., nicotinamide) cleavage reaction is irreversible and leads to the consump- via a salvage pathway in order to maintain NAD+ homeo- tion of NAD+. Consistent with this notion, in the 1970s it stasis. A major family of NAD+ consumers in mammalian was shown that NAD+ exhibits a high turnover in human cells are poly-ADP-ribose-polymerases (PARPs). PARPs cells (Rechsteiner et al. 1976). We now know that there are comprise a family of 17 enzymes in humans, 16 of which many “NAD+ consumers” (e.g., other PARP family mem- catalyze the transfer of ADP-ribose from NAD+ to macro- ber, sirtuins, etc.) beyond PARP1, which are found in molecular targets (namely, proteins, but also DNA and nearly all subcellular compartments, including the nucle- RNA). Because PARPs and the NAD+ biosynthetic en- us, cytoplasm, and mitochondria (Fig.
    [Show full text]
  • 1 | Page SARM1 Deficiency, Which Prevents Wallerian Degeneration
    bioRxiv preprint doi: https://doi.org/10.1101/485052; this version posted December 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. SARM1 deficiency, which prevents Wallerian degeneration, upregulates XAF1 and accelerates prion disease Caihong Zhu#, Bei Li#, Karl Frontzek, Yingjun Liu and Adriano Aguzzi* Institute of Neuropathology, University of Zurich, Zurich, Switzerland # equal contribution * Corresponding author: Adriano Aguzzi Institute of Neuropathology, University of Zurich Schmelzbergstrasse 12, CH-8091 Zurich, Switzerland Tel: +41-44-255-2107 Email address: [email protected] Condensed title: SARM1 deficiency accelerates prion diseases Abbreviations: CNS, central nervous system; dpi, days post inoculation; MyD88, myeloid differentiation primary response gene 88; NBH, non-infectious brain homogenates; PK, proteinase K; qRT-PCR, quantitative real-time PCR; RML6, Rocky Mountain Laboratories scrapie strain passage 6; SARM1, sterile α and HEAT/armadillo motifs containing protein; XAF1, X-linked inhibitor of apoptosis-associated factor 1. 1 | Page bioRxiv preprint doi: https://doi.org/10.1101/485052; this version posted December 3, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Abstract SARM1 (sterile α and HEAT/armadillo motifs 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.
    [Show full text]
  • Permeant Fluorescent Probes Visualize the Activation of SARM1 And
    RESEARCH ARTICLE Permeant fluorescent probes visualize the activation of SARM1 and uncover an anti- neurodegenerative drug candidate Wan Hua Li1,2, Ke Huang3, Yang Cai4, Qian Wen Wang1, Wen Jie Zhu1, Yun Nan Hou1, Sujing Wang1, Sheng Cao5, Zhi Ying Zhao1, Xu Jie Xie1, Yang Du5, Chi-Sing Lee3*, Hon Cheung Lee1*, Hongmin Zhang4*, Yong Juan Zhao1,2,6* 1State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China; 2Ciechanover Institute of Precision and Regenerative Medicine, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, China; 3Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China; 4Department of Biology, Southern University of Science and Technology, Shenzhen, China; 5Kobilka Institute of Innovative Drug Discovery, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, China; 6Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China Abstract SARM1 regulates axonal degeneration through its NAD-metabolizing activity and is a drug target for neurodegenerative disorders. We designed and synthesized fluorescent conjugates of styryl derivative with pyridine to serve as substrates of SARM1, which exhibited large red shifts after conversion. With the conjugates, SARM1 activation was visualized in live cells following elevation of endogenous NMN or treatment with a cell-permeant NMN-analog. In neurons, imaging documented mouse SARM1 activation preceded vincristine-induced axonal degeneration by hours. *For correspondence: Library screening identified a derivative of nisoldipine (NSDP) as a covalent inhibitor of SARM1 that [email protected] (C-SL); reacted with the cysteines, especially Cys311 in its ARM domain and blocked its NMN-activation, [email protected] (HCL); protecting axons from degeneration.
    [Show full text]
  • Activation of Sarm1 Produces Cadpr to Increase Intra-Axonal Calcium and Promote Axon Degeneration in CIPN
    bioRxiv preprint doi: https://doi.org/10.1101/2021.04.15.440024; this version posted April 15, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Title: Activation of Sarm1 produces cADPR to increase intra-axonal calcium and promote axon degeneration in CIPN. Yihang Li1,2, Maria F. Pazyra-Murphy1,2, Daina Avizonis3, Mariana de Sa Tavares Russo3, Sophia Tang2, Johann S. Bergholz2,4,5, Tao Jiang2, Jean J. Zhao2,4,5, Jian Zhu6, Kwang Woo Ko7, Jeffrey Milbrandt6,8, Aaron DiAntonio7,8, Rosalind A. Segal1,2 1. Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA 2. Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA 3. Metabolomics Innovation Resource, Goodman Cancer Research Centre, McGill University, Montréal, QC, Canada, H3A1A3 4. Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA 5. Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA 6. Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA, 63110 7. Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA, 63110 8. Needleman Center for Neurometabolism and Axonal Therapeutics, Washington University School of Medicine, St. Louis, MO, 63110, USA HIGHLIGHTS • Paclitaxel induces intra-axonal calcium flux • Sarm1-dependent cADPR production promotes axonal calcium elevation and degeneration • Antagonizing cADPR signaling pathway protects against paclitaxel-induced peripheral neuropathy in vitro and in vivo SUMMARY Cancer patients frequently develop chemotherapy-induced peripheral neuropathy (CIPN), a painful and long-lasting disorder with profound somatosensory deficits.
    [Show full text]
  • Primary Traumatic Axonopathy in Mice Subjected to Impact Acceleration: a Reappraisal of Pathology and Mechanisms with High-Resolution Anatomical Methods
    The Journal of Neuroscience, April 18, 2018 • 38(16):4031–4047 • 4031 Neurobiology of Disease Primary Traumatic Axonopathy in Mice Subjected to Impact Acceleration: A Reappraisal of Pathology and Mechanisms with High-Resolution Anatomical Methods Nikolaos K. Ziogas1 and Vassilis E. Koliatsos1,2,3 1Division of Neuropathology, Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, 2Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, and 3Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD 21205 Traumatic axonal injury (TAI) is a common neuropathology in traumatic brain injury and is featured by primary injury to axons. Here, wegeneratedTAIwithimpactaccelerationoftheheadinmaleThy1-eYFP-Htransgenicmiceinwhichspecificpopulationsofneuronsand their axons are labeled with yellow fluorescent protein. This model results in axonal lesions in multiple axonal tracts along with blood– brain barrier disruption and neuroinflammation. The corticospinal tract, a prototypical long tract, is severely affected and is the focus of this study. Using optimized CLARITY at single-axon resolution, we visualized the entire corticospinal tract volume from the pons to the cervical spinal cord in 3D and counted the total number of axonal lesions and their progression over time. Our results divulged the presence of progressive traumatic axonopathy that was maximal at the pyramidal decussation. The perikarya of injured corticospinal neurons atrophied, but there was no evidence of neuronal cell death. We also used CLARITY at single-axon resolution to explore the role of the NMNAT2-SARM1 axonal self-destruction pathway in traumatic axonopathy. When we interfered with this pathway by genetically ablating SARM1 or by pharmacological strategies designed to increase levels of Nicotinamide (Nam), a feedback inhibitor of SARM1, we found a significant reduction in the number of axonal lesions early after injury.
    [Show full text]
  • SARM1-Specific Motifs in the TIR Domain Enable NAD Loss and Regulate Injury-Induced SARM1 Activation
    + PNAS PLUS SARM1-specific motifs in the TIR domain enable NAD loss and regulate injury-induced SARM1 activation Daniel W. Summersa,b, Daniel A. Gibsonb, Aaron DiAntoniob,c,1, and Jeffrey Milbrandta,c,1 aDepartment of Genetics, Washington University in St. Louis, St. Louis, MO 63110; bDepartment of Developmental Biology, Washington University in St. Louis, St. Louis, MO 63110; and cHope Center for Neurological Disorders, Washington University in St. Louis, St. Louis, MO 63110 Edited by Richard H. Goodman, Vollum Institute, Portland, OR, and approved August 19, 2016 (received for review January 27, 2016) Axon injury in response to trauma or disease stimulates a self- in pathological neuron destruction, so a deeper understanding of destruction program that promotes the localized clearance of the molecular mechanism of SARM1 function may identify novel damaged axon segments. Sterile alpha and Toll/interleukin re- targets for the inhibition of this prodestructive factor. ceptor (TIR) motif-containing protein 1 (SARM1) is an evolution- Domain analysis of the SARM1 protein generated a model of arily conserved executioner of this degeneration cascade, also SARM1 activation and function (3, 11, 12). SARM1 possesses a known as Wallerian degeneration; however, the mechanism of Toll/interleukin receptor (TIR) domain that is required for SARM1-dependent neuronal destruction is still obscure. SARM1 SARM1-dependent axon degeneration and cell death. Recently, possesses a TIR domain that is necessary for SARM1 activity. In others and we demonstrated that enforced dimerization of the other proteins, dimerized TIR domains serve as scaffolds for in- SARM1 TIR domain is sufficient to induce local axon de- nate immune signaling.
    [Show full text]
  • Vincristine and Bortezomib Use Distinct Upstream Mechanisms to Activate a Common SARM1-Dependent Axon Degeneration Program
    Vincristine and bortezomib use distinct upstream mechanisms to activate a common SARM1-dependent axon degeneration program Stefanie Geisler, … , Jeffrey Milbrandt, Aaron DiAntonio JCI Insight. 2019;4(17):e129920. https://doi.org/10.1172/jci.insight.129920. Research Article Cell biology Neuroscience Graphical abstract Find the latest version: https://jci.me/129920/pdf RESEARCH ARTICLE Vincristine and bortezomib use distinct upstream mechanisms to activate a common SARM1-dependent axon degeneration program Stefanie Geisler,1,2 Ryan A. Doan,1 Galen C. Cheng,1 Aysel Cetinkaya-Fisgin,3 Shay X. Huang,1 Ahmet Höke,3 Jeffrey Milbrandt,2,4 and Aaron DiAntonio2,5 1Department of Neurology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA. 2Hope Center for Neurological Disorders, Washington University, St. Louis, Missouri, USA. 3Department of Neurology, Johns Hopkins University, Baltimore, Maryland, USA. 4Department of Genetics and 5Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA. Chemotherapy-induced peripheral neuropathy is one of the most prevalent dose-limiting toxicities of anticancer therapy. Development of effective therapies to prevent chemotherapy-induced neuropathies could be enabled by a mechanistic understanding of axonal breakdown following exposure to neuropathy-causing agents. Here, we reveal the molecular mechanisms underlying axon degeneration induced by 2 widely used chemotherapeutic agents with distinct mechanisms of action: vincristine
    [Show full text]
  • Genetic and Cell Biological Studies of Sarm1 in Zebrafish
    Genetic and Cell Biological studies of Sarm1 in Zebrafish Dissertation an der Fakultät für Biologie der Ludwig-Maximilians-Universität München Durchgeführt am Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt Vorgelegt von Weili Tian This work has been done in the laboratory of Dr. Hernán López-Schier at the Research Unit Sensory Biology and Organogenesis, Helmholtz Zentrum München (Munich, Germany) from November 2014 to September 2019. 1. Gutachter: Prof. Dr. Barbara Conradt 2. Gutachter: Prof. Dr. rer. nat. Anja Horn-Bochtler Tag der Einreichung: 17.11.2019 Tag der mündlichen Prüfung: 18.03.2020 Abstract The peripheral nerves that communicate skin, muscle and sensory organs with the brain must maintain functionality throughout life despite frequent stress and injury. Loss of integrity of peripheral neurons and associated glial cells is a common occurrence in severe neurological dysfunctions that include weakness, pain and loss of sensation. Therefore, protecting the nervous system from chronic effects of physical and chemical stress is a pressing clinical challenge. This has sparked intense efforts to exploit molecular inhibitors of axonal and glial destruction. Loss of function of the obligate pro-degenerative protein Sarm1 blocks axon degeneration across species. Thus, Sarm1 is an ideal target for clinical application aimed at reducing axonal degradation in humans. In my thesis, I addressed the impact of Sarm1 deficiency on the development of the peripheral nervous system and glial Schwann cells. Using zebrafish, I show that elimination of Sarm1 is compatible with neural homeostasis, and is glioprotective against chemotherapeutic agents after nerve injury. Specifically, I have discovered that Schwann cells are not essential for the maintenance of severed axons lacking Sarm1, which subsist well beyond the normal timeframe allowed by Sarm1 destructive activity.
    [Show full text]
  • 1 Enrichment of SARM1 Alleles Encoding Variants with Constitutively
    medRxiv preprint doi: https://doi.org/10.1101/2021.06.17.21258268; this version posted June 17, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. All rights reserved. No reuse allowed without permission. Enrichment of SARM1 alleles encoding variants with constitutively hyperactive NADase in patients with ALS and other motor nerve disorders Jonathan Gilley 1, Oscar Jackson 1, Menelaos Pipis 2, Mehrdad A. Estiar 3,4, Ziv Gan-Or 3,4,5, Stephen A. Goutman 6, Matthew B. Harms 7, Julia Kaye 8, Leandro Lima 8, Queen Square Genomics 2, John Ravits 9, Guy A. Rouleau 3,4,5, Stephan Züchner 10, Mary M. Reilly 2 and Michael P. Coleman 1†. 1 John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Forvie Site, Robinson Way, Cambridge, CB2 0PY, UK 2 Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology and The National Hospital for Neurology, London, WC1N 3BG, UK 3 Department of Human Genetics, McGill University, Montreal, H3A 1A1, Canada 4 The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montreal, H3A 2B4, Canada 5 Department of Neurology and Neurosurgery, McGill University, Montreal, H3A 1A1, Canada 6 Department of Neurology, University of Michigan, Ann Arbor, MI, USA 7 Institute for Genomic Medicine, Columbia University, New York, NY100332, USA 8 Finkbeiner lab, Center for Systems and Therapeutics, Bioinformatics Core, Institute of Data Science and Biotechnology, Gladstone Institutes, 1650 Owens Street, San Francisco, CA 94158, USA 9 Department of Neurosciences, University of California, San Diego, La Jolla, CA92093, USA 10 Dr.
    [Show full text]
  • Sarm1 Gene Deficiency Attenuates Diabetic Peripheral Neuropathy In
    Diabetes Page 2 of 37 Sarm1 gene deficiency attenuates diabetic peripheral neuropathy in mice Yalan Cheng1, Jun Liu1, Yi Luan1, Zhiyuan Liu1, Hejin Lai1, Wuling Zhong1, Yale Yang1, Huimin Yu1, Ning Feng1, Hui Wang1, Rui Huang1, Zhishui He1, Menghong Yan1, Fang Zhang1, Yan-Gang Sun2, Hao Yin1, Feifan Guo1, Qiwei Zhai1,3* From: 1 CAS Key Laboratory of Nutrition, Metabolism and Food safety, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031 Shanghai, China; 2 Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 200031 Shanghai, China; 3 School of Life Science and Technology, Shanghai Tech University, 200093 Shanghai, China Correspondence: Qiwei Zhai, PHD Shanghai Institute of Nutrition and Health Shanghai Institutes for Biological Sciences Chinese Academy of Sciences 320 Yueyang Road, Shanghai 200031, China Tel: +86 21 5492 0903 E-mail: [email protected] School of Life Science and Technology Shanghai Tech University, Shanghai 200093, China. Word count abstract: 200 Word count: 4000 Number of tables: 2 Number of figures: 7 1 Diabetes Publish Ahead of Print, published online August 22, 2019 Page 3 of 37 Diabetes Abstract Diabetic peripheral neuropathy (DPN) is the most common complication in both type 1 and type 2 diabetes, but any treatment towards the development of DPN is yet not available. Axon degeneration is an early feature of many peripheral neuropathies, including DPN. Delay of axon degeneration has beneficial effects on various neurodegenerative diseases, but its effect on DPN is yet to be elucidated.
    [Show full text]
  • Mitochondrial Dysfunction Induces Sarm1-Dependent Cell Death in Sensory Neurons
    9338 • The Journal of Neuroscience, July 9, 2014 • 34(28):9338–9350 Cellular/Molecular Mitochondrial Dysfunction Induces Sarm1-Dependent Cell Death in Sensory Neurons Daniel W. Summers,1,2 X Aaron DiAntonio,2,3 and Jeffrey Milbrandt1,3 1Department of Genetics, 2Department of Developmental Biology, and 3Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri 63110 Mitochondrialdysfunctionistheunderlyingcauseofmanyneurologicaldisorders,includingperipheralneuropathies.Mitochondriarely on a proton gradient to generate ATP and interfering with electron transport chain function can lead to the deleterious accumulation of reactive oxygen species (ROS). Notably, loss of mitochondrial potential precedes cellular demise in several programmed cell destruction pathways, including axons undergoing Wallerian degeneration. Here, we demonstrate that mitochondrial depolarization triggers axon degeneration and cell death in primary mouse sensory neurons. These degenerative events are not blocked by inhibitors of canonical programmed cell death pathways such as apoptosis, necroptosis, and parthanatos. Instead, the axodestructive factor Sarm1 is required for this axon degeneration and cell death. In the absence of Sarm1, the mitochondrial poison CCCP still induces depolarization of mitochondria, ATP depletion, calcium influx, and the accumulation of ROS, yet cell death and axon degeneration are blocked. The survivaloftheseneuronsdespitetheaccumulationofROSindicatesthatSarm1actsdownstreamofROSgeneration.Indeed,lossofSarm1
    [Show full text]
  • Absence of SARM1 Rescues Development and Survival of NMNAT2-Deficient Axons
    Report Absence of SARM1 Rescues Development and Survival of NMNAT2-Deficient Axons Graphical Abstract Authors Jonathan Gilley, Giuseppe Orsomando, Isabel Nascimento-Ferreira, Michael P. Coleman Correspondence [email protected] In Brief Gilley et al. find that SARM1 promotes axon degeneration after NMNAT2 depletion and limits outgrowth of axons constitutively lacking NMNAT2. The NMNAT substrate NMN also appears to be pro-degenerative in both situations. Restricted outgrowth of NMNAT2- deficient axons is thus mechanistically related to the degeneration of established axons. Highlights d SARM1 is required for WLDS-sensitive axon degeneration caused by NMNAT2 depletion d SARM1 deficiency rescues developmental axon defects caused by a lack of NMNAT2 d Mice lacking NMNAT2 and SARM1 are viable d Lowering NMN by inhibiting NAMPT partially rescues growth of NMNAT2-deficient axons Gilley et al., 2015, Cell Reports 10, 1974–1981 March 31, 2015 ª2015 The Authors http://dx.doi.org/10.1016/j.celrep.2015.02.060 Cell Reports Report Absence of SARM1 Rescues Development and Survival of NMNAT2-Deficient Axons Jonathan Gilley,1 Giuseppe Orsomando,2 Isabel Nascimento-Ferreira,1 and Michael P. Coleman1,* 1Signalling Programme, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK 2Department of Clinical Sciences (DISCO), Section of Biochemistry, Polytechnic University of Marche, Via Ranieri 67, Ancona 60131, Italy *Correspondence: [email protected] http://dx.doi.org/10.1016/j.celrep.2015.02.060 This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/). SUMMARY tion with opposing influences, is still unknown (Conforti et al., 2014).
    [Show full text]