SARM1

Last updated
SARM1
Identifiers
Aliases SARM1 , MyD88-5, SAMD2, SARM, sterile alpha and TIR motif containing 1, hHsTIR
External IDs OMIM: 607732 MGI: 2136419 HomoloGene: 9015 GeneCards: SARM1
Orthologs
SpeciesHumanMouse
Entrez
Ensembl
UniProt
RefSeq (mRNA)

NM_015077

NM_001168521
NM_172795

RefSeq (protein)

NP_055892
NP_055892.2

NP_001161993
NP_766383

Location (UCSC) Chr 17: 28.36 – 28.4 Mb Chr 11: 78.36 – 78.39 Mb
PubMed search [3] [4]
Wikidata
View/Edit Human View/Edit Mouse

Sterile alpha and TIR motif containing 1 Is an enzyme that in humans is encoded by the SARM1 gene. It is the most evolutionarily conserved member of the Toll/Interleukin receptor-1 (TIR) family. [5] [6] SARM1's TIR domain has intrinsic NADase enzymatic activity that is highly conserved from archaea, plants, nematode worms, fruit flies, and humans. [7] [8] [9] In mammals, SARM1 is highly expressed in neurons, where it resides in both cell bodies and axons, and can be associated with mitochondria. [10]

Contents

Function

While SARM1 has been studied as a Toll-like receptor adaptor protein in an immune context, its most well-studied function in mammals is as a sensor of metabolic stress and an executioner of neuronal cell body and axon death. [5] [11] [12] [13] [14] [15] Because SARM1 is highly expressed in the nervous system, most studies of SARM1 focus on neuron degeneration, but some SARM1 can be found in other tissues, notably macrophages and T cells. [16] [17] By generating cADPR or NAADP, SARM1 may function as a Ca2+-signaling enzyme similar to CD38. [18] [19] [20] [21] [22]

Regulation of enzymatic activity

SARM1's TIR domain is a multi-functional NAD(P)ase enzyme capable of hydrolyzing NAD+ or NADP, cyclizing NAD+ or NADP to form cADPR or cADPRP, and transglycosidation (base exchange) of NAD+ or NADP with free pyridines to form molecules such as NAADP. [6] [8] [23] [20] [24] [21] [25] For NAD+, The transglycosidation (base exchange) activity of SARM1 extends beyond simple pyridines and includes many heterocyclic nucleophilic bases. [26]

SARM1's enzymatic activity can be regulated at the TIR domain orthosteric site by naturally occurring metabolites such as nicotinamide, NADP, and nicotinic acid riboside. [6] [21] [27] Non-endogenous small chemical molecules have also been shown to inhibit SARM1's enzymatic activity at or near the orthosteric site. [26] [28] [29] [30] [31]

In addition, SARM1's enzymatic activity can be regulated by its allosteric site at the ARM domain, which can bind to NMN or NAD+. [13] [26] The ratio of NMN/NAD+ in cells determines SARM1's enzymatic activity. [13] [21] [32] [33] [34] A chemically-modified cell permeable version of NMN, CZ-48, likely activates SARM1 via interacting with this allosteric region. [20] [35] Two long-studied neurotoxins, Vacor and 3-acetylpyridine, cause neurodegeneration by activating SARM1. Both Vacor and 3-acetylpyridine can be modified by NAMPT to become their mononucleotide versions (Vacor-MN or 3-AP-MN) that bind to SARM1's allosteric ARM domain region and activate its TIR domain NADase activity. [36] [37] When NAD+ levels are low, nicotinic acid mononucleotide (NaMN) can bind to the allosteric region and inhibit SARM1 activity, [38] thus explaining the potent axon protection provided by treating neurons with the NaMN precursor nicotinic acid riboside (NaR) while inhibiting NAMPT. [39] Chemical screening approaches have also identified covalent inhibitors of SARM1's allosteric ARM domain region. [24] [40]

Other pro-degeneration signaling pathways, such as the MAP kinase pathway, have been linked to SARM1 activation. MAPK signaling has been shown to promote the loss of NMNAT2, thereby promoting SARM1 activation. [41] [42] [43] SARM1 activation also triggers the MAP kinase cascade, indicating some form of feedback loop may exist. [44]

Relevance to human disease

Possible implications of the SARM1 pathway with regard to human health may be found in animal models of neurodegeneration, where loss of SARM1 is neuroprotective in models of traumatic brain injury, [45] [46] [47] [48] [49] [31] [50] [51] chemotherapy-induced neuropathy, [52] [53] [54] [29] [55] [56] [31] diabetic neuropathy, [56] [57] degenerative eye conditions, [58] [59] [60] [61] [62] [63] [64] drug-induced Schwann cell death, [65] Charcot-Marie-Tooth disease, [66] and hereditary spastic paraplegia. [67]

Loss-of-function alleles of the SARM1 gene also occur naturally in the human population, potentially altering susceptibility to various neurological conditions. [68]

Specific mutations in the human NMNAT2 gene, encoding a key regulator of SARM1 activity, have linked the Wallerian degeneration mechanism to two human neurological diseases - fetal akinesia deformation sequence [69] and childhood-onset polyneuropathy with erythromelalgia. [70] Mutations in the human SARM1 gene that result in SARM1 protein with constitutive NADase activity have been reported in patients with amyotrophic lateral sclerosis (ALS). [71] [72]

Wallerian degeneration pathway

SARM1 protein plays a central role in the Wallerian degeneration pathway. The role for this gene in the Wallerian degeneration pathway was first identified in a Drosophila melanogaster mutagenesis screen, [11] and subsequently genetic knockout of its homologue in mice showed robust protection of transected axons comparable to that of WldS mutation (a mouse mutation resulting in delayed Wallerian degeneration). [11] [12] Loss of SARM1 in human iPSC-derived neurons is also axon protective. [73]

The SARM1 protein has a mitochondrial localization signal, an auto-inhibitory N-terminus region consisting of armadillo (ARM)/HEAT motifs, two sterile alpha motif domains (SAM) responsible for multimerization, and a C-terminal Toll/Interleukin-1 receptor (TIR) domain that possesses enzymatic activity. [12] The functional unit of SARM1 is an octameric ring. [74] In healthy neurons, SARM1's enzyme activity is mostly autoinhibited through intramolecular and intermolecular interactions between ARM-ARM, ARM-SAM and ARM-TIR domains, as well as interactions between a duplex of octameric rings. [75] [35] [15] [14] [13] [76]

SARM1's enzymatic activity is critically tuned to the activity of another axonal enzyme, NMNAT2. NMNAT2 is a labile protein in axons and is rapidly degraded after axon injury. [77] NMNAT2 is a transferase that uses ATP to convert nicotinamide mononucleotide (NMN) into NAD+. Remarkably, genetic loss of NMNAT2 in mice leads to embryonic lethality that can be fully rescued by genetic loss of SARM1, indicating that SARM1 acts downstream of NMNAT2. [78] Thus, when NMNAT2 is degraded after axon injury, SARM1 is activated. Conversely, overexpression of the WldS protein (which contains functional NMNAT1), axon-targeted NMNAT1, or NMNAT2 itself can protect axons and keep SARM1 from being activated. [79] [80] [81] [82] [83] [84] [85] [86] [87] These findings lead to the hypothesis and subsequent demonstration that NMNAT2's substrate NMN, which should increase when NMNAT2 is degraded after injury, can promote axon degeneration via SARM1. [88] [89] Further studies revealed that NMN could activate SARM1's enzymatic activity. [20] [35] Through a combination of structural, biochemical, biophysical, and cellular assays, it was revealed that SARM1 is tuned to NMNAT activity by sensing the ratio of NMN/NAD+. [13] This ratio is sensed by an allosteric region in SARM1's ARM domain region that can bind either NMN or NAD+. NAD+ binding is associated with SARM1's auto-inhibited state, [13] [14] [15] while NMN binding to the allosteric region results in a conformational change in the ARM domain that allows for multimerization of SARM1's TIR domains and enzymatic activation. [13] [26] [33] [34]

SARM1 activation locally triggers a rapid collapse of NAD+ levels in the distal section of the injured axon, which then undergoes degeneration. [90] This collapse in NAD+ levels was later shown to be due to SARM1's TIR domain having intrinsic NAD+ cleavage activity. [6] SARM1 can hydrolyze NAD+ into nicotinamide and adenosine diphosphate ribose (ADPR), generate cyclic ADPR (cADPR), or mediate a base-exchange reaction with ADPR and free pyridine-ring containing bases, like nicotinamide. [6] [19] [20] [21] Activation of SARM1's NADase activity is necessary and sufficient to collapse NAD+ levels and initiate the Wallerian degeneration pathway. [90] [6] NAD+ loss is followed by depletion of ATP, defects in mitochondrial movement and depolarization, calcium influx, externalization of phosphatidylserine, and loss of membrane permeability prior to catastrophic axonal self-destruction. [91]

SARM1 activation due to loss of NMNAT2 in neurons also elicits a pro-degenerative neuroinflammatory response from peripheral nervous system macrophages and central nervous system astrocytes and microglia. [92] [93] [ unreliable source ]

Related Research Articles

<span class="mw-page-title-main">Axon</span> Long projection on a neuron that conducts signals to other neurons

An axon or nerve fiber is a long, slender projection of a nerve cell, or neuron, in vertebrates, that typically conducts electrical impulses known as action potentials away from the nerve cell body. The function of the axon is to transmit information to different neurons, muscles, and glands. In certain sensory neurons, such as those for touch and warmth, the axons are called afferent nerve fibers and the electrical impulse travels along these from the periphery to the cell body and from the cell body to the spinal cord along another branch of the same axon. Axon dysfunction can be the cause of many inherited and acquired neurological disorders that affect both the peripheral and central neurons. Nerve fibers are classed into three types – group A nerve fibers, group B nerve fibers, and group C nerve fibers. Groups A and B are myelinated, and group C are unmyelinated. These groups include both sensory fibers and motor fibers. Another classification groups only the sensory fibers as Type I, Type II, Type III, and Type IV.

<span class="mw-page-title-main">Charcot–Marie–Tooth disease</span> Neuromuscular disease

Charcot–Marie–Tooth disease (CMT) is a hereditary motor and sensory neuropathy of the peripheral nervous system characterized by progressive loss of muscle tissue and touch sensation across various parts of the body. This disease is the most commonly inherited neurological disorder, affecting about one in 2,500 people. It is named after those who classically described it: the Frenchman Jean-Martin Charcot (1825–1893), his pupil Pierre Marie (1853–1940), and the Briton Howard Henry Tooth (1856–1925).

<span class="mw-page-title-main">Nicotinamide adenine dinucleotide</span> Chemical compound which is reduced and oxidized

Nicotinamide adenine dinucleotide (NAD) is a coenzyme central to metabolism. Found in all living cells, NAD is called a dinucleotide because it consists of two nucleotides joined through their phosphate groups. One nucleotide contains an adenine nucleobase and the other, nicotinamide. NAD exists in two forms: an oxidized and reduced form, abbreviated as NAD+ and NADH (H for hydrogen), respectively.

<span class="mw-page-title-main">Wallerian degeneration</span> Biological process of axonal degeneration

Wallerian degeneration is an active process of degeneration that results when a nerve fiber is cut or crushed and the part of the axon distal to the injury degenerates. A related process of dying back or retrograde degeneration known as 'Wallerian-like degeneration' occurs in many neurodegenerative diseases, especially those where axonal transport is impaired such as ALS and Alzheimer's disease. Primary culture studies suggest that a failure to deliver sufficient quantities of the essential axonal protein NMNAT2 is a key initiating event.

<span class="mw-page-title-main">Diffuse axonal injury</span> Medical condition

Diffuse axonal injury (DAI) is a brain injury in which scattered lesions occur over a widespread area in white matter tracts as well as grey matter. DAI is one of the most common and devastating types of traumatic brain injury and is a major cause of unconsciousness and persistent vegetative state after severe head trauma. It occurs in about half of all cases of severe head trauma and may be the primary damage that occurs in concussion. The outcome is frequently coma, with over 90% of patients with severe DAI never regaining consciousness. Those who awaken from the coma often remain significantly impaired.

Neurofilaments (NF) are classed as type IV intermediate filaments found in the cytoplasm of neurons. They are protein polymers measuring 10 nm in diameter and many micrometers in length. Together with microtubules (~25 nm) and microfilaments (7 nm), they form the neuronal cytoskeleton. They are believed to function primarily to provide structural support for axons and to regulate axon diameter, which influences nerve conduction velocity. The proteins that form neurofilaments are members of the intermediate filament protein family, which is divided into six types based on their gene organization and protein structure. Types I and II are the keratins which are expressed in epithelia. Type III contains the proteins vimentin, desmin, peripherin and glial fibrillary acidic protein (GFAP). Type IV consists of the neurofilament proteins NF-L, NF-M, NF-H and α-internexin. Type V consists of the nuclear lamins, and type VI consists of the protein nestin. The type IV intermediate filament genes all share two unique introns not found in other intermediate filament gene sequences, suggesting a common evolutionary origin from one primitive type IV gene.

<span class="mw-page-title-main">CD38</span> Protein-coding gene in the species Homo sapiens

CD38 (cluster of differentiation 38), also known as cyclic ADP ribose hydrolase is a glycoprotein found on the surface of many immune cells (white blood cells), including CD4+, CD8+, B lymphocytes and natural killer cells. CD38 also functions in cell adhesion, signal transduction and calcium signaling.

<span class="mw-page-title-main">Cyclic ADP-ribose</span> Chemical compound

Cyclic ADP-ribose, frequently abbreviated as cADPR, is a cyclic adenine nucleotide (like cAMP) with two phosphate groups present on 5' OH of the adenosine (like ADP), further connected to another ribose at the 5' position, which, in turn, closes the cycle by glycosidic bonding to the nitrogen 1 (N1) of the same adenine base (whose position N9 has the glycosidic bond to the other ribose). The N1-glycosidic bond to adenine is what distinguishes cADPR from ADP-ribose (ADPR), the non-cyclic analog. cADPR is produced from nicotinamide adenine dinucleotide (NAD+) by ADP-ribosyl cyclases (EC 3.2.2.5) as part of a second messenger system.

Neuroregeneration involves the regrowth or repair of nervous tissues, cells or cell products. Neuroregenerative mechanisms may include generation of new neurons, glia, axons, myelin, or synapses. Neuroregeneration differs between the peripheral nervous system (PNS) and the central nervous system (CNS) by the functional mechanisms involved, especially in the extent and speed of repair. When an axon is damaged, the distal segment undergoes Wallerian degeneration, losing its myelin sheath. The proximal segment can either die by apoptosis or undergo the chromatolytic reaction, which is an attempt at repair. In the CNS, synaptic stripping occurs as glial foot processes invade the dead synapse.

<span class="mw-page-title-main">MFN2</span> Protein-coding gene in the species Homo sapiens

Mitofusin-2 is a protein that in humans is encoded by the MFN2 gene. Mitofusins are GTPases embedded in the outer membrane of the mitochondria. In mammals MFN1 and MFN2 are essential for mitochondrial fusion. In addition to the mitofusins, OPA1 regulates inner mitochondrial membrane fusion, and DRP1 is responsible for mitochondrial fission.

<span class="mw-page-title-main">Nicotinamide phosphoribosyltransferase</span> Human protein and coding gene

Nicotinamide phosphoribosyltransferase, formerly known as pre-B-cell colony-enhancing factor 1 (PBEF1) or visfatin for its extracellular form (eNAMPT), is an enzyme that in humans is encoded by the NAMPT gene. The intracellular form of this protein (iNAMPT) is the rate-limiting enzyme in the nicotinamide adenine dinucleotide (NAD+) salvage pathway that converts nicotinamide to nicotinamide mononucleotide (NMN) which is responsible for most of the NAD+ formation in mammals. iNAMPT can also catalyze the synthesis of NMN from phosphoribosyl pyrophosphate (PRPP) when ATP is present. eNAMPT has been reported to be a cytokine (PBEF) that activates TLR4, that promotes B cell maturation, and that inhibits neutrophil apoptosis.

<span class="mw-page-title-main">Nicotinamide-nucleotide adenylyltransferase</span>

In enzymology, nicotinamide-nucleotide adenylyltransferase (NMNAT) (EC 2.7.7.1) are enzymes that catalyzes the chemical reaction

<span class="mw-page-title-main">NMNAT1</span> Protein-coding gene in the species Homo sapiens

Nicotinamide mononucleotide adenylyltransferase 1 (NMNAT1) is an enzyme that in humans is encoded by the nmnat1 gene. It is a member of the nicotinamide-nucleotide adenylyltransferases (NMNATs) which catalyze nicotinamide adenine dinucleotide (NAD) synthesis.

<span class="mw-page-title-main">NMNAT2</span> Protein-coding gene in the species Homo sapiens

Nicotinamide mononucleotide adenylyltransferase 2 (NMNAT2) is an enzyme that in humans is encoded by the NMNAT2 gene.

<span class="mw-page-title-main">NINJ1</span> Protein-coding gene in the species Homo sapiens

Ninjurin-1 is a protein that in humans is encoded by the NINJ1 gene.

Sirtuin-activating compounds (STAC) are chemical compounds having an effect on sirtuins, a group of enzymes that use NAD+ to remove acetyl groups from proteins. They are caloric restriction mimetic compounds that may be helpful in treating various aging-related diseases.

<span class="mw-page-title-main">Toll-interleukin receptor</span> Intracellular signaling domain

The toll-interleukin-1 receptor (TIR) homology domain is an intracellular signaling domain found in MyD88, SARM1, interleukin-1 receptors, toll receptors and many plant R proteins. It contains three highly conserved regions, and mediates protein-protein interactions between the toll-like receptors (TLRs) and signal-transduction components. TIR-like motifs are also found in plant proteins where they are involved in resistance to disease and in bacteria where they are associated with virulence. When activated, TIR domains recruit cytoplasmic adaptor proteins MyD88 (UniProt Q99836) and TOLLIP (toll-interacting protein, UniProt Q9H0E2). In turn, these associate with various kinases to set off signaling cascades. Some TIR domains have also been found to have intrinsic NAD+ cleavage activity, such as in SARM1. In the case of SARM1, the TIR NADase activity leads to the production of Nam, ADPR and cADPR and the activation of downstream pathways involved in Wallerian degeneration and neuron death.

Members of the very wide interleukin-1 receptor (IL-1R) family are characterized by extracellular immunoglobulin-like domains and intracellular Toll/Interleukin-1R (TIR) domain. It is a group of structurally homologous proteins, conserved throughout the species as it was identified from plants to mammals. Proteins of this family play important role in host defence, injury and stress. There are four main groups of TIR domain-containing proteins in animals; Toll-like receptors, Interleukin-1 receptor (IL-1R), cytosolic adaptor proteins and insect and nematode Toll. Each of these groups is involved mainly in host defence; Toll receptors are also involved in embryogenesis.

Preferential motor reinnervation (PMR) refers to the tendency of a regenerating axon in the peripheral nervous system (PNS) to reinnervate a motor pathway as opposed to a somatosensory pathway. PMR affects how nerves regenerate and reinnervate within the PNS after surgical procedures or traumatic injuries. It is important to understand in order to further develop axonal regrowth surgical techniques. Further research of preferential motor reinnervation will lead to a better understanding of peripheral nervous system function in the human body regarding cell roles and abilities.

<span class="mw-page-title-main">Nicotinamide mononucleotide</span> Chemical compound

Nicotinamide mononucleotide is a nucleotide derived from ribose, nicotinamide, nicotinamide riboside and niacin. In humans, several enzymes use NMN to generate nicotinamide adenine dinucleotide (NADH). In mice, it has been proposed that NMN is absorbed via the small intestine within 10 minutes of oral uptake and converted to nicotinamide adenine dinucleotide (NAD+) through the Slc12a8 transporter. However, this observation has been challenged, and the matter remains unsettled.

References

  1. 1 2 3 GRCh38: Ensembl release 89: ENSG00000004139 Ensembl, May 2017
  2. 1 2 3 GRCm38: Ensembl release 89: ENSMUSG00000050132 Ensembl, May 2017
  3. "Human PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  4. "Mouse PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  5. 1 2 Carty M, Bowie AG (March 2019). "SARM: From immune regulator to cell executioner". Biochemical Pharmacology. 161: 52–62. doi:10.1016/j.bcp.2019.01.005. PMID   30633870. S2CID   58613555.
  6. 1 2 3 4 5 6 Essuman K, Summers DW, Sasaki Y, Mao X, DiAntonio A, Milbrandt J (March 2017). "The SARM1 Toll/Interleukin-1 Receptor Domain Possesses Intrinsic NAD+ Cleavage Activity that Promotes Pathological Axonal Degeneration". Neuron. 93 (6): 1334–1343.e5. doi:10.1016/j.neuron.2017.02.022. PMC   6284238 . PMID   28334607.
  7. Essuman K, Summers DW, Sasaki Y, Mao X, Yim AK, DiAntonio A, et al. (February 2018). "TIR Domain Proteins Are an Ancient Family of NAD+-Consuming Enzymes". Current Biology. 28 (3): 421–430.e4. Bibcode:2018CBio...28E.421E. doi:10.1016/j.cub.2017.12.024. PMC   5802418 . PMID   29395922.
  8. 1 2 Wan L, Essuman K, Anderson RG, Sasaki Y, Monteiro F, Chung EH, et al. (August 2019). "TIR domains of plant immune receptors are NAD+-cleaving enzymes that promote cell death". Science. 365 (6455): 799–803. Bibcode:2019Sci...365..799W. doi:10.1126/science.aax1771. PMC   7045805 . PMID   31439793.
  9. Zhang Q, Zmasek CM, Cai X, Godzik A (April 2011). "TIR domain-containing adaptor SARM is a late addition to the ongoing microbe-host dialog". Developmental and Comparative Immunology. 35 (4): 461–468. doi:10.1016/j.dci.2010.11.013. PMC   3085110 . PMID   21110998.
  10. Gerdts J, Summers DW, Milbrandt J, DiAntonio A (February 2016). "Axon Self-Destruction: New Links among SARM1, MAPKs, and NAD+ Metabolism". Neuron. 89 (3): 449–460. doi:10.1016/j.neuron.2015.12.023. PMC   4742785 . PMID   26844829.
  11. 1 2 3 Osterloh JM, Yang J, Rooney TM, Fox AN, Adalbert R, Powell EH, et al. (July 2012). "dSarm/Sarm1 is required for activation of an injury-induced axon death pathway". Science. 337 (6093): 481–484. Bibcode:2012Sci...337..481O. doi:10.1126/science.1223899. PMC   5225956 . PMID   22678360.
  12. 1 2 3 Gerdts J, Summers DW, Sasaki Y, DiAntonio A, Milbrandt J (August 2013). "Sarm1-mediated axon degeneration requires both SAM and TIR interactions". The Journal of Neuroscience. 33 (33): 13569–13580. doi:10.1523/JNEUROSCI.1197-13.2013. PMC   3742939 . PMID   23946415.
  13. 1 2 3 4 5 6 7 Figley MD, Gu W, Nanson JD, Shi Y, Sasaki Y, Cunnea K, et al. (April 2021). "SARM1 is a metabolic sensor activated by an increased NMN/NAD+ ratio to trigger axon degeneration". Neuron. 109 (7): 1118–1136.e11. doi:10.1016/j.neuron.2021.02.009. PMC   8174188 . PMID   33657413.
  14. 1 2 3 Jiang Y, Liu T, Lee CH, Chang Q, Yang J, Zhang Z (December 2020). "The NAD+-mediated self-inhibition mechanism of pro-neurodegenerative SARM1". Nature. 588 (7839): 658–663. Bibcode:2020Natur.588..658J. doi:10.1038/s41586-020-2862-z. PMID   33053563. S2CID   222420804.
  15. 1 2 3 Sporny M, Guez-Haddad J, Khazma T, Yaron A, Dessau M, Shkolnisky Y, et al. (November 2020). "Structural basis for SARM1 inhibition and activation under energetic stress". eLife. 9. doi: 10.7554/eLife.62021 . PMC   7688312 . PMID   33185189.
  16. Covarrubias AJ, Perrone R, Grozio A, Verdin E (February 2021). "NAD+ metabolism and its roles in cellular processes during ageing". Nature Reviews. Molecular Cell Biology. 22 (2): 119–141. doi:10.1038/s41580-020-00313-x. PMC   7963035 . PMID   33353981.
  17. Doran CG, Sugisawa R, Carty M, Roche F, Fergus C, Hokamp K, et al. (December 2021). "CRISPR/Cas9-mediated SARM1 knockout and epitope-tagged mice reveal that SARM1 does not regulate nuclear transcription, but is expressed in macrophages". The Journal of Biological Chemistry. 297 (6): 101417. doi: 10.1016/j.jbc.2021.101417 . PMC   8661062 . PMID   34793837.
  18. Lee HC, Zhao YJ (December 2019). "Resolving the topological enigma in Ca2+ signaling by cyclic ADP-ribose and NAADP". The Journal of Biological Chemistry. 294 (52): 19831–19843. doi: 10.1074/jbc.REV119.009635 . PMC   6937575 . PMID   31672920.
  19. 1 2 Sasaki Y, Engber TM, Hughes RO, Figley MD, Wu T, Bosanac T, et al. (July 2020). "cADPR is a gene dosage-sensitive biomarker of SARM1 activity in healthy, compromised, and degenerating axons". Experimental Neurology. 329: 113252. doi:10.1016/j.expneurol.2020.113252. PMC   7302925 . PMID   32087251.
  20. 1 2 3 4 5 Zhao ZY, Xie XJ, Li WH, Liu J, Chen Z, Zhang B, et al. (May 2019). "A Cell-Permeant Mimetic of NMN Activates SARM1 to Produce Cyclic ADP-Ribose and Induce Non-apoptotic Cell Death". iScience. 15: 452–466. Bibcode:2019iSci...15..452Z. doi:10.1016/j.isci.2019.05.001. PMC   6531917 . PMID   31128467.
  21. 1 2 3 4 5 Angeletti C, Amici A, Gilley J, Loreto A, Trapanotto AG, Antoniou C, et al. (February 2022). "SARM1 is a multi-functional NAD(P)ase with prominent base exchange activity, all regulated bymultiple physiologically relevant NAD metabolites". iScience. 25 (2): 103812. Bibcode:2022iSci...25j3812A. doi:10.1016/j.isci.2022.103812. PMC   8844822 . PMID   35198877.
  22. Li Y, Pazyra-Murphy MF, Avizonis D, de Sá Tavares Russo M, Tang S, Chen CY, et al. (February 2022). "Sarm1 activation produces cADPR to increase intra-axonal Ca++ and promote axon degeneration in PIPN". The Journal of Cell Biology. 221 (2): e202106080. doi:10.1083/jcb.202106080. PMC   8704956 . PMID   34935867.
  23. Horsefield S, Burdett H, Zhang X, Manik MK, Shi Y, Chen J, et al. (August 2019). "NAD+ cleavage activity by animal and plant TIR domains in cell death pathways". Science. 365 (6455): 793–799. Bibcode:2019Sci...365..793H. doi:10.1126/science.aax1911. hdl: 10072/393098 . PMID   31439792. S2CID   201616651.
  24. 1 2 Li WH, Huang K, Cai Y, Wang QW, Zhu WJ, Hou YN, et al. (May 2021). "Permeant fluorescent probes visualize the activation of SARM1 and uncover an anti-neurodegenerative drug candidate". eLife. 10. doi: 10.7554/eLife.67381 . PMC   8143800 . PMID   33944777.
  25. Loring HS, Icso JD, Nemmara VV, Thompson PR (March 2020). "Initial Kinetic Characterization of Sterile Alpha and Toll/Interleukin Receptor Motif-Containing Protein 1". Biochemistry. 59 (8): 933–942. doi:10.1021/acs.biochem.9b01078. PMC   7085114 . PMID   32049506.
  26. 1 2 3 4 Shi Y, Kerry PS, Nanson JD, Bosanac T, Sasaki Y, Krauss R, et al. (May 2022). "Structural basis of SARM1 activation, substrate recognition, and inhibition by small molecules". Molecular Cell. 82 (9): 1643–1659.e10. doi:10.1016/j.molcel.2022.03.007. PMC   9188649 . PMID   35334231.
  27. Zhao YJ, He WM, Zhao ZY, Li WH, Wang QW, Hou YN, et al. (December 2021). "Acidic pH irreversibly activates the signaling enzyme SARM1". The FEBS Journal. 288 (23): 6783–6794. doi: 10.1111/febs.16104 . PMID   34213829. S2CID   235707670.
  28. Hughes RO, Bosanac T, Mao X, Engber TM, DiAntonio A, Milbrandt J, et al. (January 2021). "Small Molecule SARM1 Inhibitors Recapitulate the SARM1-/- Phenotype and Allow Recovery of a Metastable Pool of Axons Fated to Degenerate". Cell Reports. 34 (1): 108588. doi:10.1016/j.celrep.2020.108588. PMC   8179325 . PMID   33406435.
  29. 1 2 Bosanac T, Hughes RO, Engber T, Devraj R, Brearley A, Danker K, et al. (November 2021). "Pharmacological SARM1 inhibition protects axon structure and function in paclitaxel-induced peripheral neuropathy". Brain. 144 (10): 3226–3238. doi: 10.1093/brain/awab184 . PMC   8634121 . PMID   33964142.
  30. Loring HS, Parelkar SS, Mondal S, Thompson PR (September 2020). "Identification of the first noncompetitive SARM1 inhibitors". Bioorganic & Medicinal Chemistry. 28 (18): 115644. doi:10.1016/j.bmc.2020.115644. PMC   7443514 . PMID   32828421.
  31. 1 2 3 Bratkowski M, Burdett TC, Danao J, Wang X, Mathur P, Gu W, et al. (September 2022). "Uncompetitive, adduct-forming SARM1 inhibitors are neuroprotective in preclinical models of nerve injury and disease". Neuron. 110 (22): S0896–6273(22)00749–8. doi: 10.1016/j.neuron.2022.08.017 . PMID   36087583. S2CID   252167671.
  32. Alexandris AS, Ryu J, Rajbhandari L, Harlan R, McKenney J, Wang Y, et al. (September 2022). "Protective effects of NAMPT or MAPK inhibitors and NaR on Wallerian degeneration of mammalian axons". Neurobiology of Disease. 171: 105808. doi: 10.1016/j.nbd.2022.105808 . PMC   10621467 . PMID   35779777. S2CID   250122204.
  33. 1 2 Llobet Rosell A, Paglione M, Gilley J, Kocia M, Perillo G, Gasparrini M, et al. (December 2022). "The NAD+ precursor NMN activates dSarm to trigger axon degeneration in Drosophila". eLife. 11: e80245. doi: 10.7554/eLife.80245 . PMC   9788811 . PMID   36476387.
  34. 1 2 Hou YN, Cai Y, Li WH, He WM, Zhao ZY, Zhu WJ, et al. (December 2022). "A conformation-specific nanobody targeting the nicotinamide mononucleotide-activated state of SARM1". Nature Communications. 13 (1): 7898. Bibcode:2022NatCo..13.7898H. doi:10.1038/s41467-022-35581-y. PMC   9780360 . PMID   36550129.
  35. 1 2 3 Bratkowski M, Xie T, Thayer DA, Lad S, Mathur P, Yang YS, et al. (August 2020). "Structural and Mechanistic Regulation of the Pro-degenerative NAD Hydrolase SARM1". Cell Reports. 32 (5): 107999. doi: 10.1016/j.celrep.2020.107999 . PMID   32755591.
  36. Loreto A, Angeletti C, Gu W, Osborne A, Nieuwenhuis B, Gilley J, et al. (December 2021). "Neurotoxin-mediated potent activation of the axon degeneration regulator SARM1". eLife. 10: e72823. doi: 10.7554/eLife.72823 . PMC   8758145 . PMID   34870595.
  37. Wu T, Zhu J, Strickland A, Ko KW, Sasaki Y, Dingwall CB, et al. (October 2021). "Neurotoxins subvert the allosteric activation mechanism of SARM1 to induce neuronal loss". Cell Reports. 37 (3): 109872. doi:10.1016/j.celrep.2021.109872. PMC   8638332 . PMID   34686345.
  38. Sasaki Y, Zhu J, Shi Y, Gu W, Kobe B, Ve T, et al. (November 2021). "Nicotinic acid mononucleotide is an allosteric SARM1 inhibitor promoting axonal protection". Experimental Neurology. 345: 113842. doi:10.1016/j.expneurol.2021.113842. PMC   8571713 . PMID   34403688. S2CID   237012029.
  39. Liu HW, Smith CB, Schmidt MS, Cambronne XA, Cohen MS, Migaud ME, et al. (October 2018). "Pharmacological bypass of NAD+ salvage pathway protects neurons from chemotherapy-induced degeneration". Proceedings of the National Academy of Sciences of the United States of America. 115 (42): 10654–10659. Bibcode:2018PNAS..11510654L. doi: 10.1073/pnas.1809392115 . PMC   6196523 . PMID   30257945.
  40. Feldman HC, Merlini E, Guijas C, DeMeester KE, Njomen E, Kozina EM, et al. (August 2022). "Selective inhibitors of SARM1 targeting an allosteric cysteine in the autoregulatory ARM domain". Proceedings of the National Academy of Sciences of the United States of America. 119 (35): e2208457119. Bibcode:2022PNAS..11908457F. doi: 10.1073/pnas.2208457119 . PMC   9436332 . PMID   35994671.
  41. Walker LJ, Summers DW, Sasaki Y, Brace EJ, Milbrandt J, DiAntonio A (January 2017). "MAPK signaling promotes axonal degeneration by speeding the turnover of the axonal maintenance factor NMNAT2". eLife. 6. doi: 10.7554/eLife.22540 . PMC   5241118 . PMID   28095293.
  42. Summers DW, Milbrandt J, DiAntonio A (September 2018). "Palmitoylation enables MAPK-dependent proteostasis of axon survival factors". Proceedings of the National Academy of Sciences of the United States of America. 115 (37): E8746–E8754. Bibcode:2018PNAS..115E8746S. doi: 10.1073/pnas.1806933115 . PMC   6140512 . PMID   30150401.
  43. Summers DW, Frey E, Walker LJ, Milbrandt J, DiAntonio A (February 2020). "DLK Activation Synergizes with Mitochondrial Dysfunction to Downregulate Axon Survival Factors and Promote SARM1-Dependent Axon Degeneration". Molecular Neurobiology. 57 (2): 1146–1158. doi:10.1007/s12035-019-01796-2. PMC   7035184 . PMID   31696428.
  44. Yang J, Wu Z, Renier N, Simon DJ, Uryu K, Park DS, et al. (January 2015). "Pathological axonal death through a MAPK cascade that triggers a local energy deficit". Cell. 160 (1–2): 161–176. doi:10.1016/j.cell.2014.11.053. PMC   4306654 . PMID   25594179.
  45. Henninger N, Bouley J, Sikoglu EM, An J, Moore CM, King JA, et al. (April 2016). "Attenuated traumatic axonal injury and improved functional outcome after traumatic brain injury in mice lacking Sarm1". Brain. 139 (Pt 4): 1094–1105. doi:10.1093/brain/aww001. PMC   5006226 . PMID   26912636.
  46. Ziogas NK, Koliatsos VE (April 2018). "Primary Traumatic Axonopathy in Mice Subjected to Impact Acceleration: A Reappraisal of Pathology and Mechanisms with High-Resolution Anatomical Methods". The Journal of Neuroscience. 38 (16): 4031–4047. doi:10.1523/JNEUROSCI.2343-17.2018. PMC   6705930 . PMID   29567804.
  47. Marion CM, McDaniel DP, Armstrong RC (November 2019). "Sarm1 deletion reduces axon damage, demyelination, and white matter atrophy after experimental traumatic brain injury". Experimental Neurology. 321: 113040. doi: 10.1016/j.expneurol.2019.113040 . PMID   31445042. S2CID   201124859.
  48. Maynard ME, Redell JB, Zhao J, Hood KN, Vita SM, Kobori N, et al. (May 2020). "Sarm1 loss reduces axonal damage and improves cognitive outcome after repetitive mild closed head injury". Experimental Neurology. 327: 113207. doi:10.1016/j.expneurol.2020.113207. PMC   7959192 . PMID   31962129.
  49. Bradshaw DV, Knutsen AK, Korotcov A, Sullivan GM, Radomski KL, Dardzinski BJ, et al. (May 2021). "Genetic inactivation of SARM1 axon degeneration pathway improves outcome trajectory after experimental traumatic brain injury based on pathological, radiological, and functional measures". Acta Neuropathologica Communications. 9 (1): 89. doi: 10.1186/s40478-021-01193-8 . PMC   8130449 . PMID   34001261.
  50. Alexandris AS, Lee Y, Lehar M, Alam Z, Samineni P, Tripathi SJ, et al. (October 2022). "Traumatic axonopathy in spinal tracts after impact acceleration head injury: Ultrastructural observations and evidence of SARM1-dependent axonal degeneration". Experimental Neurology. 359: 114252. doi:10.1016/j.expneurol.2022.114252. PMC   10321775 . PMID   36244414. S2CID   252894815.
  51. Alexandris AS, Lee Y, Lehar M, Alam Z, McKenney J, Perdomo D, et al. (2023-03-14). "Traumatic Axonal Injury in the Optic Nerve: The Selective Role of SARM1 in the Evolution of Distal Axonopathy". Journal of Neurotrauma. 40 (15–16): 1743–1761. doi: 10.1089/neu.2022.0416 . ISSN   1557-9042. PMC   10460965 . PMID   36680758. S2CID   256055839.
  52. Geisler S, Doan RA, Strickland A, Huang X, Milbrandt J, DiAntonio A (December 2016). "Prevention of vincristine-induced peripheral neuropathy by genetic deletion of SARM1 in mice". Brain. 139 (Pt 12): 3092–3108. doi:10.1093/brain/aww251. PMC   5840884 . PMID   27797810.
  53. Geisler S, Doan RA, Cheng GC, Cetinkaya-Fisgin A, Huang SX, Höke A, et al. (September 2019). "Vincristine and bortezomib use distinct upstream mechanisms to activate a common SARM1-dependent axon degeneration program". JCI Insight. 4 (17). doi:10.1172/jci.insight.129920. PMC   6777905 . PMID   31484833.
  54. Gould SA, White M, Wilbrey AL, Pór E, Coleman MP, Adalbert R (April 2021). "Protection against oxaliplatin-induced mechanical and thermal hypersensitivity in Sarm1-/- mice". Experimental Neurology. 338: 113607. doi:10.1016/j.expneurol.2021.113607. PMID   33460644. S2CID   231614379.
  55. Cetinkaya-Fisgin A, Luan X, Reed N, Jeong YE, Oh BC, Hoke A (December 2020). "Cisplatin induced neurotoxicity is mediated by Sarm1 and calpain activation". Scientific Reports. 10 (1): 21889. Bibcode:2020NatSR..1021889C. doi:10.1038/s41598-020-78896-w. PMC   7736304 . PMID   33318563.
  56. 1 2 Turkiew E, Falconer D, Reed N, Höke A (September 2017). "Deletion of Sarm1 gene is neuroprotective in two models of peripheral neuropathy". Journal of the Peripheral Nervous System. 22 (3): 162–171. doi:10.1111/jns.12219. PMC   5585053 . PMID   28485482.
  57. Cheng Y, Liu J, Luan Y, Liu Z, Lai H, Zhong W, et al. (November 2019). "Sarm1 Gene Deficiency Attenuates Diabetic Peripheral Neuropathy in Mice". Diabetes. 68 (11): 2120–2130. doi:10.2337/db18-1233. PMC   6804630 . PMID   31439642.
  58. Fernandes KA, Mitchell KL, Patel A, Marola OJ, Shrager P, Zack DJ, et al. (June 2018). "Role of SARM1 and DR6 in retinal ganglion cell axonal and somal degeneration following axonal injury". Experimental Eye Research. 171: 54–61. doi:10.1016/j.exer.2018.03.007. PMC   5964014 . PMID   29526794.
  59. Ozaki E, Gibbons L, Neto NG, Kenna P, Carty M, Humphries M, et al. (May 2020). "SARM1 deficiency promotes rod and cone photoreceptor cell survival in a model of retinal degeneration". Life Science Alliance. 3 (5): e201900618. doi:10.26508/lsa.201900618. PMC   7184027 . PMID   32312889.
  60. Ko KW, Milbrandt J, DiAntonio A (August 2020). "SARM1 acts downstream of neuroinflammatory and necroptotic signaling to induce axon degeneration". The Journal of Cell Biology. 219 (8): e201912047. doi:10.1083/jcb.201912047. PMC   7401797 . PMID   32609299.
  61. Sasaki Y, Kakita H, Kubota S, Sene A, Lee TJ, Ban N, et al. (October 2020). "SARM1 depletion rescues NMNAT1-dependent photoreceptor cell death and retinal degeneration". eLife. 9: e62027. doi: 10.7554/eLife.62027 . PMC   7591247 . PMID   33107823.
  62. Finnegan LK, Chadderton N, Kenna PF, Palfi A, Carty M, Bowie AG, et al. (January 2022). "SARM1 Ablation Is Protective and Preserves Spatial Vision in an In Vivo Mouse Model of Retinal Ganglion Cell Degeneration". International Journal of Molecular Sciences. 23 (3): 1606. doi: 10.3390/ijms23031606 . PMC   8835928 . PMID   35163535.
  63. Gibbons L, Ozaki E, Greene C, Trappe A, Carty M, Coppinger JA, et al. (2022). "SARM1 Promotes Photoreceptor Degeneration in an Oxidative Stress Model of Retinal Degeneration". Frontiers in Neuroscience. 16: 852114. doi: 10.3389/fnins.2022.852114 . PMC   9012108 . PMID   35431772.
  64. Liu P, Chen W, Jiang H, Huang H, Liu L, Fang F, et al. (2023-06-13). "Differential effects of SARM1 inhibition in traumatic glaucoma and EAE optic neuropathies". Molecular Therapy. Nucleic Acids. 32: 13–27. doi:10.1016/j.omtn.2023.02.029. ISSN   2162-2531. PMC   10025007 . PMID   36950280.
  65. Tian W, Czopka T, López-Schier H (January 2020). "Systemic loss of Sarm1 protects Schwann cells from chemotoxicity by delaying axon degeneration". Communications Biology. 3 (1): 49. doi:10.1038/s42003-020-0776-9. PMC   6992705 . PMID   32001778.
  66. Yamada Y, Strickland A, Sasaki Y, Bloom AJ, DiAntonio A, Milbrandt J (October 2022). "A SARM1/mitochondrial feedback loop drives neuropathogenesis in a Charcot-Marie-Tooth disease type 2A rat model". The Journal of Clinical Investigation. 132 (23): e161566. doi:10.1172/JCI161566. PMC   9711878 . PMID   36287202. S2CID   253118335.
  67. Montoro-Gámez C, Nolte H, Molinié T, Evangelista G, Tröder S, Barth E, et al. (2023-04-22). "SARM1 deletion delays cerebellar but not spinal cord degeneration in an enhanced mouse model of SPG7 deficiency". Brain: A Journal of Neurology. 146 (10): 4117–4131. doi:10.1093/brain/awad136. ISSN   1460-2156. PMID   37086482.
  68. Ademi M, Yang X, Coleman MP, Gilley J (August 2022). "Natural variants of human SARM1 cause both intrinsic and dominant loss-of-function influencing axon survival". Scientific Reports. 12 (1): 13846. Bibcode:2022NatSR..1213846A. doi:10.1038/s41598-022-18052-8. PMC   9381744 . PMID   35974060.
  69. Lukacs M, Gilley J, Zhu Y, Orsomando G, Angeletti C, Liu J, et al. (October 2019). "Severe biallelic loss-of-function mutations in nicotinamide mononucleotide adenylyltransferase 2 (NMNAT2) in two fetuses with fetal akinesia deformation sequence". Experimental Neurology. 320: 112961. doi:10.1016/j.expneurol.2019.112961. PMC   6708453 . PMID   31136762.
  70. Huppke P, Wegener E, Gilley J, Angeletti C, Kurth I, Drenth JP, et al. (October 2019). "Homozygous NMNAT2 mutation in sisters with polyneuropathy and erythromelalgia". Experimental Neurology. 320: 112958. doi:10.1016/j.expneurol.2019.112958. PMID   31132363. S2CID   162183820.
  71. Gilley J, Jackson O, Pipis M, Estiar MA, Al-Chalabi A, Danzi MC, et al. (November 2021). "Enrichment of SARM1 alleles encoding variants with constitutively hyperactive NADase in patients with ALS and other motor nerve disorders". eLife. 10: e70905. doi: 10.7554/eLife.70905 . PMC   8735862 . PMID   34796871.
  72. Bloom AJ, Mao X, Strickland A, Sasaki Y, Milbrandt J, DiAntonio A (January 2022). "Constitutively active SARM1 variants that induce neuropathy are enriched in ALS patients". Molecular Neurodegeneration. 17 (1): 1. doi: 10.1186/s13024-021-00511-x . PMC   8739729 . PMID   34991663.
  73. Chen YH, Sasaki Y, DiAntonio A, Milbrandt J (May 2021). "SARM1 is required in human derived sensory neurons for injury-induced and neurotoxic axon degeneration". Experimental Neurology. 339: 113636. doi:10.1016/j.expneurol.2021.113636. PMC   8171232 . PMID   33548217.
  74. Sporny M, Guez-Haddad J, Lebendiker M, Ulisse V, Volf A, Mim C, et al. (September 2019). "Structural Evidence for an Octameric Ring Arrangement of SARM1". Journal of Molecular Biology. 431 (19): 3591–3605. doi:10.1016/j.jmb.2019.06.030. PMID   31278906. S2CID   195819811.
  75. Shen C, Vohra M, Zhang P, Mao X, Figley MD, Zhu J, et al. (January 2021). "Multiple domain interfaces mediate SARM1 autoinhibition". Proceedings of the National Academy of Sciences of the United States of America. 118 (4): e2023151118. Bibcode:2021PNAS..11823151S. doi: 10.1073/pnas.2023151118 . PMC   7848697 . PMID   33468661.
  76. Khazma T, Golan-Vaishenker Y, Guez-Haddad J, Grossman A, Sain R, Weitman M, et al. (December 2022). "A duplex structure of SARM1 octamers stabilized by a new inhibitor". Cellular and Molecular Life Sciences. 80 (1): 16. doi:10.1007/s00018-022-04641-3. PMID   36564647. S2CID   255085095.
  77. Gilley J, Coleman MP (January 2010). "Endogenous Nmnat2 is an essential survival factor for maintenance of healthy axons". PLOS Biology. 8 (1): e1000300. doi: 10.1371/journal.pbio.1000300 . PMC   2811159 . PMID   20126265.
  78. Gilley J, Orsomando G, Nascimento-Ferreira I, Coleman MP (March 2015). "Absence of SARM1 rescues development and survival of NMNAT2-deficient axons". Cell Reports. 10 (12): 1974–1981. doi:10.1016/j.celrep.2015.02.060. PMC   4386025 . PMID   25818290.
  79. Araki T, Sasaki Y, Milbrandt J (August 2004). "Increased nuclear NAD biosynthesis and SIRT1 activation prevent axonal degeneration". Science. 305 (5686): 1010–1013. Bibcode:2004Sci...305.1010A. doi:10.1126/science.1098014. PMID   15310905. S2CID   32370137.
  80. Sasaki Y, Vohra BP, Lund FE, Milbrandt J (April 2009). "Nicotinamide mononucleotide adenylyl transferase-mediated axonal protection requires enzymatic activity but not increased levels of neuronal nicotinamide adenine dinucleotide". The Journal of Neuroscience. 29 (17): 5525–5535. doi:10.1523/JNEUROSCI.5469-08.2009. PMC   3162248 . PMID   19403820.
  81. Sasaki Y, Vohra BP, Baloh RH, Milbrandt J (May 2009). "Transgenic mice expressing the Nmnat1 protein manifest robust delay in axonal degeneration in vivo". The Journal of Neuroscience. 29 (20): 6526–6534. doi:10.1523/JNEUROSCI.1429-09.2009. PMC   2697066 . PMID   19458223.
  82. Sasaki Y, Milbrandt J (December 2010). "Axonal degeneration is blocked by nicotinamide mononucleotide adenylyltransferase (Nmnat) protein transduction into transected axons". The Journal of Biological Chemistry. 285 (53): 41211–41215. doi: 10.1074/jbc.C110.193904 . PMC   3009846 . PMID   21071441.
  83. Sasaki Y, Nakagawa T, Mao X, DiAntonio A, Milbrandt J (October 2016). "NMNAT1 inhibits axon degeneration via blockade of SARM1-mediated NAD+ depletion". eLife. 5. doi: 10.7554/eLife.19749 . PMC   5063586 . PMID   27735788.
  84. Coleman MP, Conforti L, Buckmaster EA, Tarlton A, Ewing RM, Brown MC, et al. (August 1998). "An 85-kb tandem triplication in the slow Wallerian degeneration (Wlds) mouse". Proceedings of the National Academy of Sciences of the United States of America. 95 (17): 9985–9990. Bibcode:1998PNAS...95.9985C. doi: 10.1073/pnas.95.17.9985 . PMC   21448 . PMID   9707587.
  85. Beirowski B, Babetto E, Gilley J, Mazzola F, Conforti L, Janeckova L, et al. (January 2009). "Non-nuclear Wld(S) determines its neuroprotective efficacy for axons and synapses in vivo". The Journal of Neuroscience. 29 (3): 653–668. doi:10.1523/JNEUROSCI.3814-08.2009. PMC   6665162 . PMID   19158292.
  86. Conforti L, Wilbrey A, Morreale G, Janeckova L, Beirowski B, Adalbert R, et al. (February 2009). "Wld S protein requires Nmnat activity and a short N-terminal sequence to protect axons in mice". The Journal of Cell Biology. 184 (4): 491–500. doi:10.1083/jcb.200807175. PMC   2654131 . PMID   19237596.
  87. Babetto E, Beirowski B, Janeckova L, Brown R, Gilley J, Thomson D, et al. (October 2010). "Targeting NMNAT1 to axons and synapses transforms its neuroprotective potency in vivo". The Journal of Neuroscience. 30 (40): 13291–13304. doi:10.1523/JNEUROSCI.1189-10.2010. PMC   6634738 . PMID   20926655.
  88. Di Stefano M, Nascimento-Ferreira I, Orsomando G, Mori V, Gilley J, Brown R, et al. (May 2015). "A rise in NAD precursor nicotinamide mononucleotide (NMN) after injury promotes axon degeneration". Cell Death and Differentiation. 22 (5): 731–742. doi:10.1038/cdd.2014.164. PMC   4392071 . PMID   25323584.
  89. Loreto A, Di Stefano M, Gering M, Conforti L (December 2015). "Wallerian Degeneration Is Executed by an NMN-SARM1-Dependent Late Ca(2+) Influx but Only Modestly Influenced by Mitochondria". Cell Reports. 13 (11): 2539–2552. doi: 10.1016/j.celrep.2015.11.032 . PMID   26686637. S2CID   25667592.
  90. 1 2 Gerdts J, Brace EJ, Sasaki Y, DiAntonio A, Milbrandt J (April 2015). "SARM1 activation triggers axon degeneration locally via NAD⁺ destruction". Science. 348 (6233): 453–457. Bibcode:2015Sci...348..453G. doi:10.1126/science.1258366. PMC   4513950 . PMID   25908823.
  91. Ko KW, Devault L, Sasaki Y, Milbrandt J, DiAntonio A (November 2021). "Live imaging reveals the cellular events downstream of SARM1 activation". eLife. 10: e71148. doi: 10.7554/eLife.71148 . PMC   8612704 . PMID   34779400.
  92. Dingwall CB, Strickland A, Yum SW, Yim AK, Zhu J, Wang PL, et al. (October 2022). "Macrophage depletion blocks congenital SARM1-dependent neuropathy". The Journal of Clinical Investigation. 132 (23): e159800. doi:10.1172/JCI159800. PMC   9711884 . PMID   36287209.
  93. Niou Z, Yang S, Sri A, Rodriquez H, Gilley J, Coleman MP, et al. (2022-02-08). "NMNAT2 in cortical glutamatergic neurons exerts both cell and non-cell autonomous influences to shape cortical development and to maintain neuronal health". bioRxiv: 2022.02.05.479195. doi:10.1101/2022.02.05.479195. S2CID   246746270.