1
|
He S, Zhu Y, Wang X, Zhang G, Hou K, Xia X, Jiang Z, Gong X, Zhao P. Targeting SARM1 as a novel neuroprotective therapy in neurotropic viral infections. J Neuroinflammation 2025; 22:113. [PMID: 40254576 PMCID: PMC12010687 DOI: 10.1186/s12974-025-03423-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 03/18/2025] [Indexed: 04/22/2025] Open
Abstract
Viral encephalitis, resulting from neurotropic viral infections, leads to severe neurological impairment, inflammation, and exhibits high mortality rates with poor prognosis. Currently, there is a lack of effective targeted treatments for this disease, which poses a significant public health concern. SARM1 has been identified as the pivotal mediator of axonal degeneration and inflammation across various neuropathies, activated by an elevation in the NMN/NAD+ ratio. However, comprehensive in vivo investigations into the role of SARM1-mediated pathogenesis in viral encephalitis are still lacking. In this study, we established mouse models of viral encephalitis using Japanese encephalitis virus (JEV), herpes simplex virus-1 (HSV-1), and rabies virus (RABV) as representative pathogens. Our findings demonstrate that neurotropic virus infections elicit robust axonal degeneration, mitochondrial dysfunction, and profound neuropathological damage in cortical neurons via the activation of SARM1. In mouse models of viral encephalitis, deletion or inhibition of SARM1 effectively preserved axonal morphology and maintained mitochondrial homeostasis, while also attenuating the infiltration of CD45+ leukocytes in the cortex. Consequently, these interventions ameliorated neuropathological damage and enhanced survival outcomes in mice. Our findings suggest that SARM1-mediated axonal degeneration and brain inflammation exacerbate the pathological progression of viral encephalitis. Therapies targeting SARM1 emerge as viable and promising strategies for protecting neuronal function in the context of neurotropic viral infections.
Collapse
Affiliation(s)
- Sheng He
- Department of Laboratory Medicine, Yuebei People's Hospital, Affiliated to Shantou University Medical College, Shaoguan, 512025, China
- Laboratory for Diagnosis of Clinical Microbiology and Infection, Yuebei People's Hospital, Affiliated to Shantou University Medical College, Shaoguan, 512025, China
- Research Center for Interdisciplinary & High-quality Innovative Development in Laboratory Medicine, Shaoguan, 512025, China
- Shaoguan Municipal Quality Control Center for Laboratory Medicine, Yuebei People's Hospital, Affiliated to Shantou University Medical College, Shaoguan, 512025, China
- Shaoguan Municipal Quality Control Center for Surveillance of Bacterial Resistance, Shaoguan, 512025, China
- Shaoguan Engineering Research Center for Research and Development of Molecular and Cellular Technology in Rapid Diagnosis of Infectious Diseases and Cancer, Shaoguan, 512025, China
- Yuebei People's Hospital, Affiliated to Shantou University Medical College, No 133, Huimin Road South, Wujiang District, Shaoguan, 512025, China
| | - Yanyan Zhu
- Department of Laboratory Medicine, Yuebei People's Hospital, Affiliated to Shantou University Medical College, Shaoguan, 512025, China
- Laboratory for Diagnosis of Clinical Microbiology and Infection, Yuebei People's Hospital, Affiliated to Shantou University Medical College, Shaoguan, 512025, China
- Research Center for Interdisciplinary & High-quality Innovative Development in Laboratory Medicine, Shaoguan, 512025, China
- Shaoguan Municipal Quality Control Center for Laboratory Medicine, Yuebei People's Hospital, Affiliated to Shantou University Medical College, Shaoguan, 512025, China
- Shaoguan Municipal Quality Control Center for Surveillance of Bacterial Resistance, Shaoguan, 512025, China
- Shaoguan Engineering Research Center for Research and Development of Molecular and Cellular Technology in Rapid Diagnosis of Infectious Diseases and Cancer, Shaoguan, 512025, China
| | - Xinyue Wang
- Department of Laboratory Medicine, Yuebei People's Hospital, Affiliated to Shantou University Medical College, Shaoguan, 512025, China
- Laboratory for Diagnosis of Clinical Microbiology and Infection, Yuebei People's Hospital, Affiliated to Shantou University Medical College, Shaoguan, 512025, China
- Research Center for Interdisciplinary & High-quality Innovative Development in Laboratory Medicine, Shaoguan, 512025, China
- Shaoguan Municipal Quality Control Center for Laboratory Medicine, Yuebei People's Hospital, Affiliated to Shantou University Medical College, Shaoguan, 512025, China
- Shaoguan Municipal Quality Control Center for Surveillance of Bacterial Resistance, Shaoguan, 512025, China
- Shaoguan Engineering Research Center for Research and Development of Molecular and Cellular Technology in Rapid Diagnosis of Infectious Diseases and Cancer, Shaoguan, 512025, China
| | - Gaofeng Zhang
- Department of Laboratory Medicine, Yuebei People's Hospital, Affiliated to Shantou University Medical College, Shaoguan, 512025, China
- Laboratory for Diagnosis of Clinical Microbiology and Infection, Yuebei People's Hospital, Affiliated to Shantou University Medical College, Shaoguan, 512025, China
- Research Center for Interdisciplinary & High-quality Innovative Development in Laboratory Medicine, Shaoguan, 512025, China
- Shaoguan Municipal Quality Control Center for Laboratory Medicine, Yuebei People's Hospital, Affiliated to Shantou University Medical College, Shaoguan, 512025, China
- Shaoguan Municipal Quality Control Center for Surveillance of Bacterial Resistance, Shaoguan, 512025, China
- Shaoguan Engineering Research Center for Research and Development of Molecular and Cellular Technology in Rapid Diagnosis of Infectious Diseases and Cancer, Shaoguan, 512025, China
| | - Kaijian Hou
- School of Public Health, Shantou University, Shantou, 515041, China
| | - Xianzhu Xia
- Department of Laboratory Medicine, Yuebei People's Hospital, Affiliated to Shantou University Medical College, Shaoguan, 512025, China
- Laboratory for Diagnosis of Clinical Microbiology and Infection, Yuebei People's Hospital, Affiliated to Shantou University Medical College, Shaoguan, 512025, China
- Research Center for Interdisciplinary & High-quality Innovative Development in Laboratory Medicine, Shaoguan, 512025, China
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Zhenyou Jiang
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China
| | - Xiaoqian Gong
- Yuebei People's Hospital, Affiliated to Shantou University Medical College, No 133, Huimin Road South, Wujiang District, Shaoguan, 512025, China.
| | - Pingsen Zhao
- Department of Laboratory Medicine, Yuebei People's Hospital, Affiliated to Shantou University Medical College, Shaoguan, 512025, China.
- Laboratory for Diagnosis of Clinical Microbiology and Infection, Yuebei People's Hospital, Affiliated to Shantou University Medical College, Shaoguan, 512025, China.
- Research Center for Interdisciplinary & High-quality Innovative Development in Laboratory Medicine, Shaoguan, 512025, China.
- Shaoguan Municipal Quality Control Center for Laboratory Medicine, Yuebei People's Hospital, Affiliated to Shantou University Medical College, Shaoguan, 512025, China.
- Shaoguan Municipal Quality Control Center for Surveillance of Bacterial Resistance, Shaoguan, 512025, China.
- Shaoguan Engineering Research Center for Research and Development of Molecular and Cellular Technology in Rapid Diagnosis of Infectious Diseases and Cancer, Shaoguan, 512025, China.
- Yuebei People's Hospital, Affiliated to Shantou University Medical College, No 133, Huimin Road South, Wujiang District, Shaoguan, 512025, China.
| |
Collapse
|
2
|
Danos JA, Addemir M, McGettigan L, Summers DW. Nerve growth factor signaling tunes axon maintenance protein abundance and kinetics of Wallerian degeneration. Mol Biol Cell 2025; 36:ar46. [PMID: 39969989 PMCID: PMC12005098 DOI: 10.1091/mbc.e25-01-0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 02/20/2025] Open
Abstract
Neurotrophic factors are critical for establishing functional connectivity in the nervous system and sustaining neuronal survival through adulthood. As the first neurotrophic factor purified, nerve growth factor (NGF) is extensively studied for its prolific role in axon outgrowth, pruning, and survival. Applying NGF to diseased neuronal tissue is an exciting therapeutic option and understanding how NGF regulates local axon susceptibility to pathological degeneration is critical for exploiting its full potential. Our study identifies surprising connections between NGF signaling and proteostasis of axon maintenance factors. NGF deprivation increases Nmnat2 and Stmn2 protein levels in axon segments with a corresponding delay in Wallerian degeneration. Conversely, acute NGF stimulation reduces local abundance of these axon maintenance factors and accelerates Wallerian degeneration. Pharmacological studies implicate phospholipase C as the key effector in tropomyosin-related kinase A (TrkA) activation, which drives degradation of palmitoylated Stmn2. While seemingly opposed to neuroprotective activities well-documented for NGF, downregulating Nmnat2 and Stmn2 favors axonal outgrowth over transient hypersusceptibility to Sarm1-dependent degeneration. This new facet of NGF biology has important implications for axonal remodeling during development and sustained integrity through adulthood.
Collapse
Affiliation(s)
- Joseph A. Danos
- Department of Biology, University of Iowa, Iowa City, IA 52242
| | - Merve Addemir
- Department of Biology, University of Iowa, Iowa City, IA 52242
| | - Lily McGettigan
- Department of Biology, University of Iowa, Iowa City, IA 52242
| | - Daniel W. Summers
- Department of Biology, University of Iowa, Iowa City, IA 52242
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242
| |
Collapse
|
3
|
Herbosa CG, Perez R, Jaeger A, Dy CJ, Brogan DM. Inhibition of SARM1 Reduces Neuropathic Pain in a Spared Nerve Injury Rodent Model. Muscle Nerve 2025; 71:670-679. [PMID: 39936361 DOI: 10.1002/mus.28367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 02/13/2025]
Abstract
INTRODUCTION/AIMS The function of the sterile alpha and toll/interleukin receptor motif-containing protein 1 (SARM1) in neuropathic pain development has not yet been established. This protein has a central role in regulating axon degeneration and its depletion delays this process. This study aims to demonstrate the effects of SARM1 deletion on the development of neuropathic pain. METHODS Thirty-two wild-type (WT) or SARM1 knockout (KO) rats underwent spared nerve injury (SNI) or sham surgery. Mechanical allodynia was assessed by electronic Von Frey and cold hyperalgesia by the acetone test. Nociception was evaluated at the baseline, Day-1, Day-2, Week-1, Week-2, Week-3, and Week-4 time points. Nerve sections were examined by immunohistochemistry (IHC). RESULTS WT Injury rats were more sensitive to pain than WT Sham at all postoperative time points, validating the pain model. Injured SARM1 KO rats only demonstrated a difference in mechanical or cold nociception from KO Sham at Week 3. Injured KO rats demonstrated a clear trend of decreased sensitivity compared to WT Injury nociception, reaching significance at Week 4 (p = 0.044). Injured KO rats showed attenuated sensitivity to cold allodynia relative to WT at Week 2 (p = 0.019). IHC revealed decreased macrophages in spared sural nerves of injured KO animals at 2 and 4 weeks, and the proximal portion of tibial/peroneal nerves at Week 2. DISCUSSION This study demonstrates that SARM1 KO rats are less sensitive to mechanical and cold nociception than WT rats in an SNI model with decreased inflammatory response. Given these results, inhibition of SARM1 should be further investigated in the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Christopher G Herbosa
- Department of Orthopedic Surgery, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Ronald Perez
- Department of Orthopedic Surgery, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Alexandra Jaeger
- Department of Orthopedic Surgery, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Christopher J Dy
- Department of Orthopedic Surgery, Washington University in St. Louis, St. Louis, Missouri, USA
| | - David M Brogan
- Department of Orthopedic Surgery, Washington University in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
4
|
Karnik A, Joshi A. SARM1: The Checkpoint of Axonal Degeneration in the Nervous System Disorders. Mol Neurobiol 2025:10.1007/s12035-025-04835-3. [PMID: 40097763 DOI: 10.1007/s12035-025-04835-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 03/09/2025] [Indexed: 03/19/2025]
Abstract
Axons are metabolically active neuronal segments with well-controlled axonal degeneration and regeneration. External stress or injury displaces this equilibrium toward degeneration leading to axonal dysfunction observed in the pathology of several diseases. The demand and supply matrix of energy at the synapses are maintained by the axonal transport. Nicotinamide adenine dinucleotide (NAD+) is a major energy-driving coenzyme of cells that controls mitochondrial, cytoplasmic, and other organellar energy cycles generating high amounts of adenosine triphosphate (ATP). NAD+ participates in various cellular cycles and is consumed by several enzymes. One of the key enzymes targeting NAD+ is Sterile alpha and TIR motif-containing protein 1 (SARM1) which gets activated in response to external noxious stimuli. SARM1 is an octamer consisting of multiple domains of which the TIR domain governs NAD+ hydrolysis which eventually leads to axonal deficits. Besides its localization in neurons, SARM1 is also present in astrocytes, microglia, and macrophages in which it regulates inflammatory responses associated with disease pathology. SARM1 localization in the outer mitochondrial membrane is responsible for its association with mitochondrial dynamics. SARM1-mediated mitochondrial dysfunction further drives the axonal degeneration associated with peripheral and central nervous system disorders. Several genetic and pharmacological studies highlight the role of SARM1 in axonal degeneration. SARM1 is thus becoming a popular target for preventing axonal degeneration. Several small molecules consisting of isoquinoline, isothiazole, pyridine, and tryptoline acrylamide moieties have been tested for their activity against SARM1 with a promising foundation for drug discovery in targeting SARM1. In our review, we highlight the role of SARM1 in axonal degeneration associated with several disease pathologies focusing on genetic and pharmacological evaluation.
Collapse
Affiliation(s)
- Aaditi Karnik
- Department of Pharmacy, Birla Institute of Technology and Sciences-Pilani, Telangana State, Hyderabad Campus, Hyderabad City, India
| | - Abhijeet Joshi
- Department of Pharmacy, Birla Institute of Technology and Sciences-Pilani, Telangana State, Hyderabad Campus, Hyderabad City, India.
| |
Collapse
|
5
|
Chin MY, Joy DA, Samaddar M, Rana A, Chow J, Miyamoto T, Calvert M. Novel high-content and open-source image analysis tools for profiling mitochondrial morphology in neurological cell models. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2025; 31:100208. [PMID: 39778657 DOI: 10.1016/j.slasd.2025.100208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/16/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025]
Abstract
Mitochondria undergo dynamic morphological changes depending on cellular cues, stress, genetic factors, or disease. The structural complexity and disease-relevance of mitochondria have stimulated efforts to generate image analysis tools for describing mitochondrial morphology for therapeutic development. Using high-content analysis, we measured multiple morphological parameters and employed unbiased feature clustering to identify the most robust pair of texture metrics that described mitochondrial state. Here, we introduce a novel image analysis pipeline to enable rapid and accurate profiling of mitochondrial morphology in various cell types and pharmacological perturbations. We applied a high-content adapted implementation of our tool, MitoProfilerHC, to quantify mitochondrial morphology changes in i) a mammalian cell dose response study and ii) compartment-specific drug effects in primary neurons. Next, we expanded the usability of our pipeline by using napari, a Python-powered image analysis tool, to build an open-source version of MitoProfiler and validated its performance and applicability. In conclusion, we introduce MitoProfiler as both a high-content-based and an open-source method to accurately quantify mitochondrial morphology in cells, which we anticipate to greatly facilitate mechanistic discoveries in mitochondrial biology and disease.
Collapse
Affiliation(s)
- Marcus Y Chin
- Denali Therapeutics Inc., South San Francisco, CA 94080 USA.
| | - David A Joy
- Denali Therapeutics Inc., South San Francisco, CA 94080 USA
| | | | - Anil Rana
- Denali Therapeutics Inc., South San Francisco, CA 94080 USA
| | - Johann Chow
- Denali Therapeutics Inc., South San Francisco, CA 94080 USA
| | | | | |
Collapse
|
6
|
Nasuhidehnavi A, Zarzycka W, Górecki I, Chiao YA, Lee CF. Emerging interactions between mitochondria and NAD + metabolism in cardiometabolic diseases. Trends Endocrinol Metab 2025; 36:176-190. [PMID: 39198117 PMCID: PMC11794032 DOI: 10.1016/j.tem.2024.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 09/01/2024]
Abstract
Nicotinamide adenine dinucleotide (NAD+) is an essential coenzyme for redox reactions and regulates cellular catabolic pathways. An intertwined relationship exists between NAD+ and mitochondria, with consequences for mitochondrial function. Dysregulation in NAD+ homeostasis can lead to impaired energetics and increased oxidative stress, contributing to the pathogenesis of cardiometabolic diseases. In this review, we explore how disruptions in NAD+ homeostasis impact mitochondrial function in various cardiometabolic diseases. We discuss emerging studies demonstrating that enhancing NAD+ synthesis or inhibiting its consumption can ameliorate complications of this family of pathological conditions. Additionally, we highlight the potential role and therapeutic promise of mitochondrial NAD+ transporters in regulating cellular and mitochondrial NAD+ homeostasis.
Collapse
Affiliation(s)
- Azadeh Nasuhidehnavi
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY 13790, USA
| | - Weronika Zarzycka
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Ignacy Górecki
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Ying Ann Chiao
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Chi Fung Lee
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| |
Collapse
|
7
|
Sachar R, Lee TY, DiAntonio A, Dy CJ, Wever J, Milbrandt J, Brogan DM. SARM1 Inhibition Maintains Axonal Integrity After Rat Sciatic Nerve Transection and Repair. J Hand Surg Am 2025:S0363-5023(24)00617-8. [PMID: 39895440 DOI: 10.1016/j.jhsa.2024.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 11/06/2024] [Accepted: 12/11/2024] [Indexed: 02/04/2025]
Abstract
PURPOSE Sterile alpha and TlR motif containing-1 (SARM1) protein has been demonstrated to play a critical role in the initiation of Wallerian degeneration after nerve injury. The goal of this study was to assess whether blockade of SARM1 activity inhibits Wallerian degeneration following nerve transection, potentially promoting more rapid recovery of axonal function. METHODS An adeno-associated virus plasmid encoded with a dominant-negative SARM1 protein fused with green fluorescent protein to impair SARM1 function, was injected into 24 juvenile rats to create a SARM1 dominant-negative (SARM1-DN) phenotype. Twenty-four control rats were injected with a control plasmid expressing only green fluorescent protein. Three weeks after transfection, the rats underwent unilateral sciatic nerve transection and repair. Walking track analysis and nonsurvival surgeries were performed at 2 days, 2 weeks, or 6 weeks to assess muscle strength and compound nerve action potential. Histomorphologic and electrodiagnostic studies were evaluated with mixed-effect analysis. RESULTS Histomorphologic analysis showed maintenance of axons in the SARM1-DN animals at 2 weeks, with significantly improved compound nerve action potential amplitude. Muscle testing demonstrated greater gastrocnemius strength in SARM1 DN muscles at 2 days and 2 weeks compared to controls, although this was not maintained at 6 weeks. CONCLUSION Inhibition of SARM1 resulted in early increases in number and myelination of axons and action potential after sciatic nerve transection and repair in SARM1-DN rats. CLINICAL RELEVANCE SARM1 inhibition may offer the potential to delay Wallerian degeneration following nerve transection and enable earlier functional recovery of motor strength. .
Collapse
Affiliation(s)
- Ryan Sachar
- Department of Orthopedic Surgery, Washington University in St. Louis, St. Louis, MO
| | - Tony Y Lee
- Department of Orthopedic Surgery, Washington University in St. Louis, St. Louis, MO
| | - Aaron DiAntonio
- Department of Genetics, Washington University in St. Louis, St. Louis, MO
| | - Christopher J Dy
- Department of Orthopedic Surgery, Washington University in St. Louis, St. Louis, MO
| | - Jason Wever
- Department of Orthopedic Surgery, Washington University in St. Louis, St. Louis, MO
| | - Jeff Milbrandt
- Department of Genetics, Washington University in St. Louis, St. Louis, MO
| | - David M Brogan
- Department of Orthopedic Surgery, Washington University in St. Louis, St. Louis, MO.
| |
Collapse
|
8
|
Brüll M, Multrus S, Schäfer M, Celardo I, Karreman C, Leist M. Programmed neurite degeneration in human central nervous system neurons driven by changes in NAD + metabolism. Cell Death Dis 2025; 16:24. [PMID: 39824831 PMCID: PMC11742042 DOI: 10.1038/s41419-024-07326-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/04/2024] [Accepted: 12/27/2024] [Indexed: 01/20/2025]
Abstract
Neurite degeneration (ND) precedes cell death in many neurodegenerative diseases. However, it remains unclear how this compartmentalized cell death process is orchestrated in the central nervous system (CNS). The establishment of a CNS axotomy model (using modified 3D LUHMES cultures) allowed us to study metabolic control of ND in human midbrain-derived neurons without the use of toxicants or other direct disturbance of cellular metabolism. Axotomy lead to a loss of the NAD+ synthesis enzyme NMNAT2 within 2 h and a depletion of NAD+ within 4-6 h. This process appeared specific, as isolated neurites maintained ATP levels and a coupled mitochondrial respiration for at least 6 h. In the peripheral nervous system (PNS) many studies observed that NAD+ metabolism, in particular by the NADase SARM1, plays a major role in the ND occurring after axotomy. Since neither ferroptosis nor necroptosis, nor caspase-dependent apoptosis seemed to be involved in neurite loss, we investigated SARM1 as potential executioner (or controller). Knock-down or expression of a dominant-negative isoform of SARM1 indeed drastically delayed ND. Various modifications of NAD+ metabolism known to modulate SARM1 activity showed the corresponding effects on ND. Moreover, supplementation with NAD+ attenuated ND. As a third approach to investigate the role of altered NAD+ metabolism, we made use of the WLD(s) protein, which has been found in a mutant mouse to inhibit Wallerian degeneration of axons. This protein, which has a stable NMNAT activity, and thus can buffer the loss of NMNAT2, protected the neurites by stabilizing neurite NAD+ levels. Thus CNS-type ND was tightly linked to neurite metabolism in multiple experimental setups. Based on this knowledge, several new strategies for treating neurodegenerative diseases can be envisaged.
Collapse
Affiliation(s)
- Markus Brüll
- In vitro Toxicology and Biomedicine, Dept. inaugurated by the Doerenkamp-Zbinden foundation, University of Konstanz, 78457, Konstanz, Germany
| | - Selina Multrus
- In vitro Toxicology and Biomedicine, Dept. inaugurated by the Doerenkamp-Zbinden foundation, University of Konstanz, 78457, Konstanz, Germany
| | - Michael Schäfer
- In vitro Toxicology and Biomedicine, Dept. inaugurated by the Doerenkamp-Zbinden foundation, University of Konstanz, 78457, Konstanz, Germany
| | - Ivana Celardo
- In vitro Toxicology and Biomedicine, Dept. inaugurated by the Doerenkamp-Zbinden foundation, University of Konstanz, 78457, Konstanz, Germany
| | - Christiaan Karreman
- In vitro Toxicology and Biomedicine, Dept. inaugurated by the Doerenkamp-Zbinden foundation, University of Konstanz, 78457, Konstanz, Germany
| | - Marcel Leist
- In vitro Toxicology and Biomedicine, Dept. inaugurated by the Doerenkamp-Zbinden foundation, University of Konstanz, 78457, Konstanz, Germany.
- CAAT-Europe, University of Konstanz, 78457, Konstanz, Germany.
| |
Collapse
|
9
|
Gibbons L, Doyle S. A Role for SARM1 in Photoreceptor Cell Death. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1468:183-187. [PMID: 39930193 DOI: 10.1007/978-3-031-76550-6_30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Photoreceptor cell death is a common feature of many retinal degenerative diseases, leading to incurable vision loss. While there is evidence to support the involvement of cell death pathways such as apoptosis and necroptosis in the degeneration of photoreceptors, the inhibition of these pathways has not been sufficient to rescue photoreceptors and preserve vision in a number of models of disease. Therefore, there is a need to identify other pathways involved in photoreceptor cell death. SARM1 is a TLR adaptor protein with a novel role in the induction of axonal degeneration and neuronal cell death. Our lab and others have demonstrated a role for SARM1 in the induction of photoreceptor cell death in models of retinal degenerative disease. Here, we summarize the current knowledge on SARM1 function and its role in photoreceptor cell death.
Collapse
Affiliation(s)
- Luke Gibbons
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland.
- Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Dublin, Ireland.
| | - Sarah Doyle
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
- Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
10
|
Danos JA, Addemir M, McGettigan L, Summers DW. Nerve Growth Factor Signaling Tunes Axon Maintenance Protein Abundance and Kinetics of Wallerian Degeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.31.630780. [PMID: 39803444 PMCID: PMC11722262 DOI: 10.1101/2024.12.31.630780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Neurotrophic factors are critical for establishing functional connectivity in the nervous system and sustaining neuronal survival through adulthood. As the first neurotrophic factor purified, nerve growth factor (NGF) is extensively studied for its prolific role in axon outgrowth, pruning, and survival. Applying NGF to diseased neuronal tissue is an exciting therapeutic option and understanding how NGF regulates local axon susceptibility to pathological degeneration is critical for exploiting its full potential. Our study identifies surprising connections between NGF signaling and proteostasis of axon maintenance factors. NGF deprivation increases Nmnat2 and Stmn2 protein levels in axon segments with a corresponding delay in Wallerian degeneration. Conversely, acute NGF stimulation reduces local abundance of these axon maintenance factors and accelerates Wallerian degeneration. Pharmacological studies implicate phospholipase C as the key effector in TrkA activation, which drives degradation of palmitoylated Stmn2. While seemingly opposed to neuroprotective activities well-documented for NGF, downregulating Nmnat2 and Stmn2 favors axonal outgrowth over transient hyper-susceptibility to Sarm1-dependent degeneration. This new facet of NGF biology has important implications for axonal remodeling during development and sustained integrity through adulthood.
Collapse
Affiliation(s)
- Joseph A Danos
- Department of Biology, University of Iowa, Iowa City, IA 52242 USA
| | - Merve Addemir
- Department of Biology, University of Iowa, Iowa City, IA 52242 USA
| | - Lily McGettigan
- Department of Biology, University of Iowa, Iowa City, IA 52242 USA
| | - Daniel W Summers
- Department of Biology, University of Iowa, Iowa City, IA 52242 USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242 USA
| |
Collapse
|
11
|
Chen J, Li H. Characterization of Novel SARM1 Inhibitors for the Treatment of Chemotherapy-Induced Peripheral Neuropathy. Biomedicines 2024; 12:2123. [PMID: 39335636 PMCID: PMC11428815 DOI: 10.3390/biomedicines12092123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/05/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Sterile α and Toll/IL-1 receptor motif-containing 1 (SARM1) is a central regulator of programmed axon death and a crucial nicotinamide adenine dinucleotide (NAD+) hydrolase (NADase) in mammalian tissues, hydrolyzing NAD+ and playing an important role in cellular NAD+ recycling. Abnormal SARM1 expression is linked to axon degeneration, which causes disability and disease progression in many neurodegenerative disorders of the peripheral and central nervous systems. METHODS In this study, we use PC6 assay of hydrolase activity, DRG axon regeneration and CIPN model to screen for potent SARM1 Inhibitors. RESULTS Two novel SARM1 inhibitors (compound 174 and 331P1) are charcterized for its high potency for SARM1 NADase. In a chemotherapy-induced peripheral neuropathy (CIPN) myopathy model, compound 331P1 treatment prevented the decline in neurofilament light chain (NfL) levels caused by axonal injury in a dose-dependent manner, associated with elevated intraepidermal nerve fiber (IENF) intensity in mouse foot paw tissue, suggesting its functionality in reversing axon degeneration. CONCLUSIONS The newly designed SARM1 inhibitor 331P1 is a promising candidate due to its excellent in vivo efficacy, favorable CYP inhibition properties, and attractive safety profiles. The 331P1 compound possesses the potential to be developed as a novel neuroprotective therapy that can prevent or halt the neurodegenerative process in CIPN.
Collapse
Affiliation(s)
- Jiayu Chen
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, and School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Hao Li
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, and School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| |
Collapse
|
12
|
Nheu D, Petratos S. How does Nogo-A signalling influence mitochondrial function during multiple sclerosis pathogenesis? Neurosci Biobehav Rev 2024; 163:105767. [PMID: 38885889 DOI: 10.1016/j.neubiorev.2024.105767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 05/30/2024] [Accepted: 06/08/2024] [Indexed: 06/20/2024]
Abstract
Multiple sclerosis (MS) is a severe neurological disorder that involves inflammation in the brain, spinal cord and optic nerve with key disabling neuropathological outcomes being axonal damage and demyelination. When degeneration of the axo-glial union occurs, a consequence of inflammatory damage to central nervous system (CNS) myelin, dystrophy and death can lead to large membranous structures from dead oligodendrocytes and degenerative myelin deposited in the extracellular milieu. For the first time, this review covers mitochondrial mechanisms that may be operative during MS-related neurodegenerative changes directly activated during accumulating extracellular deposits of myelin associated inhibitory factors (MAIFs), that include the potent inhibitor of neurite outgrowth, Nogo-A. Axonal damage may occur when Nogo-A binds to and signals through its cognate receptor, NgR1, a multimeric complex, to initially stall axonal transport and limit the delivery of important growth-dependent cargo and subcellular organelles such as mitochondria for metabolic efficiency at sites of axo-glial disintegration as a consequence of inflammation. Metabolic efficiency in axons fails during active demyelination and progressive neurodegeneration, preceded by stalled transport of functional mitochondria to fuel axo-glial integrity.
Collapse
Affiliation(s)
- Danica Nheu
- Department of Neuroscience, School of Translational Medicine, Monash University, Prahran, VIC 3004, Australia
| | - Steven Petratos
- Department of Neuroscience, School of Translational Medicine, Monash University, Prahran, VIC 3004, Australia.
| |
Collapse
|
13
|
McGuinness HY, Gu W, Shi Y, Kobe B, Ve T. SARM1-Dependent Axon Degeneration: Nucleotide Signaling, Neurodegenerative Disorders, Toxicity, and Therapeutic Opportunities. Neuroscientist 2024; 30:473-492. [PMID: 37002660 PMCID: PMC11282687 DOI: 10.1177/10738584231162508] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
Axons are an essential component of the nervous system, and axon degeneration is an early feature of many neurodegenerative disorders. The NAD+ metabolome plays an essential role in regulating axonal integrity. Axonal levels of NAD+ and its precursor NMN are controlled in large part by the NAD+ synthesizing survival factor NMNAT2 and the pro-neurodegenerative NADase SARM1, whose activation triggers axon destruction. SARM1 has emerged as a promising axon-specific target for therapeutic intervention, and its function, regulation, structure, and role in neurodegenerative diseases have been extensively characterized in recent years. In this review, we first introduce the key molecular players involved in the SARM1-dependent axon degeneration program. Next, we summarize recent major advances in our understanding of how SARM1 is kept inactive in healthy neurons and how it becomes activated in injured or diseased neurons, which has involved important insights from structural biology. Finally, we discuss the role of SARM1 in neurodegenerative disorders and environmental neurotoxicity and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Helen Y. McGuinness
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Saint Lucia, Australia
| | - Weixi Gu
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Saint Lucia, Australia
| | - Yun Shi
- Institute for Glycomics, Griffith University, Gold Coast, Australia
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Saint Lucia, Australia
| | - Thomas Ve
- Institute for Glycomics, Griffith University, Gold Coast, Australia
| |
Collapse
|
14
|
Geisler S. Augustus Waller's foresight realized: SARM1 in peripheral neuropathies. Curr Opin Neurobiol 2024; 87:102884. [PMID: 38852438 PMCID: PMC11600426 DOI: 10.1016/j.conb.2024.102884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 06/11/2024]
Abstract
Peripheral neuropathy is a common neurodegenerative condition characterized by numbness, tingling, pain, and weakness that frequently starts in the distal limbs. Arising from multiple etiologies, many peripheral neuropathies exhibit a slowly progressive course due to axon degeneration for which no effective treatments exist. During the past decade, numerous crucial insights into mechanisms of axon degeneration in peripheral neuropathies emerged from experiments involving nerve-cutting procedures, revealing the central role of the SARM1 axon degeneration pathway in both. Here I review commonalities and differences in the role of SARM1 after nerve cut and in several acquired and inherited peripheral neuropathies. This new knowledge now paves the way for the development of therapeutics that directly address root causes of various kinds of neuropathies.
Collapse
Affiliation(s)
- Stefanie Geisler
- Department of Neurology, Washington University School of Medicine in St. Louis, 660S. Euclid Ave, Box 8111, St. Louis, MO 63110, USA.
| |
Collapse
|
15
|
Dingwall CB, Sasaki Y, Strickland A, Summers DW, Bloom AJ, DiAntonio A, Milbrandt J. Suppressing phagocyte activation by overexpressing the phosphatidylserine lipase ABHD12 preserves sarmopathic nerves. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.20.599919. [PMID: 38979309 PMCID: PMC11230269 DOI: 10.1101/2024.06.20.599919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Programmed axon degeneration (AxD) is a key feature of many neurodegenerative diseases. In healthy axons, the axon survival factor NMNAT2 inhibits SARM1, the central executioner of AxD, preventing it from initiating the rapid local NAD+ depletion and metabolic catastrophe that precipitates axon destruction. Because these components of the AxD pathway act within neurons, it was also assumed that the timetable of AxD was set strictly by a cell-intrinsic mechanism independent of neuron-extrinsic processes later activated by axon fragmentation. However, using a rare human disease model of neuropathy caused by hypomorphic NMNAT2 mutations and chronic SARM1 activation (sarmopathy), we demonstrated that neuronal SARM1 can initiate macrophage-mediated axon elimination long before stressed-but-viable axons would otherwise succumb to cell-intrinsic metabolic failure. Investigating potential SARM1-dependent signals that mediate macrophage recognition and/or engulfment of stressed-but-viable axons, we found that chronic SARM1 activation triggers axonal blebbing and dysregulation of phosphatidylserine (PS), a potent phagocyte immunomodulatory molecule. Neuronal expression of the phosphatidylserine lipase ABDH12 suppresses nerve macrophage activation, preserves motor axon integrity, and rescues motor function in this chronic sarmopathy model. We conclude that PS dysregulation is an early SARM1-dependent axonal stress signal, and that blockade of phagocytic recognition and engulfment of stressed-but-viable axons could be an attractive therapeutic target for management of neurological disorders involving SARM1 activation.
Collapse
|
16
|
Hinz FI, Villegas CLM, Roberts JT, Yao H, Gaddam S, Delwig A, Green SA, Fredrickson C, Adrian M, Asuncion RR, Cheung TK, Hayne M, Hackos DH, Rose CM, Richmond D, Hoogenraad CC. Context-Specific Stress Causes Compartmentalized SARM1 Activation and Local Degeneration in Cortical Neurons. J Neurosci 2024; 44:e2424232024. [PMID: 38692735 PMCID: PMC11170950 DOI: 10.1523/jneurosci.2424-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 04/21/2024] [Accepted: 04/23/2024] [Indexed: 05/03/2024] Open
Abstract
Sterile alpha and TIR motif containing 1 (SARM1) is an inducible NADase that localizes to mitochondria throughout neurons and senses metabolic changes that occur after injury. Minimal proteomic changes are observed upon either SARM1 depletion or activation, suggesting that SARM1 does not exert broad effects on neuronal protein homeostasis. However, whether SARM1 activation occurs throughout the neuron in response to injury and cell stress remains largely unknown. Using a semiautomated imaging pipeline and a custom-built deep learning scoring algorithm, we studied degeneration in both mixed-sex mouse primary cortical neurons and male human-induced pluripotent stem cell-derived cortical neurons in response to a number of different stressors. We show that SARM1 activation is differentially restricted to specific neuronal compartments depending on the stressor. Cortical neurons undergo SARM1-dependent axon degeneration after mechanical transection, and SARM1 activation is limited to the axonal compartment distal to the injury site. However, global SARM1 activation following vacor treatment causes both cell body and axon degeneration. Context-specific stressors, such as microtubule dysfunction and mitochondrial stress, induce axonal SARM1 activation leading to SARM1-dependent axon degeneration and SARM1-independent cell body death. Our data reveal that compartment-specific SARM1-mediated death signaling is dependent on the type of injury and cellular stressor.
Collapse
Affiliation(s)
- Flora I Hinz
- Department of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | | | - Jasmine T Roberts
- Department of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Heming Yao
- Biological Research | AI Development, Genentech, Inc., South San Francisco, California 94080
| | - Shreya Gaddam
- Biological Research | AI Development, Genentech, Inc., South San Francisco, California 94080
| | - Anton Delwig
- Departments of Biochemical and Cellular Pharmacology, Genentech, Inc., South San Francisco, California 94080
| | - Samantha A Green
- Discovery Chemistry, Genentech, Inc., South San Francisco, California 94080
| | - Craig Fredrickson
- Department of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Max Adrian
- Pathology, Genentech, Inc., South San Francisco, California 94080
| | - Raymond R Asuncion
- Transgenic Technology, Genentech, Inc., South San Francisco, California 94080
| | - Tommy K Cheung
- Microchemistry, Proteomics, and Lipidomics, Genentech, Inc., South San Francisco, California 94080
| | - Margaret Hayne
- Department of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - David H Hackos
- Department of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Christopher M Rose
- Microchemistry, Proteomics, and Lipidomics, Genentech, Inc., South San Francisco, California 94080
| | - David Richmond
- Biological Research | AI Development, Genentech, Inc., South San Francisco, California 94080
| | - Casper C Hoogenraad
- Department of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| |
Collapse
|
17
|
Garb J, Amitai G, Lu A, Ofir G, Brandis A, Mehlman T, Kranzusch PJ, Sorek R. The SARM1 TIR domain produces glycocyclic ADPR molecules as minor products. PLoS One 2024; 19:e0302251. [PMID: 38635746 PMCID: PMC11025887 DOI: 10.1371/journal.pone.0302251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 03/31/2024] [Indexed: 04/20/2024] Open
Abstract
Sterile alpha and TIR motif-containing 1 (SARM1) is a protein involved in programmed death of injured axons. Following axon injury or a drug-induced insult, the TIR domain of SARM1 degrades the essential molecule nicotinamide adenine dinucleotide (NAD+), leading to a form of axonal death called Wallerian degeneration. Degradation of NAD+ by SARM1 is essential for the Wallerian degeneration process, but accumulating evidence suggest that other activities of SARM1, beyond the mere degradation of NAD+, may be necessary for programmed axonal death. In this study we show that the TIR domains of both human and fruit fly SARM1 produce 1''-2' and 1''-3' glycocyclic ADP-ribose (gcADPR) molecules as minor products. As previously reported, we observed that SARM1 TIR domains mostly convert NAD+ to ADPR (for human SARM1) or cADPR (in the case of SARM1 from Drosophila melanogaster). However, we now show that human and Drosophila SARM1 additionally convert ~0.1-0.5% of NAD+ into gcADPR molecules. We find that SARM1 TIR domains produce gcADPR molecules both when purified in vitro and when expressed in bacterial cells. Given that gcADPR is a second messenger involved in programmed cell death in bacteria and likely in plants, we propose that gcADPR may play a role in SARM1-induced programmed axonal death in animals.
Collapse
Affiliation(s)
- Jeremy Garb
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Gil Amitai
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Allen Lu
- Department of Microbiology, Harvard Medical School, Boston, MA, United States of America
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, United States of America
| | - Gal Ofir
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Alexander Brandis
- Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Tevie Mehlman
- Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Philip J Kranzusch
- Department of Microbiology, Harvard Medical School, Boston, MA, United States of America
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, United States of America
- Parker Institute for Cancer Immunotherapy at Dana-Farber Cancer Institute, Boston, MA, United States of America
| | - Rotem Sorek
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
18
|
Yuan Y, Fang A, Wang H, Wang C, Sui B, Zhao J, Fu ZF, Zhou M, Zhao L. Lyssavirus M protein degrades neuronal microtubules by reprogramming mitochondrial metabolism. mBio 2024; 15:e0288023. [PMID: 38349129 PMCID: PMC10936203 DOI: 10.1128/mbio.02880-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/22/2024] [Indexed: 03/14/2024] Open
Abstract
Infection with neurotropic viruses may result in changes in host behavior, which are closely associated with degenerative changes in neurons. The lyssavirus genus comprises highly neurotropic viruses, including the rabies virus (RABV), which has been shown to induce degenerative changes in neurons, marked by the self-destruction of axons. The underlying mechanism by which the RABV degrades neuronal cytoskeletal proteins remains incomplete. In this study, we show that infection with RABV or overexpression of its M protein can disrupt mitochondrial metabolism by binding to Slc25a4. This leads to a reduction in NAD+ production and a subsequent influx of Ca2+ from the endoplasmic reticulum and mitochondria into the cytoplasm of neuronal cell lines, activating Ca2+-dependent proteinase calpains that degrade α-tubulin. We further screened the M proteins of different lyssaviruses and discovered that the M protein of the dog-derived RABV strain (DRV) does not degrade α-tubulin. Sequence analysis of the DRV M protein and that of the lab-attenuated RABV strain CVS revealed that the 57th amino acid is vital for M-induced microtubule degradation. We generated a recombinant RABV with a mutation at the 57th amino acid position in its M protein and showed that this mutation reduces α-tubulin degradation in vitro and axonal degeneration in vivo. This study elucidates the mechanism by which lyssavirus induces neuron degeneration.IMPORTANCEPrevious studies have suggested that RABV (rabies virus, the representative of lyssavirus) infection induces structural abnormalities in neurons. But there are few articles on the mechanism of lyssavirus' effect on neurons, and the mechanism of how RABV infection induces neurological dysfunction remains incomplete. The M protein of lyssavirus can downregulate cellular ATP levels by interacting with Slc25a4, and this decrease in ATP leads to a decrease in the level of NAD+ in the cytosol, which results in the release of Ca2+ from the intracellular calcium pool, the endoplasmic reticulum, and mitochondria. The presence of large amounts of Ca2+ in the cytoplasm activates Ca2+-dependent proteases and degrades microtubule proteins. The amino acid 57 of M protein is the key site determining its disruption of mitochondrial metabolism and subsequent neuron degeneration.
Collapse
Affiliation(s)
- Yueming Yuan
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - An Fang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Haoran Wang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Caiqian Wang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Baokun Sui
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Jianqing Zhao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Zhen F. Fu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Ming Zhou
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Ling Zhao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, China
- Hubei Hongshan Laboratory, Wuhan, China
| |
Collapse
|
19
|
Miyamoto T, Kim C, Chow J, Dugas JC, DeGroot J, Bagdasarian AL, Thottumkara AP, Larhammar M, Calvert ME, Fox BM, Lewcock JW, Kane LA. SARM1 is responsible for calpain-dependent dendrite degeneration in mouse hippocampal neurons. J Biol Chem 2024; 300:105630. [PMID: 38199568 PMCID: PMC10862016 DOI: 10.1016/j.jbc.2024.105630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 12/10/2023] [Accepted: 12/24/2023] [Indexed: 01/12/2024] Open
Abstract
Sterile alpha and toll/interleukin receptor motif-containing 1 (SARM1) is a critical regulator of axon degeneration that acts through hydrolysis of NAD+ following injury. Recent work has defined the mechanisms underlying SARM1's catalytic activity and advanced our understanding of SARM1 function in axons, yet the role of SARM1 signaling in other compartments of neurons is still not well understood. Here, we show in cultured hippocampal neurons that endogenous SARM1 is present in axons, dendrites, and cell bodies and that direct activation of SARM1 by the neurotoxin Vacor causes not just axon degeneration, but degeneration of all neuronal compartments. In contrast to the axon degeneration pathway defined in dorsal root ganglia, SARM1-dependent hippocampal axon degeneration in vitro is not sensitive to inhibition of calpain proteases. Dendrite degeneration downstream of SARM1 in hippocampal neurons is dependent on calpain 2, a calpain protease isotype enriched in dendrites in this cell type. In summary, these data indicate SARM1 plays a critical role in neurodegeneration outside of axons and elucidates divergent pathways leading to degeneration in hippocampal axons and dendrites.
Collapse
Affiliation(s)
| | - Chaeyoung Kim
- Denali Therapeutics Inc, South San Francisco, California, USA
| | - Johann Chow
- Denali Therapeutics Inc, South San Francisco, California, USA
| | - Jason C Dugas
- Denali Therapeutics Inc, South San Francisco, California, USA
| | - Jack DeGroot
- Denali Therapeutics Inc, South San Francisco, California, USA
| | | | | | | | | | - Brian M Fox
- Denali Therapeutics Inc, South San Francisco, California, USA
| | | | - Lesley A Kane
- Denali Therapeutics Inc, South San Francisco, California, USA.
| |
Collapse
|
20
|
Yang S, Niou ZX, Enriquez A, LaMar J, Huang JY, Ling K, Jafar-Nejad P, Gilley J, Coleman MP, Tennessen JM, Rangaraju V, Lu HC. NMNAT2 supports vesicular glycolysis via NAD homeostasis to fuel fast axonal transport. Mol Neurodegener 2024; 19:13. [PMID: 38282024 PMCID: PMC10823734 DOI: 10.1186/s13024-023-00690-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 11/28/2023] [Indexed: 01/30/2024] Open
Abstract
BACKGROUND Bioenergetic maladaptations and axonopathy are often found in the early stages of neurodegeneration. Nicotinamide adenine dinucleotide (NAD), an essential cofactor for energy metabolism, is mainly synthesized by Nicotinamide mononucleotide adenylyl transferase 2 (NMNAT2) in CNS neurons. NMNAT2 mRNA levels are reduced in the brains of Alzheimer's, Parkinson's, and Huntington's disease. Here we addressed whether NMNAT2 is required for axonal health of cortical glutamatergic neurons, whose long-projecting axons are often vulnerable in neurodegenerative conditions. We also tested if NMNAT2 maintains axonal health by ensuring axonal ATP levels for axonal transport, critical for axonal function. METHODS We generated mouse and cultured neuron models to determine the impact of NMNAT2 loss from cortical glutamatergic neurons on axonal transport, energetic metabolism, and morphological integrity. In addition, we determined if exogenous NAD supplementation or inhibiting a NAD hydrolase, sterile alpha and TIR motif-containing protein 1 (SARM1), prevented axonal deficits caused by NMNAT2 loss. This study used a combination of techniques, including genetics, molecular biology, immunohistochemistry, biochemistry, fluorescent time-lapse imaging, live imaging with optical sensors, and anti-sense oligos. RESULTS We provide in vivo evidence that NMNAT2 in glutamatergic neurons is required for axonal survival. Using in vivo and in vitro studies, we demonstrate that NMNAT2 maintains the NAD-redox potential to provide "on-board" ATP via glycolysis to vesicular cargos in distal axons. Exogenous NAD+ supplementation to NMNAT2 KO neurons restores glycolysis and resumes fast axonal transport. Finally, we demonstrate both in vitro and in vivo that reducing the activity of SARM1, an NAD degradation enzyme, can reduce axonal transport deficits and suppress axon degeneration in NMNAT2 KO neurons. CONCLUSION NMNAT2 ensures axonal health by maintaining NAD redox potential in distal axons to ensure efficient vesicular glycolysis required for fast axonal transport.
Collapse
Affiliation(s)
- Sen Yang
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA
- Program in Neuroscience, Indiana University, Bloomington, IN, 47405, USA
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, 33458, USA
| | - Zhen-Xian Niou
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA
- Program in Neuroscience, Indiana University, Bloomington, IN, 47405, USA
| | - Andrea Enriquez
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA
- Program in Neuroscience, Indiana University, Bloomington, IN, 47405, USA
| | - Jacob LaMar
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, 33458, USA
- Present address: Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, 33458, USA
| | - Jui-Yen Huang
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA
| | - Karen Ling
- Neuroscience Drug Discovery, Ionis Pharmaceuticals, Inc., 2855, Gazelle Court, Carlsbad, CA, 92010, USA
| | - Paymaan Jafar-Nejad
- Neuroscience Drug Discovery, Ionis Pharmaceuticals, Inc., 2855, Gazelle Court, Carlsbad, CA, 92010, USA
| | - Jonathan Gilley
- Department of Clinical Neuroscience, Cambridge University, Cambridge, UK
| | - Michael P Coleman
- Department of Clinical Neuroscience, Cambridge University, Cambridge, UK
| | - Jason M Tennessen
- Department of Biology, Indiana University, Bloomington, IN, 47405, USA
| | - Vidhya Rangaraju
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, 33458, USA
| | - Hui-Chen Lu
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA.
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA.
- Program in Neuroscience, Indiana University, Bloomington, IN, 47405, USA.
| |
Collapse
|
21
|
Alexandris AS, Koliatsos VE. NAD +, Axonal Maintenance, and Neurological Disease. Antioxid Redox Signal 2023; 39:1167-1184. [PMID: 37503611 PMCID: PMC10715442 DOI: 10.1089/ars.2023.0350] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 05/28/2023] [Indexed: 07/29/2023]
Abstract
Significance: The remarkable geometry of the axon exposes it to unique challenges for survival and maintenance. Axonal degeneration is a feature of peripheral neuropathies, glaucoma, and traumatic brain injury, and an early event in neurodegenerative diseases. Since the discovery of Wallerian degeneration (WD), a molecular program that hijacks nicotinamide adenine dinucleotide (NAD+) metabolism for axonal self-destruction, the complex roles of NAD+ in axonal viability and disease have become research priority. Recent Advances: The discoveries of the protective Wallerian degeneration slow (WldS) and of sterile alpha and TIR motif containing 1 (SARM1) activation as the main instructive signal for WD have shed new light on the regulatory role of NAD+ in axonal degeneration in a growing number of neurological diseases. SARM1 has been characterized as a NAD+ hydrolase and sensor of NAD+ metabolism. The discovery of regulators of nicotinamide mononucleotide adenylyltransferase 2 (NMNAT2) proteostasis in axons, the allosteric regulation of SARM1 by NAD+ and NMN, and the existence of clinically relevant windows of action of these signals has opened new opportunities for therapeutic interventions, including SARM1 inhibitors and modulators of NAD+ metabolism. Critical Issues: Events upstream and downstream of SARM1 remain unclear. Furthermore, manipulating NAD+ metabolism, an overdetermined process crucial in cell survival, for preventing the degeneration of the injured axon may be difficult and potentially toxic. Future Directions: There is a need for clarification of the distinct roles of NAD+ metabolism in axonal maintenance as contrasted to WD. There is also a need to better understand the role of NAD+ metabolism in axonal endangerment in neuropathies, diseases of the white matter, and the early stages of neurodegenerative diseases of the central nervous system. Antioxid. Redox Signal. 39, 1167-1184.
Collapse
Affiliation(s)
| | - Vassilis E. Koliatsos
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Neurology, and Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
22
|
Icso JD, Thompson PR. A phase transition reduces the threshold for nicotinamide mononucleotide-based activation of SARM1, an NAD(P) hydrolase, to physiologically relevant levels. J Biol Chem 2023; 299:105284. [PMID: 37742918 PMCID: PMC10624580 DOI: 10.1016/j.jbc.2023.105284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/04/2023] [Accepted: 09/19/2023] [Indexed: 09/26/2023] Open
Abstract
Axonal degeneration is a hallmark feature of neurodegenerative diseases. Activation of the NAD(P)ase sterile alpha and toll-interleukin receptor motif containing protein 1 (SARM1) is critical for this process. In resting neurons, SARM1 activity is inhibited, but upon damage, SARM1 is activated and catalyzes one of three NAD(P)+ dependent reactions: (1) NAD(P)+ hydrolysis to form ADP-ribose (ADPR[P]) and nicotinamide; (2) the formation of cyclic-ADPR (cADPR[P]); or (3) a base exchange reaction with nicotinic acid (NA) and NADP+ to form NA adenine dinucleotide phosphate. Production of these metabolites triggers axonal death. Two activation mechanisms have been proposed: (1) an increase in the nicotinamide mononucleotide (NMN) concentration, which leads to the allosteric activation of SARM1, and (2) a phase transition, which stabilizes the active conformation of the enzyme. However, neither of these mechanisms have been shown to occur at the same time. Using in vitro assay systems, we show that the liquid-to-solid phase transition lowers the NMN concentration required to activate the catalytic activity of SARM1 by up to 140-fold. These results unify the proposed activation mechanisms and show for the first time that a phase transition reduces the threshold for NMN-based SARM1 activation to physiologically relevant levels. These results further our understanding of SARM1 activation and will be important for the future development of therapeutics targeting SARM1.
Collapse
Affiliation(s)
- Janneke Doedée Icso
- Program in Chemical Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA; Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medial School, Worcester, Massachusetts, USA
| | - Paul Ryan Thompson
- Program in Chemical Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA; Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medial School, Worcester, Massachusetts, USA.
| |
Collapse
|
23
|
Huang D, Chen S, Xiong D, Wang H, Zhu L, Wei Y, Li Y, Zou S. Mitochondrial Dynamics: Working with the Cytoskeleton and Intracellular Organelles to Mediate Mechanotransduction. Aging Dis 2023; 14:1511-1532. [PMID: 37196113 PMCID: PMC10529762 DOI: 10.14336/ad.2023.0201] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 02/01/2023] [Indexed: 05/19/2023] Open
Abstract
Cells are constantly exposed to various mechanical environments; therefore, it is important that they are able to sense and adapt to changes. It is known that the cytoskeleton plays a critical role in mediating and generating extra- and intracellular forces and that mitochondrial dynamics are crucial for maintaining energy homeostasis. Nevertheless, the mechanisms by which cells integrate mechanosensing, mechanotransduction, and metabolic reprogramming remain poorly understood. In this review, we first discuss the interaction between mitochondrial dynamics and cytoskeletal components, followed by the annotation of membranous organelles intimately related to mitochondrial dynamic events. Finally, we discuss the evidence supporting the participation of mitochondria in mechanotransduction and corresponding alterations in cellular energy conditions. Notable advances in bioenergetics and biomechanics suggest that the mechanotransduction system composed of mitochondria, the cytoskeletal system, and membranous organelles is regulated through mitochondrial dynamics, which may be a promising target for further investigation and precision therapies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yuyu Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Shujuan Zou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
24
|
Khazma T, Grossman A, Guez-Haddad J, Feng C, Dabas H, Sain R, Weitman M, Zalk R, Isupov MN, Hammarlund M, Hons M, Opatowsky Y. Structure-function analysis of ceTIR-1/hSARM1 explains the lack of Wallerian axonal degeneration in C. elegans. Cell Rep 2023; 42:113026. [PMID: 37635352 PMCID: PMC10675840 DOI: 10.1016/j.celrep.2023.113026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 06/05/2023] [Accepted: 08/09/2023] [Indexed: 08/29/2023] Open
Abstract
Wallerian axonal degeneration (WD) does not occur in the nematode C. elegans, in contrast to other model animals. However, WD depends on the NADase activity of SARM1, a protein that is also expressed in C. elegans (ceSARM/ceTIR-1). We hypothesized that differences in SARM between species might exist and account for the divergence in WD. We first show that expression of the human (h)SARM1, but not ceTIR-1, in C. elegans neurons is sufficient to confer axon degeneration after nerve injury. Next, we determined the cryoelectron microscopy structure of ceTIR-1 and found that, unlike hSARM1, which exists as an auto-inhibited ring octamer, ceTIR-1 forms a readily active 9-mer. Enzymatically, the NADase activity of ceTIR-1 is substantially weaker (10-fold higher Km) than that of hSARM1, and even when fully active, it falls short of consuming all cellular NAD+. Our experiments provide insight into the molecular mechanisms and evolution of SARM orthologs and WD across species.
Collapse
Affiliation(s)
- Tami Khazma
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Atira Grossman
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Julia Guez-Haddad
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Chengye Feng
- Departments of Neuroscience and Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Hadas Dabas
- Departments of Neuroscience and Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Radhika Sain
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Michal Weitman
- Department of Chemistry, Bar-Ilan University, Ramat Gan, Israel
| | - Ran Zalk
- Ilse Katz Institute for Nanoscale Science & Technology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Michail N Isupov
- Henry Wellcome Building for Biocatalysis, Biosciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Marc Hammarlund
- Departments of Neuroscience and Genetics, Yale School of Medicine, New Haven, CT, USA.
| | - Michael Hons
- European Molecular Biology Laboratory, Grenoble, France.
| | - Yarden Opatowsky
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel.
| |
Collapse
|
25
|
Icso JD, Barasa L, Thompson PR. SARM1, an Enzyme Involved in Axon Degeneration, Catalyzes Multiple Activities through a Ternary Complex Mechanism. Biochemistry 2023; 62:2065-2078. [PMID: 37307562 DOI: 10.1021/acs.biochem.3c00081] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Sterile alpha and toll/interleukin receptor (TIR) motif containing protein 1 (SARM1) is an NAD+ hydrolase and cyclase involved in axonal degeneration. In addition to NAD+ hydrolysis and cyclization, SARM1 catalyzes a base exchange reaction between nicotinic acid (NA) and NADP+ to generate NAADP, which is a potent calcium signaling molecule. Herein, we describe efforts to characterize the hydrolysis, cyclization, and base exchange activities of TIR-1, the Caenorhabditis elegans ortholog of SARM1; TIR-1 also catalyzes NAD(P)+ hydrolysis and/or cyclization and regulates axonal degeneration in worms. We show that the catalytic domain of TIR-1 undergoes a liquid-to-solid phase transition that regulates not only the hydrolysis and cyclization reactions but also the base exchange reaction. We define the substrate specificities of the reactions, demonstrate that cyclization and base exchange reactions occur within the same pH range, and establish that TIR-1 uses a ternary complex mechanism. Overall, our findings will aid drug discovery efforts and provide insight into the mechanism of recently described inhibitors.
Collapse
Affiliation(s)
- Janneke D Icso
- Program in Chemical Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605, United States
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medial School, Worcester, Massachusetts 01605, United States
| | - Leonard Barasa
- Program in Chemical Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605, United States
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medial School, Worcester, Massachusetts 01605, United States
| | - Paul R Thompson
- Program in Chemical Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605, United States
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medial School, Worcester, Massachusetts 01605, United States
| |
Collapse
|
26
|
Barros LF, Ruminot I, Sandoval PY, San Martín A. Enlightening brain energy metabolism. Neurobiol Dis 2023:106211. [PMID: 37352985 DOI: 10.1016/j.nbd.2023.106211] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/06/2023] [Accepted: 06/20/2023] [Indexed: 06/25/2023] Open
Abstract
Brain tissue metabolism is distributed across several cell types and subcellular compartments, which activate at different times and with different temporal patterns. The introduction of genetically-encoded fluorescent indicators that are imaged using time-lapse microscopy has opened the possibility of studying brain metabolism at cellular and sub-cellular levels. There are indicators for sugars, monocarboxylates, Krebs cycle intermediates, amino acids, cofactors, and energy nucleotides, which inform about relative levels, concentrations and fluxes. This review offers a brief survey of the metabolic indicators that have been validated in brain cells, with some illustrative examples from the literature. Whereas only a small fraction of the metabolome is currently accessible to fluorescent probes, there are grounds to be optimistic about coming developments and the application of these tools to the study of brain disease.
Collapse
Affiliation(s)
- L F Barros
- Centro de Estudios Científicos (CECs), Valdivia, Chile; Facultad de Medicina y Ciencia, Universidad San Sebastián, Valdivia, Chile.
| | - I Ruminot
- Centro de Estudios Científicos (CECs), Valdivia, Chile; Facultad de Ciencias para el Cuidado de La Salud, Universidad San Sebastián, Valdivia, Chile
| | - P Y Sandoval
- Centro de Estudios Científicos (CECs), Valdivia, Chile; Facultad de Ciencias para el Cuidado de La Salud, Universidad San Sebastián, Valdivia, Chile
| | - A San Martín
- Centro de Estudios Científicos (CECs), Valdivia, Chile; Facultad de Ciencias para el Cuidado de La Salud, Universidad San Sebastián, Valdivia, Chile
| |
Collapse
|
27
|
Wan L. TIR enzymatic functions: signaling molecules and receptor mechanisms. ABIOTECH 2023; 4:172-175. [PMID: 37581018 PMCID: PMC10423176 DOI: 10.1007/s42994-023-00104-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 04/25/2023] [Indexed: 08/16/2023]
Abstract
The evolutionarily conserved Toll/Interleukin-1 Receptor (TIR) domains across kingdoms of prokaryotes, plants, and animals play critical roles in innate immunity. Recent studies have revealed the enzymatic functions of TIRs, the structural bases of TIRs as holoenzymes, and the identity of TIR-generated small signaling molecules and their receptors, which significantly advanced our understanding on TIR-mediated immune signaling pathways. We reviewed the most up-to-date findings in TIR enzymatic functions from the perspectives of signaling molecules and receptor mechanisms.
Collapse
Affiliation(s)
- Li Wan
- National Key Laboratory of Plant Molecular Genetics, CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, 200032 China
| |
Collapse
|
28
|
Fague L, Marsh-Armstrong N. Dual leucine zipper kinase is necessary for retinal ganglion cell axonal regeneration in Xenopus laevis. PNAS NEXUS 2023; 2:pgad109. [PMID: 37152673 PMCID: PMC10162689 DOI: 10.1093/pnasnexus/pgad109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 03/08/2023] [Indexed: 05/09/2023]
Abstract
Retinal ganglion cell (RGC) axons of the African clawed frog, Xenopus laevis, unlike those of mammals, are capable of regeneration and functional reinnervation of central brain targets following injury. Here, we describe a tadpole optic nerve crush (ONC) procedure and assessments of brain reinnervation based on live imaging of RGC-specific transgenes which, when paired with CRISPR/Cas9 injections at the one-cell stage, can be used to assess the function of regeneration-associated genes in vivo in F0 animals. Using this assay, we find that map3k12, also known as dual leucine zipper kinase (Dlk), is necessary for RGC axonal regeneration and acts in a dose-dependent manner. Loss of Dlk does not affect RGC innervation of the brain during development or visually driven behavior but does block both axonal regeneration and functional vision restoration after ONC. Dlk loss does not alter the acute changes in mitochondrial movement that occur within RGC axons hours after ONC but does completely block the phosphorylation and nuclear translocation of the transcription factor Jun within RGCs days after ONC; yet, Jun is dispensable for reinnervation. These results demonstrate that in a species fully capable of regenerating its RGC axons, Dlk is essential for the axonal injury signal to reach the nucleus but may affect regeneration through a different pathway than by which it signals in mammalian RGCs.
Collapse
Affiliation(s)
- Lindsay Fague
- Department of Ophthalmology and Vision Science, UC Davis Eye Center, University of California, Davis, 1275 Med Science Drive Rm. 3451, Davis, CA 95616, USA
| | | |
Collapse
|
29
|
Kikuchi S, Kohno T, Kojima T, Tatsumi H, Ohsaki Y, Ninomiya T. Oxygen-Glucose Deprivation Decreases the Motility and Length of Axonal Mitochondria in Cultured Dorsal Root Ganglion Cells of Rats. Cell Mol Neurobiol 2023; 43:1267-1280. [PMID: 35771293 PMCID: PMC11414435 DOI: 10.1007/s10571-022-01247-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 06/20/2022] [Indexed: 11/03/2022]
Abstract
Controlling axonal mitochondria is important for maintaining normal function of the neural network. Oxygen-glucose deprivation (OGD), a model used for mimicking ischemia, eventually induces neuronal cell death similar to axonal degeneration. Axonal mitochondria are disrupted during OGD-induced neural degeneration; however, the mechanism underlying mitochondrial dysfunction has not been completely understood. We focused on the dynamics of mitochondria in axons exposed to OGD; we observed that the number of motile mitochondria significantly reduced in 1 h following OGD exposure. In our observation, the decreased length of stationary mitochondria was affected by the following factors: first, the halt of motile mitochondria; second, the fission of longer stationary mitochondria; and third, a transformation from tubular to spherical shape in OGD-exposed axons. Motile mitochondria reduction preceded stationary mitochondria fragmentation in OGD exposure; these conditions induced the decrease of stationary mitochondria in three different ways. Our results suggest that mitochondrial morphological changes precede the axonal degeneration while ischemia-induced neurodegeneration.
Collapse
Affiliation(s)
- Shin Kikuchi
- Department of Anatomy 1, Sapporo Medical University School of Medicine, West 17, South 1, Chuo-ku, Sapporo, Hokkaido, 060-8556, Japan.
| | - Takayuki Kohno
- Department of Cell Sciences, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, West 17, South 1, Chuo-ku, Sapporo, Hokkaido, 060-8556, Japan
| | - Takashi Kojima
- Department of Cell Sciences, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, West 17, South 1, Chuo-ku, Sapporo, Hokkaido, 060-8556, Japan
| | - Haruyuki Tatsumi
- Department of Anatomy 1, Sapporo Medical University School of Medicine, West 17, South 1, Chuo-ku, Sapporo, Hokkaido, 060-8556, Japan
| | - Yuki Ohsaki
- Department of Anatomy 1, Sapporo Medical University School of Medicine, West 17, South 1, Chuo-ku, Sapporo, Hokkaido, 060-8556, Japan
| | - Takafumi Ninomiya
- Department of Anatomy 1, Sapporo Medical University School of Medicine, West 17, South 1, Chuo-ku, Sapporo, Hokkaido, 060-8556, Japan
| |
Collapse
|
30
|
Sarkar A, Dutta S, Sur M, Chakraborty S, Dey P, Mukherjee P. Early loss of endogenous NAD + following rotenone treatment leads to mitochondrial dysfunction and Sarm1 induction that is ameliorated by PARP inhibition. FEBS J 2023; 290:1596-1624. [PMID: 36239430 DOI: 10.1111/febs.16652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 08/17/2022] [Accepted: 10/13/2022] [Indexed: 11/07/2022]
Abstract
Sarm1 is an evolutionary conserved innate immune adaptor protein that has emerged as a primary regulator of programmed axonal degeneration over the past decade. In vitro structural insights have revealed that although Sarm1 induces energy depletion by breaking down nicotinamide adenine dinucleotide+ (NAD+ ), it is also allosterically inhibited by NAD+ . However, how NAD+ levels modulate the activation of intracellular Sarm1 has not been elucidated so far. This study focuses on understanding the events leading to Sarm1 activation in both neuronal and non-neuronal cells using the mitochondrial complex I inhibitor rotenone. Here, we report the regulation of rotenone-induced cell death by loss of NAD+ that may act as a 'biological trigger' of Sarm1 activation. Our study revealed that early loss of endogenous NAD+ levels arising due to PARP1 hyperactivation preceded Sarm1 induction following rotenone treatment. Interestingly, replenishing NAD+ levels by the PARP inhibitor, PJ34 restored mitochondrial complex I activity and also prevented subsequent Sarm1 activation in rotenone-treated cells. These cellular data were further validated in Drosophila melanogaster where a significant reduction in rotenone-mediated loss of locomotor abilities, and reduced dSarm expression was observed in the flies following PARP inhibition. Taken together, these observations not only uncover a novel regulation of Sarm1 induction by endogenous NAD+ levels but also point towards an important understanding on how PARP inhibitors could be repurposed in the treatment of mitochondrial complex I deficiency disorders.
Collapse
Affiliation(s)
- Ankita Sarkar
- Institute of Health Sciences, Presidency University, Kolkata, India
| | - Sourav Dutta
- Institute of Health Sciences, Presidency University, Kolkata, India
| | - Malinki Sur
- Institute of Health Sciences, Presidency University, Kolkata, India
| | | | - Puja Dey
- Institute of Health Sciences, Presidency University, Kolkata, India
| | - Piyali Mukherjee
- Institute of Health Sciences, Presidency University, Kolkata, India
| |
Collapse
|
31
|
A conformation-specific nanobody targeting the nicotinamide mononucleotide-activated state of SARM1. Nat Commun 2022; 13:7898. [PMID: 36550129 PMCID: PMC9780360 DOI: 10.1038/s41467-022-35581-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
Sterile alpha (SAM) and Toll/interleukin-1 receptor (TIR) motif containing 1 (SARM1) is an autoinhibitory NAD-consuming enzyme that is activated by the accumulation of nicotinamide mononucleotide (NMN) during axonal injury. Its activation mechanism is not fully understood. Here, we generate a nanobody, Nb-C6, that specifically recognizes NMN-activated SARM1. Nb-C6 stains only the activated SARM1 in cells stimulated with CZ-48, a permeant mimetic of NMN, and partially activates SARM1 in vitro and in cells. Cryo-EM of NMN/SARM1/Nb-C6 complex shows an octameric structure with ARM domains bending significantly inward and swinging out together with TIR domains. Nb-C6 binds to SAM domain of the activated SARM1 and stabilized its ARM domain. Mass spectrometry analyses indicate that the activated SARM1 in solution is highly dynamic and that the neighboring TIRs form transient dimers via the surface close to one BB loop. We show that Nb-C6 is a valuable tool for studies of SARM1 activation.
Collapse
|
32
|
Dingwall CB, Strickland A, Yum SW, Yim AK, Zhu J, Wang PL, Yamada Y, Schmidt RE, Sasaki Y, Bloom AJ, DiAntonio A, Milbrandt J. Macrophage depletion blocks congenital SARM1-dependent neuropathy. J Clin Invest 2022; 132:e159800. [PMID: 36287209 PMCID: PMC9711884 DOI: 10.1172/jci159800] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 10/04/2022] [Indexed: 11/17/2022] Open
Abstract
Axon loss contributes to many common neurodegenerative disorders. In healthy axons, the axon survival factor NMNAT2 inhibits SARM1, the central executioner of programmed axon degeneration. We identified 2 rare NMNAT2 missense variants in 2 brothers afflicted with a progressive neuropathy syndrome. The polymorphisms resulted in amino acid substitutions V98M and R232Q, which reduced NMNAT2 NAD+-synthetase activity. We generated a mouse model to mirror the human syndrome and found that Nmnat2V98M/R232Q compound-heterozygous CRISPR mice survived to adulthood but developed progressive motor dysfunction, peripheral axon loss, and macrophage infiltration. These disease phenotypes were all SARM1-dependent. Remarkably, macrophage depletion therapy blocked and reversed neuropathic phenotypes in Nmnat2V98M/R232Q mice, identifying a SARM1-dependent neuroimmune mechanism as a key driver of disease pathogenesis. These findings demonstrate that SARM1 induced inflammatory neuropathy and highlight the potential of immune therapy as a treatment for this rare syndrome and other neurodegenerative conditions associated with NMNAT2 loss and SARM1 activation.
Collapse
Affiliation(s)
- Caitlin B. Dingwall
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Amy Strickland
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Sabrina W. Yum
- Division of Neurology, Children’s Hospital of Philadelphia, Department of Neurology, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Aldrin K.Y. Yim
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jian Zhu
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Peter L. Wang
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Yurie Yamada
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Robert E. Schmidt
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Yo Sasaki
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - A. Joseph Bloom
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
- Needleman Center for Neurometabolism and Axonal Therapeutics, St. Louis, Missouri, USA
| | - Aaron DiAntonio
- Needleman Center for Neurometabolism and Axonal Therapeutics, St. Louis, Missouri, USA
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jeffrey Milbrandt
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
- Needleman Center for Neurometabolism and Axonal Therapeutics, St. Louis, Missouri, USA
| |
Collapse
|
33
|
Sato-Yamada Y, Strickland A, Sasaki Y, Bloom J, DiAntonio A, Milbrandt J. A SARM1-mitochondrial feedback loop drives neuropathogenesis in a Charcot-Marie-Tooth disease type 2A rat model. J Clin Invest 2022; 132:e161566. [PMID: 36287202 PMCID: PMC9711878 DOI: 10.1172/jci161566] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 10/11/2022] [Indexed: 11/17/2022] Open
Abstract
Charcot-Marie-Tooth disease type 2A (CMT2A) is an axonal neuropathy caused by mutations in the mitofusin 2 (MFN2) gene. MFN2 mutations result in profound mitochondrial abnormalities, but the mechanism underlying the axonal pathology is unknown. Sterile α and Toll/IL-1 receptor motif-containing 1 (SARM1), the central executioner of axon degeneration, can induce neuropathy and is activated by dysfunctional mitochondria. We tested the role of SARM1 in a rat model carrying a dominant CMT2A mutation (Mfn2H361Y) that exhibits progressive dying-back axonal degeneration, neuromuscular junction (NMJ) abnormalities, muscle atrophy, and mitochondrial abnormalities - all hallmarks of the human disease. We generated Sarm1-KO (Sarm1-/-) and Mfn2H361Y Sarm1 double-mutant rats and found that deletion of Sarm1 rescued axonal, synaptic, muscle, and functional phenotypes, demonstrating that SARM1 was responsible for much of the neuropathology in this model. Despite the presence of mutant MFN2 protein in these double-mutant rats, loss of SARM1 also dramatically suppressed many mitochondrial defects, including the number, size, and cristae density defects of synaptic mitochondria. This surprising finding indicates that dysfunctional mitochondria activated SARM1 and that activated SARM1 fed back on mitochondria to exacerbate the mitochondrial pathology. As such, this work identifies SARM1 inhibition as a therapeutic candidate for the treatment of CMT2A and other neurodegenerative diseases with prominent mitochondrial pathology.
Collapse
Affiliation(s)
- Yurie Sato-Yamada
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
- Center for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Science, Niigata City, Japan
| | - Amy Strickland
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Yo Sasaki
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Joseph Bloom
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
- Needleman Center for Neurometabolism and Axonal Therapeutics, St. Louis, Missouri, USA
| | - Aaron DiAntonio
- Needleman Center for Neurometabolism and Axonal Therapeutics, St. Louis, Missouri, USA
- Department of Developmental Biology and
| | - Jeffrey Milbrandt
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
- Needleman Center for Neurometabolism and Axonal Therapeutics, St. Louis, Missouri, USA
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
34
|
Eastman S, Bayless A, Guo M. The Nucleotide Revolution: Immunity at the Intersection of Toll/Interleukin-1 Receptor Domains, Nucleotides, and Ca 2. MOLECULAR PLANT-MICROBE INTERACTIONS : MPMI 2022; 35:964-976. [PMID: 35881867 DOI: 10.1094/mpmi-06-22-0132-cr] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The discovery of the enzymatic activity of the toll/interleukin-1 receptor (TIR) domain protein SARM1 five years ago preceded a flood of discoveries regarding the nucleotide substrates and products of TIR domains in plants, animals, bacteria, and archaea. These discoveries into the activity of TIR domains coincide with major advances in understanding the structure and mechanisms of NOD-like receptors and the mutual dependence of pattern recognition receptor- and effector-triggered immunity (PTI and ETI, respectively) in plants. It is quickly becoming clear that TIR domains and TIR-produced nucleotides are ancestral signaling molecules that modulate immunity and that their activity is closely associated with Ca2+ signaling. TIR domain research now bridges the separate disciplines of molecular plant- and animal-microbe interactions, neurology, and prokaryotic immunity. A cohesive framework for understanding the role of enzymatic TIR domains in diverse organisms will help unite the research of these disparate fields. Here, we review known products of TIR domains in plants, animals, bacteria, and archaea and use context gained from animal and prokaryotic TIR domain systems to present a model for TIR domains, nucleotides, and Ca2+ at the intersection of PTI and ETI in plant immunity. [Formula: see text] Copyright © 2022 The Author(s). This is an open access article distributed under the CC BY 4.0 International license.
Collapse
Affiliation(s)
- Samuel Eastman
- Department of Plant Pathology, University of Nebraska-Lincoln, Lincoln, NE 68583, U.S.A
| | - Adam Bayless
- Department of Biology, Colorado State University, Fort Collins, CO 80521, U.S.A
| | - Ming Guo
- Department of Agriculture and Horticulture, University of Nebraska-Lincoln, Lincoln, NE 68583, U.S.A
| |
Collapse
|
35
|
Tay N, Laakso EL, Schweitzer D, Endersby R, Vetter I, Starobova H. Chemotherapy-induced peripheral neuropathy in children and adolescent cancer patients. Front Mol Biosci 2022; 9:1015746. [PMID: 36310587 PMCID: PMC9614173 DOI: 10.3389/fmolb.2022.1015746] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 09/20/2022] [Indexed: 11/22/2022] Open
Abstract
Brain cancer and leukemia are the most common cancers diagnosed in the pediatric population and are often treated with lifesaving chemotherapy. However, chemotherapy causes severe adverse effects and chemotherapy-induced peripheral neuropathy (CIPN) is a major dose-limiting and debilitating side effect. CIPN can greatly impair quality of life and increases morbidity of pediatric patients with cancer, with the accompanying symptoms frequently remaining underdiagnosed. Little is known about the incidence of CIPN, its impact on the pediatric population, and the underlying pathophysiological mechanisms, as most existing information stems from studies in animal models or adult cancer patients. Herein, we aim to provide an understanding of CIPN in the pediatric population and focus on the 6 main substance groups that frequently cause CIPN, namely the vinca alkaloids (vincristine), platinum-based antineoplastics (cisplatin, carboplatin and oxaliplatin), taxanes (paclitaxel and docetaxel), epothilones (ixabepilone), proteasome inhibitors (bortezomib) and immunomodulatory drugs (thalidomide). We discuss the clinical manifestations, assessments and diagnostic tools, as well as risk factors, pathophysiological processes and current pharmacological and non-pharmacological approaches for the prevention and treatment of CIPN.
Collapse
Affiliation(s)
- Nicolette Tay
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - E-Liisa Laakso
- Mater Research Institute-The University of Queensland, South Brisbane, QLD, Australia
| | - Daniel Schweitzer
- Mater Research Institute-The University of Queensland, South Brisbane, QLD, Australia
| | - Raelene Endersby
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Irina Vetter
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
- The School of Pharmacy, The University of Queensland, Woolloongabba, QLD, Australia
| | - Hana Starobova
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
- *Correspondence: Hana Starobova,
| |
Collapse
|
36
|
Nizami HL, Minor KE, Chiao YA, Light CM, Lee CF. Sexually dimorphic effects of SARM1 deletion on cardiac NAD + metabolism and function. Am J Physiol Heart Circ Physiol 2022; 323:H774-H781. [PMID: 36053750 PMCID: PMC9529255 DOI: 10.1152/ajpheart.00370.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/17/2022] [Accepted: 08/24/2022] [Indexed: 11/22/2022]
Abstract
Nicotinamide adenine dinucleotide (NAD+) decline is repeatedly observed in heart disease and its risk factors. Although strategies promoting NAD+ synthesis to elevate NAD+ levels improve cardiac function, whether inhibition of NAD+ consumption can be therapeutic is less investigated. In this study, we examined the role of sterile-α and TIR motif containing 1 (SARM1) NAD+ hydrolase in mouse hearts, using global SARM1-knockout mice (KO). Cardiac function was assessed by echocardiography in male and female KO mice and wild-type (WT) controls. Hearts were collected for biochemical, histological, and molecular analyses. We found that the cardiac NAD+ pool was elevated in female KO mice, but only trended to increase in male KO mice. SARM1 deletion induced changes to a greater number of NAD+ metabolism transcripts in male mice than in female mice. Body weights, cardiac systolic and diastolic function, and geometry showed no changes in both male and female KO mice compared with WT counterparts. Male KO mice showed a small, but significant, elevation in cardiac collagen levels compared with WT counterparts, but no difference in collagen levels was detected in female mice. The increased collagen levels were associated with greater number of altered profibrotic and senescence-associated inflammatory genes in male KO mice, but not in female KO mice.NEW & NOTEWORTHY We examined the effects of SARM1 deletion on NAD+ pool, transcripts of NAD+ metabolism, and fibrotic pathway for the first time in mouse hearts. We observed the sexually dimorphic effects of SARM1 deletion. How these sex-dependent effects influence the outcomes of SARM1 deficiency in male and female mice in responses to cardiac stresses warrant further investigation. The elevation of cardiac NAD+ pool by SARM1 deletion provides evidence that targeting SARM1 may reverse disease-related NAD+ decline.
Collapse
Affiliation(s)
- Hina Lateef Nizami
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Keaton E Minor
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Ying Ann Chiao
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Christine M Light
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Chi Fung Lee
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
37
|
Martínez-Gil N, Maneu V, Kutsyr O, Fernández-Sánchez L, Sánchez-Sáez X, Sánchez-Castillo C, Campello L, Lax P, Pinilla I, Cuenca N. Cellular and molecular alterations in neurons and glial cells in inherited retinal degeneration. Front Neuroanat 2022; 16:984052. [PMID: 36225228 PMCID: PMC9548552 DOI: 10.3389/fnana.2022.984052] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/29/2022] [Indexed: 11/19/2022] Open
Abstract
Multiple gene mutations have been associated with inherited retinal dystrophies (IRDs). Despite the spectrum of phenotypes caused by the distinct mutations, IRDs display common physiopathology features. Cell death is accompanied by inflammation and oxidative stress. The vertebrate retina has several attributes that make this tissue vulnerable to oxidative and nitrosative imbalance. The high energy demands and active metabolism in retinal cells, as well as their continuous exposure to high oxygen levels and light-induced stress, reveal the importance of tightly regulated homeostatic processes to maintain retinal function, which are compromised in pathological conditions. In addition, the subsequent microglial activation and gliosis, which triggers the secretion of pro-inflammatory cytokines, chemokines, trophic factors, and other molecules, further worsen the degenerative process. As the disease evolves, retinal cells change their morphology and function. In disease stages where photoreceptors are lost, the remaining neurons of the retina to preserve their function seek out for new synaptic partners, which leads to a cascade of morphological alterations in retinal cells that results in a complete remodeling of the tissue. In this review, we describe important molecular and morphological changes in retinal cells that occur in response to oxidative stress and the inflammatory processes underlying IRDs.
Collapse
Affiliation(s)
- Natalia Martínez-Gil
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Victoria Maneu
- Department of Optics, Pharmacology and Anatomy, University of Alicante, Alicante, Spain
| | - Oksana Kutsyr
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | | | - Xavier Sánchez-Sáez
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Carla Sánchez-Castillo
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Laura Campello
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Pedro Lax
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
- Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
| | - Isabel Pinilla
- Aragón Institute for Health Research (IIS Aragón), Zaragoza, Spain
- Department of Ophthalmology, Lozano Blesa University Hospital, Zaragoza, Spain
- Department of Surgery, University of Zaragoza, Zaragoza, Spain
- Isabel Pinilla,
| | - Nicolás Cuenca
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
- Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
- Institute Ramón Margalef, University of Alicante, Alicante, Spain
- *Correspondence: Nicolás Cuenca,
| |
Collapse
|
38
|
Waller TJ, Collins CA. Multifaceted roles of SARM1 in axon degeneration and signaling. Front Cell Neurosci 2022; 16:958900. [PMID: 36090788 PMCID: PMC9453223 DOI: 10.3389/fncel.2022.958900] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/09/2022] [Indexed: 12/01/2022] Open
Abstract
Axons are considered to be particularly vulnerable components of the nervous system; impairments to a neuron’s axon leads to an effective silencing of a neuron’s ability to communicate with other cells. Nervous systems have therefore evolved plasticity mechanisms for adapting to axonal damage. These include acute mechanisms that promote the degeneration and clearance of damaged axons and, in some cases, the initiation of new axonal growth and synapse formation to rebuild lost connections. Here we review how these diverse processes are influenced by the therapeutically targetable enzyme SARM1. SARM1 catalyzes the breakdown of NAD+, which, when unmitigated, can lead to rundown of this essential metabolite and axonal degeneration. SARM1’s enzymatic activity also triggers the activation of downstream signaling pathways, which manifest numerous functions for SARM1 in development, innate immunity and responses to injury. Here we will consider the multiple intersections between SARM1 and the injury signaling pathways that coordinate cellular adaptations to nervous system damage.
Collapse
Affiliation(s)
- Thomas J. Waller
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| | - Catherine A. Collins
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH, United States
- *Correspondence: Catherine A. Collins,
| |
Collapse
|
39
|
Essuman K, Milbrandt J, Dangl JL, Nishimura MT. Shared TIR enzymatic functions regulate cell death and immunity across the tree of life. Science 2022; 377:eabo0001. [DOI: 10.1126/science.abo0001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In the 20th century, researchers studying animal and plant signaling pathways discovered a protein domain shared across diverse innate immune systems: the Toll/Interleukin-1/Resistance-gene (TIR) domain. The TIR domain is found in several protein architectures and was defined as an adaptor mediating protein-protein interactions in animal innate immunity and developmental signaling pathways. However, studies of nerve degeneration in animals, and subsequent breakthroughs in plant, bacterial and archaeal systems, revealed that TIR domains possess enzymatic activities. We provide a synthesis of TIR functions and the role of various related TIR enzymatic products in evolutionarily diverse immune systems. These studies may ultimately guide interventions that would span the tree of life, from treating human neurodegenerative disorders and bacterial infections, to preventing plant diseases.
Collapse
Affiliation(s)
- Kow Essuman
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Jeffrey Milbrandt
- Department of Genetics, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
- Needleman Center for Neurometabolism and Axonal Therapeutics, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
- McDonnell Genome Institute, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Jeffery L. Dangl
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Howard Hughes Medical Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Marc T. Nishimura
- Department of Biology, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
40
|
Herrmann KA, Liu Y, Llobet-Rosell A, McLaughlin CN, Neukomm LJ, Coutinho-Budd JC, Broihier HT. Divergent signaling requirements of dSARM in injury-induced degeneration and developmental glial phagocytosis. PLoS Genet 2022; 18:e1010257. [PMID: 35737721 PMCID: PMC9223396 DOI: 10.1371/journal.pgen.1010257] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 05/13/2022] [Indexed: 02/06/2023] Open
Abstract
Elucidating signal transduction mechanisms of innate immune pathways is essential to defining how they elicit distinct cellular responses. Toll-like receptors (TLR) signal through their cytoplasmic TIR domains which bind other TIR domain-containing adaptors. dSARM/SARM1 is one such TIR domain adaptor best known for its role as the central axon degeneration trigger after injury. In degeneration, SARM1's domains have been assigned unique functions: the ARM domain is auto-inhibitory, SAM-SAM domain interactions mediate multimerization, and the TIR domain has intrinsic NAD+ hydrolase activity that precipitates axonal demise. Whether and how these distinct functions contribute to TLR signaling is unknown. Here we show divergent signaling requirements for dSARM in injury-induced axon degeneration and TLR-mediated developmental glial phagocytosis through analysis of new knock-in domain and point mutations. We demonstrate intragenic complementation between reciprocal pairs of domain mutants during development, providing evidence for separability of dSARM functional domains in TLR signaling. Surprisingly, dSARM's NAD+ hydrolase activity is strictly required for both degenerative and developmental signaling, demonstrating that TLR signal transduction requires dSARM's enzymatic activity. In contrast, while SAM domain-mediated dSARM multimerization is important for axon degeneration, it is dispensable for TLR signaling. Finally, dSARM functions in a linear genetic pathway with the MAP3K Ask1 during development but not in degenerating axons. Thus, we propose that dSARM exists in distinct signaling states in developmental and pathological contexts.
Collapse
Affiliation(s)
- Kelsey A. Herrmann
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Yizhou Liu
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Arnau Llobet-Rosell
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Colleen N. McLaughlin
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Lukas J. Neukomm
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Jaeda C. Coutinho-Budd
- Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Heather T. Broihier
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
- * E-mail:
| |
Collapse
|
41
|
Brace EJ, Essuman K, Mao X, Palucki J, Sasaki Y, Milbrandt J, DiAntonio A. Distinct developmental and degenerative functions of SARM1 require NAD+ hydrolase activity. PLoS Genet 2022; 18:e1010246. [PMID: 35737728 PMCID: PMC9223315 DOI: 10.1371/journal.pgen.1010246] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 05/10/2022] [Indexed: 11/25/2022] Open
Abstract
SARM1 is the founding member of the TIR-domain family of NAD+ hydrolases and the central executioner of pathological axon degeneration. SARM1-dependent degeneration requires NAD+ hydrolysis. Prior to the discovery that SARM1 is an enzyme, SARM1 was studied as a TIR-domain adaptor protein with non-degenerative signaling roles in innate immunity and invertebrate neurodevelopment, including at the Drosophila neuromuscular junction (NMJ). Here we explore whether the NADase activity of SARM1 also contributes to developmental signaling. We developed transgenic Drosophila lines that express SARM1 variants with normal, deficient, and enhanced NADase activity and tested their function in NMJ development. We find that NMJ overgrowth scales with the amount of NADase activity, suggesting an instructive role for NAD+ hydrolysis in this developmental signaling pathway. While degenerative and developmental SARM1 signaling share a requirement for NAD+ hydrolysis, we demonstrate that these signals use distinct upstream and downstream mechanisms. These results identify SARM1-dependent NAD+ hydrolysis as a heretofore unappreciated component of developmental signaling. SARM1 now joins sirtuins and Parps as enzymes that regulate signal transduction pathways via mechanisms that involve NAD+ cleavage, greatly expanding the potential scope of SARM1 TIR NADase functions. SARM1 is the central executioner of axon loss, and inhibition of SARM1 is a therapeutic target for many devastating neurodegenerative disorders. SARM1 is the founding member of the TIR-domain family of NAD+ cleaving enzymes, destroying the essential metabolite NAD+ and inducing an energetic crisis in the axon. This was a surprising finding, as previously studied TIR-domain proteins were characterized as scaffolds that bind signaling proteins to coordinate signal transduction cascades. Indeed, before the discovery of the role of SARM1 in axon degeneration, SARM1 was studied as a regulator of intracellular signaling in immunity and neurodevelopment where it was assumed to act as a scaffold. Here we investigate whether the recently described SARM1 enzymatic activity also regulates such signal transduction pathways. Indeed, we show that a developmental signaling pathway scales with the amount of NADase activity, suggesting an instructive role for NAD+ cleavage. While degenerative and developmental SARM1 signaling share a requirement for NAD+ cleavage, they utilize distinct upstream and downstream mechanisms. With these findings, SARM1 now joins sirtuins and Parps as enzymes that regulate signal transduction pathways via mechanisms that involve NAD+ cleavage, greatly expanding the potential scope of SARM1 TIR NADase functions.
Collapse
Affiliation(s)
- E J Brace
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Kow Essuman
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Xianrong Mao
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - John Palucki
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Yo Sasaki
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Jeff Milbrandt
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America.,Needleman Center for Neurometabolism and Axonal Therapeutics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Aaron DiAntonio
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America.,Needleman Center for Neurometabolism and Axonal Therapeutics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| |
Collapse
|
42
|
Babetto E, Beirowski B. Of axons that struggle to make ends meet: Linking axonal bioenergetic failure to programmed axon degeneration. BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2022; 1863:148545. [PMID: 35339437 DOI: 10.1016/j.bbabio.2022.148545] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 03/07/2022] [Accepted: 03/16/2022] [Indexed: 02/07/2023]
Abstract
Axons are the long, fragile, and energy-hungry projections of neurons that are challenging to sustain. Together with their associated glia, they form the bulk of the neuronal network. Pathological axon degeneration (pAxD) is a driver of irreversible neurological disability in a host of neurodegenerative conditions. Halting pAxD is therefore an attractive therapeutic strategy. Here we review recent work demonstrating that pAxD is regulated by an auto-destruction program that revolves around axonal bioenergetics. We then focus on the emerging concept that axonal and glial energy metabolism are intertwined. We anticipate that these discoveries will encourage the pursuit of new treatment strategies for neurodegeneration.
Collapse
Affiliation(s)
- Elisabetta Babetto
- Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA; Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14214, USA.
| | - Bogdan Beirowski
- Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA; Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14214, USA.
| |
Collapse
|
43
|
Beirowski B. Emerging evidence for compromised axonal bioenergetics and axoglial metabolic coupling as drivers of neurodegeneration. Neurobiol Dis 2022; 170:105751. [PMID: 35569720 DOI: 10.1016/j.nbd.2022.105751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/20/2022] [Accepted: 05/09/2022] [Indexed: 10/18/2022] Open
Abstract
Impaired bioenergetic capacity of the nervous system is thought to contribute to the pathogenesis of many neurodegenerative diseases (NDD). Since neuronal synapses are believed to be the major energy consumers in the nervous system, synaptic derangements resulting from energy deficits have been suggested to play a central role for the development of many of these disorders. However, long axons constitute the largest compartment of the neuronal network, require large amounts of energy, are metabolically and structurally highly vulnerable, and undergo early injurious stresses in many NDD. These stresses likely impose additional energy demands for continuous adaptations and repair processes, and may eventually overwhelm axonal maintenance mechanisms. Indeed, pathological axon degeneration (pAxD) is now recognized as an etiological focus in a wide array of NDD associated with bioenergetic abnormalities. In this paper I first discuss the recognition that a simple experimental model for pAxD is regulated by an auto-destruction program that exhausts distressed axons energetically. Provision of the energy substrate pyruvate robustly counteracts this axonal breakdown. Importantly, energy decline in axons is not only a consequence but also an initiator of this program. This opens the intriguing possibility that axon dysfunction and pAxD can be suppressed by preemptively energizing distressed axons. Second, I focus on the emerging concept that axons communicate energetically with their flanking glia. This axoglial metabolic coupling can help offset the axonal energy decline that activates the pAxD program but also jeopardize axon integrity as a result of perturbed glial metabolism. Third, I present compelling evidence that abnormal axonal energetics and compromised axoglial metabolic coupling accompany the activation of the pAxD auto-destruction pathway in models of glaucoma, a widespread neurodegenerative condition with pathogenic overlap to other common NDD. In conclusion, I propose a novel conceptual framework suggesting that therapeutic interventions focused on bioenergetic support of the nervous system should also address axons and their metabolic interactions with glia.
Collapse
Affiliation(s)
- Bogdan Beirowski
- Institute for Myelin and Glia Exploration, New York State Center of Excellence in Bioinformatics & Life Sciences (CBLS), University at Buffalo, Buffalo, NY 14203, USA; Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14214, USA.
| |
Collapse
|
44
|
Cao Y, Wang Y, Yang J. NAD +-dependent mechanism of pathological axon degeneration. CELL INSIGHT 2022; 1:100019. [PMID: 37193131 PMCID: PMC10120281 DOI: 10.1016/j.cellin.2022.100019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 05/18/2023]
Abstract
Pathological axon degeneration is broadly observed in neurodegenerative diseases. This unique process of axonal pathology could directly interfere with the normal functions of neurocircuitries and contribute to the onset of clinical symptoms in patients. It has been increasingly recognized that functional preservation of axonal structures is an indispensable part of therapeutic strategies for treating neurological disorders. In the past decades, the research field has witnessed significant breakthroughs in understanding the stereotyped self-destruction of axons upon neurodegenerative insults, which is distinct from all the known types of programmed cell death. In particular, the novel NAD+-dependent mechanism involving the WLDs, NMNAT2, and SARM1 proteins has emerged. This review summarizes the landmark discoveries elucidating the molecular pathway of pathological axon degeneration and highlights the evolving concept that neurodegeneration would be intrinsically linked to NAD+ and energy metabolism.
Collapse
Affiliation(s)
- Ying Cao
- Center for Life Sciences, Peking University, Beijing, 100871, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Yi Wang
- Center for Life Sciences, Peking University, Beijing, 100871, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Jing Yang
- Center for Life Sciences, Peking University, Beijing, 100871, China
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, 100871, China
- IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
- Chinese Institute for Brain Research, Beijing, 102206, China
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, 518055, China
| |
Collapse
|
45
|
Knobel DL, Jackson AC, Bingham J, Ertl HCJ, Gibson AD, Hughes D, Joubert K, Mani RS, Mohr BJ, Moore SM, Rivett-Carnac H, Tordo N, Yeates JW, Zambelli AB, Rupprecht CE. A One Medicine Mission for an Effective Rabies Therapy. Front Vet Sci 2022; 9:867382. [PMID: 35372555 PMCID: PMC8967983 DOI: 10.3389/fvets.2022.867382] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
Despite the disease's long history, little progress has been made toward a treatment for rabies. The prognosis for patient recovery remains dire. For any prospect of survival, patients require aggressive critical care, which physicians in rabies endemic areas may be reluctant or unable to provide given the cost, clinical expertise required, and uncertain outcome. Systematic clinical research into combination therapies is further hampered by sporadic occurrence of cases. In this Perspective, we examine the case for a One Medicine approach to accelerate development of an effective therapy for rabies through the veterinary care and investigational treatment of naturally infected dogs in appropriate circumstances. We review the pathogenesis of rabies virus in humans and dogs, including recent advances in our understanding of the molecular basis for the severe neurological dysfunction. We propose that four categories of disease process need to be managed in patients: viral propagation, neuronal degeneration, inflammation and systemic compromise. Compassionate critical care and investigational treatment of naturally infected dogs receiving supportive therapy that mimics the human clinical scenario could increase opportunities to study combination therapies that address these processes, and to identify biomarkers for prognosis and therapeutic response. We discuss the safety and ethics of this approach, and introduce the Canine Rabies Treatment Initiative, a non-profit organization with the mission to apply a One Medicine approach to the investigation of diagnostic, prognostic, and therapeutic options for rabies in naturally infected dogs, to accelerate transformation of rabies into a treatable disease for all patients.
Collapse
Affiliation(s)
- Darryn L. Knobel
- Department of Biomedical Sciences, Ross University School of Veterinary Medicine, Basseterre, Saint Kitts and Nevis
- Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Pretoria, South Africa
- Canine Rabies Treatment Initiative, Salt Rock, South Africa
| | - Alan C. Jackson
- Department of Medicine, Northern Consultation Centre, Thompson General Hospital, Thompson, MB, Canada
- Department of Medicine, Lake of the Woods District Hospital, Kenora, ON, Canada
| | - John Bingham
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Australian Animal Health Laboratory at the Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | | | - Andrew D. Gibson
- Division of Genetics and Genomics, Easter Bush Veterinary Centre, The Roslin Institute and the Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Roslin, United Kingdom
| | - Daniela Hughes
- Canine Rabies Treatment Initiative, Salt Rock, South Africa
| | - Kenneth Joubert
- Veterinary Anaesthesia, Analgesia and Critical Care Services, Lonehill, South Africa
| | - Reeta S. Mani
- Department of Neurovirology, WHO Collaborating Centre for Reference and Research in Rabies, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Bert J. Mohr
- Canine Rabies Treatment Initiative, Salt Rock, South Africa
- Centre for Animal Research, Faculty of Health Sciences, University of Cape Town, Observatory, South Africa
| | - Susan M. Moore
- Veterinary Medical Diagnostic Laboratory, University of Missouri, Columbia, MO, United States
| | | | - Noël Tordo
- Institut Pasteur de Guinée, Conakry, Guinea
| | | | | | | |
Collapse
|
46
|
Bloom AJ, Mao X, Strickland A, Sasaki Y, Milbrandt J, DiAntonio A. Constitutively active SARM1 variants that induce neuropathy are enriched in ALS patients. Mol Neurodegener 2022; 17:1. [PMID: 34991663 PMCID: PMC8739729 DOI: 10.1186/s13024-021-00511-x] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 12/17/2021] [Indexed: 03/31/2023] Open
Abstract
Background In response to injury, neurons activate a program of organized axon self-destruction initiated by the NAD+ hydrolase, SARM1. In healthy neurons SARM1 is autoinhibited, but single amino acid changes can abolish autoinhibition leading to constitutively active SARM1 enzymes that promote degeneration when expressed in cultured neurons. Methods To investigate whether naturally occurring human variants might disrupt SARM1 autoinhibition and potentially contribute to risk for neurodegenerative disease, we assayed the enzymatic activity of all 42 rare SARM1 alleles identified among 8507 amyotrophic lateral sclerosis (ALS) patients and 9671 controls. We then intrathecally injected mice with virus expressing SARM1 constructs to test the capacity of an ALS-associated constitutively active SARM1 variant to promote neurodegeneration in vivo. Results Twelve out of 42 SARM1 missense variants or small in-frame deletions assayed exhibit constitutive NADase activity, including more than half of those that are unique to the ALS patients or that occur in multiple patients. There is a > 5-fold enrichment of constitutively active variants among patients compared to controls. Expression of constitutively active ALS-associated SARM1 alleles in cultured dorsal root ganglion (DRG) neurons is pro-degenerative and cytotoxic. Intrathecal injection of an AAV expressing the common SARM1 reference allele is innocuous to mice, but a construct harboring SARM1V184G, the constitutively active variant found most frequently among the ALS patients, causes axon loss, motor dysfunction, and sustained neuroinflammation. Conclusions These results implicate rare hypermorphic SARM1 alleles as candidate genetic risk factors for ALS and other neurodegenerative conditions. Supplementary Information The online version contains supplementary material available at 10.1186/s13024-021-00511-x.
Collapse
Affiliation(s)
- A Joseph Bloom
- Needleman Center for Neurometabolism and Axonal Therapeutics and Department of Genetics, Washington University School of Medicine in Saint Louis, St. Louis, MO, USA.
| | - Xianrong Mao
- Needleman Center for Neurometabolism and Axonal Therapeutics and Department of Genetics, Washington University School of Medicine in Saint Louis, St. Louis, MO, USA
| | - Amy Strickland
- Needleman Center for Neurometabolism and Axonal Therapeutics and Department of Genetics, Washington University School of Medicine in Saint Louis, St. Louis, MO, USA
| | - Yo Sasaki
- Needleman Center for Neurometabolism and Axonal Therapeutics and Department of Genetics, Washington University School of Medicine in Saint Louis, St. Louis, MO, USA
| | - Jeffrey Milbrandt
- Needleman Center for Neurometabolism and Axonal Therapeutics and Department of Genetics, Washington University School of Medicine in Saint Louis, St. Louis, MO, USA.
| | - Aaron DiAntonio
- Needleman Center for Neurometabolism and Axonal Therapeutics and Department of Developmental Biology, Washington University School of Medicine in Saint Louis, St. Louis, MO, USA.
| |
Collapse
|