1
|
Zhang P, Xu Z. The advancements in precision medicine for Leber congenital amaurosis: Breakthroughs from genetic diagnosis to therapy. Surv Ophthalmol 2025:S0039-6257(25)00070-0. [PMID: 40311816 DOI: 10.1016/j.survophthal.2025.04.005] [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: 12/17/2024] [Revised: 04/15/2025] [Accepted: 04/28/2025] [Indexed: 05/03/2025]
Abstract
Leber congenital amaurosis (LCA) is a hereditary retinal disease, typically manifesting as severe vision impairment in infancy. With the advancement of precision medicine, genetic diagnosis and targeted therapies offer new hope for LCA patients, significantly improving both diagnostic accuracy and therapeutic efficacy. We summarize the epidemiological characteristics, clinical manifestations, and molecular genetics underlying LCA. It also highlights recent developments in precision treatment strategies, including gene replacement therapy, CRISPR/Cas9-mediated gene editing, and antisense oligonucleotide therapies. In addition, we discuss the applications of induced pluripotent stem cells and retinal organoids in LCA treatment research. Furthermore, we explore preventive strategies and future treatment directions for LCA, including the development of novel gene therapy vectors, the optimization of combinatorial treatment strategies, and the formulation of personalized treatment approaches. These advancements hold significant potential to offer improved treatment options and enhance the quality of life for LCA patients.
Collapse
Affiliation(s)
- Pei Zhang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhuping Xu
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
2
|
Zhu S, Zhang L, Tong P, Chen J, Wang C, Wang Z, Liu J, Duan P, Jiang Q, Zhou Y, Tan G, Zhang X, Jiang B. Nicotinamide Riboside Mitigates Retinal Degeneration by Suppressing Damaged DNA-Stimulated Microglial Activation and STING-Mediated Pyroptosis. Invest Ophthalmol Vis Sci 2025; 66:14. [PMID: 40192637 PMCID: PMC11980955 DOI: 10.1167/iovs.66.4.14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 03/10/2025] [Indexed: 04/11/2025] Open
Abstract
Purpose Microglial activation plays a pivotal role in the pathogenesis of retinal degeneration, contributing to neuroinflammation within the retina. Previous studies identified that nicotinamide riboside (NR) mitigated light-induced retinal degeneration (LIRD) and inhibited microglial activation. The cGAS-STING signaling pathway has been recognized as a key mediator of inflammation in response to cellular stress and tissue damage. This study further explores the regulatory impact of NR on microglial activation and STING-mediated pyroptosis in retinal degeneration. Methods Balb/c mice were subjected to bright light exposure to induce retinal degeneration. Bioinformatics analysis was used to identify the upregulated key genes and signaling pathways involved in the progression of retinal degeneration, based on mouse transcriptomes from the LIRD model. Molecular biology techniques and immunofluorescence staining were used to assess cGAS-STING activation and expression of pyroptosis-associated molecules. Retinal function, photoreceptor apoptosis and inflammatory response were evaluated in the presence and absence of NR supplementation. Results Exposure to bright light resulted in mitochondrial dysfunction and the release of dsDNA, significantly triggering the activation of cGAS-STING pathway and microglial pyroptosis. In contrast, NR treatment preserved mitochondrial biosynthesis, inhibited STING expression in reactive microglia, and dampened the pro-inflammatory response. Additionally, intraperitoneal administration of the STING inhibitor H151 reduced light-induced microglial activation and pyroptosis, while improving retinal function and promoting photoreceptor survival. Conclusions These findings suggest that NR confers neuroprotection by attenuating damaged DNA-triggered STING-mediated microglial activation and pyroptosis. Targeting the cGAS-STING pathway presents a promising therapeutic avenue for retinal degeneration.
Collapse
Affiliation(s)
- Shanshan Zhu
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, China
| | - Lusi Zhang
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, China
| | - Ping Tong
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, China
| | - Jiawei Chen
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, China
| | - Cong Wang
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, China
| | - Zewei Wang
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, China
| | - Jingyuan Liu
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, China
| | - Peiyun Duan
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, China
| | - Qian Jiang
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, China
| | - Yubing Zhou
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, China
| | - Guangshuang Tan
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, China
| | - Xian Zhang
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, China
| | - Bing Jiang
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, China
| |
Collapse
|
3
|
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
|
4
|
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
|
5
|
Cvekl A, Vijg J. Aging of the eye: Lessons from cataracts and age-related macular degeneration. Ageing Res Rev 2024; 99:102407. [PMID: 38977082 DOI: 10.1016/j.arr.2024.102407] [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: 03/11/2024] [Revised: 06/18/2024] [Accepted: 07/01/2024] [Indexed: 07/10/2024]
Abstract
Aging is the greatest risk factor for chronic human diseases, including many eye diseases. Geroscience aims to understand the effects of the aging process on these diseases, including the genetic, molecular, and cellular mechanisms that underlie the increased risk of disease over the lifetime. Understanding of the aging eye increases general knowledge of the cellular physiology impacted by aging processes at various biological extremes. Two major diseases, age-related cataract and age-related macular degeneration (AMD) are caused by dysfunction of the lens and retina, respectively. Lens transparency and light refraction are mediated by lens fiber cells lacking nuclei and other organelles, which provides a unique opportunity to study a single aging hallmark, i.e., loss of proteostasis, within an environment of limited metabolism. In AMD, local dysfunction of the photoreceptors/retinal pigmented epithelium/Bruch's membrane/choriocapillaris complex in the macula leads to the loss of photoreceptors and eventually loss of central vision, and is driven by nearly all the hallmarks of aging and shares features with Alzheimer's disease, Parkinson's disease, cardiovascular disease, and diabetes. The aging eye can function as a model for studying basic mechanisms of aging and, vice versa, well-defined hallmarks of aging can be used as tools to understand age-related eye disease.
Collapse
Affiliation(s)
- Ales Cvekl
- Departments of Genetics and Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Jan Vijg
- Departments of Genetics and Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
6
|
Loreto A, Merlini E, Coleman MP. Programmed axon death: a promising target for treating retinal and optic nerve disorders. Eye (Lond) 2024; 38:1802-1809. [PMID: 38538779 PMCID: PMC11226669 DOI: 10.1038/s41433-024-03025-0] [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: 09/04/2023] [Revised: 02/13/2024] [Accepted: 03/08/2024] [Indexed: 07/07/2024] Open
Abstract
Programmed axon death is a druggable pathway of axon degeneration that has garnered considerable interest from pharmaceutical companies as a promising therapeutic target for various neurodegenerative disorders. In this review, we highlight mechanisms through which this pathway is activated in the retina and optic nerve, and discuss its potential significance for developing therapies for eye disorders and beyond. At the core of programmed axon death are two enzymes, NMNAT2 and SARM1, with pivotal roles in NAD metabolism. Extensive preclinical data in disease models consistently demonstrate remarkable, and in some instances, complete and enduring neuroprotection when this mechanism is targeted. Findings from animal studies are now being substantiated by genetic human data, propelling the field rapidly toward clinical translation. As we approach the clinical phase, the selection of suitable disorders for initial clinical trials targeting programmed axon death becomes crucial for their success. We delve into the multifaceted roles of programmed axon death and NAD metabolism in retinal and optic nerve disorders. We discuss the role of SARM1 beyond axon degeneration, including its potential involvement in neuronal soma death and photoreceptor degeneration. We also discuss genetic human data and environmental triggers of programmed axon death. Lastly, we touch upon potential therapeutic approaches targeting NMNATs and SARM1, as well as the nicotinamide trials for glaucoma. The extensive literature linking programmed axon death to eye disorders, along with the eye's suitability for drug delivery and visual assessments, makes retinal and optic nerve disorders strong contenders for early clinical trials targeting programmed axon death.
Collapse
Affiliation(s)
- Andrea Loreto
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Forvie Site, Robinson Way, Cambridge, UK.
- School of Medical Sciences and Save Sight Institute, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
| | - Elisa Merlini
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Forvie Site, Robinson Way, Cambridge, UK
| | - Michael P Coleman
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Forvie Site, Robinson Way, Cambridge, UK.
| |
Collapse
|
7
|
Miao X, Wu Q, Du S, Xiang L, Zhou S, Zhu J, Chen Z, Wang H, Pan X, Fan Y, Zhang L, Qian J, Xing Y, Xie Y, Hu L, Xu H, Wang W, Wang Y, Huang Z. SARM1 Promotes Neurodegeneration and Memory Impairment in Mouse Models of Alzheimer's Disease. Aging Dis 2024; 15:390-407. [PMID: 37307837 PMCID: PMC10796105 DOI: 10.14336/ad.2023.0516-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 05/16/2023] [Indexed: 06/14/2023] Open
Abstract
Neuroinflammation plays a crucial role in the pathogenesis and progression of Alzheimer's disease (AD). The Sterile Alpha and Toll Interleukin Receptor Motif-containing protein 1 (SARM1) has been shown to promote axonal degeneration and is involved in neuroinflammation. However, the role of SARM1 in AD remains unclear. In this study, we found that SARM1 was reduced in hippocampal neurons of AD model mice. Interestingly, conditional knockout (CKO) of SARM1 in the central nervous system (CNS, SARM1Nestin-CKO mice) delayed the cognitive decline in APP/PS1 AD model mice. Furthermore, SARM1 deletion reduced the Aβ deposition and inflammatory infiltration in the hippocampus and inhibited neurodegeneration in APP/PS1 AD model mice. Further investigation into the underlying mechanisms revealed that the signaling of tumor necrosis factor-α (TNF-α) was downregulated in the hippocampus tissues of APP/PS1;SARM1Nestin-CKO mice, thereby alleviating the cognitive decline, Aβ deposition and inflammatory infiltration. These findings identify unrecognized functions of SARM1 in promoting AD and reveal the SARM1-TNF-α pathway in AD model mice.
Collapse
Affiliation(s)
- Xuemeng Miao
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China.
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Qian Wu
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Siyu Du
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Ludan Xiang
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Siyao Zhou
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Junzhe Zhu
- School of the First Clinical Medical Sciences, School of Information and Engineering, Wenzhou Medical University, Wenzhou, Zhejiang 325205, China.
| | - Zirun Chen
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Hui Wang
- School of the First Clinical Medical Sciences, School of Information and Engineering, Wenzhou Medical University, Wenzhou, Zhejiang 325205, China.
| | - Xuyi Pan
- School of the First Clinical Medical Sciences, School of Information and Engineering, Wenzhou Medical University, Wenzhou, Zhejiang 325205, China.
| | - Yiren Fan
- School of the First Clinical Medical Sciences, School of Information and Engineering, Wenzhou Medical University, Wenzhou, Zhejiang 325205, China.
| | - Lihan Zhang
- School of the First Clinical Medical Sciences, School of Information and Engineering, Wenzhou Medical University, Wenzhou, Zhejiang 325205, China.
| | - Jingkang Qian
- School of the First Clinical Medical Sciences, School of Information and Engineering, Wenzhou Medical University, Wenzhou, Zhejiang 325205, China.
| | - Yuxuan Xing
- School of the First Clinical Medical Sciences, School of Information and Engineering, Wenzhou Medical University, Wenzhou, Zhejiang 325205, China.
| | - Yiyang Xie
- School of the First Clinical Medical Sciences, School of Information and Engineering, Wenzhou Medical University, Wenzhou, Zhejiang 325205, China.
| | - Lixin Hu
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Haiyun Xu
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Wei Wang
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Ying Wang
- Clinical Research Center, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China.
| | - Zhihui Huang
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China.
- School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| |
Collapse
|
8
|
Yang Y, Jiang X, Chen J, Liu L, Liu G, Sun K, Liu W, Zhu X, Guan Q. The m 6A reader YTHDC2 maintains visual function and retinal photoreceptor survival through modulating translation of PPEF2 and PDE6B. J Genet Genomics 2024; 51:208-221. [PMID: 38157933 DOI: 10.1016/j.jgg.2023.12.007] [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: 08/30/2023] [Revised: 12/22/2023] [Accepted: 12/22/2023] [Indexed: 01/03/2024]
Abstract
Inherited retinal dystrophies (IRDs) are major causes of visual impairment and irreversible blindness worldwide, while the precise molecular and genetic mechanisms are still elusive. N6-methyladenosine (m6A) modification is the most prevalent internal modification in eukaryotic mRNA. YTH domain containing 2 (YTHDC2), an m6A reader protein, has recently been identified as a key player in germline development and human cancer. However, its contribution to retinal function remains unknown. Here, we explore the role of YTHDC2 in the visual function of retinal rod photoreceptors by generating rod-specific Ythdc2 knockout mice. Results show that Ythdc2 deficiency in rods causes diminished scotopic ERG responses and progressive retinal degeneration. Multi-omics analysis further identifies Ppef2 and Pde6b as the potential targets of YTHDC2 in the retina. Specifically, via its YTH domain, YTHDC2 recognizes and binds m6A-modified Ppef2 mRNA at the coding sequence and Pde6b mRNA at the 5'-UTR, resulting in enhanced translation efficiency without affecting mRNA levels. Compromised translation efficiency of Ppef2 and Pde6b after YTHDC2 depletion ultimately leads to decreased protein levels in the retina, impaired retinal function, and progressive rod death. Collectively, our finding highlights the importance of YTHDC2 in visual function and photoreceptor survival, which provides an unreported elucidation of IRD pathogenesis via epitranscriptomics.
Collapse
Affiliation(s)
- Yeming Yang
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Xiaoyan Jiang
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Junyao Chen
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Lu Liu
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Guo Liu
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Kuanxiang Sun
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Wenjing Liu
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Xianjun Zhu
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China; Department of Geriatrics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China; Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, China; Qinghai Key Laboratory of Qinghai Tibet Plateau Biological Resources, Chinese Academy of Sciences and Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Xining, Qinghai 810008, China; Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China.
| | - Qiuyue Guan
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China; Department of Geriatrics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.
| |
Collapse
|
9
|
Lee TJ, Sasaki Y, Ruzycki PA, Ban N, Lin JB, Wu HT, Santeford A, Apte RS. Catalytic isoforms of AMP-activated protein kinase differentially regulate IMPDH activity and photoreceptor neuron function. JCI Insight 2024; 9:e173707. [PMID: 38227383 PMCID: PMC11143937 DOI: 10.1172/jci.insight.173707] [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: 07/10/2023] [Accepted: 01/10/2024] [Indexed: 01/17/2024] Open
Abstract
AMP-activated protein kinase (AMPK) plays a crucial role in maintaining ATP homeostasis in photoreceptor neurons. AMPK is a heterotrimeric protein consisting of α, β, and γ subunits. The independent functions of the 2 isoforms of the catalytic α subunit, PRKAA1 and PRKAA2, are uncharacterized in specialized neurons, such as photoreceptors. Here, we demonstrate in mice that rod photoreceptors lacking PRKAA2, but not PRKAA1, showed altered levels of cGMP, GTP, and ATP, suggesting isoform-specific regulation of photoreceptor metabolism. Furthermore, PRKAA2-deficient mice displayed visual functional deficits on electroretinography and photoreceptor outer segment structural abnormalities on transmission electron microscopy consistent with neuronal dysfunction, but not neurodegeneration. Phosphoproteomics identified inosine monophosphate dehydrogenase (IMPDH) as a molecular driver of PRKAA2-specific photoreceptor dysfunction, and inhibition of IMPDH improved visual function in Prkaa2 rod photoreceptor-knockout mice. These findings highlight a therapeutically targetable PRKAA2 isoform-specific function of AMPK in regulating photoreceptor metabolism and function through a potentially previously uncharacterized mechanism affecting IMPDH activity.
Collapse
Affiliation(s)
- Tae Jun Lee
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences
- Department of Developmental Biology; and
| | - Yo Sasaki
- Department of Genetics, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Philip A. Ruzycki
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences
- Department of Genetics, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Norimitsu Ban
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Joseph B. Lin
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences
| | | | - Andrea Santeford
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences
| | - Rajendra S. Apte
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences
- Department of Developmental Biology; and
- Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
10
|
Chiarugi A. Glaucoma: neuroprotection with NAD-based therapeutic interventions. Trends Pharmacol Sci 2023; 44:869-879. [PMID: 37880000 DOI: 10.1016/j.tips.2023.09.010] [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: 09/06/2023] [Revised: 09/22/2023] [Accepted: 09/26/2023] [Indexed: 10/27/2023]
Abstract
Clinical evidence shows that intraocular hypertension is not the primary pathogenetic event of glaucoma, whereas early neurodegeneration of retinal ganglion cells (RGCs) represents a key therapeutic target. Unfortunately, failure of clinical trials with neuroprotective agents, in particular those testing the anti-excitotoxic drug memantine, generated widespread skepticism regarding the possibility of counteracting neurodegeneration during glaucoma. New avenues for neuroprotective approaches to counteract glaucoma evolution have been opened by the identification of a programmed axonal degeneration (PAD) program triggered by increased nicotinamide mononucleotide (NMN)/NAD concentration ratio. Positive results of proof-of-concept clinical studies based on sustaining axonal NAD homeostasis facilitated the design of Phase 2/3 trials. Here, I share my opinion on how neurodegeneration in glaucoma should be put into context, together with an appraisal of the pharmacological rationale of NAD-supporting therapies for use during glaucoma progression.
Collapse
Affiliation(s)
- Alberto Chiarugi
- Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy; Headache Center and Clinical Pharmacology Unit, Careggi University Hospital, Florence, Italy.
| |
Collapse
|
11
|
Loreto A, Antoniou C, Merlini E, Gilley J, Coleman MP. NMN: The NAD precursor at the intersection between axon degeneration and anti-ageing therapies. Neurosci Res 2023; 197:18-24. [PMID: 36657725 DOI: 10.1016/j.neures.2023.01.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/18/2023]
Abstract
The past 20 years of research on axon degeneration has revealed fine details on how NAD biology controls axonal survival. Extensive data demonstrate that the NAD precursor NMN binds to and activates the pro-degenerative enzyme SARM1, so a failure to convert sufficient NMN into NAD leads to toxic NMN accumulation and axon degeneration. This involvement of NMN brings the axon degeneration field to an unexpected overlap with research into ageing and extending healthy lifespan. A decline in NAD levels throughout life, at least in some tissues, is believed to contribute to age-related functional decay and boosting NAD production with supplementation of NMN or other NAD precursors has gained attention as a potential anti-ageing therapy. Recent years have witnessed an influx of NMN-based products and related molecules on the market, sold as food supplements, with many people taking these supplements daily. While several clinical trials are ongoing to check the safety profiles and efficacy of NAD precursors, sufficient data to back their therapeutic use are still lacking. Here, we discuss NMN supplementation, SARM1 and anti-ageing strategies, with an important question in mind: considering that NMN accumulation can lead to axon degeneration, how is this compatible with its beneficial effect in ageing and are there circumstances in which NMN supplementation could become harmful?
Collapse
Affiliation(s)
- Andrea Loreto
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Forvie Site, Robinson Way, CB2 0PY Cambridge, UK.
| | - Christina Antoniou
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Forvie Site, Robinson Way, CB2 0PY Cambridge, UK
| | - Elisa Merlini
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Forvie Site, Robinson Way, CB2 0PY Cambridge, UK
| | - Jonathan Gilley
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Forvie Site, Robinson Way, CB2 0PY Cambridge, UK
| | - Michael P Coleman
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Forvie Site, Robinson Way, CB2 0PY Cambridge, UK.
| |
Collapse
|
12
|
Montoro-Gámez C, Nolte H, Molinié T, Evangelista G, Tröder SE, Barth E, Popovic M, Trifunovic A, Zevnik B, Langer T, Rugarli EI. SARM1 deletion delays cerebellar but not spinal cord degeneration in an enhanced mouse model of SPG7 deficiency. Brain 2023; 146:4117-4131. [PMID: 37086482 DOI: 10.1093/brain/awad136] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 03/16/2023] [Accepted: 04/10/2023] [Indexed: 04/24/2023] Open
Abstract
Hereditary spastic paraplegia is a neurological condition characterized by predominant axonal degeneration in long spinal tracts, leading to weakness and spasticity in the lower limbs. The nicotinamide adenine dinucleotide (NAD+)-consuming enzyme SARM1 has emerged as a key executioner of axonal degeneration upon nerve transection and in some neuropathies. An increase in the nicotinamide mononucleotide/NAD+ ratio activates SARM1, causing catastrophic NAD+ depletion and axonal degeneration. However, the role of SARM1 in the pathogenesis of hereditary spastic paraplegia has not been investigated. Here, we report an enhanced mouse model for hereditary spastic paraplegia caused by mutations in SPG7. The eSpg7 knockout mouse carries a deletion in both Spg7 and Afg3l1, a redundant homologue expressed in mice but not in humans. The eSpg7 knockout mice recapitulate the phenotypic features of human patients, showing progressive symptoms of spastic-ataxia and degeneration of axons in the spinal cord as well as the cerebellum. We show that the lack of SPG7 rewires the mitochondrial proteome in both tissues, leading to an early onset decrease in mito-ribosomal subunits and a remodelling of mitochondrial solute carriers and transporters. To interrogate mechanisms leading to axonal degeneration in this mouse model, we explored the involvement of SARM1. Deletion of SARM1 delays the appearance of ataxic signs, rescues mitochondrial swelling and axonal degeneration of cerebellar granule cells and dampens neuroinflammation in the cerebellum. The loss of SARM1 also prevents endoplasmic reticulum abnormalities in long spinal cord axons, but does not halt the degeneration of these axons. Our data thus reveal a neuron-specific interplay between SARM1 and mitochondrial dysfunction caused by lack of SPG7 in hereditary spastic paraplegia.
Collapse
Affiliation(s)
- Carolina Montoro-Gámez
- Institute for Genetics, University of Cologne, Cologne 50931, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne 50931, Germany
| | - Hendrik Nolte
- Max Planck Institute for Biology of Ageing, Cologne 50931, Germany
| | - Thibaut Molinié
- Institute for Genetics, University of Cologne, Cologne 50931, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne 50931, Germany
| | - Giovanna Evangelista
- Institute for Genetics, University of Cologne, Cologne 50931, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne 50931, Germany
| | - Simon E Tröder
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne 50931, Germany
- in vivo Research Facility, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne 50931, Germany
| | - Esther Barth
- Institute for Genetics, University of Cologne, Cologne 50931, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne 50931, Germany
| | - Milica Popovic
- Institute for Genetics, University of Cologne, Cologne 50931, Germany
- Institute for Mitochondrial Diseases and Aging, Medical Faculty, University of Cologne, Cologne 50931, Germany
| | - Aleksandra Trifunovic
- Institute for Genetics, University of Cologne, Cologne 50931, Germany
- Institute for Mitochondrial Diseases and Aging, Medical Faculty, University of Cologne, Cologne 50931, Germany
- Center for Molecular Medicine (CMMC), University of Cologne, Cologne 50931, Germany
| | - Branko Zevnik
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne 50931, Germany
- in vivo Research Facility, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne 50931, Germany
| | - Thomas Langer
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne 50931, Germany
- Max Planck Institute for Biology of Ageing, Cologne 50931, Germany
| | - Elena I Rugarli
- Institute for Genetics, University of Cologne, Cologne 50931, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne 50931, Germany
- Center for Molecular Medicine (CMMC), University of Cologne, Cologne 50931, Germany
| |
Collapse
|
13
|
Liu S, Zhang W. NAD + metabolism and eye diseases: current status and future directions. Mol Biol Rep 2023; 50:8653-8663. [PMID: 37540459 DOI: 10.1007/s11033-023-08692-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: 05/16/2023] [Accepted: 07/18/2023] [Indexed: 08/05/2023]
Abstract
Currently, there are no truly effective treatments for a variety of eye diseases, such as glaucoma, age-related macular degeneration (AMD), and inherited retinal degenerations (IRDs). These conditions have a significant impact on patients' quality of life and can be a burden on society. However, these diseases share a common pathological process of NAD+ metabolism disorders. They are either associated with genetically induced primary NAD+ synthase deficiency, decreased NAD+ levels due to aging, or enhanced NAD+ consuming enzyme activity during disease pathology. In this discussion, we explore the role of NAD+ metabolic disorders in the development of associated ocular diseases and the potential advantages and disadvantages of various methods to increase NAD+ levels. It is essential to carefully evaluate the possible adverse effects of these methods and conduct a more comprehensive and objective assessment of their function before considering their use.
Collapse
Affiliation(s)
- Siyuan Liu
- Department of Ophthalmology, Second Clinical Medical College, Lanzhou University, 730030, Lanzhou, VA, China
| | - Wenfang Zhang
- Department of Ophthalmology, The Second Hospital of Lanzhou University, 730030, Lanzhou, VA, China.
| |
Collapse
|
14
|
Xiang L, Yang QL, Xie BT, Zeng HY, Ding LJ, Rao FQ, Yan T, Lu F, Chen Q, Huang XF. Dysregulated Arginine Metabolism Is Linked to Retinal Degeneration in Cep250 Knockout Mice. Invest Ophthalmol Vis Sci 2023; 64:2. [PMID: 37656476 PMCID: PMC10479211 DOI: 10.1167/iovs.64.12.2] [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: 03/06/2023] [Accepted: 08/04/2023] [Indexed: 09/02/2023] Open
Abstract
Purpose Degeneration of retinal photoreceptors is frequently observed in diverse ciliopathy disorders, and photoreceptor cilium gates the molecular trafficking between the inner and the outer segment (OS). This study aims to generate a homozygous global Cep250 knockout (KO) mouse and study the resulting phenotype. Methods We used Cep250 KO mice and untargeted metabolomics to uncover potential mechanisms underlying retinal degeneration. Long-term follow-up studies using optical coherence tomography (OCT) and electroretinography (ERG) were performed. Results OCT and ERG results demonstrated gradual thinning of the outer nuclear layer (ONL) and progressive attenuation of the scotopic ERG responses in Cep250-/- mice. More TUNEL signal was observed in the ONL of these mice. Immunostaining of selected OS proteins revealed mislocalization of these proteins in the ONL of Cep250-/- mice. Interestingly, untargeted metabolomics analysis revealed arginine-related metabolic pathways were altered and enriched in Cep250-/- mice. Mis-localization of a key protein in the arginine metabolism pathway, arginase 1 (ARG1), in the ONL of KO mice further supports this model. Moreover, adeno-associated virus (AAV)-based retinal knockdown of Arg1 led to similar architectural and functional alterations in wild-type retinas. Conclusions Altogether, these results suggest that dysregulated arginine metabolism contributes to retinal degeneration in Cep250-/- mice. Our findings provide novel insights that increase understanding of retinal degeneration in ciliopathy disorders.
Collapse
Affiliation(s)
- Lue Xiang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qiao-Li Yang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Bin-Tao Xie
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hui-Yi Zeng
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Liu-Jun Ding
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Feng-Qin Rao
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- School of Pharmaceutical Sciences of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Tong Yan
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Fan Lu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qi Chen
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiu-Feng Huang
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
15
|
Tribble JR, Hui F, Quintero H, El Hajji S, Bell K, Di Polo A, Williams PA. Neuroprotection in glaucoma: Mechanisms beyond intraocular pressure lowering. Mol Aspects Med 2023; 92:101193. [PMID: 37331129 DOI: 10.1016/j.mam.2023.101193] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/25/2023] [Accepted: 06/04/2023] [Indexed: 06/20/2023]
Abstract
Glaucoma is a common, complex, multifactorial neurodegenerative disease characterized by progressive dysfunction and then loss of retinal ganglion cells, the output neurons of the retina. Glaucoma is the most common cause of irreversible blindness and affects ∼80 million people worldwide with many more undiagnosed. The major risk factors for glaucoma are genetics, age, and elevated intraocular pressure. Current strategies only target intraocular pressure management and do not directly target the neurodegenerative processes occurring at the level of the retinal ganglion cell. Despite strategies to manage intraocular pressure, as many as 40% of glaucoma patients progress to blindness in at least one eye during their lifetime. As such, neuroprotective strategies that target the retinal ganglion cell and these neurodegenerative processes directly are of great therapeutic need. This review will cover the recent advances from basic biology to on-going clinical trials for neuroprotection in glaucoma covering degenerative mechanisms, metabolism, insulin signaling, mTOR, axon transport, apoptosis, autophagy, and neuroinflammation. With an increased understanding of both the basic and clinical mechanisms of the disease, we are closer than ever to a neuroprotective strategy for glaucoma.
Collapse
Affiliation(s)
- James R Tribble
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Flora Hui
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia; Department of Optometry & Vision Sciences, The University of Melbourne, Melbourne, Australia
| | - Heberto Quintero
- Department of Neuroscience, University of Montreal, Montreal, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada
| | - Sana El Hajji
- Department of Neuroscience, University of Montreal, Montreal, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada
| | - Katharina Bell
- NHMRC Clinical Trials Centre, University of Sydney, Australia; Eye ACP Duke-NUS, Singapore
| | - Adriana Di Polo
- Department of Neuroscience, University of Montreal, Montreal, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada
| | - Pete A Williams
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
16
|
Brown EE, Scandura MJ, Pierce EA. Expression of NMNAT1 in the photoreceptors is sufficient to prevent NMNAT1-associated retinal degeneration. Mol Ther Methods Clin Dev 2023; 29:319-328. [PMID: 37214313 PMCID: PMC10193288 DOI: 10.1016/j.omtm.2023.04.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 04/12/2023] [Indexed: 05/24/2023]
Abstract
Nicotinamide nucleotide adenylyltransferase 1 (NMNAT1) is a ubiquitously expressed enzyme involved in nuclear NAD+ production throughout the body. However, mutations in the NMNAT1 gene lead to retina-specific disease with few reports of systemic effects. We have previously demonstrated that AAV-mediated gene therapy using self-complementary AAV (scAAV) to ubiquitously express NMNAT1 throughout the retina prevents retinal degeneration in a mouse model of NMNAT1-associated disease. We aimed to develop a better understanding of the cell types in the retina that contribute to disease pathogenesis in NMNAT1-associated disease, and to identify the cell types that require NMNAT1 expression for therapeutic benefit. To achieve this goal, we treated Nmnat1V9M/V9M mice with scAAV using cell type-specific promoters to restrict NMNAT1 expression to distinct retinal cell types. We hypothesized that photoreceptors are uniquely vulnerable to NAD+ depletion due to mutations in NMNAT1. Consistent with this hypothesis, we identified that treatments that drove NMNAT1 expression in the photoreceptors led to preservation of retinal morphology. These findings suggest that gene therapies for NMNAT1-associated disease should aim to express NMNAT1 in the photoreceptor cells.
Collapse
Affiliation(s)
- Emily E. Brown
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear Harvard Medical School, Boston, MA 02114, USA
| | - Michael J. Scandura
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear Harvard Medical School, Boston, MA 02114, USA
| | - Eric A. Pierce
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
17
|
Fazal SV, Mutschler C, Chen CZ, Turmaine M, Chen CY, Hsueh YP, Ibañez-Grau A, Loreto A, Casillas-Bajo A, Cabedo H, Franklin RJM, Barker RA, Monk KR, Steventon BJ, Coleman MP, Gomez-Sanchez JA, Arthur-Farraj P. SARM1 detection in myelinating glia: sarm1/ Sarm1 is dispensable for PNS and CNS myelination in zebrafish and mice. Front Cell Neurosci 2023; 17:1158388. [PMID: 37091921 PMCID: PMC10113485 DOI: 10.3389/fncel.2023.1158388] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 03/14/2023] [Indexed: 04/08/2023] Open
Abstract
Since SARM1 mutations have been identified in human neurological disease, SARM1 inhibition has become an attractive therapeutic strategy to preserve axons in a variety of disorders of the peripheral (PNS) and central nervous system (CNS). While SARM1 has been extensively studied in neurons, it remains unknown whether SARM1 is present and functional in myelinating glia? This is an important question to address. Firstly, to identify whether SARM1 dysfunction in other cell types in the nervous system may contribute to neuropathology in SARM1 dependent diseases? Secondly, to ascertain whether therapies altering SARM1 function may have unintended deleterious impacts on PNS or CNS myelination? Surprisingly, we find that oligodendrocytes express sarm1 mRNA in the zebrafish spinal cord and that SARM1 protein is readily detectable in rodent oligodendrocytes in vitro and in vivo. Furthermore, activation of endogenous SARM1 in cultured oligodendrocytes induces rapid cell death. In contrast, in peripheral glia, SARM1 protein is not detectable in Schwann cells and satellite glia in vivo and sarm1/Sarm1 mRNA is detected at very low levels in Schwann cells, in vivo, in zebrafish and mouse. Application of specific SARM1 activators to cultured mouse Schwann cells does not induce cell death and nicotinamide adenine dinucleotide (NAD) levels remain unaltered suggesting Schwann cells likely contain no functionally relevant levels of SARM1. Finally, we address the question of whether SARM1 is required for myelination or myelin maintenance. In the zebrafish and mouse PNS and CNS, we show that SARM1 is not required for initiation of myelination and myelin sheath maintenance is unaffected in the adult mouse nervous system. Thus, strategies to inhibit SARM1 function to treat neurological disease are unlikely to perturb myelination in humans.
Collapse
Affiliation(s)
- Shaline V. Fazal
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, United Kingdom
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Clara Mutschler
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, United Kingdom
| | - Civia Z. Chen
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Mark Turmaine
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Chiung-Ya Chen
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Yi-Ping Hsueh
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Andrea Ibañez-Grau
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández, Alicante, Spain
| | - Andrea Loreto
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, United Kingdom
| | - Angeles Casillas-Bajo
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández, Alicante, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
| | - Hugo Cabedo
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández, Alicante, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
| | - Robin J. M. Franklin
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
- Altos Labs - Cambridge Institute of Science, Cambridge, United Kingdom
| | - Roger A. Barker
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, United Kingdom
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Kelly R. Monk
- Vollum Institute, Oregon Health & Science University, Portland, OR, United States
| | | | - Michael P. Coleman
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, United Kingdom
| | - Jose A. Gomez-Sanchez
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández, Alicante, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago, Chile
| | - Peter Arthur-Farraj
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
18
|
Brown EE, Scandura MJ, Pierce E. Role of Nuclear NAD + in Retinal Homeostasis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1415:235-239. [PMID: 37440039 DOI: 10.1007/978-3-031-27681-1_34] [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: 07/14/2023]
Abstract
The retina is one of the most metabolically active tissues and maintenance of metabolic homeostasis is critical for retinal function. Nicotinamide adenine dinucleotide (NAD+) is a cofactor that is required for key processes, including the electron transport chain, glycolysis, fatty acid oxidation, and redox reactions. NAD+ also acts as a co-substrate for enzymes involved in maintaining genomic DNA integrity and cellular homeostasis, including poly-ADP ribose polymerases (PARPs) and Sirtuins. This review highlights the importance of NAD+ in the retina, including the role of enzymes involved in NAD+ production in the retina and how NAD+-consuming enzymes may play a role in disease pathology. We also suggest a cell death pathway that may be common in multiple models of photoreceptor degeneration and highlight the role that NAD+ likely plays in this process. Finally, we explore future experimental approaches to enhance our understanding of the role of NAD+ in the retina.
Collapse
Affiliation(s)
- Emily E Brown
- Ocular Genomics Institute, Massachusetts Eye and Ear, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Michael J Scandura
- Ocular Genomics Institute, Massachusetts Eye and Ear, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Eric Pierce
- Ocular Genomics Institute, Massachusetts Eye and Ear, Boston, MA, USA.
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
19
|
Sarkar A, Kumari N, Mukherjee P. The curious case of SARM1: Dr. Jekyll and Mr. Hyde in cell death and immunity? FEBS J 2023; 290:340-358. [PMID: 34710262 DOI: 10.1111/febs.16256] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/21/2021] [Accepted: 10/27/2021] [Indexed: 02/06/2023]
Abstract
Sterile alpha and toll/interleukin-1 receptor motif-containing protein 1 (SARM1) was first identified as a novel ortholog of Drosophila protein CG7915 and was subsequently placed as the fifth member of the human TIR-containing adaptor protein. SARM1 holds a unique position in this family where, unlike other members, it downregulates NFκB activity in response to immunogenic stimulation, interacts with another member of the family, TRIF, to negatively regulate its function, and it also mediates cell death responses. Over the past decade, SARM1 has emerged as one of the primary mediators of programmed axonal degeneration and this robust regulation of axonal degeneration-especially in models of peripheral neuropathy and traumatic injury-makes it an attractive target for therapeutic intervention. The TIR domain of SARM1 possesses an intrinsic NADase activity resulting in cellular energy deficits within the axons, a striking deviation from its other family members of human TLR adaptors. Interestingly, the TIR NADase activity, as seen in SARM1, is also observed in several prokaryotic TIR-containing proteins where they are involved in immune evasion once within the host. Although the immune function of SARM1 is yet to be conclusively discerned, this closeness in function with the prokaryotic TIR-domain containing proteins, places it at an interesting juncture of evolution raising questions about its origin and function in cell death and immunity. In this review, we discuss how a conserved immune adaptor protein like SARM1 switches to a pro-neurodegenerative function and the evolutionarily significance of the process.
Collapse
Affiliation(s)
- Ankita Sarkar
- School of Biotechnology, Presidency University, Kolkata, West Bengal, India
| | - Nripa Kumari
- School of Biotechnology, Presidency University, Kolkata, West Bengal, India
| | - Piyali Mukherjee
- School of Biotechnology, Presidency University, Kolkata, West Bengal, India
| |
Collapse
|
20
|
Khazma T, Golan-Vaishenker Y, Guez-Haddad J, Grossman A, Sain R, Weitman M, Plotnikov A, Zalk R, Yaron A, Hons M, Opatowsky Y. A duplex structure of SARM1 octamers stabilized by a new inhibitor. Cell Mol Life Sci 2022; 80:16. [PMID: 36564647 PMCID: PMC11072711 DOI: 10.1007/s00018-022-04641-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 11/16/2022] [Accepted: 11/19/2022] [Indexed: 12/25/2022]
Abstract
In recent years, there has been growing interest in SARM1 as a potential breakthrough drug target for treating various pathologies of axon degeneration. SARM1-mediated axon degeneration relies on its TIR domain NADase activity, but recent structural data suggest that the non-catalytic ARM domain could also serve as a pharmacological site as it has an allosteric inhibitory function. Here, we screened for synthetic small molecules that inhibit SARM1, and tested a selected set of these compounds in a DRG axon degeneration assay. Using cryo-EM, we found that one of the newly discovered inhibitors, a calmidazolium designated TK106, not only stabilizes the previously reported inhibited conformation of the octamer, but also a meta-stable structure: a duplex of octamers (16 protomers), which we have now determined to 4.0 Å resolution. In the duplex, each ARM domain protomer is engaged in lateral interactions with neighboring protomers, and is further stabilized by contralateral contacts with the opposing octamer ring. Mutagenesis of the duplex contact sites leads to a moderate increase in SARM1 activation in cultured cells. Based on our data we propose that the duplex assembly constitutes an additional auto-inhibition mechanism that tightly prevents pre-mature activation and axon degeneration.
Collapse
Affiliation(s)
- Tami Khazma
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | | | - Julia Guez-Haddad
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Atira Grossman
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Radhika Sain
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Michal Weitman
- Department of Chemistry, Bar-Ilan University, Ramat Gan, Israel
| | - Alexander Plotnikov
- The Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Ran Zalk
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Avraham Yaron
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Michael Hons
- European Molecular Biology Laboratory, Grenoble, France.
| | - Yarden Opatowsky
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel.
| |
Collapse
|
21
|
Multiomic Mass Spectrometry Imaging to Advance Future Pathological Understanding of Ocular Disease. Metabolites 2022; 12:metabo12121239. [PMID: 36557277 PMCID: PMC9786289 DOI: 10.3390/metabo12121239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/01/2022] [Accepted: 12/04/2022] [Indexed: 12/13/2022] Open
Abstract
Determining the locations of proteins within the eye thought to be involved in ocular pathogenesis is important to determine how best to target them for therapeutic benefits. However, immunohistochemistry is limited by the availability and specificity of antibodies. Additionally, the perceived role of both essential and non-essential metals within ocular tissue has been at the forefront of age-related macular degeneration (AMD) pathology for decades, yet even key metals such as copper and zinc have yet to have their roles deconvoluted. Here, mass spectrometry imaging (MSI) is employed to identify and spatially characterize both proteomic and metallomic species within ocular tissue to advance the application of a multiomic imaging methodology for the investigation of ocular diseases.
Collapse
|
22
|
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
|
23
|
Innate immunity dysregulation in aging eye and therapeutic interventions. Ageing Res Rev 2022; 82:101768. [PMID: 36280210 DOI: 10.1016/j.arr.2022.101768] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 09/29/2022] [Accepted: 10/20/2022] [Indexed: 01/31/2023]
Abstract
The prevalence of eye diseases increases considerably with age, resulting in significant vision impairment. Although the pathobiology of age-related eye diseases has been studied extensively, the contribution of immune-related changes due to aging remains elusive. In the eye, tissue-resident cells and infiltrating immune cells regulate innate responses during injury or infection. But due to aging, these cells lose their protective functions and acquire pathological phenotypes. Thus, dysregulated ocular innate immunity in the elderly increases the susceptibility and severity of eye diseases. Herein, we emphasize the impact of aging on the ocular innate immune system in the pathogenesis of infectious and non-infectious eye diseases. We discuss the role of age-related alterations in cellular metabolism, epigenetics, and cellular senescence as mechanisms underlying altered innate immune functions. Finally, we describe approaches to restore protective innate immune functions in the aging eye. Overall, the review summarizes our current understanding of innate immune functions in eye diseases and their dysregulation during aging.
Collapse
|
24
|
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
|
25
|
Lai MY, Li J, Zhang XX, Wu W, Li ZP, Sun ZX, Zhao MY, Yang DM, Wang DD, Li W, Zhao DM, Zhou XM, Yang LF. SARM1 participates in axonal degeneration and mitochondrial dysfunction in prion disease. Neural Regen Res 2022; 17:2293-2299. [PMID: 35259852 PMCID: PMC9083142 DOI: 10.4103/1673-5374.337051] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Prion disease represents a group of fatal neurogenerative diseases in humans and animals that are associated with energy loss, axonal degeneration, and mitochondrial dysfunction. Axonal degeneration is an early hallmark of neurodegeneration and is triggered by SARM1. We found that depletion or dysfunctional mutation of SARM1 protected against NAD+ loss, axonal degeneration, and mitochondrial functional disorder induced by the neurotoxic peptide PrP106-126. NAD+ supplementation rescued prion-triggered axonal degeneration and mitochondrial dysfunction and SARM1 overexpression suppressed this protective effect. NAD+ supplementation in PrP106-126-incubated N2a cells, SARM1 depletion, and SARM1 dysfunctional mutation each blocked neuronal apoptosis and increased cell survival. Our results indicate that the axonal degeneration and mitochondrial dysfunction triggered by PrP106-126 are partially dependent on SARM1 NADase activity. This pathway has potential as a therapeutic target in the early stages of prion disease.
Collapse
Affiliation(s)
- Meng-Yu Lai
- National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jie Li
- National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xi-Xi Zhang
- National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Wei Wu
- National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Zhi-Ping Li
- National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Zhi-Xin Sun
- National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Meng-Yang Zhao
- National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Dong-Ming Yang
- National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Dong-Dong Wang
- National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Wen Li
- National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - De-Ming Zhao
- National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xiang-Mei Zhou
- National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Li-Feng Yang
- National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
26
|
Lee D, Tomita Y, Miwa Y, Shinojima A, Ban N, Yamaguchi S, Nishioka K, Negishi K, Yoshino J, Kurihara T. Nicotinamide Mononucleotide Prevents Retinal Dysfunction in a Mouse Model of Retinal Ischemia/Reperfusion Injury. Int J Mol Sci 2022; 23:ijms231911228. [PMID: 36232528 PMCID: PMC9570481 DOI: 10.3390/ijms231911228] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 09/21/2022] [Accepted: 09/22/2022] [Indexed: 11/16/2022] Open
Abstract
Retinal ischemia/reperfusion (I/R) injury can cause severe vision impairment. Retinal I/R injury is associated with pathological increases in reactive oxygen species and inflammation, resulting in retinal neuronal cell death. To date, effective therapies have not been developed. Nicotinamide mononucleotide (NMN), a key nicotinamide adenine dinucleotide (NAD+) intermediate, has been shown to exert neuroprotection for retinal diseases. However, it remains unclear whether NMN can prevent retinal I/R injury. Thus, we aimed to determine whether NMN therapy is useful for retinal I/R injury-induced retinal degeneration. One day after NMN intraperitoneal (IP) injection, adult mice were subjected to retinal I/R injury. Then, the mice were injected with NMN once every day for three days. Electroretinography and immunohistochemistry were used to measure retinal functional alterations and retinal inflammation, respectively. The protective effect of NMN administration was further examined using a retinal cell line, 661W, under CoCl2-induced oxidative stress conditions. NMN IP injection significantly suppressed retinal functional damage, as well as inflammation. NMN treatment showed protective effects against oxidative stress-induced cell death. The antioxidant pathway (Nrf2 and Hmox-1) was activated by NMN treatment. In conclusion, NMN could be a promising preventive neuroprotective drug for ischemic retinopathy.
Collapse
Affiliation(s)
- Deokho Lee
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Yohei Tomita
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Yukihiro Miwa
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
- Aichi Animal Eye Clinic, Nagoya 466-0827, Japan
| | - Ari Shinojima
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Norimitsu Ban
- Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Shintaro Yamaguchi
- Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Ken Nishioka
- Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Kazuno Negishi
- Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Jun Yoshino
- Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Toshihide Kurihara
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
- Correspondence:
| |
Collapse
|
27
|
Ademi M, Yang X, Coleman MP, Gilley J. Natural variants of human SARM1 cause both intrinsic and dominant loss-of-function influencing axon survival. Sci Rep 2022; 12:13846. [PMID: 35974060 PMCID: PMC9381744 DOI: 10.1038/s41598-022-18052-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/04/2022] [Indexed: 11/08/2022] Open
Abstract
SARM1 is a central executioner of programmed axon death, and this role requires intrinsic NAD(P)ase or related enzyme activity. A complete absence of SARM1 robustly blocks axon degeneration in mice, but even a partial depletion confers meaningful protection. Since axon loss contributes substantially to the onset and progression of multiple neurodegenerative disorders, lower inherent SARM1 activity is expected to reduce disease susceptibility in some situations. We, therefore, investigated whether there are naturally occurring SARM1 alleles within the human population that encode SARM1 variants with loss-of-function. Out of the 18 natural SARM1 coding variants we selected as candidates, we found that 10 display loss-of-function in three complimentary assays: they fail to robustly deplete NAD in transfected HEK 293T cells; they lack constitutive and NMN-induced NADase activity; and they fail to promote axon degeneration in primary neuronal cultures. Two of these variants are also able to block axon degeneration in primary culture neurons in the presence of endogenous, wild-type SARM1, indicative of dominant loss-of-function. These results demonstrate that SARM1 loss-of-function variants occur naturally in the human population, and we propose that carriers of these alleles will have different degrees of reduced susceptibility to various neurological conditions.
Collapse
Affiliation(s)
- Mirlinda Ademi
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Forvie Site, Robinson Way, Cambridge, CB2 0PY, UK
| | - Xiuna Yang
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Forvie Site, Robinson Way, Cambridge, CB2 0PY, UK
| | - Michael P Coleman
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Forvie Site, Robinson Way, Cambridge, CB2 0PY, UK.
| | - Jonathan Gilley
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Forvie Site, Robinson Way, Cambridge, CB2 0PY, UK.
| |
Collapse
|
28
|
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
|
29
|
Coleman MP. Axon Biology in ALS: Mechanisms of Axon Degeneration and Prospects for Therapy. Neurotherapeutics 2022; 19:1133-1144. [PMID: 36207571 PMCID: PMC9587191 DOI: 10.1007/s13311-022-01297-6] [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] [Accepted: 09/02/2022] [Indexed: 10/10/2022] Open
Abstract
This review addresses the longstanding debate over whether amyotrophic lateral sclerosis (ALS) is a 'dying back' or 'dying forward' disorder in the light of new gene identifications and the increased understanding of mechanisms of action for previously identified ALS genes. While the topological pattern of pathology in animal models, and more anecdotally in patients is indeed 'dying back', this review discusses how this fits with the fact that many of the major initiating events are thought to occur within the soma. It also discusses how widely varying ALS risk factors, including some impacting axons directly, may combine to drive a common pathway involving TAR DNA binding protein 43 (TDP-43) and neuromuscular junction (NMJ) denervation. The emerging association between sterile alpha and TIR motif-containing 1 (SARM1), a protein so far mostly associated with axon degeneration, and sporadic ALS is another major theme. The strengths and limitations of the current evidence supporting an association are considered, along with ways in which SARM1 could become activated in ALS. The final section addresses SARM1-based therapies along with the prospects for targeting other axonal steps in ALS pathogenesis.
Collapse
Affiliation(s)
- Michael P Coleman
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Robinson Way, Cambridge, CB2 0PY, UK.
| |
Collapse
|
30
|
Zhang Y, Guo X, Peng Z, Liu C, Ren L, Liang J, Wang P. Nicotinamide Mononucleotide Adenylyltransferase 1 Regulates Cerebral Ischemia-Induced Blood-Brain Barrier Disruption Through NAD +/SIRT1 Signaling Pathway. Mol Neurobiol 2022; 59:4879-4891. [PMID: 35657458 DOI: 10.1007/s12035-022-02903-6] [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: 11/09/2021] [Accepted: 05/26/2022] [Indexed: 10/18/2022]
Abstract
The molecular mechanisms of blood-brain barrier (BBB) disruption in the early stage after ischemic stroke are poorly understood. In the present study, we investigated the potential role of nicotinamide mononucleotide adenylyltransferase 1 (NMNAT1) in ischemia-induced BBB damage using an animal middle cerebral artery occlusion (MCAO) model of ischemic stroke. Recombinant human NMNAT1 (rh-NMNAT1) was administered intranasally and Sirtuin 1 (SIRT1) siRNA was administered by intracerebroventricular injection. Our results indicate that rh-NMNAT1 reduced infarct volume, improved functional outcome, and decreased BBB permeability in mice after ischemic stroke. Furthermore, rh-NMNAT1 prevented the loss of tight junction proteins (occludin and claudin-5) and reduced cell apoptosis in ischemic microvessels. NMNAT1-mediated BBB permeability was correlated with the elevation of nicotinamide adenine dinucleotide (NAD+)/NADH ratio and SIRT1 level in brain microvascular endothelial cells. In addition, rh-NMNAT1 treatment significantly decreased the levels of acetylated nuclear factor-κB, acetylated p53, and matrix metalloproteinase-9 in ischemic microvessels. Moreover, the protective effects of rh-NMNAT1 could be reversed by SIRT1 siRNA. In conclusion, these findings indicate that rh-NMNAT1 protects BBB integrity after cerebral ischemia via the NAD+/SIRT1 signaling pathway in brain microvascular endothelial cells. NMNAT1 may be a novel potential therapeutic target for reducing BBB disruption after ischemic stroke.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Neurobiology and Key Laboratory of Neurodegenerative Diseases of Liaoning Province, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Xun Guo
- Department of Neurobiology and Key Laboratory of Neurodegenerative Diseases of Liaoning Province, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Zhifeng Peng
- Department of Physiology, Shanxi Datong University, Datong, 037009, Shanxi, China
| | - Chang Liu
- Department of Neurobiology and Key Laboratory of Neurodegenerative Diseases of Liaoning Province, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Lili Ren
- Department of Neurobiology and Key Laboratory of Neurodegenerative Diseases of Liaoning Province, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China
| | - Jia Liang
- Institute of Life Science, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China.
| | - Peng Wang
- Department of Neurobiology and Key Laboratory of Neurodegenerative Diseases of Liaoning Province, Jinzhou Medical University, Jinzhou, 121001, Liaoning, China.
| |
Collapse
|
31
|
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
|
32
|
Fang F, Zhuang P, Feng X, Liu P, Liu D, Huang H, Li L, Chen W, Liu L, Sun Y, Jiang H, Ye J, Hu Y. NMNAT2 is downregulated in glaucomatous RGCs, and RGC-specific gene therapy rescues neurodegeneration and visual function. Mol Ther 2022; 30:1421-1431. [PMID: 35114390 PMCID: PMC9077370 DOI: 10.1016/j.ymthe.2022.01.035] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 12/17/2021] [Accepted: 01/27/2022] [Indexed: 11/19/2022] Open
Abstract
The lack of neuroprotective treatments for retinal ganglion cells (RGCs) and optic nerve (ON) is a central challenge for glaucoma management. Emerging evidence suggests that redox factor NAD+ decline is a hallmark of aging and neurodegenerative diseases. Supplementation with NAD+ precursors and overexpression of NMNAT1, the key enzyme in the NAD+ biosynthetic process, have significant neuroprotective effects. We first profile the translatomes of RGCs in naive mice and mice with silicone oil-induced ocular hypertension (SOHU)/glaucoma by RiboTag mRNA sequencing. Intriguingly, only NMNAT2, but not NMNAT1 or NMNAT3, is significantly decreased in SOHU glaucomatous RGCs, which we confirm by in situ hybridization. We next demonstrate that AAV2 intravitreal injection-mediated overexpression of long half-life NMNAT2 mutant driven by RGC-specific mouse γ-synuclein (mSncg) promoter restores decreased NAD+ levels in glaucomatous RGCs and ONs. Moreover, this RGC-specific gene therapy strategy delivers significant neuroprotection of both RGC soma and axon and preservation of visual function in the traumatic ON crush model and the SOHU glaucoma model. Collectively, our studies suggest that the weakening of NMNAT2 expression in glaucomatous RGCs contributes to a deleterious NAD+ decline, and that modulating RGC-intrinsic NMNAT2 levels by AAV2-mSncg vector is a promising gene therapy for glaucomatous neurodegeneration.
Collapse
Affiliation(s)
- Fang Fang
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA 94304, USA; Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Pei Zhuang
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Xue Feng
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Pingting Liu
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Dong Liu
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Haoliang Huang
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Liang Li
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Wei Chen
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Liang Liu
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Yang Sun
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Haowen Jiang
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jiangbin Ye
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Yang Hu
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA 94304, USA.
| |
Collapse
|
33
|
Gibbons L, Ozaki E, Greene C, Trappe A, Carty M, Coppinger JA, Bowie AG, Campbell M, Doyle SL. SARM1 Promotes Photoreceptor Degeneration in an Oxidative Stress Model of Retinal Degeneration. Front Neurosci 2022; 16:852114. [PMID: 35431772 PMCID: PMC9012108 DOI: 10.3389/fnins.2022.852114] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/04/2022] [Indexed: 11/13/2022] Open
Abstract
SARM1 (sterile alpha and armadillo motif-containing protein) is a highly conserved Toll/IL-1 Receptor (TIR) adaptor with important roles in mediating immune responses. Studies in the brain have shown that SARM1 plays a role in induction of neuronal axon degeneration in response to a variety of injuries. We recently demonstrated that SARM1 is pro-degenerative in a genetic model of inherited retinopathy. This current study aimed to characterise the effect of SARM1 deletion in an alternative model of retinal degeneration (RD) in which the retinal pigment epithelium (RPE) fragments following administration of oxidising agent, sodium iodate (NaIO3), leading to subsequent photoreceptor cell death. Following administration of NaIO3, we observed no apparent difference in rate of loss of RPE integrity in SARM1 deficient mice compared to WT counterparts. However, despite no differences in RPE degeneration, photoreceptor cell number and retinal thickness were increased in Sarm1–/– mice compared to WT counterparts. This apparent protection of the photoreceptors in SARM1 deficient mice is supported by an observed decrease in pro-apoptotic caspase-3 in the photoreceptor layer of Sarm1–/– mice compared to WT. Together these data indicate a pro-degenerative role for SARM1 in the photoreceptors, but not in the RPE, in an oxidative stress induced model of retinal degeneration consistent with its known degenerative role in neurons in a range of neurodegenerative settings.
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
| | - Ema Ozaki
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
- Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Chris Greene
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Anne Trappe
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Michael Carty
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Judith A. Coppinger
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Andrew G. Bowie
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Matthew Campbell
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Sarah L. Doyle
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
- Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Dublin, Ireland
- *Correspondence: Sarah L. Doyle,
| |
Collapse
|
34
|
Li X, Li Y, Li F, Chen Q, Zhao Z, Liu X, Zhang N, Li H. NAD + Anabolism Disturbance Causes Glomerular Mesangial Cell Injury in Diabetic Nephropathy. Int J Mol Sci 2022; 23:ijms23073458. [PMID: 35408818 PMCID: PMC8998683 DOI: 10.3390/ijms23073458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/17/2022] [Accepted: 03/19/2022] [Indexed: 12/04/2022] Open
Abstract
The homeostasis of NAD+ anabolism is indispensable for maintaining the NAD+ pool. In mammals, the mainly synthetic pathway of NAD+ is the salvage synthesis, a reaction catalyzed by nicotinamide mononucleotide adenylyltransferase (NAMPT) and nicotinamide mononucleotide adenylyltransferase (NMNATs) successively, converting nicotinamide (NAM) to nicotinamide mononucleotide (NMN) and NMN to NAD+, respectively. However, the relationship between NAD+ anabolism disturbance and diabetic nephropathy (DN) remains elusive. Here our study found that the disruption of NAD+ anabolism homeostasis caused an elevation in both oxidative stress and fibronectin expression, along with a decrease in Sirt1 and an increase in both NF-κB P65 expression and acetylation, culminating in extracellular matrix deposition and globular fibrosis in DN. More importantly, through constitutively overexpressing NMNAT1 or NAMPT in human mesangial cells, we revealed NAD+ levels altered inversely with NMN levels in the context of DN and, further, their changes affect Sirt1/NF-κB P65, thus playing a crucial role in the pathogenesis of DN. Accordingly, FK866, a NAMPT inhibitor, and quercetin, a Sirt1 agonist, have favorable effects on the maintenance of NAD+ homeostasis and renal function in db/db mice. Collectively, our findings suggest that NMN accumulation may provide a causal link between NAD+ anabolism disturbance and diabetic nephropathy (DN) as well as a promising therapeutic target for DN treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Hui Li
- Correspondence: ; Tel.: +86-021-54237528
| |
Collapse
|
35
|
Kayazawa T, Kuniyoshi K, Hatsukawa Y, Fujinami K, Yoshitake K, Tsunoda K, Shimojo H, Iwata T, Kusaka S. Clinical course of a Japanese girl with Leber congenital amaurosis associated with a novel nonsense pathogenic variant in NMNAT1: a case report and mini review. Ophthalmic Genet 2022; 43:400-408. [PMID: 35026968 DOI: 10.1080/13816810.2021.2023195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Leber congenital amaurosis (LCA), although rare, is one of the most severe forms of early-onset inherited retinal dystrophy (IRD). Here, we review the molecular genetics and phenotypic characteristics of patients with NMNAT1-associated IRD. The longitudinal clinical and molecular findings of a Japanese girl diagnosed with LCA associated with pathogenic variants in NMNAT1 c.648delG, (p.Trp216Ter*) and c.709C>T (p.Arg237Cys) have been described to highlight the salient clinical features of NMNAT1-associated IRD.
Collapse
Affiliation(s)
- Tomoyasu Kayazawa
- Department of Ophthalmology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Kazuki Kuniyoshi
- Department of Ophthalmology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Yoshikazu Hatsukawa
- Department of Ophthalmology, Osaka Women's and Children's Hospital, Osaka, Japan
| | - Kaoru Fujinami
- Laboratory of Visual Physiology, Division of Vision Research, National Institute of Sensory Organs, National Hospital Organization Tokyo Medical Center, Tokyo, Japan.,Genetics, UCL Institute of Ophthalmology Associated with Moorfields Eye Hospital, London, UK.,Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Kazutoshi Yoshitake
- Division of Molecular and Cellular Biology, National Institute of Sensory Organs, National Hospital Organization Tokyo Medical Center, Tokyo, Japan.,Graduate School of Agricultural and Life Science, Faculty of Agriculture, The University of Tokyo, Tokyo, Japan
| | - Kazushige Tsunoda
- Laboratory of Visual Physiology, Division of Vision Research, National Institute of Sensory Organs, National Hospital Organization Tokyo Medical Center, Tokyo, Japan
| | - Hiroshi Shimojo
- Department of Ophthalmology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Takeshi Iwata
- Division of Molecular and Cellular Biology, National Institute of Sensory Organs, National Hospital Organization Tokyo Medical Center, Tokyo, Japan
| | - Shunji Kusaka
- Department of Ophthalmology, Kindai University Faculty of Medicine, Osaka, Japan
| |
Collapse
|
36
|
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
|
37
|
Sokolov D, Sechrest ER, Wang Y, Nevin C, Du J, Kolandaivelu S. Nuclear NAD +-biosynthetic enzyme NMNAT1 facilitates development and early survival of retinal neurons. eLife 2021; 10:e71185. [PMID: 34878972 PMCID: PMC8754432 DOI: 10.7554/elife.71185] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 12/07/2021] [Indexed: 11/13/2022] Open
Abstract
Despite mounting evidence that the mammalian retina is exceptionally reliant on proper NAD+ homeostasis for health and function, the specific roles of subcellular NAD+ pools in retinal development, maintenance, and disease remain obscure. Here, we show that deletion of the nuclear-localized NAD+ synthase nicotinamide mononucleotide adenylyltransferase-1 (NMNAT1) in the developing murine retina causes early and severe degeneration of photoreceptors and select inner retinal neurons via multiple distinct cell death pathways. This severe phenotype is associated with disruptions to retinal central carbon metabolism, purine nucleotide synthesis, and amino acid pathways. Furthermore, transcriptomic and immunostaining approaches reveal dysregulation of a collection of photoreceptor and synapse-specific genes in NMNAT1 knockout retinas prior to detectable morphological or metabolic alterations. Collectively, our study reveals previously unrecognized complexity in NMNAT1-associated retinal degeneration and suggests a yet-undescribed role for NMNAT1 in gene regulation during photoreceptor terminal differentiation.
Collapse
Affiliation(s)
- David Sokolov
- Department of Ophthalmology and Visual Sciences, Eye Institute, One Medical Center Drive, West Virginia UniversityMorgantownUnited States
| | - Emily R Sechrest
- Department of Ophthalmology and Visual Sciences, Eye Institute, One Medical Center Drive, West Virginia UniversityMorgantownUnited States
| | - Yekai Wang
- Department of Ophthalmology and Visual Sciences, Eye Institute, One Medical Center Drive, West Virginia UniversityMorgantownUnited States
- Department of Biochemistry, One Medical Center Drive, West Virginia UniversityMorgantownUnited States
| | - Connor Nevin
- Department of Ophthalmology and Visual Sciences, Eye Institute, One Medical Center Drive, West Virginia UniversityMorgantownUnited States
| | - Jianhai Du
- Department of Ophthalmology and Visual Sciences, Eye Institute, One Medical Center Drive, West Virginia UniversityMorgantownUnited States
- Department of Biochemistry, One Medical Center Drive, West Virginia UniversityMorgantownUnited States
| | - Saravanan Kolandaivelu
- Department of Ophthalmology and Visual Sciences, Eye Institute, One Medical Center Drive, West Virginia UniversityMorgantownUnited States
- Department of Biochemistry, One Medical Center Drive, West Virginia UniversityMorgantownUnited States
| |
Collapse
|
38
|
Loreto A, Angeletti C, Gu W, Osborne A, Nieuwenhuis B, Gilley J, Merlini E, Arthur-Farraj P, Amici A, Luo Z, Hartley-Tassell L, Ve T, Desrochers LM, Wang Q, Kobe B, Orsomando G, Coleman MP. Neurotoxin-mediated potent activation of the axon degeneration regulator SARM1. eLife 2021; 10:72823. [PMID: 34870595 PMCID: PMC8758145 DOI: 10.7554/elife.72823] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 12/05/2021] [Indexed: 11/13/2022] Open
Abstract
Axon loss underlies symptom onset and progression in many neurodegenerative disorders. Axon degeneration in injury and disease is promoted by activation of the NAD-consuming enzyme SARM1. Here, we report a novel activator of SARM1, a metabolite of the pesticide and neurotoxin vacor. Removal of SARM1 completely rescues mouse neurons from vacor-induced neuron and axon death in vitro and in vivo. We present the crystal structure of the Drosophila SARM1 regulatory domain complexed with this activator, the vacor metabolite VMN, which as the most potent activator yet known is likely to support drug development for human SARM1 and NMNAT2 disorders. This study indicates the mechanism of neurotoxicity and pesticide action by vacor, raises important questions about other pyridines in wider use today, provides important new tools for drug discovery, and demonstrates that removing SARM1 can robustly block programmed axon death induced by toxicity as well as genetic mutation.
Collapse
Affiliation(s)
- Andrea Loreto
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Carlo Angeletti
- Department of Clinical Sciences (DISCO), Section of Biochemistry, Polytechnic University of Marche, Ancona, Italy
| | - Weixi Gu
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, Australia
| | - Andrew Osborne
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Bart Nieuwenhuis
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Jonathan Gilley
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Elisa Merlini
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Peter Arthur-Farraj
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Adolfo Amici
- Department of Clinical Sciences (DISCO), Section of Biochemistry, Polytechnic University of Marche, Ancona, Italy
| | - Zhenyao Luo
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, Australia
| | | | - Thomas Ve
- Institute for Glycomics, Griffith University, Southport, Australia
| | - Laura M Desrochers
- Neuroscience, BioPharmaceuticals R and D, AstraZeneca, Waltham, United States
| | - Qi Wang
- Neuroscience, BioPharmaceuticals R and D, AstraZeneca, Waltham, United States
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, Australia
| | - Giuseppe Orsomando
- Department of Clinical Sciences (DISCO), Section of Biochemistry, Polytechnic University of Marche, Ancona, Italy
| | - Michael P Coleman
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
39
|
Gilley J, Jackson O, Pipis M, Estiar MA, Al-Chalabi A, Danzi MC, van Eijk KR, Goutman SA, Harms MB, Houlden H, Iacoangeli A, Kaye J, Lima L, Ravits J, Rouleau GA, Schüle R, Xu J, Züchner S, Cooper-Knock J, Gan-Or Z, Reilly MM, Coleman MP. Enrichment of SARM1 alleles encoding variants with constitutively hyperactive NADase in patients with ALS and other motor nerve disorders. eLife 2021; 10:e70905. [PMID: 34796871 PMCID: PMC8735862 DOI: 10.7554/elife.70905] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 11/18/2021] [Indexed: 11/13/2022] Open
Abstract
SARM1, a protein with critical NADase activity, is a central executioner in a conserved programme of axon degeneration. We report seven rare missense or in-frame microdeletion human SARM1 variant alleles in patients with amyotrophic lateral sclerosis (ALS) or other motor nerve disorders that alter the SARM1 auto-inhibitory ARM domain and constitutively hyperactivate SARM1 NADase activity. The constitutive NADase activity of these seven variants is similar to that of SARM1 lacking the entire ARM domain and greatly exceeds the activity of wild-type SARM1, even in the presence of nicotinamide mononucleotide (NMN), its physiological activator. This rise in constitutive activity alone is enough to promote neuronal degeneration in response to otherwise non-harmful, mild stress. Importantly, these strong gain-of-function alleles are completely patient-specific in the cohorts studied and show a highly significant association with disease at the single gene level. These findings of disease-associated coding variants that alter SARM1 function build on previously reported genome-wide significant association with ALS for a neighbouring, more common SARM1 intragenic single nucleotide polymorphism (SNP) to support a contributory role of SARM1 in these disorders. A broad phenotypic heterogeneity and variable age-of-onset of disease among patients with these alleles also raises intriguing questions about the pathogenic mechanism of hyperactive SARM1 variants.
Collapse
Affiliation(s)
- Jonathan Gilley
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of CambridgeCambridgeUnited Kingdom
| | - Oscar Jackson
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of CambridgeCambridgeUnited Kingdom
| | - Menelaos Pipis
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology and The National Hospital for NeurologyLondonUnited Kingdom
| | - Mehrdad A Estiar
- Department of Human Genetics, McGill UniversityMontrealCanada
- The Neuro (Montreal Neurological Institute-Hospital), McGill UniversityMontrealCanada
| | - Ammar Al-Chalabi
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College LondonLondonUnited Kingdom
- Department of Neurology, King's College Hospital, King’s College LondonLondonUnited Kingdom
| | - Matt C Danzi
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of MedicineMiamiUnited States
| | - Kristel R van Eijk
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht UniversityUtrechtNetherlands
| | - Stephen A Goutman
- Department of Neurology, University of MichiganAnn ArborUnited States
| | - Matthew B Harms
- Institute for Genomic Medicine, Columbia UniversityNew YorkUnited States
| | - Henry Houlden
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology and The National Hospital for NeurologyLondonUnited Kingdom
| | - Alfredo Iacoangeli
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College LondonLondonUnited Kingdom
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology & Neuroscience, King's College LondonLondonUnited Kingdom
- National Institute for Health Research Biomedical Research Centre and Dementia Unit at South London and Maudsley NHS Foundation Trust and King's College LondonLondonUnited Kingdom
| | - Julia Kaye
- Center for Systems and Therapeutics, Gladstone InstitutesSan FranciscoUnited States
| | - Leandro Lima
- Center for Systems and Therapeutics, Gladstone InstitutesSan FranciscoUnited States
- Gladstone Institute of Data Science and Biotechnology, Gladstone InstitutesSan FranciscoUnited States
| | - Queen Square Genomics
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology and The National Hospital for NeurologyLondonUnited Kingdom
| | - John Ravits
- Department of Neurosciences, University of California, San DiegoLa JollaUnited States
| | - Guy A Rouleau
- Department of Human Genetics, McGill UniversityMontrealCanada
- The Neuro (Montreal Neurological Institute-Hospital), McGill UniversityMontrealCanada
- Department of Neurology and Neurosurgery, McGill UniversityMontrealCanada
| | - Rebecca Schüle
- Center for Neurology and Hertie Institute für Clinical Brain Research, University of Tübingen, German Center for Neurodegenerative DiseasesTübingenGermany
| | - Jishu Xu
- Center for Neurology and Hertie Institute für Clinical Brain Research, University of Tübingen, German Center for Neurodegenerative DiseasesTübingenGermany
| | - Stephan Züchner
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of MedicineMiamiUnited States
| | - Johnathan Cooper-Knock
- Sheffield Institute for Translational Neuroscience, University of SheffieldSheffieldUnited Kingdom
| | - Ziv Gan-Or
- Department of Human Genetics, McGill UniversityMontrealCanada
- The Neuro (Montreal Neurological Institute-Hospital), McGill UniversityMontrealCanada
- Department of Neurology and Neurosurgery, McGill UniversityMontrealCanada
| | - Mary M Reilly
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology and The National Hospital for NeurologyLondonUnited Kingdom
| | - Michael P Coleman
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of CambridgeCambridgeUnited Kingdom
| |
Collapse
|
40
|
Ko KW, Devault L, Sasaki Y, Milbrandt J, DiAntonio A. Live imaging reveals the cellular events downstream of SARM1 activation. eLife 2021; 10:e71148. [PMID: 34779400 PMCID: PMC8612704 DOI: 10.7554/elife.71148] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 11/12/2021] [Indexed: 12/15/2022] Open
Abstract
SARM1 is an inducible NAD+ hydrolase that triggers axon loss and neuronal cell death in the injured and diseased nervous system. While SARM1 activation and enzyme function are well defined, the cellular events downstream of SARM1 activity but prior to axonal demise are much less well understood. Defects in calcium, mitochondria, ATP, and membrane homeostasis occur in injured axons, but the relationships among these events have been difficult to disentangle because prior studies analyzed large collections of axons in which cellular events occur asynchronously. Here, we used live imaging of mouse sensory neurons with single axon resolution to investigate the cellular events downstream of SARM1 activity. Our studies support a model in which SARM1 NADase activity leads to an ordered sequence of events from loss of cellular ATP, to defects in mitochondrial movement and depolarization, followed by calcium influx, externalization of phosphatidylserine, and loss of membrane permeability prior to catastrophic axonal self-destruction.
Collapse
Affiliation(s)
- Kwang Woo Ko
- Washington University School of MedicineSt LouisUnited States
| | - Laura Devault
- Washington University School of MedicineSt LouisUnited States
| | - Yo Sasaki
- Genetics, Washington University School of MedicineSt LouisUnited States
| | - Jeffrey Milbrandt
- Genetics, Hope Center for Neurological Disorders, Washington University School of MedicineSt LouisUnited States
| | - Aaron DiAntonio
- Developmental Biology, Needleman Center for Neurometabolism and Axonal Therapeutics, Washington University School of MedicineSt LouisUnited States
| |
Collapse
|
41
|
Brown EE, Scandura MJ, Mehrotra S, Wang Y, Du J, Pierce EA. Reduced nuclear NAD+ drives DNA damage and subsequent immune activation in the retina. Hum Mol Genet 2021; 31:1370-1388. [PMID: 34750622 DOI: 10.1093/hmg/ddab324] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/14/2021] [Accepted: 11/04/2021] [Indexed: 11/13/2022] Open
Abstract
Mutations in NMNAT1, a key enzyme involved in the synthesis of NAD+ in the nucleus, lead to an early onset severe inherited retinal degeneration (IRD). We aimed to understand the role of nuclear NAD+ in the retina and to identify the molecular mechanisms underlying NMNAT1-associated disease, using a mouse model that harbors the p.V9M mutation in Nmnat1 (Nmnat1V9M/V9M). We identified temporal transcriptional reprogramming in the retinas of Nmnat1V9M/V9M mice prior to retinal degeneration, which begins at 4 weeks of age, with no significant alterations in gene expression at 2 weeks of age and over 2600 differentially expressed genes by 3 weeks of age. Expression of the primary consumer of NAD+ in the nucleus, PARP1, an enzyme involved in DNA damage repair and transcriptional regulation, as well as 7 other PARP family enzymes, was elevated in the retinas of Nmnat1V9M/V9M. This was associated with elevated levels of DNA damage, PARP-mediated NAD+ consumption, and migration of Iba1+/CD45+ microglia/macrophages to the subretinal space in the retinas of Nmnat1V9M/V9M mice. These findings suggest that photoreceptor cells are especially sensitive to perturbation of genome homeostasis, and that PARP-mediated cell death may play a role in other genetic forms of IRDs, and potentially other forms of neurodegeneration.
Collapse
Affiliation(s)
- Emily E Brown
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA
| | - Michael J Scandura
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA
| | - Sudeep Mehrotra
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA
| | - Yekai Wang
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, WV, 26506, USA.,Department of Biochemistry, West Virginia University, Morgantown, WV, 26506, USA
| | - Jianhai Du
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, WV, 26506, USA.,Department of Biochemistry, West Virginia University, Morgantown, WV, 26506, USA
| | - Eric A Pierce
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA
| |
Collapse
|
42
|
Sasaki Y, Zhu J, Shi Y, Gu W, Kobe B, Ve T, DiAntonio A, Milbrandt J. Nicotinic acid mononucleotide is an allosteric SARM1 inhibitor promoting axonal protection. Exp Neurol 2021; 345:113842. [PMID: 34403688 PMCID: PMC8571713 DOI: 10.1016/j.expneurol.2021.113842] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/09/2021] [Accepted: 08/12/2021] [Indexed: 12/31/2022]
Abstract
SARM1 is an inducible NAD+ hydrolase that is the central executioner of pathological axon loss. Recently, we elucidated the molecular mechanism of SARM1 activation, demonstrating that SARM1 is a metabolic sensor regulated by the levels of NAD+ and its precursor, nicotinamide mononucleotide (NMN), via their competitive binding to an allosteric site within the SARM1 N-terminal ARM domain. In healthy neurons with abundant NAD+, binding of NAD+ blocks access of NMN to this allosteric site. However, with injury or disease the levels of the NAD+ biosynthetic enzyme NMNAT2 drop, increasing the NMN/ NAD+ ratio and thereby promoting NMN binding to the SARM1 allosteric site, which in turn induces a conformational change activating the SARM1 NAD+ hydrolase. Hence, NAD+ metabolites both regulate the activation of SARM1 and, in turn, are regulated by the SARM1 NAD+ hydrolase. This dual upstream and downstream role for NAD+ metabolites in SARM1 function has hindered mechanistic understanding of axoprotective mechanisms that manipulate the NAD+ metabolome. Here we reevaluate two methods that potently block axon degeneration via modulation of NAD+ related metabolites, 1) the administration of the NMN biosynthesis inhibitor FK866 in conjunction with the NAD+ precursor nicotinic acid riboside (NaR) and 2) the neuronal expression of the bacterial enzyme NMN deamidase. We find that these approaches not only lead to a decrease in the levels of the SARM1 activator NMN, but also an increase in the levels of the NAD+ precursor nicotinic acid mononucleotide (NaMN). We show that NaMN inhibits SARM1 activation, and demonstrate that this NaMN-mediated inhibition is important for the long-term axon protection induced by these treatments. Analysis of the NaMN-ARM domain co-crystal structure shows that NaMN competes with NMN for binding to the SARM1 allosteric site and promotes the open, autoinhibited configuration of SARM1 ARM domain. Together, these results demonstrate that the SARM1 allosteric pocket can bind a diverse set of metabolites including NMN, NAD+, and NaMN to monitor cellular NAD+ homeostasis and regulate SARM1 NAD+ hydrolase activity. The relative promiscuity of the allosteric site may enable the development of potent pharmacological inhibitors of SARM1 activation for the treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Yo Sasaki
- Washington University School of Medicine in Saint Louis, Department of Genetics, St. Louis, MO, USA; Needleman Center for Neurometabolism and Axonal Therapeutics, USA.
| | - Jian Zhu
- Washington University School of Medicine in Saint Louis, Department of Genetics, St. Louis, MO, USA; Needleman Center for Neurometabolism and Axonal Therapeutics, USA
| | - Yun Shi
- Institute for Glycomics, Griffith University, Southport, QLD 4222, Australia
| | - Weixi Gu
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, QLD 4072, Australia
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, QLD 4072, Australia
| | - Thomas Ve
- Institute for Glycomics, Griffith University, Southport, QLD 4222, Australia
| | - Aaron DiAntonio
- Washington University School of Medicine in Saint Louis, Department of Developmental Biology, St. Louis, MO, USA; Needleman Center for Neurometabolism and Axonal Therapeutics, USA
| | - Jeffrey Milbrandt
- Washington University School of Medicine in Saint Louis, Department of Genetics, St. Louis, MO, USA; Needleman Center for Neurometabolism and Axonal Therapeutics, USA
| |
Collapse
|
43
|
DiAntonio A, Milbrandt J, Figley MD. The SARM1 TIR NADase: Mechanistic Similarities to Bacterial Phage Defense and Toxin-Antitoxin Systems. Front Immunol 2021; 12:752898. [PMID: 34630431 PMCID: PMC8494770 DOI: 10.3389/fimmu.2021.752898] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 08/26/2021] [Indexed: 01/19/2023] Open
Abstract
The Toll/interleukin-1 receptor (TIR) domain is the signature signalling motif of innate immunity, with essential roles in innate immune signalling in bacteria, plants, and animals. TIR domains canonically function as scaffolds, with stimulus-dependent multimerization generating binding sites for signalling molecules such as kinases and ligases that activate downstream immune mechanisms. Recent studies have dramatically expanded our understanding of the TIR domain, demonstrating that the primordial function of the TIR domain is to metabolize NAD+. Mammalian SARM1, the central executioner of pathological axon degeneration, is the founding member of the TIR-domain class of NAD+ hydrolases. This unexpected NADase activity of TIR domains is evolutionarily conserved, with archaeal, bacterial, and plant TIR domains all sharing this catalytic function. Moreover, this enzymatic activity is essential for the innate immune function of these proteins. These evolutionary relationships suggest a link between SARM1 and ancient self-defense mechanisms that has only been strengthened by the recent discovery of the SARM1 activation mechanism which, we will argue, is strikingly similar to bacterial toxin-antitoxin systems. In this brief review we will describe the regulation and function of SARM1 in programmed axon self-destruction, and highlight the parallels between the SARM1 axon degeneration pathway and bacterial innate immune mechanisms.
Collapse
Affiliation(s)
- Aaron DiAntonio
- Department of Developmental Biology, Washington University School of Medicine in Saint Louis, St. Louis, MO, United States
- Needleman Center for Neurometabolism and Axonal Therapeutics, Washington University School of Medicine in Saint Louis, St. Louis, MO, United States
| | - Jeffrey Milbrandt
- Needleman Center for Neurometabolism and Axonal Therapeutics, Washington University School of Medicine in Saint Louis, St. Louis, MO, United States
- Department of Genetics, Washington University School of Medicine in Saint Louis, St. Louis, MO, United States
| | - Matthew D Figley
- Department of Developmental Biology, Washington University School of Medicine in Saint Louis, St. Louis, MO, United States
| |
Collapse
|
44
|
SARM1 signaling mechanisms in the injured nervous system. Curr Opin Neurobiol 2021; 69:247-255. [PMID: 34175654 DOI: 10.1016/j.conb.2021.05.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/18/2021] [Accepted: 05/24/2021] [Indexed: 11/21/2022]
Abstract
Axon degeneration is a prominent feature of the injured nervous system, occurs across neurological diseases, and drives functional loss in neural circuits. We have seen a paradigm shift in the last decade with the realization that injured axons are capable of actively driving their own destruction through the sterile-alpha and TIR motif containing 1 (SARM1) protein. Early studies of Wallerian degeneration highlighted a central role for NAD+ metabolites in axon survival, and this association has grown even stronger in recent years with a deeper understanding of SARM1 biology. Here, we review our current knowledge of SARM1 function in vivo and our evolving understanding of its complex architecture and regulation by injury-dependent changes in the local metabolic environment. The field is converging on a model whereby SARM1 acts as a sensor for metabolic changes that occur after injury and then drives catastrophic NAD+ loss to promote degeneration. However, a number of observations suggest that SARM1 biology is more complicated, and there remains much to learn about how SARM1 governs nervous system responses to injury or disease.
Collapse
|
45
|
Abstract
Axon degeneration is an active program of self-destruction mediated by the protein SARM1. In healthy neurons, SARM1 is autoinhibited and, upon injury autoinhibition is relieved, activating the SARM1 enzyme to deplete NAD+ and induce axon degeneration. SARM1 forms a homomultimeric octamer with each monomer composed of an N-terminal autoinhibitory ARM domain, tandem SAM domains that mediate multimerization, and a C-terminal TIR domain encoding the NADase enzyme. Here we discovered multiple intramolecular and intermolecular domain interfaces required for SARM1 autoinhibition using peptide mapping and cryo-electron microscopy (cryo-EM). We identified a candidate autoinhibitory region by screening a panel of peptides derived from the SARM1 ARM domain, identifying a peptide mediating high-affinity inhibition of the SARM1 NADase. Mutation of residues in full-length SARM1 within the region encompassed by the peptide led to loss of autoinhibition, rendering SARM1 constitutively active and inducing spontaneous NAD+ and axon loss. The cryo-EM structure of SARM1 revealed 1) a compact autoinhibited SARM1 octamer in which the TIR domains are isolated and prevented from oligomerization and enzymatic activation and 2) multiple candidate autoinhibitory interfaces among the domains. Mutational analysis demonstrated that five distinct interfaces are required for autoinhibition, including intramolecular and intermolecular ARM-SAM interfaces, an intermolecular ARM-ARM interface, and two ARM-TIR interfaces formed between a single TIR and two distinct ARM domains. These autoinhibitory regions are not redundant, as point mutants in each led to constitutively active SARM1. These studies define the structural basis for SARM1 autoinhibition and may enable the development of SARM1 inhibitors that stabilize the autoinhibited state.
Collapse
|
46
|
Abstract
The outer retina is nourished from the choroid, a capillary bed just inside the sclera. O2, glucose, and other nutrients diffuse out of the choroid and then filter through a monolayer of retinal pigment epithelium (RPE) cells to fuel the retina. Recent studies of energy metabolism have revealed striking differences between retinas and RPE cells in the ways that they extract energy from fuels. The purpose of this review is to suggest and evaluate the hypothesis that the retina and RPE have complementary metabolic roles that make them depend on each other for survival and for their abilities to perform essential and specialized functions. Expected final online publication date for the Annual Review of Vision Science, Volume 7 is September 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- James B Hurley
- Departments of Biochemistry and Ophthalmology, University of Washington, Seattle, Washington 98115, USA;
| |
Collapse
|
47
|
Figley MD, Gu W, Nanson JD, Shi Y, Sasaki Y, Cunnea K, Malde AK, Jia X, Luo Z, Saikot FK, Mosaiab T, Masic V, Holt S, Hartley-Tassell L, McGuinness HY, Manik MK, Bosanac T, Landsberg MJ, Kerry PS, Mobli M, Hughes RO, Milbrandt J, Kobe B, DiAntonio A, Ve T. SARM1 is a metabolic sensor activated by an increased NMN/NAD + ratio to trigger axon degeneration. Neuron 2021; 109:1118-1136.e11. [PMID: 33657413 PMCID: PMC8174188 DOI: 10.1016/j.neuron.2021.02.009] [Citation(s) in RCA: 184] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 01/15/2021] [Accepted: 02/08/2021] [Indexed: 12/16/2022]
Abstract
Axon degeneration is a central pathological feature of many neurodegenerative diseases. Sterile alpha and Toll/interleukin-1 receptor motif-containing 1 (SARM1) is a nicotinamide adenine dinucleotide (NAD+)-cleaving enzyme whose activation triggers axon destruction. Loss of the biosynthetic enzyme NMNAT2, which converts nicotinamide mononucleotide (NMN) to NAD+, activates SARM1 via an unknown mechanism. Using structural, biochemical, biophysical, and cellular assays, we demonstrate that SARM1 is activated by an increase in the ratio of NMN to NAD+ and show that both metabolites compete for binding to the auto-inhibitory N-terminal armadillo repeat (ARM) domain of SARM1. We report structures of the SARM1 ARM domain bound to NMN and of the homo-octameric SARM1 complex in the absence of ligands. We show that NMN influences the structure of SARM1 and demonstrate via mutagenesis that NMN binding is required for injury-induced SARM1 activation and axon destruction. Hence, SARM1 is a metabolic sensor responding to an increased NMN/NAD+ ratio by cleaving residual NAD+, thereby inducing feedforward metabolic catastrophe and axonal demise.
Collapse
Affiliation(s)
- Matthew D Figley
- Department of Developmental Biology, Washington University School of Medicine in Saint Louis, St. Louis, MO, USA; Needleman Center for Neurometabolism and Axonal Therapeutics, Washington University School of Medicine in Saint Louis, St. Louis, MO, USA
| | - Weixi Gu
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD 4072, Australia
| | - Jeffrey D Nanson
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD 4072, Australia
| | - Yun Shi
- Institute for Glycomics, Griffith University, Southport, QLD 4222, Australia
| | - Yo Sasaki
- Needleman Center for Neurometabolism and Axonal Therapeutics, Washington University School of Medicine in Saint Louis, St. Louis, MO, USA; Department of Genetics, Washington University School of Medicine in Saint Louis, St. Louis, MO, USA
| | - Katie Cunnea
- Evotec (UK) Ltd., 114 Innovation Drive, Milton Park, Abingdon, Oxfordshire OX14 4RZ, UK; Evotec SE, Manfred Eigen Campus, Essener Bogen 7, 22419 Hamburg, Germany
| | - Alpeshkumar K Malde
- Institute for Glycomics, Griffith University, Southport, QLD 4222, Australia
| | - Xinying Jia
- Centre for Advanced Imaging, University of Queensland, Brisbane, QLD 4072, Australia
| | - Zhenyao Luo
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD 4072, Australia
| | - Forhad K Saikot
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD 4072, Australia
| | - Tamim Mosaiab
- Institute for Glycomics, Griffith University, Southport, QLD 4222, Australia
| | - Veronika Masic
- Institute for Glycomics, Griffith University, Southport, QLD 4222, Australia
| | - Stephanie Holt
- Institute for Glycomics, Griffith University, Southport, QLD 4222, Australia
| | | | - Helen Y McGuinness
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD 4072, Australia
| | - Mohammad K Manik
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD 4072, Australia
| | - Todd Bosanac
- Disarm Therapeutics, a wholly owned subsidiary of Eli Lilly & Co., Cambridge, MA, USA
| | - Michael J Landsberg
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD 4072, Australia
| | - Philip S Kerry
- Evotec (UK) Ltd., 114 Innovation Drive, Milton Park, Abingdon, Oxfordshire OX14 4RZ, UK; Evotec SE, Manfred Eigen Campus, Essener Bogen 7, 22419 Hamburg, Germany
| | - Mehdi Mobli
- Centre for Advanced Imaging, University of Queensland, Brisbane, QLD 4072, Australia
| | - Robert O Hughes
- Disarm Therapeutics, a wholly owned subsidiary of Eli Lilly & Co., Cambridge, MA, USA
| | - Jeffrey Milbrandt
- Needleman Center for Neurometabolism and Axonal Therapeutics, Washington University School of Medicine in Saint Louis, St. Louis, MO, USA; Department of Genetics, Washington University School of Medicine in Saint Louis, St. Louis, MO, USA.
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD 4072, Australia.
| | - Aaron DiAntonio
- Department of Developmental Biology, Washington University School of Medicine in Saint Louis, St. Louis, MO, USA; Needleman Center for Neurometabolism and Axonal Therapeutics, Washington University School of Medicine in Saint Louis, St. Louis, MO, USA.
| | - Thomas Ve
- Institute for Glycomics, Griffith University, Southport, QLD 4222, Australia.
| |
Collapse
|
48
|
Asgharsharghi A, Tian W, Haehnel-Taguchi M, López-Schier H. Sarm1 is dispensable for mechanosensory-motor transformations in zebrafish. MICROPUBLICATION BIOLOGY 2021; 2021. [PMID: 33688624 PMCID: PMC7930918 DOI: 10.17912/micropub.biology.000369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Sarm1 is an evolutionary conserved protein that is essential for Wallerian axon degeneration. Sarm1 has emerged as a therapeutic target to treat neuropathies derived from metabolic or chemical stress and physical injury of axons. Yet, the full repertoire of consequences of inhibiting Sarm1 remains unknown. Here we show that loss of Sarm1 in zebrafish does not affect the sensorimotor transformations that underlie rheotaxis. In addition, Sarm1 deficit accelerates the re-growth of regenerating axons. These data indicate that systemic inhibition of Sarm1 is a viable therapeutic option compatible with sustained nervous system function.
Collapse
Affiliation(s)
| | - Weili Tian
- Helmholtz Zentrum Munich, Munich, Germany
| | | | | |
Collapse
|
49
|
Pan WW, Wubben TJ, Besirli CG. Photoreceptor metabolic reprogramming: current understanding and therapeutic implications. Commun Biol 2021; 4:245. [PMID: 33627778 PMCID: PMC7904922 DOI: 10.1038/s42003-021-01765-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 01/28/2021] [Indexed: 02/06/2023] Open
Abstract
Acquired and inherited retinal disorders are responsible for vision loss in an increasing proportion of individuals worldwide. Photoreceptor (PR) death is central to the vision loss individuals experience in these various retinal diseases. Unfortunately, there is a lack of treatment options to prevent PR loss, so an urgent unmet need exists for therapies that improve PR survival and ultimately, vision. The retina is one of the most energy demanding tissues in the body, and this is driven in large part by the metabolic needs of PRs. Recent studies suggest that disruption of nutrient availability and regulation of cell metabolism may be a unifying mechanism in PR death. Understanding retinal cell metabolism and how it is altered in disease has been identified as a priority area of research. The focus of this review is on the recent advances in the understanding of PR metabolism and how it is critical to reduction-oxidation (redox) balance, the outer retinal metabolic ecosystem, and retinal disease. The importance of these metabolic processes is just beginning to be realized and unraveling the metabolic and redox pathways integral to PR health may identify novel targets for neuroprotective strategies that prevent blindness in the heterogenous group of retinal disorders.
Collapse
Affiliation(s)
- Warren W Pan
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, USA
| | - Thomas J Wubben
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, USA.
| | - Cagri G Besirli
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
50
|
Chen YH, Sasaki Y, DiAntonio A, Milbrandt J. SARM1 is required in human derived sensory neurons for injury-induced and neurotoxic axon degeneration. Exp Neurol 2021; 339:113636. [PMID: 33548217 DOI: 10.1016/j.expneurol.2021.113636] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 01/12/2021] [Accepted: 01/31/2021] [Indexed: 11/16/2022]
Abstract
Axonal degeneration contributes to the pathogenesis of many neurodegenerative disorders, motivating efforts to dissect the mechanism of pathological axon loss in order to develop therapies for axonal preservation. SARM1 is a particularly attractive therapeutic target, as it is an inducible NAD+ cleaving enzyme that is required for axon loss in multiple mouse models of traumatic and degenerative neurological disease. However, it is essential to establish whether SARM1 triggers axon degeneration in human neurons before proceeding with the development of SARM1-directed therapeutics. Here we combine genome engineering with the production of human stem cell-derived neurons to test the role of human SARM1 in traumatic and neurotoxic axon degeneration. We have generated two independent SARM1 knockout human iPSC lines that do not express SARM1 protein upon differentiation into neurons. We have developed a modified sensory neuron differentiation protocol that generates human sensory neurons with high yield and purity. We find that SARM1 is required for axon degeneration in response to both physical trauma and in a cellular model of chemotherapy-induced peripheral neuropathy. Finally, we identify cADPR as a biomarker of SARM1 enzyme activity in both healthy and injured human sensory neurons. These findings are consistent with prior molecular and cellular studies in mouse neurons, and highlight the therapeutic potential of SARM1 inhibition for the prevention and treatment of human neurological disease.
Collapse
Affiliation(s)
- Yi-Hsien Chen
- Needleman Center for Neurometabolism and Axonal Therapeutics, Washington University School of Medicine, St. Louis, MO 63110, USA; Genome Engineering and iPSC Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yo Sasaki
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Aaron DiAntonio
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Needleman Center for Neurometabolism and Axonal Therapeutics, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Jeffrey Milbrandt
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Needleman Center for Neurometabolism and Axonal Therapeutics, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|