1
|
Chen X, Lv S, Liu J, Guan Y, Xu C, Ma X, Li M, Bai X, Liu K, Zhang H, Yan Q, Zhou F, Chen Y. Exploring the Role of Axons in ALS from Multiple Perspectives. Cells 2024; 13:2076. [PMID: 39768167 PMCID: PMC11674045 DOI: 10.3390/cells13242076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Amyotrophic lateral sclerosis (ALS), commonly known as motor neuron disease, is a neurodegenerative disorder characterized by the progressive degeneration of both upper and lower motor neurons. This pathological process results in muscle weakness and can culminate in paralysis. To date, the precise etiology of ALS remains unclear. However, a burgeoning body of research indicates that axonal dysfunction is a pivotal element in the pathogenesis of ALS and significantly influences the progression of disease. Dysfunction of axons in ALS can result in impediments to nerve impulse transmission, leading to motor impairment, muscle atrophy, and other associated complications that severely compromise patients' quality of life and survival prognosis. In this review, we concentrate on several key areas: the ultrastructure of axons, the mechanisms of axonal degeneration in ALS, the impact of impaired axonal transport on disease progression in ALS, and the potential for axonal regeneration within the central nervous system (CNS). Our objective is to achieve a more holistic and profound understanding of the multifaceted role that axons play in ALS, thereby offering a more intricate and refined perspective on targeted axonal therapeutic interventions.
Collapse
Affiliation(s)
- Xiaosu Chen
- Department of Histology and Embryology, Shandong Second Medical University, Weifang 261053, China; (X.C.); (S.L.); (C.X.); (X.M.); (X.B.)
- Neurologic Disorders and Regenerative Repair Lab of Shandong Higher Education, Shandong Second Medical University, Weifang 261053, China; (J.L.); (M.L.); (K.L.); (H.Z.); (Q.Y.); (F.Z.)
| | - Shuchang Lv
- Department of Histology and Embryology, Shandong Second Medical University, Weifang 261053, China; (X.C.); (S.L.); (C.X.); (X.M.); (X.B.)
- Neurologic Disorders and Regenerative Repair Lab of Shandong Higher Education, Shandong Second Medical University, Weifang 261053, China; (J.L.); (M.L.); (K.L.); (H.Z.); (Q.Y.); (F.Z.)
| | - Jinmeng Liu
- Neurologic Disorders and Regenerative Repair Lab of Shandong Higher Education, Shandong Second Medical University, Weifang 261053, China; (J.L.); (M.L.); (K.L.); (H.Z.); (Q.Y.); (F.Z.)
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao 266000, China
| | - Yingjun Guan
- Department of Histology and Embryology, Shandong Second Medical University, Weifang 261053, China; (X.C.); (S.L.); (C.X.); (X.M.); (X.B.)
- Neurologic Disorders and Regenerative Repair Lab of Shandong Higher Education, Shandong Second Medical University, Weifang 261053, China; (J.L.); (M.L.); (K.L.); (H.Z.); (Q.Y.); (F.Z.)
| | - Chunjie Xu
- Department of Histology and Embryology, Shandong Second Medical University, Weifang 261053, China; (X.C.); (S.L.); (C.X.); (X.M.); (X.B.)
- Neurologic Disorders and Regenerative Repair Lab of Shandong Higher Education, Shandong Second Medical University, Weifang 261053, China; (J.L.); (M.L.); (K.L.); (H.Z.); (Q.Y.); (F.Z.)
| | - Xiaonan Ma
- Department of Histology and Embryology, Shandong Second Medical University, Weifang 261053, China; (X.C.); (S.L.); (C.X.); (X.M.); (X.B.)
- Neurologic Disorders and Regenerative Repair Lab of Shandong Higher Education, Shandong Second Medical University, Weifang 261053, China; (J.L.); (M.L.); (K.L.); (H.Z.); (Q.Y.); (F.Z.)
| | - Mu Li
- Neurologic Disorders and Regenerative Repair Lab of Shandong Higher Education, Shandong Second Medical University, Weifang 261053, China; (J.L.); (M.L.); (K.L.); (H.Z.); (Q.Y.); (F.Z.)
| | - Xue Bai
- Department of Histology and Embryology, Shandong Second Medical University, Weifang 261053, China; (X.C.); (S.L.); (C.X.); (X.M.); (X.B.)
- Neurologic Disorders and Regenerative Repair Lab of Shandong Higher Education, Shandong Second Medical University, Weifang 261053, China; (J.L.); (M.L.); (K.L.); (H.Z.); (Q.Y.); (F.Z.)
| | - Kexin Liu
- Neurologic Disorders and Regenerative Repair Lab of Shandong Higher Education, Shandong Second Medical University, Weifang 261053, China; (J.L.); (M.L.); (K.L.); (H.Z.); (Q.Y.); (F.Z.)
| | - Haoyun Zhang
- Neurologic Disorders and Regenerative Repair Lab of Shandong Higher Education, Shandong Second Medical University, Weifang 261053, China; (J.L.); (M.L.); (K.L.); (H.Z.); (Q.Y.); (F.Z.)
| | - Qiupeng Yan
- Neurologic Disorders and Regenerative Repair Lab of Shandong Higher Education, Shandong Second Medical University, Weifang 261053, China; (J.L.); (M.L.); (K.L.); (H.Z.); (Q.Y.); (F.Z.)
| | - Fenghua Zhou
- Neurologic Disorders and Regenerative Repair Lab of Shandong Higher Education, Shandong Second Medical University, Weifang 261053, China; (J.L.); (M.L.); (K.L.); (H.Z.); (Q.Y.); (F.Z.)
| | - Yanchun Chen
- Department of Histology and Embryology, Shandong Second Medical University, Weifang 261053, China; (X.C.); (S.L.); (C.X.); (X.M.); (X.B.)
- Neurologic Disorders and Regenerative Repair Lab of Shandong Higher Education, Shandong Second Medical University, Weifang 261053, China; (J.L.); (M.L.); (K.L.); (H.Z.); (Q.Y.); (F.Z.)
| |
Collapse
|
2
|
Marín-Blázquez M, Rovira J, Ramírez-Bajo MJ, Zapata-Pérez R, Rabadán-Ros R. NAD + enhancers as therapeutic agents in the cardiorenal axis. Cell Commun Signal 2024; 22:537. [PMID: 39516787 PMCID: PMC11546376 DOI: 10.1186/s12964-024-01903-4] [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: 08/01/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Cardiorenal diseases represent a complex interplay between heart failure and renal dysfunction, being clinically classified as cardiorenal syndromes (CRS). Recently, the contributions of altered nicotinamide adenine dinucleotide (NAD+) metabolism, through deficient NAD+ synthesis and/or elevated consumption, have proved to be decisive in the onset and progress of cardiorenal disease. NAD+ is a pivotal coenzyme in cellular metabolism, being significant in various signaling pathways, such as energy metabolism, DNA damage repair, gene expression, and stress response. Convincing evidence suggests that strategies designed to boost cellular NAD+ levels are a promising therapeutic option to address cardiovascular and renal disorders. Here, we review and discuss the implications of NAD+ metabolism in cardiorenal diseases, focusing on the propitious NAD+ boosting therapeutic strategies, based on the use of NAD+ precursors, poly(ADP-ribose) polymerase inhibitors, sirtuin activators, and other alternative approaches, such as CD38 blockade, nicotinamide phosphoribosyltransferase activation and combined interventions.
Collapse
Affiliation(s)
- Mariano Marín-Blázquez
- Group of Metabolism and Genetic Regulation of Disease, UCAM HiTech Sport & Health Innovation Hub, Universidad Católica de Murcia, 30107 Guadalupe de Maciascoque, Murcia, Spain
| | - Jordi Rovira
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Casanova 143 CRB CELLEX sector 2B, Barcelona, 08036, Spain
- Red de Investigación Cooperativa Orientada a Resultados en Salud (RICORS 2040), Madrid, Spain
| | - María José Ramírez-Bajo
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Casanova 143 CRB CELLEX sector 2B, Barcelona, 08036, Spain
- Red de Investigación Cooperativa Orientada a Resultados en Salud (RICORS 2040), Madrid, Spain
| | - Rubén Zapata-Pérez
- Group of Metabolism and Genetic Regulation of Disease, UCAM HiTech Sport & Health Innovation Hub, Universidad Católica de Murcia, 30107 Guadalupe de Maciascoque, Murcia, Spain.
| | - Rubén Rabadán-Ros
- Group of Metabolism and Genetic Regulation of Disease, UCAM HiTech Sport & Health Innovation Hub, Universidad Católica de Murcia, 30107 Guadalupe de Maciascoque, Murcia, Spain.
| |
Collapse
|
3
|
Krukenberg S, Möckl F, Weiß M, Dekiert P, Hofmann M, Gerlach F, Winterberg KJ, Kovacevic D, Khansahib I, Troost B, Hinrichs M, Granato V, Nawrocki M, Hub T, Tsvilovskyy V, Medert R, Woelk LM, Förster F, Li H, Werner R, Altfeld M, Huber S, Clarke OB, Freichel M, Diercks BP, Meier C, Guse AH. MASTER-NAADP: a membrane permeable precursor of the Ca 2+ mobilizing second messenger NAADP. Nat Commun 2024; 15:8008. [PMID: 39271671 PMCID: PMC11399135 DOI: 10.1038/s41467-024-52024-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 08/20/2024] [Indexed: 09/15/2024] Open
Abstract
Upon stimulation of membrane receptors, nicotinic acid adenine dinucleotide phosphate (NAADP) is formed as second messenger within seconds and evokes Ca2+ signaling in many different cell types. Here, to directly stimulate NAADP signaling, MASTER-NAADP, a Membrane permeAble, STabilized, bio-rEversibly pRotected precursor of NAADP is synthesized and release of its active NAADP mimetic, benzoic acid C-nucleoside, 2'-phospho-3'F-adenosine-diphosphate, by esterase digestion is confirmed. In the presence of NAADP receptor HN1L/JPT2 (hematological and neurological expressed 1-like protein, HN1L, also known as Jupiter microtubule-associated homolog 2, JPT2), this active NAADP mimetic releases Ca2+ and increases the open probability of type 1 ryanodine receptor. When added to intact cells, MASTER-NAADP initially evokes single local Ca2+ signals of low amplitude. Subsequently, also global Ca2+ signaling is observed in T cells, natural killer cells, and Neuro2A cells. In contrast, control compound MASTER-NADP does not stimulate Ca2+ signaling. Likewise, in cells devoid of HN1L/JPT2, MASTER-NAADP does not affect Ca2+ signaling, confirming that the product released from MASTER-NAADP is a bona fide NAADP mimetic.
Collapse
Affiliation(s)
- Sarah Krukenberg
- Organic Chemistry, University of Hamburg, 20146, Hamburg, Germany
| | - Franziska Möckl
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Mariella Weiß
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Patrick Dekiert
- Organic Chemistry, University of Hamburg, 20146, Hamburg, Germany
| | - Melanie Hofmann
- Organic Chemistry, University of Hamburg, 20146, Hamburg, Germany
| | - Fynn Gerlach
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Kai J Winterberg
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Dejan Kovacevic
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Imrankhan Khansahib
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Berit Troost
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Macarena Hinrichs
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Viviana Granato
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Mikolaj Nawrocki
- Section of Molecular Immunology and Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Tobis Hub
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Volodymyr Tsvilovskyy
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Rebekka Medert
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Lena-Marie Woelk
- Department of Applied Medical Informatics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Fritz Förster
- Department of Applied Medical Informatics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Huan Li
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - René Werner
- Department of Applied Medical Informatics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Marcus Altfeld
- Department of Immunology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Samuel Huber
- Section of Molecular Immunology and Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Oliver Biggs Clarke
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Marc Freichel
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Björn-Philipp Diercks
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Chris Meier
- Organic Chemistry, University of Hamburg, 20146, Hamburg, Germany
| | - Andreas H Guse
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.
| |
Collapse
|
4
|
McGuinness HY, Gu W, Shi Y, Kobe B, Ve T. SARM1-Dependent Axon Degeneration: Nucleotide Signaling, Neurodegenerative Disorders, Toxicity, and Therapeutic Opportunities. Neuroscientist 2024; 30:473-492. [PMID: 37002660 PMCID: PMC11282687 DOI: 10.1177/10738584231162508] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
Axons are an essential component of the nervous system, and axon degeneration is an early feature of many neurodegenerative disorders. The NAD+ metabolome plays an essential role in regulating axonal integrity. Axonal levels of NAD+ and its precursor NMN are controlled in large part by the NAD+ synthesizing survival factor NMNAT2 and the pro-neurodegenerative NADase SARM1, whose activation triggers axon destruction. SARM1 has emerged as a promising axon-specific target for therapeutic intervention, and its function, regulation, structure, and role in neurodegenerative diseases have been extensively characterized in recent years. In this review, we first introduce the key molecular players involved in the SARM1-dependent axon degeneration program. Next, we summarize recent major advances in our understanding of how SARM1 is kept inactive in healthy neurons and how it becomes activated in injured or diseased neurons, which has involved important insights from structural biology. Finally, we discuss the role of SARM1 in neurodegenerative disorders and environmental neurotoxicity and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Helen Y. McGuinness
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Saint Lucia, Australia
| | - Weixi Gu
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Saint Lucia, Australia
| | - Yun Shi
- Institute for Glycomics, Griffith University, Gold Coast, Australia
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Saint Lucia, Australia
| | - Thomas Ve
- Institute for Glycomics, Griffith University, Gold Coast, Australia
| |
Collapse
|
5
|
Choi S, Lee Y, Park S, Jang SY, Park J, Oh DW, Kim SM, Kim TH, Lee GS, Cho C, Kim BS, Lee D, Kim EH, Cheong HK, Moon JH, Song JJ, Hwang J, Kim MH. Dissemination of pathogenic bacteria is reinforced by a MARTX toxin effector duet. Nat Commun 2024; 15:6218. [PMID: 39043696 PMCID: PMC11266601 DOI: 10.1038/s41467-024-50650-0] [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: 02/02/2024] [Accepted: 07/15/2024] [Indexed: 07/25/2024] Open
Abstract
Multiple bacterial genera take advantage of the multifunctional autoprocessing repeats-in-toxin (MARTX) toxin to invade host cells. Secretion of the MARTX toxin by Vibrio vulnificus, a deadly opportunistic pathogen that causes primary septicemia, the precursor of sepsis, is a major driver of infection; however, the molecular mechanism via which the toxin contributes to septicemia remains unclear. Here, we report the crystal and cryo-electron microscopy (EM) structures of a toxin effector duet comprising the domain of unknown function in the first position (DUF1)/Rho inactivation domain (RID) complexed with human targets. These structures reveal how the duet is used by bacteria as a potent weapon. The data show that DUF1 acts as a RID-dependent transforming NADase domain (RDTND) that disrupts NAD+ homeostasis by hijacking calmodulin. The cryo-EM structure of the RDTND-RID duet complexed with calmodulin and Rac1, together with immunological analyses in vitro and in mice, provide mechanistic insight into how V. vulnificus uses the duet to suppress ROS generation by depleting NAD(P)+ and modifying Rac1 in a mutually-reinforcing manner that ultimately paralyzes first line immune responses, promotes dissemination of invaders, and induces sepsis. These data may allow development of tools or strategies to combat MARTX toxin-related human diseases.
Collapse
Affiliation(s)
- Sanghyeon Choi
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Korea
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
| | - Youngjin Lee
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
| | - Shinhye Park
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, 34134, Korea
| | - Song Yee Jang
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
- Core Research Facility & Analysis Center, KRIBB, Daejeon, 34141, Korea
| | - Jongbin Park
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
| | - Do Won Oh
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, Korea
| | - Su-Man Kim
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
- Department of Biology Education, Chonnam National University, Gwangju, 61186, Korea
| | - Tae-Hwan Kim
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
- College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, Korea
| | - Ga Seul Lee
- Core Research Facility & Analysis Center, KRIBB, Daejeon, 34141, Korea
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk, 28644, Korea
| | - Changyi Cho
- Department of Food Science and Biotechnology, Ewha Womans University, Seoul, 03760, Korea
| | - Byoung Sik Kim
- Department of Food Science and Biotechnology, Ewha Womans University, Seoul, 03760, Korea
| | - Donghan Lee
- Korea Basic Science Institute, Cheongju, Chungbuk, 28119, Korea
| | - Eun-Hee Kim
- Korea Basic Science Institute, Cheongju, Chungbuk, 28119, Korea
| | - Hae-Kap Cheong
- Korea Basic Science Institute, Cheongju, Chungbuk, 28119, Korea
| | - Jeong Hee Moon
- Core Research Facility & Analysis Center, KRIBB, Daejeon, 34141, Korea
| | - Ji-Joon Song
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Korea
| | - Jungwon Hwang
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea.
| | - Myung Hee Kim
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea.
| |
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
|
Chakraborty J. A comprehensive review of soybean RNL and TIR domain proteins. PLANT MOLECULAR BIOLOGY 2024; 114:78. [PMID: 38922375 DOI: 10.1007/s11103-024-01473-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 05/29/2024] [Indexed: 06/27/2024]
Abstract
Both prokaryotic and eukaryotic organisms use the nucleotide-binding domain/leucine-rich repeat (NBD/LRR)-triggered immunity (NLR-triggered immunity) signaling pathway to defend against pathogens. Plant NLRs are intracellular immune receptors that can bind to effector proteins secreted by pathogens. Dicotyledonous plants express a type of NLR known as TIR domain-containing NLRs (TNLs). TIR domains are enzymes that catalyze the production of small molecules that are essential for immune signaling and lead to plant cell death. The activation of downstream TNL signaling components, such as enhanced disease susceptibility 1 (EDS1), phytoalexin deficient 4 (PAD4), and senescence-associated gene 101 (SAG101), is facilitated by these small molecules. Helper NLRs (hNLRs) and the EDS1-PAD4/SAG101 complex associate after activation, causing the hNLRs to oligomerize, translocate to the plasma membrane (PM), and produce cation-selective channels. According to a recent theory, cations enter cells through pores created by oligomeric hNLRs and trigger cell death. Occasionally, TNLs can self-associate to create higher-order oligomers. Here, we categorized soybean TNLs based on the protein domains that they possess. We believe that TNLs may help soybean plants effectively fight pathogens by acting as a source of genetic resistance. In summary, the purpose of this review is to elucidate the range of TNLs that are expressed in soybean.
Collapse
Affiliation(s)
- Joydeep Chakraborty
- School of Plant Sciences and Food Security, Tel Aviv University, Tel-Aviv, Israel.
| |
Collapse
|
8
|
Hinz FI, Villegas CLM, Roberts JT, Yao H, Gaddam S, Delwig A, Green SA, Fredrickson C, Adrian M, Asuncion RR, Cheung TK, Hayne M, Hackos DH, Rose CM, Richmond D, Hoogenraad CC. Context-Specific Stress Causes Compartmentalized SARM1 Activation and Local Degeneration in Cortical Neurons. J Neurosci 2024; 44:e2424232024. [PMID: 38692735 PMCID: PMC11170950 DOI: 10.1523/jneurosci.2424-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 04/21/2024] [Accepted: 04/23/2024] [Indexed: 05/03/2024] Open
Abstract
Sterile alpha and TIR motif containing 1 (SARM1) is an inducible NADase that localizes to mitochondria throughout neurons and senses metabolic changes that occur after injury. Minimal proteomic changes are observed upon either SARM1 depletion or activation, suggesting that SARM1 does not exert broad effects on neuronal protein homeostasis. However, whether SARM1 activation occurs throughout the neuron in response to injury and cell stress remains largely unknown. Using a semiautomated imaging pipeline and a custom-built deep learning scoring algorithm, we studied degeneration in both mixed-sex mouse primary cortical neurons and male human-induced pluripotent stem cell-derived cortical neurons in response to a number of different stressors. We show that SARM1 activation is differentially restricted to specific neuronal compartments depending on the stressor. Cortical neurons undergo SARM1-dependent axon degeneration after mechanical transection, and SARM1 activation is limited to the axonal compartment distal to the injury site. However, global SARM1 activation following vacor treatment causes both cell body and axon degeneration. Context-specific stressors, such as microtubule dysfunction and mitochondrial stress, induce axonal SARM1 activation leading to SARM1-dependent axon degeneration and SARM1-independent cell body death. Our data reveal that compartment-specific SARM1-mediated death signaling is dependent on the type of injury and cellular stressor.
Collapse
Affiliation(s)
- Flora I Hinz
- Department of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | | | - Jasmine T Roberts
- Department of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Heming Yao
- Biological Research | AI Development, Genentech, Inc., South San Francisco, California 94080
| | - Shreya Gaddam
- Biological Research | AI Development, Genentech, Inc., South San Francisco, California 94080
| | - Anton Delwig
- Departments of Biochemical and Cellular Pharmacology, Genentech, Inc., South San Francisco, California 94080
| | - Samantha A Green
- Discovery Chemistry, Genentech, Inc., South San Francisco, California 94080
| | - Craig Fredrickson
- Department of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Max Adrian
- Pathology, Genentech, Inc., South San Francisco, California 94080
| | - Raymond R Asuncion
- Transgenic Technology, Genentech, Inc., South San Francisco, California 94080
| | - Tommy K Cheung
- Microchemistry, Proteomics, and Lipidomics, Genentech, Inc., South San Francisco, California 94080
| | - Margaret Hayne
- Department of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - David H Hackos
- Department of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Christopher M Rose
- Microchemistry, Proteomics, and Lipidomics, Genentech, Inc., South San Francisco, California 94080
| | - David Richmond
- Biological Research | AI Development, Genentech, Inc., South San Francisco, California 94080
| | - Casper C Hoogenraad
- Department of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| |
Collapse
|
9
|
Velma G, Krider IS, Alves ETM, Courey JM, Laham MS, Thatcher GRJ. Channeling Nicotinamide Phosphoribosyltransferase (NAMPT) to Address Life and Death. J Med Chem 2024; 67:5999-6026. [PMID: 38580317 PMCID: PMC11056997 DOI: 10.1021/acs.jmedchem.3c02112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 02/22/2024] [Accepted: 03/11/2024] [Indexed: 04/07/2024]
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT) catalyzes the rate-limiting step in NAD+ biosynthesis via salvage of NAM formed from catabolism of NAD+ by proteins with NADase activity (e.g., PARPs, SIRTs, CD38). Depletion of NAD+ in aging, neurodegeneration, and metabolic disorders is addressed by NAD+ supplementation. Conversely, NAMPT inhibitors have been developed for cancer therapy: many discovered by phenotypic screening for cancer cell death have low nanomolar potency in cellular models. No NAMPT inhibitor is yet FDA-approved. The ability of inhibitors to act as NAMPT substrates may be associated with efficacy and toxicity. Some 3-pyridyl inhibitors become 4-pyridyl activators or "NAD+ boosters". NAMPT positive allosteric modulators (N-PAMs) and boosters may increase enzyme activity by relieving substrate/product inhibition. Binding to a "rear channel" extending from the NAMPT active site is key for inhibitors, boosters, and N-PAMs. A deeper understanding may fulfill the potential of NAMPT ligands to regulate cellular life and death.
Collapse
Affiliation(s)
- Ganga
Reddy Velma
- Department
of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, Arizona 85721, United States
| | - Isabella S. Krider
- Department
of Chemistry & Biochemistry, University
of Arizona, Tucson, Arizona 85721, United States
| | - Erick T. M. Alves
- Department
of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, Arizona 85721, United States
| | - Jenna M. Courey
- Department
of Chemistry & Biochemistry, University
of Arizona, Tucson, Arizona 85721, United States
| | - Megan S. Laham
- Department
of Chemistry & Biochemistry, University
of Arizona, Tucson, Arizona 85721, United States
| | - Gregory R. J. Thatcher
- Department
of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, Arizona 85721, United States
- Department
of Chemistry & Biochemistry, University
of Arizona, Tucson, Arizona 85721, United States
| |
Collapse
|
10
|
Wenzl SJ, de Oliveira Mann CC. How enzyme-centered approaches are advancing research on cyclic oligo-nucleotides. FEBS Lett 2024; 598:839-863. [PMID: 38453162 DOI: 10.1002/1873-3468.14838] [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: 12/03/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 03/09/2024]
Abstract
Cyclic nucleotides are the most diversified category of second messengers and are found in all organisms modulating diverse pathways. While cAMP and cGMP have been studied over 50 years, cyclic di-nucleotide signaling in eukaryotes emerged only recently with the anti-viral molecule 2´3´cGAMP. Recent breakthrough discoveries have revealed not only the astonishing chemical diversity of cyclic nucleotides but also surprisingly deep-rooted evolutionary origins of cyclic oligo-nucleotide signaling pathways and structural conservation of the proteins involved in their synthesis and signaling. Here we discuss how enzyme-centered approaches have paved the way for the identification of several cyclic nucleotide signals, focusing on the advantages and challenges associated with deciphering the activation mechanisms of such enzymes.
Collapse
Affiliation(s)
- Simon J Wenzl
- Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, Garching, Germany
| | - Carina C de Oliveira Mann
- Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, Garching, Germany
| |
Collapse
|
11
|
Norambuena-Soto I, Deng Y, Brenner C, Lavandero S, Wang ZV. NAD in pathological cardiac remodeling: Metabolic regulation and beyond. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167038. [PMID: 38281710 PMCID: PMC10922927 DOI: 10.1016/j.bbadis.2024.167038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 01/05/2024] [Accepted: 01/19/2024] [Indexed: 01/30/2024]
Abstract
Nicotinamide adenine dinucleotide (NAD) coenzymes are carriers of high energy electrons in metabolism and also play critical roles in numerous signaling pathways. NAD metabolism is decreased in various cardiovascular diseases. Importantly, stimulation of NAD biosynthesis protects against heart disease under different pathological conditions. In this review, we describe pathways for both generation and catabolism of NAD coenzymes and the respective changes of these pathways in the heart under cardiac diseases, including pressure overload, myocardial infarction, cardiometabolic disease, cancer treatment cardiotoxicity, and heart failure. We next provide an update on the strategies and treatments to increase NAD levels, such as supplementation of NAD precursors, in the heart that prevent or reverse cardiomyopathy. We also introduce the approaches to manipulate NAD consumption enzymes to ameliorate cardiac disease. Finally, we discuss the mechanisms associated with improvements in cardiac function by NAD coenzymes, differentiating between mitochondria-dependent effects and those independent of mitochondrial metabolism.
Collapse
Affiliation(s)
- Ignacio Norambuena-Soto
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA; Advanced Center for Chronic Diseases (ACCDiS), Facultad Ciencias Químicas y Farmacéuticas & Facultad Medicina, Universidad de Chile, Santiago 8380494, Chile
| | - Yingfeng Deng
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Charles Brenner
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases (ACCDiS), Facultad Ciencias Químicas y Farmacéuticas & Facultad Medicina, Universidad de Chile, Santiago 8380494, Chile; Cardiology Division, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA.
| | - Zhao V Wang
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA.
| |
Collapse
|
12
|
Cirilli I, Amici A, Gilley J, Coleman MP, Orsomando G. Adaptation of a Commercial NAD + Quantification Kit to Assay the Base-Exchange Activity and Substrate Preferences of SARM1. Molecules 2024; 29:847. [PMID: 38398599 PMCID: PMC10891823 DOI: 10.3390/molecules29040847] [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: 01/10/2024] [Revised: 02/06/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Here, we report an adapted protocol using the Promega NAD/NADH-Glo™ Assay kit. The assay normally allows quantification of trace amounts of both oxidized and reduced forms of nicotinamide adenine dinucleotide (NAD) by enzymatic cycling, but we now show that the NAD analog 3-acetylpyridine adenine dinucleotide (AcPyrAD) also acts as a substrate for this enzyme-cycling assay. In fact, AcPyrAD generates amplification signals of a larger amplitude than those obtained with NAD. We exploited this finding to devise and validate a novel method for assaying the base-exchange activity of SARM1 in reactions containing NAD and an excess of the free base 3-acetylpyridine (AcPyr), where the product is AcPyrAD. We then used this assay to study competition between AcPyr and other free bases to rank the preference of SARM1 for different base-exchange substrates, identifying isoquinoline as a highly effect substrate that completely outcompetes even AcPyr. This has significant advantages over traditional HPLC methods for assaying SARM1 base exchange as it is rapid, sensitive, cost-effective, and easily scalable. This could represent a useful tool given current interest in the role of SARM1 base exchange in programmed axon death and related human disorders. It may also be applicable to other multifunctional NAD glycohydrolases (EC 3.2.2.6) that possess similar base-exchange activity.
Collapse
Affiliation(s)
- Ilenia Cirilli
- Department of Clinical Sciences (DISCO), Section of Biochemistry, Polytechnic University of Marche, Via Ranieri 67, 60131 Ancona, Italy; (I.C.); (A.A.)
| | - Adolfo Amici
- Department of Clinical Sciences (DISCO), Section of Biochemistry, Polytechnic University of Marche, Via Ranieri 67, 60131 Ancona, Italy; (I.C.); (A.A.)
| | - Jonathan Gilley
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Forvie Site, Robinson Way, Cambridge CB2 0PY, UK; (J.G.); (M.P.C.)
| | - Michael P. Coleman
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Forvie Site, Robinson Way, Cambridge CB2 0PY, UK; (J.G.); (M.P.C.)
| | - Giuseppe Orsomando
- Department of Clinical Sciences (DISCO), Section of Biochemistry, Polytechnic University of Marche, Via Ranieri 67, 60131 Ancona, Italy; (I.C.); (A.A.)
| |
Collapse
|
13
|
Miyamoto T, Kim C, Chow J, Dugas JC, DeGroot J, Bagdasarian AL, Thottumkara AP, Larhammar M, Calvert ME, Fox BM, Lewcock JW, Kane LA. SARM1 is responsible for calpain-dependent dendrite degeneration in mouse hippocampal neurons. J Biol Chem 2024; 300:105630. [PMID: 38199568 PMCID: PMC10862016 DOI: 10.1016/j.jbc.2024.105630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 12/10/2023] [Accepted: 12/24/2023] [Indexed: 01/12/2024] Open
Abstract
Sterile alpha and toll/interleukin receptor motif-containing 1 (SARM1) is a critical regulator of axon degeneration that acts through hydrolysis of NAD+ following injury. Recent work has defined the mechanisms underlying SARM1's catalytic activity and advanced our understanding of SARM1 function in axons, yet the role of SARM1 signaling in other compartments of neurons is still not well understood. Here, we show in cultured hippocampal neurons that endogenous SARM1 is present in axons, dendrites, and cell bodies and that direct activation of SARM1 by the neurotoxin Vacor causes not just axon degeneration, but degeneration of all neuronal compartments. In contrast to the axon degeneration pathway defined in dorsal root ganglia, SARM1-dependent hippocampal axon degeneration in vitro is not sensitive to inhibition of calpain proteases. Dendrite degeneration downstream of SARM1 in hippocampal neurons is dependent on calpain 2, a calpain protease isotype enriched in dendrites in this cell type. In summary, these data indicate SARM1 plays a critical role in neurodegeneration outside of axons and elucidates divergent pathways leading to degeneration in hippocampal axons and dendrites.
Collapse
Affiliation(s)
| | - Chaeyoung Kim
- Denali Therapeutics Inc, South San Francisco, California, USA
| | - Johann Chow
- Denali Therapeutics Inc, South San Francisco, California, USA
| | - Jason C Dugas
- Denali Therapeutics Inc, South San Francisco, California, USA
| | - Jack DeGroot
- Denali Therapeutics Inc, South San Francisco, California, USA
| | | | | | | | | | - Brian M Fox
- Denali Therapeutics Inc, South San Francisco, California, USA
| | | | - Lesley A Kane
- Denali Therapeutics Inc, South San Francisco, California, USA.
| |
Collapse
|
14
|
Reiss AB, Gulkarov S, Jacob B, Srivastava A, Pinkhasov A, Gomolin IH, Stecker MM, Wisniewski T, De Leon J. Mitochondria in Alzheimer's Disease Pathogenesis. Life (Basel) 2024; 14:196. [PMID: 38398707 PMCID: PMC10890468 DOI: 10.3390/life14020196] [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: 01/05/2024] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive and incurable neurodegenerative disorder that primarily affects persons aged 65 years and above. It causes dementia with memory loss and deterioration in thinking and language skills. AD is characterized by specific pathology resulting from the accumulation in the brain of extracellular plaques of amyloid-β and intracellular tangles of phosphorylated tau. The importance of mitochondrial dysfunction in AD pathogenesis, while previously underrecognized, is now more and more appreciated. Mitochondria are an essential organelle involved in cellular bioenergetics and signaling pathways. Mitochondrial processes crucial for synaptic activity such as mitophagy, mitochondrial trafficking, mitochondrial fission, and mitochondrial fusion are dysregulated in the AD brain. Excess fission and fragmentation yield mitochondria with low energy production. Reduced glucose metabolism is also observed in the AD brain with a hypometabolic state, particularly in the temporo-parietal brain regions. This review addresses the multiple ways in which abnormal mitochondrial structure and function contribute to AD. Disruption of the electron transport chain and ATP production are particularly neurotoxic because brain cells have disproportionately high energy demands. In addition, oxidative stress, which is extremely damaging to nerve cells, rises dramatically with mitochondrial dyshomeostasis. Restoring mitochondrial health may be a viable approach to AD treatment.
Collapse
Affiliation(s)
- Allison B. Reiss
- Department of Medicine and Biomedical Research Institute, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (S.G.); (B.J.); (A.S.); (A.P.); (I.H.G.); (J.D.L.)
| | - Shelly Gulkarov
- Department of Medicine and Biomedical Research Institute, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (S.G.); (B.J.); (A.S.); (A.P.); (I.H.G.); (J.D.L.)
| | - Benna Jacob
- Department of Medicine and Biomedical Research Institute, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (S.G.); (B.J.); (A.S.); (A.P.); (I.H.G.); (J.D.L.)
| | - Ankita Srivastava
- Department of Medicine and Biomedical Research Institute, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (S.G.); (B.J.); (A.S.); (A.P.); (I.H.G.); (J.D.L.)
| | - Aaron Pinkhasov
- Department of Medicine and Biomedical Research Institute, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (S.G.); (B.J.); (A.S.); (A.P.); (I.H.G.); (J.D.L.)
| | - Irving H. Gomolin
- Department of Medicine and Biomedical Research Institute, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (S.G.); (B.J.); (A.S.); (A.P.); (I.H.G.); (J.D.L.)
| | - Mark M. Stecker
- The Fresno Institute of Neuroscience, Fresno, CA 93730, USA;
| | - Thomas Wisniewski
- Center for Cognitive Neurology, Departments of Neurology, Pathology and Psychiatry, New York University Grossman School of Medicine, New York, NY 10016, USA;
| | - Joshua De Leon
- Department of Medicine and Biomedical Research Institute, NYU Grossman Long Island School of Medicine, Mineola, NY 11501, USA; (S.G.); (B.J.); (A.S.); (A.P.); (I.H.G.); (J.D.L.)
| |
Collapse
|
15
|
Yang S, Niou ZX, Enriquez A, LaMar J, Huang JY, Ling K, Jafar-Nejad P, Gilley J, Coleman MP, Tennessen JM, Rangaraju V, Lu HC. NMNAT2 supports vesicular glycolysis via NAD homeostasis to fuel fast axonal transport. Mol Neurodegener 2024; 19:13. [PMID: 38282024 PMCID: PMC10823734 DOI: 10.1186/s13024-023-00690-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 11/28/2023] [Indexed: 01/30/2024] Open
Abstract
BACKGROUND Bioenergetic maladaptations and axonopathy are often found in the early stages of neurodegeneration. Nicotinamide adenine dinucleotide (NAD), an essential cofactor for energy metabolism, is mainly synthesized by Nicotinamide mononucleotide adenylyl transferase 2 (NMNAT2) in CNS neurons. NMNAT2 mRNA levels are reduced in the brains of Alzheimer's, Parkinson's, and Huntington's disease. Here we addressed whether NMNAT2 is required for axonal health of cortical glutamatergic neurons, whose long-projecting axons are often vulnerable in neurodegenerative conditions. We also tested if NMNAT2 maintains axonal health by ensuring axonal ATP levels for axonal transport, critical for axonal function. METHODS We generated mouse and cultured neuron models to determine the impact of NMNAT2 loss from cortical glutamatergic neurons on axonal transport, energetic metabolism, and morphological integrity. In addition, we determined if exogenous NAD supplementation or inhibiting a NAD hydrolase, sterile alpha and TIR motif-containing protein 1 (SARM1), prevented axonal deficits caused by NMNAT2 loss. This study used a combination of techniques, including genetics, molecular biology, immunohistochemistry, biochemistry, fluorescent time-lapse imaging, live imaging with optical sensors, and anti-sense oligos. RESULTS We provide in vivo evidence that NMNAT2 in glutamatergic neurons is required for axonal survival. Using in vivo and in vitro studies, we demonstrate that NMNAT2 maintains the NAD-redox potential to provide "on-board" ATP via glycolysis to vesicular cargos in distal axons. Exogenous NAD+ supplementation to NMNAT2 KO neurons restores glycolysis and resumes fast axonal transport. Finally, we demonstrate both in vitro and in vivo that reducing the activity of SARM1, an NAD degradation enzyme, can reduce axonal transport deficits and suppress axon degeneration in NMNAT2 KO neurons. CONCLUSION NMNAT2 ensures axonal health by maintaining NAD redox potential in distal axons to ensure efficient vesicular glycolysis required for fast axonal transport.
Collapse
Affiliation(s)
- Sen Yang
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA
- Program in Neuroscience, Indiana University, Bloomington, IN, 47405, USA
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, 33458, USA
| | - Zhen-Xian Niou
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA
- Program in Neuroscience, Indiana University, Bloomington, IN, 47405, USA
| | - Andrea Enriquez
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA
- Program in Neuroscience, Indiana University, Bloomington, IN, 47405, USA
| | - Jacob LaMar
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, 33458, USA
- Present address: Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, 33458, USA
| | - Jui-Yen Huang
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA
| | - Karen Ling
- Neuroscience Drug Discovery, Ionis Pharmaceuticals, Inc., 2855, Gazelle Court, Carlsbad, CA, 92010, USA
| | - Paymaan Jafar-Nejad
- Neuroscience Drug Discovery, Ionis Pharmaceuticals, Inc., 2855, Gazelle Court, Carlsbad, CA, 92010, USA
| | - Jonathan Gilley
- Department of Clinical Neuroscience, Cambridge University, Cambridge, UK
| | - Michael P Coleman
- Department of Clinical Neuroscience, Cambridge University, Cambridge, UK
| | - Jason M Tennessen
- Department of Biology, Indiana University, Bloomington, IN, 47405, USA
| | - Vidhya Rangaraju
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, 33458, USA
| | - Hui-Chen Lu
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA.
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA.
- Program in Neuroscience, Indiana University, Bloomington, IN, 47405, USA.
| |
Collapse
|
16
|
Guo Y, Mehrabian Z, Milbrandt J, DiAntonio A, Bernstein SL. Synergistic Protection of Retinal Ganglion Cells (RGCs) by SARM1 Inactivation with CNTF in a Rodent Model of Nonarteritic Anterior Ischemic Optic Neuropathy. Cells 2024; 13:202. [PMID: 38334594 PMCID: PMC10854792 DOI: 10.3390/cells13030202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 02/10/2024] Open
Abstract
We evaluated whether inhibiting sterile alpha and (Toll/interleukin receptor (TIR)) motif-containing 1 (SARM1) activity protects retinal ganglion cells (RGCs) following ischemic axonopathy (rodent nonarteritic anterior ischemic optic neuropathy: rNAION) by itself and combined with ciliary neurotrophic factor (CNTF). Genetically modified SARM1(-) rats were rNAION-induced in one eye and compared against equivalently induced wild-type animals of the same background. Optic nerve (ON) diameters were quantified using optical coherence tomography (SD-OCT). RGCs were quantified 30 d post-induction using retinal stereology for Brn3a(+) nuclei. ON sections were analyzed by TEM and immunohistochemistry. SARM1(-)(-) and WT animals were then bilaterally sequentially rNAION-induced. One eye received intravitreal vehicle injection following induction; the contralateral side received CNTF and was analyzed 30 d post-induction. Inhibiting SARM1 activity suppressed axonal collapse following ischemic axonopathy. SARM1(-) animals significantly reduced RGC loss, compared with WT animals (49.4 ± 6.8% RGC loss in SARM1(-) vs. 63.6 ± 3.2% sem RGC loss in WT; Mann-Whitney one-tailed U-test, (p = 0.049)). IVT-CNTF treatment vs. IVT-vehicle in SARM1(-) animals further reduced RGC loss by 24% at 30 d post-induction, but CNTF did not, by itself, improve long-term RGC survival in WT animals compared with vehicle (Mann-Whitney one-tailed t-test; p = 0.033). While inhibiting SARM1 activity is itself neuroprotective, combining SARM1 inhibition and CNTF treatment generated a long-term, synergistic neuroprotective effect in ischemic neuropathy. Combinatorial treatments for NAION utilizing independent neuroprotective mechanisms may thus provide a greater effect than individual treatment modalities.
Collapse
Affiliation(s)
- Yan Guo
- Departments of Ophthalmology and Visual Sciences, School of Medicine, University of Maryland, Baltimore, MD 21201, USA; (Y.G.); (Z.M.)
| | - Zara Mehrabian
- Departments of Ophthalmology and Visual Sciences, School of Medicine, University of Maryland, Baltimore, MD 21201, USA; (Y.G.); (Z.M.)
| | - Jeffrey Milbrandt
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA;
- Needleman Center for Neurometabolism and Axonal Therapeutics, St. Louis, MO 63110, USA;
| | - Aaron DiAntonio
- Needleman Center for Neurometabolism and Axonal Therapeutics, St. Louis, MO 63110, USA;
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Steven L. Bernstein
- Departments of Ophthalmology and Visual Sciences, School of Medicine, University of Maryland, Baltimore, MD 21201, USA; (Y.G.); (Z.M.)
- Anatomy and Neurobiology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| |
Collapse
|
17
|
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
|
18
|
Alexandris AS, Koliatsos VE. NAD +, Axonal Maintenance, and Neurological Disease. Antioxid Redox Signal 2023; 39:1167-1184. [PMID: 37503611 PMCID: PMC10715442 DOI: 10.1089/ars.2023.0350] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 05/28/2023] [Indexed: 07/29/2023]
Abstract
Significance: The remarkable geometry of the axon exposes it to unique challenges for survival and maintenance. Axonal degeneration is a feature of peripheral neuropathies, glaucoma, and traumatic brain injury, and an early event in neurodegenerative diseases. Since the discovery of Wallerian degeneration (WD), a molecular program that hijacks nicotinamide adenine dinucleotide (NAD+) metabolism for axonal self-destruction, the complex roles of NAD+ in axonal viability and disease have become research priority. Recent Advances: The discoveries of the protective Wallerian degeneration slow (WldS) and of sterile alpha and TIR motif containing 1 (SARM1) activation as the main instructive signal for WD have shed new light on the regulatory role of NAD+ in axonal degeneration in a growing number of neurological diseases. SARM1 has been characterized as a NAD+ hydrolase and sensor of NAD+ metabolism. The discovery of regulators of nicotinamide mononucleotide adenylyltransferase 2 (NMNAT2) proteostasis in axons, the allosteric regulation of SARM1 by NAD+ and NMN, and the existence of clinically relevant windows of action of these signals has opened new opportunities for therapeutic interventions, including SARM1 inhibitors and modulators of NAD+ metabolism. Critical Issues: Events upstream and downstream of SARM1 remain unclear. Furthermore, manipulating NAD+ metabolism, an overdetermined process crucial in cell survival, for preventing the degeneration of the injured axon may be difficult and potentially toxic. Future Directions: There is a need for clarification of the distinct roles of NAD+ metabolism in axonal maintenance as contrasted to WD. There is also a need to better understand the role of NAD+ metabolism in axonal endangerment in neuropathies, diseases of the white matter, and the early stages of neurodegenerative diseases of the central nervous system. Antioxid. Redox Signal. 39, 1167-1184.
Collapse
Affiliation(s)
| | - Vassilis E. Koliatsos
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Neurology, and Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
19
|
Bhasin S, Seals D, Migaud M, Musi N, Baur JA. Nicotinamide Adenine Dinucleotide in Aging Biology: Potential Applications and Many Unknowns. Endocr Rev 2023; 44:1047-1073. [PMID: 37364580 DOI: 10.1210/endrev/bnad019] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 05/28/2023] [Accepted: 06/22/2023] [Indexed: 06/28/2023]
Abstract
Recent research has unveiled an expansive role of NAD+ in cellular energy generation, redox reactions, and as a substrate or cosubstrate in signaling pathways that regulate health span and aging. This review provides a critical appraisal of the clinical pharmacology and the preclinical and clinical evidence for therapeutic effects of NAD+ precursors for age-related conditions, with a particular focus on cardiometabolic disorders, and discusses gaps in current knowledge. NAD+ levels decrease throughout life; age-related decline in NAD+ bioavailability has been postulated to be a contributor to many age-related diseases. Raising NAD+ levels in model organisms by administration of NAD+ precursors improves glucose and lipid metabolism; attenuates diet-induced weight gain, diabetes, diabetic kidney disease, and hepatic steatosis; reduces endothelial dysfunction; protects heart from ischemic injury; improves left ventricular function in models of heart failure; attenuates cerebrovascular and neurodegenerative disorders; and increases health span. Early human studies show that NAD+ levels can be raised safely in blood and some tissues by oral NAD+ precursors and suggest benefit in preventing nonmelanotic skin cancer, modestly reducing blood pressure and improving lipid profile in older adults with obesity or overweight; preventing kidney injury in at-risk patients; and suppressing inflammation in Parkinson disease and SARS-CoV-2 infection. Clinical pharmacology, metabolism, and therapeutic mechanisms of NAD+ precursors remain incompletely understood. We suggest that these early findings provide the rationale for adequately powered randomized trials to evaluate the efficacy of NAD+ augmentation as a therapeutic strategy to prevent and treat metabolic disorders and age-related conditions.
Collapse
Affiliation(s)
- Shalender Bhasin
- Department of Medicine, Harvard Medical School, Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Douglas Seals
- Department of Integrative Physiology and Medicine, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Marie Migaud
- Department of Pharmacology, Mitchell Cancer Institute, College of Medicine, University of Southern Alabama, Mobile, AL 36688, USA
| | - Nicolas Musi
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Joseph A Baur
- Department of Physiology, Institute for Diabetes, Obesity & Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
20
|
Zuo Z, Zhang Z, Zhang S, Fan B, Li G. The Molecular Mechanisms Involved in Axonal Degeneration and Retrograde Retinal Ganglion Cell Death. DNA Cell Biol 2023; 42:653-667. [PMID: 37819746 DOI: 10.1089/dna.2023.0180] [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] [Indexed: 10/13/2023] Open
Abstract
Axonal degeneration is a pathologic change common to multiple retinopathies and optic neuropathies. Various pathologic factors, such as mechanical injury, inflammation, and ischemia, can damage retinal ganglion cell (RGC) somas and axons, eventually triggering axonal degeneration and RGC death. The molecular mechanisms of somal and axonal degeneration are distinct but also overlap, and axonal degeneration can result in retrograde somal degeneration. While the mitogen-activated protein kinase pathway acts as a central node in RGC axon degeneration, several newly discovered molecules, such as sterile alpha and Toll/interleukin-1 receptor motif-containing protein 1 and nicotinamide mononucleotide adenylyltransferase 2, also play a critical role in this pathological process following different types of injury. Therefore, we summarize the types of injury that cause RGC axon degeneration and retrograde RGC death and important underlying molecular mechanisms, providing a reference for the identification of targets for protecting axons and RGCs.
Collapse
Affiliation(s)
- Zhaoyang Zuo
- Department of Ophthalmology, Second Hospital of Jilin University, Changchun, China
| | - Ziyuan Zhang
- Department of Ophthalmology, Second Hospital of Jilin University, Changchun, China
| | - Siming Zhang
- Department of Ophthalmology, Second Hospital of Jilin University, Changchun, China
| | - Bin Fan
- Department of Ophthalmology, Second Hospital of Jilin University, Changchun, China
| | - Guangyu Li
- Department of Ophthalmology, Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
21
|
Icso JD, Thompson PR. A phase transition reduces the threshold for nicotinamide mononucleotide-based activation of SARM1, an NAD(P) hydrolase, to physiologically relevant levels. J Biol Chem 2023; 299:105284. [PMID: 37742918 PMCID: PMC10624580 DOI: 10.1016/j.jbc.2023.105284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/04/2023] [Accepted: 09/19/2023] [Indexed: 09/26/2023] Open
Abstract
Axonal degeneration is a hallmark feature of neurodegenerative diseases. Activation of the NAD(P)ase sterile alpha and toll-interleukin receptor motif containing protein 1 (SARM1) is critical for this process. In resting neurons, SARM1 activity is inhibited, but upon damage, SARM1 is activated and catalyzes one of three NAD(P)+ dependent reactions: (1) NAD(P)+ hydrolysis to form ADP-ribose (ADPR[P]) and nicotinamide; (2) the formation of cyclic-ADPR (cADPR[P]); or (3) a base exchange reaction with nicotinic acid (NA) and NADP+ to form NA adenine dinucleotide phosphate. Production of these metabolites triggers axonal death. Two activation mechanisms have been proposed: (1) an increase in the nicotinamide mononucleotide (NMN) concentration, which leads to the allosteric activation of SARM1, and (2) a phase transition, which stabilizes the active conformation of the enzyme. However, neither of these mechanisms have been shown to occur at the same time. Using in vitro assay systems, we show that the liquid-to-solid phase transition lowers the NMN concentration required to activate the catalytic activity of SARM1 by up to 140-fold. These results unify the proposed activation mechanisms and show for the first time that a phase transition reduces the threshold for NMN-based SARM1 activation to physiologically relevant levels. These results further our understanding of SARM1 activation and will be important for the future development of therapeutics targeting SARM1.
Collapse
Affiliation(s)
- Janneke Doedée Icso
- Program in Chemical Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA; Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medial School, Worcester, Massachusetts, USA
| | - Paul Ryan Thompson
- Program in Chemical Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA; Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medial School, Worcester, Massachusetts, USA.
| |
Collapse
|
22
|
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
|
23
|
Heßling LD, Troost-Kind B, Weiß M. NAADP-binding proteins - Linking NAADP signaling to cancer and immunity. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119531. [PMID: 37394011 DOI: 10.1016/j.bbamcr.2023.119531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/02/2023] [Accepted: 06/26/2023] [Indexed: 07/04/2023]
Abstract
NAADP is one of the most potent calcium mobilizing second messengers. Only recently, two NAADP-binding proteins have been identified: HN1L/JPT2 and LSM12. Further, ASPDH was suggested as a less selective binding partner. Apart from this newly uncovered link, little is known about the shared mechanisms between these proteins. The aim of this review is to assess potential functional connections between NAADP and its binding proteins. We here give a description of two major links. For one, HN1L/JPT2 and LSM12 both have potent oncogenic functions in several cancer types. Second, they are involved in similar cellular pathways in both cancer and immunity.
Collapse
Affiliation(s)
- Louisa D Heßling
- The Calcium Signaling Group, Dept. of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.
| | - Berit Troost-Kind
- The Calcium Signaling Group, Dept. of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Mariella Weiß
- The Calcium Signaling Group, Dept. of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| |
Collapse
|
24
|
Khazma T, Grossman A, Guez-Haddad J, Feng C, Dabas H, Sain R, Weitman M, Zalk R, Isupov MN, Hammarlund M, Hons M, Opatowsky Y. Structure-function analysis of ceTIR-1/hSARM1 explains the lack of Wallerian axonal degeneration in C. elegans. Cell Rep 2023; 42:113026. [PMID: 37635352 PMCID: PMC10675840 DOI: 10.1016/j.celrep.2023.113026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 06/05/2023] [Accepted: 08/09/2023] [Indexed: 08/29/2023] Open
Abstract
Wallerian axonal degeneration (WD) does not occur in the nematode C. elegans, in contrast to other model animals. However, WD depends on the NADase activity of SARM1, a protein that is also expressed in C. elegans (ceSARM/ceTIR-1). We hypothesized that differences in SARM between species might exist and account for the divergence in WD. We first show that expression of the human (h)SARM1, but not ceTIR-1, in C. elegans neurons is sufficient to confer axon degeneration after nerve injury. Next, we determined the cryoelectron microscopy structure of ceTIR-1 and found that, unlike hSARM1, which exists as an auto-inhibited ring octamer, ceTIR-1 forms a readily active 9-mer. Enzymatically, the NADase activity of ceTIR-1 is substantially weaker (10-fold higher Km) than that of hSARM1, and even when fully active, it falls short of consuming all cellular NAD+. Our experiments provide insight into the molecular mechanisms and evolution of SARM orthologs and WD across species.
Collapse
Affiliation(s)
- Tami Khazma
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Atira Grossman
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Julia Guez-Haddad
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Chengye Feng
- Departments of Neuroscience and Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Hadas Dabas
- Departments of Neuroscience and Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Radhika Sain
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Michal Weitman
- Department of Chemistry, Bar-Ilan University, Ramat Gan, Israel
| | - Ran Zalk
- Ilse Katz Institute for Nanoscale Science & Technology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Michail N Isupov
- Henry Wellcome Building for Biocatalysis, Biosciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Marc Hammarlund
- Departments of Neuroscience and Genetics, Yale School of Medicine, New Haven, CT, USA.
| | - Michael Hons
- European Molecular Biology Laboratory, Grenoble, France.
| | - Yarden Opatowsky
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel.
| |
Collapse
|
25
|
Yang S, Park JH, Lu HC. Axonal energy metabolism, and the effects in aging and neurodegenerative diseases. Mol Neurodegener 2023; 18:49. [PMID: 37475056 PMCID: PMC10357692 DOI: 10.1186/s13024-023-00634-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 06/08/2023] [Indexed: 07/22/2023] Open
Abstract
Human studies consistently identify bioenergetic maladaptations in brains upon aging and neurodegenerative disorders of aging (NDAs), such as Alzheimer's disease, Parkinson's disease, Huntington's disease, and Amyotrophic lateral sclerosis. Glucose is the major brain fuel and glucose hypometabolism has been observed in brain regions vulnerable to aging and NDAs. Many neurodegenerative susceptible regions are in the topological central hub of the brain connectome, linked by densely interconnected long-range axons. Axons, key components of the connectome, have high metabolic needs to support neurotransmission and other essential activities. Long-range axons are particularly vulnerable to injury, neurotoxin exposure, protein stress, lysosomal dysfunction, etc. Axonopathy is often an early sign of neurodegeneration. Recent studies ascribe axonal maintenance failures to local bioenergetic dysregulation. With this review, we aim to stimulate research in exploring metabolically oriented neuroprotection strategies to enhance or normalize bioenergetics in NDA models. Here we start by summarizing evidence from human patients and animal models to reveal the correlation between glucose hypometabolism and connectomic disintegration upon aging/NDAs. To encourage mechanistic investigations on how axonal bioenergetic dysregulation occurs during aging/NDAs, we first review the current literature on axonal bioenergetics in distinct axonal subdomains: axon initial segments, myelinated axonal segments, and axonal arbors harboring pre-synaptic boutons. In each subdomain, we focus on the organization, activity-dependent regulation of the bioenergetic system, and external glial support. Second, we review the mechanisms regulating axonal nicotinamide adenine dinucleotide (NAD+) homeostasis, an essential molecule for energy metabolism processes, including NAD+ biosynthetic, recycling, and consuming pathways. Third, we highlight the innate metabolic vulnerability of the brain connectome and discuss its perturbation during aging and NDAs. As axonal bioenergetic deficits are developing into NDAs, especially in asymptomatic phase, they are likely exaggerated further by impaired NAD+ homeostasis, the high energetic cost of neural network hyperactivity, and glial pathology. Future research in interrogating the causal relationship between metabolic vulnerability, axonopathy, amyloid/tau pathology, and cognitive decline will provide fundamental knowledge for developing therapeutic interventions.
Collapse
Affiliation(s)
- Sen Yang
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA
- Program in Neuroscience, Indiana University, Bloomington, IN, 47405, USA
| | - Jung Hyun Park
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA
- Program in Neuroscience, Indiana University, Bloomington, IN, 47405, USA
| | - Hui-Chen Lu
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA.
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA.
- Program in Neuroscience, Indiana University, Bloomington, IN, 47405, USA.
| |
Collapse
|
26
|
Icso JD, Barasa L, Thompson PR. SARM1, an Enzyme Involved in Axon Degeneration, Catalyzes Multiple Activities through a Ternary Complex Mechanism. Biochemistry 2023; 62:2065-2078. [PMID: 37307562 DOI: 10.1021/acs.biochem.3c00081] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Sterile alpha and toll/interleukin receptor (TIR) motif containing protein 1 (SARM1) is an NAD+ hydrolase and cyclase involved in axonal degeneration. In addition to NAD+ hydrolysis and cyclization, SARM1 catalyzes a base exchange reaction between nicotinic acid (NA) and NADP+ to generate NAADP, which is a potent calcium signaling molecule. Herein, we describe efforts to characterize the hydrolysis, cyclization, and base exchange activities of TIR-1, the Caenorhabditis elegans ortholog of SARM1; TIR-1 also catalyzes NAD(P)+ hydrolysis and/or cyclization and regulates axonal degeneration in worms. We show that the catalytic domain of TIR-1 undergoes a liquid-to-solid phase transition that regulates not only the hydrolysis and cyclization reactions but also the base exchange reaction. We define the substrate specificities of the reactions, demonstrate that cyclization and base exchange reactions occur within the same pH range, and establish that TIR-1 uses a ternary complex mechanism. Overall, our findings will aid drug discovery efforts and provide insight into the mechanism of recently described inhibitors.
Collapse
Affiliation(s)
- Janneke D Icso
- Program in Chemical Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605, United States
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medial School, Worcester, Massachusetts 01605, United States
| | - Leonard Barasa
- Program in Chemical Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605, United States
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medial School, Worcester, Massachusetts 01605, United States
| | - Paul R Thompson
- Program in Chemical Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605, United States
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medial School, Worcester, Massachusetts 01605, United States
| |
Collapse
|
27
|
Maruta N, Sorbello M, Lim BYJ, McGuinness HY, Shi Y, Ve T, Kobe B. TIR domain-associated nucleotides with functions in plant immunity and beyond. CURRENT OPINION IN PLANT BIOLOGY 2023; 73:102364. [PMID: 37086529 DOI: 10.1016/j.pbi.2023.102364] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/19/2023] [Accepted: 03/09/2023] [Indexed: 05/03/2023]
Abstract
TIR (Toll/interlukin-1 receptor) domains are found in archaea, bacteria and eukaryotes, featured in proteins generally associated with immune functions. In plants, they are found in a large group of NLRs (nucleotide-binding leucine-rich repeat receptors), NLR-like proteins and TIR-only proteins. They are also present in effector proteins from phytopathogenic bacteria that are associated with suppression of host immunity. TIR domains from plants and bacteria are enzymes that cleave NAD+ (nicotinamide adenine dinucleotide, oxidized form) and other nucleotides. In dicot plants, TIR-derived signalling molecules activate downstream immune signalling proteins, the EDS1 (enhanced disease susceptibility 1) family proteins, and in turn helper NLRs. Recent work has brought major advances in understanding how TIR domains work, how they produce signalling molecules and how these products signal.
Collapse
Affiliation(s)
- Natsumi Maruta
- The University of Queensland, School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, Brisbane, QLD 4072, Australia
| | - Mitchell Sorbello
- The University of Queensland, School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, Brisbane, QLD 4072, Australia
| | - Bryan Y J Lim
- The University of Queensland, School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, Brisbane, QLD 4072, Australia
| | - Helen Y McGuinness
- The University of Queensland, School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, Brisbane, QLD 4072, Australia
| | - Yun Shi
- Institute for Glycomics, Griffith University, Southport, QLD 4222, Australia
| | - Thomas Ve
- Institute for Glycomics, Griffith University, Southport, QLD 4222, Australia
| | - Bostjan Kobe
- The University of Queensland, School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, Brisbane, QLD 4072, Australia.
| |
Collapse
|
28
|
Jia A, Huang S, Ma S, Chang X, Han Z, Chai J. TIR-catalyzed nucleotide signaling molecules in plant defense. CURRENT OPINION IN PLANT BIOLOGY 2023; 73:102334. [PMID: 36702016 DOI: 10.1016/j.pbi.2022.102334] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 12/26/2022] [Accepted: 12/26/2022] [Indexed: 06/10/2023]
Abstract
Toll and interleukin-1 receptor (TIR) domain is a conserved immune module in prokaryotes and eukaryotes. Signaling regulated by TIR-only proteins or TIR domain-containing intracellular immune receptors is critical for plant immunity. Recent studies demonstrated that TIR domains function as enzymes encoding a variety of activities, which manifest different mechanisms for regulation of plant immunity. These enzymatic activities catalyze metabolism of NAD+, ATP and other nucleic acids, generating structurally diversified nucleotide metabolites. Signaling roles have been revealed for some TIR enzymatic products that can act as second messengers to induce plant immunity. Herein, we summarize our current knowledge about catalytic production of these nucleotide metabolites and their roles in plant immune signaling. We also highlight outstanding questions that are likely to be the focus of future investigations about TIR-produced signaling molecules.
Collapse
Affiliation(s)
- Aolin Jia
- Beijing Frontier Research Center for Biological Structure, Center for Plant Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Shijia Huang
- Beijing Frontier Research Center for Biological Structure, Center for Plant Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Shoucai Ma
- Beijing Frontier Research Center for Biological Structure, Center for Plant Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xiaoyu Chang
- Beijing Frontier Research Center for Biological Structure, Center for Plant Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Zhifu Han
- Beijing Frontier Research Center for Biological Structure, Center for Plant Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China.
| | - Jijie Chai
- Beijing Frontier Research Center for Biological Structure, Center for Plant Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; Institute of Biochemistry, University of Cologne, Cologne 50674, Germany; Max Planck Institute for Plant Breeding Research, Department of Plant-Microbe Interactions, Cologne 50829, Germany.
| |
Collapse
|
29
|
Yang S, Niou ZX, Enriquez A, LaMar J, Huang JY, Ling K, Jafar-Nejad P, Gilley J, Coleman MP, Tennessen JM, Rangaraju V, Lu HC. NMNAT2 supports vesicular glycolysis via NAD homeostasis to fuel fast axonal transport. RESEARCH SQUARE 2023:rs.3.rs-2859584. [PMID: 37292715 PMCID: PMC10246254 DOI: 10.21203/rs.3.rs-2859584/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Background Bioenergetic maladaptations and axonopathy are often found in the early stages of neurodegeneration. Nicotinamide adenine dinucleotide (NAD), an essential cofactor for energy metabolism, is mainly synthesized by Nicotinamide mononucleotide adenylyl transferase 2 (NMNAT2) in CNS neurons. NMNAT2 mRNA levels are reduced in the brains of Alzheimer's, Parkinson's, and Huntington's disease. Here we addressed whether NMNAT2 is required for axonal health of cortical glutamatergic neurons, whose long-projecting axons are often vulnerable in neurodegenerative conditions. We also tested if NMNAT2 maintains axonal health by ensuring axonal ATP levels for axonal transport, critical for axonal function. Methods We generated mouse and cultured neuron models to determine the impact of NMNAT2 loss from cortical glutamatergic neurons on axonal transport, energetic metabolism, and morphological integrity. In addition, we determined if exogenous NAD supplementation or inhibiting a NAD hydrolase, sterile alpha and TIR motif-containing protein 1 (SARM1), prevented axonal deficits caused by NMNAT2 loss. This study used a combination of genetics, molecular biology, immunohistochemistry, biochemistry, fluorescent time-lapse imaging, live imaging with optical sensors, and anti-sense oligos. Results We provide in vivo evidence that NMNAT2 in glutamatergic neurons is required for axonal survival. Using in vivo and in vitro studies, we demonstrate that NMNAT2 maintains the NAD-redox potential to provide "on-board" ATP via glycolysis to vesicular cargos in distal axons. Exogenous NAD+ supplementation to NMNAT2 KO neurons restores glycolysis and resumes fast axonal transport. Finally, we demonstrate both in vitro and in vivo that reducing the activity of SARM1, an NAD degradation enzyme, can reduce axonal transport deficits and suppress axon degeneration in NMNAT2 KO neurons. Conclusion NMNAT2 ensures axonal health by maintaining NAD redox potential in distal axons to ensure efficient vesicular glycolysis required for fast axonal transport.
Collapse
|
30
|
Huang K, Zhu WJ, Li WH, Lee HC, Zhao YJ, Lee CS. Base-Exchange Enabling the Visualization of SARM1 Activities in Sciatic Nerve-Injured Mice. ACS Sens 2023; 8:767-773. [PMID: 36689294 PMCID: PMC9972468 DOI: 10.1021/acssensors.2c02317] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Enzymes are important in homeostasis in living organisms. Since abnormal enzyme activities are highly associated with many human diseases, detection of in vivo activities of a specific enzyme is important to study the pathology of the related diseases. In this work, we have designed and synthesized a series of new small-molecule-activatable fluorescent probes for the imaging of Sterile Alpha and TIR Motif-containing 1 (SARM1) activities based on its transglycosidase activities (base-exchange reactions of NAD+). Probe 1a was found to undergo base-exchange reactions with NAD+ in the presence of activated SARM1 but not CD38 nor NADase and formed a highly emissive product AD-1a [about a 100-fold fluorescence enhancement in 20 min with a 150 nm (5665 cm-1) Stokes shift and a 100 nm (3812 cm-1) red shift]. This probe exhibited a higher reactivity and sensitivity than those commonly used for SARM1 imaging. The utilities of 1a have also been demonstrated in live-cell imaging and detection of in vivo activities of SARM1 in a sciatic nerve injury mouse model.
Collapse
Affiliation(s)
- Ke Huang
- Department of Chemistry, Hong Kong Baptist University, Waterloo Road, Kowloon Tong, Kowloon, Hong Kong SAR 999077, China
| | - Wen Jie Zhu
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen University Town, Lishui Road, Shenzhen 518055, China
| | - Wan Hua Li
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen University Town, Lishui Road, Shenzhen 518055, China.,Ciechanover Institute of Precision and Regenerative Medicine, School of Life and Health Sciences, School of Medicine, The Chinese University of Hong Kong Shenzhen, Shenzhen 518172, China.,School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Hon Cheung Lee
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen University Town, Lishui Road, Shenzhen 518055, China
| | - Yong Juan Zhao
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen University Town, Lishui Road, Shenzhen 518055, China.,Ciechanover Institute of Precision and Regenerative Medicine, School of Life and Health Sciences, School of Medicine, The Chinese University of Hong Kong Shenzhen, Shenzhen 518172, China.,Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Chi-Sing Lee
- Department of Chemistry, Hong Kong Baptist University, Waterloo Road, Kowloon Tong, Kowloon, Hong Kong SAR 999077, China
| |
Collapse
|
31
|
Guse AH. Enzymology of Ca 2+-Mobilizing Second Messengers Derived from NAD: From NAD Glycohydrolases to (Dual) NADPH Oxidases. Cells 2023; 12:cells12040675. [PMID: 36831342 PMCID: PMC9954121 DOI: 10.3390/cells12040675] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 02/23/2023] Open
Abstract
Nicotinamide adenine dinucleotide (NAD) and its 2'-phosphorylated cousin NADP are precursors for the enzymatic formation of the Ca2+-mobilizing second messengers adenosine diphosphoribose (ADPR), 2'-deoxy-ADPR, cyclic ADPR, and nicotinic acid adenine dinucleotide phosphate (NAADP). The enzymes involved are either NAD glycohydrolases CD38 or sterile alpha toll/interleukin receptor motif containing-1 (SARM1), or (dual) NADPH oxidases (NOX/DUOX). Enzymatic function(s) are reviewed and physiological role(s) in selected cell systems are discussed.
Collapse
Affiliation(s)
- Andreas H Guse
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| |
Collapse
|
32
|
Gao L, Du X, Li J, Qin FXF. Evolving roles of CD38 metabolism in solid tumour microenvironment. Br J Cancer 2023; 128:492-504. [PMID: 36396822 PMCID: PMC9938187 DOI: 10.1038/s41416-022-02052-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/20/2022] [Accepted: 10/27/2022] [Indexed: 11/19/2022] Open
Abstract
Given that plenty of clinical findings and reviews have already explained in detail on the progression of CD38 in multiple myeloma and haematological system tumours, here we no longer give unnecessary discussion on the above progression. Though therapeutic antibodies have been regarded as a greatest breakthrough in multiple myeloma immunotherapies due to the durable anti-tumour responses in the clinic, but the role of CD38 in the immunologic regulation and evasion of non-hematopoietic solid tumours are just initiated and controversial. Therefore, we will focus on the bio-function of CD38 enzymatic substrates or metabolites in the variety of non-hematopoietic malignancies and the potential therapeutic value of targeting the CD38-NAD+ or CD38-cADPR/ADPR signal axis. Though limited, we review some ongoing researches and clinical trials on therapeutic approaches in solid tumour as well.
Collapse
Affiliation(s)
- Long Gao
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, 230022, Hefei, China
| | - Xiaohong Du
- Institute of Clinical Medicine Research, Suzhou Science and Technology Town Hospital, Suzhou, China
| | - Jiabin Li
- Department of Infectious Disease, The First Affiliated Hospital of Anhui Medical University, 230022, Hefei, China.
| | - F Xiao-Feng Qin
- Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 100005, Beijing, China.
- Suzhou Institute of Systems Medicine, 215123, Suzhou, China.
| |
Collapse
|
33
|
Abstract
The discovery of NAADP-evoked Ca2+ release in sea urchin eggs and then as a ubiquitous Ca2+ mobilizing messenger has introduced several novel paradigms to our understanding of Ca2+ signalling, not least in providing a link between cell stimulation and Ca2+ release from lysosomes and other acidic Ca2+ storage organelles. In addition, the hallmark concentration-response relationship of NAADP-mediated Ca2+ release, shaped by striking activation/desensitization mechanisms, influences its actions as an intracellular messenger. There has been recent progress in our understanding of the molecular mechanisms underlying NAADP-evoked Ca2+ release, such as the identification of the endo-lysosomal two-pore channel family of cation channels (TPCs) as their principal target and the identity of NAADP-binding proteins that complex with them. The NAADP/TPC signalling axis has gained recent prominence in pathophysiology for their roles in such disease processes as neurodegeneration, tumorigenesis and cellular viral entry.
Collapse
Affiliation(s)
- Antony Galione
- Department of Pharmacology, University of Oxford, Oxford, UK.
| | - Lianne C Davis
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Lora L Martucci
- Department of Pharmacology, University of Oxford, Oxford, UK
| | | |
Collapse
|
34
|
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
|
35
|
NAD + Metabolism and Interventions in Premature Renal Aging and Chronic Kidney Disease. Cells 2022; 12:cells12010021. [PMID: 36611814 PMCID: PMC9818486 DOI: 10.3390/cells12010021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/17/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Premature aging causes morphological and functional changes in the kidney, leading to chronic kidney disease (CKD). CKD is a global public health issue with far-reaching consequences, including cardio-vascular complications, increased frailty, shortened lifespan and a heightened risk of kidney failure. Dialysis or transplantation are lifesaving therapies, but they can also be debilitating. Currently, no cure is available for CKD, despite ongoing efforts to identify clinical biomarkers of premature renal aging and molecular pathways of disease progression. Kidney proximal tubular epithelial cells (PTECs) have high energy demand, and disruption of their energy homeostasis has been linked to the progression of kidney disease. Consequently, metabolic reprogramming of PTECs is gaining interest as a therapeutic tool. Preclinical and clinical evidence is emerging that NAD+ homeostasis, crucial for PTECs' oxidative metabolism, is impaired in CKD, and administration of dietary NAD+ precursors could have a prophylactic role against age-related kidney disease. This review describes the biology of NAD+ in the kidney, including its precursors and cellular roles, and discusses the importance of NAD+ homeostasis for renal health. Furthermore, we provide a comprehensive summary of preclinical and clinical studies aimed at increasing NAD+ levels in premature renal aging and CKD.
Collapse
|
36
|
Dai Y, Lin J, Ren J, Zhu B, Wu C, Yu L. NAD + metabolism in peripheral neuropathic pain. Neurochem Int 2022; 161:105435. [PMID: 36273706 DOI: 10.1016/j.neuint.2022.105435] [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: 04/17/2022] [Revised: 10/04/2022] [Accepted: 10/16/2022] [Indexed: 11/07/2022]
Abstract
Nicotinamide adenine dinucleotide (NAD+) is an omnipresent metabolite that participates in redox reactions. Multiple NAD+-consuming enzymes are implicated in numerous biological processes, including transcription, signaling, and cell survival. Multiple pieces of evidence have demonstrated that NAD+-consuming enzymes, including poly(ADP-ribose) polymerases (PARPs), sirtuins (SIRTs), and sterile alpha and TIR motif-containing 1 (SARM1), play major roles in peripheral neuropathic pain of various etiologies. These NAD+ consumers primarily participate in peripheral neuropathic pain via mechanisms such as mitochondrial dysfunction, oxidative stress, and inflammation. Furthermore, NAD+ synthase and nicotinamide phosphoribosyltransferase (NAMPT) have recently been found to contribute to the regulation of pain. Here, we review the evidence indicating the involvement of NAD+ metabolism in the pathological mechanisms of peripheral neuropathic pain. Advanced understanding of the molecular and cellular mechanisms associated with NAD+ in peripheral neuropathic pain will facilitate the development of novel treatment options for diverse types of peripheral neuropathic pain.
Collapse
Affiliation(s)
- Yi Dai
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, PR China
| | - Jiaqi Lin
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, PR China
| | - Jinxuan Ren
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, PR China
| | - Bin Zhu
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, PR China
| | - Chengwei Wu
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, PR China
| | - Lina Yu
- Department of Anesthesiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, PR China.
| |
Collapse
|
37
|
Bratkowski M, Burdett TC, Danao J, Wang X, Mathur P, Gu W, Beckstead JA, Talreja S, Yang YS, Danko G, Park JH, Walton M, Brown SP, Tegley CM, Joseph PRB, Reynolds CH, Sambashivan S. Uncompetitive, adduct-forming SARM1 inhibitors are neuroprotective in preclinical models of nerve injury and disease. Neuron 2022; 110:3711-3726.e16. [PMID: 36087583 DOI: 10.1016/j.neuron.2022.08.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 07/06/2022] [Accepted: 08/10/2022] [Indexed: 12/15/2022]
Abstract
Axon degeneration is an early pathological event in many neurological diseases. The identification of the nicotinamide adenine dinucleotide (NAD) hydrolase SARM1 as a central metabolic sensor and axon executioner presents an exciting opportunity to develop novel neuroprotective therapies that can prevent or halt the degenerative process, yet limited progress has been made on advancing efficacious inhibitors. We describe a class of NAD-dependent active-site SARM1 inhibitors that function by intercepting NAD hydrolysis and undergoing covalent conjugation with the reaction product adenosine diphosphate ribose (ADPR). The resulting small-molecule ADPR adducts are highly potent and confer compelling neuroprotection in preclinical models of neurological injury and disease, validating this mode of inhibition as a viable therapeutic strategy. Additionally, we show that the most potent inhibitor of CD38, a related NAD hydrolase, also functions by the same mechanism, further underscoring the broader applicability of this mechanism in developing therapies against this class of enzymes.
Collapse
Affiliation(s)
| | - Thomas C Burdett
- Biology Department, Nura Bio Inc., South San Francisco, CA 94080, USA
| | - Jean Danao
- Biology Department, Nura Bio Inc., South San Francisco, CA 94080, USA
| | - Xidao Wang
- Biology Department, Nura Bio Inc., South San Francisco, CA 94080, USA
| | - Prakhyat Mathur
- Biology Department, Nura Bio Inc., South San Francisco, CA 94080, USA
| | - Weijing Gu
- Biology Department, Nura Bio Inc., South San Francisco, CA 94080, USA
| | | | - Santosh Talreja
- Biology Department, Nura Bio Inc., South San Francisco, CA 94080, USA
| | - Yu-San Yang
- Biology Department, Nura Bio Inc., South San Francisco, CA 94080, USA
| | - Gregory Danko
- Biology Department, Nura Bio Inc., South San Francisco, CA 94080, USA
| | - Jae Hong Park
- Biology Department, Nura Bio Inc., South San Francisco, CA 94080, USA
| | - Mary Walton
- Chemistry Department, Nura Bio Inc., South San Francisco, CA 94080, USA
| | - Sean P Brown
- Chemistry Department, Nura Bio Inc., South San Francisco, CA 94080, USA
| | | | - Prem Raj B Joseph
- WuXi AppTec, Research Services Division, 6 Cedarbrook Drive, Cranbury, NJ 08512, USA
| | | | | |
Collapse
|
38
|
Eastman S, Bayless A, Guo M. The Nucleotide Revolution: Immunity at the Intersection of Toll/Interleukin-1 Receptor Domains, Nucleotides, and Ca 2. MOLECULAR PLANT-MICROBE INTERACTIONS : MPMI 2022; 35:964-976. [PMID: 35881867 DOI: 10.1094/mpmi-06-22-0132-cr] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The discovery of the enzymatic activity of the toll/interleukin-1 receptor (TIR) domain protein SARM1 five years ago preceded a flood of discoveries regarding the nucleotide substrates and products of TIR domains in plants, animals, bacteria, and archaea. These discoveries into the activity of TIR domains coincide with major advances in understanding the structure and mechanisms of NOD-like receptors and the mutual dependence of pattern recognition receptor- and effector-triggered immunity (PTI and ETI, respectively) in plants. It is quickly becoming clear that TIR domains and TIR-produced nucleotides are ancestral signaling molecules that modulate immunity and that their activity is closely associated with Ca2+ signaling. TIR domain research now bridges the separate disciplines of molecular plant- and animal-microbe interactions, neurology, and prokaryotic immunity. A cohesive framework for understanding the role of enzymatic TIR domains in diverse organisms will help unite the research of these disparate fields. Here, we review known products of TIR domains in plants, animals, bacteria, and archaea and use context gained from animal and prokaryotic TIR domain systems to present a model for TIR domains, nucleotides, and Ca2+ at the intersection of PTI and ETI in plant immunity. [Formula: see text] Copyright © 2022 The Author(s). This is an open access article distributed under the CC BY 4.0 International license.
Collapse
Affiliation(s)
- Samuel Eastman
- Department of Plant Pathology, University of Nebraska-Lincoln, Lincoln, NE 68583, U.S.A
| | - Adam Bayless
- Department of Biology, Colorado State University, Fort Collins, CO 80521, U.S.A
| | - Ming Guo
- Department of Agriculture and Horticulture, University of Nebraska-Lincoln, Lincoln, NE 68583, U.S.A
| |
Collapse
|
39
|
Alexandris AS, Ryu J, Rajbhandari L, Harlan R, McKenney J, Wang Y, Aja S, Graham D, Venkatesan A, Koliatsos VE. Protective effects of NAMPT or MAPK inhibitors and NaR on Wallerian degeneration of mammalian axons. Neurobiol Dis 2022; 171:105808. [PMID: 35779777 PMCID: PMC10621467 DOI: 10.1016/j.nbd.2022.105808] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/14/2022] [Accepted: 06/25/2022] [Indexed: 01/23/2023] Open
Abstract
Wallerian degeneration (WD) is a conserved axonal self-destruction program implicated in several neurological diseases. WD is driven by the degradation of the NAD+ synthesizing enzyme NMNAT2, the buildup of its substrate NMN, and the activation of the NAD+ degrading SARM1, eventually leading to axonal fragmentation. The regulation and amenability of these events to therapeutic interventions remain unclear. Here we explored pharmacological strategies that modulate NMN and NAD+ metabolism, namely the inhibition of the NMN-synthesizing enzyme NAMPT, activation of the nicotinic acid riboside (NaR) salvage pathway and inhibition of the NMNAT2-degrading DLK MAPK pathway in an axotomy model in vitro. Results show that NAMPT and DLK inhibition cause a significant but time-dependent delay of WD. These time-dependent effects are related to NMNAT2 degradation and changes in NMN and NAD+ levels. Supplementation of NAMPT inhibition with NaR has an enhanced effect that does not depend on timing of intervention and leads to robust protection up to 4 days. Additional DLK inhibition extends this even further to 6 days. Metabolite analyses reveal complex effects indicating that NAMPT and MAPK inhibition act by reducing NMN levels, ameliorating NAD+ loss and suppressing SARM1 activity. Finally, the axonal NAD+/NMN ratio is highly predictive of cADPR levels, extending previous cell-free evidence on the allosteric regulation of SARM1. Our findings establish a window of axon protection extending several hours following injury. Moreover, we show prolonged protection by mixed treatments combining MAPK and NAMPT inhibition that proceed via complex effects on NAD+ metabolism and inhibition of SARM1.
Collapse
Affiliation(s)
| | - Jiwon Ryu
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Labchan Rajbhandari
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert Harlan
- The Molecular Determinants Center and Core, Johns Hopkins All Children's Hospital, St. Petersburg, FL, USA
| | - James McKenney
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yiqing Wang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Susan Aja
- The Molecular Determinants Center and Core, Johns Hopkins All Children's Hospital, St. Petersburg, FL, USA
| | - David Graham
- The Molecular Determinants Center and Core, Johns Hopkins All Children's Hospital, St. Petersburg, FL, USA
| | - Arun Venkatesan
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vassilis E Koliatsos
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
40
|
Waller TJ, Collins CA. Multifaceted roles of SARM1 in axon degeneration and signaling. Front Cell Neurosci 2022; 16:958900. [PMID: 36090788 PMCID: PMC9453223 DOI: 10.3389/fncel.2022.958900] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/09/2022] [Indexed: 12/01/2022] Open
Abstract
Axons are considered to be particularly vulnerable components of the nervous system; impairments to a neuron’s axon leads to an effective silencing of a neuron’s ability to communicate with other cells. Nervous systems have therefore evolved plasticity mechanisms for adapting to axonal damage. These include acute mechanisms that promote the degeneration and clearance of damaged axons and, in some cases, the initiation of new axonal growth and synapse formation to rebuild lost connections. Here we review how these diverse processes are influenced by the therapeutically targetable enzyme SARM1. SARM1 catalyzes the breakdown of NAD+, which, when unmitigated, can lead to rundown of this essential metabolite and axonal degeneration. SARM1’s enzymatic activity also triggers the activation of downstream signaling pathways, which manifest numerous functions for SARM1 in development, innate immunity and responses to injury. Here we will consider the multiple intersections between SARM1 and the injury signaling pathways that coordinate cellular adaptations to nervous system damage.
Collapse
Affiliation(s)
- Thomas J. Waller
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| | - Catherine A. Collins
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH, United States
- *Correspondence: Catherine A. Collins,
| |
Collapse
|
41
|
Poljšak B, Kovač V, Milisav I. Current Uncertainties and Future Challenges Regarding NAD+ Boosting Strategies. Antioxidants (Basel) 2022; 11:1637. [PMID: 36139711 PMCID: PMC9495723 DOI: 10.3390/antiox11091637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 11/23/2022] Open
Abstract
Precursors of nicotinamide adenine dinucleotide (NAD+), modulators of enzymes of the NAD+ biosynthesis pathways and inhibitors of NAD+ consuming enzymes, are the main boosters of NAD+. Increasing public awareness and interest in anti-ageing strategies and health-promoting lifestyles have grown the interest in the use of NAD+ boosters as dietary supplements, both in scientific circles and among the general population. Here, we discuss the current trends in NAD+ precursor usage as well as the uncertainties in dosage, timing, safety, and side effects. There are many unknowns regarding pharmacokinetics and pharmacodynamics, particularly bioavailability, metabolism, and tissue specificity of NAD+ boosters. Given the lack of long-term safety studies, there is a need for more clinical trials to determine the proper dose of NAD+ boosters and treatment duration for aging prevention and as disease therapy. Further research will also need to address the long-term consequences of increased NAD+ and the best approaches and combinations to increase NAD+ levels. The answers to the above questions will contribute to the more efficient and safer use of NAD+ boosters.
Collapse
Affiliation(s)
- Borut Poljšak
- Laboratory of Oxidative Stress Research, Faculty of Health Sciences, University of Ljubljana, Zdravstvena pot 5, SI-1000 Ljubljana, Slovenia
| | - Vito Kovač
- Laboratory of Oxidative Stress Research, Faculty of Health Sciences, University of Ljubljana, Zdravstvena pot 5, SI-1000 Ljubljana, Slovenia
| | - Irina Milisav
- Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Zaloska 4, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
42
|
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
|
43
|
Two-pore channels: going with the flows. Biochem Soc Trans 2022; 50:1143-1155. [PMID: 35959977 PMCID: PMC9444070 DOI: 10.1042/bst20220229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/21/2022] [Accepted: 07/25/2022] [Indexed: 11/26/2022]
Abstract
In recent years, our understanding of the structure, mechanisms and functions of the endo-lysosomal TPC (two-pore channel) family have grown apace. Gated by the second messengers, NAADP and PI(3,5)P2, TPCs are an integral part of fundamental signal-transduction pathways, but their array and plasticity of cation conductances (Na+, Ca2+, H+) allow them to variously signal electrically, osmotically or chemically. Their relative tissue- and organelle-selective distribution, together with agonist-selective ion permeabilities provides a rich palette from which extracellular stimuli can choose. TPCs are emerging as mediators of immunity, cancer, metabolism, viral infectivity and neurodegeneration as this short review attests.
Collapse
|
44
|
Pozo Devoto VM, Onyango IG, Stokin GB. Mitochondrial behavior when things go wrong in the axon. Front Cell Neurosci 2022; 16:959598. [PMID: 35990893 PMCID: PMC9389222 DOI: 10.3389/fncel.2022.959598] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Axonal homeostasis is maintained by processes that include cytoskeletal regulation, cargo transport, synaptic activity, ionic balance, and energy supply. Several of these processes involve mitochondria to varying degrees. As a transportable powerplant, the mitochondria deliver ATP and Ca2+-buffering capabilities and require fusion/fission to maintain proper functioning. Taking into consideration the long distances that need to be covered by mitochondria in the axons, their transport, distribution, fusion/fission, and health are of cardinal importance. However, axonal homeostasis is disrupted in several disorders of the nervous system, or by traumatic brain injury (TBI), where the external insult is translated into physical forces that damage nervous tissue including axons. The degree of damage varies and can disconnect the axon into two segments and/or generate axonal swellings in addition to cytoskeletal changes, membrane leakage, and changes in ionic composition. Cytoskeletal changes and increased intra-axonal Ca2+ levels are the main factors that challenge mitochondrial homeostasis. On the other hand, a proper function and distribution of mitochondria can determine the recovery or regeneration of the axonal physiological state. Here, we discuss the current knowledge regarding mitochondrial transport, fusion/fission, and Ca2+ regulation under axonal physiological or pathological conditions.
Collapse
Affiliation(s)
- Victorio M. Pozo Devoto
- Translational Neuroscience and Ageing Program, Centre for Translational Medicine, International Clinical Research Centre, St. Anne's University Hospital, Brno, Czechia
| | - Isaac G. Onyango
- Translational Neuroscience and Ageing Program, Centre for Translational Medicine, International Clinical Research Centre, St. Anne's University Hospital, Brno, Czechia
| | - Gorazd B. Stokin
- Translational Neuroscience and Ageing Program, Centre for Translational Medicine, International Clinical Research Centre, St. Anne's University Hospital, Brno, Czechia
- Division of Neurology, University Medical Centre, Ljubljana, Slovenia
- Department of Neurosciences, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
45
|
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
|
46
|
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: 9] [Impact Index Per Article: 3.0] [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
|
47
|
Icso JD, Thompson PR. The chemical biology of NAD + regulation in axon degeneration. Curr Opin Chem Biol 2022; 69:102176. [PMID: 35780654 PMCID: PMC10084848 DOI: 10.1016/j.cbpa.2022.102176] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/24/2022] [Accepted: 05/27/2022] [Indexed: 11/26/2022]
Abstract
During axon degeneration, NAD+ levels are largely controlled by two enzymes: nicotinamide mononucleotide adenylyltransferase 2 (NMNAT2) and sterile alpha and toll interleukin motif containing protein 1 (SARM1). NMNAT2, which catalyzes the formation of NAD+ from NMN and ATP, is actively degraded leading to decreased NAD+ levels. SARM1 activity further decreases the concentration of NAD+ by catalyzing its hydrolysis to form nicotinamide and a mixture of ADPR and cADPR. Notably, SARM1 knockout mice show decreased neurodegeneration in animal models of axon degeneration, highlighting the therapeutic potential of targeting this novel NAD+ hydrolase. This review discusses recent advances in the SARM1 field, including SARM1 structure, regulation, and catalysis as well as the identification of the first SARM1 inhibitors.
Collapse
Affiliation(s)
- Janneke D Icso
- Program in Chemical Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA; Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Paul R Thompson
- Program in Chemical Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA; Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
48
|
Ryu WI, Shen M, Lee Y, Healy RA, Bormann MK, Cohen BM, Sonntag KC. Nicotinamide riboside and caffeine partially restore diminished NAD availability but not altered energy metabolism in Alzheimer's disease. Aging Cell 2022; 21:e13658. [PMID: 35730144 PMCID: PMC9282847 DOI: 10.1111/acel.13658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 05/20/2022] [Accepted: 06/02/2022] [Indexed: 12/03/2022] Open
Abstract
The redox co‐factor nicotinamide adenine dinucleotide (NAD) declines with age, and NAD deficits are specifically associated with dysfunctional energy metabolism in late‐onset Alzheimer's disease (LOAD). Nicotinamide riboside (NR), a dietary NAD precursor, has been suggested to ameliorate the aging process or neurodegeneration. We assessed whether NR with or without caffeine, which increases nicotinamide mononucleotide transferase subtype 2 (NMNAT2), an essential enzyme in NAD production, modulates bioenergetic functions in LOAD. In LOAD patients—and young or old control individuals—derived dermal fibroblasts as well as in induced pluripotent stem cell‐differentiated neural progenitors and astrocytes, NR and caffeine cell type‐specifically increased the NAD pool, transiently enhanced mitochondrial respiration or glycolysis and altered the expression of genes in the NAD synthesis or consumption pathways. However, continued treatment led to reversed bioenergetic effects. Importantly, NR and caffeine did not alter the characteristics of a previously documented inherent LOAD‐associated bioenergetic phenotype. Thus, although NR and caffeine can partially restore diminished NAD availability, increasing NAD alone may not be sufficient to boost or restore energy metabolism in brain aging or alter aberrant energy management in LOAD. Nicotinamide riboside might still be of value in combination with other agents in preventive or therapeutic intervention strategies to address the aging process or age‐associated dementia.
Collapse
Affiliation(s)
- Woo-In Ryu
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA.,Basic Neuroscience Division, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA.,Program for Neuropsychiatric Research, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA
| | - Minqi Shen
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA.,Basic Neuroscience Division, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA.,Program for Neuropsychiatric Research, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA
| | - Yoon Lee
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA.,Basic Neuroscience Division, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA.,Program for Neuropsychiatric Research, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA
| | - Ryan A Healy
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA.,Basic Neuroscience Division, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA.,Program for Neuropsychiatric Research, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA
| | - Mariana K Bormann
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA.,Basic Neuroscience Division, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA.,Program for Neuropsychiatric Research, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA
| | - Bruce M Cohen
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA.,Program for Neuropsychiatric Research, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA
| | - Kai-Christian Sonntag
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA.,Basic Neuroscience Division, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA.,Program for Neuropsychiatric Research, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA
| |
Collapse
|
49
|
Shi Y, Kerry PS, Nanson JD, Bosanac T, Sasaki Y, Krauss R, Saikot FK, Adams SE, Mosaiab T, Masic V, Mao X, Rose F, Vasquez E, Furrer M, Cunnea K, Brearley A, Gu W, Luo Z, Brillault L, Landsberg MJ, DiAntonio A, Kobe B, Milbrandt J, Hughes RO, Ve T. Structural basis of SARM1 activation, substrate recognition, and inhibition by small molecules. Mol Cell 2022; 82:1643-1659.e10. [PMID: 35334231 PMCID: PMC9188649 DOI: 10.1016/j.molcel.2022.03.007] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 01/11/2022] [Accepted: 03/01/2022] [Indexed: 01/04/2023]
Abstract
The NADase SARM1 (sterile alpha and TIR motif containing 1) is a key executioner of axon degeneration and a therapeutic target for several neurodegenerative conditions. We show that a potent SARM1 inhibitor undergoes base exchange with the nicotinamide moiety of nicotinamide adenine dinucleotide (NAD+) to produce the bona fide inhibitor 1AD. We report structures of SARM1 in complex with 1AD, NAD+ mimetics and the allosteric activator nicotinamide mononucleotide (NMN). NMN binding triggers reorientation of the armadillo repeat (ARM) domains, which disrupts ARM:TIR interactions and leads to formation of a two-stranded TIR domain assembly. The active site spans two molecules in these assemblies, explaining the requirement of TIR domain self-association for NADase activity and axon degeneration. Our results reveal the mechanisms of SARM1 activation and substrate binding, providing rational avenues for the design of new therapeutics targeting SARM1.
Collapse
Affiliation(s)
- Yun Shi
- Institute for Glycomics, Griffith University, Southport, QLD 4222, Australia
| | - Philip S Kerry
- Evotec (UK) Ltd., 114 Innovation Drive, Milton Park, Abingdon, Oxfordshire OX14 4RZ, UK
| | - Jeffrey D Nanson
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, QLD 4072, Australia
| | - Todd Bosanac
- Disarm Therapeutics, a wholly-owned subsidiary of Eli Lilly & Co., Cambridge, MA, 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
| | - Raul Krauss
- Disarm Therapeutics, a wholly-owned subsidiary of Eli Lilly & Co., Cambridge, MA, USA
| | - Forhad K Saikot
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, QLD 4072, Australia
| | - Sarah E Adams
- Evotec (UK) Ltd., 114 Innovation Drive, Milton Park, Abingdon, Oxfordshire OX14 4RZ, UK
| | - Tamim Mosaiab
- Institute for Glycomics, Griffith University, Southport, QLD 4222, Australia
| | - Veronika Masic
- Institute for Glycomics, Griffith University, Southport, QLD 4222, Australia
| | - 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
| | - Faith Rose
- Institute for Glycomics, Griffith University, Southport, QLD 4222, Australia
| | - Eduardo Vasquez
- Institute for Glycomics, Griffith University, Southport, QLD 4222, Australia
| | - Marieke Furrer
- Evotec SE, Manfred Eigen Campus, Essener Bogen 7, 22419 Hamburg, Germany
| | - Katie Cunnea
- Evotec (UK) Ltd., 114 Innovation Drive, Milton Park, Abingdon, Oxfordshire OX14 4RZ, UK
| | - Andrew Brearley
- Evotec (UK) Ltd., 114 Innovation Drive, Milton Park, Abingdon, Oxfordshire OX14 4RZ, UK
| | - Weixi Gu
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, QLD 4072, Australia
| | - Zhenyao Luo
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, QLD 4072, Australia
| | - Lou Brillault
- Centre for Microscopy and Microanalysis, The University of Queensland, Brisbane, Australia
| | - Michael J Landsberg
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, QLD 4072, Australia
| | - 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
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, QLD 4072, Australia
| | - 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
| | - Robert O Hughes
- Disarm Therapeutics, a wholly-owned subsidiary of Eli Lilly & Co., Cambridge, MA, USA.
| | - Thomas Ve
- Institute for Glycomics, Griffith University, Southport, QLD 4222, Australia.
| |
Collapse
|
50
|
Crawford CL, Antoniou C, Komarek L, Schultz V, Donald CL, Montague P, Barnett SC, Linington C, Willison HJ, Kohl A, Coleman MP, Edgar JM. SARM1 Depletion Slows Axon Degeneration in a CNS Model of Neurotropic Viral Infection. Front Mol Neurosci 2022; 15:860410. [PMID: 35493328 PMCID: PMC9043327 DOI: 10.3389/fnmol.2022.860410] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 02/18/2022] [Indexed: 01/30/2023] Open
Abstract
Zika virus (ZIKV) is a neurotropic flavivirus recently linked to congenital ZIKV syndrome in children and encephalitis and Guillain-Barré syndrome in adults. Neurotropic viruses often use axons to traffic to neuronal or glial cell somas where they either remain latent or replicate and proceed to infect new cells. Consequently, it has been suggested that axon degeneration could represent an evolutionarily conserved mechanism to limit viral spread. Whilst it is not known if ZIKV transits in axons, we previously reported that ZIKV infection of glial cells in a murine spinal cord-derived cell culture model of the CNS is associated with a profound loss of neuronal cell processes. This, despite that postmitotic neurons are relatively refractory to infection and death. Here, we tested the hypothesis that ZIKV-associated degeneration of neuronal processes is dependent on activation of Sterile alpha and armadillo motif-containing protein 1 (SARM1), an NADase that acts as a central executioner in a conserved axon degeneration pathway. To test this, we infected wild type and Sarm1 homozygous or heterozygous null cell cultures with ZIKV and examined NAD+ levels as well as the survival of neurons and their processes. Unexpectedly, ZIKV infection led to a rapid SARM1-independent reduction in NAD+. Nonetheless, the subsequent profound loss of neuronal cell processes was SARM1-dependent and was preceded by early changes in the appearance of β-tubulin III staining. Together, these data identify a role for SARM1 in the pathogenesis of ZIKV infection, which may reflect SARM1's conserved prodegenerative function, independent of its NADase activity.
Collapse
Affiliation(s)
- Colin L. Crawford
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | | | - Lina Komarek
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Verena Schultz
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Claire L. Donald
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Paul Montague
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Susan C. Barnett
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Christopher Linington
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Hugh J. Willison
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Alain Kohl
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Michael P. Coleman
- John van Geest Centre for Brain Repair, Cambridge, United Kingdom
- Michael P. Coleman
| | - Julia M. Edgar
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
- *Correspondence: Julia M. Edgar
| |
Collapse
|