1
|
Chen Y, Deng H, Zhang N. Autophagy-targeting modulation to promote peripheral nerve regeneration. Neural Regen Res 2025; 20:1864-1882. [PMID: 39254547 DOI: 10.4103/nrr.nrr-d-23-01948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 03/29/2024] [Indexed: 09/11/2024] Open
Abstract
Nerve regeneration following traumatic peripheral nerve injuries and neuropathies is a complex process modulated by diverse factors and intricate molecular mechanisms. Past studies have focused on factors that stimulate axonal outgrowth and myelin regeneration. However, recent studies have highlighted the pivotal role of autophagy in peripheral nerve regeneration, particularly in the context of traumatic injuries. Consequently, autophagy-targeting modulation has emerged as a promising therapeutic approach to enhancing peripheral nerve regeneration. Our current understanding suggests that activating autophagy facilitates the rapid clearance of damaged axons and myelin sheaths, thereby enhancing neuronal survival and mitigating injury-induced oxidative stress and inflammation. These actions collectively contribute to creating a favorable microenvironment for structural and functional nerve regeneration. A range of autophagy-inducing drugs and interventions have demonstrated beneficial effects in alleviating peripheral neuropathy and promoting nerve regeneration in preclinical models of traumatic peripheral nerve injuries. This review delves into the regulation of autophagy in cell types involved in peripheral nerve regeneration, summarizing the potential drugs and interventions that can be harnessed to promote this process. We hope that our review will offer novel insights and perspectives on the exploitation of autophagy pathways in the treatment of peripheral nerve injuries and neuropathies.
Collapse
Affiliation(s)
- Yan Chen
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Key Laboratory of Birth Defects and Women and Children's Diseases, Ministry of Education, Sichuan University, Chengdu, Sichuan Province, China
- Laboratory of Reproductive Endocrinology and Reproductive Regulation, Sichuan University, Chengdu, Sichuan Province, China
| | - Hongxia Deng
- Key Laboratory of Birth Defects and Women and Children's Diseases, Ministry of Education, Sichuan University, Chengdu, Sichuan Province, China
- Laboratory of Reproductive Endocrinology and Reproductive Regulation, Sichuan University, Chengdu, Sichuan Province, China
| | - Nannan Zhang
- Key Laboratory of Birth Defects and Women and Children's Diseases, Ministry of Education, Sichuan University, Chengdu, Sichuan Province, China
- National Center for Birth Defect Monitoring, West China Second University Hospital, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
2
|
Hardy PB, Wang BY, Chan KM, Webber CA, Senger JLB. The use of electrical stimulation to enhance recovery following peripheral nerve injury. Muscle Nerve 2024; 70:1151-1162. [PMID: 39347555 DOI: 10.1002/mus.28262] [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: 01/17/2024] [Revised: 09/04/2024] [Accepted: 09/07/2024] [Indexed: 10/01/2024]
Abstract
Peripheral nerve injury is common and can have devastating consequences. In severe cases, functional recovery is often poor despite surgery. This is primarily due to the exceedingly slow rate of nerve regeneration at only 1-3 mm/day. The local environment in the distal nerve stump supportive of nerve regrowth deteriorates over time and the target end organs become atrophic. To overcome these challenges, investigations into treatments capable of accelerating nerve regrowth are of great clinical relevance and are an active area of research. One intervention that has shown great promise is perioperative electrical stimulation. Postoperative stimulation helps to expedite the Wallerian degeneration process and reduces delays caused by staggered regeneration at the site of nerve injury. By contrast, preoperative "conditioning" stimulation increases the rate of nerve regrowth along the nerve trunk. Over the past two decades, a rich body of literature has emerged that provides molecular insights into the mechanism by which electrical stimulation impacts nerve regeneration. The end result is upregulation of regeneration-associated genes in the neuronal body and accelerated transport to the axon front for regrowth. The efficacy of brief electrical stimulation on patients with peripheral nerve injuries was demonstrated in a number of randomized controlled trials on compressive, transection and traction injuries. As approved equipment to deliver this treatment is becoming available, it may be feasible to deploy this novel treatment in a wide range of clinical settings.
Collapse
Affiliation(s)
- Paige B Hardy
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Bonnie Y Wang
- Division of Physical Medicine and Rehabilitation, University of Alberta, Edmonton, Alberta, Canada
| | - K Ming Chan
- Division of Physical Medicine and Rehabilitation, University of Alberta, Edmonton, Alberta, Canada
| | | | - Jenna-Lynn B Senger
- Division of Plastic & Reconstructive Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
3
|
Ye Y, Song F. SARM1 in the pathogenesis of immune-related disease. Toxicol Res (Camb) 2024; 13:tfae208. [PMID: 39664502 PMCID: PMC11631086 DOI: 10.1093/toxres/tfae208] [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: 02/21/2024] [Revised: 09/10/2024] [Accepted: 12/02/2024] [Indexed: 12/13/2024] Open
Abstract
Background Sterile alpha and toll interleukin receptor motif-containing protein 1 (SARM1) are primarily expressed in the mammalian nervous system, with their presence in neurons being associated with mitochondrial aggregation. SARM1 functions as a mediator of cell death and morphological changes, while also regulating Waller degeneration in nerve fibers and influencing glial cell formation. Purpose Recent reports demonstrate SARM1 serves as a connector in the Toll-like receptor (TLR) pathway and plays a role in regulating inflammation during periods of stress such as infection, trauma, and hypoxia. These findings offer new insights into pathogenesis research and the prevention and treatment of neurodegenerative diseases and pathogen infections. Methods This review synthesizes recent findings on the immune-related mechanisms of SARM1, emphasizing its roles in inflammation and its functional impact on the nervous system and other bodily systems. Conclusions Understanding the multifaceted roles of SARM1 in immune regulation and neuronal health provides novel insights into its involvement in disease pathogenesis. These insights hold promise for advancing research into the prevention and treatment of neurodegenerative diseases and pathogen-induced conditions.
Collapse
Affiliation(s)
- Yihan Ye
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, P. R. China
| | - Fuyong Song
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, P. R. China
| |
Collapse
|
4
|
Avetisyan A, Barria R, Sheehan A, Freeman MR. An Ionic Sensor acts in Parallel to dSarm to Promote Neurodegeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.29.620922. [PMID: 39651259 PMCID: PMC11623519 DOI: 10.1101/2024.10.29.620922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
How neurons to sense when they are terminally dysfunctional and activate neurodegeneration remains poorly defined. The pro-degenerative NAD + hydrolase dSarm/SARM1 can act as a metabolic sensor by detecting pathological changes in NAD + /NMN and subsequently induce catastrophic axon degeneration. Here we show Drosophila with-no-lysine kinase (dWnk), which can directly sense Cl - , K + and osmotic pressure, is required for neurodegeneration induced by depletion of the NAD + biosynthetic enzyme dNmnat. dWnk functions in parallel to dSarm and acts through the downstream kinase Frayed to promote axon degeneration and neuronal cell death. dWnk and dSarm ultimately converge on the BTB-Back domain molecule Axundead (Axed) to execute neurodegeneration. Our work argues that neurons use direct sensors of both metabolism (dSarm/SARM1) and ionic/osmotic status (dWnk) to evaluate cellular health and, when dysfunctional, promote neurodegeneration though a common axon death signaling molecule, Axundead.
Collapse
|
5
|
Paglione M, Restivo L, Zakhia S, Llobet Rosell A, Terenzio M, Neukomm LJ. Local translatome sustains synaptic function in impaired Wallerian degeneration. EMBO Rep 2024:10.1038/s44319-024-00301-8. [PMID: 39482489 DOI: 10.1038/s44319-024-00301-8] [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: 02/10/2024] [Revised: 10/07/2024] [Accepted: 10/17/2024] [Indexed: 11/03/2024] Open
Abstract
After injury, severed axons separated from their somas activate programmed axon degeneration, a conserved pathway to initiate their degeneration within a day. Conversely, severed projections deficient in programmed axon degeneration remain morphologically preserved with functional synapses for weeks to months after axotomy. How this synaptic function is sustained remains currently unknown. Here, we show that dNmnat overexpression attenuates programmed axon degeneration in distinct neuronal populations. Severed projections remain morphologically preserved for weeks. When evoked, they elicit a postsynaptic behavior, a readout for preserved synaptic function. We used ribosomal pulldown to isolate the translatome from these projections 1 week after axotomy. Translatome candidates of enriched biological classes identified by transcriptional profiling are validated in a screen using a novel automated system to detect evoked antennal grooming as a proxy for preserved synaptic function. RNAi-mediated knockdown reveals that transcripts of the mTORC1 pathway, a mediator of protein synthesis, and of candidate genes involved in protein ubiquitination and Ca2+ homeostasis are required for preserved synaptic function. Our translatome dataset also uncovers several uncharacterized Drosophila genes associated with human disease. It may offer insights into novel avenues for therapeutic treatments.
Collapse
Affiliation(s)
- Maria Paglione
- Department of Fundamental Neurosciences, University of Lausanne, 1005, Lausanne, Switzerland
- Lemanic Neuroscience Doctoral School (LNDS), Lausanne, Switzerland
| | - Leonardo Restivo
- Department of Fundamental Neurosciences, University of Lausanne, 1005, Lausanne, Switzerland
| | - Sarah Zakhia
- Molecular Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, Kunigami-gun, Okinawa, 904-0412, Japan
| | - Arnau Llobet Rosell
- Department of Fundamental Neurosciences, University of Lausanne, 1005, Lausanne, Switzerland
| | - Marco Terenzio
- Molecular Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, Kunigami-gun, Okinawa, 904-0412, Japan
| | - Lukas J Neukomm
- Department of Fundamental Neurosciences, University of Lausanne, 1005, Lausanne, Switzerland.
| |
Collapse
|
6
|
Asadollahi E, Trevisiol A, Saab AS, Looser ZJ, Dibaj P, Ebrahimi R, Kusch K, Ruhwedel T, Möbius W, Jahn O, Lee JY, Don AS, Khalil MA, Hiller K, Baes M, Weber B, Abel ED, Ballabio A, Popko B, Kassmann CM, Ehrenreich H, Hirrlinger J, Nave KA. Oligodendroglial fatty acid metabolism as a central nervous system energy reserve. Nat Neurosci 2024; 27:1934-1944. [PMID: 39251890 PMCID: PMC11452346 DOI: 10.1038/s41593-024-01749-6] [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: 06/22/2023] [Accepted: 08/05/2024] [Indexed: 09/11/2024]
Abstract
Brain function requires a constant supply of glucose. However, the brain has no known energy stores, except for glycogen granules in astrocytes. In the present study, we report that continuous oligodendroglial lipid metabolism provides an energy reserve in white matter tracts. In the isolated optic nerve from young adult mice of both sexes, oligodendrocytes survive glucose deprivation better than astrocytes. Under low glucose, both axonal ATP levels and action potentials become dependent on fatty acid β-oxidation. Importantly, ongoing oligodendroglial lipid degradation feeds rapidly into white matter energy metabolism. Although not supporting high-frequency spiking, fatty acid β-oxidation in mitochondria and oligodendroglial peroxisomes protects axons from conduction blocks when glucose is limiting. Disruption of the glucose transporter GLUT1 expression in oligodendrocytes of adult mice perturbs myelin homeostasis in vivo and causes gradual demyelination without behavioral signs. This further suggests that the imbalance of myelin synthesis and degradation can underlie myelin thinning in aging and disease.
Collapse
Affiliation(s)
- Ebrahim Asadollahi
- Max Planck Institute for Multidisciplinary Sciences, Department of Neurogenetics, Göttingen, Germany.
| | - Andrea Trevisiol
- Max Planck Institute for Multidisciplinary Sciences, Department of Neurogenetics, Göttingen, Germany
- University of Toronto, Sunnybrook Health Sciences Centre, Department of Physical Sciences, North York, Ontario, Canada
| | - Aiman S Saab
- Max Planck Institute for Multidisciplinary Sciences, Department of Neurogenetics, Göttingen, Germany
- University of Zurich, Institute of Pharmacology and Toxicology, Zurich, Switzerland
| | - Zoe J Looser
- University of Zurich, Institute of Pharmacology and Toxicology, Zurich, Switzerland
| | - Payam Dibaj
- Max Planck Institute for Multidisciplinary Sciences, Department of Neurogenetics, Göttingen, Germany
- Center for Rare Diseases Göttingen, Department of Pediatrics and Pediatric Neurology, Georg August University Göttingen, Göttingen, Germany
| | - Reyhane Ebrahimi
- Max Planck Institute for Multidisciplinary Sciences, Department of Neurogenetics, Göttingen, Germany
| | - Kathrin Kusch
- Max Planck Institute for Multidisciplinary Sciences, Department of Neurogenetics, Göttingen, Germany
- University of Göttingen Medical School, Institute for Auditory Neuroscience and Inner Ear Lab, Göttingen, Germany
| | - Torben Ruhwedel
- Max Planck Institute for Multidisciplinary Sciences, Department of Neurogenetics, Göttingen, Germany
| | - Wiebke Möbius
- Max Planck Institute for Multidisciplinary Sciences, Department of Neurogenetics, Göttingen, Germany
| | - Olaf Jahn
- Max Planck Institute for Multidisciplinary Sciences, Department of Molecular Neurobiology, Neuroproteomics Group, Göttingen, Germany
- University Medical Center Göttingen, Department of Psychiatry and Psychotherapy, Translational Neuroproteomics Group, Göttingen, Germany
| | - Jun Yup Lee
- School of Medical Sciences and Charles Perkins Centre, The University of Sydney, Camperdown, New South Wales, Australia
| | - Anthony S Don
- School of Medical Sciences and Charles Perkins Centre, The University of Sydney, Camperdown, New South Wales, Australia
| | - Michelle-Amirah Khalil
- Department for Bioinformatics and Biochemistry, Braunschweig Integrated Center of System Biology, Technische Universität Braunschweig, Braunschweig, Germany
| | - Karsten Hiller
- Department for Bioinformatics and Biochemistry, Braunschweig Integrated Center of System Biology, Technische Universität Braunschweig, Braunschweig, Germany
| | - Myriam Baes
- Lab of Cell Metabolism, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Bruno Weber
- University of Zurich, Institute of Pharmacology and Toxicology, Zurich, Switzerland
| | - E Dale Abel
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine, Naples, Italy
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Brian Popko
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Celia M Kassmann
- Max Planck Institute for Multidisciplinary Sciences, Department of Neurogenetics, Göttingen, Germany
| | - Hannelore Ehrenreich
- Max Planck Institute for Multidisciplinary Sciences, Clinical Neuroscience, Göttingen, Germany
- Central Institute of Mental Health, Mannheim, Germany
| | - Johannes Hirrlinger
- Max Planck Institute for Multidisciplinary Sciences, Department of Neurogenetics, Göttingen, Germany
- Carl-Ludwig-Institute for Physiology, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Klaus-Armin Nave
- Max Planck Institute for Multidisciplinary Sciences, Department of Neurogenetics, Göttingen, Germany.
| |
Collapse
|
7
|
Knoerl R, Mazzola E, Pazyra-Murphy M, Ryback B, Frazier AL, Freeman RL, Hammer M, LaCasce A, Ligibel J, Luskin MR, Berry DL, Segal RA. Exploring clinical markers of Axon degeneration processes in Chemotherapy-induced peripheral neuropathy among young adults receiving vincristine or paclitaxel. BMC Neurol 2024; 24:366. [PMID: 39342135 PMCID: PMC11438373 DOI: 10.1186/s12883-024-03877-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Approximately 70% of patients receiving neurotoxic chemotherapy (e.g., paclitaxel or vincristine) will develop chemotherapy-induced peripheral neuropathy. Despite the known negative effects of CIPN on physical functioning and chemotherapy dosing, little is known about how to prevent CIPN. The development of efficacious CIPN prevention interventions is hindered by the lack of knowledge surrounding CIPN mechanisms. Nicotinamide adenine dinucleotide (NAD+) and cyclic-adenosine diphosphate ribose (cADPR) are potential markers of axon degeneration following neurotoxic chemotherapy, however, such markers have been exclusively measured in preclinical models of chemotherapy-induced peripheral neuropathy (CIPN). The overall objective of this longitudinal, observational study was to determine the association between plasma NAD+, cADPR, and ADPR with CIPN severity in young adults receiving vincristine or paclitaxel. METHODS Young adults (18-39 years old) beginning vincristine or paclitaxel were recruited from Dana-Farber Cancer Institute. Young adults completed the QLQ-CIPN20 sensory and motor subscales and provided a blood sample prior to starting chemotherapy (T1) and at increasing cumulative vincristine (T2: 3-5 mg, T3: 7-9 mg) and paclitaxel (T2: 300-500 mg/m2, T3: 700-900 mg/m2) dosages. NAD+, cADPR, and ADPR were quantified from plasma using mass spectrometry. Metabolite levels and QLQ-CIPN20 scores over time were compared using mixed-effects linear regression models and/or paired two-sample tests. RESULTS Participants (N = 50) were mainly female (88%), white (80%), and receiving paclitaxel (78%). Sensory and motor CIPN severity increased from T1-T3 (p < 0.001). NAD+ (p = 0.28), cADPR (p = 0.62), and ADPR (p = 0.005) values decreased, while cADPR/NAD+ ratio increased from T1-T3 (p = 0.50). There were no statistically significant associations between NAD + and QLQ-CIPN20 scores over time. CONCLUSIONS To our knowledge, this is the first study to measure plasma NAD+, cADPR, and ADPR among patients receiving neurotoxic chemotherapy. Although, no meaningful changes in NAD+, cADPR, or cADPR/NAD+ were observed among young adults receiving paclitaxel or vincristine. Future research in an adequately powered sample is needed to explore the clinical utility of biomarkers of axon degeneration among patients receiving neurotoxic chemotherapy to guide mechanistic research and novel CIPN treatments.
Collapse
Affiliation(s)
- Robert Knoerl
- Phyllis F. Cantor Center for Research in Nursing and Patient Care Services, Dana-Farber Cancer Institute, Boston, MA, USA.
- University of Michigan School of Nursing, 400 North Ingalls St, Office 2350, Ann Arbor, MI, 48109, USA.
| | - Emanuele Mazzola
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Birgitta Ryback
- Metabolomics Core, Dana-Farber Cancer Institute, Boston, MA, USA
| | - A Lindsay Frazier
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Roy L Freeman
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Marilyn Hammer
- Phyllis F. Cantor Center for Research in Nursing and Patient Care Services, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ann LaCasce
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jennifer Ligibel
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Marlise R Luskin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Donna L Berry
- Biobehavioral Nursing and Health Informatics, University of Washington, Seattle, WA, USA
| | - Rosalind A Segal
- Deparment of Neurobiology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
8
|
Liu Y, Wang A, Chen C, Zhang Q, Shen Q, Zhang D, Xiao X, Chen S, Lian L, Le Z, Liu S, Liang T, Zheng Q, Xu P, Zou J. Microglial cGAS-STING signaling underlies glaucoma pathogenesis. Proc Natl Acad Sci U S A 2024; 121:e2409493121. [PMID: 39190350 PMCID: PMC11388346 DOI: 10.1073/pnas.2409493121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 07/24/2024] [Indexed: 08/28/2024] Open
Abstract
Characterized by progressive degeneration of retinal ganglion cells (RGCs) and vision loss, glaucoma is the primary cause of irreversible blindness, incurable and affecting over 78 million patients. However, pathogenic mechanisms leading to glaucoma-induced RGC loss are incompletely understood. Unexpectedly, we found that cGAS-STING (2'3'-cyclic GMP-AMP-stimulator of interferon genes) signaling, which surveils displaced double-stranded DNA (dsDNA) in the cytosol and initiates innate immune responses, was robustly activated during glaucoma in retinal microglia in distinct murine models. Global or microglial deletion of STING markedly relieved glaucoma symptoms and protected RGC degeneration and vision loss, while mice bearing genetic cGAS-STING supersensitivity aggravated retinal neuroinflammation and RGC loss. Mechanistically, dsDNA from tissue injury activated microglial cGAS-STING signaling, causing deleterious macroglia reactivity in retinas by cytokine-mediated microglia-macroglia interactions, progressively driving apoptotic death of RGCs. Remarkably, preclinical investigations of targeting cGAS-STING signaling by intraocular injection of TBK1i or anti-IFNAR1 antibody prevented glaucoma-induced losses of RGCs and vision. Therefore, we unravel an essential role of cGAS-STING signaling underlying glaucoma pathogenesis and suggest promising therapeutic strategies for treating this devastating disease.
Collapse
Affiliation(s)
- Yutong Liu
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, University School of Medicine, Zhejiang University, Hangzhou 310058, China
- Institute of Intelligent Medicine, Zhejiang University-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou 310058, China
- Ministry of Education Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Ailian Wang
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, University School of Medicine, Zhejiang University, Hangzhou 310058, China
- Institute of Intelligent Medicine, Zhejiang University-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou 310058, China
- Ministry of Education Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Chen Chen
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, University School of Medicine, Zhejiang University, Hangzhou 310058, China
- Institute of Intelligent Medicine, Zhejiang University-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou 310058, China
- Ministry of Education Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Qian Zhang
- Institute of Intelligent Medicine, Zhejiang University-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou 310058, China
- Ministry of Education Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Qin Shen
- Institute of Intelligent Medicine, Zhejiang University-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou 310058, China
| | - Dan Zhang
- Institute of Intelligent Medicine, Zhejiang University-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou 310058, China
| | - Xueqi Xiao
- Eye Center of the Second Affiliated Hospital School of Medicine, Center for Genetic Medicine, Zhejiang University International Institute of Medicine, Hangzhou 310029, China
| | - Shasha Chen
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Lili Lian
- National Clinical Research Center for Ocular Diseases, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Zhenmin Le
- National Clinical Research Center for Ocular Diseases, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Shengduo Liu
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, University School of Medicine, Zhejiang University, Hangzhou 310058, China
- Institute of Intelligent Medicine, Zhejiang University-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou 310058, China
- Ministry of Education Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Qinxiang Zheng
- National Clinical Research Center for Ocular Diseases, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Pinglong Xu
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, University School of Medicine, Zhejiang University, Hangzhou 310058, China
- Institute of Intelligent Medicine, Zhejiang University-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou 310058, China
- Ministry of Education Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Jian Zou
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, University School of Medicine, Zhejiang University, Hangzhou 310058, China
- Eye Center of the Second Affiliated Hospital School of Medicine, Center for Genetic Medicine, Zhejiang University International Institute of Medicine, Hangzhou 310029, China
| |
Collapse
|
9
|
Bhatt A, Mishra BP, Gu W, Sorbello M, Xu H, Ve T, Kobe B. Structural characterization of TIR-domain signalosomes through a combination of structural biology approaches. IUCRJ 2024; 11:695-707. [PMID: 39190506 PMCID: PMC11364022 DOI: 10.1107/s2052252524007693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 08/05/2024] [Indexed: 08/29/2024]
Abstract
The TIR (Toll/interleukin-1 receptor) domain represents a vital structural element shared by proteins with roles in immunity signalling pathways across phyla (from humans and plants to bacteria). Decades of research have finally led to identifying the key features of the molecular basis of signalling by these domains, including the formation of open-ended (filamentous) assemblies (responsible for the signalling by cooperative assembly formation mechanism, SCAF) and enzymatic activities involving the cleavage of nucleotides. We present a historical perspective of the research that led to this understanding, highlighting the roles that different structural methods played in this process: X-ray crystallography (including serial crystallography), microED (micro-crystal electron diffraction), NMR (nuclear magnetic resonance) spectroscopy and cryo-EM (cryogenic electron microscopy) involving helical reconstruction and single-particle analysis. This perspective emphasizes the complementarity of different structural approaches.
Collapse
Affiliation(s)
- Akansha Bhatt
- Institute for GlycomicsGriffith UniversitySouthportQLD4222Australia
- School of Pharmacy and Medical SciencesGriffith UniversitySouthportQLD4222Australia
| | - Biswa P. Mishra
- Institute for GlycomicsGriffith UniversitySouthportQLD4222Australia
| | - Weixi Gu
- School of Chemistry and Molecular BiosciencesUniversity of QueenslandBrisbaneQLD4072Australia
- Institute for Molecular BioscienceThe University of QueenslandBrisbaneQLD4072Australia
- Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQLD4072Australia
| | - Mitchell Sorbello
- School of Chemistry and Molecular BiosciencesUniversity of QueenslandBrisbaneQLD4072Australia
- Institute for Molecular BioscienceThe University of QueenslandBrisbaneQLD4072Australia
- Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQLD4072Australia
| | - Hongyi Xu
- School of Chemistry and Molecular BiosciencesUniversity of QueenslandBrisbaneQLD4072Australia
- Department of Materials and Environmental ChemistryStockholm UniversityStockholmSweden
| | - Thomas Ve
- Institute for GlycomicsGriffith UniversitySouthportQLD4222Australia
| | - Bostjan Kobe
- School of Chemistry and Molecular BiosciencesUniversity of QueenslandBrisbaneQLD4072Australia
- Institute for Molecular BioscienceThe University of QueenslandBrisbaneQLD4072Australia
- Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQLD4072Australia
| |
Collapse
|
10
|
Cimaglia G, Tribble JR, Votruba M, Williams PA, Morgan JE. Oral nicotinamide provides robust, dose-dependent structural and metabolic neuroprotection of retinal ganglion cells in experimental glaucoma. Acta Neuropathol Commun 2024; 12:137. [PMID: 39180087 PMCID: PMC11342512 DOI: 10.1186/s40478-024-01850-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 08/12/2024] [Indexed: 08/26/2024] Open
Abstract
A compromised capacity to maintain NAD pools is recognized as a key underlying pathophysiological feature of neurodegenerative diseases. NAD acts as a substrate in major cell functions including mitochondrial homeostasis, cell signalling, axonal transport, axon/Wallerian degeneration, and neuronal energy supply. Dendritic degeneration is an early marker of neuronal stress and precedes cell loss. However, little is known about dendritic structural preservation in pathologic environments and remodelling in mature neurons. Retinal ganglion cell dendritic atrophy is an early pathological feature in animal models of the disease and has been demonstrated in port-mortem human glaucoma samples. Here we report that a nicotinamide (a precursor to NAD through the NAD salvage pathway) enriched diet provides robust retinal ganglion cell dendritic protection and preserves dendritic structure in a rat model of experimental glaucoma. Metabolomic analysis of optic nerve samples from the same animals demonstrates that nicotinamide provides robust metabolic neuroprotection in glaucoma. Advances in our understanding of retinal ganglion cell metabolic profiles shed light on the energetic shift that triggers early neuronal changes in neurodegenerative diseases. As nicotinamide can improve visual function short term in existing glaucoma patients, we hypothesize that a portion of this visual recovery may be due to dendritic preservation in stressed, but not yet fully degenerated, retinal ganglion cells.
Collapse
Affiliation(s)
- Gloria Cimaglia
- School of Optometry and Vision Sciences, Cardiff University, Maindy Road, Cardiff, Wales, UK
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - James R Tribble
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Marcela Votruba
- School of Optometry and Vision Sciences, Cardiff University, Maindy Road, Cardiff, Wales, UK
- University Hospital of Wales, Heath Park, Cardiff, Wales, UK
| | - Pete A Williams
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden.
| | - James E Morgan
- School of Optometry and Vision Sciences, Cardiff University, Maindy Road, Cardiff, Wales, UK.
- University Hospital of Wales, Heath Park, Cardiff, Wales, UK.
| |
Collapse
|
11
|
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
|
12
|
Geisler S. Augustus Waller's foresight realized: SARM1 in peripheral neuropathies. Curr Opin Neurobiol 2024; 87:102884. [PMID: 38852438 PMCID: PMC11600426 DOI: 10.1016/j.conb.2024.102884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 06/11/2024]
Abstract
Peripheral neuropathy is a common neurodegenerative condition characterized by numbness, tingling, pain, and weakness that frequently starts in the distal limbs. Arising from multiple etiologies, many peripheral neuropathies exhibit a slowly progressive course due to axon degeneration for which no effective treatments exist. During the past decade, numerous crucial insights into mechanisms of axon degeneration in peripheral neuropathies emerged from experiments involving nerve-cutting procedures, revealing the central role of the SARM1 axon degeneration pathway in both. Here I review commonalities and differences in the role of SARM1 after nerve cut and in several acquired and inherited peripheral neuropathies. This new knowledge now paves the way for the development of therapeutics that directly address root causes of various kinds of neuropathies.
Collapse
Affiliation(s)
- Stefanie Geisler
- Department of Neurology, Washington University School of Medicine in St. Louis, 660S. Euclid Ave, Box 8111, St. Louis, MO 63110, USA.
| |
Collapse
|
13
|
Petriti B, Rabiolo A, Chau KY, Williams PA, Montesano G, Lascaratos G, Garway-Heath DF. Peripheral blood mononuclear cell respiratory function is associated with progressive glaucomatous vision loss. Nat Med 2024; 30:2362-2370. [PMID: 38886621 PMCID: PMC11333286 DOI: 10.1038/s41591-024-03068-6] [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: 09/13/2023] [Accepted: 05/14/2024] [Indexed: 06/20/2024]
Abstract
Intraocular pressure (IOP) is currently the only modifiable risk factor for glaucoma and all licensed treatments lower IOP. However, many patients continue to lose vision despite IOP-lowering treatment. Identifying biomarkers for progressive vision loss would have considerable clinical utility. We demonstrate that lower peripheral blood mononuclear cell (PBMC) oxygen consumption rate (OCR) is strongly associated with faster visual field (VF) progression in patients treated by lowering IOP (P < 0.001, 229 eyes of 139 participants), explaining 13% of variance in the rate of progression. In a separate reference cohort of untreated patients with glaucoma (213 eyes of 213 participants), IOP explained 16% of VF progression variance. OCR is lower in patients with glaucoma (n = 168) than in controls (n = 50; P < 0.001) and is lower in patients with low baseline IOP (n = 99) than those with high baseline IOP (n = 69; P < 0.01). PBMC nicotinamide adenine dinucleotide (NAD) levels are lower in patients with glaucoma (n = 29) compared to controls (n = 25; P < 0.001) and strongly associated with OCR (P < 0.001). Our results support PBMC OCR and NAD levels as new biomarkers for progressive glaucoma.
Collapse
Affiliation(s)
- Bledi Petriti
- NIHR Biomedical Research Centre, Moorfields Eye Hospital and UCL Institute of Ophthalmology, London, UK
- Department of Clinical and Movement Neurosciences, UCL Queens Square Institute of Neurology, London, UK
| | - Alessandro Rabiolo
- Department of Health Sciences, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy
| | - Kai-Yin Chau
- Department of Clinical and Movement Neurosciences, UCL Queens Square Institute of Neurology, London, UK
| | - Pete A Williams
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Giovanni Montesano
- NIHR Biomedical Research Centre, Moorfields Eye Hospital and UCL Institute of Ophthalmology, London, UK
| | | | - David F Garway-Heath
- NIHR Biomedical Research Centre, Moorfields Eye Hospital and UCL Institute of Ophthalmology, London, UK.
| |
Collapse
|
14
|
Bazarek SF, Krenn MJ, Shah SB, Mandeville RM, Brown JM. Novel Technologies to Address the Lower Motor Neuron Injury and Augment Reconstruction in Spinal Cord Injury. Cells 2024; 13:1231. [PMID: 39056812 PMCID: PMC11274462 DOI: 10.3390/cells13141231] [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: 06/19/2024] [Revised: 07/11/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Lower motor neuron (LMN) damage results in denervation of the associated muscle targets and is a significant yet under-appreciated component of spinal cord injury (SCI). Denervated muscle undergoes a progressive degeneration and fibro-fatty infiltration that eventually renders the muscle non-viable unless reinnervated within a limited time window. The distal nerve deprived of axons also undergoes degeneration and fibrosis making it less receptive to axons. In this review, we describe the LMN injury associated with SCI and its clinical consequences. The process of degeneration of the muscle and nerve is broken down into the primary components of the neuromuscular circuit and reviewed, including the nerve and Schwann cells, the neuromuscular junction, and the muscle. Finally, we discuss three promising strategies to reverse denervation atrophy. These include providing surrogate axons from local sources; introducing stem cell-derived spinal motor neurons into the nerve to provide the missing axons; and finally, instituting a training program of high-energy electrical stimulation to directly rehabilitate these muscles. Successful interventions for denervation atrophy would significantly expand reconstructive options for cervical SCI and could be transformative for the predominantly LMN injuries of the conus medullaris and cauda equina.
Collapse
Affiliation(s)
- Stanley F. Bazarek
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (S.F.B.); (M.J.K.); (R.M.M.)
- Department of Neurological Surgery, University Hospitals-Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Matthias J. Krenn
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (S.F.B.); (M.J.K.); (R.M.M.)
- Department of Neurosurgery, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Center for Neuroscience and Neurological Recovery, Methodist Rehabilitation Center, Jackson, MS 39216, USA
- Spinal Cord Injury Medicine and Research Services, VA Medical Center, Jackson, MS 39216, USA
| | - Sameer B. Shah
- Departments of Orthopedic Surgery and Bioengineering, University of California-San Diego, La Jolla, CA 92093, USA;
- Research Division, VA San Diego Medical Center, San Diego, CA 92161, USA
| | - Ross M. Mandeville
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (S.F.B.); (M.J.K.); (R.M.M.)
| | - Justin M. Brown
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (S.F.B.); (M.J.K.); (R.M.M.)
| |
Collapse
|
15
|
Taub DG, Woolf CJ. Age-dependent small fiber neuropathy: Mechanistic insights from animal models. Exp Neurol 2024; 377:114811. [PMID: 38723859 PMCID: PMC11131160 DOI: 10.1016/j.expneurol.2024.114811] [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: 01/26/2024] [Revised: 04/07/2024] [Accepted: 05/05/2024] [Indexed: 05/28/2024]
Abstract
Small fiber neuropathy (SFN) is a common and debilitating disease in which the terminals of small diameter sensory axons degenerate, producing sensory loss, and in many patients neuropathic pain. While a substantial number of cases are attributable to diabetes, almost 50% are idiopathic. An underappreciated aspect of the disease is its late onset in most patients. Animal models of human genetic mutations that produce SFN also display age-dependent phenotypes suggesting that aging is an important contributor to the risk of development of the disease. In this review we define how particular sensory neurons are affected in SFN and discuss how aging may drive the disease. We also evaluate how animal models of SFN can define disease mechanisms that will provide insight into early risk detection and suggest novel therapeutic interventions.
Collapse
Affiliation(s)
- Daniel G Taub
- F. M. Kirby Neurobiology Center and Department of Neurology, Boston Children's Hospital, Boston, MA, USA; Department of Neurobiology, Harvard Medical School, Boston, MA, USA.
| | - Clifford J Woolf
- F. M. Kirby Neurobiology Center and Department of Neurology, Boston Children's Hospital, Boston, MA, USA; Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
16
|
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
|
17
|
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
|
18
|
Stassart RM, Gomez-Sanchez JA, Lloyd AC. Schwann Cells as Orchestrators of Nerve Repair: Implications for Tissue Regeneration and Pathologies. Cold Spring Harb Perspect Biol 2024; 16:a041363. [PMID: 38199866 PMCID: PMC11146315 DOI: 10.1101/cshperspect.a041363] [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] [Indexed: 01/12/2024]
Abstract
Peripheral nerves exist in a stable state in adulthood providing a rapid bidirectional signaling system to control tissue structure and function. However, following injury, peripheral nerves can regenerate much more effectively than those of the central nervous system (CNS). This multicellular process is coordinated by peripheral glia, in particular Schwann cells, which have multiple roles in stimulating and nurturing the regrowth of damaged axons back to their targets. Aside from the repair of damaged nerves themselves, nerve regenerative processes have been linked to the repair of other tissues and de novo innervation appears important in establishing an environment conducive for the development and spread of tumors. In contrast, defects in these processes are linked to neuropathies, aging, and pain. In this review, we focus on the role of peripheral glia, especially Schwann cells, in multiple aspects of nerve regeneration and discuss how these findings may be relevant for pathologies associated with these processes.
Collapse
Affiliation(s)
- Ruth M Stassart
- Paul-Flechsig-Institute of Neuropathology, University Clinic Leipzig, Leipzig 04103, Germany
| | - Jose A Gomez-Sanchez
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante 03010, Spain
- Instituto de Neurociencias CSIC-UMH, Sant Joan de Alicante 03550, Spain
| | - Alison C Lloyd
- UCL Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, United Kingdom
| |
Collapse
|
19
|
Bou Ghanem GO, Wareham LK, Calkins DJ. Addressing neurodegeneration in glaucoma: Mechanisms, challenges, and treatments. Prog Retin Eye Res 2024; 100:101261. [PMID: 38527623 DOI: 10.1016/j.preteyeres.2024.101261] [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: 12/28/2023] [Revised: 03/15/2024] [Accepted: 03/19/2024] [Indexed: 03/27/2024]
Abstract
Glaucoma is the leading cause of irreversible blindness globally. The disease causes vision loss due to neurodegeneration of the retinal ganglion cell (RGC) projection to the brain through the optic nerve. Glaucoma is associated with sensitivity to intraocular pressure (IOP). Thus, mainstay treatments seek to manage IOP, though many patients continue to lose vision. To address neurodegeneration directly, numerous preclinical studies seek to develop protective or reparative therapies that act independently of IOP. These include growth factors, compounds targeting metabolism, anti-inflammatory and antioxidant agents, and neuromodulators. Despite success in experimental models, many of these approaches fail to translate into clinical benefits. Several factors contribute to this challenge. Firstly, the anatomic structure of the optic nerve head differs between rodents, nonhuman primates, and humans. Additionally, animal models do not replicate the complex glaucoma pathophysiology in humans. Therefore, to enhance the success of translating these findings, we propose two approaches. First, thorough evaluation of experimental targets in multiple animal models, including nonhuman primates, should precede clinical trials. Second, we advocate for combination therapy, which involves using multiple agents simultaneously, especially in the early and potentially reversible stages of the disease. These strategies aim to increase the chances of successful neuroprotective treatment for glaucoma.
Collapse
Affiliation(s)
- Ghazi O Bou Ghanem
- Vanderbilt Eye Institute, Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Lauren K Wareham
- Vanderbilt Eye Institute, Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - David J Calkins
- Vanderbilt Eye Institute, Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
20
|
Zhao R, Deng X, Dong J, Liang C, Yang X, Tang Y, Du J, Ge Z, Wang D, Shen Y, Jiang L, Lin W, Zhu T, Wang G. Highly Bioadaptable Hybrid Conduits with Spatially Bidirectional Structure for Precision Nerve Fiber Regeneration via Gene Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309306. [PMID: 38483934 PMCID: PMC11109652 DOI: 10.1002/advs.202309306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/20/2024] [Indexed: 05/23/2024]
Abstract
Peripheral nerve deficits give rise to motor and sensory impairments within the limb. The clinical restoration of extensive segmental nerve defects through autologous nerve transplantation often encounters challenges such as axonal mismatch and suboptimal functional recovery. These issues may stem from the limited regenerative capacity of proximal axons and the subsequent Wallerian degeneration of distal axons. To achieve the integration of sensory and motor functions, a spatially differential plasmid DNA (pDNA) dual-delivery nanohydrogel conduit scaffold is devised. This innovative scaffold facilitates the localized administration of the transforming growth factor β (TGF-β) gene in the proximal region to accelerate nerve regeneration, while simultaneously delivering nicotinamide mononucleotide adenylyltransferase 2 (NMNAT2) to the distal region to mitigate Wallerian degeneration. By promoting autonomous and selective alignment of nerve fiber gap sutures via structure design, the approach aims to achieve a harmonious unification of nerve regeneration, neuromotor function, and sensory recovery. It is anticipated that this groundbreaking technology will establish a robust platform for gene delivery in tissue engineering.
Collapse
Affiliation(s)
- Renliang Zhao
- Orthopedics Research InstituteDepartment of OrthopedicsWest China HospitalSichuan UniversityChengdu610041P. R. China
- Trauma Medical CenterDepartment of Orthopedics SurgeryWest China HospitalSichuan UniversityChengdu610041China
| | - Xiangtian Deng
- Orthopedics Research InstituteDepartment of OrthopedicsWest China HospitalSichuan UniversityChengdu610041P. R. China
- Trauma Medical CenterDepartment of Orthopedics SurgeryWest China HospitalSichuan UniversityChengdu610041China
| | - Jizhao Dong
- Multidisciplinary Centre for Advanced MaterialsInstitute for Frontier Medical TechnologySchool of Chemistry and Chemical EngineeringShanghai University of Engineering Science333 Longteng Rd.Shanghai201620P. R. China
| | - Chen Liang
- Multidisciplinary Centre for Advanced MaterialsInstitute for Frontier Medical TechnologySchool of Chemistry and Chemical EngineeringShanghai University of Engineering Science333 Longteng Rd.Shanghai201620P. R. China
| | - Xiaozhong Yang
- Orthopedics Research InstituteDepartment of OrthopedicsWest China HospitalSichuan UniversityChengdu610041P. R. China
- Trauma Medical CenterDepartment of Orthopedics SurgeryWest China HospitalSichuan UniversityChengdu610041China
| | - Yunfeng Tang
- Head & Neck Oncology WardCancer CenterWest China HospitalCancer CenterSichuan UniversityChengdu610041P. R. China
| | - Juan Du
- Multidisciplinary Centre for Advanced MaterialsInstitute for Frontier Medical TechnologySchool of Chemistry and Chemical EngineeringShanghai University of Engineering Science333 Longteng Rd.Shanghai201620P. R. China
| | - Zilu Ge
- Orthopedics Research InstituteDepartment of OrthopedicsWest China HospitalSichuan UniversityChengdu610041P. R. China
- Trauma Medical CenterDepartment of Orthopedics SurgeryWest China HospitalSichuan UniversityChengdu610041China
| | - Dong Wang
- Orthopedics Research InstituteDepartment of OrthopedicsWest China HospitalSichuan UniversityChengdu610041P. R. China
- Trauma Medical CenterDepartment of Orthopedics SurgeryWest China HospitalSichuan UniversityChengdu610041China
| | - Yifan Shen
- Spine LabDepartment of Orthopedic SurgeryThe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003China
| | - Lianghua Jiang
- Department of Orthopedic TraumaThe First People's Hospital of Kunshan affiliated with Jiangsu UniversitySuzhouJiangsu215300P. R. China
| | - Wei Lin
- Department of GynecologyWest China Second HospitalSichuan UniversityChengdu610041P. R. China
| | - Tonghe Zhu
- Multidisciplinary Centre for Advanced MaterialsInstitute for Frontier Medical TechnologySchool of Chemistry and Chemical EngineeringShanghai University of Engineering Science333 Longteng Rd.Shanghai201620P. R. China
| | - Guanglin Wang
- Orthopedics Research InstituteDepartment of OrthopedicsWest China HospitalSichuan UniversityChengdu610041P. R. China
- Trauma Medical CenterDepartment of Orthopedics SurgeryWest China HospitalSichuan UniversityChengdu610041China
| |
Collapse
|
21
|
Garb J, Amitai G, Lu A, Ofir G, Brandis A, Mehlman T, Kranzusch PJ, Sorek R. The SARM1 TIR domain produces glycocyclic ADPR molecules as minor products. PLoS One 2024; 19:e0302251. [PMID: 38635746 PMCID: PMC11025887 DOI: 10.1371/journal.pone.0302251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 03/31/2024] [Indexed: 04/20/2024] Open
Abstract
Sterile alpha and TIR motif-containing 1 (SARM1) is a protein involved in programmed death of injured axons. Following axon injury or a drug-induced insult, the TIR domain of SARM1 degrades the essential molecule nicotinamide adenine dinucleotide (NAD+), leading to a form of axonal death called Wallerian degeneration. Degradation of NAD+ by SARM1 is essential for the Wallerian degeneration process, but accumulating evidence suggest that other activities of SARM1, beyond the mere degradation of NAD+, may be necessary for programmed axonal death. In this study we show that the TIR domains of both human and fruit fly SARM1 produce 1''-2' and 1''-3' glycocyclic ADP-ribose (gcADPR) molecules as minor products. As previously reported, we observed that SARM1 TIR domains mostly convert NAD+ to ADPR (for human SARM1) or cADPR (in the case of SARM1 from Drosophila melanogaster). However, we now show that human and Drosophila SARM1 additionally convert ~0.1-0.5% of NAD+ into gcADPR molecules. We find that SARM1 TIR domains produce gcADPR molecules both when purified in vitro and when expressed in bacterial cells. Given that gcADPR is a second messenger involved in programmed cell death in bacteria and likely in plants, we propose that gcADPR may play a role in SARM1-induced programmed axonal death in animals.
Collapse
Affiliation(s)
- Jeremy Garb
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Gil Amitai
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Allen Lu
- Department of Microbiology, Harvard Medical School, Boston, MA, United States of America
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, United States of America
| | - Gal Ofir
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Alexander Brandis
- Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Tevie Mehlman
- Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Philip J Kranzusch
- Department of Microbiology, Harvard Medical School, Boston, MA, United States of America
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, United States of America
- Parker Institute for Cancer Immunotherapy at Dana-Farber Cancer Institute, Boston, MA, United States of America
| | - Rotem Sorek
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
22
|
Zhang M, Liu T, Yang J. Skin neuropathy and immunomodulation in diseases. FUNDAMENTAL RESEARCH 2024; 4:218-225. [PMID: 38933512 PMCID: PMC11197692 DOI: 10.1016/j.fmre.2022.08.016] [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: 07/31/2022] [Revised: 08/14/2022] [Accepted: 08/30/2022] [Indexed: 12/01/2022] Open
Abstract
Skin is a vital barrier tissue of the body. Immune responses in the skin must be precisely controlled, which would otherwise cause severe disease conditions such as psoriasis, atopic dermatitis, or pathogenic infection. Research evidence has increasingly demonstrated the essential roles of neural innervations, i.e., sensory and sympathetic signals, in modulating skin immunity. Notably, neuropathic changes of such neural structures have been observed in skin disease conditions, implicating their direct involvement in various pathological processes. An in-depth understanding of the mechanism underlying skin neuropathy and its immunomodulatory effects could help reveal novel entry points for therapeutic interventions. Here, we summarize the neuroimmune interactions between neuropathic events and skin immunity, highlighting the current knowledge and future perspectives of this emerging research frontier.
Collapse
Affiliation(s)
- Manze Zhang
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Tingting Liu
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065, USA
| | - Jing Yang
- IDG/McGovern Institute for Brain Research, Center for Life Sciences, School of Life Sciences, Peking University, Beijing 100871, China
- Peking University Third Hospital Cancer Center, Beijing 100191, China
| |
Collapse
|
23
|
Hwang CD, Hoftiezer YAJ, Raasveld FV, Gomez-Eslava B, van der Heijden EPA, Jayakar S, Black BJ, Johnston BR, Wainger BJ, Renthal W, Woolf CJ, Eberlin KR. Biology and pathophysiology of symptomatic neuromas. Pain 2024; 165:550-564. [PMID: 37851396 DOI: 10.1097/j.pain.0000000000003055] [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: 11/28/2022] [Accepted: 06/07/2023] [Indexed: 10/19/2023]
Abstract
ABSTRACT Neuromas are a substantial cause of morbidity and reduction in quality of life. This is not only caused by a disruption in motor and sensory function from the underlying nerve injury but also by the debilitating effects of neuropathic pain resulting from symptomatic neuromas. A wide range of surgical and therapeutic modalities have been introduced to mitigate this pain. Nevertheless, no single treatment option has been successful in completely resolving the associated constellation of symptoms. While certain novel surgical techniques have shown promising results in reducing neuroma-derived and phantom limb pain, their effectiveness and the exact mechanism behind their pain-relieving capacities have not yet been defined. Furthermore, surgery has inherent risks, may not be suitable for many patients, and may yet still fail to relieve pain. Therefore, there remains a great clinical need for additional therapeutic modalities to further improve treatment for patients with devastating injuries that lead to symptomatic neuromas. However, the molecular mechanisms and genetic contributions behind the regulatory programs that drive neuroma formation-as well as the resulting neuropathic pain-remain incompletely understood. Here, we review the histopathological features of symptomatic neuromas, our current understanding of the mechanisms that favor neuroma formation, and the putative contributory signals and regulatory programs that facilitate somatic pain, including neurotrophic factors, neuroinflammatory peptides, cytokines, along with transient receptor potential, and ionotropic channels that suggest possible approaches and innovations to identify novel clinical therapeutics.
Collapse
Affiliation(s)
- Charles D Hwang
- Division of Plastic and Reconstructive Surgery, Department of General Surgery, Massachusetts General Hospital, Harvard University, Boston, MA, United States
| | - Yannick Albert J Hoftiezer
- Hand and Arm Center, Department of Orthopaedic Surgery, Massachusetts General Hospital, Boston, MA, United States
- Department of Plastic, Reconstructive and Hand Surgery, Radboudumc, Nijmegen, the Netherlands
| | - Floris V Raasveld
- Hand and Arm Center, Department of Orthopaedic Surgery, Massachusetts General Hospital, Boston, MA, United States
- Department of Plastic, Reconstructive and Hand Surgery, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Barbara Gomez-Eslava
- Hand and Arm Center, Department of Orthopaedic Surgery, Massachusetts General Hospital, Boston, MA, United States
- F.M. Kirby Neurobiology Center, Boston Children's Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA, United States
| | - E P A van der Heijden
- Department of Plastic, Reconstructive and Hand Surgery, Radboudumc, Nijmegen, the Netherlands
- Department of Plastic, Reconstructive and Hand Surgery, Jeroen Bosch Ziekenhuis, Den Bosch, the Netherlands
| | - Selwyn Jayakar
- F.M. Kirby Neurobiology Center, Boston Children's Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA, United States
| | - Bryan James Black
- Department of Biomedical Engineering, UMass Lowell, Lowell, MA, United States
| | - Benjamin R Johnston
- Department of Neurosurgery, Brigham and Women's Hospital, Boston, MA, United States
| | - Brian J Wainger
- Departments of Anesthesia, Critical Care & Pain Medicine and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | | | - Clifford J Woolf
- F.M. Kirby Neurobiology Center, Boston Children's Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA, United States
| | - Kyle R Eberlin
- Division of Plastic and Reconstructive Surgery, Department of General Surgery, Massachusetts General Hospital, Harvard University, Boston, MA, United States
| |
Collapse
|
24
|
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
|
25
|
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
|
26
|
Zhou K, Tan Y, Zhang G, Li J, Xing S, Chen X, Wen J, Li G, Fan Y, Zeng J, Zhang J. Loss of SARM1 ameliorates secondary thalamic neurodegeneration after cerebral infarction. J Cereb Blood Flow Metab 2024; 44:224-238. [PMID: 37898107 PMCID: PMC10993876 DOI: 10.1177/0271678x231210694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 08/07/2023] [Accepted: 10/07/2023] [Indexed: 10/30/2023]
Abstract
Ischemic stroke causes secondary neurodegeneration in the thalamus ipsilateral to the infarction site and impedes neurological recovery. Axonal degeneration of thalamocortical fibers and autophagy overactivation are involved in thalamic neurodegeneration after ischemic stroke. However, the molecular mechanisms underlying thalamic neurodegeneration remain unclear. Sterile /Armadillo/Toll-Interleukin receptor homology domain protein (SARM1) can induce Wallerian degeneration. Herein, we aimed to investigate the role of SARM1 in thalamic neurodegeneration and autophagy activation after photothrombotic infarction. Neurological deficits measured using modified neurological severity scores and adhesive-removal test were ameliorated in Sarm1-/- mice after photothrombotic infarction. Compared with wild-type mice, Sarm1-/- mice exhibited unaltered infarct volume; however, there were markedly reduced neuronal death and gliosis in the ipsilateral thalamus. In parallel, autophagy activation was attenuated in the thalamus of Sarm1-/- mice after cerebral infarction. Thalamic Sarm1 re-expression in Sarm1-/- mice increased thalamic neurodegeneration and promoted autophagy activation. Auotophagic inhibitor 3-methyladenine partially alleviated thalamic damage induced by SARM1. Moreover, autophagic initiation through rapamycin treatment aggravated post-stroke neuronal death and gliosis in Sarm1-/- mice. Taken together, SARM1 contributes to secondary thalamic neurodegeneration after cerebral infarction, at least partly through autophagy inhibition. SARM1 deficiency is a potential therapeutic strategy for secondary thalamic neurodegeneration and functional deficits after stroke.
Collapse
Affiliation(s)
- Kun Zhou
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases; National Key Clinical Department and Key Discipline of Neurology; Guangzhou, China
| | - Yan Tan
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases; National Key Clinical Department and Key Discipline of Neurology; Guangzhou, China
| | - Guofen Zhang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases; National Key Clinical Department and Key Discipline of Neurology; Guangzhou, China
| | - Jingjing Li
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases; National Key Clinical Department and Key Discipline of Neurology; Guangzhou, China
| | - Shihui Xing
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases; National Key Clinical Department and Key Discipline of Neurology; Guangzhou, China
| | - Xinran Chen
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases; National Key Clinical Department and Key Discipline of Neurology; Guangzhou, China
| | - Jiali Wen
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases; National Key Clinical Department and Key Discipline of Neurology; Guangzhou, China
| | - Ge Li
- Guangdong Provincial Key Laboratory of Laboratory Animals, Guangdong Laboratory Animals Monitoring Institute, Guangzhou, China
| | - Yuhua Fan
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases; National Key Clinical Department and Key Discipline of Neurology; Guangzhou, China
| | - Jinsheng Zeng
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases; National Key Clinical Department and Key Discipline of Neurology; Guangzhou, China
| | - Jian Zhang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases; National Key Clinical Department and Key Discipline of Neurology; Guangzhou, China
| |
Collapse
|
27
|
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
|
28
|
Brüll M, Geese N, Celardo I, Laumann M, Leist M. Preparation of Viable Human Neurites for Neurobiological and Neurodegeneration Studies. Cells 2024; 13:242. [PMID: 38334634 PMCID: PMC10854604 DOI: 10.3390/cells13030242] [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: 12/21/2023] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 02/10/2024] Open
Abstract
Few models allow the study of neurite damage in the human central nervous system. We used here dopaminergic LUHMES neurons to establish a culture system that allows for (i) the observation of highly enriched neurites, (ii) the preparation of the neurite fraction for biochemical studies, and (iii) the measurement of neurite markers and metabolites after axotomy. LUHMES-based spheroids, plated in culture dishes, extended neurites of several thousand µm length, while all somata remained aggregated. These cultures allowed an easy microscopic observation of live or fixed neurites. Neurite-only cultures (NOC) were produced by cutting out the still-aggregated somata. The potential application of such cultures was exemplified by determinations of their protein and RNA contents. For instance, the mitochondrial TOM20 protein was highly abundant, while nuclear histone H3 was absent. Similarly, mitochondrial-encoded RNAs were found at relatively high levels, while the mRNA for a histone or the neuronal nuclear marker NeuN (RBFOX3) were relatively depleted in NOC. Another potential use of NOC is the study of neurite degeneration. For this purpose, an algorithm to quantify neurite integrity was developed. Using this tool, we found that the addition of nicotinamide drastically reduced neurite degeneration. Also, the chelation of Ca2+ in NOC delayed the degeneration, while inhibitors of calpains had no effect. Thus, NOC proved to be suitable for biochemical analysis and for studying degeneration processes after a defined cut injury.
Collapse
Affiliation(s)
- Markus Brüll
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78457 Konstanz, Germany; (M.B.); (N.G.); (I.C.)
| | - Nils Geese
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78457 Konstanz, Germany; (M.B.); (N.G.); (I.C.)
| | - Ivana Celardo
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78457 Konstanz, Germany; (M.B.); (N.G.); (I.C.)
| | - Michael Laumann
- Electron Microscopy Centre, University of Konstanz, 78457 Konstanz, Germany;
| | - Marcel Leist
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78457 Konstanz, Germany; (M.B.); (N.G.); (I.C.)
- Center for Alternatives to Animal Testing in Europe (CAAT-Europe), University of Konstanz, 78457 Konstanz, Germany
| |
Collapse
|
29
|
Tarasiuk O, Molteni L, Malacrida A, Nicolini G. The Role of NMNAT2/SARM1 in Neuropathy Development. BIOLOGY 2024; 13:61. [PMID: 38275737 PMCID: PMC10813049 DOI: 10.3390/biology13010061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/15/2024] [Accepted: 01/19/2024] [Indexed: 01/27/2024]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) commonly arises as a side effect of diverse cancer chemotherapy treatments. This condition presents symptoms such as numbness, tingling, and altered sensation in patients, often accompanied by neuropathic pain. Pathologically, CIPN is characterized by an intensive "dying-back" axonopathy, starting at the intra-epidermal sensory innervations and advancing retrogradely. The lack of comprehensive understanding regarding its underlying mechanisms explains the absence of effective treatments for CIPN. Recent investigations into axon degeneration mechanisms have pinpointed nicotinamide mononucleotide adenylyltransferase 2 (NMNAT2) and sterile alpha and TIR motif-containing 1 protein (SARM1) as pivotal mediators of injury-induced axonal degeneration. In this review, we aim to explore various studies shedding light on the interplay between NMNAT2 and SARM1 proteins and their roles in the progression of CIPN.
Collapse
Affiliation(s)
- Olga Tarasiuk
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (L.M.); (A.M.); (G.N.)
| | | | | | | |
Collapse
|
30
|
Chandrasekaran K, Najimi N, Sagi AR, Yarlagadda S, Salimian M, Arvas MI, Hedayat AF, Kevas Y, Kadakia A, Kristian T, Russell JW. NAD + Precursors Reverse Experimental Diabetic Neuropathy in Mice. Int J Mol Sci 2024; 25:1102. [PMID: 38256175 PMCID: PMC10816262 DOI: 10.3390/ijms25021102] [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: 11/16/2023] [Revised: 01/05/2024] [Accepted: 01/13/2024] [Indexed: 01/24/2024] Open
Abstract
Abnormal NAD+ signaling has been implicated in axonal degeneration in diabetic peripheral neuropathy (DPN). We hypothesized that supplementing NAD+ precursors could alleviate DPN symptoms through increasing the NAD+ levels and activating the sirtuin-1 (SIRT1) protein. To test this, we exposed cultured Dorsal Root Ganglion neurons (DRGs) to Nicotinamide Riboside (NR) or Nicotinamide Mononucleotide (NMN), which increased the levels of NAD+, the SIRT1 protein, and the deacetylation activity that is associated with increased neurite growth. A SIRT1 inhibitor blocked the neurite growth induced via NR or NMN. We then induced neuropathy in C57BL6 mice with streptozotocin (STZ) or a high fat diet (HFD) and administered NR or NMN for two months. Both the STZ and HFD mice developed neuropathy, which was reversed through the NR or NMN administration: sensory function improved, nerve conduction velocities normalized, and intraepidermal nerve fibers were restored. The NAD+ levels and SIRT1 activity were reduced in the DRGs from diabetic mice but were preserved with the NR or NMN treatment. We also tested the effect of NR or NMN administration in mice that overexpress the SIRT1 protein in neurons (nSIRT1 OE) and found no additional benefit from the addition of the drug. These findings suggest that supplementing with NAD+ precursors or activating SIRT1 may be a promising treatment for DPN.
Collapse
Affiliation(s)
- Krish Chandrasekaran
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (S.Y.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
| | - Neda Najimi
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (S.Y.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
| | - Avinash R. Sagi
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (S.Y.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
| | - Sushuma Yarlagadda
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (S.Y.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
| | - Mohammad Salimian
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (S.Y.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
| | - Muhammed Ikbal Arvas
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (S.Y.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
| | - Ahmad F. Hedayat
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (S.Y.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
| | - Yanni Kevas
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (S.Y.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
| | - Anand Kadakia
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (S.Y.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
| | - Tibor Kristian
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
- Veterans Affairs Medical Center, Baltimore, MD 21201, USA
| | - James W. Russell
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (S.Y.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
- Veterans Affairs Medical Center, Baltimore, MD 21201, USA
- CAMC Institute for Academic Medicine, 415 Morris Street Suite 300, Charleston, WV 25301, USA
| |
Collapse
|
31
|
Zhu L, Deng F, Bai D, Hou J, Jia Q, Zhang C, Ou K, Li S, Li XJ, Yin P. Loss of TDP-43 mediates severe neurotoxicity by suppressing PJA1 gene transcription in the monkey brain. Cell Mol Life Sci 2024; 81:16. [PMID: 38194085 PMCID: PMC11072099 DOI: 10.1007/s00018-023-05066-2] [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: 09/12/2023] [Revised: 11/19/2023] [Accepted: 11/27/2023] [Indexed: 01/10/2024]
Abstract
The nuclear loss and cytoplasmic accumulation of TDP-43 (TAR DNA/RNA binding protein 43) are pathological hallmarks of amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). Previously, we reported that the primate-specific cleavage of TDP-43 accounts for its cytoplasmic mislocalization in patients' brains. This prompted us to investigate further whether and how the loss of nuclear TDP-43 mediates neuropathology in primate brain. In this study, we report that TDP-43 knockdown at the similar effectiveness, induces more damage to neuronal cells in the monkey brain than rodent mouse. Importantly, the loss of TDP-43 suppresses the E3 ubiquitin ligase PJA1 expression in the monkey brain at transcriptional level, but yields an opposite upregulation of PJA1 in the mouse brain. This distinct effect is due to the species-dependent binding of nuclear TDP-43 to the unique promoter sequences of the PJA1 genes. Further analyses reveal that the reduction of PJA1 accelerates neurotoxicity, whereas overexpressing PJA1 diminishes neuronal cell death by the TDP-43 knockdown in vivo. Our findings not only uncover a novel primate-specific neurotoxic contribution to the loss of function theory of TDP-43 proteinopathy, but also underscore a potential therapeutic approach of PJA1 to the loss of nuclear TDP-43.
Collapse
Affiliation(s)
- Longhong Zhu
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Fuyu Deng
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Dazhang Bai
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, North Sichuan Medical College, Nanchong, 637000, China
- Institute of Neurological Diseases, North Sichuan Medical College, Nanchong, 637000, China
| | - Junqi Hou
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Qingqing Jia
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Chen Zhang
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Kaili Ou
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Shihua Li
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
- Ministry of Education Key Laboratory of CNS Regeneration, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Xiao-Jiang Li
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China.
- Ministry of Education Key Laboratory of CNS Regeneration, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China.
| | - Peng Yin
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China.
- Ministry of Education Key Laboratory of CNS Regeneration, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
32
|
López-Erauskin J, Bravo-Hernandez M, Presa M, Baughn MW, Melamed Z, Beccari MS, Agra de Almeida Quadros AR, Arnold-Garcia O, Zuberi A, Ling K, Platoshyn O, Niño-Jara E, Ndayambaje IS, McAlonis-Downes M, Cabrera L, Artates JW, Ryan J, Hermann A, Ravits J, Bennett CF, Jafar-Nejad P, Rigo F, Marsala M, Lutz CM, Cleveland DW, Lagier-Tourenne C. Stathmin-2 loss leads to neurofilament-dependent axonal collapse driving motor and sensory denervation. Nat Neurosci 2024; 27:34-47. [PMID: 37996528 PMCID: PMC10842032 DOI: 10.1038/s41593-023-01496-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 10/16/2023] [Indexed: 11/25/2023]
Abstract
The mRNA transcript of the human STMN2 gene, encoding for stathmin-2 protein (also called SCG10), is profoundly impacted by TAR DNA-binding protein 43 (TDP-43) loss of function. The latter is a hallmark of several neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). Using a combination of approaches, including transient antisense oligonucleotide-mediated suppression, sustained shRNA-induced depletion in aging mice, and germline deletion, we show that stathmin-2 has an important role in the establishment and maintenance of neurofilament-dependent axoplasmic organization that is critical for preserving the caliber and conduction velocity of myelinated large-diameter axons. Persistent stathmin-2 loss in adult mice results in pathologies found in ALS, including reduced interneurofilament spacing, axonal caliber collapse that drives tearing within outer myelin layers, diminished conduction velocity, progressive motor and sensory deficits, and muscle denervation. These findings reinforce restoration of stathmin-2 as an attractive therapeutic approach for ALS and other TDP-43-dependent neurodegenerative diseases.
Collapse
Affiliation(s)
- Jone López-Erauskin
- Ludwig Institute and Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Mariana Bravo-Hernandez
- Department of Anesthesiology and Stem Cell Program and Institute for Genomic Medicine, University of California at San Diego, La Jolla, CA, USA
- Ionis Pharmaceuticals Inc., Carlsbad, CA, USA
| | | | - Michael W Baughn
- Ludwig Institute and Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Ze'ev Melamed
- Ludwig Institute and Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
- Department of Medical Neurobiology, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Melinda S Beccari
- Ludwig Institute and Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Ana Rita Agra de Almeida Quadros
- Department of Neurology, The Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Olatz Arnold-Garcia
- Ludwig Institute and Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
- Department of Neurosciences, Biodonostia Health Research Institute, San Sebastián, Spain
- CIBERNED, ISCIII (CIBER, Carlos III Institute, Spanish Ministry of Sciences and Innovation), Madrid, Spain
| | | | - Karen Ling
- Ionis Pharmaceuticals Inc., Carlsbad, CA, USA
| | - Oleksandr Platoshyn
- Department of Anesthesiology and Stem Cell Program and Institute for Genomic Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Elkin Niño-Jara
- Department of Anesthesiology and Stem Cell Program and Institute for Genomic Medicine, University of California at San Diego, La Jolla, CA, USA
| | - I Sandra Ndayambaje
- Department of Neurology, The Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Melissa McAlonis-Downes
- Ludwig Institute and Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Larissa Cabrera
- Ludwig Institute and Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Jonathan W Artates
- Ludwig Institute and Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
| | | | - Anita Hermann
- Department of Neurosciences, School of Medicine, University of California at San Diego, La Jolla, CA, USA
| | - John Ravits
- Department of Neurosciences, School of Medicine, University of California at San Diego, La Jolla, CA, USA
| | | | | | - Frank Rigo
- Ionis Pharmaceuticals Inc., Carlsbad, CA, USA
| | - Martin Marsala
- Department of Anesthesiology and Stem Cell Program and Institute for Genomic Medicine, University of California at San Diego, La Jolla, CA, USA
| | | | - Don W Cleveland
- Ludwig Institute and Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA.
| | - Clotilde Lagier-Tourenne
- Department of Neurology, The Sean M. Healey and AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
33
|
Ma Y, Jia T, Qin F, He Y, Han F, Zhang C. Abnormal Brain Protein Abundance and Cross-tissue mRNA Expression in Amyotrophic Lateral Sclerosis. Mol Neurobiol 2024; 61:510-518. [PMID: 37639066 PMCID: PMC10791788 DOI: 10.1007/s12035-023-03587-2] [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: 02/20/2023] [Accepted: 08/13/2023] [Indexed: 08/29/2023]
Abstract
Due to the limitations of the present risk genes in understanding the etiology of amyotrophic lateral sclerosis (ALS), it is necessary to find additional causative genes utilizing novel approaches. In this study, we conducted a two-stage proteome-wide association study (PWAS) using ALS genome-wide association study (GWAS) data (N = 152,268) and two distinct human brain protein quantitative trait loci (pQTL) datasets (ROSMAP N = 376 and Banner N = 152) to identify ALS risk genes and prioritized candidate genes with Mendelian randomization (MR) and Bayesian colocalization analysis. Next, we verified the aberrant expression of risk genes in multiple tissues, including lower motor neurons, skeletal muscle, and whole blood. Six ALS risk genes (SCFD1, SARM1, TMEM175, BCS1L, WIPI2, and DHRS11) were found during the PWAS discovery phase, and SARM1 and BCS1L were confirmed during the validation phase. The following MR (p = 2.10 × 10-7) and Bayesian colocalization analysis (ROSMAP PP4 = 0.999, Banner PP4 = 0.999) confirmed the causal association between SARM1 and ALS. Further differential expression analysis revealed that SARM1 was markedly downregulated in lower motor neurons (p = 7.64 × 10-3), skeletal muscle (p = 9.34 × 10-3), and whole blood (p = 1.94 × 10-3). Our findings identified some promising protein candidates for future investigation as therapeutic targets. The dysregulation of SARM1 in multiple tissues provides a new way to explain ALS pathology.
Collapse
Affiliation(s)
- Yanni Ma
- Mental Health Center and Psychiatric Laboratory, The State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan, China
| | - Tingting Jia
- Mental Health Center and Psychiatric Laboratory, The State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan, China
| | - Fengqin Qin
- Department of Neurology, The 3Rd Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Yongji He
- Clinical Trial Center, National Medical Products Administration Key Laboratory for Clinical Research and Evaluation of Innovative Drugs, West China Hospital Sichuan University, Chengdu, People's Republic of China
| | - Feng Han
- Department of Emergency Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Chengcheng Zhang
- Mental Health Center and Psychiatric Laboratory, The State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
34
|
Liang J, Huang F, Song Z, Tang R, Zhang P, Chen R. Impact of NAD+ metabolism on ovarian aging. Immun Ageing 2023; 20:70. [PMID: 38041117 PMCID: PMC10693113 DOI: 10.1186/s12979-023-00398-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 11/21/2023] [Indexed: 12/03/2023]
Abstract
Nicotinamide adenine dinucleotide (NAD+), a crucial coenzyme in cellular redox reactions, is closely associated with age-related functional degeneration and metabolic diseases. NAD exerts direct and indirect influences on many crucial cellular functions, including metabolic pathways, DNA repair, chromatin remodeling, cellular senescence, and immune cell functionality. These cellular processes and functions are essential for maintaining tissue and metabolic homeostasis, as well as healthy aging. Causality has been elucidated between a decline in NAD levels and multiple age-related diseases, which has been confirmed by various strategies aimed at increasing NAD levels in the preclinical setting. Ovarian aging is recognized as a natural process characterized by a decline in follicle number and function, resulting in decreased estrogen production and menopause. In this regard, it is necessary to address the many factors involved in this complicated procedure, which could improve fertility in women of advanced maternal age. Concerning the decrease in NAD+ levels as ovarian aging progresses, promising and exciting results are presented for strategies using NAD+ precursors to promote NAD+ biosynthesis, which could substantially improve oocyte quality and alleviate ovarian aging. Hence, to acquire further insights into NAD+ metabolism and biology, this review aims to probe the factors affecting ovarian aging, the characteristics of NAD+ precursors, and the current research status of NAD+ supplementation in ovarian aging. Specifically, by gaining a comprehensive understanding of these aspects, we are optimistic about the prominent progress that will be made in both research and therapy related to ovarian aging.
Collapse
Affiliation(s)
- Jinghui Liang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing, 100730, China
| | - Feiling Huang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing, 100730, China
| | - Zhaoqi Song
- School of Medical Technology and Engineering, Fujian Medical University, Fuzhou, Fujian, China
| | - Ruiyi Tang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing, 100730, China
| | - Peng Zhang
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, MOE Key Laboratory of Major Diseases in Children, Rare Disease Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China.
| | - Rong Chen
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing, 100730, China.
| |
Collapse
|
35
|
Chiarugi A. Glaucoma: neuroprotection with NAD-based therapeutic interventions. Trends Pharmacol Sci 2023; 44:869-879. [PMID: 37880000 DOI: 10.1016/j.tips.2023.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 09/22/2023] [Accepted: 09/26/2023] [Indexed: 10/27/2023]
Abstract
Clinical evidence shows that intraocular hypertension is not the primary pathogenetic event of glaucoma, whereas early neurodegeneration of retinal ganglion cells (RGCs) represents a key therapeutic target. Unfortunately, failure of clinical trials with neuroprotective agents, in particular those testing the anti-excitotoxic drug memantine, generated widespread skepticism regarding the possibility of counteracting neurodegeneration during glaucoma. New avenues for neuroprotective approaches to counteract glaucoma evolution have been opened by the identification of a programmed axonal degeneration (PAD) program triggered by increased nicotinamide mononucleotide (NMN)/NAD concentration ratio. Positive results of proof-of-concept clinical studies based on sustaining axonal NAD homeostasis facilitated the design of Phase 2/3 trials. Here, I share my opinion on how neurodegeneration in glaucoma should be put into context, together with an appraisal of the pharmacological rationale of NAD-supporting therapies for use during glaucoma progression.
Collapse
Affiliation(s)
- Alberto Chiarugi
- Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy; Headache Center and Clinical Pharmacology Unit, Careggi University Hospital, Florence, Italy.
| |
Collapse
|
36
|
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: 4.0] [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
|
37
|
Otsuki M, Terenzio M. Mechanisms of axonal degeneration and regeneration of the nervous system. Neurosci Res 2023; 197:1-2. [PMID: 37839523 DOI: 10.1016/j.neures.2023.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Affiliation(s)
- Miki Otsuki
- Molecular Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, Kunigami-gun, Okinawa 904-0412, Japan
| | - Marco Terenzio
- Molecular Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, Kunigami-gun, Okinawa 904-0412, Japan.
| |
Collapse
|
38
|
Estera LA, Walsh SP, Headen JA, Williamson RE, Kalinski AL. Neuroinflammation: Breaking barriers and bridging gaps. Neurosci Res 2023; 197:9-17. [PMID: 34748905 DOI: 10.1016/j.neures.2021.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 11/01/2021] [Indexed: 01/04/2023]
Abstract
Neurons are the cells of the nervous system and are responsible for every thought, movement and perception. Immune cells are the cells of the immune system, constantly protecting from foreign pathogens. Understanding the interaction between the two systems is especially important in disease states such as autoimmune or neurodegenerative disease. Unfortunately, this interaction is typically detrimental to the host. However, recent efforts have focused on how neurons and immune cells interact, either directly or indirectly, following traumatic injury to the nervous system. The outcome of this interaction can be beneficial - leading to successful neural repair, or detrimental - leading to functional deficits, depending on where the injury occurs. This review will discuss our understanding of neuron-immune cell interactions after traumatic injury to both the peripheral and central nervous system.
Collapse
Affiliation(s)
- Lora A Estera
- Department of Biology, Ball State University, Muncie, IN 47306, USA
| | - Sam P Walsh
- Department of Biology, Ball State University, Muncie, IN 47306, USA
| | - Jordan A Headen
- Department of Biology, Ball State University, Muncie, IN 47306, USA
| | | | - Ashley L Kalinski
- Department of Biology, Ball State University, Muncie, IN 47306, USA.
| |
Collapse
|
39
|
Mimic S, Aru B, Pehlivanoğlu C, Sleiman H, Andjus PR, Yanıkkaya Demirel G. Immunology of amyotrophic lateral sclerosis - role of the innate and adaptive immunity. Front Neurosci 2023; 17:1277399. [PMID: 38105925 PMCID: PMC10723830 DOI: 10.3389/fnins.2023.1277399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 11/07/2023] [Indexed: 12/19/2023] Open
Abstract
This review aims to summarize the latest evidence about the role of innate and adaptive immunity in Amyotrophic Lateral Sclerosis (ALS). ALS is a devastating neurodegenerative disease affecting upper and lower motor neurons, which involves essential cells of the immune system that play a basic role in innate or adaptive immunity, that can be neurotoxic or neuroprotective for neurons. However, distinguishing between the sole neurotoxic or neuroprotective function of certain cells such as astrocytes can be challenging due to intricate nature of these cells, the complexity of the microenvironment and the contextual factors. In this review, in regard to innate immunity we focus on the involvement of monocytes/macrophages, microglia, the complement, NK cells, neutrophils, mast cells, and astrocytes, while regarding adaptive immunity, in addition to humoral immunity the most important features and roles of T and B cells are highlighted, specifically different subsets of CD4+ as well as CD8+ T cells. The role of autoantibodies and cytokines is also discussed in distinct sections of this review.
Collapse
Affiliation(s)
- Stefan Mimic
- Centre for Laser Microscopy, Institute of Physiology and Biochemistry “Jean Giaja”, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Başak Aru
- Immunology Department, Faculty of Medicine, Yeditepe University, Istanbul, Türkiye
| | - Cemil Pehlivanoğlu
- Immunology Department, Faculty of Medicine, Yeditepe University, Istanbul, Türkiye
| | - Hadi Sleiman
- Faculty of Medicine, Yeditepe University, Istanbul, Türkiye
| | - Pavle R. Andjus
- Centre for Laser Microscopy, Institute of Physiology and Biochemistry “Jean Giaja”, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | | |
Collapse
|
40
|
Murata H, Phoo MTZ, Ochi T, Tomonobu N, Yamamoto KI, Kinoshita R, Miyazaki I, Nishibori M, Asanuma M, Sakaguchi M. Phosphorylated SARM1 is involved in the pathological process of rotenone-induced neurodegeneration. J Biochem 2023; 174:533-548. [PMID: 37725528 PMCID: PMC11033528 DOI: 10.1093/jb/mvad068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 08/29/2023] [Accepted: 09/10/2023] [Indexed: 09/21/2023] Open
Abstract
Sterile alpha and Toll/interleukin receptor motif-containing protein 1 (SARM1) is a NAD+ hydrolase that plays a key role in axonal degeneration and neuronal cell death. We reported that c-Jun N-terminal kinase (JNK) activates SARM1 through phosphorylation at Ser-548. The importance of SARM1 phosphorylation in the pathological process of Parkinson's disease (PD) has not been determined. We thus conducted the present study by using rotenone (an inducer of PD-like pathology) and neurons derived from induced pluripotent stem cells (iPSCs) from healthy donors and a patient with familial PD PARK2 (FPD2). The results showed that compared to the healthy neurons, FPD2 neurons were more vulnerable to rotenone-induced stress and had higher levels of SARM1 phosphorylation. Similar cellular events were obtained when we used PARK2-knockdown neurons derived from healthy donor iPSCs. These events in both types of PD-model neurons were suppressed in neurons treated with JNK inhibitors, Ca2+-signal inhibitors, or by a SARM1-knockdown procedure. The degenerative events were enhanced in neurons overexpressing wild-type SARM1 and conversely suppressed in neurons overexpressing the SARM1-S548A mutant. We also detected elevated SARM1 phosphorylation in the midbrain of PD-model mice. The results indicate that phosphorylated SARM1 plays an important role in the pathological process of rotenone-induced neurodegeneration.
Collapse
Key Words
- JNK
- PARK2
- Parkinson’s disease
- Phosphorylation
- SARM1.Abbreviations: ARM, armadillo/HEAT motif; DMSO, dimethyl sulfoxide; EGTA, ethylene glycol-bis(2-aminoethelether)-N: N: N: N-tetraacetic acid; iPSC, induced pluripotent stem cell; JNK, c-Jun N-terminal kinase; NAD, nicotinamide adenine dinucleotide; NSC, neural stem cell; NF-L, neurofilament-L; NF-M, neurofilament-M; PD, Parkinson’s disease; PINK1, PTEN-induced kinase 1; ROS, reactive oxygen species; SAM, sterile alpha motif; SARM1, sterile alpha and Toll/interleukin receptor motif-containing protein 1; SNpc, substantia nigra pars compacta; TH, tyrosine hydroxylase; TIR, Toll/interleukin receptor; WT, wild type
Collapse
Affiliation(s)
- Hitoshi Murata
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - May Tha Zin Phoo
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Toshiki Ochi
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Nahoko Tomonobu
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Ken-ichi Yamamoto
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Rie Kinoshita
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Ikuko Miyazaki
- Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Masahiro Nishibori
- Department of Translational Research and Drug Development, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Masato Asanuma
- Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Masakiyo Sakaguchi
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| |
Collapse
|
41
|
Waller TJ, Collins CA. Opposing roles of Fos, Raw, and SARM1 in the regulation of axonal degeneration and synaptic structure. Front Cell Neurosci 2023; 17:1283995. [PMID: 38099151 PMCID: PMC10719852 DOI: 10.3389/fncel.2023.1283995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 10/30/2023] [Indexed: 12/17/2023] Open
Abstract
Introduction The degeneration of injured axons is driven by conserved molecules, including the sterile armadillo TIR domain-containing protein SARM1, the cJun N-terminal kinase JNK, and regulators of these proteins. These molecules are also implicated in the regulation of synapse development though the mechanistic relationship of their functions in degeneration vs. development is poorly understood. Results and discussion Here, we uncover disparate functional relationships between SARM1 and the transmembrane protein Raw in the regulation of Wallerian degeneration and synaptic growth in motoneurons of Drosophila melanogaster. Our genetic data suggest that Raw antagonizes the downstream output MAP kinase signaling mediated by Drosophila SARM1 (dSarm). This relationship is revealed by dramatic synaptic overgrowth phenotypes at the larval neuromuscular junction when motoneurons are depleted for Raw or overexpress dSarm. While Raw antagonizes the downstream output of dSarm to regulate synaptic growth, it shows an opposite functional relationship with dSarm for axonal degeneration. Loss of Raw leads to decreased levels of dSarm in axons and delayed axonal degeneration that is rescued by overexpression of dSarm, supporting a model that Raw promotes the activation of dSarm in axons. However, inhibiting Fos also decreases dSarm levels in axons but has the opposite outcome of enabling Wallerian degeneration. The combined genetic data suggest that Raw, dSarm, and Fos influence each other's functions through multiple points of regulation to control the structure of synaptic terminals and the resilience of axons to degeneration.
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
| |
Collapse
|
42
|
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
|
43
|
Park SB, Cetinkaya-Fisgin A, Argyriou AA, Höke A, Cavaletti G, Alberti P. Axonal degeneration in chemotherapy-induced peripheral neurotoxicity: clinical and experimental evidence. J Neurol Neurosurg Psychiatry 2023; 94:962-972. [PMID: 37015772 PMCID: PMC10579520 DOI: 10.1136/jnnp-2021-328323] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 02/15/2023] [Indexed: 04/06/2023]
Abstract
Multiple pathological mechanisms are involved in the development of chemotherapy-induced peripheral neurotoxicity (CIPN). Recent work has provided insights into the molecular mechanisms underlying chemotherapy-induced axonal degeneration. This review integrates evidence from preclinical and clinical work on the onset, progression and outcome of axonal degeneration in CIPN. We review likely triggers of axonal degeneration in CIPN and highlight evidence of molecular pathways involved in axonal degeneration and their relevance to CIPN, including SARM1-mediated axon degeneration pathway. We identify potential clinical markers of axonal dysfunction to provide early identification of toxicity as well as present potential treatment strategies to intervene in axonal degeneration pathways. A greater understanding of axonal degeneration processes in CIPN will provide important information regarding the development and progression of axonal dysfunction more broadly and will hopefully assist in the development of successful interventions for CIPN and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Susanna B Park
- Brain and Mind Centre, Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Camperdown, New South Wales, Australia
| | - Aysel Cetinkaya-Fisgin
- Department of Neurology, Neuromuscular Division, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Andreas A Argyriou
- Department of Neurology, "Agios Andreas" State General Hospital of Patras, Patras, Greece
| | - Ahmet Höke
- Department of Neurology, Neuromuscular Division, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Guido Cavaletti
- Experimental Neurology Unit and Milan Center for Neuroscience, University of Milano-Bicocca, Monza, Italy
| | - Paola Alberti
- Experimental Neurology Unit and Milan Center for Neuroscience, University of Milano-Bicocca, Monza, Italy
| |
Collapse
|
44
|
Sun L, Bian F, Xu D, Luo Y, Wang Y, Zhao Y. Tailoring biomaterials for biomimetic organs-on-chips. MATERIALS HORIZONS 2023; 10:4724-4745. [PMID: 37697735 DOI: 10.1039/d3mh00755c] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/13/2023]
Abstract
Organs-on-chips are microengineered microfluidic living cell culture devices with continuously perfused chambers penetrating to cells. By mimicking the biological features of the multicellular constructions, interactions among organs, vascular perfusion, physicochemical microenvironments, and so on, these devices are imparted with some key pathophysiological function levels of living organs that are difficult to be achieved in conventional 2D or 3D culture systems. In this technology, biomaterials are extremely important because they affect the microstructures and functionalities of the organ cells and the development of the organs-on-chip functions. Thus, herein, we provide an overview on the advances of biomaterials for the construction of organs-on-chips. After introducing the general components, structures, and fabrication techniques of the biomaterials, we focus on the studies of the functions and applications of these biomaterials in the organs-on-chips systems. Applications of the biomaterial-based organs-on-chips as alternative animal models for pharmaceutical, chemical, and environmental tests are described and highlighted. The prospects for exciting future directions and the challenges of biomaterials for realizing the further functionalization of organs-on-chips are also presented.
Collapse
Affiliation(s)
- Lingyu Sun
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Feika Bian
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Dongyu Xu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Yuan Luo
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.
| | - Yongan Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
- Southeast University Shenzhen Research Institute, Shenzhen 518071, China
| |
Collapse
|
45
|
Cao Y, Chen H, Yang J. Neuroanatomy of lymphoid organs: Lessons learned from whole-tissue imaging studies. Eur J Immunol 2023; 53:e2250136. [PMID: 37377338 DOI: 10.1002/eji.202250136] [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: 12/16/2022] [Revised: 06/06/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023]
Abstract
Decades of extensive research have documented the presence of neural innervations of sensory, sympathetic, or parasympathetic origin in primary and secondary lymphoid organs. Such neural inputs can release neurotransmitters and neuropeptides to directly modulate the functions of various immune cells, which represents one of the essential aspects of the body's neuroimmune network. Notably, recent studies empowered by state-of-the-art imaging techniques have comprehensively assessed neural distribution patterns in BM, thymus, spleen, and LNs of rodents and humans, helping clarify several controversies lingering in the field. In addition, it has become evident that neural innervations in lymphoid organs are not static but undergo alterations in pathophysiological contexts. This review aims to update the current information on the neuroanatomy of lymphoid organs obtained through whole-tissue 3D imaging and genetic approaches, focusing on anatomical features that may designate the functional modulation of immune responses. Moreover, we discuss several critical questions that call for future research, which will advance our in-depth understanding of the importance and complexity of neural control of lymphoid organs.
Collapse
Affiliation(s)
- Ying Cao
- Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Hongjie Chen
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Jing Yang
- Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
- IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
- Shenzhen Bay Laboratory, Institute of Molecular Physiology, Shenzhen, China
| |
Collapse
|
46
|
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
|
47
|
Kim M, Kim H, Kang BG, Lee J, Kim T, Lee H, Jung J, Oh MJ, Seo S, Ryu MJ, Sung Y, Lee Y, Yeom J, Han G, Cha SS, Jung H, Kim HS. Discovery of a novel NAMPT inhibitor that selectively targets NAPRT-deficient EMT-subtype cancer cells and alleviates chemotherapy-induced peripheral neuropathy. Theranostics 2023; 13:5075-5098. [PMID: 37771778 PMCID: PMC10526665 DOI: 10.7150/thno.85356] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 09/05/2023] [Indexed: 09/30/2023] Open
Abstract
Background: Exploiting synthetic lethality (SL) relationships between protein pairs has emerged as an important avenue for the development of anti-cancer drugs. Nicotinamide phosphoribosyltransferase (NAMPT) is the rate-limiting enzyme of the NAD+ salvage pathway, having an SL relationship with nicotinic acid phosphoribosyltransferase (NAPRT), the key enzyme in the NAD+ Preiss-Handler pathway. NAMPT inhibitor holds clinical potential not only as a promising cancer treatment but also as a means of protection against chemotherapy-induced-peripheral-neuropathy (CIPN). However, as NAD+ is essential for normal cells, the clinical use of NAMPT inhibitors is challenging. This study aimed to identify a novel NAMPT inhibitor with enhanced selective cytotoxicity against NAPRT-deficient cancer cells as well as prominent efficacy in alleviating CIPN. Methods: We began by conducting drug derivatives screening in a panel of lung cancer cell lines to select an agent with the broadest therapeutic window between the NAPRT-negative and-positive cancer cell lines. Both in vitro and In vivo comparative analyses were conducted between A4276 and other NAMPT inhibitors to evaluate the NAPRT-negative cancer cell selectivity and the underlying distinct NAMPT inhibition mechanism of A4276. Patient-derived tumor transcriptomic data and protein levels in various cancer cell lines were analyzed to confirm the correlation between NAPRT depletion and epithelial-to-mesenchymal transition (EMT)-like features in various cancer types. Finally, the efficacy of A4276 for axonal protection and CIPN remedy was examined in vitro and in vivo. Results: The biomarker-driven phenotypic screening led to a discovery of A4276 with prominent selectivity against NAPRT-negative cancer cells compared with NAPRT-positive cancer cells and normal cells. The cytotoxic effect of A4276 on NAPRT-negative cells is achieved through its direct binding to NAMPT, inhibiting its enzymatic function at an optimal and balanced level allowing NAPRT-positive cells to survive through NAPRT-dependent NAD+ synthesis. NAPRT deficiency serves as a biomarker for the response to A4276 as well as an indicator of EMT-subtype cancer in various tumor types. Notably, A4276 protects axons from Wallerian degeneration more effectively than other NAMPT inhibitors by decreasing NMN-to-NAD+ ratio. Conclusion: This study demonstrates that A4276 selectively targets NAPRT-deficient EMT-subtype cancer cells and prevents chemotherapy-induced peripheral neuropathy, highlighting its potential as a promising anti-cancer agent for use in cancer monotherapy or combination therapy with conventional chemotherapeutics.
Collapse
Affiliation(s)
- Minjee Kim
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Hyeyoung Kim
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Department of Anatomy, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Bu-Gyeong Kang
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Jooyoung Lee
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Checkmate Therapeutics Inc., Seoul, 07207, Republic of Korea
| | - Taegun Kim
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Hwanho Lee
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jane Jung
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Department of Anatomy, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Myung Joon Oh
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Seungyoon Seo
- Prometabio Research Institute, Prometabio Co., Ltd. Hanam-si, Gyeonggi-do 12939, Republic of Korea
| | - Myung-Jeom Ryu
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Yeojin Sung
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Yunji Lee
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jeonghun Yeom
- Prometabio Research Institute, Prometabio Co., Ltd. Hanam-si, Gyeonggi-do 12939, Republic of Korea
| | - Gyoonhee Han
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Sun-Shin Cha
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Hosung Jung
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Department of Anatomy, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Hyun Seok Kim
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Checkmate Therapeutics Inc., Seoul, 07207, Republic of Korea
| |
Collapse
|
48
|
Griffioen G. Calcium Dyshomeostasis Drives Pathophysiology and Neuronal Demise in Age-Related Neurodegenerative Diseases. Int J Mol Sci 2023; 24:13243. [PMID: 37686048 PMCID: PMC10487569 DOI: 10.3390/ijms241713243] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 08/21/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
This review postulates that age-related neurodegeneration entails inappropriate activation of intrinsic pathways to enable brain plasticity through deregulated calcium (Ca2+) signalling. Ca2+ in the cytosol comprises a versatile signal controlling neuronal cell physiology to accommodate adaptive structural and functional changes of neuronal networks (neuronal plasticity) and, as such, is essential for brain function. Although disease risk factors selectively affect different neuronal cell types across age-related neurodegenerative diseases (NDDs), these appear to have in common the ability to impair the specificity of the Ca2+ signal. As a result, non-specific Ca2+ signalling facilitates the development of intraneuronal pathophysiology shared by age-related NDDs, including mitochondrial dysfunction, elevated reactive oxygen species (ROS) levels, impaired proteostasis, and decreased axonal transport, leading to even more Ca2+ dyshomeostasis. These core pathophysiological processes and elevated cytosolic Ca2+ levels comprise a self-enforcing feedforward cycle inevitably spiralling toward high levels of cytosolic Ca2+. The resultant elevated cytosolic Ca2+ levels ultimately gear otherwise physiological effector pathways underlying plasticity toward neuronal demise. Ageing impacts mitochondrial function indiscriminately of the neuronal cell type and, therefore, contributes to the feedforward cycle of pathophysiology development seen in all age-related NDDs. From this perspective, therapeutic interventions to safely restore Ca2+ homeostasis would mitigate the excessive activation of neuronal destruction pathways and, therefore, are expected to have promising neuroprotective potential.
Collapse
|
49
|
Habek M, Blazekovic A, Gotovac Jercic K, Pivac N, Outero TF, Borovecki F, Brinar V. Genome-Wide Expression Profile in People with Optic Neuritis Associated with Multiple Sclerosis. Biomedicines 2023; 11:2209. [PMID: 37626706 PMCID: PMC10452153 DOI: 10.3390/biomedicines11082209] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/13/2023] [Accepted: 07/20/2023] [Indexed: 08/27/2023] Open
Abstract
The aim of this study was to perform a genome-wide expression analysis of whole-blood samples from people with optic neuritis (ON) and to determine differentially expressed mRNAs compared to healthy control subjects. The study included eight people with acute ON and six healthy control subjects. Gene expression was analyzed using DNA microarrays for whole-human-genome analysis, which contain 54,675 25-base pairs. The additional biostatistical analysis included gene ontology analysis and gene set enrichment analysis (GSEA). Quantitative RT-PCR (qPCR) was used to confirm selected differentially expressed genes. In total, 722 differently expressed genes were identified, with 377 exhibiting increased, and 345 decreased, expression. Gene ontology analysis and GSEA revealed that protein phosphorylation and intracellular compartment, apoptosis inhibition, pathways involved in cell cycles, T and B cell functions, and anti-inflammatory central nervous system (CNS) pathways are implicated in ON pathology. qPCR confirmed the differential expression of eight selected genes, with SLPI, CR3, and ITGA4 exhibiting statistically significant results. In conclusion, whole-blood gene expression analysis showed significant differences in the expression profiles of people with ON compared to healthy control subjects. Additionally, pathways involved in T cell regulation and anti-inflammatory pathways within CNS were identified as important in the early phases of MS.
Collapse
Affiliation(s)
- Mario Habek
- Department of Neurology, Referral Center for Autonomic Nervous System Disorders, University Hospital Centre Zagreb, 10000 Zagreb, Croatia
| | - Antonela Blazekovic
- Department for Functional Genomics, Center for Translational and Clinical Research, University of Zagreb School of Medicine, University Hospital Centre Zagreb, 10000 Zagreb, Croatia
- Department for Anatomy and Clinical Anatomy, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Kristina Gotovac Jercic
- Department for Functional Genomics, Center for Translational and Clinical Research, University of Zagreb School of Medicine, University Hospital Centre Zagreb, 10000 Zagreb, Croatia
- Department of Neurology, University Hospital Centre Zagreb, 10000 Zagreb, Croatia
| | - Nela Pivac
- Division of Molecular Medicine, Rudjer Bošković Institute, 10002 Zagreb, Croatia
| | - Tiago Fleming Outero
- Department of Experimental Neurodegeneration, Centre for Biostructural Imaging of Neurodegeneration, University Medical Centre Göttingen, 37075 Göttingen, Germany
- Max Planck Institute for Experimental Medicine, 37075 Göttingen, Germany
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle upon Tyne NE1 7RU, UK
- German Centre for Neurodegenerative Diseases (DZNE), 17475 Göttingen, Germany
| | - Fran Borovecki
- Department for Functional Genomics, Center for Translational and Clinical Research, University of Zagreb School of Medicine, University Hospital Centre Zagreb, 10000 Zagreb, Croatia
- Department of Neurology, University Hospital Centre Zagreb, 10000 Zagreb, Croatia
| | - Vesna Brinar
- Department of Neurology, University Hospital Centre Zagreb, 10000 Zagreb, Croatia
| |
Collapse
|
50
|
Crawford KL, Lee YJ, Hom M, Rosenthal EL, Orosco RK, Nguyen QT. Identification of Degenerated Murine Facial Nerves With Fluorescence Labeling After Transection Injury. Otolaryngol Head Neck Surg 2023; 169:234-242. [PMID: 36758958 DOI: 10.1002/ohn.262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 12/08/2022] [Accepted: 12/24/2022] [Indexed: 02/11/2023]
Abstract
OBJECTIVE Delayed peripheral nerve repair is complicated by nerve degeneration and atrophy that can prevent identification. We use a murine facial nerve transection model to demonstrate the efficacy of ALM-488 (bevonescein) in labeling degenerated facial nerves with quantitative image analysis and qualitative survey data. STUDY DESIGN Prospective cohort study. SETTING Laboratory. METHODS Ten wild-type mice underwent transection of the lower facial nerve division with subsequent degeneration. Either 9 (n = 5 mice) or 12 (n = 5 mice) weeks later, mice underwent intravenous infusion of ALM-488 with in vivo real-time fluorescence imaging (FL) of the facial nerve. Using ImageJ, the mean gray value of each nerve segment under white light reflectance (WLR) and FL was compared to that of adjacent soft tissue to calculate the signal-to-background ratio (SBR). A survey was distributed to evaluate the perceived utility of ALM-488 in surgeon identification of degenerated nerves. RESULTS The mean SBR of degenerated nerves was 1.08 (standard deviation [SD]: 0.07) under WLR and 2.11 (SD: 0.31) under FL (p < 0.001). In mice with degenerated nerves, survey participants identified on average 3.01 (SD: 1.84) nerve branches under WLR and 5.73 (SD: 1.88) under FL (p < 0.0001). Under FL, 47 of 48 survey responses correctly identified isolated, degenerated nerves; in contrast, only 12 responses identified degenerated nerves under WLR (p < 0.0001). CONCLUSION Preoperative intravenous infusion of ALM-488 with FL improves the identification of degenerated facial nerves. ALM-488 also improves surgeon confidence in nerve identification, particularly in degenerated nerve branches that are not visible with WLR.
Collapse
Affiliation(s)
- Kayva L Crawford
- Department of Otolaryngology-Head and Neck Surgery, University of California San Diego, San Diego, California, USA
| | - Yu-Jin Lee
- Department of Otolaryngology-Head and Neck Surgery, Stanford University, Palo Alto, California, USA
| | - Marisa Hom
- Department of Otolaryngology-Head and Neck Surgery, Stanford University, Palo Alto, California, USA
| | - Eben L Rosenthal
- Department of Otolaryngology-Head and Neck Surgery, Vanderbilt University, Nashville, Tennessee, USA
| | - Ryan K Orosco
- Department of Otolaryngology-Head and Neck Surgery, University of California San Diego, San Diego, California, USA
- Hanna and Mark Gleiberman Head and Neck Cancer Center, Moores Cancer Center, University of California San Diego, California, San Diego, USA
| | - Quyen T Nguyen
- Department of Otolaryngology-Head and Neck Surgery, University of California San Diego, San Diego, California, USA
| |
Collapse
|