1
|
Fu Y, Rui X, Zhu S, Guo C, Li H, Pan Z, Wu X, He W. Research status of regenerative difficulties after central nervous system injury. Regen Ther 2025; 29:493-498. [PMID: 40390864 PMCID: PMC12088777 DOI: 10.1016/j.reth.2025.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 04/08/2025] [Accepted: 04/17/2025] [Indexed: 05/21/2025] Open
Abstract
Multiple studies have shown that permanent functional disabilities caused after nerve damage are mainly due to the limited ability of damaged neurons in the central nervous system (CNS) to regenerate axons and re-establish functional connections. Most axons in the CNS of adult mammals cannot reactivate their intrinsic growth program after injury, making axonal regeneration difficult when damaged. This article provides a systematic review of the response processes following CNS injury and the factors affecting repair and regeneration, focusing on the molecular mechanisms that regulate the regeneration of damaged axons, in hopes of offering new insights for the repair of CNS injuries.
Collapse
Affiliation(s)
- Yunxia Fu
- School of Stomatology, Jinan University, Guangzhou 510632, China
| | - Xi Rui
- Hospital of Stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- School of Stomatology, Jinan University, Guangzhou 510632, China
| | - Shumin Zhu
- Hospital of Stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- School of Stomatology, Jinan University, Guangzhou 510632, China
| | - Chenqu Guo
- School of Stomatology, Jinan University, Guangzhou 510632, China
| | - Haoyang Li
- School of Stomatology, Jinan University, Guangzhou 510632, China
| | - Zhenhao Pan
- School of Stomatology, Jinan University, Guangzhou 510632, China
| | - Xuanhao Wu
- School of Stomatology, Jinan University, Guangzhou 510632, China
| | - Wenpeng He
- Hospital of Stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- School of Stomatology, Jinan University, Guangzhou 510632, China
| |
Collapse
|
2
|
Tohda C. Pharmacological intervention for chronic phase of spinal cord injury. Neural Regen Res 2025; 20:1377-1389. [PMID: 38934397 PMCID: PMC11624870 DOI: 10.4103/nrr.nrr-d-24-00176] [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: 02/08/2024] [Revised: 04/24/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
Spinal cord injury is an intractable traumatic injury. The most common hurdles faced during spinal cord injury are failure of axonal regrowth and reconnection to target sites. These also tend to be the most challenging issues in spinal cord injury. As spinal cord injury progresses to the chronic phase, lost motor and sensory functions are not recovered. Several reasons may be attributed to the failure of recovery from chronic spinal cord injury. These include factors that inhibit axonal growth such as activated astrocytes, chondroitin sulfate proteoglycan, myelin-associated proteins, inflammatory microglia, and fibroblasts that accumulate at lesion sites. Skeletal muscle atrophy due to denervation is another chronic and detrimental spinal cord injury-specific condition. Although several intervention strategies based on multiple outlooks have been attempted for treating spinal cord injury, few approaches have been successful. To treat chronic spinal cord injury, neural cells or tissue substitutes may need to be supplied in the cavity area to enable possible axonal growth. Additionally, stimulating axonal growth activity by extrinsic factors is extremely important and essential for maintaining the remaining host neurons and transplanted neurons. This review focuses on pharmacotherapeutic approaches using small compounds and proteins to enable axonal growth in chronic spinal cord injury. This review presents some of these candidates that have shown promising outcomes in basic research ( in vivo animal studies) and clinical trials: AA-NgR(310)ecto-Fc (AXER-204), fasudil, phosphatase and tensin homolog protein antagonist peptide 4, chondroitinase ABC, intracellular sigma peptide, (-)-epigallocatechin gallate, matrine, acteoside, pyrvate kinase M2, diosgenin, granulocyte-colony stimulating factor, and fampridine-sustained release. Although the current situation suggests that drug-based therapies to recover function in chronic spinal cord injury are limited, potential candidates have been identified through basic research, and these candidates may be subjects of clinical studies in the future. Moreover, cocktail therapy comprising drugs with varied underlying mechanisms may be effective in treating the refractory status of chronic spinal cord injury.
Collapse
Affiliation(s)
- Chihiro Tohda
- Section of Neuromedical Science, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| |
Collapse
|
3
|
Ruscher K, Michalettos G, Abu Hamdeh S, Clausen F, Nolan AL, Flygt J, Özen I, Marklund N. Persistent increase of Nogo-A-positive cells and dynamic reduction in oligodendroglia lineage cells in white matter regions following experimental and clinical traumatic brain injury. J Neuropathol Exp Neurol 2025; 84:423-435. [PMID: 40085014 PMCID: PMC12012378 DOI: 10.1093/jnen/nlaf017] [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] [Indexed: 03/16/2025] Open
Abstract
White matter (WM) disruption and atrophy is a consequence of traumatic brain injury (TBI) and contributes to persisting cognitive impairment. An increased expression of the myelin-associated axonal outgrowth inhibitor Nogo-A and oligodendrocyte pathology might be negatively associated with postinjury WM changes. Here, we analyzed brain tissue from severe TBI patients, obtained by surgical decompression in the early postinjury phase and postmortem brain tissue of long-term TBI survivors and observed an increased number of Nogo-A+ cells in WM tracts such as the corpus callosum (CC). Likewise, the number of Nogo-A+ cells in the CC was increased from day 7 postinjury to 6 months postinjury (mpi) following central fluid percussion injury (cFPI) in mice. In addition, the number of Olig2+ cells in the CC and capsula externa remained constant, while the numbers of Olig2+/CC1+ and GST-π+ mature oligodendrocytes declined throughout the observation time of 18 months. A significantly lower number of Olig2+/CC1+ cells was found in cFPI mice compared to controls at 18 mpi. Persistent vulnerability of oligodendrocytes in combination with dynamic alterations of Nogo-A expression may have implications for the WM atrophy and insufficient recovery observed after TBI.
Collapse
Affiliation(s)
- Karsten Ruscher
- Department of Clinical Sciences Lund, Neurosurgery, Lund University, Lund, Sweden
- Department of Clinical Sciences, Division of Neurosurgery, Laboratory for Experimental Brain Research, Lund University, Lund, Sweden
| | - Georgios Michalettos
- Department of Clinical Sciences Lund, Neurosurgery, Lund University, Lund, Sweden
| | - Sami Abu Hamdeh
- Department of Medical Sciences, Section of Neurosurgery, Uppsala University, Uppsala, Sweden
| | - Fredrik Clausen
- Department of Medical Sciences, Section of Neurosurgery, Uppsala University, Uppsala, Sweden
| | - Amber L Nolan
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Johanna Flygt
- Department of Medical Sciences, Section of Neurosurgery, Uppsala University, Uppsala, Sweden
| | - Ilknur Özen
- Department of Clinical Sciences Lund, Neurosurgery, Lund University, Lund, Sweden
| | - Niklas Marklund
- Department of Clinical Sciences Lund, Neurosurgery, Lund University, Lund, Sweden
- Department of Clinical Sciences, Neurosurgery, Lund University, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
4
|
Sun W, Dion E, Laredo F, Okonak A, Sepeda JA, Haykal E, Zhou M, El-Hodiri HM, Fischer AJ, Silver J, Peng J, Sas A, Tedeschi A. In vivo programming of adult pericytes aids axon regeneration by providing cellular bridges for SCI repair. Mol Ther 2025:S1525-0016(25)00294-1. [PMID: 40253585 DOI: 10.1016/j.ymthe.2025.04.020] [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: 06/21/2024] [Revised: 03/04/2025] [Accepted: 04/16/2025] [Indexed: 04/22/2025] Open
Abstract
Pericytes are contractile cells of the microcirculation that participate in wound healing after spinal cord injury (SCI). Thus far, the extent to which pericytes cause or contribute to axon growth and regeneration failure after SCI remains controversial. Here, we found that SCI leads to profound changes in vasculature architecture and pericyte coverage. We demonstrated that pericytes constrain sensory axons on their surface, causing detrimental structural and functional changes in adult dorsal root ganglion neurons that contribute to axon regeneration failure after SCI. Perhaps more excitingly, we discovered that in vivo programming of adult pericytes via local administration of platelet-derived growth factor BB (PDGF-BB) effectively promotes axon regeneration and recovery of hindlimb function by contributing to the formation of cellular bridges that span the lesion. Ultrastructural analysis showed that PDGF-BB induced fibronectin fibril alignment and extension, effectively converting adult pericytes into a permissive substrate for axon growth. In addition, PDGF-BB localized delivery positively affects the physical and chemical nature of the lesion environment, thereby creating more favorable conditions for SCI repair. Thus, therapeutic manipulation rather than wholesale ablation of pericytes can be exploited to prime axon regeneration and SCI repair.
Collapse
Affiliation(s)
- Wenjing Sun
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Elliot Dion
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Fabio Laredo
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA; University of Nottingham, Nottingham NG7 2QL, UK
| | - Allyson Okonak
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Jesse A Sepeda
- Department of Neurology, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Esraa Haykal
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Min Zhou
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Heithem M El-Hodiri
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Andy J Fischer
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Jerry Silver
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Juan Peng
- Center for Biostatistics and Bioinformatics, The Ohio State University, Columbus, OH 43210, USA
| | - Andrew Sas
- Department of Neurology, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA; Chronic Brain Injury Program, The Ohio State University, Columbus, OH 43210, USA
| | - Andrea Tedeschi
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA; Chronic Brain Injury Program, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
5
|
Zhang J, Chen X, Du K, Zhang Z, Ma Y, Kuang Y, Yu S, Shu H. NgR1 knockout increased neuronal excitability and altered seizure pattern in traumatic brain injury mice brain after PTZ-induced seizure. PLoS One 2025; 20:e0321447. [PMID: 40233041 PMCID: PMC11999111 DOI: 10.1371/journal.pone.0321447] [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: 09/10/2024] [Accepted: 03/06/2025] [Indexed: 04/17/2025] Open
Abstract
The recovery process from traumatic brain injury (TBI) is significantly impeded by inhibitors such as Nogo-A, myelin associated glycoprotein, and oligodendrocyte myelin glycoprotein, which exert an impact on the regeneration and repair of neuronal axons through their binding to Nogo-66 receptor 1 (NgR1). Recent research findings have revealed that NgR1 signaling may play a pivotal role in various seizure mechanisms, including the regulation of synaptic plasticity and migration of neural precursor cells. In this study, wild type (WT) and NgR1 knockout (KO) mice were utilized to establish craniocerebral injury models, while pentylenetetrazol (PTZ) was employed to induce seizures in both groups of mice following TBI. The results revealed that NgR1 KO mice exhibited heightened levels of neuronal electrical activity, along with elevated seizure scores compared to WT controls. Immunofluorescence staining demonstrated an increase in the number of excitatory synapses (P < 0.001) and a decrease in inhibitory synaptic density (P < 0.001) in NgR1 KO mice. Furthermore, the NgR1 KO model mice also displayed an augmentation in the number of presynaptic vesicles (P < 0.001), a narrowing of the synaptic gap (P < 0.001), and an elongation of the synaptic active region (P < 0.001). Our findings have demonstrated that in the previous single cognition of NgR1 inhibition in nerve function repair following TBI, revealing the potential risks associated with inhibiting NgR1 activity in nerve function repair following TBI, and providing a new perspective for understanding the role of NgR1 in the nervous system.
Collapse
Affiliation(s)
- Jinwei Zhang
- Department of Neurosurgery, General Hospital of Western Theater Command of PLA, Chengdu, China
- Department of Neurosurgery, Sichuan Tianfu New Area People’s Hospital, Chengdu, China
| | - Xin Chen
- Department of Neurosurgery, General Hospital of Western Theater Command of PLA, Chengdu, China
| | - Kejun Du
- Department of Neurosurgery, General Hospital of Western Theater Command of PLA, Chengdu, China
| | - Zhi Zhang
- Department of Neurosurgery, General Hospital of Western Theater Command of PLA, Chengdu, China
| | - Yuan Ma
- Department of Neurosurgery, General Hospital of Western Theater Command of PLA, Chengdu, China
| | - Yongqin Kuang
- Department of Neurosurgery, General Hospital of Western Theater Command of PLA, Chengdu, China
| | - Sixun Yu
- Department of Neurosurgery, General Hospital of Western Theater Command of PLA, Chengdu, China
| | - Haifeng Shu
- Department of Neurosurgery, General Hospital of Western Theater Command of PLA, Chengdu, China
| |
Collapse
|
6
|
Sun X, Li L, Huang L, Li Y, Wang L, Wei Q. Harnessing spinal circuit reorganization for targeted functional recovery after spinal cord injury. Neurobiol Dis 2025; 207:106854. [PMID: 40010611 DOI: 10.1016/j.nbd.2025.106854] [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: 01/10/2025] [Revised: 02/13/2025] [Accepted: 02/23/2025] [Indexed: 02/28/2025] Open
Abstract
Spinal cord injury (SCI) disrupts the communication between the brain and spinal cord, resulting in the loss of motor function below the injury site. However, spontaneous structural and functional plasticity occurs in neural circuits after SCI, with unaffected synaptic inputs forming new connections and detour pathways to support recovery. The review discusses various mechanisms of circuit reorganization post-SCI, including supraspinal pathways, spinal interneurons, and spinal central pattern generators. Functional recovery may rely on maintaining a balance between excitatory and inhibitory neural activity, as well as enhancing proprioceptive input, which plays a key role in limb stability. The review emphasizes the importance of endogenous neuronal regeneration, neuromodulation therapies (such as electrical stimulation) and proprioception in SCI treatment. Future research should integrate advanced technologies such as gene targeting, imaging, and single-cell mapping to better understand the mechanisms underpinning SCI recovery, aiming to identify key neuronal subpopulations for targeted reconstruction and enhanced functional recovery. By harnessing spinal circuit reorganization, these efforts hold the potential to pave the way for more precise and effective strategies for functional recovery after SCI.
Collapse
Affiliation(s)
- Xin Sun
- Department of Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, PR China
| | - Lijuan Li
- Department of Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, PR China
| | - Liyi Huang
- Department of Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, PR China
| | - Yangan Li
- Department of Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, PR China
| | - Lu Wang
- Department of Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, PR China
| | - Quan Wei
- Department of Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, PR China.
| |
Collapse
|
7
|
Kondiles BR, Rana S, Weiner D, Blesch A, St. John J, Haag-Molkenteller C, Freund P, Guest J, Mikol DD, Harkema S, Trumbower RD, Fehlings MG, Weidner N, Hogge GS, Field-Fote EC, Baptista MA, Curt A, Hsieh J, Jones L. Lessons Learned and Recommendations from a SCOPE Spinal Cord Injury Neurorestorative Clinical Trials Update. Neurotrauma Rep 2025; 6:210-231. [PMID: 40309157 PMCID: PMC12040541 DOI: 10.1089/neur.2024.0163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025] Open
Abstract
The Spinal Cord Outcomes Partnership Endeavors presented a clinical trials update (CTU) in collaboration with the International Spinal Research Trust as a precourse to their annual meeting. Selected trials adhered to a priori considerations, prioritizing novelty and a focus on neurorestorative approaches. The sessions featured 13 speakers, covering 4 in-preparation, 4 in-progress, and 4 recently completed trials. In addition to in-person attendance, individuals worldwide viewed a live stream of the presentations. Approximately 1600 participants, comprising clinicians, researchers, industry stakeholders, foundations, and individuals with lived experiences, engaged in the CTU through both in-person and virtual channels. Presentations represented a variety of approaches, including drug, biological, and device-based therapeutics. This summary provides high-level summaries of the trials presented and the resulting discussions including lessons learned. Rather than recapitulating published data, the presentations and discussions emphasized the novelty and strengths of each trial, practical aspects of translation, and lessons learned. Throughout the day, several discussion themes surfaced. These included reflections on the suitability of outcome measures and the distinction between statistically or clinically meaningful effects and meaningful changes in quality of life. Additional topics included novel trial designs, selection of inclusion criteria, recognizing the indispensable role of rehabilitation, tailoring approaches to individual needs, the importance of integrating lived experience, and emphasizing the importance of establishing robust pre-clinical data packages before venturing into clinical translation. Importantly, strategic directives are summarized to address these challenges, focusing resources and efforts to steer forthcoming trials effectively.
Collapse
Affiliation(s)
- Bethany R. Kondiles
- International Collaboration on Repair Discoveries, Department of Zoology, University of British Columbia, Vancouver, Canada
| | - Sabhya Rana
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | | | - Armin Blesch
- Department of Neurosciences, University of California-San Diego, and VA San Diego Healthcare System, La Jolla, California, USA
| | - James St. John
- Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith Sciences and Griffith Health, Griffith University, Australia
| | | | - Patrick Freund
- Spinal Cord Injury Center, Research, Balgrist University Hospital, Zürich, Switzerland
| | - James Guest
- Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
| | | | | | - Randy D. Trumbower
- Department of Physical Medicine & Rehabilitation, Spaulding Research Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael G. Fehlings
- Division of Neurosurgery and Spine Program, Department of Surgery, University of Toronto, Toronto, Canada
| | - Norbert Weidner
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Gary S. Hogge
- GSH Biomedical Consulting, San Francisco, California, USA
| | | | | | - Armin Curt
- Balgrist University Hospital, Zürich, Switzerland
| | | | - Linda Jones
- Christopher & Dana Reeve Foundation Short Hills, New Jersey, USA; Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
8
|
Kunz PB, Maurer MA, Vollmer J, Machacek M, Weinmann O, Klisic J, Schwab ME. Intrathecal administration of Anti-Nogo-A antibody in macaque monkeys: Pharmacokinetics, tissue penetration and target interaction. Neurotherapeutics 2025; 22:e00484. [PMID: 39572304 PMCID: PMC12014330 DOI: 10.1016/j.neurot.2024.e00484] [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: 05/11/2024] [Revised: 10/11/2024] [Accepted: 11/06/2024] [Indexed: 03/05/2025] Open
Abstract
Intrathecal drug administration represents a promising method to deliver biologics effectively to the central nervous system (CNS). However, little is known about the tolerability and pharmacokinetics of intrathecally applied antibodies. Hence, the focus of this study was to evaluate the toxicity, pharmacokinetic, and pharmacodynamic properties of an intrathecally administered human monoclonal antibody against the growth inhibitory CNS membrane protein Nogo-A in the non-human primate (NHP). The antibody was repeatedly injected into the lumbar cerebrospinal fluid (CSF) sack of NHPs, Macaca fascicularis (N = 18), at three dose levels (placebo, 75 and 150 mg antibody/injection, n = 6/group). CSF and serum samples were collected for pharmacokinetic analysis. The health status was constantly monitored to detect any treatment-related abnormalities. After sacrifice, the CNS tissues were evaluated by immunohistochemistry and biochemistry to study the antibody distribution and target interaction in the spinal cord and brain. No treatment-related side effects were observed, and the treatment was well tolerated by NHPs. After administration, the antibody was rapidly cleared from the CSF with a half-life of 6.4 h and accumulated in the serum where it showed a half-life of 13.7 days. The antibody distributed over the spinal cord and brain, penetrated into the CNS parenchyma where it bound to Nogo-A expressing neurons and oligodendrocytes, and induced significant (P < 0.05) downregulation of the target antigen Nogo-A. Collectively, these results support the direct administration of therapeutic antibodies into the CSF and are of relevance for the antibody-based therapeutics currently in development for different CNS diseases.
Collapse
Affiliation(s)
- Pascal B Kunz
- NovaGo Therapeutics AG, 8952 Zurich-Schlieren, Switzerland
| | | | | | | | | | - Jelena Klisic
- NovaGo Therapeutics AG, 8952 Zurich-Schlieren, Switzerland
| | | |
Collapse
|
9
|
Yang H, Gao J, Wang HY, Ma XM, Liu BY, Song QZ, Cheng H, Li S, Long ZY, Lu XM, Wang YT. The effects and possible mechanisms of whole-body vibration on cognitive function: A narrative review. Brain Res 2025; 1850:149392. [PMID: 39662790 DOI: 10.1016/j.brainres.2024.149392] [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: 09/21/2024] [Revised: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 12/13/2024]
Abstract
Whole-body vibration (WBV) is a physical stimulation method that transmits mechanical oscillations to the entire body through a vibration platform or device. Biokinetic and epidemiologic studies have shown that prolonged exposure to high-intensity WBV increases health risks, primarily to the lumbar spine and the nervous system connected to it. There is currently insufficient evidence to demonstrate a quantitative relationship between vibration exposure and risk of health effects. The positive effects of WBV on increasing muscle strength and improving balance and flexibility are well known, but its effects on cognitive function are more complex, with mixed findings, largely related to vibration conditions, including frequency, amplitude, and duration. Studies have shown that short-term low-frequency WBV may have a positive impact on cognitive function, demonstrates potential rehabilitation benefits in enhancing learning and memory, possibly by promoting neuromuscular coordination and enhancing neural plasticity. However, long term exposure to vibration may lead to chronic stress in nerve tissue, affecting nerve conduction efficiency and potentially interfering with neuroprotective mechanisms, thereby having a negative impact on cognitive ability, even causes symptoms such as cognitive decline, mental fatigue, decreased attention, and drowsiness. This literature review aimed to explore the effects of WBV on cognitive function and further to analyze the possible mechanisms. Based on the analysis of literatures, we came to the conclusion that the impact of WBV on cognitive function depends mainly on the frequency and duration of vibration, short-term low-frequency WBV may have a positive impact on cognitive function, while long term exposure to WBV may lead to cognitive decline, and the mechanisms may be involved in neuroinflammation, oxidative stress, synaptic plasticity, and neurotransmitter changes. This review may provide some theoretical foundations and guidance for the prevention and treatment of WBV induced cognitive impairment.
Collapse
Affiliation(s)
- Huan Yang
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Jie Gao
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Hai-Yan Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Xin-Mei Ma
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Bing-Yao Liu
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Qian-Zhong Song
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Hui Cheng
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Sen Li
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Zai-Yun Long
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Xiu-Min Lu
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China.
| | - Yong-Tang Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing 400042, China.
| |
Collapse
|
10
|
Mao D, Chen Q, Tong S, Xu Z, Yu G, Chang C, Lv Y. Integrated bioinformatics analysis identified cuproptosis-related hub gene Mpeg1 as potential biomarker in spinal cord injury. Sci Rep 2025; 15:1993. [PMID: 39814871 PMCID: PMC11736097 DOI: 10.1038/s41598-025-86170-0] [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: 09/04/2024] [Accepted: 01/08/2025] [Indexed: 01/18/2025] Open
Abstract
Spinal cord injury (SCI) is a profound ailment lacking a well-defined molecular mechanism and effective treatments. Cuproptosis, identified as a recently discovered cell death pathway, exhibits diverse roles in various cancers. Nevertheless, its involvement in SCI is yet to be elucidated. Firstly, the RNA sequencing data of 1, 3, 7 dpi SCI samples were collected from GEO database. We performed differential expression analysis on these samples with varying cuproptosis-related scores calculating by ssGSEA. Subsequently, we conducted enrichment analyses with KEGG, GO, and GSEA. Simultaneously, we executed WGCNA analysis using cuproptosis-related scores, selecting the most relevant module for enrichment analysis. Hub genes were identified at the intersection of PPI analysis results from two modules and cuproptosis-related DEGs. Additionally, relying on the immune infiltration landscape associated with cuproptosis, we carried out immune cell correlation analysis on hub genes. Finally, to corroborate our earlier findings, we utilized single-cell RNA-seq analysis and in vitro experimental validation. Based on ssGSEA, differential expression analysis and WGCNA analysis, we identified two modules that were highly relevant to cell division and immune processes, respectively. From these modules, we identified two hub genes, Cd48 and Mpeg1, which exhibited a strong positive correlation (R = 0.92) and shared similar pathways. Furthermore, we observed a positive correlation between M2 macrophages and Cd48/Mpeg1. To validate our findings, we performed external cohort validation using a single-cell RNA sequencing dataset. The results confirmed that Mpeg1 was highly expressed in microglia (macrophages in center nervous system) following spinal cord injury. Additionally, we conducted in vitro experiments to further validate the molecular functions of Mpeg1 in SCI. In summary, targeting Mpeg1, as well as cuproptosis and immune cell infiltration, holds promise as a potential strategy for reducing spinal cord tissue damage and promoting recovery after SCI. These findings provide valuable insights for future therapeutic interventions.
Collapse
Affiliation(s)
- Dandan Mao
- Department of Neurosurgery, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Qi Chen
- Department of Nursing, Huashan Hospital, Fudan University, Shanghai, China
| | - Shuolan Tong
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, China
| | - Zixia Xu
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, China
| | - Guofeng Yu
- Department of Neurosurgery, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Chuan Chang
- Department of Neurosurgery, Huashan hospital, Fudan University, Shanghai, China.
| | - Yao Lv
- Department of Neurosurgery, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China.
| |
Collapse
|
11
|
Weidner N, Abel R, Maier D, Röhl K, Röhrich F, Baumberger M, Hund-Georgiadis M, Saur M, Benito J, Rehahn K, Aach M, Badke A, Kriz J, Barkovits K, Killeen T, Farner L, Seif M, Hubli M, Marcus K, Maurer MA, Robert B, Rupp R, Scheuren PS, Schubert M, Schuld C, Sina C, Steiner B, Weis T, Hug A, Bolliger M, Weiskopf N, Freund P, Hothorn T, Schwab ME, Curt A. Safety and efficacy of intrathecal antibodies to Nogo-A in patients with acute cervical spinal cord injury: a randomised, double-blind, multicentre, placebo-controlled, phase 2b trial. Lancet Neurol 2025; 24:42-53. [PMID: 39706632 DOI: 10.1016/s1474-4422(24)00447-2] [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: 07/01/2024] [Revised: 10/29/2024] [Accepted: 10/31/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND Spinal cord injury results in permanent neurological impairment and disability due to the absence of spontaneous regeneration. NG101, a recombinant human antibody, neutralises the neurite growth-inhibiting protein Nogo-A, promoting neural repair and motor recovery in animal models of spinal cord injury. We aimed to evaluate the efficacy of intrathecal NG101 on recovery in patients with acute cervical traumatic spinal cord injury. METHODS This randomised, double-blind, placebo-controlled phase 2b clinical trial was done at 13 hospitals in the Czech Republic, Germany, Spain, and Switzerland. Patients aged 18-70 years with acute, complete or incomplete cervical spinal cord injury (neurological level of injury C1-C8) within 4-28 days of injury were eligible for inclusion. Participants were initially randomly assigned 1:1 to intrathecal treatment with 45 mg NG101 or placebo (phosphate-buffered saline); 18 months into the study, the ratio was adjusted to 3:1 to achieve a final distribution of 2:1 to improve enrolment and drug exposure. Randomisation was done using a centralised, computer-based randomisation system and was stratified according to nine distinct outcome categories with a validated upper extremity motor score (UEMS) prediction model based on clinical parameters at screening. Six intrathecal injections were administered every 5 days over 4 weeks, starting within 28 days of injury. Investigators, study personnel, and study participants were masked to treatment allocation. The primary outcome was change in UEMS at 6 months, analysed alongside safety in the full analysis set. The completed trial was registered at ClinicalTrials.gov, NCT03935321. FINDINGS From May 20, 2019, to July 20, 2022, 463 patients with acute traumatic cervical spinal cord injury were screened, 334 were deemed ineligible and excluded, and 129 were randomly assigned to an intervention (80 patients in the NG101 group and 49 in the placebo group). The full analysis set comprised 78 patients from the NG101 group and 48 patients from the placebo group. 107 (85%) patients were male and 19 (15%) patients were female, with a median age of 51·5 years (IQR 30·0-60·0). Across all patients, the primary endpoint showed no significant difference between groups (with UEMS change at 6 months 1·37 [95% CI -1·44 to 4·18]; placebo group mean 19·20 [SD 11·78] at baseline and 30·91 [SD 15·49] at day 168; NG101 group mean 18·23 [SD 15·14] at baseline and 31·31 [19·54] at day 168). Treatment-related adverse events were similar between groups (nine in the NG101 group and six in the placebo group). 25 severe adverse events were reported: 18 in 11 (14%) patients in the NG101 group and seven in six (13%) patients in the placebo group. Although no treatment-related fatalities were reported in the NG101 group, one fatality not related to treatment occurred in the placebo group. Infections were the most common adverse event affecting 44 (92%) patients in the placebo group and 65 (83%) patients in the NG101 group. INTERPRETATION NG101 did not improve UEMS in patients with acute spinal cord injury. Post-hoc subgroup analyses assessing UEMS and Spinal Cord Independence Measure of self-care in patients with motor-incomplete injury indicated potential beneficial effects that require investigation in future studies. FUNDING EU program Horizon2020; Swiss State Secretariat for Education, Research and Innovation; Wings for Life; the Swiss Paraplegic Foundation; and the CeNeReg project of Wyss Zurich (University of Zurich and Eidgenössische Technische Hochschule Zurich).
Collapse
Affiliation(s)
- Norbert Weidner
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany.
| | - Rainer Abel
- Clinic for Paraplegia, Klinikum Bayreuth GmbH, Bayreuth, Germany
| | - Doris Maier
- Spinal Cord Injuries, Berufsgenossenschaftliche Unfallklinik Murnau, Murnau, Germany
| | - Klaus Röhl
- BG Klinikum Bergmannstrost Halle gGmbH, Zentrum für Rückenmarkverletzte und Klinik für Orthopädie, Halle, Germany
| | - Frank Röhrich
- BG Klinikum Bergmannstrost Halle gGmbH, Zentrum für Rückenmarkverletzte und Klinik für Orthopädie, Halle, Germany
| | | | | | - Marion Saur
- Orthopädische Klinik, Hessisch Lichtenau, Germany
| | - Jesús Benito
- Fundacio Institut d'Investigacio en Ciencies de la Salut Germans Trias i Pujol, Barcelona, Spain
| | - Kerstin Rehahn
- Treatment Centre for Spinal Cord Injuries, Trauma Hospital Berlin, Berlin, Germany
| | - Mirko Aach
- Department of Spinal Cord Injuries, BG University Hospital Bergmannsheil, Bochum, Germany
| | | | - Jiri Kriz
- Spinal Cord Unit, Department of Rehabilitation and Sports Medicine, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Katalin Barkovits
- Medical Proteome Analysis, Center for Proteindiagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
| | - Tim Killeen
- Spinal Cord Injury Center, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - Lynn Farner
- Spinal Cord Injury Center, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - Maryam Seif
- Spinal Cord Injury Center, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - Michèle Hubli
- Spinal Cord Injury Center, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - Katrin Marcus
- Medical Proteome Analysis, Center for Proteindiagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
| | - Michael A Maurer
- Institute for Regenerative Medicine, University of Zurich, Switzerland
| | - Bérénice Robert
- Clinical Study Coordination Center, University of Heidelberg, Heidelberg, Germany
| | - Rüdiger Rupp
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Paulina S Scheuren
- Spinal Cord Injury Center, Balgrist University Hospital, University of Zurich, Zurich, Switzerland; International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, BC, Canada
| | - Martin Schubert
- Spinal Cord Injury Center, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - Christian Schuld
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Christina Sina
- Wyss Zurich Translational Center, University and ETH Zurich, Zurich, Switzerland
| | - Bettina Steiner
- Wyss Zurich Translational Center, University and ETH Zurich, Zurich, Switzerland
| | - Tanja Weis
- Department of Cardiology, Angiology and Pneumonology, Heidelberg University Hospital, Heidelberg, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, Heidelberg/Mannheim, Germany
| | - Andreas Hug
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Marc Bolliger
- Spinal Cord Injury Center, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - Nikolaus Weiskopf
- Department of Neurophysics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany; Felix Bloch Institute for Solid State Physics, Faculty of Physics and Earth Sciences, Leipzig University, Leipzig, Germany; Wellcome Centre for Human Neuroimaging, Institute of Neurology, University College London, London, UK
| | - Patrick Freund
- Spinal Cord Injury Center, Balgrist University Hospital, University of Zurich, Zurich, Switzerland; Wellcome Centre for Human Neuroimaging, Institute of Neurology, University College London, London, UK
| | - Torsten Hothorn
- Epidemiology, Biostatistics, and Prevention Institute, University of Zurich, Switzerland
| | - Martin E Schwab
- Institute for Regenerative Medicine, University of Zurich, Switzerland; Wyss Zurich Translational Center, University and ETH Zurich, Zurich, Switzerland
| | - Armin Curt
- Spinal Cord Injury Center, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| |
Collapse
|
12
|
Wang HY, Lin ZY, Lei JF, Ouyang JY, Lu Y, Zhuang YM, Liu XN, Yang L, Zhao H. Trillium tschonoskii rhizome saponin improves spatial learning and memory by enhancing neurovascular restorative in ischemic rats. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156096. [PMID: 39396404 DOI: 10.1016/j.phymed.2024.156096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/26/2024] [Accepted: 09/26/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND Trillium tschonoskii rhizome saponins (TSTT) has been significantly effective in treating traumatic injury, neurasthenia, cancer and inflammatory diseases as a folk medicine. However, the mechanism regarding to TSTT induced the neurovascular restorative after ischemia is without fully elucidated. PURPOSE This research was constructed to study the value of TSTT in promoting endogenous repair of neurovascular and augmenting the ability of spatial study and memory retention in ischaemic rats. STUDY DESIGN The improvement of TSTT on cerebral infraction and perfusion was observed by magnetic resonance imaging (MRI) experiments and the molecular mechanisms were further explored. METHODS First, rats were ligated the middle cerebral artery to construct a permanent ischaemia model, subsequently intragastric injection administrated with TSTT (120, 60, 30 mg kg-1) at 6 h after operation, then once a day during next 30 days. Morris water maze was applied to observe the neurobehavioral changes. Multimodal MRI sequences were performed to monitoring brain injuries as well as cerebral blood flow. Histopathological staining was employed to evaluate the morphological changes of neurons. Transmission electron microscopy (TEM) was employed to detect the neurons, vascular structure, and synapse. Immunofluorescent staining was utilized to evaluate the endogenous repair progress. The axonal growth-inhibitors and axonal guidance cues were analyzed using western blotting. RESULTS Contrast to the model group, TSTT declined the infarction and elevated the parenchymal volume. Notably, treated with TSTT significantly decreased the ADC (ipsilateral/contralateral). In histopathologic examination, TSTT prominently boosted amounts of cortical and striatal nerve cells and protected ultrastructure of neurovascular unit. According with results of nuclear magnetic imaging, TSTT enhanced endogenous repair progress. Especially, TSTT treatments obviously inhibited protein levels of NogoA/NgR/RhoA/ROCK2, accompanied by increased expression of Netrin/DCC and Slit2/Robo1. CONCLUSION To sum up, our data illustrated that TSTT promoted cerebral reestablishment. The above result was in line with improving cerebral blood flow, elevated integrity of neurovascular structure, accelerating endogenous restoration and impairing the axonal growth inhibitors NogoA/NgR/RhoA/ROCK2 signaling, thereby improving poststroke learning and memory.
Collapse
Affiliation(s)
- Han-Yu Wang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Zi-Yue Lin
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Jian-Feng Lei
- Medical Imaging Laboratory of Core Facility Center, Capital Medical University, Beijing, China
| | - Jun-Yao Ouyang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Yun Lu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Yu-Ming Zhuang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China
| | - Xiao-Nan Liu
- Department of Laboratory Animal, Capital Medical University, Beijing 100069, China
| | - Le Yang
- The Second Hospital of Dalian Medical University, Dalian Medical University, #467 Zhongshan Road, Dalian 116023, China.
| | - Hui Zhao
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, China.
| |
Collapse
|
13
|
Pagella P, Lai CF, Pirenne L, Cantù C, Schwab ME, Mitsiadis TA. An unexpected role of neurite outgrowth inhibitor A as regulator of tooth enamel formation. Int J Oral Sci 2024; 16:60. [PMID: 39426966 PMCID: PMC11490607 DOI: 10.1038/s41368-024-00323-x] [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: 06/06/2024] [Revised: 09/09/2024] [Accepted: 09/09/2024] [Indexed: 10/21/2024] Open
Abstract
Neurite outgrowth inhibitor A (Nogo-A) is a major player in neural development and regeneration and the target of clinical trials aiming at promoting the regeneration of the central nervous system upon traumatic and ischemic injury. In this work, we investigated the functions of Nogo-A during tooth development to determine its role in dental physiology and pathology. Using immunohistochemistry and in situ hybridization techniques, we showed that Nogo-A is highly expressed in the developing mouse teeth and, most specifically, in the ameloblasts that are responsible for the formation of enamel. Using both Nogo-A knockout and K14-Cre;Nogo-A fl/fl transgenic mice, we showed that Nogo-A deletion in the dental epithelium leads to the formation of defective enamel. This phenotype is associated with overexpression of a set of specific genes involved in ameloblast differentiation and enamel matrix production, such as amelogenin, ameloblastin and enamelin. By characterising the interactome of Nogo-A in the dental epithelium of wild-type and mutant animals, we found that Nogo-A directly interacts with molecules important for regulating gene expression, and its deletion disturbs their cellular localisation. Furthermore, we demonstrated that inhibition of the intracellular, but not cell-surface, Nogo-A is responsible for gene expression modulation in ameloblasts. Taken together, these results reveal an unexpected function for Nogo-A in tooth enamel formation by regulating gene expression and cytodifferentiation events.
Collapse
Affiliation(s)
- Pierfrancesco Pagella
- Orofacial Development and Regeneration, Institute of Oral Biology, University of Zürich, Zürich, Switzerland
- Laboratory of Molecular Materials, Division of Biophysics and Bioengineering, Department of Physics, Chemistry, and Biology (IFM), Linköping University, Linköping, Sweden
| | - Chai Foong Lai
- Orofacial Development and Regeneration, Institute of Oral Biology, University of Zürich, Zürich, Switzerland
| | - Laurence Pirenne
- Orofacial Development and Regeneration, Institute of Oral Biology, University of Zürich, Zürich, Switzerland
| | - Claudio Cantù
- Wallenberg Centre for Molecular Medicine, Linköping University, Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Division of Molecular Medicine and Virology; Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | - Martin E Schwab
- Institute for Regenerative Medicine, University of Zürich, Zürich, Switzerland
| | - Thimios A Mitsiadis
- Orofacial Development and Regeneration, Institute of Oral Biology, University of Zürich, Zürich, Switzerland.
| |
Collapse
|
14
|
Pavon MV, Navakkode S, Sajikumar S. Nogo-A-mediated constraints on activity-dependent synaptic plasticity and associativity in rat hippocampal CA2 synapses. Hippocampus 2024; 34:491-502. [PMID: 39091158 DOI: 10.1002/hipo.23625] [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: 12/02/2023] [Revised: 06/13/2024] [Accepted: 07/15/2024] [Indexed: 08/04/2024]
Abstract
Hippocampal area CA2 has garnered attention in recent times owing to its significant involvement in social memory and distinctive plasticity characteristics. Research has revealed that the CA2 region demonstrates a remarkable resistance to plasticity, particularly in the Schaffer Collateral (SC)-CA2 pathway. In this study we investigated the role of Nogo-A, a well-known axon growth inhibitor and more recently discovered plasticity regulator, in modulating plasticity within the CA2 region. The findings demonstrate that blocking Nogo-A in male rat hippocampal slices facilitates the establishment of both short-term and long-term plasticity in the SC-CA2 pathway, while having no impact on the Entorhinal Cortical (EC)-CA2 pathway. Additionally, the study reveals that inhibiting Nogo-A enables association between the SC and EC pathways. Mechanistically, we confirm that Nogo-A operates through its well-known co-receptor, p75 neurotrophin receptor (p75NTR), and its downstream signaling factor such as Rho-associated protein kinase (ROCK), as their inhibition also allows plasticity induction in the SC-CA2 pathway. Additionally, the induction of long-term depression (LTD) in both the EC and SC-CA2 pathways led to persistent LTD, which was not affected by Nogo-A inhibition. Our study demonstrates the involvement of Nogo-A mediated signaling mechanisms in limiting synaptic plasticity within the CA2 region.
Collapse
Affiliation(s)
- Maria Vazquez Pavon
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Neurobiology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Sheeja Navakkode
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sreedharan Sajikumar
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Neurobiology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
15
|
Hao W, Luo D, Jiang Y, Wan S, Li X. An overview of sphingosine-1-phosphate receptor 2: Structure, biological function, and small-molecule modulators. Med Res Rev 2024; 44:2331-2362. [PMID: 38665010 DOI: 10.1002/med.22044] [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: 01/03/2024] [Revised: 04/02/2024] [Accepted: 04/14/2024] [Indexed: 08/09/2024]
Abstract
Over the past decade, there has been a notable increase in research on sphingosine-1-phosphate receptor 2 (S1PR2), which is a type of G-protein-coupled receptor. Upon activation by S1P or other ligands, S1PR2 initiates downstream signaling pathways such as phosphoinositide 3-kinase (PI3K), Mitogen-activated protein kinase (MAPK), Rho/Rho-associated coiled-coil containing kinases (ROCK), and others, contributing to the diverse biological functions of S1PR2 and playing a pivotal role in various physiological processes and disease progressions, such as multiple sclerosis, fibrosis, inflammation, and tumors. Due to the extensive biological functions of S1PR2, many S1PR2 modulators, including agonists and antagonists, have been developed and discovered by pharmaceutical companies (e.g., Novartis and Galapagos NV) and academic medicinal chemists for disease diagnosis and treatment. However, few reviews have been published that comprehensively overview the functions and regulators of S1PR2. Herein, we provide an in-depth review of the advances in the function of S1PR2 and its modulators. We first summarize the structure and biological function of S1PR2 and its pathological role in human diseases. We then focus on the discovery approach, design strategy, development process, and biomedical application of S1PR2 modulators. Additionally, we outline the major challenges and future directions in this field. Our comprehensive review will aid in the discovery and development of more effective and clinically applicable S1PR2 modulators.
Collapse
Affiliation(s)
- Wanting Hao
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Dongdong Luo
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Yuqi Jiang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Shengbiao Wan
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Xiaoyang Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- Marine Biomedical Research, Institute of Qingdao, Qingdao, China
| |
Collapse
|
16
|
Dong X, Xiong YT, He T, Zheng C, Li J, Zhuang Y, Xu Y, Xiu Y, Wu Z, Zhao X, Xiao X, Bai Z, Gao L. Protective effects of Nogo-B deficiency in NAFLD mice and its multiomics analysis of gut microbiology and metabolism. GENES & NUTRITION 2024; 19:17. [PMID: 39182019 PMCID: PMC11344411 DOI: 10.1186/s12263-024-00754-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 08/11/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is a prevalent chronic liver ailment that can lead to serious conditions such as cirrhosis and hepatocellular carcinoma. Hepatic Nogo-B regulates glucose and lipid metabolism, and its inhibition has been shown to be protective against metabolic syndrome. Increasing evidence suggests that imbalances in the gut microbiota (GM) and lipid metabolism disorders are significant contributors to NAFLD progression. Nevertheless, it is not yet known whether Nogo-B can affect NAFLD by influencing the gut microbiota and metabolites. Hence, the aim of the present study was to characterize this process and explore its possible underlying mechanisms. METHODS A NAFLD model was constructed by administering a high-fat diet (HFD) to Nogo-B-/- and WT mice from the same litter, and body weight was measured weekly in each group. The glucose tolerance test (GTT) and insulin tolerance test (ITT) were performed to assess blood glucose levels. At the end of the 12-week period, samples of serum, liver, and intestinal contents were collected and used for serum biochemical marker and inflammatory factor detection; pathology evaluation; and gut microbiome and metabolomics analysis. Spearman's correlation analysis was performed to determine possible correlations between differential gut microbiota and differential serum metabolites between groups. RESULTS Nogo-B deficiency attenuated the effects of the HFD, including weight gain, liver weight gain, impaired glucose tolerance, hepatic steatosis, elevated serum lipid biochemicals levels, and liver function. Nogo-B deficiency suppressed M1 polarization and promoted M2 polarization, thus inhibiting inflammatory responses. Furthermore, Nogo-B-/--HFD-fed mice presented increased gut microbiota richness and diversity, decreased Firmicutes/Bacteroidota (F/B) ratios, and altered serum metabolites compared with those of WT-HFD-fed mice. During analysis, several differential gut microbiota, including Lachnoclostridium, Harryflintia, Odoribacter, UCG-009, and unclassified_f_Butyricoccaceae, were screened between groups. These microbiota were found to be positively correlated with upregulated purine metabolism and bile acid metabolites in Nogo-B deficiency, while they were negatively correlated with downregulated corticosterone and tricarboxylic acid cyclic metabolites in Nogo-B deficiency. CONCLUSION Nogo-B deficiency delayed NAFLD progression, as demonstrated by reduced hepatocellular lipid accumulation, attenuated inflammation and liver injury, and ameliorated gut microbiota dysbiosis and metabolic disorders. Importantly, Odoribacter was strongly positively correlated with ALB and taurodeoxycholic acid, suggesting that it played a considerable role in the influence of Nogo-B on the progression of NAFLD, a specific feature of NAFLD in Nogo-B-/- mice. The regulation of bile acid metabolism by the gut microbiota may be a potential target for Nogo-B deficiency to ameliorate NAFLD.
Collapse
Affiliation(s)
- Xu Dong
- Medical School of Chinese PLA, Beijing, China
- Department of Gastroenterology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
- Department of Hepatology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yu-Ting Xiong
- 307 Clinical Medical College of PLA, Anhui Medical University, Beijing, China
| | - Tingting He
- Department of Hepatology Medicine of Traditional Chinese Medicine, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Congyang Zheng
- Medical School of Chinese PLA, Beijing, China
- Department of Gastroenterology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
- Department of Hepatology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Junjie Li
- Department of Hepatology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
- School of Chengde Medical University, Chengdeshi, China
| | - Yingjie Zhuang
- Department of Gastroenterology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yingjie Xu
- Department of Hepatology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Ye Xiu
- Department of Hepatology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhixin Wu
- Department of Hepatology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xiaomei Zhao
- Department of Hepatology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xiaohe Xiao
- Department of Hepatology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China.
- China Military Institute of Chinese Materia, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China.
| | - Zhaofang Bai
- Department of Hepatology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China.
- China Military Institute of Chinese Materia, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China.
| | - Lili Gao
- Medical School of Chinese PLA, Beijing, China.
- Department of Gastroenterology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
17
|
Kurihara Y, Kawaguchi Y, Ohta Y, Kawasaki N, Fujita Y, Takei K. Nogo Receptor Antagonist LOTUS Promotes Neurite Outgrowth through Its Interaction with Teneurin-4. Cells 2024; 13:1369. [PMID: 39195260 PMCID: PMC11352776 DOI: 10.3390/cells13161369] [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: 07/08/2024] [Revised: 08/02/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024] Open
Abstract
Neurite outgrowth is a crucial process for organizing neuronal circuits in neuronal development and regeneration after injury. Regenerative failure in the adult mammalian central nervous system (CNS) is attributed to axonal growth inhibitors such as the Nogo protein that commonly binds to Nogo receptor-1 (NgR1). We previously reported that lateral olfactory tract usher substance (LOTUS) functions as an endogenous antagonist for NgR1 in forming neuronal circuits in the developing brain and improving axonal regeneration in the adult injured CNS. However, another molecular and cellular function of LOTUS remains unknown. In this study, we found that cultured retinal explant neurons extend their neurites on the LOTUS-coating substrate. This action was also observed in cultured retinal explant neurons derived from Ngr1-deficient mouse embryos, indicating that the promoting action of LOTUS on neurite outgrowth may be mediated by unidentified LOTUS-binding protein(s). We therefore screened the binding partner(s) of LOTUS by using a liquid chromatography-tandem mass spectrometry (LC-MS/MS). LC-MS/MS analysis and pull-down assay showed that LOTUS interacts with Teneurin-4 (Ten-4), a cell adhesion molecule. RNAi knockdown of Ten-4 inhibited neurite outgrowth on the LOTUS substrate in retinoic acid (RA)-treated Neuro2A cells. Furthermore, a soluble form of Ten-4 attenuates the promoting action on neurite outgrowth in cultured retinal explant neurons on the LOTUS substrate. These results suggest that LOTUS promotes neurite outgrowth by interacting with Ten-4. Our findings may provide a new molecular mechanism of LOTUS to contribute to neuronal circuit formation in development and to enhance axonal regeneration after CNS injury.
Collapse
Affiliation(s)
- Yuji Kurihara
- Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Yokohama 230-0045, Japan
- Department of Anatomy & Developmental Biology, Faculty of Medicine, Shimane University, Izumo 693-8501, Japan
| | - Yuki Kawaguchi
- Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Yokohama 230-0045, Japan
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama 230-0045, Japan
| | - Yuki Ohta
- Laboratory of Biopharmaceutical and Regenerative Sciences, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Yokohama 230-0045, Japan
| | - Nana Kawasaki
- Laboratory of Biopharmaceutical and Regenerative Sciences, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Yokohama 230-0045, Japan
| | - Yuki Fujita
- Department of Anatomy & Developmental Biology, Faculty of Medicine, Shimane University, Izumo 693-8501, Japan
| | - Kohtaro Takei
- Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Yokohama 230-0045, Japan
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Yokohama 230-0045, Japan
| |
Collapse
|
18
|
Lu Y, Lin Z, Li M, Zhuang Y, Nie B, Lei J, Zhao Y, Zhao H. Three-phase Enriched Environment Improves Post-stroke Gait Dysfunction via Facilitating Neuronal Plasticity in the Bilateral Sensorimotor Cortex: A Multimodal MRI/PET Analysis in Rats. Neurosci Bull 2024; 40:719-731. [PMID: 38055107 PMCID: PMC11178725 DOI: 10.1007/s12264-023-01155-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 08/22/2023] [Indexed: 12/07/2023] Open
Abstract
The three-phase Enriched Environment (EE) paradigm has been shown to promote post-stroke functional improvement, but the neuronal mechanisms are still unclear. In this study, we applied a multimodal neuroimaging protocol combining magnetic resonance imaging (MRI) and positron emission tomography (PET) to examine the effects of post-ischemic EE treatment on structural and functional neuroplasticity in the bilateral sensorimotor cortex. Rats were subjected to permanent middle cerebral artery occlusion. The motor function of the rats was examined using the DigiGait test. MRI was applied to investigate the EE-induced structural modifications of the bilateral sensorimotor cortex. [18F]-fluorodeoxyglucose PET was used to detect glucose metabolism. Blood oxygen level-dependent (BOLD)-functional MRI (fMRI) was used to identify the regional brain activity and functional connectivity (FC). In addition, the expression of neuroplasticity-related signaling pathways including neurotrophic factors (BDNF/CREB), axonal guidance proteins (Robo1/Slit2), and axonal growth-inhibitory proteins (NogoA/NgR) as well as downstream proteins (RhoA/ROCK) in the bilateral sensorimotor cortex were measured by Western blots. Our results showed the three-phase EE improved the walking ability. Structural T2 mapping imaging and diffusion tensor imaging demonstrated that EE benefited structure integrity in the bilateral sensorimotor cortex. PET-MRI fused images showed improved glucose metabolism in the corresponding regions after EE intervention. Specifically, the BOLD-based amplitude of low-frequency fluctuations showed that EE increased spontaneous activity in the bilateral motor cortex and ipsilateral sensory cortex. In addition, FC results showed increased sensorimotor connectivity in the ipsilateral hemisphere and increased interhemispheric motor cortical connectivity and motor cortical-thalamic connectivity following EE intervention. In addition, a strong correlation was found between increased functional connectivity and improved motor performance of limbs. Specifically, EE regulated the expression of neuroplasticity-related signaling, involving BDNF/CREB, Slit2/Robo1, as well as the axonal growth-inhibitory pathways Nogo-A/Nogo receptor and RhoA/ROCK in the bilateral sensorimotor cortex. Our results indicated that the three-phase enriched environment paradigm enhances neuronal plasticity of the bilateral sensorimotor cortex and consequently ameliorates post-stroke gait deficits. These findings might provide some new clues for the development of EE and thus facilitate the clinical translation of EE.
Collapse
Affiliation(s)
- Yun Lu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
- Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, 100069, China
| | - Ziyue Lin
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
- Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, 100069, China
| | - Mingcong Li
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
- Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, 100069, China
| | - Yuming Zhuang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
- Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, 100069, China
| | - Binbin Nie
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Jianfeng Lei
- Medical Imaging Laboratory of Core Facility Center, Capital Medical University, Beijing, 100069, China
| | - Yuanyuan Zhao
- Medical Imaging Laboratory of Core Facility Center, Capital Medical University, Beijing, 100069, China
| | - Hui Zhao
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China.
- Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, 100069, China.
| |
Collapse
|
19
|
Rust R, Holm MM, Egger M, Weinmann O, van Rossum D, Walter FR, Santa-Maria AR, Grönnert L, Maurer MA, Kraler S, Akhmedov A, Cideciyan R, Lüscher TF, Deli MA, Herrmann IK, Schwab ME. Nogo-A is secreted in extracellular vesicles, occurs in blood and can influence vascular permeability. J Cereb Blood Flow Metab 2024; 44:938-954. [PMID: 38000040 PMCID: PMC11318402 DOI: 10.1177/0271678x231216270] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 10/10/2023] [Accepted: 10/20/2023] [Indexed: 11/26/2023]
Abstract
Nogo-A is a transmembrane protein with multiple functions in the central nervous system (CNS), including restriction of neurite growth and synaptic plasticity. Thus far, Nogo-A has been predominantly considered a cell contact-dependent ligand signaling via cell surface receptors. Here, we show that Nogo-A can be secreted by cultured cells of neuronal and glial origin in association with extracellular vesicles (EVs). Neuron- and oligodendrocyte-derived Nogo-A containing EVs inhibited fibroblast spreading, and this effect was partially reversed by Nogo-A receptor S1PR2 blockage. EVs purified from HEK cells only inhibited fibroblast spreading upon Nogo-A over-expression. Nogo-A-containing EVs were found in vivo in the blood of healthy mice and rats, as well as in human plasma. Blood Nogo-A concentrations were elevated after acute stroke lesions in mice and rats. Nogo-A active peptides decreased barrier integrity in an in vitro blood-brain barrier model. Stroked mice showed increased dye permeability in peripheral organs when tested 2 weeks after injury. In the Miles assay, an in vivo test to assess leakage of the skin vasculature, a Nogo-A active peptide increased dye permeability. These findings suggest that blood borne, possibly EV-associated Nogo-A could exert long-range regulatory actions on vascular permeability.
Collapse
Affiliation(s)
- Ruslan Rust
- Brain Research Institute, University of Zürich, Switzerland
- Department of Health Sciences and Technology, ETH Zürich, Switzerland
- Institute for Regenerative Medicine (IREM), University of Zurich, Switzerland
| | - Mea M Holm
- Brain Research Institute, University of Zürich, Switzerland
- Department of Health Sciences and Technology, ETH Zürich, Switzerland
| | - Matteo Egger
- Department of Health Sciences and Technology, ETH Zürich, Switzerland
| | | | | | - Fruzsina R Walter
- Biological Barriers Research Group, ELKH Biological Research Centre, Szeged, Hungary
| | | | - Lisa Grönnert
- Brain Research Institute, University of Zürich, Switzerland
| | | | - Simon Kraler
- Center for Molecular Cardiology, University of Zurich, Switzerland
| | | | - Rose Cideciyan
- Center for Molecular Cardiology, University of Zurich, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Switzerland
- Royal Brompton and Harefield Hospitals and Imperial College, London, United Kingdom
| | - Maria A Deli
- Biological Barriers Research Group, ELKH Biological Research Centre, Szeged, Hungary
| | - Inge K Herrmann
- Particles Biology Interactions Laboratory, Swiss Federal Laboratories for Materials Science and Technology (Empa), St. Gallen, Switzerland
- Nanoparticle Systems Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland
| | - Martin E Schwab
- Brain Research Institute, University of Zürich, Switzerland
- Department of Health Sciences and Technology, ETH Zürich, Switzerland
- Institute for Regenerative Medicine (IREM), University of Zurich, Switzerland
| |
Collapse
|
20
|
Chen T, Chen Z, Wu P, Luo J, Liu Q, Yang H, Peng C, Zhang G, Lin H, Ji Z. The Interaction between ADK and SCG10 Regulate the Repair of Nerve Damage. Neuroscience 2024; 544:75-87. [PMID: 38423163 DOI: 10.1016/j.neuroscience.2024.02.023] [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: 07/24/2023] [Revised: 02/22/2024] [Accepted: 02/23/2024] [Indexed: 03/02/2024]
Abstract
The cytoskeleton must be remodeled during neurite outgrowth, and Superior Cervical Ganglion 10 (SCG10) plays a critical role in this process by depolymerizing Microtubules (MTs), conferring highly dynamic properties to the MTs. However, the precise mechanism of action of SCG10 in the repair of injured neurons remains largely uncertain. Using transcriptomic identification, we discovered that SCG10 expression was downregulated in neurons after Spinal Cord Injury (SCI). Additionally, through mass spectrometry identification, immunoprecipitation, and pull-down assays, we established that SCG10 could interact with Adenosine Kinase (ADK). Furthermore, we developed an excitotoxicity-induced neural injury model and discovered that ADK suppressed injured neurite re-growth, whereas, through overexpression and small molecule interference experiments, SCG10 enhanced it. Moreover, we discovered ADK to be the upstream of SCG10. More importantly, the application of the ADK inhibitor called 5-Iodotubercidin (5-ITu) was found to significantly enhance the recovery of motor function in mice with SCI. Consequently, our findings suggest that ADK plays a negative regulatory role in the repair of injured neurons. Herein, we propose a molecular interaction model of the SCG10-ADK axis to regulate neuronal recovery.
Collapse
Affiliation(s)
- Tianjun Chen
- Department of Orthopedics, The First Affiliated Hospital of Jinan University, Guangdong Province, Guangzhou 510630, People's Republic of China
| | - Zhiwan Chen
- Department of Orthopedics, The First Affiliated Hospital of Jinan University, Guangdong Province, Guangzhou 510630, People's Republic of China
| | - Ping Wu
- Department of Orthopedics, The First Affiliated Hospital of Jinan University, Guangdong Province, Guangzhou 510630, People's Republic of China
| | - Jianxian Luo
- Department of Orthopedics, The First Affiliated Hospital of Jinan University, Guangdong Province, Guangzhou 510630, People's Republic of China
| | - Qiuling Liu
- Department of Orthopedics, The First Affiliated Hospital of Jinan University, Guangdong Province, Guangzhou 510630, People's Republic of China
| | - Hua Yang
- Department of Orthopedics, The First Affiliated Hospital of Jinan University, Guangdong Province, Guangzhou 510630, People's Republic of China
| | - Cheng Peng
- Department of Orthopedics, The First Affiliated Hospital of Jinan University, Guangdong Province, Guangzhou 510630, People's Republic of China
| | - Guowei Zhang
- Department of Orthopedics, The First Affiliated Hospital of Jinan University, Guangdong Province, Guangzhou 510630, People's Republic of China
| | - Hongsheng Lin
- Department of Orthopedics, The First Affiliated Hospital of Jinan University, Guangdong Province, Guangzhou 510630, People's Republic of China
| | - Zhisheng Ji
- Department of Orthopedics, The First Affiliated Hospital of Jinan University, Guangdong Province, Guangzhou 510630, People's Republic of China.
| |
Collapse
|
21
|
Luppi AI, Rosas FE, Noonan MP, Mediano PAM, Kringelbach ML, Carhart-Harris RL, Stamatakis EA, Vernon AC, Turkheimer FE. Oxygen and the Spark of Human Brain Evolution: Complex Interactions of Metabolism and Cortical Expansion across Development and Evolution. Neuroscientist 2024; 30:173-198. [PMID: 36476177 DOI: 10.1177/10738584221138032] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Scientific theories on the functioning and dysfunction of the human brain require an understanding of its development-before and after birth and through maturation to adulthood-and its evolution. Here we bring together several accounts of human brain evolution by focusing on the central role of oxygen and brain metabolism. We argue that evolutionary expansion of human transmodal association cortices exceeded the capacity of oxygen delivery by the vascular system, which led these brain tissues to rely on nonoxidative glycolysis for additional energy supply. We draw a link between the resulting lower oxygen tension and its effect on cytoarchitecture, which we posit as a key driver of genetic developmental programs for the human brain-favoring lower intracortical myelination and the presence of biosynthetic materials for synapse turnover. Across biological and temporal scales, this protracted capacity for neural plasticity sets the conditions for cognitive flexibility and ongoing learning, supporting complex group dynamics and intergenerational learning that in turn enabled improved nutrition to fuel the metabolic costs of further cortical expansion. Our proposed model delineates explicit mechanistic links among metabolism, molecular and cellular brain heterogeneity, and behavior, which may lead toward a clearer understanding of brain development and its disorders.
Collapse
Affiliation(s)
- Andrea I Luppi
- Department of Clinical Neurosciences and Division of Anaesthesia, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Leverhulme Centre for the Future of Intelligence, University of Cambridge, Cambridge, UK
- The Alan Turing Institute, London, UK
| | - Fernando E Rosas
- Department of Informatics, University of Sussex, Brighton, UK
- Centre for Psychedelic Research, Department of Brain Science, Imperial College London, London, UK
- Centre for Complexity Science, Imperial College London, London, UK
- Centre for Eudaimonia and Human Flourishing, University of Oxford, Oxford, UK
| | - MaryAnn P Noonan
- Department of Experimental Psychology, University of Oxford, Oxford, UK
| | - Pedro A M Mediano
- Department of Psychology, University of Cambridge, Cambridge, UK
- Department of Psychology, Queen Mary University of London, London, UK
- Department of Computing, Imperial College London, London, UK
| | - Morten L Kringelbach
- Centre for Eudaimonia and Human Flourishing, University of Oxford, Oxford, UK
- Center for Music in the Brain, Aarhus University, Aarhus, Denmark
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Robin L Carhart-Harris
- Psychedelics Division-Neuroscape, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Emmanuel A Stamatakis
- Department of Clinical Neurosciences and Division of Anaesthesia, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Anthony C Vernon
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Federico E Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
22
|
Wang XW, Yang SG, Hu MW, Wang RY, Zhang C, Kosanam AR, Ochuba AJ, Jiang JJ, Luo X, Guan Y, Qian J, Liu CM, Zhou FQ. Histone methyltransferase Ezh2 coordinates mammalian axon regeneration via regulation of key regenerative pathways. J Clin Invest 2024; 134:e163145. [PMID: 38015636 PMCID: PMC10849760 DOI: 10.1172/jci163145] [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: 06/29/2022] [Accepted: 11/21/2023] [Indexed: 11/30/2023] Open
Abstract
Current treatments for neurodegenerative diseases and neural injuries face major challenges, primarily due to the diminished regenerative capacity of neurons in the mammalian CNS as they mature. Here, we investigated the role of Ezh2, a histone methyltransferase, in regulating mammalian axon regeneration. We found that Ezh2 declined in the mouse nervous system during maturation but was upregulated in adult dorsal root ganglion neurons following peripheral nerve injury to facilitate spontaneous axon regeneration. In addition, overexpression of Ezh2 in retinal ganglion cells in the CNS promoted optic nerve regeneration via both histone methylation-dependent and -independent mechanisms. Further investigation revealed that Ezh2 fostered axon regeneration by orchestrating the transcriptional silencing of genes governing synaptic function and those inhibiting axon regeneration, while concurrently activating various factors that support axon regeneration. Notably, we demonstrated that GABA transporter 2, encoded by Slc6a13, acted downstream of Ezh2 to control axon regeneration. Overall, our study underscores the potential of modulating chromatin accessibility as a promising strategy for promoting CNS axon regeneration.
Collapse
Affiliation(s)
- Xue-Wei Wang
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, Florida, USA
| | - Shu-Guang Yang
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Rui-Ying Wang
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Chi Zhang
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Anish R. Kosanam
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Arinze J. Ochuba
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jing-Jing Jiang
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Yun Guan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Chang-Mei Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Feng-Quan Zhou
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
23
|
Yang R, Zhang Y, Kang J, Zhang C, Ning B. Chondroitin Sulfate Proteoglycans Revisited: Its Mechanism of Generation and Action for Spinal Cord Injury. Aging Dis 2024; 15:153-168. [PMID: 37307832 PMCID: PMC10796098 DOI: 10.14336/ad.2023.0512] [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: 02/12/2023] [Accepted: 05/12/2023] [Indexed: 06/14/2023] Open
Abstract
Reactive astrocytes (RAs) produce chondroitin sulfate proteoglycans (CSPGs) in large quantities after spinal cord injury (SCI) and inhibit axon regeneration through the Rho-associated protein kinase (ROCK) pathway. However, the mechanism of producing CSPGs by RAs and their roles in other aspects are often overlooked. In recent years, novel generation mechanisms and functions of CSPGs have gradually emerged. Extracellular traps (ETs), a new recently discovered phenomenon in SCI, can promote secondary injury. ETs are released by neutrophils and microglia, which activate astrocytes to produce CSPGs after SCI. CSPGs inhibit axon regeneration and play an important role in regulating inflammation as well as cell migration and differentiation; some of these regulations are beneficial. The current review summarized the process of ET-activated RAs to generate CSPGs at the cellular signaling pathway level. Moreover, the roles of CSPGs in inhibiting axon regeneration, regulating inflammation, and regulating cell migration and differentiation were discussed. Finally, based on the above process, novel potential therapeutic targets were proposed to eliminate the adverse effects of CSPGs.
Collapse
Affiliation(s)
- Rui Yang
- Jinan Central Hospital, Shandong University, Jinan, Shandong, China.
| | - Ying Zhang
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jianning Kang
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Ce Zhang
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Bin Ning
- Jinan Central Hospital, Shandong University, Jinan, Shandong, China.
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
24
|
Zhu Y, Hui Q, Zhang Z, Fu H, Qin Y, Zhao Q, Li Q, Zhang J, Guo L, He W, Han C. Advancements in the study of synaptic plasticity and mitochondrial autophagy relationship. J Neurosci Res 2024; 102:e25309. [PMID: 38400573 DOI: 10.1002/jnr.25309] [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: 10/09/2023] [Revised: 01/26/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024]
Abstract
Synapses serve as the points of communication between neurons, consisting primarily of three components: the presynaptic membrane, synaptic cleft, and postsynaptic membrane. They transmit signals through the release and reception of neurotransmitters. Synaptic plasticity, the ability of synapses to undergo structural and functional changes, is influenced by proteins such as growth-associated proteins, synaptic vesicle proteins, postsynaptic density proteins, and neurotrophic growth factors. Furthermore, maintaining synaptic plasticity consumes more than half of the brain's energy, with a significant portion of this energy originating from ATP generated through mitochondrial energy metabolism. Consequently, the quantity, distribution, transport, and function of mitochondria impact the stability of brain energy metabolism, thereby participating in the regulation of fundamental processes in synaptic plasticity, including neuronal differentiation, neurite outgrowth, synapse formation, and neurotransmitter release. This article provides a comprehensive overview of the proteins associated with presynaptic plasticity, postsynaptic plasticity, and common factors between the two, as well as the relationship between mitochondrial energy metabolism and synaptic plasticity.
Collapse
Affiliation(s)
- Yousong Zhu
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Qinlong Hui
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Zheng Zhang
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Hao Fu
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Yali Qin
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Qiong Zhao
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Qinqing Li
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Junlong Zhang
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
| | - Lei Guo
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
| | - Wenbin He
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
| | - Cheng Han
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| |
Collapse
|
25
|
Herzog H, Glöckler S, Flamm J, Ladel S, Maigler F, Pitzer C, Schindowski K. Intranasal Nose-to-Brain Drug Delivery via the Olfactory Region in Mice: Two In-Depth Protocols for Region-Specific Intranasal Application of Antibodies and for Expression Analysis of Fc Receptors via In Situ Hybridization in the Nasal Mucosa. Methods Mol Biol 2024; 2754:387-410. [PMID: 38512678 DOI: 10.1007/978-1-0716-3629-9_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2024]
Abstract
A region-specific catheter-based intranasal administration method was successfully developed, established, and validated as reported previously. By using this method, drugs can be applicated specifically to the olfactory region. Thereby, intranasally administered drugs could be delivered via neuronal connections to the central nervous system. Here, we present a detailed protocol with a step-by-step procedure for nose-to-brain delivery via the olfactory mucosa.Fc receptors such as the neonatal Fc receptor (FcRn) and potentially Fcγ receptor IIb (FcγRIIb) are involved in the uptake and transport of antibodies via the olfactory nasal mucosa. To better characterize their expression levels and their role in CNS drug delivery via the nose, an in situ hybridization (ISH) protocol was adapted for nasal mucosa samples and described in abundant details.
Collapse
Affiliation(s)
- Helena Herzog
- Institute of Applied Biotechnology, University of Applied Science Biberach, Biberach, Germany
- Faculty of Natural Science, University of Ulm, Ulm, Germany
| | - Sara Glöckler
- Institute of Applied Biotechnology, University of Applied Science Biberach, Biberach, Germany
- Faculty of Natural Science, University of Ulm, Ulm, Germany
| | - Johannes Flamm
- Institute of Applied Biotechnology, University of Applied Science Biberach, Biberach, Germany
- Faculty of Natural Science, University of Ulm, Ulm, Germany
| | - Simone Ladel
- Institute of Applied Biotechnology, University of Applied Science Biberach, Biberach, Germany
- Faculty of Natural Science, University of Ulm, Ulm, Germany
| | - Frank Maigler
- Institute of Applied Biotechnology, University of Applied Science Biberach, Biberach, Germany
- Faculty of Natural Science, University of Ulm, Ulm, Germany
| | - Claudia Pitzer
- Interdisciplinary Neurobehavioral Core, Heidelberg University, Heidelberg, Germany
| | - Katharina Schindowski
- Institute of Applied Biotechnology, University of Applied Science Biberach, Biberach, Germany.
| |
Collapse
|
26
|
Reyes C, Mokalled MH. Astrocyte-Neuron Interactions in Spinal Cord Injury. ADVANCES IN NEUROBIOLOGY 2024; 39:213-231. [PMID: 39190077 PMCID: PMC11684398 DOI: 10.1007/978-3-031-64839-7_9] [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] [Indexed: 08/28/2024]
Abstract
Spinal cord injuries cause irreversible loss of sensory and motor functions. In mammals, intrinsic and extrinsic inhibitions of neuronal regeneration obstruct neural repair after spinal cord injury. Although astrocytes have been involved in a growing list of vital homeostatic functions in the nervous system, their roles after injury have fascinated and puzzled scientists for decades. Astrocytes undergo long-lasting morphological and functional changes after injury, referred to as reactive astrogliosis. Although reactive astrogliosis is required to contain spinal cord lesions and restore the blood-spinal cord barrier, reactive astrocytes have detrimental effects that inhibit neuronal repair and remyelination. Intriguingly, elevated regenerative capacity is preserved in some non-mammalian vertebrates, where astrocyte-like glial cells display exclusively pro-regenerative effects after injury. A detailed molecular and phenotypic catalog of the continuum of astrocyte reactivity states is an essential first step toward the development of glial cell manipulations for spinal cord repair.
Collapse
Affiliation(s)
- Catrina Reyes
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Mayssa H Mokalled
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
27
|
Hirt J, Khanteymoori A, Hohenhaus M, Kopp MA, Howells DW, Schwab JM, Watzlawick R. Inhibition of the Nogo-pathway in experimental spinal cord injury: a meta-analysis of 76 experimental treatments. Sci Rep 2023; 13:22898. [PMID: 38129508 PMCID: PMC10739940 DOI: 10.1038/s41598-023-49260-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023] Open
Abstract
Recovery after spinal cord injury (SCI) may be propagated by plasticity-enhancing treatments. The myelin-associated nerve outgrowth inhibitor Nogo-A (Reticulon 4, RTN4) pathway has been shown to restrict neuroaxonal plasticity in experimental SCI models. Early randomized controlled trials are underway to investigate the effect of Nogo-A/Nogo-Receptor (NgR1) pathway blockers. This systematic review and meta-analysis of therapeutic approaches blocking the Nogo-A pathway interrogated the efficacy of functional locomotor recovery after experimental SCI according to a pre-registered study protocol. A total of 51 manuscripts reporting 76 experiments in 1572 animals were identified for meta-analysis. Overall, a neurobehavioral improvement by 18.9% (95% CI 14.5-23.2) was observed. Subgroup analysis (40 experiments, N = 890) revealed SCI-modelling factors associated with outcome variability. Lack of reported randomization and smaller group sizes were associated with larger effect sizes. Delayed treatment start was associated with lower effect sizes. Trim and Fill assessment as well as Egger regression suggested the presence of publication bias. Factoring in theoretically missing studies resulted in a reduced effect size [8.8% (95% CI 2.6-14.9)]. The available data indicates that inhibition of the Nogo-A/NgR1pathway alters functional recovery after SCI in animal studies although substantial differences appear for the applied injury mechanisms and other study details. Mirroring other SCI interventions assessed earlier we identify similar factors associated with outcome heterogeneity.
Collapse
Affiliation(s)
- Julian Hirt
- Department of Neurology and Experimental Neurology, Charité Campus Mitte, Clinical and Experimental Spinal Cord Injury Research Laboratory (Neuroparaplegiology), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Alireza Khanteymoori
- Department of Neurosurgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Breisacher Straße 64, 79106, Freiburg, Germany
| | - Marc Hohenhaus
- Department of Neurosurgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Breisacher Straße 64, 79106, Freiburg, Germany
| | - Marcel A Kopp
- Department of Neurology and Experimental Neurology, Charité Campus Mitte, Clinical and Experimental Spinal Cord Injury Research Laboratory (Neuroparaplegiology), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - David W Howells
- Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC, Australia
| | - Jan M Schwab
- Department of Neurology and Experimental Neurology, Charité Campus Mitte, Clinical and Experimental Spinal Cord Injury Research Laboratory (Neuroparaplegiology), Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department of Neurology, Spinal Cord Injury Division (Paraplegiology), The Neurological Institute, The Ohio State University, Wexner Medical Center, Columbus, OH, USA
- Belford Center for Spinal Cord Injury, Departments of Neuroscience and Physical Medicine and Rehabilitation, The Neurological Institute, The Ohio State University, Wexner Medical Center, Columbus, OH, USA
| | - Ralf Watzlawick
- Department of Neurology and Experimental Neurology, Charité Campus Mitte, Clinical and Experimental Spinal Cord Injury Research Laboratory (Neuroparaplegiology), Charité - Universitätsmedizin Berlin, Berlin, Germany.
- Department of Neurosurgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Breisacher Straße 64, 79106, Freiburg, Germany.
| |
Collapse
|
28
|
Ikeda T, Takahashi K, Higashi M, Komiya H, Asano T, Ogasawara A, Kubota S, Hashiguchi S, Kunii M, Tanaka K, Tada M, Doi H, Takeuchi H, Takei K, Tanaka F. Lateral olfactory tract usher substance (LOTUS), an endogenous Nogo receptor antagonist, ameliorates disease progression in amyotrophic lateral sclerosis model mice. Cell Death Discov 2023; 9:454. [PMID: 38097540 PMCID: PMC10721829 DOI: 10.1038/s41420-023-01758-7] [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: 10/04/2023] [Revised: 11/22/2023] [Accepted: 11/29/2023] [Indexed: 12/17/2023] Open
Abstract
Nogo-Nogo receptor 1 (NgR1) signaling is significantly implicated in neurodegeneration in amyotrophic lateral sclerosis (ALS). We previously showed that lateral olfactory tract usher substance (LOTUS) is an endogenous antagonist of NgR1 that prevents all myelin-associated inhibitors (MAIs), including Nogo, from binding to NgR1. Here we investigated the role of LOTUS in ALS pathogenesis by analyzing G93A-mutated human superoxide dismutase 1 (SOD1) transgenic (Tg) mice, as an ALS model, as well as newly generated LOTUS-overexpressing SOD1 Tg mice. We examined expression profiles of LOTUS and MAIs and compared motor functions and survival periods in these mice. We also investigated motor neuron survival, glial proliferation in the lumbar spinal cord, and neuromuscular junction (NMJ) morphology. We analyzed downstream molecules of NgR1 signaling such as ROCK2, LIMK1, cofilin, and ataxin-2, and also neurotrophins. In addition, we investigated LOTUS protein levels in the ventral horn of ALS patients. We found significantly decreased LOTUS expression in both SOD1 Tg mice and ALS patients. LOTUS overexpression in SOD1 Tg mice increased lifespan and improved motor function, in association with prevention of motor neuron loss, reduced gliosis, increased NMJ innervation, maintenance of cofilin phosphorylation dynamics, decreased levels of ataxin-2, and increased levels of brain-derived neurotrophic factor (BDNF). Reduced LOTUS expression may enhance neurodegeneration in SOD1 Tg mice and ALS patients by activating NgR1 signaling, and in this study LOTUS overexpression significantly ameliorated ALS pathogenesis. LOTUS might serve as a promising therapeutic target for ALS.
Collapse
Affiliation(s)
- Takuya Ikeda
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Keita Takahashi
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan.
| | - Minatsu Higashi
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Hiroyasu Komiya
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Tetsuya Asano
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Akihiro Ogasawara
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Shun Kubota
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Shunta Hashiguchi
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Misako Kunii
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Kenichi Tanaka
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Mikiko Tada
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Hiroshi Doi
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Hideyuki Takeuchi
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Kohtaro Takei
- Molecular Medical Bioscience Laboratory, Yokohama City University Graduate School of Medical Life Science, Yokohama, 236-0004, Japan
| | - Fumiaki Tanaka
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan.
| |
Collapse
|
29
|
Çetin E, Sancak T, Keleş ÖF, Ünlü İ, Akyol ME, Arabacı Ö. Histopathological and immunohistochemical investigation of the effect of Shilajit in rats with experimental spinal cord injury. ULUS TRAVMA ACIL CER 2023; 29:1329-1334. [PMID: 38073457 PMCID: PMC10767290 DOI: 10.14744/tjtes.2023.60621] [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: 06/13/2023] [Revised: 10/20/2023] [Accepted: 11/03/2023] [Indexed: 12/18/2023]
Abstract
BACKGROUND This experimental study was designed to investigate the histopathological and immunohistochemical effects of Shilajit in rats with experimentally induced spinal cord injury (SCI). METHODS The rats were divided into three groups: Control group: The group in which spinal cord damage was created but no drug was administered. Low-dose group: This is the group in which intraperitoneal Shilajit is given at a dose of 150 mg/kg at the 1st h, 1st day, 2nd day, and 3rd day after spinal cord damage was induced. High-dose group: This is the group in which intraperitoneal Shilajit is given at a dose of 250 mg/kg at the 1st h, 1st day, 2nd day, and 3rd day after spinal cord damage was induced. Thin sections taken from the spinal cord after euthanasia were sent for histopathological and immunohistochemical examination. RESULTS Histopathological examination of the high-dose group showed lower amounts of morphological findings compared to the low-dose group and control group. While a significant CD68 immune reaction was observed in the control group of rats with spinal injury, the positive immune reaction was found to be significantly decreased in the Shilajit-applied groups. CONCLUSION It is thought that the use of Shilajit in SCI will reduce the effects of secondary damage in SCI and that its administra-tion to such patients will have positive effects on the results.
Collapse
Affiliation(s)
- Eyüp Çetin
- Department of Neurosurgery, Health Sciences University Haydarpaşa Numune Training and Research Hospital, İstanbul-Türkiye
| | - Tunahan Sancak
- Department of Surgery, Faculty of Veterinary Medicine, Sivas Cumhuriyet University, Sivas-Türkiye
| | - Ömer Faruk Keleş
- Department of Patology, Faculty of Veterinary Medicine, Van Yüzüncü Yıl University, Van-Türkiye
| | - İlker Ünlü
- Department of Neurosurgery, Faculty of Health Sciences, İstanbul Esenyurt University, İstanbul-Türkiye
| | - Mehmet Edip Akyol
- Department of Neurosurgery, Faculty of Medicine, Van Yüzüncü Yıl University, Van-Türkiye
| | - Özkan Arabacı
- Department of Neurosurgery, Faculty of Medicine, Van Yüzüncü Yıl University, Van-Türkiye
| |
Collapse
|
30
|
Howard EM, Strittmatter SM. Development of neural repair therapy for chronic spinal cord trauma: soluble Nogo receptor decoy from discovery to clinical trial. Curr Opin Neurol 2023; 36:516-522. [PMID: 37865850 PMCID: PMC10841037 DOI: 10.1097/wco.0000000000001205] [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] [Indexed: 10/23/2023]
Abstract
PURPOSE OF REVIEW After traumatic spinal cord injury (SCI), neurological deficits persist due to the disconnection of surviving neurons. While repair of connectivity may restore function, no medical therapy exists today.This review traces the development of the neural repair-based therapeutic AXER-204 from animal studies to the recent clinical trial for chronic cervical SCI. RECENT FINDINGS Molecular studies reveal a Nogo-66 Receptor 1 (NgR1, RTN4R) pathway inhibiting axon regeneration, sprouting, and plasticity in the adult mammalian central nervous system (CNS). Rodent and nonhuman primate studies demonstrate that the soluble receptor decoy NgR(310)ecto-Fc or AXER-204 promotes neural repair and functional recovery in transection and contusion SCI. Recently, this biological agent completed a first-in-human and randomized clinical trial for chronic cervical SCI. The intervention was safe and well tolerated. Across all participants, upper extremity strength did not improve with treatment. However, posthoc and biomarker analyses suggest that AXER-204 may benefit treatment-naïve patients with incomplete SCI in the chronic stage. SUMMARY NgR1 signaling restricts neurological recovery in animal studies of CNS injury. The recent clinical trial of AXER-204 provides encouraging signals supporting future focused trials of this neural repair therapeutic. Further, AXER-204 studies provide a roadmap for the development of additional and synergistic therapies for chronic SCI.
Collapse
Affiliation(s)
- Elisa M. Howard
- Departments of Neuroscience and Neurology, Yale School of Medicine, New Haven, CT, USA
| | | |
Collapse
|
31
|
Golan N, Ehrlich D, Bonanno J, O'Brien RF, Murillo M, Kauer SD, Ravindra N, Van Dijk D, Cafferty WB. Anatomical Diversity of the Adult Corticospinal Tract Revealed by Single-Cell Transcriptional Profiling. J Neurosci 2023; 43:7929-7945. [PMID: 37748862 PMCID: PMC10669816 DOI: 10.1523/jneurosci.0811-22.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 07/28/2023] [Accepted: 08/01/2023] [Indexed: 09/27/2023] Open
Abstract
The corticospinal tract (CST) forms a central part of the voluntary motor apparatus in all mammals. Thus, injury, disease, and subsequent degeneration within this pathway result in chronic irreversible functional deficits. Current strategies to repair the damaged CST are suboptimal in part because of underexplored molecular heterogeneity within the adult tract. Here, we combine spinal retrograde CST tracing with single-cell RNA sequencing (scRNAseq) in adult male and female mice to index corticospinal neuron (CSN) subtypes that differentially innervate the forelimb and hindlimb. We exploit publicly available datasets to confer anatomic specialization among CSNs and show that CSNs segregate not only along the forelimb and hindlimb axis but also by supraspinal axon collateralization. These anatomically defined transcriptional data allow us to use machine learning tools to build classifiers that discriminate between CSNs and cortical layer 2/3 and nonspinally terminating layer 5 neurons in M1 and separately identify limb-specific CSNs. Using these tools, CSN subtypes can be differentially identified to study postnatal patterning of the CST in vivo, leveraged to screen for novel limb-specific axon growth survival and growth activators in vitro, and ultimately exploited to repair the damaged CST after injury and disease.SIGNIFICANCE STATEMENT Therapeutic interventions designed to repair the damaged CST after spinal cord injury have remained functionally suboptimal in part because of an incomplete understanding of the molecular heterogeneity among subclasses of CSNs. Here, we combine spinal retrograde labeling with scRNAseq and annotate a CSN index by the termination pattern of their primary axon in the cervical or lumbar spinal cord and supraspinal collateral terminal fields. Using machine learning we have confirmed the veracity of our CSN gene lists to train classifiers to identify CSNs among all classes of neurons in primary motor cortex to study the development, patterning, homeostasis, and response to injury and disease, and ultimately target streamlined repair strategies to this critical motor pathway.
Collapse
Affiliation(s)
- Noa Golan
- Interdepartmental Neuroscience Program, Yale University School, New Haven, Connecticut 06511
- Department of Neurology, Yale University School, New Haven, Connecticut 06511
| | - Daniel Ehrlich
- Interdepartmental Neuroscience Program, Yale University School, New Haven, Connecticut 06511
- Department of Psychiatry, Yale University School, New Haven, Connecticut 06511
| | - James Bonanno
- Interdepartmental Neuroscience Program, Yale University School, New Haven, Connecticut 06511
- Department of Neurology, Yale University School, New Haven, Connecticut 06511
| | - Rory F O'Brien
- Department of Neurology, Yale University School, New Haven, Connecticut 06511
| | - Matias Murillo
- Interdepartmental Neuroscience Program, Yale University School, New Haven, Connecticut 06511
- Department of Neurology, Yale University School, New Haven, Connecticut 06511
| | - Sierra D Kauer
- Department of Neurology, Yale University School, New Haven, Connecticut 06511
| | - Neal Ravindra
- Department of Internal Medicine, Yale University School, New Haven, Connecticut 06511
- Department of Computer Science, Yale University School, New Haven, Connecticut 06511
| | - David Van Dijk
- Department of Internal Medicine, Yale University School, New Haven, Connecticut 06511
- Department of Computer Science, Yale University School, New Haven, Connecticut 06511
| | - William B Cafferty
- Department of Neurology, Yale University School, New Haven, Connecticut 06511
- Department of Neuroscience, Yale University School, New Haven, Connecticut 06511
| |
Collapse
|
32
|
Frase S, Steddin J, Paschen E, Lenz M, Conforti P, Haas CA, Vlachos A, Schachtrup C, Hosp JA. Dense dopaminergic innervation of the peri-infarct cortex despite dopaminergic cell loss after a pure motor-cortical stroke in rats. J Neurochem 2023; 167:427-440. [PMID: 37735852 DOI: 10.1111/jnc.15970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 08/28/2023] [Accepted: 09/12/2023] [Indexed: 09/23/2023]
Abstract
After ischemic stroke, the cortex directly adjacent to the ischemic core (i.e., the peri-infarct cortex, PIC) undergoes plastic changes that facilitate motor recovery. Dopaminergic signaling is thought to support this process. However, ischemic stroke also leads to the remote degeneration of dopaminergic midbrain neurons, possibly interfering with this beneficial effect. In this study, we assessed the reorganization of dopaminergic innervation of the PIC in a rat model of focal cortical stroke. Adult Sprague-Dawley rats either received a photothrombotic stroke (PTS) in the primary motor cortex (M1) or a sham operation. 30 days after PTS or sham procedure, the retrograde tracer Micro Ruby (MR) was injected into the PIC of stroke animals or into homotopic cortical areas of matched sham rats. Thus, dopaminergic midbrain neurons projecting into the PIC were identified based on MR signal and immunoreactivity against tyrosine hydroxylase (TH), a marker for dopaminergic neurons. The density of dopaminergic innervation within the PIC was assessed by quantification of dopaminergic boutons indicated by TH-immunoreactivity. Regarding postsynaptic processes, expression of dopamine receptors (D1- and D2) and a marker of the functional signal cascade (DARPP-32) were visualized histologically. Despite a 25% ipsilesional loss of dopaminergic midbrain neurons after PTS, the number and spatial distribution of dopaminergic neurons projecting to the PIC was not different compared to sham controls. Moreover, the density of dopaminergic innervation in the PIC was significantly higher than in homotopic cortical areas of the sham group. Within the PIC, D1-receptors were expressed in neurons, whereas D2-receptors were confined to astrocytes. The intensity of D1- and DARPP-32 expression appeared to be higher in the PIC compared to the contralesional homotopic cortex. Our data suggest a sprouting of dopaminergic fibers into the PIC and point to a role for dopaminergic signaling in reparative mechanisms post-stroke, potentially related to recovery.
Collapse
Affiliation(s)
- Sibylle Frase
- Department of Neurology and Neuroscience, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Julius Steddin
- Department of Neurology and Neuroscience, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Enya Paschen
- Experimental Epilepsy Research, Department of Neurosurgery, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Maximilian Lenz
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Pasquale Conforti
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Carola A Haas
- Experimental Epilepsy Research, Department of Neurosurgery, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christian Schachtrup
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jonas A Hosp
- Department of Neurology and Neuroscience, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
33
|
Chen SY, Yang RL, Wu XC, Zhao DZ, Fu SP, Lin FQ, Li LY, Yu LM, Zhang Q, Zhang T. Mesenchymal Stem Cell Transplantation: Neuroprotection and Nerve Regeneration After Spinal Cord Injury. J Inflamm Res 2023; 16:4763-4776. [PMID: 37881652 PMCID: PMC10595983 DOI: 10.2147/jir.s428425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 10/03/2023] [Indexed: 10/27/2023] Open
Abstract
Spinal Cord Injury (SCI), with its morbidity characteristics of high disability rate and high mortality rate, is a disease that is highly destructive to both the physiology and psychology of the patient, and for which there is still a lack of effective treatment. Following spinal cord injury, a cascade of secondary injury reactions known as ischemia, peripheral inflammatory cell infiltration, oxidative stress, etc. create a microenvironment that is unfavorable to neural recovery and ultimately results in apoptosis and necrosis of neurons and glial cells. Mesenchymal stem cell (MSC) transplantation has emerged as a more promising therapeutic options in recent years. MSC can promote spinal cord injury repair through a variety of mechanisms, including immunomodulation, neuroprotection, and nerve regeneration, giving patients with spinal cord injury hope. In this paper, it is discussed the neuroprotection and nerve regeneration components of MSCs' therapeutic method for treating spinal cord injuries.
Collapse
Affiliation(s)
- Si-Yu Chen
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - Rui-Lin Yang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - Xiang-Chong Wu
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - De-Zhi Zhao
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - Sheng-Ping Fu
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - Feng-Qin Lin
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - Lin-Yan Li
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - Li-Mei Yu
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - Qian Zhang
- Department of Human Anatomy, Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - Tao Zhang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| |
Collapse
|
34
|
Glotfelty EJ, Hsueh SC, Claybourne Q, Bedolla A, Kopp KO, Wallace T, Zheng B, Luo Y, Karlsson TE, McDevitt RA, Olson L, Greig NH. Microglial Nogo delays recovery following traumatic brain injury in mice. Glia 2023; 71:2473-2494. [PMID: 37401784 PMCID: PMC10528455 DOI: 10.1002/glia.24436] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/16/2023] [Accepted: 06/20/2023] [Indexed: 07/05/2023]
Abstract
Nogo-A, B, and C are well described members of the reticulon family of proteins, most well known for their negative regulatory effects on central nervous system (CNS) neurite outgrowth and repair following injury. Recent research indicates a relationship between Nogo-proteins and inflammation. Microglia, the brain's immune cells and inflammation-competent compartment, express Nogo protein, although specific roles of the Nogo in these cells is understudied. To examine inflammation-related effects of Nogo, we generated a microglial-specific inducible Nogo KO (MinoKO) mouse and challenged the mouse with a controlled cortical impact (CCI) traumatic brain injury (TBI). Histological analysis shows no difference in brain lesion sizes between MinoKO-CCI and Control-CCI mice, although MinoKO-CCI mice do not exhibit the levels of ipsilateral lateral ventricle enlargement as injury matched controls. Microglial Nogo-KO results in decreased lateral ventricle enlargement, microglial and astrocyte immunoreactivity, and increased microglial morphological complexity compared to injury matched controls, suggesting decreased tissue inflammation. Behaviorally, healthy MinoKO mice do not differ from control mice, but automated tracking of movement around the home cage and stereotypic behavior, such as grooming and eating (termed cage "activation"), following CCI is significantly elevated. Asymmetrical motor function, a deficit typical of unilaterally brain lesioned rodents, was not detected in CCI injured MinoKO mice, while the phenomenon was present in CCI injured controls 1-week post-injury. Overall, our studies show microglial Nogo as a negative regulator of recovery following brain injury. To date, this is the first evaluation of the roles microglial specific Nogo in a rodent injury model.
Collapse
Affiliation(s)
- Elliot J. Glotfelty
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, NIH, Baltimore, MD 21224, USA
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Shih-Chang Hsueh
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Quia Claybourne
- Comparative Medicine Section, National Institute on Aging, NIH, Baltimore, Maryland 21224, USA
| | - Alicia Bedolla
- Department of Molecular Genetics and Biochemistry, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Katherine O. Kopp
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Tonya Wallace
- Flow Cytometry Unit, National Institute on Aging, Baltimore, MD, USA
| | - Binhai Zheng
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Yu Luo
- Department of Molecular Genetics and Biochemistry, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | | | - Ross A. McDevitt
- Comparative Medicine Section, National Institute on Aging, NIH, Baltimore, Maryland 21224, USA
| | - Lars Olson
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Nigel H. Greig
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, NIH, Baltimore, MD 21224, USA
| |
Collapse
|
35
|
Powers BE, Ton ST, Farrer RG, Chaudhary S, Nockels RP, Kartje GL, Tsai SY. Anti-Nogo-A Antibody Therapy Improves Functional Outcome Following Traumatic Brain Injury. Neurorehabil Neural Repair 2023; 37:682-693. [PMID: 37837331 PMCID: PMC10843026 DOI: 10.1177/15459683231203194] [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] [Indexed: 10/16/2023]
Abstract
BACKGROUND Traumatic brain injury (TBI) can cause sensorimotor deficits, and recovery is slow and incomplete. There are no effective pharmacological treatments for recovery from TBI, but research indicates potential for anti-Nogo-A antibody (Ab) therapy. This Ab neutralizes Nogo-A, an endogenous transmembrane protein that inhibits neuronal plasticity and regeneration. OBJECTIVE We hypothesized that anti-Nogo-A Ab treatment following TBI results in disinhibited axonal growth from the contralesional cortex, the establishment of new compensatory neuronal connections, and improved function. METHODS We modeled TBI in rats using the controlled cortical impact method, resulting in focal brain damage and motor deficits like those observed in humans with a moderate cortical TBI. Rats were trained on the skilled forelimb reaching task and the horizontal ladder rung walking task. They were then given a TBI, targeting the caudal forelimb motor cortex, and randomly divided into 3 groups: TBI-only, TBI + Anti-Nogo-A Ab, and TBI + Control Ab. Testing resumed 3 days after TBI and continued for 8 weeks, when rats received an injection of the anterograde neuronal tracer, biotinylated dextran amine (BDA), into the corresponding area contralateral to the TBI. RESULTS We observed significant improvement in rats that received anti-Nogo-A Ab treatment post-TBI compared to controls. Analysis of BDA-positive axons revealed that anti-Nogo-A Ab treatment resulted in cortico-rubral plasticity to the deafferented red nucleus. Conclusions. Anti-Nogo-A Ab treatment may improve functional recovery via neuronal plasticity to brain areas important for skilled movements, and this treatment shows promise to improve outcomes in humans who have suffered a TBI.
Collapse
Affiliation(s)
- Brian E Powers
- Edward Hines Jr. Veteran Affairs Hospital, Hines, IL, USA
| | - Son T Ton
- Edward Hines Jr. Veteran Affairs Hospital, Hines, IL, USA
| | | | | | - Russ P Nockels
- Department of Neurological Surgery, Loyola University Medical Center, Maywood, IL, USA
| | - Gwendolyn L Kartje
- Edward Hines Jr. Veteran Affairs Hospital, Hines, IL, USA
- Department of Molecular Pharmacology and Neuroscience, Loyola University Health Sciences Division, Maywood, IL, USA
| | - Shih-Yen Tsai
- Edward Hines Jr. Veteran Affairs Hospital, Hines, IL, USA
| |
Collapse
|
36
|
Sekine Y, Wang X, Kikkawa K, Honda S, Strittmatter SM. Amino-terminal proteolytic fragment of the axon growth inhibitor Nogo-A (Rtn4A) is upregulated by injury and promotes axon regeneration. J Biol Chem 2023; 299:105232. [PMID: 37690690 PMCID: PMC10622843 DOI: 10.1016/j.jbc.2023.105232] [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: 04/21/2023] [Revised: 08/28/2023] [Accepted: 08/31/2023] [Indexed: 09/12/2023] Open
Abstract
After adult mammalian central nervous system injury, axon regeneration is extremely limited or absent, resulting in persistent neurological deficits. Axon regeneration failure is due in part to the presence of inhibitory proteins, including NogoA (Rtn4A), from which two inhibitory domains have been defined. When these inhibitory domains are deleted, but an amino-terminal domain is still expressed in a gene trap line, mice show axon regeneration and enhanced recovery from injury. In contrast, when there is no amino-terminal Nogo-A fragment in the setting of inhibitory domain deletion, then axon regeneration and recovery are indistinguishable from WT. These data indicated that an amino-terminal Nogo-A fragment derived from the gene trap might promote axon regeneration, but this had not been tested directly and production of this fragment without gene targeting was unclear. Here, we describe posttranslation production of an amino-terminal fragment of Nogo-A from the intact gene product. This fragment is created by proteolysis near amino acid G214-N215 and levels are enhanced by axotomy. Furthermore, this fragment promotes axon regeneration in vitro and acts cell autonomously in neurons, in contrast to the inhibitory extracellular action of other Nogo-A domains.Proteins interacting with the amino-terminal Nogo-A fragment by immunoprecipitation include HSPA8 (HSC70, HSP7C). Suppression of HSPA8 expression by shRNA decreases axon regeneration from cerebral cortical neurons and overexpression increases axon regeneration. Moreover, the amino-terminal Nogo-A fragment increases HSPA8 chaperone activity. These data provide an explanation for varied results in different gene-targeted Nogo-A mice, as well as revealing an axon regeneration promoting domain of Nogo-A.
Collapse
Affiliation(s)
- Yuichi Sekine
- Department of Neuroscience and Neurology, Cellular Neuroscience, Neurodegeneration & Repair Program, Yale School of Medicine, New Haven, Connecticut, USA; Department of Cell Biology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Xingxing Wang
- Department of Neuroscience and Neurology, Cellular Neuroscience, Neurodegeneration & Repair Program, Yale School of Medicine, New Haven, Connecticut, USA
| | - Kazuna Kikkawa
- Department of Cell Biology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Sachie Honda
- Department of Cell Biology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Stephen M Strittmatter
- Department of Neuroscience and Neurology, Cellular Neuroscience, Neurodegeneration & Repair Program, Yale School of Medicine, New Haven, Connecticut, USA.
| |
Collapse
|
37
|
Patil V, Bohara R, Krishna Kanala V, McMahon S, Pandit A. Models and approaches to comprehend and address glial inflammation following spinal cord injury. Drug Discov Today 2023; 28:103722. [PMID: 37482236 DOI: 10.1016/j.drudis.2023.103722] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/10/2023] [Accepted: 07/18/2023] [Indexed: 07/25/2023]
Abstract
Spinal cord injury (SCI) culminates in chronic inflammation and glial scar formation driven by the activation of microglia and astrocytes. Current anti-inflammatory strategies to treat glial activation associated with SCI have several limitations. Existing in vitro and ex vivo models studying molecular mechanisms associated with inflammation focus only on the acute phase. However, the progression of glial cell-derived inflammation over the acute-to-chronic phases has not been assessed. Understanding this progression will help establish a framework for evaluating therapeutic strategies. Additionally, new models could be useful as high-throughput screening (HTS) platforms. This review aims to highlight currently available models and future methods that could facilitate screening of novel therapeutics for SCI.
Collapse
Affiliation(s)
- Vaibhav Patil
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| | - Raghvendra Bohara
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| | - Vijaya Krishna Kanala
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| | - Siobhan McMahon
- Anatomy, School of Medicine, University of Galway, Galway, Ireland
| | - Abhay Pandit
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland.
| |
Collapse
|
38
|
Zeng CW, Tsai HJ. The Promising Role of a Zebrafish Model Employed in Neural Regeneration Following a Spinal Cord Injury. Int J Mol Sci 2023; 24:13938. [PMID: 37762240 PMCID: PMC10530783 DOI: 10.3390/ijms241813938] [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: 07/27/2023] [Revised: 09/07/2023] [Accepted: 09/09/2023] [Indexed: 09/29/2023] Open
Abstract
Spinal cord injury (SCI) is a devastating event that results in a wide range of physical impairments and disabilities. Despite the advances in our understanding of the biological response to injured tissue, no effective treatments are available for SCIs at present. Some studies have addressed this issue by exploring the potential of cell transplantation therapy. However, because of the abnormal microenvironment in injured tissue, the survival rate of transplanted cells is often low, thus limiting the efficacy of such treatments. Many studies have attempted to overcome these obstacles using a variety of cell types and animal models. Recent studies have shown the utility of zebrafish as a model of neural regeneration following SCIs, including the proliferation and migration of various cell types and the involvement of various progenitor cells. In this review, we discuss some of the current challenges in SCI research, including the accurate identification of cell types involved in neural regeneration, the adverse microenvironment created by SCIs, attenuated immune responses that inhibit nerve regeneration, and glial scar formation that prevents axonal regeneration. More in-depth studies are needed to fully understand the neural regeneration mechanisms, proteins, and signaling pathways involved in the complex interactions between the SCI microenvironment and transplanted cells in non-mammals, particularly in the zebrafish model, which could, in turn, lead to new therapeutic approaches to treat SCIs in humans and other mammals.
Collapse
Affiliation(s)
- Chih-Wei Zeng
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Huai-Jen Tsai
- Department of Life Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan
| |
Collapse
|
39
|
Bandla AC, Sheth AS, Zarate SM, Uskamalla S, Hager EC, Villarreal VA, González-García M, Ballestero RP. Enhancing structural plasticity of PC12 neurons during differentiation and neurite regeneration with a catalytically inactive mutant version of the zRICH protein. BMC Neurosci 2023; 24:43. [PMID: 37612637 PMCID: PMC10463786 DOI: 10.1186/s12868-023-00808-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 06/23/2023] [Indexed: 08/25/2023] Open
Abstract
BACKGROUND Studies of the molecular mechanisms of nerve regeneration have led to the discovery of several proteins that are induced during successful nerve regeneration. RICH proteins were identified as proteins induced during the regeneration of the optic nerve of teleost fish. These proteins are 2',3'-cyclic nucleotide, 3'-phosphodiesterases that can bind to cellular membranes through a carboxy-terminal membrane localization domain. They interact with the tubulin cytoskeleton and are able to enhance neuronal structural plasticity by promoting the formation of neurite branches. RESULTS PC12 stable transfectant cells expressing a fusion protein combining a red fluorescent protein with a catalytically inactive mutant version of zebrafish RICH protein were generated. These cells were used as a model to analyze effects of the protein on neuritogenesis. Differentiation experiments showed a 2.9 fold increase in formation of secondary neurites and a 2.4 fold increase in branching points. A 2.2 fold increase in formation of secondary neurites was observed in neurite regeneration assays. CONCLUSIONS The use of a fluorescent fusion protein facilitated detection of expression levels. Two computer-assisted morphometric analysis methods indicated that the catalytically inactive RICH protein induced the formation of branching points and secondary neurites both during differentiation and neurite regeneration. A procedure based on analysis of random field images provided comparable results to classic neurite tracing methods.
Collapse
Affiliation(s)
- Ashoka C Bandla
- Department of Biological and Health Sciences, Texas A&M University-Kingsville, 700 University Blvd, Kingsville, TX, 78363, USA
| | - Aditya S Sheth
- Department of Biological and Health Sciences, Texas A&M University-Kingsville, 700 University Blvd, Kingsville, TX, 78363, USA
| | - Sara M Zarate
- Department of Biological and Health Sciences, Texas A&M University-Kingsville, 700 University Blvd, Kingsville, TX, 78363, USA
| | - Suraj Uskamalla
- Department of Biological and Health Sciences, Texas A&M University-Kingsville, 700 University Blvd, Kingsville, TX, 78363, USA
| | - Elizabeth C Hager
- Department of Biological and Health Sciences, Texas A&M University-Kingsville, 700 University Blvd, Kingsville, TX, 78363, USA
| | - Victor A Villarreal
- Department of Chemistry, Texas A&M University-Kingsville, Kingsville, TX, 78363, USA
| | | | - Rafael P Ballestero
- Department of Biological and Health Sciences, Texas A&M University-Kingsville, 700 University Blvd, Kingsville, TX, 78363, USA.
| |
Collapse
|
40
|
Maynard G, Kannan R, Liu J, Wang W, Lam TKT, Wang X, Adamson C, Hackett C, Schwab JM, Liu C, Leslie DP, Chen D, Marino R, Zafonte R, Flanders A, Block G, Smith E, Strittmatter SM. Soluble Nogo-Receptor-Fc decoy (AXER-204) in patients with chronic cervical spinal cord injury in the USA: a first-in-human and randomised clinical trial. Lancet Neurol 2023; 22:672-684. [PMID: 37479373 PMCID: PMC10410101 DOI: 10.1016/s1474-4422(23)00215-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/13/2023] [Accepted: 06/02/2023] [Indexed: 07/23/2023]
Abstract
BACKGROUND Spinal cord injury (SCI) causes neural disconnection and persistent neurological deficits, so axon sprouting and plasticity might promote recovery. Soluble Nogo-Receptor-Fc decoy (AXER-204) blocks inhibitors of axon growth and promotes recovery of motor function after SCI in animals. This first-in-human and randomised trial sought to determine primarily the safety and pharmacokinetics of AXER-204 in individuals with chronic SCI, and secondarily its effect on recovery. METHODS We conducted a two-part study in adults (aged 18-65 years) with chronic (>1 year) cervical traumatic SCI at six rehabilitation centres in the USA. In part 1, AXER-204 was delivered open label as single intrathecal doses of 3 mg, 30 mg, 90 mg, or 200 mg, with primary outcomes of safety and pharmacokinetics. Part 2 was a randomised, parallel, double-blind comparison of six intrathecal doses of 200 mg AXER-204 over 104 days versus placebo. Participants were randomly allocated (1:1) by investigators using a central electronic system, stratified in blocks of four by American Spinal Injury Association Impairment Scale grade and receipt of AXER-204 in part 1. All investigators and patients were masked to treatment allocation until at least day 169. The part 2 primary objectives were safety and pharmacokinetics, with a key secondary objective to assess change in International Standards for Neurological Classification of SCI (ISNCSCI) Upper Extremity Motor Score (UEMS) at day 169 for all enrolled participants. This trial is registered with ClinicalTrials.gov, NCT03989440, and is completed. FINDINGS We treated 24 participants in part 1 (six per dose; 18 men, six women), and 27 participants in part 2 (13 placebo, 14 AXER-204; 23 men, four women), between June 20, 2019, and June 21, 2022. There were no deaths and no discontinuations from the study due to an adverse event in part 1 and 2. In part 2, treatment-related adverse events were of similar incidence in AXER-204 and placebo groups (ten [71%] vs nine [69%]). Headache was the most common treatment-related adverse event (five [21%] in part 1, 11 [41%] in part 2). In part 1, AXER-204 reached mean maximal CSF concentration 1 day after dosing with 200 mg of 412 000 ng/mL (SD 129 000), exceeding those concentrations that were efficacious in animal studies. In part 2, mean changes from baseline to day 169 in ISNCSCI UEMS were 1·5 (SD 3·3) for AXER-204 and 0·9 (2·3) for placebo (mean difference 0·54, 95% CI -1·48 to 2·55; p=0·59). INTERPRETATION This study delivers the first, to our knowledge, clinical trial of a rationally designed pharmacological treatment intended to promote neural repair in chronic SCI. AXER-204 appeared safe and reached target CSF concentrations; exploratory biomarker results were consistent with target engagement and synaptic stabilisation. Post-hoc subgroup analyses suggest that future trials could investigate efficacy in patients with moderately severe SCI without prior AXER-204 exposure. FUNDING Wings for Life Foundation, National Institute of Neurological Disorders and Stroke, National Center for Advancing Translational Sciences, National Institute on Drug Abuse, and ReNetX Bio.
Collapse
Affiliation(s)
| | - Ramakrishnan Kannan
- Departments of Neuroscience and Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Jian Liu
- Departments of Neuroscience and Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Weiwei Wang
- Keck MS and Proteomic Resource, Yale School of Medicine, New Haven, CT, USA
| | - Tu Kiet T Lam
- Keck MS and Proteomic Resource, Yale School of Medicine, New Haven, CT, USA; Department of Molecular Biophysics and Biochemistry, Yale School of Medicine, New Haven, CT, USA
| | - Xingxing Wang
- Departments of Neuroscience and Neurology, Yale School of Medicine, New Haven, CT, USA
| | | | | | - Jan M Schwab
- Belford Center for Spinal Cord Injury and Departments of Neurology and Neuroscience, The Ohio State University, Wexner Medical Center, Columbus, OH, USA
| | - Charles Liu
- USC Neurorestoration Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | - David Chen
- Shirley Ryan AbilityLab, Chicago, IL, USA
| | - Ralph Marino
- Department of Rehabilitation Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Ross Zafonte
- Spaulding Rehabilitation Hospital, Massachusetts General Hospital, Brigham and Womens Hospital, Harvard Medical School, Boston, MA, USA
| | - Adam Flanders
- Department of Rehabilitation Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | | | | | | |
Collapse
|
41
|
Shafqat A, Albalkhi I, Magableh HM, Saleh T, Alkattan K, Yaqinuddin A. Tackling the glial scar in spinal cord regeneration: new discoveries and future directions. Front Cell Neurosci 2023; 17:1180825. [PMID: 37293626 PMCID: PMC10244598 DOI: 10.3389/fncel.2023.1180825] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/08/2023] [Indexed: 06/10/2023] Open
Abstract
Axonal regeneration and functional recovery are poor after spinal cord injury (SCI), typified by the formation of an injury scar. While this scar was traditionally believed to be primarily responsible for axonal regeneration failure, current knowledge takes a more holistic approach that considers the intrinsic growth capacity of axons. Targeting the SCI scar has also not reproducibly yielded nearly the same efficacy in animal models compared to these neuron-directed approaches. These results suggest that the major reason behind central nervous system (CNS) regeneration failure is not the injury scar but a failure to stimulate axon growth adequately. These findings raise questions about whether targeting neuroinflammation and glial scarring still constitute viable translational avenues. We provide a comprehensive review of the dual role of neuroinflammation and scarring after SCI and how future research can produce therapeutic strategies targeting the hurdles to axonal regeneration posed by these processes without compromising neuroprotection.
Collapse
|
42
|
Current Advancements in Spinal Cord Injury Research—Glial Scar Formation and Neural Regeneration. Cells 2023; 12:cells12060853. [PMID: 36980193 PMCID: PMC10046908 DOI: 10.3390/cells12060853] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/01/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023] Open
Abstract
Spinal cord injury (SCI) is a complex tissue injury resulting in permanent and degenerating damage to the central nervous system (CNS). Detrimental cellular processes occur after SCI, including axonal degeneration, neuronal loss, neuroinflammation, reactive gliosis, and scar formation. The glial scar border forms to segregate the neural lesion and isolate spreading inflammation, reactive oxygen species, and excitotoxicity at the injury epicenter to preserve surrounding healthy tissue. The scar border is a physicochemical barrier composed of elongated astrocytes, fibroblasts, and microglia secreting chondroitin sulfate proteoglycans, collogen, and the dense extra-cellular matrix. While this physiological response preserves viable neural tissue, it is also detrimental to regeneration. To overcome negative outcomes associated with scar formation, therapeutic strategies have been developed: the prevention of scar formation, the resolution of the developed scar, cell transplantation into the lesion, and endogenous cell reprogramming. This review focuses on cellular/molecular aspects of glial scar formation, and discusses advantages and disadvantages of strategies to promote regeneration after SCI.
Collapse
|
43
|
Oh SC, Kim SE, Jang IH, Kim SM, Lee SY, Lee S, Chu IS, Yoon SR, Jung H, Choi I, Doh J, Kim TD. NgR1 is an NK cell inhibitory receptor that destabilizes the immunological synapse. Nat Immunol 2023; 24:463-473. [PMID: 36624164 DOI: 10.1038/s41590-022-01394-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 11/22/2022] [Indexed: 01/10/2023]
Abstract
The formation of an immunological synapse (IS) is essential for natural killer (NK) cells to eliminate target cells. Despite an advanced understanding of the characteristics of the IS and its formation processes, the mechanisms that regulate its stability via the cytoskeleton are unclear. Here, we show that Nogo receptor 1 (NgR1) has an important function in modulating NK cell-mediated killing by destabilization of IS formation. NgR1 deficiency or blockade resulted in improved tumor control of NK cells by enhancing NK-to-target cell contact stability and regulating F-actin dynamics during IS formation. Patients with tumors expressing abundant NgR1 ligand had poor prognosis despite high levels of NK cell infiltration. Thus, our study identifies NgR1 as an immune checkpoint in IS formation and indicates a potential approach to improve the cytolytic function of NK cells in cancer immunotherapy.
Collapse
Affiliation(s)
- Se-Chan Oh
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.,Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Seong-Eun Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - In-Hwan Jang
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.,Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Seok-Min Kim
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.,Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Soo Yun Lee
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Sunyoung Lee
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.,Department of Life Sciences, Korea University, Seoul, Republic of Korea
| | - In-Sun Chu
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.,Department of Bioinformatics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Suk Ran Yoon
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.,Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Haiyoung Jung
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.,Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Inpyo Choi
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Junsang Doh
- Department of Materials Science and Engineering, Research Institute of Advanced Materials, Institute of Engineering Research, Bio-MAX Institute, Soft Foundry Institute, Seoul National University, Seoul, Republic of Korea.
| | - Tae-Don Kim
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea. .,Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea. .,Biomedical Mathematics Group, Institute for Basic Science, Daejeon, Republic of Korea. .,Department of Biopharmaceutical Convergence, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea.
| |
Collapse
|
44
|
Czech VL, O'Connor LC, Philippon B, Norman E, Byrne AB. TIR-1/SARM1 inhibits axon regeneration and promotes axon degeneration. eLife 2023; 12:80856. [PMID: 37083456 PMCID: PMC10121217 DOI: 10.7554/elife.80856] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 01/31/2023] [Indexed: 03/03/2023] Open
Abstract
Growth and destruction are central components of the neuronal injury response. Injured axons that are capable of repair, including axons in the mammalian peripheral nervous system and in many invertebrate animals, often regenerate and degenerate on either side of the injury. Here we show that TIR-1/dSarm/SARM1, a key regulator of axon degeneration, also inhibits regeneration of injured motor axons. The increased regeneration in tir-1 mutants is not a secondary consequence of its effects on degeneration, nor is it determined by the NADase activity of TIR-1. Rather, we found that TIR-1 functions cell-autonomously to regulate each of the seemingly opposite processes through distinct interactions with two MAP kinase pathways. On one side of the injury, TIR-1 inhibits axon regeneration by activating the NSY-1/ASK1 MAPK signaling cascade, while on the other side of the injury, TIR-1 simultaneously promotes axon degeneration by interacting with the DLK-1 mitogen-activated protein kinase (MAPK) signaling cascade. In parallel, we found that the ability to cell-intrinsically inhibit axon regeneration is conserved in human SARM1. Our finding that TIR-1/SARM1 regulates axon regeneration provides critical insight into how axons coordinate a multidimensional response to injury, consequently informing approaches to manipulate the response toward repair.
Collapse
Affiliation(s)
- Victoria L Czech
- Department of Neurobiology, UMass Chan Massachusetts Medical School
| | | | | | - Emily Norman
- Department of Neurobiology, UMass Chan Massachusetts Medical School
| | | |
Collapse
|
45
|
Nogo-A and LINGO-1: Two Important Targets for Remyelination and Regeneration. Int J Mol Sci 2023; 24:ijms24054479. [PMID: 36901909 PMCID: PMC10003089 DOI: 10.3390/ijms24054479] [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: 01/24/2023] [Revised: 02/13/2023] [Accepted: 02/22/2023] [Indexed: 02/26/2023] Open
Abstract
Multiple sclerosis (MS) is an inflammatory disease of the central nervous system (CNS) that causes progressive neurological disability in most patients due to neurodegeneration. Activated immune cells infiltrate the CNS, triggering an inflammatory cascade that leads to demyelination and axonal injury. Non-inflammatory mechanisms are also involved in axonal degeneration, although they are not fully elucidated yet. Current therapies focus on immunosuppression; however, no therapies to promote regeneration, myelin repair, or maintenance are currently available. Two different negative regulators of myelination have been proposed as promising targets to induce remyelination and regeneration, namely the Nogo-A and LINGO-1 proteins. Although Nogo-A was first discovered as a potent neurite outgrowth inhibitor in the CNS, it has emerged as a multifunctional protein. It is involved in numerous developmental processes and is necessary for shaping and later maintaining CNS structure and functionality. However, the growth-restricting properties of Nogo-A have negative effects on CNS injury or disease. LINGO-1 is also an inhibitor of neurite outgrowth, axonal regeneration, oligodendrocyte differentiation, and myelin production. Inhibiting the actions of Nogo-A or LINGO-1 promotes remyelination both in vitro and in vivo, while Nogo-A or LINGO-1 antagonists have been suggested as promising therapeutic approaches for demyelinating diseases. In this review, we focus on these two negative regulators of myelination while also providing an overview of the available data on the effects of Nogo-A and LINGO-1 inhibition on oligodendrocyte differentiation and remyelination.
Collapse
|
46
|
Zheng B, Tuszynski MH. Regulation of axonal regeneration after mammalian spinal cord injury. Nat Rev Mol Cell Biol 2023; 24:396-413. [PMID: 36604586 DOI: 10.1038/s41580-022-00562-y] [Citation(s) in RCA: 117] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2022] [Indexed: 01/06/2023]
Abstract
One hundred years ago, Ramón y Cajal, considered by many as the founder of modern neuroscience, stated that neurons of the adult central nervous system (CNS) are incapable of regenerating. Yet, recent years have seen a tremendous expansion of knowledge in the molecular control of axon regeneration after CNS injury. We now understand that regeneration in the adult CNS is limited by (1) a failure to form cellular or molecular substrates for axon attachment and elongation through the lesion site; (2) environmental factors, including inhibitors of axon growth associated with myelin and the extracellular matrix; (3) astrocyte responses, which can both limit and support axon growth; and (4) intraneuronal mechanisms controlling the establishment of an active cellular growth programme. We discuss these topics together with newly emerging hypotheses, including the surprising finding from transcriptomic analyses of the corticospinal system in mice that neurons revert to an embryonic state after spinal cord injury, which can be sustained to promote regeneration with neural stem cell transplantation. These gains in knowledge are steadily advancing efforts to develop effective treatment strategies for spinal cord injury in humans.
Collapse
Affiliation(s)
- Binhai Zheng
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, CA, USA. .,VA San Diego Research Service, San Diego, CA, USA.
| | - Mark H Tuszynski
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, CA, USA. .,VA San Diego Research Service, San Diego, CA, USA.
| |
Collapse
|
47
|
Hao DL, Li JM, Xie R, Huo HR, Xiong XJ, Sui F, Wang PQ. The role of traditional herbal medicine for ischemic stroke: from bench to clinic-A critical review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 109:154609. [PMID: 36610141 DOI: 10.1016/j.phymed.2022.154609] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 09/29/2022] [Accepted: 12/16/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Ischemic stroke (IS) is a leading cause of death and severe long-term disability worldwide. Over the past few decades, considerable progress has been made in anti-ischemic therapies. However, IS remains a tremendous challenge, with favourable clinical outcomes being generally difficult to achieve from candidate drugs in preclinical phase testing. Traditional herbal medicine (THM) has been used to treat stroke for over 2,000 years in China. In modern times, THM as an alternative and complementary therapy have been prescribed in other Asian countries and have gained increasing attention for their therapeutic effects. These millennia of clinical experience allow THM to be a promising avenue for improving clinical efficacy and accelerating drug discovery. PURPOSE To summarise the clinical evidence and potential mechanisms of THMs in IS. METHODS A comprehensive literature search was conducted in seven electronic databases, including PubMed, EMBASE, the Cochrane Central Register of Controlled Trials, the Chinese National Knowledge Infrastructure, the VIP Information Database, the Chinese Biomedical Literature Database, and the Wanfang Database, from inception to 17 June 2022 to examine the efficacy and safety of THM for IS, and to investigate experimental studies regarding potential mechanisms. RESULTS THM is widely prescribed for IS alone or as adjuvant therapy. In clinical trials, THM is generally administered within 72 h of stroke onset and are continuously prescribed for over 3 months. Compared with Western medicine (WM), THM combined with routine WM can significantly improve neurological function defect scores, promote clinical total effective rate, and accelerate the recovery time of stroke with fewer adverse effects (AEs). These effects can be attributed to multiple mechanisms, mainly anti-inflammation, antioxidative stress, anti-apoptosis, brain blood barrier (BBB) modulation, inhibition of platelet activation and thrombus formation, and promotion of neurogenesis and angiogenesis. CONCLUSIONS THM may be a promising candidate for IS management to guide clinical applications and as a reference for drug development.
Collapse
Affiliation(s)
- Dan-Li Hao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Jia-Meng Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Ran Xie
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Hai-Ru Huo
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xing-Jiang Xiong
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China.
| | - Feng Sui
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Peng-Qian Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| |
Collapse
|
48
|
Schwaiger C, Haider T, Endmayr V, Zrzavy T, Gruber VE, Ricken G, Simonovska A, Hametner S, Schwab JM, Höftberger R. Dynamic induction of the myelin-associated growth inhibitor Nogo-A in perilesional plasticity regions after human spinal cord injury. Brain Pathol 2023; 33:e13098. [PMID: 35698271 PMCID: PMC9836369 DOI: 10.1111/bpa.13098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 05/29/2022] [Indexed: 01/21/2023] Open
Abstract
The myelin-associated inhibitor Nogo-A (Reticulon 4, RTN4) restricts axonal outgrowth, plasticity, and neural circuitry formation in experimental models of spinal cord injury (SCI) and is targeted in clinical interventions starting treatment within 4 weeks post-SCI. Specifically, Nogo-A expressed by oligodendroglia restricts compensatory neurite sprouting. To interrogate the hypothesis of an inducible, lesion reactive Nogo-A expression over time, we analyzed the spatiotemporal Nogo-A expression at the spinal lesion core (region of tissue necrosis and axonal damage/pruning) and perilesional rim (region of plasticity formation). Spinal cord specimens of SCI subjects (n = 22) were compared to neuropathologically unaltered controls (n = 9). Nogo-A expression was investigated ranging from acute (0-3 days), early subacute (4-21 days), late subacute (22-90 days) to early chronic-chronic (91 days to 1.5 years after SCI) stages after SCI. Nogo-A expression in controls is confined to motoneurons in the anterior horn and to oligodendrocytes in gray and white matter. After SCI, the number of Nogo-A+ and TPPP/p25+ oligodendrocytes (i) inclined at the organizing perilesional rim specifically, (ii) increased further over time, and (iii) peaked at chronic stages after SCI. By contrast, at the lesion core, the number of Nogo-A+ and TPPP/p25+ oligodendrocytes did not increase. Increasing numbers of Nogo-A+ oligodendrocytes coincided with oligodendrogenesis corroborated by Nogo-A coexpression of Ki67+ , TPPP/p25+ proliferating oligodendrocytes. Nogo-A oligodendrocyte expression emerges at perilesional (plasticity) regions over time and suggests an extended therapeutical window for anti-Nogo-A pathway targeting interventions beyond 4 weeks in patients after SCI.
Collapse
Affiliation(s)
- Carmen Schwaiger
- Division of Neuropathology and Neurochemistry, Department of NeurologyMedical University of ViennaViennaAustria
| | - Thomas Haider
- Department of Orthopedics and Trauma SurgeryMedical University of ViennaViennaAustria
| | - Verena Endmayr
- Division of Neuropathology and Neurochemistry, Department of NeurologyMedical University of ViennaViennaAustria
| | - Tobias Zrzavy
- Department of NeurologyMedical University of ViennaViennaAustria
| | - Victoria E. Gruber
- Department of Pediatrics and Adolescent MedicineMedical University of Vienna (Affiliated Partner of the ERN EpiCARE)ViennaAustria
| | - Gerda Ricken
- Division of Neuropathology and Neurochemistry, Department of NeurologyMedical University of ViennaViennaAustria
| | - Anika Simonovska
- Center for Medical Physics and Biomedical EngineeringMedical University of ViennaViennaAustria
| | - Simon Hametner
- Division of Neuropathology and Neurochemistry, Department of NeurologyMedical University of ViennaViennaAustria
| | - Jan M. Schwab
- The Belford Center for Spinal Cord Injury and Departments of Neurology, Physical Medicine and Rehabilitation and NeurosciencesThe Ohio State UniversityColumbusOhioUSA
| | - Romana Höftberger
- Division of Neuropathology and Neurochemistry, Department of NeurologyMedical University of ViennaViennaAustria
| |
Collapse
|
49
|
Zochodne DW. Growth factors and molecular-driven plasticity in neurological systems. HANDBOOK OF CLINICAL NEUROLOGY 2023; 196:569-598. [PMID: 37620091 DOI: 10.1016/b978-0-323-98817-9.00017-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/26/2023]
Abstract
It has been almost 70 years since the discovery of nerve growth factor (NGF), a period of a dramatic evolution in our understanding of dynamic growth, regeneration, and rewiring of the nervous system. In 1953, the extraordinary finding that a protein found in mouse submandibular glands generated a halo of outgrowing axons has now redefined our concept of the nervous system connectome. Central and peripheral neurons and their axons or dendrites are no longer considered fixed or static "wiring." Exploiting this molecular-driven plasticity as a therapeutic approach has arrived in the clinic with a slate of new trials and ideas. Neural growth factors (GFs), soluble proteins that alter the behavior of neurons, have expanded in numbers and our understanding of the complexity of their signaling and interactions with other proteins has intensified. However, beyond these "extrinsic" determinants of neuron growth and function are the downstream pathways that impact neurons, ripe for translational development and potentially more important than individual growth factors that may trigger them. Persistent and ongoing nuances in clinical trial design in some of the most intractable and irreversible neurological conditions give hope for connecting new biological ideas with clinical benefits. This review is a targeted update on neural GFs, their signals, and new therapeutic ideas, selected from an expansive literature.
Collapse
Affiliation(s)
- Douglas W Zochodne
- Division of Neurology, Department of Medicine and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
50
|
Matsubayashi J, Kawaguchi Y, Kawakami Y, Takei K. Brain-derived neurotrophic factor (BDNF) induces antagonistic action to Nogo signaling by the upregulation of lateral olfactory tract usher substance (LOTUS) expression. J Neurochem 2023; 164:29-43. [PMID: 36448220 DOI: 10.1111/jnc.15732] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 11/10/2022] [Accepted: 11/16/2022] [Indexed: 12/03/2022]
Abstract
Neurons in the central nervous system (CNS) have limited capacity for axonal regeneration after trauma and neurological disorders due to an endogenous nonpermissive environment for axon regrowth in the CNS. Lateral olfactory tract usher substance (LOTUS) contributes to axonal tract formation in the developing brain and axonal regeneration in the adult brain as an endogenous Nogo receptor-1 (NgR1) antagonist. However, how LOTUS expression is regulated remains unclarified. This study examined molecular mechanism of regulation in LOTUS expression and found that brain-derived neurotrophic factor (BDNF) increased LOTUS expression in cultured hippocampal neurons. Exogenous application of BDNF increased LOTUS expression at both mRNA and protein levels in a dose-dependent manner. We also found that pharmacological inhibition with K252a and gene knockdown by siRNA of tropomyosin-related kinase B (TrkB), BDNF receptor suppressed BDNF-induced increase in LOTUS expression. Further pharmacological analysis of the TrkB signaling pathway revealed that BDNF increased LOTUS expression through mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kinase (PI3K) cascades, but not phospholipase C-γ (PLCγ) cascade. Additionally, treatment with c-AMP response element binding protein (CREB) inhibitor partially suppressed BDNF-induced LOTUS expression. Finally, neurite outgrowth assay in cultured hippocampal neurons revealed that BDNF treatment-induced antagonism for NgR1 by up-regulating LOTUS expression. These findings suggest that BDNF may acts as a positive regulator of LOTUS expression through the TrkB signaling, thereby inducing an antagonistic action for NgR1 function by up-regulating LOTUS expression. Also, BDNF may synergistically affect axon regrowth through the upregulation of LOTUS expression.
Collapse
Affiliation(s)
- Junpei Matsubayashi
- Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Yokohama, Japan
| | - Yuki Kawaguchi
- Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Yokohama, Japan
| | - Yutaka Kawakami
- Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Yokohama, Japan.,Department of Anesthesiology, National Center for Neurology and Psychiatry, Kodaira, Japan
| | - Kohtaro Takei
- Molecular Medical Bioscience Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, Yokohama, Japan
| |
Collapse
|