1
|
Asami Y, Tokutake K, Kurimoto S, Saeki S, Yamamoto M, Hirata H. Transplantation of embryonic spinal motor neurons into peripheral nerves enables functional reconstruction of a denervated diaphragm. Neuroscience 2024; 559:113-122. [PMID: 39216747 DOI: 10.1016/j.neuroscience.2024.08.039] [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: 08/20/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Respiratory muscle paralysis due to trauma or neurodegenerative diseases can have devastating consequences. Only a few studies have investigated the reconstruction of motor function in denervated diaphragms caused by such conditions. Here, we studied the efficacy of transplanting E14 embryonic spinal motor neurons (SMNs) into peripheral nerve grafts for functionally reconstructing a denervated diaphragm in a rat model. The diaphragms of 8-week-old male Fischer 344 rats were first denervated by transecting the phrenic nerves. Subsequently, peripheral nerve grafts taken from the lower limb were used for neurotization of the denervated diaphragms. One week later, fetal E14 SMNs were transplanted into the peripheral nerve grafts. After 3 months, we observed functional contraction of the diaphragm following neuromuscular electrical stimulation (NMES) of the peripheral nerve graft. Additionally, we confirmed that SMN transplantation into the peripheral nerve graft had an inhibitory effect on diaphragm muscle atrophy. The SMNs transplanted into the peripheral nerve grafts formed a structure similar to the spinal cord, and the neuromuscular junction of the denervated diaphragm was reinnervated. These findings suggest the establishment of an ectopic motor neuron pool in the peripheral nerve graft. Free peripheral intra-nerve SMN transplantation in combination with NMES, which can be applied for diaphragmatic pacing, offers novel insights into the development of neuroregenerative therapies for treating life-threatening and intractable respiratory muscle paralysis caused by severe nerve damage and degenerative diseases.
Collapse
Affiliation(s)
- Yuta Asami
- Department of Human Enhancement and Hand Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan.
| | - Katsuhiro Tokutake
- Department of Human Enhancement and Hand Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Shigeru Kurimoto
- Department of Human Enhancement and Hand Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Sota Saeki
- Department of Human Enhancement and Hand Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Michiro Yamamoto
- Department of Human Enhancement and Hand Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Hitoshi Hirata
- Department of Human Enhancement and Hand Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| |
Collapse
|
2
|
Li W, Li L, Hu J, Zhou D, Su H. Design and Applications of Supramolecular Peptide Hydrogel as Artificial Extracellular Matrix. Biomacromolecules 2024. [PMID: 39418328 DOI: 10.1021/acs.biomac.4c00971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Supramolecular peptide hydrogels (SPHs) consist of peptides containing hydrogelators and functional epitopes, which can first self-assemble into nanofibers and then physically entangle together to form dynamic three-dimensional networks. Their porous structures, excellent bioactivity, and high dynamicity, similar to an extracellular matrix (ECM), have great potential in artificial ECM. The properties of the hydrogel are largely dependent on peptides. The noncovalent interactions among hydrogelators drive the formation of assemblies and further transition into hydrogels, while bioactive epitopes modulate cell-cell and cell-ECM interactions. Therefore, SPHs can support cell growth, making them ideal biomaterials for ECM mimics. This Review outlines the classical molecular design of SPHs from hydrogelators to functional epitopes and summarizes the recent advancements of SPHs as artificial ECMs in nervous system repair, wound healing, bone and cartilage regeneration, and organoid culture. This emerging SPH platform could provide an alternative strategy for developing more effective biomaterials for tissue engineering.
Collapse
Affiliation(s)
- Wenting Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Longjie Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Jiale Hu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Dongdong Zhou
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Hao Su
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| |
Collapse
|
3
|
Ho MH, Tsai YJ, Chen CY, Yang A, Burnouf T, Wang Y, Chiang YH, Hoffer BJ, Chou SY. CCL5 is essential for axonogenesis and neuronal restoration after brain injury. J Biomed Sci 2024; 31:91. [PMID: 39285280 PMCID: PMC11406852 DOI: 10.1186/s12929-024-01083-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 09/04/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) causes axon tearing and synapse degradation, resulting in multiple neurological dysfunctions and exacerbation of early neurodegeneration; the repair of axonal and synaptic structures is critical for restoring neuronal function. C-C Motif Chemokine Ligand 5 (CCL5) shows many neuroprotective activities. METHOD A close-head weight-drop system was used to induce mild brain trauma in C57BL/6 (wild-type, WT) and CCL5 knockout (CCL5-KO) mice. The mNSS score, rotarod, beam walking, and sticker removal tests were used to assay neurological function after mTBI in different groups of mice. The restoration of motor and sensory functions was impaired in CCL5-KO mice after one month of injury, with swelling of axons and synapses from Golgi staining and reduced synaptic proteins-synaptophysin and PSD95. Administration of recombinant CCL5 (Pre-treatment: 300 pg/g once before injury; or post-treatment: 30 pg/g every 2 days, since 3 days after injury for 1 month) through intranasal delivery into mouse brain improved the motor and sensory neurological dysfunctions in CCL5-KO TBI mice. RESULTS Proteomic analysis using LC-MS/MS identified that the "Nervous system development and function"-related proteins, including axonogenesis, synaptogenesis, and myelination signaling pathways, were reduced in injured cortex of CCL5-KO mice; both pre-treatment and post-treatment with CCL5 augmented those pathways. Immunostaining and western blot analysis confirmed axonogenesis and synaptogenesis related Semaphorin, Ephrin, p70S6/mTOR signaling, and myelination-related Neuregulin/ErbB and FGF/FAK signaling pathways were up-regulated in the cortical tissue by CCL5 after brain injury. We also noticed cortex redevelopment after long-term administration of CCL5 after brain injury with increased Reelin positive Cajal-Rerzius Cells and CXCR4 expression. CCL5 enhanced the growth of cone filopodia in a primary neuron culture system; blocking CCL5's receptor CCR5 by Maraviroc reduced the intensity of filopodia in growth cone and also CCL5 mediated mTOR and Rho signalling activation. Inhibiting mTOR and Rho signaling abolished CCL5 induced growth cone formation. CONCLUSIONS CCL5 plays a critical role in starting the intrinsic neuronal regeneration system following TBI, which includes growth cone formation, axonogenesis and synaptogensis, remyelination, and the subsequent proper wiring of cortical circuits. Our study underscores the potential of CCL5 as a robust therapeutic stratagem in treating axonal injury and degeneration during the chronic phase after mild brain injury.
Collapse
Affiliation(s)
- Man-Hau Ho
- Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research Institute, Taipei, 11031, Taiwan
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, 250 Wu-Xing Street, Taipei City, 11031, Taiwan
| | - Yih-Jeng Tsai
- Department of Otolaryngology Head and Neck Surgery, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, 11160, Taiwan
- School of Medicine, Fu Jen Catholic University, New Taipei City, 24352, Taiwan
| | - Chia-Yen Chen
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, 250 Wu-Xing Street, Taipei City, 11031, Taiwan
| | - Anastasia Yang
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, 250 Wu-Xing Street, Taipei City, 11031, Taiwan
- Department of Molecular and Cell Biology, University of California, Berkeley, LA, 94720, USA
| | - Thierry Burnouf
- Neuroscience Research Center, Taipei Medical University, Taipei, 11031, Taiwan
- International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan
- NeuroTMULille International Laboratory, Taipei Medical University, Taipei, 11031, Taiwan
- International PhD Program in Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Yun Wang
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli County, Zhunan, 350401, Taiwan
| | - Yung-Hsiao Chiang
- Neuroscience Research Center, Taipei Medical University, Taipei, 11031, Taiwan
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei, 11031, Taiwan
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, 11031, Taipei, Taiwan
| | - Barry J Hoffer
- Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research Institute, Taipei, 11031, Taiwan
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, 250 Wu-Xing Street, Taipei City, 11031, Taiwan
- Department of Neurosurgery, University Hospitals of Cleveland, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
- Scientist Emeritus, National Institutes of Health, Maryland, 20892, USA
| | - Szu-Yi Chou
- Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research Institute, Taipei, 11031, Taiwan.
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, 250 Wu-Xing Street, Taipei City, 11031, Taiwan.
- Neuroscience Research Center, Taipei Medical University, Taipei, 11031, Taiwan.
- NeuroTMULille International Laboratory, Taipei Medical University, Taipei, 11031, Taiwan.
- International Master Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan.
| |
Collapse
|
4
|
Romero-Ramírez L, García-Rama C, Mey J. Janus Kinase Inhibitor Brepocitinib Rescues Myelin Phagocytosis Under Inflammatory Conditions: In Vitro Evidence from Microglia and Macrophage Cell Lines. Mol Neurobiol 2024; 61:6423-6434. [PMID: 38308667 DOI: 10.1007/s12035-024-03963-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 01/16/2024] [Indexed: 02/05/2024]
Abstract
Central nervous system (CNS) injuries induce cell death and consequently the release of myelin and other cellular debris. Microglia as well as hematogenous macrophages actively collaborate to phagocyte them and undergo their degradation. However, myelin accumulation persists in the lesion site long after the injury with detrimental effects on axonal regeneration. This might be due to the presence of inhibitors of phagocytosis in the injury site. As we recently published that some proinflammatory stimuli, like interferon-γ (IFNγ) and lipopolysaccharide (LPS), inhibit myelin phagocytosis in macrophages, we have now studied the signaling pathways involved. A phagocytosis assay in Raw264.7 macrophages and N13 microglia cell lines with labeled myelin was developed with the pHrodo reagent that emits fluorescence in acidic cellular compartments (e.g.lysosomes). Pharmacological inhibition of Janus kinases (Jak) with Brepocitinib restored myelin phagocytosis and rescued the expression of genes related to phagocytosis, like triggering receptor expressed on myeloid cells 2 (TREM2), induced by IFNγ or LPS. In addition, while pharmacological inhibition of the signal transducer and activator of transcription 3 (STAT3) rescued myelin phagocytosis and the expression of phagocytosis related genes in the presence of LPS, it did not have any effect on IFNγ-treated cells. Our results show that Jak pathways participate in the inhibition of myelin phagocytosis by IFNγ and LPS. They also indicate that the resolution of inflammation is important for the clearance of cellular debris by macrophages and subsequent regenerative processes.
Collapse
Affiliation(s)
- Lorenzo Romero-Ramírez
- Laboratorio de Regeneración Neuronal, Hospital Nacional de Parapléjicos, SESCAM, Finca La Peraleda S/N, 45071, Toledo, Spain.
| | - Concepción García-Rama
- Laboratorio de Regeneración Neuronal, Hospital Nacional de Parapléjicos, SESCAM, Finca La Peraleda S/N, 45071, Toledo, Spain
| | - Jörg Mey
- Laboratorio de Regeneración Neuronal, Hospital Nacional de Parapléjicos, SESCAM, Finca La Peraleda S/N, 45071, Toledo, Spain
- School of Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
5
|
Zhai C, Wang Z, Cai J, Fang L, Li X, Jiang K, Shen Y, Wang Y, Xu X, Liu W, Wang T, Wu Q. Repeated trans-spinal magnetic stimulation promotes microglial phagocytosis of myelin debris after spinal cord injury through LRP-1. Exp Neurol 2024; 379:114844. [PMID: 38830500 DOI: 10.1016/j.expneurol.2024.114844] [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: 02/20/2024] [Revised: 05/07/2024] [Accepted: 05/28/2024] [Indexed: 06/05/2024]
Abstract
Spinal cord injury (SCI) is a serious trauma of the central nervous system. The clearance of myelin debris is a critical step in the functional recovery following spinal cord injury (SCI). Recent studies have begun to reveal critical roles for professional phagocytes in the central nervous system, microglia, and their receptors in the control of myelin debris in neurodegenerative disease. Repeated trans-spinal magnetic stimulation (rTSMS) has been demonstrated as a noninvasive SCI treatment that enhances tissue repair and functional recovery. In this study, we investigated the role and molecular mechanism of rTSMS on microglial phagocytosis of myelin debris in a rat SCI model. In our studies, we found that rTSMS significantly promoted the motor function recovery of SCI rats associated with the inhibition the neuroinflammation and glia scar formation. Immunofluorescence results further showed that the rTSMS promotes the clearance of myelin debris by microglia in vivo and in vitro. Additionally, receptor-associated protein (RAP), a Low-density lipoprotein receptor-related protein-1 (LRP-1) inhibitor, could cancel the accelerated microglial phagocytosis of myelin debris after rTSMS in vitro experiments. Simultaneously, Elisa's results and western blotting respectively showed that rTSMS significantly decreased the levels of soluble LRP-1(sLRP-1) and the LRP-1 splicing enzyme of ADAM17. In conclusion, rTSMS could promote the clearance of myelin debris by microglia through LRP-1 to improve the functional recovery of SCI rats.
Collapse
Affiliation(s)
- Chenyuan Zhai
- Rehabilitation Medicine Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Zun Wang
- Rehabilitation Medicine Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; Rehabilitation medicine department, School of Acupuncture and Tuina, School of Health and Rehabilitation, Nanjing university of Chinese medicine, Nanjing 210023, China
| | - Jili Cai
- Rehabilitation Medicine Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Lu Fang
- Rehabilitation Medicine Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xiangzhe Li
- Rehabilitation Medicine Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; Rehabilitation Medicine Center, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, Jiangsu 215153, China
| | - Kunmao Jiang
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Ying Shen
- Rehabilitation Medicine Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yu Wang
- Rehabilitation Medicine Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xingjun Xu
- Department of Rehabilitation, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Wentao Liu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu 211166, China.
| | - Tong Wang
- Rehabilitation Medicine Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| | - Qi Wu
- Department of Rehabilitation, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang 421000, China.
| |
Collapse
|
6
|
Wang X, Cheng Z, Tai W, Shi M, Ayazi M, Liu Y, Sun L, Yu C, Fan Z, Guo B, He X, Sun D, Young W, Ren Y. Targeting foamy macrophages by manipulating ABCA1 expression to facilitate lesion healing in the injured spinal cord. Brain Behav Immun 2024; 119:431-453. [PMID: 38636566 DOI: 10.1016/j.bbi.2024.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 04/03/2024] [Accepted: 04/16/2024] [Indexed: 04/20/2024] Open
Abstract
Spinal cord injury (SCI) triggers a complex cascade of events, including myelin loss, neuronal damage, neuroinflammation, and the accumulation of damaged cells and debris at the injury site. Infiltrating bone marrow derived macrophages (BMDMϕ) migrate to the epicenter of the SCI lesion, where they engulf cell debris including abundant myelin debris to become pro-inflammatory foamy macrophages (foamy Mϕ), participate neuroinflammation, and facilitate the progression of SCI. This study aimed to elucidate the cellular and molecular mechanisms underlying the functional changes in foamy Mϕ and their potential implications for SCI. Contusion at T10 level of the spinal cord was induced using a New York University (NYU) impactor (5 g rod from a height of 6.25 mm) in male mice. ABCA1, an ATP-binding cassette transporter expressed by Mϕ, plays a crucial role in lipid efflux from foamy cells. We observed that foamy Mϕ lacking ABCA1 exhibited increased lipid accumulation and a higher presence of lipid-accumulated foamy Mϕ as well as elevated pro-inflammatory response in vitro and in injured spinal cord. We also found that both genetic and pharmacological enhancement of ABCA1 expression accelerated lipid efflux from foamy Mϕ, reduced lipid accumulation and inhibited the pro-inflammatory response of foamy Mϕ, and accelerated clearance of cell debris and necrotic cells, which resulted in functional recovery. Our study highlights the importance of understanding the pathologic role of foamy Mϕ in SCI progression and the potential of ABCA1 as a therapeutic target for modulating the inflammatory response, promoting lipid metabolism, and facilitating functional recovery in SCI.
Collapse
Affiliation(s)
- Xi Wang
- W. M. Keck Center for Collaborative Neuroscience, Rutgers, State University of New Jersey, New Brunswick, NJ 08854, USA; Institute of Neurosciences, Fourth Military Medical University, Xi'an 710032, China; College of Life Sciences and Medicine, Northwest University, Xi'an 710069, China.
| | - Zhijian Cheng
- Department of Biomedical Sciences, Florida State University College of Medicine, 1115 West Call Street, Tallahassee, FL 32306, USA
| | - Wenjiao Tai
- Department of Biomedical Sciences, Florida State University College of Medicine, 1115 West Call Street, Tallahassee, FL 32306, USA
| | - Mingjun Shi
- Department of Biomedical Sciences, Florida State University College of Medicine, 1115 West Call Street, Tallahassee, FL 32306, USA
| | - Maryam Ayazi
- Department of Biomedical Sciences, Florida State University College of Medicine, 1115 West Call Street, Tallahassee, FL 32306, USA
| | - Yang Liu
- Department of Biomedical Sciences, Florida State University College of Medicine, 1115 West Call Street, Tallahassee, FL 32306, USA
| | - Li Sun
- Department of Biomedical Sciences, Florida State University College of Medicine, 1115 West Call Street, Tallahassee, FL 32306, USA
| | - Caiyong Yu
- Institute of Neurosciences, Fourth Military Medical University, Xi'an 710032, China
| | - Zhongmin Fan
- Department of Critical Care Medicine and Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Xi'an 710032, China
| | - Bin Guo
- Department of Pathology, Guizhou Medical University, Guiyang 550025, China
| | - Xijing He
- Department of Orthopedics, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Dongming Sun
- W. M. Keck Center for Collaborative Neuroscience, Rutgers, State University of New Jersey, New Brunswick, NJ 08854, USA
| | - Wise Young
- W. M. Keck Center for Collaborative Neuroscience, Rutgers, State University of New Jersey, New Brunswick, NJ 08854, USA
| | - Yi Ren
- W. M. Keck Center for Collaborative Neuroscience, Rutgers, State University of New Jersey, New Brunswick, NJ 08854, USA; Department of Biomedical Sciences, Florida State University College of Medicine, 1115 West Call Street, Tallahassee, FL 32306, USA.
| |
Collapse
|
7
|
Hou X, Qu X, Chen W, Sang X, Ye Y, Wang C, Guo Y, Shi H, Yang C, Zhu K, Zhang Y, Xu H, Lv L, Zhang D, Hou L. CD36 deletion prevents white matter injury by modulating microglia polarization through the Traf5-MAPK signal pathway. J Neuroinflammation 2024; 21:148. [PMID: 38840180 PMCID: PMC11155181 DOI: 10.1186/s12974-024-03143-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 05/27/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND White matter injury (WMI) represents a significant etiological factor contributing to neurological impairment subsequent to Traumatic Brain Injury (TBI). CD36 receptors are recognized as pivotal participants in the pathogenesis of neurological disorders, including stroke and spinal cord injury. Furthermore, dynamic fluctuations in the phenotypic polarization of microglial cells have been intimately associated with the regenerative processes within the injured tissue following TBI. Nevertheless, there is a paucity of research addressing the impact of CD36 receptors on WMI and microglial polarization. This investigation aims to elucidate the functional role and mechanistic underpinnings of CD36 in modulating microglial polarization and WMI following TBI. METHODS TBI models were induced in murine subjects via controlled cortical impact (CCI). The spatiotemporal patterns of CD36 expression were examined through quantitative polymerase chain reaction (qPCR), Western blot analysis, and immunofluorescence staining. The extent of white matter injury was assessed via transmission electron microscopy, Luxol Fast Blue (LFB) staining, and immunofluorescence staining. Transcriptome sequencing was employed to dissect the molecular mechanisms underlying CD36 down-regulation and its influence on white matter damage. Microglial polarization status was ascertained using qPCR, Western blot analysis, and immunofluorescence staining. In vitro, a Transwell co-culture system was employed to investigate the impact of CD36-dependent microglial polarization on oligodendrocytes subjected to oxygen-glucose deprivation (OGD). RESULTS Western blot and qPCR analyses revealed that CD36 expression reached its zenith at 7 days post-TBI and remained sustained at this level thereafter. Immunofluorescence staining exhibited robust CD36 expression in astrocytes and microglia following TBI. Genetic deletion of CD36 ameliorated TBI-induced white matter injury, as evidenced by a reduced SMI-32/MBP ratio and G-ratio. Transcriptome sequencing unveiled differentially expressed genes enriched in processes linked to microglial activation, regulation of neuroinflammation, and the TNF signaling pathway. Additionally, bioinformatics analysis pinpointed the Traf5-p38 axis as a critical signaling pathway. In vivo and in vitro experiments indicated that inhibition of the CD36-Traf5-MAPK axis curtailed microglial polarization toward the pro-inflammatory phenotype. In a Transwell co-culture system, BV2 cells treated with LPS + IFN-γ exacerbated the damage of post-OGD oligodendrocytes, which could be rectified through CD36 knockdown in BV2 cells. CONCLUSIONS This study illuminates that the suppression of CD36 mitigates WMI by constraining microglial polarization towards the pro-inflammatory phenotype through the down-regulation of the Traf5-MAPK signaling pathway. Our findings present a potential therapeutic strategy for averting neuroinflammatory responses and ensuing WMI damage resulting from TBI.
Collapse
Affiliation(s)
- Xiaoxiang Hou
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, 415 Fengyang Rd, Shanghai, China
| | - Xiaolin Qu
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, China
| | - Wen Chen
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, 415 Fengyang Rd, Shanghai, China
| | - Xianzheng Sang
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, 415 Fengyang Rd, Shanghai, China
| | - Yichao Ye
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, 415 Fengyang Rd, Shanghai, China
| | - Chengqing Wang
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, 415 Fengyang Rd, Shanghai, China
| | - Yangu Guo
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, 415 Fengyang Rd, Shanghai, China
| | - Hantong Shi
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, 415 Fengyang Rd, Shanghai, China
| | - Chengzi Yang
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, 415 Fengyang Rd, Shanghai, China
| | - Kaixin Zhu
- Department of Neurosurgery, The First Naval Hospital of Southern Theater Command, Zhanjiang, China
| | - Yelei Zhang
- Department of Neurosurgery, Xishan People's Hospital of Wuxi City, Wuxi, China
| | - Haoxiang Xu
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, 415 Fengyang Rd, Shanghai, China
| | - Liquan Lv
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, 415 Fengyang Rd, Shanghai, China
| | - Danfeng Zhang
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, 415 Fengyang Rd, Shanghai, China.
| | - Lijun Hou
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, 415 Fengyang Rd, Shanghai, China.
| |
Collapse
|
8
|
Talsma AD, Niemi JP, Zigmond RE. Neither injury induced macrophages within the nerve, nor the environment created by Wallerian degeneration is necessary for enhanced in vivo axon regeneration after peripheral nerve injury. J Neuroinflammation 2024; 21:134. [PMID: 38802868 PMCID: PMC11131297 DOI: 10.1186/s12974-024-03132-5] [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: 04/23/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Since the 1990s, evidence has accumulated that macrophages promote peripheral nerve regeneration and are required for enhancing regeneration in the conditioning lesion (CL) response. After a sciatic nerve injury, macrophages accumulate in the injury site, the nerve distal to that site, and the axotomized dorsal root ganglia (DRGs). In the peripheral nervous system, as in other tissues, the macrophage response is derived from both resident macrophages and recruited monocyte-derived macrophages (MDMs). Unresolved questions are: at which sites do macrophages enhance nerve regeneration, and is a particular population needed. METHODS Ccr2 knock-out (KO) and Ccr2gfp/gfp knock-in/KO mice were used to prevent MDM recruitment. Using these strains in a sciatic CL paradigm, we examined the necessity of MDMs and residents for CL-enhanced regeneration in vivo and characterized injury-induced nerve inflammation. CL paradigm variants, including the addition of pharmacological macrophage depletion methods, tested the role of various macrophage populations in initiating or sustaining the CL response. In vivo regeneration, measured from bilateral proximal test lesions (TLs) after 2 d, and macrophages were quantified by immunofluorescent staining. RESULTS Peripheral CL-enhanced regeneration was equivalent between crush and transection CLs and was sustained for 28 days in both Ccr2 KO and WT mice despite MDM depletion. Similarly, the central CL response measured in dorsal roots was unchanged in Ccr2 KO mice. Macrophages at both the TL and CL, but not between them, stained for the pro-regenerative marker, arginase 1. TL macrophages were primarily CCR2-dependent MDMs and nearly absent in Ccr2 KO and Ccr2gfp/gfp KO mice. However, there were only slightly fewer Arg1+ macrophages in CCR2 null CLs than controls due to resident macrophage compensation. Zymosan injection into an intact WT sciatic nerve recruited Arg1+ macrophages but did not enhance regeneration. Finally, clodronate injection into Ccr2gfp KO CLs dramatically reduced CL macrophages. Combined with the Ccr2gfp KO background, depleting MDMs and TL macrophages, and a transection CL, physically removing the distal nerve environment, nearly all macrophages in the nerve were removed, yet CL-enhanced regeneration was not impaired. CONCLUSIONS Macrophages in the sciatic nerve are neither necessary nor sufficient to produce a CL response.
Collapse
Affiliation(s)
- Aaron D Talsma
- Department of Neurosciences, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106-4975, USA
| | - Jon P Niemi
- Department of Neurosciences, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106-4975, USA
| | - Richard E Zigmond
- Department of Neurosciences, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106-4975, USA.
| |
Collapse
|
9
|
Fischer G, Bättig L, Stienen MN, Curt A, Fehlings MG, Hejrati N. Advancements in neuroregenerative and neuroprotective therapies for traumatic spinal cord injury. Front Neurosci 2024; 18:1372920. [PMID: 38812974 PMCID: PMC11133582 DOI: 10.3389/fnins.2024.1372920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 04/10/2024] [Indexed: 05/31/2024] Open
Abstract
Traumatic spinal cord injuries (SCIs) continue to be a major healthcare concern, with a rising prevalence worldwide. In response to this growing medical challenge, considerable scientific attention has been devoted to developing neuroprotective and neuroregenerative strategies aimed at improving the prognosis and quality of life for individuals with SCIs. This comprehensive review aims to provide an up-to-date and thorough overview of the latest neuroregenerative and neuroprotective therapies currently under investigation. These strategies encompass a multifaceted approach that include neuropharmacological interventions, cell-based therapies, and other promising strategies such as biomaterial scaffolds and neuro-modulation therapies. In addition, the review discusses the importance of acute clinical management, including the role of hemodynamic management as well as timing and technical aspects of surgery as key factors mitigating the secondary injury following SCI. In conclusion, this review underscores the ongoing scientific efforts to enhance patient outcomes and quality of life, focusing on upcoming strategies for the management of traumatic SCI. Each section provides a working knowledge of the fundamental preclinical and patient trials relevant to clinicians while underscoring the pathophysiologic rationale for the therapies.
Collapse
Affiliation(s)
- Gregor Fischer
- Department of Neurosurgery, Cantonal Hospital St.Gallen, Medical School of St.Gallen, St.Gallen, Switzerland
- Spine Center of Eastern Switzerland, Cantonal Hospital St.Gallen, Medical School of St.Gallen, St.Gallen, Switzerland
| | - Linda Bättig
- Department of Neurosurgery, Cantonal Hospital St.Gallen, Medical School of St.Gallen, St.Gallen, Switzerland
- Spine Center of Eastern Switzerland, Cantonal Hospital St.Gallen, Medical School of St.Gallen, St.Gallen, Switzerland
| | - Martin N. Stienen
- Department of Neurosurgery, Cantonal Hospital St.Gallen, Medical School of St.Gallen, St.Gallen, Switzerland
- Spine Center of Eastern Switzerland, Cantonal Hospital St.Gallen, Medical School of St.Gallen, St.Gallen, Switzerland
| | - Armin Curt
- Spinal Cord Injury Center, University Hospital Balgrist, Zurich, Switzerland
| | - Michael G. Fehlings
- Division of Neurosurgery and Spine Program, Department of Surgery, University of Toronto, Toronto, ON, Canada
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Nader Hejrati
- Department of Neurosurgery, Cantonal Hospital St.Gallen, Medical School of St.Gallen, St.Gallen, Switzerland
- Spine Center of Eastern Switzerland, Cantonal Hospital St.Gallen, Medical School of St.Gallen, St.Gallen, Switzerland
| |
Collapse
|
10
|
Sun Z, Chen Z, Yin M, Wu X, Guo B, Cheng X, Quan R, Sun Y, Zhang Q, Fan Y, Jin C, Yin Y, Hou X, Liu W, Shu M, Xue X, Shi Y, Chen B, Xiao Z, Dai J, Zhao Y. Harnessing developmental dynamics of spinal cord extracellular matrix improves regenerative potential of spinal cord organoids. Cell Stem Cell 2024; 31:772-787.e11. [PMID: 38565140 DOI: 10.1016/j.stem.2024.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 02/07/2024] [Accepted: 03/11/2024] [Indexed: 04/04/2024]
Abstract
Neonatal spinal cord tissues exhibit remarkable regenerative capabilities as compared to adult spinal cord tissues after injury, but the role of extracellular matrix (ECM) in this process has remained elusive. Here, we found that early developmental spinal cord had higher levels of ECM proteins associated with neural development and axon growth, but fewer inhibitory proteoglycans, compared to those of adult spinal cord. Decellularized spinal cord ECM from neonatal (DNSCM) and adult (DASCM) rabbits preserved these differences. DNSCM promoted proliferation, migration, and neuronal differentiation of neural progenitor cells (NPCs) and facilitated axonal outgrowth and regeneration of spinal cord organoids more effectively than DASCM. Pleiotrophin (PTN) and Tenascin (TNC) in DNSCM were identified as contributors to these abilities. Furthermore, DNSCM demonstrated superior performance as a delivery vehicle for NPCs and organoids in spinal cord injury (SCI) models. This suggests that ECM cues from early development stages might significantly contribute to the prominent regeneration ability in spinal cord.
Collapse
Affiliation(s)
- Zheng Sun
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhenni Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Man Yin
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xianming Wu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Bo Guo
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiaokang Cheng
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Rui Quan
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuting Sun
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qi Zhang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yongheng Fan
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chen Jin
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yanyun Yin
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xianglin Hou
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Weiyuan Liu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Muya Shu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiaoyu Xue
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Ya Shi
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Bing Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhifeng Xiao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jianwu Dai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China.
| | - Yannan Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
11
|
Ramanujan A, Li Z, Ma Y, Lin Z, Ibáñez CF. RhoGDI phosphorylation by PKC promotes its interaction with death receptor p75 NTR to gate axon growth and neuron survival. EMBO Rep 2024; 25:1490-1512. [PMID: 38253689 PMCID: PMC10933337 DOI: 10.1038/s44319-024-00064-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/21/2023] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
How receptors juggle their interactions with multiple downstream effectors remains poorly understood. Here we show that the outcome of death receptor p75NTR signaling is determined through competition of effectors for interaction with its intracellular domain, in turn dictated by the nature of the ligand. While NGF induces release of RhoGDI through recruitment of RIP2, thus decreasing RhoA activity in favor of NFkB signaling, MAG induces PKC-mediated phosphorylation of the RhoGDI N-terminus, promoting its interaction with the juxtamembrane domain of p75NTR, disengaging RIP2, and enhancing RhoA activity in detriment of NF-kB. This results in stunted neurite outgrowth and apoptosis in cerebellar granule neurons. If presented simultaneously, MAG prevails over NGF. The NMR solution structure of the complex between the RhoGDI N-terminus and p75NTR juxtamembrane domain reveals previously unknown structures of these proteins and clarifies the mechanism of p75NTR activation. These results show how ligand-directed competition between RIP2 and RhoGDI for p75NTR engagement determine axon growth and neuron survival. Similar principles are likely at work in other receptors engaging multiple effectors and signaling pathways.
Collapse
Affiliation(s)
- Ajeena Ramanujan
- Department of Physiology and Life Sciences Institute, National University of Singapore, 117456, Singapore, Singapore
| | - Zhen Li
- Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, School of Life Sciences, Tianjin University, Tianjin, 300072, China
| | - Yanchen Ma
- Peking University School of Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, 100871, Beijing, China
- Chinese Institute for Brain Research, Life Science Park, 102206, Beijing, China
| | - Zhi Lin
- Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, School of Life Sciences, Tianjin University, Tianjin, 300072, China
| | - Carlos F Ibáñez
- Department of Physiology and Life Sciences Institute, National University of Singapore, 117456, Singapore, Singapore.
- Peking University School of Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, 100871, Beijing, China.
- Chinese Institute for Brain Research, Life Science Park, 102206, Beijing, China.
- Department of Neuroscience, Karolinska Institute, Stockholm, 17177, Sweden.
- Stellenbosch Institute for Advanced Study, Wallenberg Research Centre at Stellenbosch University, Stellenbosch, 7600, South Africa.
| |
Collapse
|
12
|
Chen L, Zhang S, Duan Y, Song X, Chang M, Feng W, Chen Y. Silicon-containing nanomedicine and biomaterials: materials chemistry, multi-dimensional design, and biomedical application. Chem Soc Rev 2024; 53:1167-1315. [PMID: 38168612 DOI: 10.1039/d1cs01022k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
The invention of silica-based bioactive glass in the late 1960s has sparked significant interest in exploring a wide range of silicon-containing biomaterials from the macroscale to the nanoscale. Over the past few decades, these biomaterials have been extensively explored for their potential in diverse biomedical applications, considering their remarkable bioactivity, excellent biocompatibility, facile surface functionalization, controllable synthesis, etc. However, to expedite the clinical translation and the unexpected utilization of silicon-composed nanomedicine and biomaterials, it is highly desirable to achieve a thorough comprehension of their characteristics and biological effects from an overall perspective. In this review, we provide a comprehensive discussion on the state-of-the-art progress of silicon-composed biomaterials, including their classification, characteristics, fabrication methods, and versatile biomedical applications. Additionally, we highlight the multi-dimensional design of both pure and hybrid silicon-composed nanomedicine and biomaterials and their intrinsic biological effects and interactions with biological systems. Their extensive biomedical applications span from drug delivery and bioimaging to therapeutic interventions and regenerative medicine, showcasing the significance of their rational design and fabrication to meet specific requirements and optimize their theranostic performance. Additionally, we offer insights into the future prospects and potential challenges regarding silicon-composed nanomedicine and biomaterials. By shedding light on these exciting research advances, we aspire to foster further progress in the biomedical field and drive the development of innovative silicon-composed nanomedicine and biomaterials with transformative applications in biomedicine.
Collapse
Affiliation(s)
- Liang Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Shanshan Zhang
- Department of Ultrasound Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P. R. China
| | - Yanqiu Duan
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, P. R. China.
| | - Xinran Song
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Meiqi Chang
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, P. R. China.
| | - Wei Feng
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| |
Collapse
|
13
|
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
|
14
|
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
|
15
|
Qin C, Qi Z, Pan S, Xia P, Kong W, Sun B, Du H, Zhang R, Zhu L, Zhou D, Yang X. Advances in Conductive Hydrogel for Spinal Cord Injury Repair and Regeneration. Int J Nanomedicine 2023; 18:7305-7333. [PMID: 38084124 PMCID: PMC10710813 DOI: 10.2147/ijn.s436111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 11/08/2023] [Indexed: 12/18/2023] Open
Abstract
Spinal cord injury (SCI) treatment represents a major challenge in clinical practice. In recent years, the rapid development of neural tissue engineering technology has provided a new therapeutic approach for spinal cord injury repair. Implanting functionalized electroconductive hydrogels (ECH) in the injury area has been shown to promote axonal regeneration and facilitate the generation of neuronal circuits by reshaping the microenvironment of SCI. ECH not only facilitate intercellular electrical signaling but, when combined with electrical stimulation, enable the transmission of electrical signals to electroactive tissue and activate bioelectric signaling pathways, thereby promoting neural tissue repair. Therefore, the implantation of ECH into damaged tissues can effectively restore physiological functions related to electrical conduction. This article focuses on the dynamic pathophysiological changes in the SCI microenvironment and discusses the mechanisms of electrical stimulation/signal in the process of SCI repair. By examining electrical activity during nerve repair, we provide insights into the mechanisms behind electrical stimulation and signaling during SCI repair. We classify conductive biomaterials, and offer an overview of the current applications and research progress of conductive hydrogels in spinal cord repair and regeneration, aiming to provide a reference for future explorations and developments in spinal cord regeneration strategies.
Collapse
Affiliation(s)
- Cheng Qin
- Department of Orthopedic Surgery, the Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
| | - Zhiping Qi
- Department of Orthopedic Surgery, the Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
| | - Su Pan
- Department of Orthopedic Surgery, the Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
| | - Peng Xia
- Department of Orthopedic Surgery, the Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
| | - Weijian Kong
- Department of Orthopedic Surgery, the Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
| | - Bin Sun
- Department of Orthopedic Surgery, the Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
| | - Haorui Du
- Department of Orthopedic Surgery, the Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
| | - Renfeng Zhang
- Department of Orthopedic Surgery, the Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
| | - Longchuan Zhu
- Department of Orthopedic Surgery, the Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
| | - Dinghai Zhou
- Department of Orthopedic Surgery, the Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
| | - Xiaoyu Yang
- Department of Orthopedic Surgery, the Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
| |
Collapse
|
16
|
Chambel SS, Cruz CD. Axonal growth inhibitors and their receptors in spinal cord injury: from biology to clinical translation. Neural Regen Res 2023; 18:2573-2581. [PMID: 37449592 DOI: 10.4103/1673-5374.373674] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023] Open
Abstract
Axonal growth inhibitors are released during traumatic injuries to the adult mammalian central nervous system, including after spinal cord injury. These molecules accumulate at the injury site and form a highly inhibitory environment for axonal regeneration. Among these inhibitory molecules, myelin-associated inhibitors, including neurite outgrowth inhibitor A, oligodendrocyte myelin glycoprotein, myelin-associated glycoprotein, chondroitin sulfate proteoglycans and repulsive guidance molecule A are of particular importance. Due to their inhibitory nature, they represent exciting molecular targets to study axonal inhibition and regeneration after central injuries. These molecules are mainly produced by neurons, oligodendrocytes, and astrocytes within the scar and in its immediate vicinity. They exert their effects by binding to specific receptors, localized in the membranes of neurons. Receptors for these inhibitory cues include Nogo receptor 1, leucine-rich repeat, and Ig domain containing 1 and p75 neurotrophin receptor/tumor necrosis factor receptor superfamily member 19 (that form a receptor complex that binds all myelin-associated inhibitors), and also paired immunoglobulin-like receptor B. Chondroitin sulfate proteoglycans and repulsive guidance molecule A bind to Nogo receptor 1, Nogo receptor 3, receptor protein tyrosine phosphatase σ and leucocyte common antigen related phosphatase, and neogenin, respectively. Once activated, these receptors initiate downstream signaling pathways, the most common amongst them being the RhoA/ROCK signaling pathway. These signaling cascades result in actin depolymerization, neurite outgrowth inhibition, and failure to regenerate after spinal cord injury. Currently, there are no approved pharmacological treatments to overcome spinal cord injuries other than physical rehabilitation and management of the array of symptoms brought on by spinal cord injuries. However, several novel therapies aiming to modulate these inhibitory proteins and/or their receptors are under investigation in ongoing clinical trials. Investigation has also been demonstrating that combinatorial therapies of growth inhibitors with other therapies, such as growth factors or stem-cell therapies, produce stronger results and their potential application in the clinics opens new venues in spinal cord injury treatment.
Collapse
Affiliation(s)
- Sílvia Sousa Chambel
- Experimental Biology Unit, Department of Biomedicine, Faculty of Medicine of Porto; Translational NeuroUrology, Instituto de Investigação e Inovação em Saúde-i3S and IBMC, Universidade do Porto, Porto, Portugal
| | - Célia Duarte Cruz
- Experimental Biology Unit, Department of Biomedicine, Faculty of Medicine of Porto; Translational NeuroUrology, Instituto de Investigação e Inovação em Saúde-i3S and IBMC, Universidade do Porto, Porto, Portugal
| |
Collapse
|
17
|
Takahashi Y, Kajitani T, Endo T, Nakayashiki A, Inoue T, Niizuma K, Tominaga T. Intravenous Administration of Human Muse Cells Ameliorates Deficits in a Rat Model of Subacute Spinal Cord Injury. Int J Mol Sci 2023; 24:14603. [PMID: 37834052 PMCID: PMC10572998 DOI: 10.3390/ijms241914603] [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: 08/25/2023] [Revised: 09/21/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Multilineage-differentiating stress-enduring (Muse) cells are newly established pluripotent stem cells. The aim of the present study was to examine the potential of the systemic administration of Muse cells as an effective treatment for subacute SCI. We intravenously administered the clinical product "CL2020" containing Muse cells to a rat model two weeks after mid-thoracic spinal cord contusion. Eight experimental animals received CL2020, and twelve received the vehicle. Behavioral analyses were conducted over 20 weeks. Histological evaluations were performed. After 20 weeks of observation, diphtheria toxin was administered to three CL2020-treated animals to selectively ablate human cell functions. Hindlimb motor functions significantly improved from 6 to 20 weeks after the administration of CL2020. The cystic cavity was smaller in the CL2020 group. Furthermore, larger numbers of descending 5-HT fibers were preserved in the distal spinal cord. Muse cells in CL2020 were considered to have differentiated into neuronal and neural cells in the injured spinal cord. Neuronal and neural cells were identified in the gray and white matter, respectively. Importantly, these effects were reversed by the selective ablation of human cells by diphtheria toxin. Intravenously administered Muse cells facilitated the therapeutic potential of CL2020 for severe subacute spinal cord injury.
Collapse
Affiliation(s)
- Yoshiharu Takahashi
- Department of Neurosurgery, Graduate School of Medicine, Tohoku University, Sendai 980-8572, Japan; (Y.T.); (A.N.)
- Department of Neurosurgery, Tohoku Medical and Pharmaceutical University, Sendai 981-8558, Japan
| | - Takumi Kajitani
- Department of Neurosurgery, Graduate School of Medicine, Tohoku University, Sendai 980-8572, Japan; (Y.T.); (A.N.)
| | - Toshiki Endo
- Department of Neurosurgery, Graduate School of Medicine, Tohoku University, Sendai 980-8572, Japan; (Y.T.); (A.N.)
- Department of Neurosurgery, Tohoku Medical and Pharmaceutical University, Sendai 981-8558, Japan
| | - Atsushi Nakayashiki
- Department of Neurosurgery, Graduate School of Medicine, Tohoku University, Sendai 980-8572, Japan; (Y.T.); (A.N.)
| | - Tomoo Inoue
- Department of Neurosurgery, Saitama Red Cross Hospital, Saitama 330-8553, Japan;
| | - Kuniyasu Niizuma
- Department of Neurosurgery, Graduate School of Medicine, Tohoku University, Sendai 980-8572, Japan; (Y.T.); (A.N.)
- Department of Neurosurgical Engineering and Translational Neuroscience, Graduate School of Medicine, Tohoku University, Sendai 980-8576, Japan
- Department of Neurosurgical Engineering and Translational Neuroscience, Graduate School of Biomedical Engineering, Tohoku University, Sendai 980-8572, Japan
| | - Teiji Tominaga
- Department of Neurosurgery, Graduate School of Medicine, Tohoku University, Sendai 980-8572, Japan; (Y.T.); (A.N.)
| |
Collapse
|
18
|
Schaeffer J, Vilallongue N, Decourt C, Blot B, El Bakdouri N, Plissonnier E, Excoffier B, Paccard A, Diaz JJ, Humbert S, Catez F, Saudou F, Nawabi H, Belin S. Customization of the translational complex regulates mRNA-specific translation to control CNS regeneration. Neuron 2023; 111:2881-2898.e12. [PMID: 37442131 PMCID: PMC10522804 DOI: 10.1016/j.neuron.2023.06.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 03/30/2023] [Accepted: 06/15/2023] [Indexed: 07/15/2023]
Abstract
In the adult mammalian central nervous system (CNS), axons fail to regenerate spontaneously after injury because of a combination of extrinsic and intrinsic factors. Despite recent advances targeting the intrinsic regenerative properties of adult neurons, the molecular mechanisms underlying axon regeneration are not fully understood. Here, we uncover a regulatory mechanism that controls the expression of key proteins involved in regeneration at the translational level. Our results show that mRNA-specific translation is critical for promoting axon regeneration. Indeed, we demonstrate that specific ribosome-interacting proteins, such as the protein Huntingtin (HTT), selectively control the translation of a specific subset of mRNAs. Moreover, modulating the expression of these translationally regulated mRNAs is crucial for promoting axon regeneration. Altogether, our findings highlight that selective translation through the customization of the translational complex is a key mechanism of axon regeneration with major implications in the development of therapeutic strategies for CNS repair.
Collapse
Affiliation(s)
- Julia Schaeffer
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Noemie Vilallongue
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Charlotte Decourt
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Beatrice Blot
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Nacera El Bakdouri
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Elise Plissonnier
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Blandine Excoffier
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Antoine Paccard
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Jean-Jacques Diaz
- Inserm U1052, CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Centre Léon Bérard, 69008 Lyon, France; Université de Lyon 1, 69000 Lyon, France
| | - Sandrine Humbert
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Frederic Catez
- Inserm U1052, CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, 69000 Lyon, France; Centre Léon Bérard, 69008 Lyon, France; Université de Lyon 1, 69000 Lyon, France
| | - Frederic Saudou
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Homaira Nawabi
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France.
| | - Stephane Belin
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France.
| |
Collapse
|
19
|
Siddiq MM, Johnson NP, Zorina Y, Yadaw AS, Toro CA, Hansen J, Rabinovich V, Gregorich SM, Xiong Y, Tolentino RE, Hannila SS, Kaplan E, Blitzer RD, Filbin MT, Cardozo CP, Passaglia CL, Iyengar R. A spatially specified systems pharmacology therapy for axonal recovery after injury. Front Pharmacol 2023; 14:1225759. [PMID: 37799971 PMCID: PMC10547904 DOI: 10.3389/fphar.2023.1225759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 09/06/2023] [Indexed: 10/07/2023] Open
Abstract
There are no known drugs or drug combinations that promote substantial central nervous system axonal regeneration after injury. We used systems pharmacology approaches to model pathways underlying axonal growth and identify a four-drug combination that regulates multiple subcellular processes in the cell body and axons using the optic nerve crush model in rats. We intravitreally injected agonists HU-210 (cannabinoid receptor-1) and IL-6 (interleukin 6 receptor) to stimulate retinal ganglion cells for axonal growth. We applied, in gel foam at the site of nerve injury, Taxol to stabilize growing microtubules, and activated protein C to clear the debris field since computational models predicted that this drug combination regulating two subcellular processes at the growth cone produces synergistic growth. Physiologically, drug treatment restored or preserved pattern electroretinograms and some of the animals had detectable visual evoked potentials in the brain and behavioral optokinetic responses. Morphology experiments show that the four-drug combination protects axons or promotes axonal regrowth to the optic chiasm and beyond. We conclude that spatially targeted drug treatment is therapeutically relevant and can restore limited functional recovery.
Collapse
Affiliation(s)
- Mustafa M. Siddiq
- Department of Pharmacological Sciences, Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Nicholas P. Johnson
- Department of Pharmacological Sciences, Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Departments of Chemical and Biomedical Engineering, University of South Florida, Tampa, FL, United States
- National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters VA Medical Center, New York, NY, United States
| | - Yana Zorina
- Department of Pharmacological Sciences, Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Arjun Singh Yadaw
- Department of Pharmacological Sciences, Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Carlos A. Toro
- National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters VA Medical Center, New York, NY, United States
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jens Hansen
- Department of Pharmacological Sciences, Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Vera Rabinovich
- Department of Pharmacological Sciences, Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Sarah M. Gregorich
- Departments of Chemical and Biomedical Engineering, University of South Florida, Tampa, FL, United States
| | - Yuguang Xiong
- Department of Pharmacological Sciences, Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Rosa E. Tolentino
- Department of Pharmacological Sciences, Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Sari S. Hannila
- Department of Human Anatomy and Cell Science, Basic Medical Sciences Building, Winnipeg, NM, United States
| | - Ehud Kaplan
- Department of Pharmacological Sciences, Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Philosophy of Science, Prague and the National Institute of Mental Health, Charles University, Prague, CZ, United States
| | - Robert D. Blitzer
- Department of Pharmacological Sciences, Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Marie T. Filbin
- Department of Biological Sciences, Hunter College, City University of New York, New York, NY, United States
| | - Christopher P. Cardozo
- National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters VA Medical Center, New York, NY, United States
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Rehabilitation Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Christopher L. Passaglia
- Departments of Chemical and Biomedical Engineering, University of South Florida, Tampa, FL, United States
| | - Ravi Iyengar
- Department of Pharmacological Sciences, Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
20
|
Siddiq MM, Toro CA, Johnson NP, Hansen J, Xiong Y, Mellado W, Tolentino RE, Johnson K, Jayaraman G, Suhail Z, Harlow L, Dai J, Beaumont KG, Sebra R, Willis DE, Cardozo CP, Iyengar R. Spinal cord injury regulates circular RNA expression in axons. Front Mol Neurosci 2023; 16:1183315. [PMID: 37692100 PMCID: PMC10483835 DOI: 10.3389/fnmol.2023.1183315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 07/04/2023] [Indexed: 09/12/2023] Open
Abstract
Introduction Neurons transport mRNA and translational machinery to axons for local translation. After spinal cord injury (SCI), de novo translation is assumed to enable neurorepair. Knowledge of the identity of axonal mRNAs that participate in neurorepair after SCI is limited. We sought to identify and understand how axonal RNAs play a role in axonal regeneration. Methods We obtained preparations enriched in axonal mRNAs from control and SCI rats by digesting spinal cord tissue with cold-active protease (CAP). The digested samples were then centrifuged to obtain a supernatant that was used to identify mRNA expression. We identified differentially expressed genes (DEGS) after SCI and mapped them to various biological processes. We validated the DEGs by RT-qPCR and RNA-scope. Results The supernatant fraction was highly enriched for mRNA from axons. Using Gene Ontology, the second most significant pathway for all DEGs was axonogenesis. Among the DEGs was Rims2, which is predominately a circular RNA (circRNA) in the CNS. We show that Rims2 RNA within spinal cord axons is circular. We found an additional 200 putative circRNAs in the axonal-enriched fraction. Knockdown in primary rat cortical neurons of the RNA editing enzyme ADAR1, which inhibits formation of circRNAs, significantly increased axonal outgrowth and increased the expression of circRims2. Using Rims2 as a prototype we used Circular RNA Interactome to predict miRNAs that bind to circRims2 also bind to the 3'UTR of GAP-43, PTEN or CREB1, all known regulators of axonal outgrowth. Axonally-translated GAP-43 supports axonal elongation and we detect GAP-43 mRNA in the rat axons by RNAscope. Discussion By enriching for axonal RNA, we detect SCI induced DEGs, including circRNA such as Rims2. Ablation of ADAR1, the enzyme that regulates circRNA formation, promotes axonal outgrowth of cortical neurons. We developed a pathway model using Circular RNA Interactome that indicates that Rims2 through miRNAs can regulate the axonal translation GAP-43 to regulate axonal regeneration. We conclude that axonal regulatory pathways will play a role in neurorepair.
Collapse
Affiliation(s)
- Mustafa M. Siddiq
- Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Carlos A. Toro
- Spinal Cord Damage Research Center, James J. Peters VA Medical Center, Bronx, NY, United States
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Nicholas P. Johnson
- Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jens Hansen
- Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Yuguang Xiong
- Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | | | - Rosa E. Tolentino
- Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Kaitlin Johnson
- Spinal Cord Damage Research Center, James J. Peters VA Medical Center, Bronx, NY, United States
| | - Gomathi Jayaraman
- Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Zaara Suhail
- Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Lauren Harlow
- Spinal Cord Damage Research Center, James J. Peters VA Medical Center, Bronx, NY, United States
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jinye Dai
- Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Kristin G. Beaumont
- Department of Genetics and Genomic Studies, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Icahn Genomics Institute, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Robert Sebra
- Department of Genetics and Genomic Studies, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Icahn Genomics Institute, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Dianna E. Willis
- Burke Neurological Institute, White Plains, NY, United States
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| | - Christopher P. Cardozo
- Spinal Cord Damage Research Center, James J. Peters VA Medical Center, Bronx, NY, United States
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Rehabilitation Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Ravi Iyengar
- Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
21
|
Lear BP, Moore DL. Moving CNS axon growth and regeneration research into human model systems. Front Neurosci 2023; 17:1198041. [PMID: 37425013 PMCID: PMC10324669 DOI: 10.3389/fnins.2023.1198041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/25/2023] [Indexed: 07/11/2023] Open
Abstract
Axon regeneration is limited in the adult mammalian central nervous system (CNS) due to both intrinsic and extrinsic factors. Rodent studies have shown that developmental age can drive differences in intrinsic axon growth ability, such that embryonic rodent CNS neurons extend long axons while postnatal and adult CNS neurons do not. In recent decades, scientists have identified several intrinsic developmental regulators in rodents that modulate growth. However, whether this developmentally programmed decline in CNS axon growth is conserved in humans is not yet known. Until recently, there have been limited human neuronal model systems, and even fewer age-specific human models. Human in vitro models range from pluripotent stem cell-derived neurons to directly reprogrammed (transdifferentiated) neurons derived from human somatic cells. In this review, we discuss the advantages and disadvantages of each system, and how studying axon growth in human neurons can provide species-specific knowledge in the field of CNS axon regeneration with the goal of bridging basic science studies to clinical trials. Additionally, with the increased availability and quality of 'omics datasets of human cortical tissue across development and lifespan, scientists can mine these datasets for developmentally regulated pathways and genes. As there has been little research performed in human neurons to study modulators of axon growth, here we provide a summary of approaches to begin to shift the field of CNS axon growth and regeneration into human model systems to uncover novel drivers of axon growth.
Collapse
Affiliation(s)
| | - Darcie L. Moore
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
22
|
Sousa CS, Lima R, Cibrão JR, Gomes ED, Fernandes LS, Pinho TS, Silva D, Campos J, Salgado AJ, Silva NA. Pre-Clinical Assessment of Roflumilast Therapy in a Thoracic Model of Spinal Cord Injury. Pharmaceutics 2023; 15:1556. [PMID: 37242797 PMCID: PMC10222626 DOI: 10.3390/pharmaceutics15051556] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/14/2023] [Accepted: 05/17/2023] [Indexed: 05/28/2023] Open
Abstract
The failure of axons to regenerate after a spinal cord injury (SCI) remains one of the greatest challenges in neuroscience. The initial mechanical trauma is followed by a secondary injury cascade, creating a hostile microenvironment, which not only is not permissive to regeneration but also leads to further damage. One of the most promising approaches for promoting axonal regeneration is to maintain the levels of cyclic adenosine monophosphate (cAMP), specifically by a phosphodiesterase-4 (PDE4) inhibitor expressed in neural tissues. Therefore, in our study, we evaluated the therapeutic effect of an FDA-approved PDE4 inhibitor, Roflumilast (Rof), in a thoracic contusion rat model. Results indicate that the treatment was effective in promoting functional recovery. Rof-treated animals showed improvements in both gross and fine motor function. Eight weeks post-injury, the animals significantly recovered by achieving occasional weight-supported plantar steps. Histological assessment revealed a significant decrease in cavity size, less reactive microglia, as well as higher axonal regeneration in treated animals. Molecular analysis revealed that IL-10 and IL-13 levels, as well as VEGF, were increased in the serum of Rof-treated animals. Overall, Roflumilast promotes functional recovery and supports neuroregeneration in a severe thoracic contusion injury model and may be important in SCI treatment.
Collapse
Affiliation(s)
- Carla S Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, 4805-017 Guimarães, Portugal
- Department of Neurosurgery, Hospital Garcia de Orta, 2805-267 Almada, Portugal
| | - Rui Lima
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, 4805-017 Guimarães, Portugal
| | - Jorge R Cibrão
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, 4805-017 Guimarães, Portugal
| | - Eduardo D Gomes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, 4805-017 Guimarães, Portugal
| | - Luís S Fernandes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, 4805-017 Guimarães, Portugal
| | - Tiffany S Pinho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, 4805-017 Guimarães, Portugal
| | - Deolinda Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, 4805-017 Guimarães, Portugal
| | - Jonas Campos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, 4805-017 Guimarães, Portugal
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, 4805-017 Guimarães, Portugal
| | - Nuno A Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's Associate Lab, PT Government Associated Lab, 4805-017 Guimarães, Portugal
| |
Collapse
|
23
|
Wang F, Ruppell KT, Zhou S, Qu Y, Gong J, Shang Y, Wu J, Liu X, Diao W, Li Y, Xiang Y. Gliotransmission and adenosine signaling promote axon regeneration. Dev Cell 2023; 58:660-676.e7. [PMID: 37028426 PMCID: PMC10173126 DOI: 10.1016/j.devcel.2023.03.007] [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: 01/04/2022] [Revised: 11/18/2022] [Accepted: 03/08/2023] [Indexed: 04/08/2023]
Abstract
How glia control axon regeneration remains incompletely understood. Here, we investigate glial regulation of regenerative ability differences of closely related Drosophila larval sensory neuron subtypes. Axotomy elicits Ca2+ signals in ensheathing glia, which activates regenerative neurons through the gliotransmitter adenosine and mounts axon regenerative programs. However, non-regenerative neurons do not respond to glial stimulation or adenosine. Such neuronal subtype-specific responses result from specific expressions of adenosine receptors in regenerative neurons. Disrupting gliotransmission impedes axon regeneration of regenerative neurons, and ectopic adenosine receptor expression in non-regenerative neurons suffices to activate regenerative programs and induce axon regeneration. Furthermore, stimulating gliotransmission or activating the mammalian ortholog of Drosophila adenosine receptors in retinal ganglion cells (RGCs) promotes axon regrowth after optic nerve crush in adult mice. Altogether, our findings demonstrate that gliotransmission orchestrates neuronal subtype-specific axon regeneration in Drosophila and suggest that targeting gliotransmission or adenosine signaling is a strategy for mammalian central nervous system repair.
Collapse
Affiliation(s)
- Fei Wang
- Department of Neurobiology, Program of Neuroscience, University of Massachusetts Chan Medical School, 364 Plantation Street, Worcester, MA 01605, USA
| | - Kendra Takle Ruppell
- Department of Neurobiology, Program of Neuroscience, University of Massachusetts Chan Medical School, 364 Plantation Street, Worcester, MA 01605, USA
| | - Songlin Zhou
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China; Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Yun Qu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Jiaxin Gong
- Department of Neurobiology, Program of Neuroscience, University of Massachusetts Chan Medical School, 364 Plantation Street, Worcester, MA 01605, USA
| | - Ye Shang
- Department of Neurobiology, Program of Neuroscience, University of Massachusetts Chan Medical School, 364 Plantation Street, Worcester, MA 01605, USA
| | - Jinglin Wu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Xin Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Wenlin Diao
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yi Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China; The National Clinical Research Center for Aging and Medicine, Fudan University, Shanghai, China.
| | - Yang Xiang
- Department of Neurobiology, Program of Neuroscience, University of Massachusetts Chan Medical School, 364 Plantation Street, Worcester, MA 01605, USA.
| |
Collapse
|
24
|
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: 7.0] [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
|
25
|
Pourkhodadad S, Hosseinkazemi H, Bonakdar S, Nekounam H. Biomimetic engineered approaches for neural tissue engineering: Spinal cord injury. J Biomed Mater Res B Appl Biomater 2023; 111:701-716. [PMID: 36214332 DOI: 10.1002/jbm.b.35171] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 07/16/2022] [Accepted: 09/03/2022] [Indexed: 01/21/2023]
Abstract
The healing process for spinal cord injuries is complex and presents many challenges. Current advances in nerve regeneration are based on promising tissue engineering techniques, However, the chances of success depend on better mimicking the extracellular matrix (ECM) of neural tissue and better supporting neurons in a three-dimensional environment. The ECM provides excellent biological conditions, including desirable morphological features, electrical conductivity, and chemical compositions for neuron attachment, proliferation and function. This review outlines the rationale for developing a construct for neuron regrowth in spinal cord injury using appropriate biomaterials and scaffolding techniques.
Collapse
Affiliation(s)
| | - Hessam Hosseinkazemi
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Shahin Bonakdar
- National Cell Bank Department, Pasteur Institute of Iran, Tehran, Iran
| | - Houra Nekounam
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
26
|
Gurdita A, Kwiecien JM, Choh V. Development of a new surgical technique to infuse kynurenic acid to optic nerves in chickens for studying loss of myelination. Heliyon 2023; 9:e14361. [PMID: 36938412 PMCID: PMC10020079 DOI: 10.1016/j.heliyon.2023.e14361] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 02/14/2023] [Accepted: 03/01/2023] [Indexed: 03/09/2023] Open
Abstract
Prolonged infusion of a high dose of kynurenic acid (KYNA) reduces the myelin content in the rat spinal cord with preservation of the axonal integrity and without inducing an inflammatory response. We hypothesized that subdural infusion of a high concentration of KYNA can induce myelin loss in the optic nerves (ONs) of chickens. However, existing methods to deliver agents to the ON are inefficient, unlocalized and provide only acute exposure. Thus, we developed a surgical approach for sustained delivery of KYNA to the chicken ON. In brief, the novel surgical technique, which does not include excision of the extraocular muscles, involves incision of the skin and underlying fascial sheath to access the optic nerve within the muscle cone, implantation of a catheter in the dura of the optic nerve, the other end of which exits the orbit under the skin. The catheter runs under the skin near the lateral canthus, over the ears to the back of the neck, where a second incision is made to both implant the osmotic pump and to attach the catheter to the osmotic pump. India ink was used to confirm prolonged sustained administration to the optic nerves and across the chiasm. This surgical model was used to investigate KYNA's effect(s) on myelin loss in the ON. ONs of 7-day old chickens were infused with 50 mM KYNA or phosphate buffered saline (PBS) for seven days. Analysis of KYNA-infused contralateral ON g-ratios and protein levels indicated a reduction in myelin. These findings demonstrate the utility of our surgical approach for sustained delivery of KYNA into the ON and suggest a role for KYNA in modulating CNS myelination.
Collapse
Affiliation(s)
- Akshay Gurdita
- School of Optometry and Vision Science, University of Waterloo, Waterloo, ON, Canada
| | - Jacek M. Kwiecien
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Vivian Choh
- School of Optometry and Vision Science, University of Waterloo, Waterloo, ON, Canada
- Corresponding author. University of Waterloo, 200 Columbia St W, Waterloo, ON N2L 3G1
| |
Collapse
|
27
|
Yao F, Luo Y, Chen Y, Li Y, Hu X, You X, Li Z, Yu S, Tian D, Zheng M, Cheng L, Jing J. Myelin Debris Impairs Tight Junctions and Promotes the Migration of Microvascular Endothelial Cells in the Injured Spinal Cord. Cell Mol Neurobiol 2023; 43:741-756. [PMID: 35147836 DOI: 10.1007/s10571-022-01203-w] [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: 07/07/2021] [Accepted: 01/31/2022] [Indexed: 01/08/2023]
Abstract
Clearance of myelin debris caused by acute demyelination is an essential process for functional restoration following spinal cord injury (SCI). Microvascular endothelial cells, acting as "amateur" phagocytes, have been confirmed to engulf and degrade myelin debris, promoting the inflammatory response, robust angiogenesis, and persistent fibrosis. However, the effect of myelin debris engulfment on the function of endothelial tight junctions (TJs) remains unclear. Here, we demonstrate that myelin debris uptake impairs TJs and gap junctions of endothelial cells in the lesion core of the injured spinal cord and in vitro, resulting in increased permeability and leakage. We further show that myelin debris acts as an inducer to regulate the endothelial-to-mesenchymal transition in a dose-dependent manner and promotes endothelial cell migration through the PI3K/AKT and ERK signaling pathways. Together, our results indicate that myelin debris engulfment impairs TJs and promotes the migration of endothelial cells. Accelerating myelin debris clearance may help maintain blood-spinal cord barrier integrity, thus facilitating restoration of motor and sensory function following SCI.
Collapse
Affiliation(s)
- Fei Yao
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Yang Luo
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Yihao Chen
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Yiteng Li
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Xuyang Hu
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Xingyu You
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Ziyu Li
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Shuisheng Yu
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Dasheng Tian
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Meige Zheng
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China.
| | - Li Cheng
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China.
- School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, China.
| | - Juehua Jing
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
28
|
Qiu X, Ping S, Kyle M, Chin L, Zhao LR. Stem Cell Factor and Granulocyte Colony-Stimulating Factor Promote Remyelination in the Chronic Phase of Severe Traumatic Brain Injury. Cells 2023; 12:705. [PMID: 36899841 PMCID: PMC10000780 DOI: 10.3390/cells12050705] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/14/2023] [Accepted: 02/17/2023] [Indexed: 02/25/2023] Open
Abstract
Severe traumatic brain injury (TBI) causes long-term disability and death in young adults. White matter is vulnerable to TBI damage. Demyelination is a major pathological change of white matter injury after TBI. Demyelination, which is characterized by myelin sheath disruption and oligodendrocyte cell death, leads to long-term neurological function deficits. Stem cell factor (SCF) and granulocyte colony-stimulating factor (G-CSF) treatments have shown neuroprotective and neurorestorative effects in the subacute and chronic phases of experimental TBI. Our previous study has revealed that combined SCF and G-CSF treatment (SCF + G-CSF) enhances myelin repair in the chronic phase of TBI. However, the long-term effect and mechanism of SCF + G-CSF-enhanced myelin repair remain unclear. In this study, we uncovered persistent and progressive myelin loss in the chronic phase of severe TBI. SCF + G-CSF treatment in the chronic phase of severe TBI enhanced remyelination in the ipsilateral external capsule and striatum. The SCF + G-CSF-enhanced myelin repair is positively correlated with the proliferation of oligodendrocyte progenitor cells in the subventricular zone. These findings reveal the therapeutic potential of SCF + G-CSF in myelin repair in the chronic phase of severe TBI and shed light on the mechanism underlying SCF + G-CSF-enhanced remyelination in chronic TBI.
Collapse
Affiliation(s)
- Xuecheng Qiu
- Department of Neurosurgery, State University of New York Upstate Medical University, Syracuse, NY 13210, USA
| | - Suning Ping
- Department of Neurosurgery, State University of New York Upstate Medical University, Syracuse, NY 13210, USA
| | - Michele Kyle
- Department of Neurosurgery, State University of New York Upstate Medical University, Syracuse, NY 13210, USA
| | - Lawrence Chin
- Department of Neurosurgery, State University of New York Upstate Medical University, Syracuse, NY 13210, USA
| | - Li-Ru Zhao
- Department of Neurosurgery, State University of New York Upstate Medical University, Syracuse, NY 13210, USA
| |
Collapse
|
29
|
Qiu X, Ping S, Kyle M, Chin L, Zhao LR. Stem cell factor and granulocyte colony-stimulating factor promote remyelination in the chronic phase of severe traumatic brain injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.24.525450. [PMID: 36747858 PMCID: PMC9900822 DOI: 10.1101/2023.01.24.525450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Severe traumatic brain injury (TBI) causes long-term disability and death in young adults. White matter is vulnerable to TBI damage. Demyelination is a major pathological change of white matter injury after TBI. Demyelination which is characterized by myelin sheath disruption and oligodendrocyte cell death leads to long-term neurological function deficits. Stem cell factor (SCF) and granulocyte colonyâ€"stimulating factor (G-CSF) treatments have shown neuroprotective and neurorestorative effects in the subacute and chronic phases of experimental TBI. Our previous study has revealed that combined SCF and G-CSF treatment (SCF+G-CSF) enhances myelin repair in the chronic phase of TBI. However, the long-term effect and mechanism of SCF+G-CSF-enhanced myelin repair remain unclear. In this study, we uncovered persistent and progressive myelin loss in the chronic phase of severe TBI. SCF+G-CSF treatment in the chronic phase of severe TBI enhanced remyelination in the ipsilateral external capsule and striatum. The SCF+G-CSF-enhanced myelin repair is positively correlated with the proliferation of oligodendrocyte progenitor cells in the subventricular zone. These findings reveal the therapeutic potential of SCF+G-CSF in myelin repair in the chronic phase of severe TBI and shed light on the mechanism underlying SCF+G-CSF-enhanced remyelination in chronic TBI.
Collapse
|
30
|
Li Z, Jiang Z, Lu L, Liu Y. Microfluidic Manipulation for Biomedical Applications in the Central and Peripheral Nervous Systems. Pharmaceutics 2023; 15:pharmaceutics15010210. [PMID: 36678839 PMCID: PMC9862045 DOI: 10.3390/pharmaceutics15010210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 01/04/2023] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Physical injuries and neurodegenerative diseases often lead to irreversible damage to the organizational structure of the central nervous system (CNS) and peripheral nervous system (PNS), culminating in physiological malfunctions. Investigating these complex and diverse biological processes at the macro and micro levels will help to identify the cellular and molecular mechanisms associated with nerve degeneration and regeneration, thereby providing new options for the development of new therapeutic strategies for the functional recovery of the nervous system. Due to their distinct advantages, modern microfluidic platforms have significant potential for high-throughput cell and organoid cultures in vitro, the synthesis of a variety of tissue engineering scaffolds and drug carriers, and observing the delivery of drugs at the desired speed to the desired location in real time. In this review, we first introduce the types of nerve damage and the repair mechanisms of the CNS and PNS; then, we summarize the development of microfluidic platforms and their application in drug carriers. We also describe a variety of damage models, tissue engineering scaffolds, and drug carriers for nerve injury repair based on the application of microfluidic platforms. Finally, we discuss remaining challenges and future perspectives with regard to the promotion of nerve injury repair based on engineered microfluidic platform technology.
Collapse
|
31
|
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: 57] [Impact Index Per Article: 57.0] [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
|
32
|
Chari D, Basit R, Wiseman J, Chowdhury F. Simulating traumatic brain injury in vitro: developing high throughput models to test biomaterial based therapies. Neural Regen Res 2023; 18:289-292. [PMID: 35900405 PMCID: PMC9396524 DOI: 10.4103/1673-5374.346465] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Traumatic brain injuries are serious clinical incidents associated with some of the poorest outcomes in neurological practice. Coupled with the limited regenerative capacity of the brain, this has significant implications for patients, carers, and healthcare systems, and the requirement for life-long care in some cases. Clinical treatment currently focuses on limiting the initial neural damage with long-term care/support from multidisciplinary teams. Therapies targeting neuroprotection and neural regeneration are not currently available but are the focus of intensive research. Biomaterial-based interventions are gaining popularity for a range of applications including biomolecule and drug delivery, and to function as cellular scaffolds. Experimental investigations into the development of such novel therapeutics for traumatic brain injury will be critically underpinned by the availability of appropriate high throughput, facile, ethically viable, and pathomimetic biological model systems. This represents a significant challenge for researchers given the pathological complexity of traumatic brain injury. Specifically, there is a concerted post-injury response mounted by multiple neural cell types which includes microglial activation and astroglial scarring with the expression of a range of growth inhibitory molecules and cytokines in the lesion environment. Here, we review common models used for the study of traumatic brain injury (ranging from live animal models to in vitro systems), focusing on penetrating traumatic brain injury models. We discuss their relative advantages and drawbacks for the developmental testing of biomaterial-based therapies.
Collapse
|
33
|
Omura T. Editorial: Axonal growth in normal and pathological conditions. Front Mol Neurosci 2022; 15:1107999. [PMID: 36583083 PMCID: PMC9792978 DOI: 10.3389/fnmol.2022.1107999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022] Open
|
34
|
Modulation of the Microglial Nogo-A/NgR Signaling Pathway as a Therapeutic Target for Multiple Sclerosis. Cells 2022; 11:cells11233768. [PMID: 36497029 PMCID: PMC9737582 DOI: 10.3390/cells11233768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/23/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
Current therapeutics targeting chronic phases of multiple sclerosis (MS) are considerably limited in reversing the neural damage resulting from repeated inflammation and demyelination insults in the multi-focal lesions. This inflammation is propagated by the activation of microglia, the endogenous immune cell aiding in the central nervous system homeostasis. Activated microglia may transition into polarized phenotypes; namely, the classically activated proinflammatory phenotype (previously categorized as M1) and the alternatively activated anti-inflammatory phenotype (previously, M2). These transitional microglial phenotypes are dynamic states, existing as a continuum. Shifting microglial polarization to an anti-inflammatory status may be a potential therapeutic strategy that can be harnessed to limit neuroinflammation and further neurodegeneration in MS. Our research has observed that the obstruction of signaling by inhibitory myelin proteins such as myelin-associated inhibitory factor, Nogo-A, with its receptor (NgR), can regulate microglial cell function and activity in pre-clinical animal studies. Our review explores the microglial role and polarization in MS pathology. Additionally, the potential therapeutics of targeting Nogo-A/NgR cellular mechanisms on microglia migration, polarization and phagocytosis for neurorepair in MS and other demyelination diseases will be discussed.
Collapse
|
35
|
Hammel G, Zivkovic S, Ayazi M, Ren Y. Consequences and mechanisms of myelin debris uptake and processing by cells in the central nervous system. Cell Immunol 2022; 380:104591. [PMID: 36030093 DOI: 10.1016/j.cellimm.2022.104591] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/29/2022] [Accepted: 08/15/2022] [Indexed: 11/28/2022]
Abstract
Central nervous system (CNS) disorders and trauma involving changes to the neuronal myelin sheath have long been a topic of great interest. One common pathological change in these diseases is the generation of myelin debris resulting from the breakdown of the myelin sheath. Myelin debris contains many inflammatory and neurotoxic factors that inhibit remyelination and make its clearance a prerequisite for healing in CNS disorders. Many professional and semiprofessional phagocytes participate in the clearance of myelin debris in the CNS. These cells use various mechanisms for the uptake of myelin debris, and each cell type produces its own unique set of pathologic consequences resulting from the debris uptake. Examining these cells' phagocytosis of myelin debris will contribute to a more complete understanding of CNS disease pathogenesis and help us conceptualize how the necessary clearance of myelin debris must be balanced with the detrimental consequences brought about by its clearance.
Collapse
Affiliation(s)
- Grace Hammel
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, United States.
| | - Sandra Zivkovic
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, United States.
| | - Maryam Ayazi
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, United States.
| | - Yi Ren
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, United States.
| |
Collapse
|
36
|
Efficacy of Nerve-Derived Hydrogels to Promote Axon Regeneration Is Influenced by the Method of Tissue Decellularization. Int J Mol Sci 2022; 23:ijms23158746. [PMID: 35955880 PMCID: PMC9369339 DOI: 10.3390/ijms23158746] [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: 07/05/2022] [Revised: 08/01/2022] [Accepted: 08/05/2022] [Indexed: 11/30/2022] Open
Abstract
Injuries to large peripheral nerves are often associated with tissue defects and require reconstruction using autologous nerve grafts, which have limited availability and result in donor site morbidity. Peripheral nerve-derived hydrogels could potentially supplement or even replace these grafts. In this study, three decellularization protocols based on the ionic detergents sodium dodecyl sulfate (P1) and sodium deoxycholate (P2), or the organic solvent tri-n-butyl phosphate (P3), were used to prepare hydrogels. All protocols resulted in significantly decreased amounts of genomic DNA, but the P2 hydrogel showed the best preservation of extracellular matrix proteins, cytokines, and chemokines, and reduced levels of sulfated glycosaminoglycans. In vitro P1 and P2 hydrogels supported Schwann cell viability, secretion of VEGF, and neurite outgrowth. Surgical repair of a 10 mm-long rat sciatic nerve gap was performed by implantation of tubular polycaprolactone conduits filled with hydrogels followed by analyses using diffusion tensor imaging and immunostaining for neuronal and glial markers. The results demonstrated that the P2 hydrogel considerably increased the number of axons and the distance of regeneration into the distal nerve stump. In summary, the method used to decellularize nerve tissue affects the efficacy of the resulting hydrogels to support regeneration after nerve injury.
Collapse
|
37
|
Development of Neurogenic Detrusor Overactivity after Thoracic Spinal Cord Injury Is Accompanied by Time-Dependent Changes in Lumbosacral Expression of Axonal Growth Regulators. Int J Mol Sci 2022; 23:ijms23158667. [PMID: 35955811 PMCID: PMC9368817 DOI: 10.3390/ijms23158667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 07/29/2022] [Accepted: 08/01/2022] [Indexed: 12/04/2022] Open
Abstract
Thoracic spinal cord injury (SCI) results in urinary dysfunction, which majorly affects the quality of life of SCI patients. Abnormal sprouting of lumbosacral bladder afferents plays a crucial role in this condition. Underlying mechanisms may include changes in expression of regulators of axonal growth, including chondroitin sulphate proteoglycans (CSPGs), myelin-associated inhibitors (MAIs) and repulsive guidance molecules, known to be upregulated at the injury site post SCI. Here, we confirmed lumbosacral upregulation of the growth-associated protein GAP43 in SCI animals with bladder dysfunction, indicating the occurrence of axonal sprouting. Neurocan and Phosphacan (CSPGs), as well as Nogo-A (MAI), at the same spinal segments were upregulated 7 days post injury (dpi) but returned to baseline values 28 dpi. In turn, qPCR analysis of the mRNA levels for receptors of those repulsive molecules in dorsal root ganglia (DRG) neurons showed a time-dependent decrease in receptor expression. In vitro assays with DRG neurons from SCI rats demonstrated that exposure to high levels of NGF downregulated the expression of some, but not all, receptors for those regulators of axonal growth. The present results, therefore, show significant molecular changes at the lumbosacral cord and DRGs after thoracic lesion, likely critically involved in neuroplastic events leading to urinary impairment.
Collapse
|
38
|
Samejima S, Henderson R, Pradarelli J, Mondello SE, Moritz CT. Activity-dependent plasticity and spinal cord stimulation for motor recovery following spinal cord injury. Exp Neurol 2022; 357:114178. [PMID: 35878817 DOI: 10.1016/j.expneurol.2022.114178] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/22/2022] [Accepted: 07/16/2022] [Indexed: 02/07/2023]
Abstract
Spinal cord injuries lead to permanent physical impairment despite most often being anatomically incomplete disruptions of the spinal cord. Remaining connections between the brain and spinal cord create the potential for inducing neural plasticity to improve sensorimotor function, even many years after injury. This narrative review provides an overview of the current evidence for spontaneous motor recovery, activity-dependent plasticity, and interventions for restoring motor control to residual brain and spinal cord networks via spinal cord stimulation. In addition to open-loop spinal cord stimulation to promote long-term neuroplasticity, we also review a more targeted approach: closed-loop stimulation. Lastly, we review mechanisms of spinal cord neuromodulation to promote sensorimotor recovery, with the goal of advancing the field of rehabilitation for physical impairments following spinal cord injury.
Collapse
Affiliation(s)
- Soshi Samejima
- International Collaboration on Repair Discoveries, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada; Department of Medicine, Division of Physical Medicine and Rehabilitation, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Richard Henderson
- Department of Electrical and Computer Engineering, University of Washington, Seattle, WA, USA; Department of Rehabilitation Medicine, University of Washington, Seattle, WA, USA
| | - Jared Pradarelli
- Department of Rehabilitation Medicine, University of Washington, Seattle, WA, USA
| | - Sarah E Mondello
- Department of Rehabilitation Medicine, University of Washington, Seattle, WA, USA
| | - Chet T Moritz
- Department of Electrical and Computer Engineering, University of Washington, Seattle, WA, USA; Department of Rehabilitation Medicine, University of Washington, Seattle, WA, USA; Center for Neurotechnology, Seattle, WA, USA; Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA.
| |
Collapse
|
39
|
Abstract
Schwann cells in the peripheral nervous system (PNS) are essential for the support and myelination of axons, ensuring fast and accurate communication between the central nervous system and the periphery. Schwann cells and related glia accompany innervating axons in virtually all tissues in the body, where they exhibit remarkable plasticity and the ability to modulate pathology in extraordinary, and sometimes surprising, ways. Here, we provide a brief overview of the various glial cell types in the PNS and describe the cornerstone cellular and molecular processes that enable Schwann cells to perform their canonical functions. We then dive into discussing exciting noncanonical functions of Schwann cells and related PNS glia, which include their role in organizing the PNS, in regulating synaptic activity and pain, in modulating immunity, in providing a pool of stem cells for different organs, and, finally, in influencing cancer.
Collapse
Affiliation(s)
- Carla Taveggia
- Axo-Glial Interaction Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy;
| | - M. Laura Feltri
- Institute for Myelin and Glia Exploration, Departments of Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, USA
| |
Collapse
|
40
|
Winter CC, He Z, Jacobi A. Axon Regeneration: A Subcellular Extension in Multiple Dimensions. Cold Spring Harb Perspect Biol 2022; 14:a040923. [PMID: 34518340 PMCID: PMC8886981 DOI: 10.1101/cshperspect.a040923] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Axons are a unique cellular structure that allows for the communication between neurons. Axon damage compromises neuronal communications and often leads to functional deficits. Thus, developing strategies that promote effective axon regeneration for functional restoration is highly desirable. One fruitful approach is to dissect the regenerative mechanisms used by some types of neurons in both mammalian and nonmammalian systems that exhibit spontaneous regenerative capacity. Additionally, numerous efforts have been devoted to deciphering the barriers that prevent successful axon regeneration in the most regeneration-refractory system-the adult mammalian central nervous system. As a result, several regeneration-promoting strategies have been developed, but significant limitations remain. This review is aimed to summarize historic progression and current understanding of this exciting yet incomplete endeavor.
Collapse
Affiliation(s)
- Carla C Winter
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, Massachusetts 02115, USA
- Department of Neurology and Ophthalmology, Harvard Medical School, Boston, Massachusetts 02115, USA
- PhD Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Zhigang He
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, Massachusetts 02115, USA
- Department of Neurology and Ophthalmology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Anne Jacobi
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, Massachusetts 02115, USA
- Department of Neurology and Ophthalmology, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
41
|
Siddiq MM, Hannila SS, Zorina Y, Nikulina E, Rabinovich V, Hou J, Huq R, Richman EL, Tolentino RE, Hansen J, Velenosi A, Kwon BK, Tsirka SE, Maze I, Sebra R, Beaumont KG, Toro CA, Cardozo CP, Iyengar R, Filbin MT. Extracellular histones, a new class of inhibitory molecules of CNS axonal regeneration. Brain Commun 2022; 3:fcab271. [PMID: 34993473 PMCID: PMC8728726 DOI: 10.1093/braincomms/fcab271] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/05/2021] [Accepted: 08/30/2021] [Indexed: 12/26/2022] Open
Abstract
Axonal regeneration in the mature CNS is limited by extracellular inhibitory factors. Triple knockout mice lacking the major myelin-associated inhibitors do not display spontaneous regeneration after injury, indicating the presence of other inhibitors. Searching for such inhibitors, we have detected elevated levels of histone H3 in human CSF 24 h after spinal cord injury. Following dorsal column lesions in mice and optic nerve crushes in rats, elevated levels of extracellular histone H3 were detected at the injury site. Similar to myelin-associated inhibitors, these extracellular histones induced growth cone collapse and inhibited neurite outgrowth. Histones mediate inhibition through the transcription factor Y-box-binding protein 1 and Toll-like receptor 2, and these effects are independent of the Nogo receptor. Histone-mediated inhibition can be reversed by the addition of activated protein C in vitro, and activated protein C treatment promotes axonal regeneration in the crushed optic nerve in vivo. These findings identify extracellular histones as a new class of nerve regeneration-inhibiting molecules within the injured CNS.
Collapse
Affiliation(s)
- Mustafa M Siddiq
- Department of Biological Sciences, Hunter College, City University of New York, New York, NY 10065, USA.,Department of Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sari S Hannila
- Department of Biological Sciences, Hunter College, City University of New York, New York, NY 10065, USA.,Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Manitoba R3E 0J9, Canada
| | - Yana Zorina
- Department of Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Gene Editing and Screening Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Elena Nikulina
- Department of Biological Sciences, Hunter College, City University of New York, New York, NY 10065, USA.,Department of Physiology and Pharmacology, SUNY Downstate Health Science University, Brooklyn, NY 11203, USA
| | - Vera Rabinovich
- Department of Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jianwei Hou
- Department of Biological Sciences, Hunter College, City University of New York, New York, NY 10065, USA
| | - Rumana Huq
- Department of Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Erica L Richman
- Department of Biological Sciences, Hunter College, City University of New York, New York, NY 10065, USA
| | - Rosa E Tolentino
- Department of Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jens Hansen
- Department of Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | - Brian K Kwon
- International Collaboration on Repair Discoveries, University of British Columbia (UBC), Vancouver, BC, Canada
| | - Stella E Tsirka
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794-8651, USA
| | - Ian Maze
- Department of Neuroscience and Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Robert Sebra
- Department of Genetics and Genomic Studies, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Icahn Institute for Data Science and Genomic Technology, Black Family Stem Cell Institute, New York, NY 10029, USA.,Sema4, a Mount Sinai Venture, Stamford, CT, USA
| | - Kristin G Beaumont
- Department of Genetics and Genomic Studies, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Icahn Institute for Data Science and Genomic Technology, Black Family Stem Cell Institute, New York, NY 10029, USA
| | - Carlos A Toro
- National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters VA Medical Center, New York, NY 10468, USA.,Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Christopher P Cardozo
- National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters VA Medical Center, New York, NY 10468, USA.,Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Department of Rehabilitation Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ravi Iyengar
- Department of Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Marie T Filbin
- Department of Biological Sciences, Hunter College, City University of New York, New York, NY 10065, USA
| |
Collapse
|
42
|
Varadarajan SG, Hunyara JL, Hamilton NR, Kolodkin AL, Huberman AD. Central nervous system regeneration. Cell 2022; 185:77-94. [PMID: 34995518 PMCID: PMC10896592 DOI: 10.1016/j.cell.2021.10.029] [Citation(s) in RCA: 84] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/26/2021] [Accepted: 10/27/2021] [Indexed: 02/06/2023]
Abstract
Neurons of the mammalian central nervous system fail to regenerate. Substantial progress has been made toward identifying the cellular and molecular mechanisms that underlie regenerative failure and how altering those pathways can promote cell survival and/or axon regeneration. Here, we summarize those findings while comparing the regenerative process in the central versus the peripheral nervous system. We also highlight studies that advance our understanding of the mechanisms underlying neural degeneration in response to injury, as many of these mechanisms represent primary targets for restoring functional neural circuits.
Collapse
Affiliation(s)
| | - John L Hunyara
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Natalie R Hamilton
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Alex L Kolodkin
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Andrew D Huberman
- Department of Neurobiology, Stanford University, Stanford, CA 94305, USA; Department of Ophthalmology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
43
|
Pandey S, Mudgal J. A Review on the Role of Endogenous Neurotrophins and Schwann Cells in Axonal Regeneration. J Neuroimmune Pharmacol 2022; 17:398-408. [PMID: 34843075 PMCID: PMC9810669 DOI: 10.1007/s11481-021-10034-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 11/13/2021] [Indexed: 01/13/2023]
Abstract
Injury to the peripheral nerve is traditionally referred to acquired nerve injury as they are the result of physical trauma due to laceration, stretch, crush and compression of nerves. However, peripheral nerve injury may not be completely limited to acquired physical trauma. Peripheral nerve injury equally implies clinical conditions like Guillain-Barré syndrome (GBS), Carpal tunnel syndrome, rheumatoid arthritis and diabetes. Physical trauma is commonly mono-neuropathic as it engages a single nerve and produces focal damage, while in the context of pathological conditions the damage is divergent involving a group of the nerve causing polyneuropathy. Damage to the peripheral nerve can cause a diverse range of manifestations from sensory impairment to loss of function with unpredictable recovery patterns. Presently no treatment option provides complete or functional recovery in nerve injury, as nerve cells are highly differentiated and inert to regeneration. However, the regenerative phenotypes in Schwann cells get expressed when a signalling cascade is triggered by neurotrophins. Neurotrophins are one of the promising biomolecules that are released naturally post-injury with the potential to exhibit better functional recovery. Pharmacological intervention modulating the expression of these neurotrophins such as brain-derived neurotrophic factor (BDNF) and pituitary adenylyl cyclase-activating peptide (PACAP) can prove to be a significant treatment option as endogenous compounds which may have remarkable innate advantage showing maximum 'biological relevance'.
Collapse
Affiliation(s)
- Samyak Pandey
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India, 576104
| | - Jayesh Mudgal
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India, 576104.
| |
Collapse
|
44
|
Salvador AFM, Kipnis J. Immune response after central nervous system injury. Semin Immunol 2022; 59:101629. [PMID: 35753867 DOI: 10.1016/j.smim.2022.101629] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 05/07/2022] [Accepted: 06/13/2022] [Indexed: 01/15/2023]
Abstract
Traumatic injuries of the central nervous system (CNS) affect millions of people worldwide, and they can lead to severely damaging consequences such as permanent disability and paralysis. Multiple factors can obstruct recovery after CNS injury. One of the most significant is the progressive neuronal death that follows the initial mechanical impact, leading to the loss of undamaged cells via a process termed secondary neurodegeneration. Efforts to define treatments that limit the spread of damage, while important, have been largely ineffectual owing to gaps in the mechanistic understanding that underlies the persisting neuronal cell death. Inflammation, with its influx of immune cells that occurs shortly after injury, has been associated with secondary neurodegeneration. However, the role of the immune system after CNS injury is far more complex. Studies have indicated that the immune response after CNS injury is detrimental, owing to immune cell-produced factors (e.g., pro-inflammatory cytokines, free radicals, neurotoxic glutamate) that worsen tissue damage. Our lab and others have also demonstrated the beneficial immune response that occurs after CNS injury, with the release of growth factors such as brain-derived growth factor (BDNF) and interleukin (IL-10) and the clearance of apoptotic and myelin debris by immune cells1-4. In this review, we first discuss the multifaceted roles of the immune system after CNS injury. We then speculate on how advancements in single-cell RNA technologies can dramatically change our understanding of the immune response, how the spinal cord meninges serve as an important site for hosting immunological processes critical for recovery, and how the origin of peripherally recruited immune cells impacts their function in the injured CNS.
Collapse
Affiliation(s)
- Andrea Francesca M Salvador
- Department of Pathology & Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA; Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22908, USA.
| | - Jonathan Kipnis
- Department of Pathology & Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
45
|
The role of ubiquitin-specific peptidases in glioma progression. Biomed Pharmacother 2021; 146:112585. [PMID: 34968923 DOI: 10.1016/j.biopha.2021.112585] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 12/21/2021] [Accepted: 12/23/2021] [Indexed: 11/24/2022] Open
Abstract
The balance between ubiquitination and deubiquitination is crucial for protein stability, function and location under physiological conditions. Dysregulation of E1/E2/E3 ligases or deubiquitinases (DUBs) results in malfunction of the ubiquitin system and is involved in many diseases. Increasing reports have indicated that ubiquitin-specific peptidases (USPs) play a part in the progression of many kinds of cancers and could be good targets for anticancer treatment. Glioma is the most common malignant tumor in the central nervous system. Clinical treatment for high-grade glioma is unsatisfactory thus far. Multiple USPs are dysregulated in glioma and have the potential to be therapeutic targets. In this review, we collected studies on the roles of USPs in glioma progression and summarized the mechanisms of USPs in glioma tumorigenesis, malignancy and chemoradiotherapy resistance.
Collapse
|
46
|
Zhao H, Liu ZD, Zhang YB, Gao XY, Wang C, Liu Y, Wang XF. NEP1‑40 promotes myelin regeneration via upregulation of GAP‑43 and MAP‑2 expression after focal cerebral ischemia in rats. Mol Med Rep 2021; 24:844. [PMID: 34643252 PMCID: PMC8524407 DOI: 10.3892/mmr.2021.12484] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 09/08/2021] [Indexed: 01/26/2023] Open
Abstract
Axon regeneration after lesions to the central nervous system (CNS) is largely limited by the presence of growth inhibitory molecules expressed in myelin. Nogo‑A is a principal inhibitor of neurite outgrowth, and blocking the activity of Nogo‑A can induce axonal sprouting and functional recovery. However, there are limited data on the expression of Nogo‑A after CNS lesions, and the mechanism underlying its influences on myelin growth remains unknown. The aim of the present study was to observe the time course of Nogo‑A after cerebral ischemia/reperfusion in rats using immunohistochemistry and western blot techniques, and to test the effect of its inhibitor Nogo extracellular peptide 1‑40 (NEP1‑40) on neural plasticity proteins, growth‑associated binding protein 43 (GAP‑43) and microtubule associated protein 2 (MAP‑2), as a possible mechanism underlying myelin suppression. A classic model of middle cerebral artery occlusion (MCAO) was established in Sprague‑Dawley rats, which were divided into three groups: i) MCAO model group; ii) MCAO + saline group; and iii) MCAO + NEP1‑40 group. Rats of each group were divided into five subgroups by time points as follows: days 1, 3, 7, 14 and 28. Animals that only received sham operation were used as controls. The Nogo‑A immunoreactivity was located primarily in the cytoplasm of oligodendrocytes. The number of Nogo‑A immunoreactive cells significantly increased from day 1 to day 3 after MCAO, nearly returning to the control level at day 7, increased again at day 14 and decreased at day 28. Myelin basic protein (MBP) immunoreactivity in the ipsilateral striatum gradually decreased from day 1 to day 28 after ischemia, indicating myelin loss appeared at early time points and continuously advanced during ischemia. Then, intracerebroventricular infusion of NEP1‑40, which is a Nogo‑66 receptor antagonist peptide, was administered at days 1, 3 and 14 after MCAO. It was observed that GAP‑43 considerably increased from day 1 to day 7 and then decreased to a baseline level at day 28 compared with the control. MAP‑2 expression across days 1‑28 significantly decreased after MCAO. Administration of NEP1‑40 attenuated the reduction of MBP, and upregulated GAP‑43 and MAP‑2 expression at the corresponding time points after MCAO compared with the MCAO + saline group. The present results indicated that NEP1‑40 ameliorated myelin damage and promoted regeneration by upregulating the expression of GAP‑43 and MAP‑2 related to neuronal and axonal plasticity, which may aid with the identification of a novel molecular mechanism of restriction in CNS regeneration mediated by Nogo‑A after ischemia in rats.
Collapse
Affiliation(s)
- Hong Zhao
- Department of Neurology, Dalian Municipal Central Hospital Affiliated to Dalian Medical University, Dalian, Liaoning 116033, P.R. China,Correspondence to: Professor Hong Zhao, Department of Neurology, Dalian Municipal Central Hospital Affiliated to Dalian Medical University, 826 Xi Nan Road, Dalian, Liaoning 116033, P.R. China, E-mail:
| | - Zhen-Dong Liu
- Department of General Medicine, Central Hospital Affiliated to Shaoxing University, Shaoxing, Zhejiang 312000, P.R. China
| | - Yong-Bo Zhang
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Xiao-Yu Gao
- Department of Neurology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Cui Wang
- Department of Neurology, Dalian Municipal Central Hospital Affiliated to Dalian Medical University, Dalian, Liaoning 116033, P.R. China
| | - Yi Liu
- Department of Neurology, Dalian Municipal Central Hospital Affiliated to Dalian Medical University, Dalian, Liaoning 116033, P.R. China
| | - Xun-Fen Wang
- Department of Neurology, Dalian Medical University, Dalian, Liaoning 116033, P.R. China
| |
Collapse
|
47
|
Li HX, Feng J, Liu Q, Ou BQ, Lu SY, Ma Y. PACAP-derived mutant peptide MPAPO protects trigeminal ganglion cell and the retina from hypoxic injury through anti-oxidative stress, anti-apoptosis, and promoting axon regeneration. Biochim Biophys Acta Gen Subj 2021; 1865:130018. [PMID: 34597723 DOI: 10.1016/j.bbagen.2021.130018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 08/30/2021] [Accepted: 09/20/2021] [Indexed: 12/20/2022]
Abstract
The purpose of this study was to determine whether the MPAPO, derived peptide of pituitary adenylate cyclase-activating polypeptide (PACAP), would protect trigeminal ganglion cells (TGCs) and the mice retinas from a hypoxic insult. The nerve endings of the ophthalmic nerve of the trigeminal nerve are widely distributed in eye tissues. In TGCs after hypoxia exposure, we discovered that reactive oxygen species level, the contents of cytosolic cytochrome c and cleaved-caspase-3 were significantly increased, in the meanwhile, m-Calpain was activated and cytoskeleton proteins (αII-spectrin and Synapsin) were degraded, neurites of TGCs disappeared, but these effects were reversed in TGCs treated with MPAPO. The structure of the mice retinas after hypoxic exposure was disordered. Increased lipid peroxidation (LPO), decreased glutathione (GSH) levels, and decreased superoxide dismutase (SOD) activity, positive cells of terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL), the disintegration of nerve fibers was examined in the retinas following a hypoxic insult. Disordered retina was attenuated with MPAPO eye drops, as well as hypoxia-induced apoptosis in the developing retina, increase in LPO, and decrease in GSH levels and SOD activity of the retina. Moreover, the disintegrated retinal nerve fibers were reassembled after MPAPO treatment. These results suggest that hypoxia induces oxidative stress, apoptosis, and neurites disruption, while MPAPO is remarkably protective against these adverse effects of hypoxia in TGCs and the developing retinas by specifically activating PAC1 receptor.
Collapse
Affiliation(s)
- Hui-Xian Li
- Institute of Biomedicine, Department of Cellular Biology, National Engineering Research Center of Genetic Medicine, Key Laboratory of Bioengineering Medicine of Guangdong Province, The national Demonstration center for Experimental Education of Life Science and Technology, Jinan University, 601 Huangpu Ave West, Guangzhou, 510632, Guangdong, People's Republic of China
| | - Jia Feng
- Institute of Biomedicine, Department of Cellular Biology, National Engineering Research Center of Genetic Medicine, Key Laboratory of Bioengineering Medicine of Guangdong Province, The national Demonstration center for Experimental Education of Life Science and Technology, Jinan University, 601 Huangpu Ave West, Guangzhou, 510632, Guangdong, People's Republic of China
| | - Qian Liu
- Institute of Biomedicine, Department of Cellular Biology, National Engineering Research Center of Genetic Medicine, Key Laboratory of Bioengineering Medicine of Guangdong Province, The national Demonstration center for Experimental Education of Life Science and Technology, Jinan University, 601 Huangpu Ave West, Guangzhou, 510632, Guangdong, People's Republic of China
| | - Bi-Qian Ou
- Institute of Biomedicine, Department of Cellular Biology, National Engineering Research Center of Genetic Medicine, Key Laboratory of Bioengineering Medicine of Guangdong Province, The national Demonstration center for Experimental Education of Life Science and Technology, Jinan University, 601 Huangpu Ave West, Guangzhou, 510632, Guangdong, People's Republic of China
| | - Shi-Yin Lu
- Institute of Biomedicine, Department of Cellular Biology, National Engineering Research Center of Genetic Medicine, Key Laboratory of Bioengineering Medicine of Guangdong Province, The national Demonstration center for Experimental Education of Life Science and Technology, Jinan University, 601 Huangpu Ave West, Guangzhou, 510632, Guangdong, People's Republic of China
| | - Yi Ma
- Institute of Biomedicine, Department of Cellular Biology, National Engineering Research Center of Genetic Medicine, Key Laboratory of Bioengineering Medicine of Guangdong Province, The national Demonstration center for Experimental Education of Life Science and Technology, Jinan University, 601 Huangpu Ave West, Guangzhou, 510632, Guangdong, People's Republic of China.
| |
Collapse
|
48
|
Linke AC, Slušná D, Kohli JS, Álvarez-Linera Prado J, Müller RA, Hinzen W. Morphometry and functional connectivity of auditory cortex in school-age children with profound language disabilities: Five comparative case studies. Brain Cogn 2021; 155:105822. [PMID: 34837801 DOI: 10.1016/j.bandc.2021.105822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 10/27/2021] [Accepted: 11/02/2021] [Indexed: 10/19/2022]
Abstract
Many neurodevelopmental conditions imply absent or severely reduced language capacities at school age. Evidence from functional magnetic resonance imaging is highly limited. We selected a series of five cases scanned with the same fMRI paradigm and the aim of relating individual language profiles onto underlying patterns of functional connectivity (FC) across auditory language cortex: three with neurogenetic syndromes (Coffin-Siris, Landau-Kleffner, and Fragile-X), one with idiopathic intellectual disability, one with autism spectrum disorder (ASD). Compared to both a group with typical development (TD) and a verbal ASD group (total N = 110), they all showed interhemispheric FC below two standard deviations of the TD mean. Children with higher language scores showed higher intrahemispheric FC between Heschl's gyrus and other auditory language regions, as well as an increase of FC during language stimulation compared to rest. An increase of FC in forward vs. reversed speech in the posterior and middle temporal gyri was seen across all cases. The Coffin-Siris case, the most severe, also had the most anomalous FC patterns and showed reduced myelin content, while the Landau-Kleffner case showed reduced cortical thickness. These results suggest potential for neural markers and mechanisms of severe language processing deficits under highly heterogeneous etiological conditions.
Collapse
Affiliation(s)
- Annika Carola Linke
- Brain Development Imaging Laboratories, Department of Psychology, San Diego State University, San Diego, CA, USA.
| | - Dominika Slušná
- Department of Translation and Language Sciences, Campus Poblenou, Pompeu Fabra University, Barcelona 08018, Barcelona, Spain
| | - Jiwandeep Singh Kohli
- Brain Development Imaging Laboratories, Department of Psychology, San Diego State University, San Diego, CA, USA
| | | | - Ralph-Axel Müller
- Brain Development Imaging Laboratories, Department of Psychology, San Diego State University, San Diego, CA, USA; San Diego State University/University of California San Diego Joint Doctoral Program in Clinical Psychology, San Diego, CA, USA
| | - Wolfram Hinzen
- Department of Translation and Language Sciences, Campus Poblenou, Pompeu Fabra University, Barcelona 08018, Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats, ICREA, 08010 Barcelona, Spain
| |
Collapse
|
49
|
Roy A, Pathak Z, Kumar H. Strategies to neutralize RhoA/ROCK pathway after spinal cord injury. Exp Neurol 2021; 343:113794. [PMID: 34166685 DOI: 10.1016/j.expneurol.2021.113794] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/01/2021] [Accepted: 06/19/2021] [Indexed: 01/22/2023]
Abstract
Regeneration is bungled following CNS injuries, including spinal cord injury (SCI). Inherent decay of permissive conditions restricts the regrowth of the mature CNS after an injury. Hypertrophic scarring, insignificant intrinsic axon-growth activity, and axon-growth inhibitory molecules such as myelin inhibitors and scar inhibitors constitute a significant hindrance to spinal cord repair. Besides these molecules, a combined absence of various mechanisms responsible for axonal regeneration is the main reason behind the dereliction of the adult CNS to regenerate. The neutralization of specific inhibitors/proteins by stymieing antibodies or encouraging enzymatic degradation results in improved axon regeneration. Previous efforts to induce regeneration after SCI have stimulated axonal development in or near lesion sites, but not beyond them. Several pathways are responsible for the axonal growth obstruction after a CNS injury, including SCI. Herein, we summarize the axonal, glial, and intrinsic factor which impedes the regeneration. We have also discussed the methods to stabilize microtubules and through this to maintain the proper cytoskeletal dynamics of growth cone as disorganized microtubules lead to the failure of axonal regeneration. Moreover, we primarily focus on diverse inhibitors of axonal growth and molecular approaches to counteract them and their downstream intracellular signaling through the RhoA/ROCK pathway.
Collapse
Affiliation(s)
- Abhishek Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India
| | - Zarna Pathak
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India
| | - Hemant Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India.
| |
Collapse
|
50
|
Li L, Luo L, Chen T, Cao W, Xu X, Zhang Y, Yue P, Fan Y, Chen J, Liu M, Ma M, Tao L, Peng Y, Dong Y, Li B, Luo S, Kong J, Zhou G, Wen S, Liu A, Bao F. Proteomic Analysis of Rhesus Macaque Brain Explants Treated With Borrelia burgdorferi Identifies Host GAP-43 as a Potential Factor Associated With Lyme Neuroborreliosis. Front Cell Infect Microbiol 2021; 11:647662. [PMID: 34178719 PMCID: PMC8224226 DOI: 10.3389/fcimb.2021.647662] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 05/25/2021] [Indexed: 12/02/2022] Open
Abstract
Background Lyme neuroborreliosis (LNB) is one of the most dangerous manifestations of Lyme disease, but the pathogenesis and inflammatory mechanisms are not fully understood. Methods Cultured explants from the frontal cortex of rhesus monkey brain (n=3) were treated with live Borrelia burgdorferi (Bb) or phosphate-buffered saline (PBS) for 6, 12, and 24 h. Total protein was collected for sequencing and bioinformatics analysis. In addition, changes in protein expression in the explants over time following Bb treatment were screened. Results We identified 1237 differentially expressed proteins (DEPs; fold change ≥1.5 or ≤0.67, P-value ≤0.05). One of these, growth-associated protein 43 (GAP-43), was highly expressed at all time points in the explants. The results of the protein-protein interaction network analysis of DEPs suggested that GAP-43 plays a role in the neuroinflammation associated with LNB. In HMC3 cells incubated with live Bb or PBS for 6, 12, and 24 h, real-time PCR and western blot analyses confirmed the increase of GAP-43 mRNA and protein, respectively. Conclusions Elevated GAP-43 expression is a potential marker for LNB that may be useful for diagnosis or treatment.
Collapse
Affiliation(s)
- Lianbao Li
- Department of Microbiology and Immunology, Kunming Medical University, Kunming, China
| | - Lisha Luo
- Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, China
| | - Taigui Chen
- Department of Microbiology and Immunology, Kunming Medical University, Kunming, China
| | - Wenjing Cao
- Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, China
| | - Xin Xu
- Department of Microbiology and Immunology, Kunming Medical University, Kunming, China
| | - Yu Zhang
- Department of Microbiology and Immunology, Kunming Medical University, Kunming, China
| | - Peng Yue
- Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, China
| | - Yuxin Fan
- Department of Microbiology and Immunology, Kunming Medical University, Kunming, China
| | - Jingjing Chen
- Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, China
| | - Meixiao Liu
- Department of Microbiology and Immunology, Kunming Medical University, Kunming, China
| | - Mingbiao Ma
- Department of Microbiology and Immunology, Kunming Medical University, Kunming, China
| | - Lvyan Tao
- Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, China
| | - Yun Peng
- Department of Microbiology and Immunology, Kunming Medical University, Kunming, China
| | - Yan Dong
- Department of Microbiology and Immunology, Kunming Medical University, Kunming, China
| | - Bingxue Li
- Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, China
| | - Suyi Luo
- Department of Microbiology and Immunology, Kunming Medical University, Kunming, China
| | - Jing Kong
- Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, China
| | - Guozhong Zhou
- Department of Microbiology and Immunology, Kunming Medical University, Kunming, China
| | - Shiyuan Wen
- Department of Microbiology and Immunology, Kunming Medical University, Kunming, China
| | - Aihua Liu
- Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, China.,Yunnan Province Key Laboratory of Children's Major Diseases Research, The Children's Hospital of Kunming/Kunming Medical University, Kunming, China.,The Institute for Tropical Medicine, Kunming Medical University, Kunming, China.,Yunnan Demonstration Base of International Science and Technology Cooperation for Tropical Diseases, Kunming, China
| | - Fukai Bao
- Department of Microbiology and Immunology, Kunming Medical University, Kunming, China.,Yunnan Province Key Laboratory of Children's Major Diseases Research, The Children's Hospital of Kunming/Kunming Medical University, Kunming, China.,The Institute for Tropical Medicine, Kunming Medical University, Kunming, China.,Yunnan Demonstration Base of International Science and Technology Cooperation for Tropical Diseases, Kunming, China
| |
Collapse
|