1
|
Castro R, Lopes M, De Biase L, Valdez G. Aging spinal cord microglia become phenotypically heterogeneous and preferentially target motor neurons and their synapses. Glia 2024; 72:206-221. [PMID: 37737058 PMCID: PMC10773989 DOI: 10.1002/glia.24470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 08/07/2023] [Accepted: 09/03/2023] [Indexed: 09/23/2023]
Abstract
Microglia have been found to acquire unique region-dependent deleterious features with age and diseases that contribute to neuronal dysfunction and degeneration in the brain. However, it remains unknown whether microglia exhibit similar phenotypic heterogeneity in the spinal cord. Here, we performed a regional analysis of spinal cord microglia in 3-, 16-, 23-, and 30-month-old mice. Using light and electron microscopy, we discovered that spinal cord microglia acquire an increasingly activated phenotype during the course of aging regardless of regional location. However, aging causes microglia in the ventral but not dorsal horn to lose their spatial organization. Aged ventral horn microglia also aggregate around the somata of motor neurons and increase their contacts with motor synapses, which have been shown to be lost with age. These findings suggest that microglia may affect the ability of motor neurons to receive and relay motor commands during aging. To generate additional insights about aging spinal cord microglia, we performed RNA-sequencing on FACS-isolated microglia from 3-, 18-, 22-, and 29-month-old mice. We found that spinal cord microglia acquire a similar transcriptional identity as those in the brain during aging that includes altered expression of genes with roles in microglia-neuron interactions and inflammation. By 29 months of age, spinal cord microglia exhibit additional and unique transcriptional changes known and predicted to cause senescence and to alter lysosomal and ribosomal regulation. Altogether, this work provides the foundation to target microglia to ameliorate aged-related changes in the spinal cord, and particularly on the motor circuit.
Collapse
Affiliation(s)
- Ryan Castro
- Neuroscience Graduate Program, Brown University, Providence, Rhode Island, USA
| | - Mikayla Lopes
- Molecular Biology, Cell Biology & Biochemistry Graduate Program, Brown University, Providence, Rhode Island, USA
| | - Lindsay De Biase
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, USA
| | - Gregorio Valdez
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, USA
- Center for Translational Neuroscience, Robert J. and Nancy D. Carney Institute for Brain Science and Brown Institute for Translational Science, Brown University, Providence, Rhode Island, USA
- Department of Neurology, Warren Alpert Medical School of Medicine, Brown University, Providence, USA
| |
Collapse
|
2
|
Starinets A, Ponomarenko A, Tyrtyshnaia A, Manzhulo I. Synaptamide modulates glial and neurotransmitter activity in the spinal cord during neuropathic pain. J Chem Neuroanat 2023; 134:102361. [PMID: 37935251 DOI: 10.1016/j.jchemneu.2023.102361] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/03/2023] [Accepted: 11/03/2023] [Indexed: 11/09/2023]
Abstract
N-docosahexaenoylethanolamine, or synaptamide, is an endogenous metabolite of docosahexaenoic acid that is known for synaptogenic and neurogenic effects. In our previous studies we have shown that synaptamide attenuates neuropathic pain, facilitates remyelination, and reduces neuroinflammation after the chronic constriction injury (CCI) of the sciatic nerve in rats. In the current study, we show that daily synaptamide administration (4 mg/kg/day) within 14 days post-surgery: (1) decreases micro- and astroglia activity in the dorsal and ventral horns of the lumbar spinal cord; (2) modulates pro-inflammatory (IL1β, IL6) and anti-inflammatory (IL4, IL10) cytokine level in the serum and spinal cord; (3) leads to a rise in synaptamide and anandamide concentration in the spinal cord; (4) enhances IL10, CD206 and N-acylethanolamine-hydrolyzing acid amidase synthesis in macrophage cell culture following LPS-induced inflammation. Thus, the ability of synaptamide to modulate glial and cytokine activity indicates its potential for implementation in the treatment peripheral nerve injury.
Collapse
Affiliation(s)
- Anna Starinets
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok 690041, Russia
| | - Arina Ponomarenko
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok 690041, Russia
| | - Anna Tyrtyshnaia
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok 690041, Russia
| | - Igor Manzhulo
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok 690041, Russia.
| |
Collapse
|
3
|
Shang J, Ma C, Ding H, Gu G, Zhang J, Wang M, Fang K, Wei Z, Feng S. Development and validation of a differentiation-related signature based on single-cell RNA sequencing data of immune cells in spinal cord injury. Heliyon 2023; 9:e19853. [PMID: 37809933 PMCID: PMC10559254 DOI: 10.1016/j.heliyon.2023.e19853] [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: 04/11/2023] [Revised: 08/02/2023] [Accepted: 09/04/2023] [Indexed: 10/10/2023] Open
Abstract
Background After spinal cord injury (SCI), the native immune surveillance function of the central nervous system is activated, resulting in a substantial infiltration of immune cells into the affected tissue. While numerous studies have explored the transcriptome data following SCI and revealed certain diagnostic biomarkers, there remains a paucity of research pertaining the identification of immune subtypes and molecular markers related to the immune system post-spinal cord injury using single-cell sequencing data of immune cells. Methods The researchers conducted an analysis of spinal cord samples obtained at three time points (3,10, and 21 days) following SCI using the GSE159638 dataset. The SCI subsets were delineated through pseudo-time analysis, and differentiation related genes were identified after principal component analysis (PCA), cell clustering, and annotation techniques. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were employed to assess the differentiation-related genes (DRGs) across different subsets. The molecular subtypes of SCI were determined using consensus clustering analysis. To further explore and validate the correlation between the molecular subtypes and the immune microenvironment, the CIBERSORT algorithm was employed. High-value diagnostic gene markers were identified using LASSO regression, and their diagnostic sensitivity was assessed using receiver operating characteristic curves (ROC) and quantitative real-time polymerase chain reaction (qRT-PCR). Results Three SCI subsets were obtained, and differentiation-related genes were characterized. Within these subsets, two distinct molecular subtypes, namely C1 and C2, were identified. These subtypes demonstrated significant variations in terms of immune cell infiltration levels and the expression of immune checkpoint genes. Through further analysis, three candidate biomarkers (C1qa, Lgals3 and Cd63) were identified and subsequently validated. Conclusions Our study revealed a diverse immune microenvironment in SCI samples, highlighting the potential significance of C1qa, Lgals3 and Cd63 as immune biomarkers for diagnosing SCI. Moreover, the identification of immune checkpoints corresponding to the two molecular subtypes suggests their potential as targets for immunotherapy to enhance SCI repair in future interventions.
Collapse
Affiliation(s)
- Jun Shang
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- Department of Orthopaedics, The Second Hospital of Shandong University, Shandong University, Jinan, Shandong, China
| | - Chao Ma
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Han Ding
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Guangjin Gu
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Jianping Zhang
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Min Wang
- Tianjin Key Laboratory of Lung Cancer Metastasis and the Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Ke Fang
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhijian Wei
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Shiqing Feng
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- Department of Orthopaedics, The Second Hospital of Shandong University, Shandong University, Jinan, Shandong, China
| |
Collapse
|
4
|
Zhu Y, Luan C, Gong L, Gu Y, Wang X, Sun H, Chen Z, Zhou Q, Liu C, Shan Q, Gu X, Zhou S. SnRNA-seq reveals the heterogeneity of spinal ventral horn and mechanism of motor neuron axon regeneration. iScience 2023; 26:107264. [PMID: 37502257 PMCID: PMC10368823 DOI: 10.1016/j.isci.2023.107264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 05/02/2023] [Accepted: 06/27/2023] [Indexed: 07/29/2023] Open
Abstract
Spinal motor neurons, the distinctive neurons of the central nervous system, extend into the peripheral nervous system and have outstanding ability of axon regeneration after injury. Here, we explored the heterogeneity of spinal ventral horn cells after rat sciatic nerve crush via single-nuclei RNA sequencing. Interestingly, regeneration mainly occurred in a Sncg+ and Anxa2+ motor neuron subtype (MN2) surrounded by a newly emerged microglia subtype (Mg6) after injury. Subsequently, microglia depletion slowed down the regeneration of sciatic nerve. OPCs were also involved into the regeneration process. Knockdown of Cacna2d2 in vitro and systemic blocking of Cacna2d2 in vivo improved the axon growth ability, hinting us the importance of Ca2+. Ultimately, we proposed three possible phases of motor neuron axon regeneration: preparation stage, early regeneration stage, and regeneration stage. Taken together, our study provided a resource for deciphering the underlying mechanism of motor neuron axon regeneration in a single cell dimension.
Collapse
Affiliation(s)
- Ye Zhu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300000, China
| | - Chengcheng Luan
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300000, China
| | - Leilei Gong
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu 226001, China
| | - Yun Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu 226001, China
| | - Xinghui Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu 226001, China
| | - Hualin Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu 226001, China
| | - Zhifeng Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu 226001, China
| | - Qiang Zhou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu 226001, China
| | - Chang Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu 226001, China
| | - Qi Shan
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300000, China
| | - Xiaosong Gu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300000, China
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu 226001, China
| | - Songlin Zhou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu 226001, China
| |
Collapse
|
5
|
He W, Wang Y, Zhang Y, Zhang Y, Zhou J. The status of knowledge on migraines: The role of microglia. J Neuroimmunol 2023; 381:578118. [PMID: 37295033 DOI: 10.1016/j.jneuroim.2023.578118] [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: 02/28/2023] [Revised: 05/01/2023] [Accepted: 05/28/2023] [Indexed: 06/11/2023]
Abstract
Migraines are a considerable social problem and economic burden worldwide. Current acute treatments are based on inhibiting meningeal neurogenic inflammation which has poor results in some patients, whereas the site of action of prophylactic medicines are unknown; therefore, exploring new treatment mechanisms and methods is increasingly needed. Recent evidence suggests that microglia and microglia-mediated neuroinflammation are important in migraine pathogenesis. In the cortical spreading depression (CSD) migraine model, microglia were activated after multiple CSD stimulations, suggesting that microglial activation may be associated with recurrent attacks of migraine with aura. In the nitroglycerin-induced chronic migraine model, the microglial response to extracellular stimuli leads to the activation of surface purine receptors P2X4、P2X7、P2Y12, which mediate signal transduction through intracellular signalling cascades, such as the BDNF/TrkB, NLRP3/IL-1β and RhoA/ROCK signalling pathways, and release inflammatory mediators and cytokines that enhance pain by increasing the excitability of nearby neurons. Inhibition of the expression or function of these microglial receptors and pathways inhibits the abnormal excitability of TNC (trigeminal nucleus caudalis) neurons and intracranial as well as extracranial hyperalgesia in migraine animal models. These findings suggest that microglia may be central in migraine recurrent attacks and a potential target for the treatment of chronic headaches.
Collapse
Affiliation(s)
- Wei He
- Department of Neurology, The First Branch of The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yanyun Wang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yixin Zhang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yinan Zhang
- Department of Neurology, The First Branch of The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiying Zhou
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
6
|
Zhang L, Liu J, Zhou C. Current aspects of small extracellular vesicles in pain process and relief. Biomater Res 2023; 27:78. [PMID: 37563666 PMCID: PMC10416402 DOI: 10.1186/s40824-023-00417-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/04/2023] [Indexed: 08/12/2023] Open
Abstract
Small extracellular vesicles (sEVs) have been identified as a noteworthy paracrine mechanism of intercellular communication in diagnosing and managing neurological disorders. Current research suggests that sEVs play a pivotal role in the pathological progression of pain, emphasizing their critical function in the pathological progression of pain in acute and chronic pain models. By facilitating the transfer of diverse molecules, such as proteins, nucleic acids, and metabolites, sEVs can modulate pain signaling transmission in both the central and peripheral nervous systems. Furthermore, the unique molecules conveyed by sEVs in pain disorders indicate their potential as diagnostic biomarkers. The application of sEVs derived from mesenchymal stem cells (MSCs) in regenerative pain medicine has emerged as a promising strategy for pain management. Moreover, modified sEVs have garnered considerable attention in the investigation of pathological processes and therapeutic interventions. This review presents a comprehensive overview of the current knowledge regarding the involvement of sEVs in pain pathogenesis and treatment. Nevertheless, additional research is imperative to facilitate their clinical implementation. Schematic diagram of sEVs in the biogenesis, signal transmission, diagnosis, and treatment of pain disorders. Small extracellular vesicles (sEVs) are secreted by multiple cells, loading with various biomolecules, such as miRNAs, transmembrane proteins, and amino acids. They selectively target other cells and regulating pain signal transmission. The composition of sEVs can serve as valuable biomarkers for pain diagnosis. In particular, mesenchymal stem cell-derived sEVs have shown promise as regenerative medicine for managing multiple pain disorders. Furthermore, by modifying the structure or contents of sEVs, they could potentially be used as a potent analgesic method.
Collapse
Affiliation(s)
- Lanyu Zhang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Anesthesia & Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jin Liu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Cheng Zhou
- Laboratory of Anesthesia & Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
7
|
Gu X, Chen A, You M, Guo H, Tan S, He Q, Hu B. Extracellular vesicles: a new communication paradigm of complement in neurological diseases. Brain Res Bull 2023; 199:110667. [PMID: 37192717 DOI: 10.1016/j.brainresbull.2023.110667] [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: 12/10/2022] [Revised: 03/25/2023] [Accepted: 05/13/2023] [Indexed: 05/18/2023]
Abstract
The complement system is crucial to the innate immune system. It has the function of destroying pathogens by activating the classical, alternative, and lectin pathways. The complement system is important in nervous system diseases such as cerebrovascular and neurodegenerative diseases. Activation of the complement system involves a series of intercellular signaling and cascade reactions. However, research on the source and transport mechanisms of the complement system in neurological diseases is still in its infancy. Studies have increasingly found that extracellular vesicles (EVs), a classic intercellular communication paradigm, may play a role in complement signaling disorders. Here, we systematically review the EV-mediated activation of complement pathways in different neurological diseases. We also discuss the prospect of EVs as future immunotherapy targets.
Collapse
Affiliation(s)
- Xinmei Gu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022
| | - Anqi Chen
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022
| | - Mingfeng You
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022
| | - Hongxiu Guo
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022
| | - Senwei Tan
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022
| | - Quanwei He
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022.
| | - Bo Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022.
| |
Collapse
|
8
|
Castro RW, Lopes MC, Settlage RE, Valdez G. Aging alters mechanisms underlying voluntary movements in spinal motor neurons of mice, primates, and humans. JCI Insight 2023; 8:e168448. [PMID: 37154159 PMCID: PMC10243831 DOI: 10.1172/jci.insight.168448] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 03/15/2023] [Indexed: 05/10/2023] Open
Abstract
Spinal motor neurons have been implicated in the loss of motor function that occurs with advancing age. However, the cellular and molecular mechanisms that impair the function of these neurons during aging remain unknown. Here, we show that motor neurons do not die in old female and male mice, rhesus monkeys, and humans. Instead, these neurons selectively and progressively shed excitatory synaptic inputs throughout the soma and dendritic arbor during aging. Thus, aged motor neurons contain a motor circuitry with a reduced ratio of excitatory to inhibitory synapses that may be responsible for the diminished ability to activate motor neurons to commence movements. An examination of the motor neuron translatome (ribosomal transcripts) in male and female mice reveals genes and molecular pathways with roles in glia-mediated synaptic pruning, inflammation, axonal regeneration, and oxidative stress that are upregulated in aged motor neurons. Some of these genes and pathways are also found altered in motor neurons affected with amyotrophic lateral sclerosis (ALS) and responding to axotomy, demonstrating that aged motor neurons are under significant stress. Our findings show mechanisms altered in aged motor neurons that could serve as therapeutic targets to preserve motor function during aging.
Collapse
Affiliation(s)
- Ryan W. Castro
- Neuroscience Graduate Program
- Department of Molecular Biology, Cellular Biology, and Biochemistry
- Center for Translational Neuroscience, Robert J. and Nancy D. Carney Institute for Brain Science and Brown Institute for Translational Science, and
| | - Mikayla C. Lopes
- Department of Molecular Biology, Cellular Biology, and Biochemistry
- Center for Translational Neuroscience, Robert J. and Nancy D. Carney Institute for Brain Science and Brown Institute for Translational Science, and
- Molecular Biology, Cell Biology, and Biochemistry Graduate Program, Brown University, Providence, Rhode Island, USA
| | - Robert E. Settlage
- Department of Advanced Research Computing, Virginia Tech, Blacksburg, Virginia, USA
| | - Gregorio Valdez
- Department of Molecular Biology, Cellular Biology, and Biochemistry
- Center for Translational Neuroscience, Robert J. and Nancy D. Carney Institute for Brain Science and Brown Institute for Translational Science, and
- Department of Neurology, Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| |
Collapse
|
9
|
Yu L, Liu M, Li F, Wang Q, Wang M, So KF, Qu Y, Zhou L. Celsr2 Knockout Alleviates Inhibitory Synaptic Stripping and Benefits Motoneuron Survival and Axon Regeneration After Branchial Plexus Avulsion. Mol Neurobiol 2023; 60:1884-1900. [PMID: 36593433 PMCID: PMC9984348 DOI: 10.1007/s12035-022-03198-3] [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: 10/13/2022] [Accepted: 12/23/2022] [Indexed: 01/04/2023]
Abstract
Axotomy-induced synaptic stripping modulates survival and axon regeneration of injured motoneurons. Celsr2 is supposed to mediate homophilic interactions of neighboring cells during development, and its role in synaptic stripping remains unknow. In a model of brachial plexus avulsion, Celsr2 knockout improved functional recovery, motoneuron survival, and axon regeneration. Celsr2 was indicated to express in spinal motoneurons, excitatory and inhibitory interneurons, astrocytes, and a subset of oligodendrocytes using Celsr2LacZ mice. Double immunostaining showed that the coverage of inhibitory and excitatory vesicles on injured motoneurons were remarkably reduced after injury, whereas more inhibitory vesicles were maintained in Celsr2-/- mutants than control mice. In the ultrastructure, the density of inhibitory F-boutons on injured motoneurons was higher in Celsr2-/- mutants than controls. Conditional knockout of Celsr2 in astrocytes or oligodendrocytes showed the similar axotomy-induced synaptic withdrawal to the control. RNAseq of injured spinal samples identified 12 MHC I molecules with significant changes between Celsr2-/- and control mice. After injury, expression of MHC I surrounding injured motoneurons was increased, particularly high in Celsr2-/- mutants. In conclusion, Celsr2 knockout enhances MHC I signaling, alleviates inhibitory synaptic stripping cell-autonomously, and contributes to motoneuron survival and regeneration, and Celsr2 is a potential target for neural repair.
Collapse
Affiliation(s)
- Lingtai Yu
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangzhou, 510632, People's Republic of China
| | - Mengfan Liu
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangzhou, 510632, People's Republic of China
| | - Fuxiang Li
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangzhou, 510632, People's Republic of China
| | - Qianghua Wang
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangzhou, 510632, People's Republic of China
| | - Meizhi Wang
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangzhou, 510632, People's Republic of China
| | - Kwok-Fai So
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangzhou, 510632, People's Republic of China.,Department of Neurology and Stroke Center, The First Affiliated Hospital & Clinical Neuroscience Institute of Jinan University, Guangzhou, 510632, People's Republic of China.,Neuroscience and Neurorehabilitation Institute, University of Health and Rehabilitation Sciences, Qingdao, 266071, Shandong, People's Republic of China.,Co-Innovation Center of Neuroregeneration, Nantong University, Jiangsu, People's Republic of China
| | - Yibo Qu
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangzhou, 510632, People's Republic of China.,Co-Innovation Center of Neuroregeneration, Nantong University, Jiangsu, People's Republic of China
| | - Libing Zhou
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangzhou, 510632, People's Republic of China. .,Department of Neurology and Stroke Center, The First Affiliated Hospital & Clinical Neuroscience Institute of Jinan University, Guangzhou, 510632, People's Republic of China. .,Neuroscience and Neurorehabilitation Institute, University of Health and Rehabilitation Sciences, Qingdao, 266071, Shandong, People's Republic of China. .,Co-Innovation Center of Neuroregeneration, Nantong University, Jiangsu, People's Republic of China.
| |
Collapse
|
10
|
Liu Y, Zhang F, Sun Q, Liang L. Adalimumab combined with erythropoietin improves recovery from spinal cord injury by suppressing microglial M1 polarization-mediated neural inflammation and apoptosis. Inflammopharmacology 2023; 31:887-897. [PMID: 36642757 DOI: 10.1007/s10787-022-01090-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 10/17/2022] [Indexed: 01/16/2023]
Abstract
OBJECTIVE Adalimumab (ADM), a humanized antibody against tumour necrosis factor (TNF), is widely applied in treating inflammatory and autoimmune diseases, but its usage in spinal cord injury (SCI) is rarely reported. Hence, this study aimed to explore the effect of ADM with or without erythropoietin (EPO) on microglial polarization, neuroinflammation, neural apoptosis, and functional recovery in SCI. METHODS Primary microglia were stimulated with lipopolysaccharide (LPS) and then treated with ADM, EPO, or ADM combined with EPO. Then, primary neurons were incubated in the microglial culture medium. SCI rats were established and then treated with ADM, EPO or ADM combined with EPO. RESULTS ADM suppressed LPS-induced microglial M1 polarization, as reflected by downregulated iNOS and CD86 expression, and neuroinflammation, as reflected by decreased TNF-α, IL-1β, and IL-6 expression, in a dose-dependent manner. Moreover, ADM inhibited microglia-induced neural apoptosis, as reflected by TUNEL assay results and the expression of apoptotic markers (C-Caspase3 and Bcl2), in a dose-dependent manner. EPO monotherapy displayed an effect similar to that of ADM monotherapy. Furthermore, ADM combined with EPO therapy exhibited greater effects than either monotherapy in terms of inhibiting microglial M1 polarization, neuroinflammation, and neural apoptosis. In vivo experiments confirmed the findings of the in vitro experiments and showed that ADM combined with EPO improved SCI functional recovery and neural injury compared with monotherapy. CONCLUSION ADM combined with EPO improves recovery from SCI by suppressing microglial M1 polarization-mediated neural inflammation and apoptosis.
Collapse
Affiliation(s)
- Yanming Liu
- Department of Physical Medicine and Rehabilitation, Zibo Central Hospital, No. 54 Communist Youth League West Road, Zhangdian District, Zibo, 255000, Shandong, China
| | - Feiyue Zhang
- Obstetrics Department, Zibo Central Hospital, Zibo, Shandong, China
| | - Qi Sun
- Department of Scientific Research and International Cooperation, Zibo Central Hospital, Zibo, Shandong, China
| | - Lichao Liang
- Department of Physical Medicine and Rehabilitation, Zibo Central Hospital, No. 54 Communist Youth League West Road, Zhangdian District, Zibo, 255000, Shandong, China.
| |
Collapse
|
11
|
Liao Y, Guo C, Wen A, Bai M, Ran Z, Hu J, Wang J, Yang J, Ding Y. Frankincense-Myrrh treatment alleviates neuropathic pain via the inhibition of neuroglia activation mediated by the TLR4/MyD88 pathway and TRPV1 signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 108:154540. [PMID: 36379093 DOI: 10.1016/j.phymed.2022.154540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 10/26/2022] [Accepted: 11/04/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Neuroglia are important modulators of neuronal functionality, and thus play an integral role in the pathogenesis and treatment of neuropathic pain (NP). According to traditional Chinese medicine, Frankincense-Myrrh is capable of "activating blood and dissipating blood stasis", and as such these two biological compounds are commonly used to treat NP, however, the mechanisms underlying the efficacy of such treatment are unclear. PURPOSE This study aimed to further elucidate the protective effects associated with the Frankincense-Myrrh treatment of NP. METHODS A chronic sciatic nerve compression injury (CCI) model of NP was established, after which animals were gavaged with Frankincense, Myrrh, Frankincense-Myrrh, or the positive control drug pregabalin for 14 days. Network pharmacology approaches were used to identify putative pathways and targets associated with the Frankincense-Myrrh-mediated treatment of NP, after which these targets were subjected to in-depth analyses. The impact of TLR4 blockade on NP pathogenesis was assessed by intrathecally administering a TLR4 antagonist (LRU) or the MyD88 homodimerization inhibitory peptide (MIP). RESULTS Significant alleviation of thermal and mechanical hypersensitivity in response to Frankincense and Myrrh treatment was observed in NP model mice, while network pharmacology analyses suggested that the pathogenesis of NP may be related to TLR4/MyD88-mediated neuroinflammation. Consistently, Frankincense-Myrrh treatment was found to reduce TLR4, MyD88, and p-p65 expression in spinal dorsal horn neuroglia from treated animals, in addition to inhibiting neuronal TRPV1 and inflammatory factor expression. Intrathecal LRU and MIP delivery were sufficient to alleviate thermal and mechanical hyperalgesia in these CCI model mice, with concomitant reductions in neuronal TRPV1 expression and neuroglial activation in the spinal dorsal horn. CONCLUSION These data suggest that Frankincense-Myrrh treatment was sufficient to alleviate NP in part via inhibiting TLR4/MyD88 pathway and TRPV1 signaling activity. Blocking TLR4 and MyD88 activation may thus hold value as a means of treating NP.
Collapse
Affiliation(s)
- Yucheng Liao
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, China; School of Pharmacy, Xinjiang Medical University, Urumqi, China
| | - Chao Guo
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Aidong Wen
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Min Bai
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zheng Ran
- School of Pharmacy, Xinjiang Medical University, Urumqi, China
| | - Junping Hu
- School of Pharmacy, Xinjiang Medical University, Urumqi, China
| | - Jingwen Wang
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| | - Jianhua Yang
- School of Pharmacy, Xinjiang Medical University, Urumqi, China; Department of Pharmacy, The First Affiliated Hospital, Xinjiang Medical University, Urumqi, China.
| | - Yi Ding
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
12
|
Theme 04 - In Vivo Experimetal Models. Amyotroph Lateral Scler Frontotemporal Degener 2022. [DOI: 10.1080/21678421.2022.2120680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
13
|
Myatich A, Haque A, Sole C, Banik NL. Clemastine in remyelination and protection of neurons and skeletal muscle after spinal cord injury. Neural Regen Res 2022; 18:940-946. [PMID: 36254972 PMCID: PMC9827778 DOI: 10.4103/1673-5374.355749] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Spinal cord injuries affect nearly five to ten individuals per million every year. Spinal cord injury causes damage to the nerves, muscles, and the tissue surrounding the spinal cord. Depending on the severity, spinal injuries are linked to degeneration of axons and myelin, resulting in neuronal impairment and skeletal muscle weakness and atrophy. The protection of neurons and promotion of myelin regeneration during spinal cord injury is important for recovery of function following spinal cord injury. Current treatments have little to no effect on spinal cord injury and neurogenic muscle loss. Clemastine, an Food and Drug Administration-approved antihistamine drug, reduces inflammation, protects cells, promotes remyelination, and preserves myelin integrity. Recent clinical evidence suggests that clemastine can decrease the loss of axons after spinal cord injury, stimulating the differentiation of oligodendrocyte progenitor cells into mature oligodendrocytes that are capable of myelination. While clemastine can aid not only in the remyelination and preservation of myelin sheath integrity, it also protects neurons. However, its role in neurogenic muscle loss remains unclear. This review discusses the pathophysiology of spinal cord injury, and the role of clemastine in the protection of neurons, myelin, and axons as well as attenuation of skeletal muscle loss following spinal cord injury.
Collapse
Affiliation(s)
- Ali Myatich
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA,Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, USA
| | - Azizul Haque
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA,Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, USA,Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, USA,Correspondence to: Azizul Haque, ; Naren L. Banik, .
| | - Christopher Sole
- Department of Health and Human Performance, The Citadel, Charleston, SC, USA
| | - Naren L. Banik
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA,Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, USA,Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, USA,Correspondence to: Azizul Haque, ; Naren L. Banik, .
| |
Collapse
|
14
|
Chu XL, Song XZ, Li Q, Li YR, He F, Gu XS, Ming D. Basic mechanisms of peripheral nerve injury and treatment via electrical stimulation. Neural Regen Res 2022; 17:2185-2193. [PMID: 35259827 PMCID: PMC9083151 DOI: 10.4103/1673-5374.335823] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Previous studies on the mechanisms of peripheral nerve injury (PNI) have mainly focused on the pathophysiological changes within a single injury site. However, recent studies have indicated that within the central nervous system, PNI can lead to changes in both injury sites and target organs at the cellular and molecular levels. Therefore, the basic mechanisms of PNI have not been comprehensively understood. Although electrical stimulation was found to promote axonal regeneration and functional rehabilitation after PNI, as well as to alleviate neuropathic pain, the specific mechanisms of successful PNI treatment are unclear. We summarize and discuss the basic mechanisms of PNI and of treatment via electrical stimulation. After PNI, activity in the central nervous system (spinal cord) is altered, which can limit regeneration of the damaged nerve. For example, cell apoptosis and synaptic stripping in the anterior horn of the spinal cord can reduce the speed of nerve regeneration. The pathological changes in the posterior horn of the spinal cord can modulate sensory abnormalities after PNI. This can be observed in cases of ectopic discharge of the dorsal root ganglion leading to increased pain signal transmission. The injured site of the peripheral nerve is also an important factor affecting post-PNI repair. After PNI, the proximal end of the injured site sends out axial buds to innervate both the skin and muscle at the injury site. A slow speed of axon regeneration leads to low nerve regeneration. Therefore, it can take a long time for the proximal nerve to reinnervate the skin and muscle at the injured site. From the perspective of target organs, long-term denervation can cause atrophy of the corresponding skeletal muscle, which leads to abnormal sensory perception and hyperalgesia, and finally, the loss of target organ function. The mechanisms underlying the use of electrical stimulation to treat PNI include the inhibition of synaptic stripping, addressing the excessive excitability of the dorsal root ganglion, alleviating neuropathic pain, improving neurological function, and accelerating nerve regeneration. Electrical stimulation of target organs can reduce the atrophy of denervated skeletal muscle and promote the recovery of sensory function. Findings from the included studies confirm that after PNI, a series of physiological and pathological changes occur in the spinal cord, injury site, and target organs, leading to dysfunction. Electrical stimulation may address the pathophysiological changes mentioned above, thus promoting nerve regeneration and ameliorating dysfunction.
Collapse
Affiliation(s)
- Xiao-Lei Chu
- Academy of Medical Engineering and Translational Medicine, Tianjin University; Department of Rehabilitation, Tianjin Hospital, Tianjin, China
| | - Xi-Zi Song
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Qi Li
- Academy of Medical Engineering and Translational Medicine, Tianjin University; Department of Rehabilitation, Tianjin Hospital, Tianjin, China
| | - Yu-Ru Li
- College of Exercise & Health Sciences, Tianjin University of Sport, Tianjin, China
| | - Feng He
- College of Precision Instruments & Optoelectronics Engineering, Tianjin University, Tianjin, China
| | - Xiao-Song Gu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Dong Ming
- Academy of Medical Engineering and Translational Medicine; College of Precision Instruments & Optoelectronics Engineering, Tianjin University, Tianjin, China
| |
Collapse
|
15
|
Pottorf TS, Rotterman TM, McCallum WM, Haley-Johnson ZA, Alvarez FJ. The Role of Microglia in Neuroinflammation of the Spinal Cord after Peripheral Nerve Injury. Cells 2022; 11:cells11132083. [PMID: 35805167 PMCID: PMC9265514 DOI: 10.3390/cells11132083] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 06/26/2022] [Accepted: 06/27/2022] [Indexed: 12/12/2022] Open
Abstract
Peripheral nerve injuries induce a pronounced immune reaction within the spinal cord, largely governed by microglia activation in both the dorsal and ventral horns. The mechanisms of activation and response of microglia are diverse depending on the location within the spinal cord, type, severity, and proximity of injury, as well as the age and species of the organism. Thanks to recent advancements in neuro-immune research techniques, such as single-cell transcriptomics, novel genetic mouse models, and live imaging, a vast amount of literature has come to light regarding the mechanisms of microglial activation and alluding to the function of microgliosis around injured motoneurons and sensory afferents. Herein, we provide a comparative analysis of the dorsal and ventral horns in relation to mechanisms of microglia activation (CSF1, DAP12, CCR2, Fractalkine signaling, Toll-like receptors, and purinergic signaling), and functionality in neuroprotection, degeneration, regeneration, synaptic plasticity, and spinal circuit reorganization following peripheral nerve injury. This review aims to shed new light on unsettled controversies regarding the diversity of spinal microglial-neuronal interactions following injury.
Collapse
Affiliation(s)
- Tana S. Pottorf
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA; (T.S.P.); (W.M.M.); (Z.A.H.-J.)
| | - Travis M. Rotterman
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30318, USA;
| | - William M. McCallum
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA; (T.S.P.); (W.M.M.); (Z.A.H.-J.)
| | - Zoë A. Haley-Johnson
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA; (T.S.P.); (W.M.M.); (Z.A.H.-J.)
| | - Francisco J. Alvarez
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA; (T.S.P.); (W.M.M.); (Z.A.H.-J.)
- Correspondence:
| |
Collapse
|
16
|
Salvany S, Casanovas A, Piedrafita L, Gras S, Calderó J, Esquerda JE. Accumulation of misfolded SOD1 outlines distinct patterns of motor neuron pathology and death during disease progression in a SOD1 G93A mouse model of amyotrophic lateral sclerosis. Brain Pathol 2022; 32:e13078. [PMID: 35584812 PMCID: PMC9616096 DOI: 10.1111/bpa.13078] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 04/11/2022] [Indexed: 12/12/2022] Open
Abstract
Early misfolded superoxide dismutase 1 (mfSOD1) accumulation, motor neuron (MN) degeneration, and microgliosis are hallmark pathological features in SOD1G93A amyotrophic lateral sclerosis (ALS) mice. Because of the different vulnerabilities of distinct MN subtypes, degenerating and surviving MNs coexist in different proportions during disease progression. By examining the expression of misfolded conformers of SOD1 using specific antibodies, we defined distinct MN phenotypes that were evaluated during disease progression and the local neuroinflammatory reaction. The most severe phenotype corresponded to somata of fast‐twitch subtype MNs, which exhibited highly positive mfSOD1 immunostaining and an extreme degree of vacuolar degeneration. Vacuoles, which are of mitochondrial origin, contain mfSOD1 in conjunction with nonmitochondrial proteins, such as chromogranin, CD81, and flotillin. The fusion of ER‐derived vesicles enriched in mfSOD1 with outer mitochondrial membranes is thought to be the primary mechanism for vacuole formation. In addition, the ulterior coalescence of enlarged mitochondria may lead to the formation of giant vacuoles. Vacuolar degeneration is a transient degenerative process occurring early during the presymptomatic stages of the disease in ALS mice. Some vacuolated MNs are also positive for pMLKL, the effector protein of necroptosis. This indicates a newly described mechanism in which extracellular vesicles derived from damaged MNs, via cellular secretion or necroptotic disruption, may be the triggers for initiating neuroinflammation, glial‐mediated neurotoxicity, and disease spreading. Furthermore, as MN degeneration in mutant SOD1 mice is noncell autonomous, the effects of experimentally increasing or decreasing the microglial response on the expression of MN phenotypes were also evaluated, demonstrating bidirectional cross talk signaling between the degree of expression of mfSOD1 and local neuroinflammation. More detailed knowledge regarding these processes occurring long before the end stages of the disease is necessary to identify novel molecular targets for future preclinical testing.
Collapse
Affiliation(s)
- Sara Salvany
- Patologia Neuromuscular Experimental, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Catalonia, Spain
| | - Anna Casanovas
- Patologia Neuromuscular Experimental, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Catalonia, Spain
| | - Lídia Piedrafita
- Patologia Neuromuscular Experimental, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Catalonia, Spain
| | - Sílvia Gras
- Patologia Neuromuscular Experimental, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Catalonia, Spain
| | - Jordi Calderó
- Patologia Neuromuscular Experimental, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Catalonia, Spain
| | - Josep E Esquerda
- Patologia Neuromuscular Experimental, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Catalonia, Spain
| |
Collapse
|
17
|
Zeng J, Bao T, Yang K, Zhu X, Wang S, Xiang W, Ge A, Zeng L, Ge J. The mechanism of microglia-mediated immune inflammation in ischemic stroke and the role of natural botanical components in regulating microglia: A review. Front Immunol 2022; 13:1047550. [PMID: 36818470 PMCID: PMC9933144 DOI: 10.3389/fimmu.2022.1047550] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 12/05/2022] [Indexed: 02/05/2023] Open
Abstract
Ischemic stroke (IS) is one of the most fatal diseases. Neuroimmunity, inflammation, and oxidative stress play important roles in various complex mechanisms of IS. In particular, the early proinflammatory response resulting from the overactivation of resident microglia and the infiltration of circulating monocytes and macrophages in the brain after cerebral ischemia leads to secondary brain injury. Microglia are innate immune cells in the brain that constantly monitor the brain microenvironment under normal conditions. Once ischemia occurs, microglia are activated to produce dual effects of neurotoxicity and neuroprotection, and the balance of the two effects determines the fate of damaged neurons. The activation of microglia is defined as the classical activation (M1 type) or alternative activation (M2 type). M1 type microglia secrete pro-inflammatory cytokines and neurotoxic mediators to exacerbate neuronal damage, while M2 type microglia promote a repairing anti-inflammatory response. Fine regulation of M1/M2 microglial activation to minimize damage and maximize protection has important therapeutic value. This review focuses on the interaction between M1/M2 microglia and other immune cells involved in the regulation of IS phenotypic characteristics, and the mechanism of natural plant components regulating microglia after IS, providing novel candidate drugs for regulating microglial balance and IS drug development.
Collapse
Affiliation(s)
- Jinsong Zeng
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Tingting Bao
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Kailin Yang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | | | - Shanshan Wang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Wang Xiang
- Department of Rheumatology, The First People's Hospital Changde City, Changde, Hunan, China
| | - Anqi Ge
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Liuting Zeng
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Jinwen Ge
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China.,Hunan Academy of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
18
|
Wofford KL, Shultz RB, Burrell JC, Cullen DK. Neuroimmune interactions and immunoengineering strategies in peripheral nerve repair. Prog Neurobiol 2022; 208:102172. [PMID: 34492307 PMCID: PMC8712351 DOI: 10.1016/j.pneurobio.2021.102172] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 08/11/2021] [Accepted: 09/02/2021] [Indexed: 01/03/2023]
Abstract
Peripheral nerve injuries result in disrupted cellular communication between the central nervous system and somatic distal end targets. The peripheral nervous system is capable of independent and extensive regeneration; however, meaningful target muscle reinnervation and functional recovery remain limited and may result in chronic neuropathic pain and diminished quality of life. Macrophages, the primary innate immune cells of the body, are critical contributors to regeneration of the injured peripheral nervous system. However, in some clinical scenarios, macrophages may fail to provide adequate support with optimal timing, duration, and location. Here, we review the history of immunosuppressive and immunomodulatory strategies to treat nerve injuries. Thereafter, we enumerate the ways in which macrophages contribute to successful nerve regeneration. We argue that implementing macrophage-based immunomodulatory therapies is a promising treatment strategy for nerve injuries across a wide range of clinical presentations.
Collapse
Affiliation(s)
- Kathryn L Wofford
- Center for Brain Injury & Repair, Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA, 19104, United States; Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, 19104, United States
| | - Robert B Shultz
- Center for Brain Injury & Repair, Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA, 19104, United States; Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, 19104, United States; Axonova Medical, LLC, Philadelphia, PA, 19104, United States
| | - Justin C Burrell
- Center for Brain Injury & Repair, Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA, 19104, United States; Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, 19104, United States; Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - D Kacy Cullen
- Center for Brain Injury & Repair, Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA, 19104, United States; Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, 19104, United States; Axonova Medical, LLC, Philadelphia, PA, 19104, United States; Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, United States.
| |
Collapse
|
19
|
Berkowitz S, Chapman J, Dori A, Gofrit SG, Maggio N, Shavit-Stein E. Complement and Coagulation System Crosstalk in Synaptic and Neural Conduction in the Central and Peripheral Nervous Systems. Biomedicines 2021; 9:biomedicines9121950. [PMID: 34944766 PMCID: PMC8698364 DOI: 10.3390/biomedicines9121950] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 12/12/2022] Open
Abstract
Complement and coagulation are both key systems that defend the body from harm. They share multiple features and are similarly activated. They each play individual roles in the systemic circulation in physiology and pathophysiology, with significant crosstalk between them. Components from both systems are mapped to important structures in the central nervous system (CNS) and peripheral nervous system (PNS). Complement and coagulation participate in critical functions in neuronal development and synaptic plasticity. During pathophysiological states, complement and coagulation factors are upregulated and can modulate synaptic transmission and neuronal conduction. This review summarizes the current evidence regarding the roles of the complement system and the coagulation cascade in the CNS and PNS. Possible crosstalk between the two systems regarding neuroinflammatory-related effects on synaptic transmission and neuronal conduction is explored. Novel treatment based on the modulation of crosstalk between complement and coagulation may perhaps help to alleviate neuroinflammatory effects in diseased states of the CNS and PNS.
Collapse
Affiliation(s)
- Shani Berkowitz
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan 5266202, Israel; (S.B.); (J.C.); (A.D.); (S.G.G.); (N.M.)
- Department of Neurology and Neurosurgery, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Joab Chapman
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan 5266202, Israel; (S.B.); (J.C.); (A.D.); (S.G.G.); (N.M.)
- Department of Neurology and Neurosurgery, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Robert and Martha Harden Chair in Mental and Neurological Diseases, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Amir Dori
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan 5266202, Israel; (S.B.); (J.C.); (A.D.); (S.G.G.); (N.M.)
- Department of Neurology and Neurosurgery, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Talpiot Medical Leadership Program, The Chaim Sheba Medical Center, Ramat Gan 6997801, Israel
| | - Shany Guly Gofrit
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan 5266202, Israel; (S.B.); (J.C.); (A.D.); (S.G.G.); (N.M.)
| | - Nicola Maggio
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan 5266202, Israel; (S.B.); (J.C.); (A.D.); (S.G.G.); (N.M.)
- Department of Neurology and Neurosurgery, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Talpiot Medical Leadership Program, The Chaim Sheba Medical Center, Ramat Gan 6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Efrat Shavit-Stein
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan 5266202, Israel; (S.B.); (J.C.); (A.D.); (S.G.G.); (N.M.)
- Department of Neurology and Neurosurgery, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Correspondence: ; Tel.: +972-50-921-0400
| |
Collapse
|
20
|
Salvany S, Casanovas A, Piedrafita L, Tarabal O, Hernández S, Calderó J, Esquerda JE. Microglial recruitment and mechanisms involved in the disruption of afferent synaptic terminals on spinal cord motor neurons after acute peripheral nerve injury. Glia 2021; 69:1216-1240. [PMID: 33386754 PMCID: PMC7986680 DOI: 10.1002/glia.23959] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022]
Abstract
Peripheral nerve section with subsequent disconnection of motor neuron (MN) cell bodies from their skeletal muscle targets leads to a rapid reactive response involving the recruitment and activation of microglia. In addition, the loss of afferent synapses on MNs occurs in concomitance with microglial reaction by a process described as synaptic stripping. However, the way in which postaxotomy-activated microglia adjacent to MNs are involved in synaptic removal is less defined. Here, we used confocal and electron microscopy to examine interactions between recruited microglial cells and presynaptic terminals in axotomized MNs between 1 and 15 days after sciatic nerve transection in mice. We did not observe any bulk engulfment of synaptic boutons by microglia. Instead, microglial cells internalized small membranous-vesicular fragments which originated from the acute disruption of synaptic terminals involving the activation of the necroptotic pathway. The presence of abundant extracellular vesicles in the perineuronal space after axotomy, together with the increased expression of phospho-mixed lineage kinase domain-like protein and, later, of extracellular vesicle markers, such as CD9, CD63, and flotillin, indicate that the vesicles mainly originated in synapses and were transferred to microglia. The upregulation of Rab7 and Rab10 in microglia interacting with injured MNs, indicated the activation of endocytosis. As activated microglia and synaptic boutons displayed positive C1q immunoreactivity, a complement-mediated opsonization may also contribute to microglial-mediated synaptic disruption. In addition to the relevance of our data in the context of neuroinflammation and MN disease, they should also be taken into account for understanding functional recovery after peripheral nerve injury.
Collapse
Affiliation(s)
- Sara Salvany
- Patologia Neuromuscular Experimental Departament de Medicina Experimental, Facultat de MedicinaUniversitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida)LleidaCataloniaSpain
| | - Anna Casanovas
- Patologia Neuromuscular Experimental Departament de Medicina Experimental, Facultat de MedicinaUniversitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida)LleidaCataloniaSpain
| | - Lídia Piedrafita
- Patologia Neuromuscular Experimental Departament de Medicina Experimental, Facultat de MedicinaUniversitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida)LleidaCataloniaSpain
| | - Olga Tarabal
- Patologia Neuromuscular Experimental Departament de Medicina Experimental, Facultat de MedicinaUniversitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida)LleidaCataloniaSpain
| | - Sara Hernández
- Patologia Neuromuscular Experimental Departament de Medicina Experimental, Facultat de MedicinaUniversitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida)LleidaCataloniaSpain
| | - Jordi Calderó
- Patologia Neuromuscular Experimental Departament de Medicina Experimental, Facultat de MedicinaUniversitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida)LleidaCataloniaSpain
| | - Josep E. Esquerda
- Patologia Neuromuscular Experimental Departament de Medicina Experimental, Facultat de MedicinaUniversitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida)LleidaCataloniaSpain
| |
Collapse
|