201
|
Fu SP, Chen SY, Pang QM, Zhang M, Wu XC, Wan X, Wan WH, Ao J, Zhang T. Advances in the research of the role of macrophage/microglia polarization-mediated inflammatory response in spinal cord injury. Front Immunol 2022; 13:1014013. [PMID: 36532022 PMCID: PMC9751019 DOI: 10.3389/fimmu.2022.1014013] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/18/2022] [Indexed: 12/04/2022] Open
Abstract
It is often difficult to regain neurological function following spinal cord injury (SCI). Neuroinflammation is thought to be responsible for this failure. Regulating the inflammatory response post-SCI may contribute to the recovery of neurological function. Over the past few decades, studies have found that macrophages/microglia are one of the primary effector cells in the inflammatory response following SCI. Growing evidence has documented that macrophages/microglia are plastic cells that can polarize in response to microenvironmental signals into M1 and M2 macrophages/microglia. M1 produces pro-inflammatory cytokines to induce inflammation and worsen tissue damage, while M2 has anti-inflammatory activities in wound healing and tissue regeneration. Recent studies have indicated that the transition from the M1 to the M2 phenotype of macrophage/microglia supports the regression of inflammation and tissue repair. Here, we will review the role of the inflammatory response and macrophages/microglia in SCI and repair. In addition, we will discuss potential molecular mechanisms that induce macrophage/microglia polarization, with emphasis on neuroprotective therapies that modulate macrophage/microglia polarization, which will provide new insights into therapeutic strategies for SCI.
Collapse
Affiliation(s)
- Sheng-Ping Fu
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China,Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Si-Yu Chen
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Qi-Ming Pang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Meng Zhang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Xiang-Chong Wu
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Xue Wan
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China,Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Wei-Hong Wan
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China,Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Jun Ao
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Tao Zhang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China,Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China,Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China,The Clinical Stem Cell Research Institute, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China,*Correspondence: Tao Zhang,
| |
Collapse
|
202
|
Astrocyte-derived sEVs alleviate fibrosis and promote functional recovery after spinal cord injury in rats. Int Immunopharmacol 2022; 113:109322. [DOI: 10.1016/j.intimp.2022.109322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 10/03/2022] [Accepted: 10/04/2022] [Indexed: 11/05/2022]
|
203
|
Paredes-Espinosa MB, Paluh JL. Human stem cell-derived neurons and neural circuitry therapeutics: Next frontier in spinal cord injury repair. Exp Biol Med (Maywood) 2022; 247:2142-2151. [PMID: 35974701 PMCID: PMC9837306 DOI: 10.1177/15353702221114099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Spinal cord injury (SCI) remains a life-altering event that devastates those injured and the families that support them. Numerous laboratories are engaged in preclinical and clinical trials to repair the injured spinal cord with stem cell-derived therapeutics. A new developmental paradigm reveals early bifurcation of brain or trunk neurons in mammals via neuromesodermal progenitors (NMPs) relevant to therapies requiring homotypic spinal cord neural populations. Human-induced pluripotent stem cell (hiPSC) NMP-derived spinal motor neurons generated ex vivo following this natural developmental route demonstrate robust survival in vivo when delivered as suspension grafts or as in vitro preformed encapsulated neuronal circuitry when transplanted into a rat C4-C5 hemicontusion injury site. Use of in vitro matured neurons avoids in vivo differentiation challenges of using pluripotent hiPSC or multipotent neural stem cell (NSC) or mesenchymal stem cell therapeutics. In this review, we provide an injury to therapeutics overview focusing on how stem cell and developmental fields are merging to generate exquisitely matched spinal motor neurons for SCI therapeutic studies. The complexity of the SCI microenvironment generated by trauma to neurons and vasculature, along with infiltrating inflammatory cells and scarring, underlies the challenging cytokine microenvironment that therapeutic cells encounter. An overview of evolving but limited stem cell-based SCI therapies that have progressed from preclinical to clinical trials illustrates the challenges and need for additional stem cell-based therapeutic approaches. The focus here on neurons describes how NMP-based neurotechnologies are advancing parallel strategies such as transplantation of preformed neuronal circuitry as well as human in vitro gastruloid multicellular models of trunk central and peripheral nervous system integration with organs. NMP-derived neurons are expected to be powerful drivers of the next generation of SCI therapeutics and integrate well with combination therapies that may utilize alternate biomimetic scaffolds for bridging injuries or flexible biodegradable electronics for electrostimulation.
Collapse
|
204
|
Hu Y, Cao K, Wang F, Wu W, Mai W, Qiu L, Luo Y, Ge WP, Sun B, Shi L, Zhu J, Zhang J, Wu Z, Xie Y, Duan S, Gao Z. Dual roles of hexokinase 2 in shaping microglial function by gating glycolytic flux and mitochondrial activity. Nat Metab 2022; 4:1756-1774. [PMID: 36536134 DOI: 10.1038/s42255-022-00707-5] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 11/04/2022] [Indexed: 12/24/2022]
Abstract
Microglia continuously survey the brain parenchyma and actively shift status following stimulation. These processes demand a unique bioenergetic programme; however, little is known about the metabolic determinants in microglia. By mining large datasets and generating transgenic tools, here we show that hexokinase 2 (HK2), the most active isozyme associated with mitochondrial membrane, is selectively expressed in microglia in the brain. Genetic ablation of HK2 reduced microglial glycolytic flux and energy production, suppressed microglial repopulation, and attenuated microglial surveillance and damage-triggered migration in male mice. HK2 elevation is prominent in immune-challenged or disease-associated microglia. In ischaemic stroke models, however, HK2 deletion promoted neuroinflammation and potentiated cerebral damages. The enhanced inflammatory responses after HK2 ablation in microglia are associated with aberrant mitochondrial function and reactive oxygen species accumulation. Our study demonstrates that HK2 gates both glycolytic flux and mitochondrial activity to shape microglial functions, changes of which contribute to metabolic abnormalities and maladaptive inflammation in brain diseases.
Collapse
Affiliation(s)
- Yaling Hu
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Kelei Cao
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Fang Wang
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Weiying Wu
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Weihao Mai
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Liyao Qiu
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Yuxiang Luo
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Woo-Ping Ge
- Chinese Institute for Brain Research, Beijing, Beijing, China
| | - Binggui Sun
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
- Department of Neurobiology and Department of Anesthesiology, the Children's Hospital, Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, China
| | - Ligen Shi
- Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Junming Zhu
- Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianmin Zhang
- Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhiying Wu
- Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yicheng Xie
- The Children's Hospital, Zhejiang, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Shumin Duan
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China.
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China.
| | - Zhihua Gao
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China.
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China.
| |
Collapse
|
205
|
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
|
206
|
Restoring After Central Nervous System Injuries: Neural Mechanisms and Translational Applications of Motor Recovery. Neurosci Bull 2022; 38:1569-1587. [DOI: 10.1007/s12264-022-00959-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 06/29/2022] [Indexed: 11/06/2022] Open
Abstract
AbstractCentral nervous system (CNS) injuries, including stroke, traumatic brain injury, and spinal cord injury, are leading causes of long-term disability. It is estimated that more than half of the survivors of severe unilateral injury are unable to use the denervated limb. Previous studies have focused on neuroprotective interventions in the affected hemisphere to limit brain lesions and neurorepair measures to promote recovery. However, the ability to increase plasticity in the injured brain is restricted and difficult to improve. Therefore, over several decades, researchers have been prompted to enhance the compensation by the unaffected hemisphere. Animal experiments have revealed that regrowth of ipsilateral descending fibers from the unaffected hemisphere to denervated motor neurons plays a significant role in the restoration of motor function. In addition, several clinical treatments have been designed to restore ipsilateral motor control, including brain stimulation, nerve transfer surgery, and brain–computer interface systems. Here, we comprehensively review the neural mechanisms as well as translational applications of ipsilateral motor control upon rehabilitation after CNS injuries.
Collapse
|
207
|
Fisher ES, Amarante MA, Lowry N, Lotz S, Farjood F, Temple S, Hill CE, Kiehl TR. Single cell profiling of CD45+ spinal cord cells reveals microglial and B cell heterogeneity and crosstalk following spinal cord injury. J Neuroinflammation 2022; 19:266. [PMID: 36333772 PMCID: PMC9635187 DOI: 10.1186/s12974-022-02627-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022] Open
Abstract
Abstract
Background
Immune cells play crucial roles after spinal cord injury (SCI). However, incomplete knowledge of immune contributions to injury and repair hinders development of SCI therapies. We leveraged single-cell observations to describe key populations of immune cells present in the spinal cord and changes in their transcriptional profiles from uninjured to subacute and chronic stages of SCI.
Methods
Deep-read single-cell sequencing was performed on CD45+ cells from spinal cords of uninjured and injured Swiss-webster mice. After T9 thoracic contusion, cells were collected 3-, 7-, and 60-day post-injury (dpi). Subpopulations of CD45+ immune cells were identified informatically, and their transcriptional responses characterized with time. We compared gene expression in spinal cord microglia and B cell subpopulations with those in published models of disease and injury. Microglia were compared with Disease Associated Microglia (DAM) and Injury Responsive Microglia (IRM). B cells were compared to developmental lineage states and to an Amyotrophic Lateral Sclerosis (ALS) model.
Results
In uninjured and 7 dpi spinal cord, most CD45+ cells isolated were microglia while chronically B cells predominated. B cells accumulating in the spinal cord following injury included immature B to mature stages and were predominantly found in the injury zone. We defined diverse subtypes of microglia and B cells with altered gene expression with time after SCI. Spinal cord microglia gene expression indicates differences from brain microglia at rest and in inflammatory states. Expression analysis of signaling ligand–receptor partners identified microglia–B cell interactions at acute and chronic stages that may be involved in B cell recruitment, retention, and formation of ectopic lymphoid follicles.
Conclusions
Immune cell responses to SCI have region-specific aspects and evolve with time. Developmentally diverse populations of B cells accumulate in the spinal cord following injury. Microglia at subacute stages express B cell recruitment factors, while chronically, they express factors predicted to reduce B cell inflammatory state. In the injured spinal cord, B cells create ectopic lymphoid structures, and express secreted factors potentially acting on microglia. Our study predicts previously unidentified crosstalk between microglia and B cells post-injury at acute and chronic stages, revealing new potential targets of inflammatory responses for SCI repair warranting future functional analyses.
Collapse
|
208
|
Smith AN, Shaughness M, Collier S, Hopkins D, Byrnes KR. Therapeutic targeting of microglia mediated oxidative stress after neurotrauma. Front Med (Lausanne) 2022; 9:1034692. [PMID: 36405593 PMCID: PMC9671221 DOI: 10.3389/fmed.2022.1034692] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/12/2022] [Indexed: 10/06/2023] Open
Abstract
Inflammation is a primary component of the central nervous system injury response. Traumatic brain and spinal cord injury are characterized by a pronounced microglial response to damage, including alterations in microglial morphology and increased production of reactive oxygen species (ROS). The acute activity of microglia may be beneficial to recovery, but continued inflammation and ROS production is deleterious to the health and function of other cells. Microglial nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX), mitochondria, and changes in iron levels are three of the most common sources of ROS. All three play a significant role in post-traumatic brain and spinal cord injury ROS production and the resultant oxidative stress. This review will evaluate the current state of therapeutics used to target these avenues of microglia-mediated oxidative stress after injury and suggest avenues for future research.
Collapse
Affiliation(s)
- Austin N. Smith
- Neuroscience Program, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Michael Shaughness
- Neuroscience Program, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Sean Collier
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Deanna Hopkins
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Kimberly R. Byrnes
- Neuroscience Program, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
209
|
Hong Z, Cheng J, Ye Y, Chen X, Zhang F. MicroRNA-451 Attenuates the Inflammatory Response of Activated Microglia by Downregulating Nucleotide Binding Oligomerization Domain-Like Receptor Protein 3. World Neurosurg 2022; 167:e1128-e1137. [PMID: 36087911 DOI: 10.1016/j.wneu.2022.08.139] [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: 04/18/2022] [Revised: 08/28/2022] [Accepted: 08/30/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND Spinal cord injury is the most common problem encountered during spinal surgery. After the initial trauma, the disruption of the blood-brain barrier and subsequent microglia activation result in extensive inflammatory responses. Inflammasomes are large protein complexes that are essential during inflammation. One of the most studied inflammasome components, nucleotide binding oligomerization domain-like receptor protein 3 (NLRP; nucleotide binding oligomerization domain-, leucine-rich repeat-, and pyrin domain-containing 3), is widely expressed in the central nervous system. Previous research has shown that microRNA-451 (miR-451) might play a role in regulating inflammatory conditions. METHODS Using bioinformatics analysis, we found that NLRP3 is a direct target of miR-451. This in silico prediction was confirmed using dual-luciferase reporter gene assays. To further demonstrate that miR-451 influenced microglial NLRP3 production, we activated microglial cells with lipopolysaccharides. RESULTS Activating microglial cells with lipopolysaccharides resulted in the production of NLRP3 inflammasomes and the secretion of the proinflammatory cytokines interleukin-1β and interleukin-18. We were able to demonstrate that overexpression of miR-451 suppressed this NLRP3-induced proinflammatory cascade of events. CONCLUSIONS Our findings have highlighted the potential anti-inflammatory role of miR-451 in reducing the secondary neuronal damage after spinal cord injury.
Collapse
Affiliation(s)
- Zhou Hong
- Medical School of Nantong University, Nantong, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Jiaqi Cheng
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China; Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong, China
| | - Yong Ye
- Medical School of Nantong University, Nantong, China
| | - Xiaoqing Chen
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong, China
| | - Feng Zhang
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong, China.
| |
Collapse
|
210
|
Sun J, Song Y, Chen Z, Qiu J, Zhu S, Wu L, Xing L. Heterogeneity and Molecular Markers for CNS Glial Cells Revealed by Single-Cell Transcriptomics. Cell Mol Neurobiol 2022; 42:2629-2642. [PMID: 34704168 PMCID: PMC11421601 DOI: 10.1007/s10571-021-01159-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 10/17/2021] [Indexed: 12/11/2022]
Abstract
Glial cells, including astrocytes, oligodendrocytes, and microglia, are the major components in the central nervous system (CNS). Studies have revealed the heterogeneity of each glial cell type and that they each may play distinct roles in physiological processes and/or neurological diseases. Single-cell sequencing (scRNA-seq) technology developed in recent years has extended our understanding of glial cell heterogeneity from the perspective of transcriptome profiling. This review summarizes the marker genes of major glial cells in the CNS and reveals their heterogeneity in different species, CNS regions, developmental stages, and pathological states (Alzheimer's disease and spinal cord injury), expanding our knowledge of glial cell heterogeneity on both molecular and functional levels.
Collapse
Affiliation(s)
- Junjie Sun
- 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, 19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Yixing Song
- 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, 19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Zhiheng Chen
- 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, 19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Jiaying Qiu
- Department of Prenatal Screening and Diagnosis Center, Nantong Maternal and Child Health Hospital affiliated to Nantong University, Nantong, 226001, Jiangsu, China
| | - Shunxing Zhu
- Laboratory Animal Center, Nantong University, Nantong, 226001, China
| | - Liucheng Wu
- Laboratory Animal Center, Nantong University, Nantong, 226001, China.
| | - Lingyan Xing
- 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, 19 Qixiu Road, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
211
|
West PK, Viengkhou B, Campbell IL, Hofer MJ. Microglia shield the murine brain from damage mediated by the cytokines IL-6 and IFN-α. Front Immunol 2022; 13:1036799. [PMID: 36389783 PMCID: PMC9650248 DOI: 10.3389/fimmu.2022.1036799] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 10/13/2022] [Indexed: 12/10/2023] Open
Abstract
Sustained production of elevated levels of the cytokines interleukin (IL)-6 or interferon (IFN)-α in the central nervous system (CNS) is detrimental and directly contributes to the pathogenesis of neurological diseases such as neuromyelitis optica spectrum disorders or cerebral interferonopathies, respectively. Using transgenic mice with CNS-targeted production of IL-6 (GFAP-IL6) or IFN-α (GFAP-IFN), we have recently demonstrated that microglia are prominent target and effector cells and mount stimulus-specific responses to these cytokines. In order to further clarify the phenotype and function of these cells, we treated GFAP-IL6 and GFAP-IFN mice with the CSF1R inhibitor PLX5622 to deplete microglia. We examined their ability to recover from acute microglia depletion, as well as the impact of chronic microglia depletion on the progression of disease. Following acute depletion in the brains of GFAP-IL6 mice, microglia repopulation was enhanced, while in GFAP-IFN mice, microglia did not repopulate the brain. Furthermore, chronic CSF1R inhibition was detrimental to the brain of GFAP-IL6 and GFAP-IFN mice and gave rise to severe CNS calcification which strongly correlated with the absence of microglia. In addition, PLX5622-treated GFAP-IFN mice had markedly reduced survival. Our findings provide evidence for novel microglia functions to protect against IFN-α-mediated neurotoxicity and neuronal dysregulation, as well as restrain calcification as a result of both IL-6- and IFN-α-induced neuroinflammation. Taken together, we demonstrate that CSF1R inhibition may be an undesirable target for therapeutic treatment of neuroinflammatory diseases that are driven by elevated IL-6 and IFN-α production.
Collapse
Affiliation(s)
| | | | | | - Markus J. Hofer
- School of Life and Environmental Sciences, Charles Perkins Centre and the Sydney Institute for Infectious Diseases, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
212
|
Liu S, Lin G, Yang Q, Wang P, Ma C, Qian X, He X, Dong Z, Liu Y, Liu M, Wu R, Yang L. Depletion of SASH1, an astrocyte differentiation-related gene, contributes to functional recovery in spinal cord injury. CNS Neurosci Ther 2022; 29:228-238. [PMID: 36286186 PMCID: PMC9804067 DOI: 10.1111/cns.13998] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 08/26/2022] [Accepted: 09/30/2022] [Indexed: 02/06/2023] Open
Abstract
AIMS This study aimed to evaluate the effects of the depletion of SAM and SH3 domain-containing protein 1 (SASH1) on functional recovery after spinal cord injury (SCI) and to investigate the possible mechanism of SASH1 knockdown in astrocytes facilitating axonal growth. METHODS SCI model was established in adult rats. SASH1 small interfering RNA (siSASH1) was used to investigate its function. Hindlimb motor function was evaluated by the Basso-Bresnahan-Beattie (BBB) assay. The gene expressions were evaluated by the methods of qRT-PCR, Western-blotting, ELISA, and immunohistochemistry. RESULTS SASH1 knockdown improved the BBB scores after SCI and significantly reduced GFAP expression. In cultured spinal astrocytes, siSASH1 treatment decreased interferon-γ release and increased brain-derived neurotrophic factor (BDNF) release. When cocultured with SASH1-knockdown astrocytes, axonal growth increased. The neuronal tropomyosin receptor kinase B (BDNF receptor) expression increased, especially in the axonal tips. SASH1 expression increased while NSCs differentiated into glial cells, instead of neurons. After SASH1 depletion, differentiated NSCs maintained a higher level of Nestin protein and an increase in BDNF release. CONCLUSIONS These results indicate that SASH1 acts as an astrocytic differentiation-maintaining protein, and SASH1 downregulation limits glial activation and contributes toward functional recovery after SCI.
Collapse
Affiliation(s)
- Siyi Liu
- Department of NeurosurgeryAffiliated Hospital of Nantong UniversityNantongChina
| | - Ge Lin
- 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 ProductsNantong UniversityNantongChina
| | - Qiao Yang
- Department of NeurosurgeryAffiliated Hospital of Nantong UniversityNantongChina
| | - Penghui Wang
- Department of NeurosurgeryAffiliated Hospital of Nantong UniversityNantongChina
| | - Chao Ma
- 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 ProductsNantong UniversityNantongChina
| | - Xiaowei Qian
- 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 ProductsNantong UniversityNantongChina
| | - Xiaomei He
- 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 ProductsNantong UniversityNantongChina
| | - Zhangji Dong
- 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 ProductsNantong UniversityNantongChina
| | - Yan 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 ProductsNantong UniversityNantongChina
| | - Mei 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 ProductsNantong UniversityNantongChina
| | - Ronghua Wu
- 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 ProductsNantong UniversityNantongChina
| | - Liu Yang
- Department of NeurosurgeryAffiliated Hospital of Nantong UniversityNantongChina
| |
Collapse
|
213
|
Zhou ZL, Xie H, Tian XB, Xu HL, Li W, Yao S, Zhang H. Microglial depletion impairs glial scar formation and aggravates inflammation partly by inhibiting STAT3 phosphorylation in astrocytes after spinal cord injury. Neural Regen Res 2022; 18:1325-1331. [PMID: 36453419 PMCID: PMC9838173 DOI: 10.4103/1673-5374.357912] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Astrocytes and microglia play an orchestrated role following spinal cord injury; however, the molecular mechanisms through which microglia regulate astrocytes after spinal cord injury are not yet fully understood. Herein, microglia were pharmacologically depleted and the effects on the astrocytic response were examined. We further explored the potential mechanisms involving the signal transducers and activators of transcription 3 (STAT3) pathway. For in vivo experiments, we constructed a contusion spinal cord injury model in C57BL/6 mice. To deplete microglia, all mice were treated with colony-stimulating factor 1 receptor inhibitor PLX3397, starting 2 weeks prior to surgery until they were sacrificed. Cell proliferation was examined by 5-ethynyl-2-deoxyuridine (EdU) and three pivotal inflammatory cytokines were detected by a specific Bio-Plex ProTM Reagent Kit. Locomotor function, neuroinflammation, astrocyte activation and phosphorylated STAT3 (pSTAT3, a maker of activation of STAT3 signaling) levels were determined. For in vitro experiments, a microglia and astrocyte coculture system was established, and the small molecule STA21, which blocks STAT3 activation, was applied to investigate whether STAT3 signaling is involved in mediating astrocyte proliferation induced by microglia. PLX3397 administration disrupted glial scar formation, increased inflammatory spillover, induced diffuse tissue damage and impaired functional recovery after spinal cord injury. Microglial depletion markedly reduced EdU+ proliferating cells, especially proliferating astrocytes at 7 days after spinal cord injury. RNA sequencing analysis showed that the JAK/STAT3 pathway was downregulated in mice treated with PLX3397. Double immunofluorescence staining confirmed that PLX3397 significantly decreased STAT3 expression in astrocytes. Importantly, in vitro coculture of astrocytes and microglia showed that microglia-induced astrocyte proliferation was abolished by STA21 administration. These findings suggest that microglial depletion impaired astrocyte proliferation and astrocytic scar formation, and induced inflammatory diffusion partly by inhibiting STAT3 phosphorylation in astrocytes following spinal cord injury.
Collapse
Affiliation(s)
- Zhi-Lai Zhou
- The Spine Surgery Department, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong Province, China,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Huan Xie
- The Spine Surgery Department, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong Province, China,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Xiao-Bo Tian
- The Spine Surgery Department, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong Province, China
| | - Hua-Li Xu
- Department of Anesthesiology, ZhuJiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Wei Li
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Shun Yao
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Hui Zhang
- The Spine Surgery Department, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong Province, China,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, China,Correspondence to: Hui Zhang, .
| |
Collapse
|
214
|
Miyata S. Glial functions in the blood-brain communication at the circumventricular organs. Front Neurosci 2022; 16:991779. [PMID: 36278020 PMCID: PMC9583022 DOI: 10.3389/fnins.2022.991779] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
The circumventricular organs (CVOs) are located around the brain ventricles, lack a blood-brain barrier (BBB) and sense blood-derived molecules. This review discusses recent advances in the importance of CVO functions, especially glial cells transferring periphery inflammation signals to the brain. The CVOs show size-limited vascular permeability, allowing the passage of molecules with molecular weight <10,000. This indicates that the lack of an endothelial cell barrier does not mean the free movement of blood-derived molecules into the CVO parenchyma. Astrocytes and tanycytes constitute a dense barrier at the distal CVO subdivision, preventing the free diffusion of blood-derived molecules into neighboring brain regions. Tanycytes in the CVOs mediate communication between cerebrospinal fluid and brain parenchyma via transcytosis. Microglia and macrophages of the CVOs are essential for transmitting peripheral information to other brain regions via toll-like receptor 2 (TLR2). Inhibition of TLR2 signaling or depletion of microglia and macrophages in the brain eliminates TLR2-dependent inflammatory responses. In contrast to TLR2, astrocytes and tanycytes in the CVOs of the brain are crucial for initiating lipopolysaccharide (LPS)-induced inflammatory responses via TLR4. Depletion of microglia and macrophages augments LPS-induced fever and chronic sickness responses. Microglia and macrophages in the CVOs are continuously activated, even under normal physiological conditions, as they exhibit activated morphology and express the M1/M2 marker proteins. Moreover, the microglial proliferation occurs in various regions, such as the hypothalamus, medulla oblongata, and telencephalon, with a marked increase in the CVOs, due to low-dose LPS administration, and after high-dose LPS administration, proliferation is seen in most brain regions, except for the cerebral cortex and hippocampus. A transient increase in the microglial population is beneficial during LPS-induced inflammation for attenuating sickness response. Transient receptor potential receptor vanilloid 1 expressed in astrocytes and tanycytes of the CVOs is responsible for thermoregulation upon exposure to a warm environment less than 37°C. Alternatively, Nax expressed in astrocytes and tanycytes of the CVOs is crucial for maintaining body fluid homeostasis. Thus, recent findings indicate that glial cells in the brain CVOs are essential for initiating neuroinflammatory responses and maintaining body fluid and thermal homeostasis.
Collapse
|
215
|
Bañuelos MM, Zavaleta YJA, Roldan A, Reyes RJ, Guardado M, Chavez Rojas B, Nyein T, Rodriguez Vega A, Santos M, Huerta-Sanchez E, Rohlfs RV. Associations between forensic loci and expression levels of neighboring genes may compromise medical privacy. Proc Natl Acad Sci U S A 2022; 119:e2121024119. [PMID: 36166477 PMCID: PMC9546536 DOI: 10.1073/pnas.2121024119] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 08/29/2022] [Indexed: 11/18/2022] Open
Abstract
A set of 20 short tandem repeats (STRs) is used by the US criminal justice system to identify suspects and to maintain a database of genetic profiles for individuals who have been previously convicted or arrested. Some of these STRs were identified in the 1990s, with a preference for markers in putative gene deserts to avoid forensic profiles revealing protected medical information. We revisit that assumption, investigating whether forensic genetic profiles reveal information about gene-expression variation or potential medical information. We find six significant correlations (false discovery rate = 0.23) between the forensic STRs and the expression levels of neighboring genes in lymphoblastoid cell lines. We explore possible mechanisms for these associations, showing evidence compatible with forensic STRs causing expression variation or being in linkage disequilibrium with a causal locus in three cases and weaker or potentially spurious associations in the other three cases. Together, these results suggest that forensic genetic loci may reveal expression levels and, perhaps, medical information.
Collapse
Affiliation(s)
- Mayra M. Bañuelos
- Department of Mathematics, San Francisco State University, San Francisco, CA 94132
- Ecology, Evolution and Organismal Biology, Brown University, Providence, RI 02912
- Center for Computational and Molecular Biology, Brown University, Providence, RI 02912
| | | | - Alennie Roldan
- Department of Biology, San Francisco State University, San Francisco, CA 94132
| | - Rochelle-Jan Reyes
- Department of Biology, San Francisco State University, San Francisco, CA 94132
| | - Miguel Guardado
- Department of Mathematics, San Francisco State University, San Francisco, CA 94132
| | | | - Thet Nyein
- Department of Mathematics, San Francisco State University, San Francisco, CA 94132
| | - Ana Rodriguez Vega
- Department of Biology, San Francisco State University, San Francisco, CA 94132
| | - Maribel Santos
- Department of Biology, San Francisco State University, San Francisco, CA 94132
| | - Emilia Huerta-Sanchez
- Ecology, Evolution and Organismal Biology, Brown University, Providence, RI 02912
- Center for Computational and Molecular Biology, Brown University, Providence, RI 02912
| | - Rori V. Rohlfs
- Department of Biology, San Francisco State University, San Francisco, CA 94132
| |
Collapse
|
216
|
The alarmin interleukin-1α triggers secondary degeneration through reactive astrocytes and endothelium after spinal cord injury. Nat Commun 2022; 13:5786. [PMID: 36184639 PMCID: PMC9527244 DOI: 10.1038/s41467-022-33463-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 09/16/2022] [Indexed: 01/18/2023] Open
Abstract
Spinal cord injury (SCI) triggers neuroinflammation, and subsequently secondary degeneration and oligodendrocyte (OL) death. We report that the alarmin interleukin (IL)-1α is produced by damaged microglia after SCI. Intra-cisterna magna injection of IL-1α in mice rapidly induces neutrophil infiltration and OL death throughout the spinal cord, mimicking the injury cascade seen in SCI sites. These effects are abolished through co-treatment with the IL-1R1 antagonist anakinra, as well as in IL-1R1-knockout mice which demonstrate enhanced locomotor recovery after SCI. Conditional restoration of IL-1R1 expression in astrocytes or endothelial cells (ECs), but not in OLs or microglia, restores IL-1α-induced effects, while astrocyte- or EC-specific Il1r1 deletion reduces OL loss. Conditioned medium derived from IL-1α-stimulated astrocytes results in toxicity for OLs; further, IL-1α-stimulated astrocytes generate reactive oxygen species (ROS), and blocking ROS production in IL-1α-treated or SCI mice prevented OL loss. Thus, after SCI, microglia release IL-1α, inducing astrocyte- and EC-mediated OL degeneration.
Collapse
|
217
|
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: 25] [Impact Index Per Article: 8.3] [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
|
218
|
Anti-inflammatory effect of glucagon-like Peptide-1 receptor agonist on the neurosensory retina in an acute optic nerve injury rat model. Eur J Pharmacol 2022; 933:175269. [PMID: 36103932 DOI: 10.1016/j.ejphar.2022.175269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 09/06/2022] [Accepted: 09/07/2022] [Indexed: 11/23/2022]
Abstract
PURPOSE To explore the possibility of using glucagon-like peptide-1 receptor agonist (GLP-1RA) as a new treatment for neuroinflammation, by analyzing retinal pathological changes in an optic nerve crush rat model. METHODS Eight-week-old male Sprague-Dawley rats were divided into lixisenatide (LIX, n = 10), traumatic control (T-CON, n = 10), and normal control (n = 5) groups. The optic nerves of left eyes in the LIX and T-CON groups were crushed in a standardized manner. The LIX group was treated with subcutaneous injections of lixisenatide (200 μg/kg/day) for 5 days. One week after initiating treatment, quantitative polymerase chain reaction, Western blot, and immunohistochemistry analyses were performed on the retinal tissues of each group to identify inflammatory markers. RESULTS The LIX group showed significantly lower mRNA levels of interleukin 1 beta (IL-1β), tumor necrosis factor-alpha (TNF-α), thioredoxin interacting protein (TXNIP), and glial fibrillary acidic protein (GFAP) than the T-CON group. Also, the LIX group exhibited decreased TXNIP and GFAP expression compared with the T-CON group, and similar expression to the normal control group, according to Western blot analysis. Significantly increased immunohistochemistry staining of Brn3a and decreased TUNEL staining were seen in the LIX group compared with the T-CON group, indicating that lixisenatide contributes to retinal ganglion cell survival in cases of acute optic nerve injury. CONCLUSIONS Neuroinflammation was significantly reduced in lixisenatide-treated retinas compared with untreated retinas in our acute optic nerve injury rat model. The neuroprotective effect of lixisenatide indicates that it can serve a new treatment option against clinically intractable traumatic optic neuropathy.
Collapse
|
219
|
Madalena KM, Brennan FH, Popovich PG. Genetic deletion of the glucocorticoid receptor in Cx 3cr1 + myeloid cells is neuroprotective and improves motor recovery after spinal cord injury. Exp Neurol 2022; 355:114114. [PMID: 35568187 PMCID: PMC10034962 DOI: 10.1016/j.expneurol.2022.114114] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/29/2022] [Accepted: 05/08/2022] [Indexed: 11/23/2022]
Abstract
Glucocorticoid receptors (GRs), part of the nuclear receptor superfamily of transcription factors (TFs), are ubiquitously expressed in all cell types and regulate cellular responses to glucocorticoids (e.g., cortisol in humans; corticosterone in rodents). In myeloid cells, glucocorticoids binding to GRs can enhance or repress gene transcription, thereby imparting distinct and context-dependent functions in macrophages at sites of inflammation. In experimental models and in humans, glucocorticoids are widely used as anti-inflammatory treatments to promote recovery of function after SCI. Thus, we predicted that deleting GR in mouse myeloid lineage cells (i.e., microglia and monocyte-derived macrophages) would enhance inflammation at the site of injury and worsen functional recovery after traumatic spinal cord injury (SCI). Contrary to our prediction, the intraspinal macrophage response to a moderate (75 kdyne) spinal contusion SCI was reduced in Cx3cr1-Cre;GRf/f conditional knockout mice (with GR specifically deleted in myeloid cells). This phenotype was associated with improvements in hindlimb motor recovery, myelin sparing, axon sparing/regeneration, and microvascular protection/plasticity relative to SCI mice with normal myeloid cell GR expression. Further analysis revealed that macrophage GR deletion impaired lipid and myelin phagocytosis and foamy macrophage formation. Together, these data reveal endogenous GR signaling as a key pathway that normally inhibits mechanisms of macrophage-mediated repair after SCI.
Collapse
Affiliation(s)
- Kathryn M Madalena
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; Belford Center for Spinal Cord Injury, Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Faith H Brennan
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; Belford Center for Spinal Cord Injury, Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Phillip G Popovich
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; Belford Center for Spinal Cord Injury, Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| |
Collapse
|
220
|
Tabarestani TQ, Lewis NE, Kelly-Hedrick M, Zhang N, Cellini BR, Marrotte EJ, Williamson T, Wang H, Laskowitz DT, Faw TD, Abd-El-Barr MM. Surgical Considerations to Improve Recovery in Acute Spinal Cord Injury. Neurospine 2022; 19:689-702. [PMID: 36203295 PMCID: PMC9537855 DOI: 10.14245/ns.2244616.308] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 09/07/2022] [Indexed: 12/14/2022] Open
Abstract
Acute traumatic spinal cord injury (SCI) can be a devastating and costly event for individuals, their families, and the health system as a whole. Prognosis is heavily dependent on the physical extent of the injury and the severity of neurological dysfunction. If not treated urgently, individuals can suffer exacerbated secondary injury cascades that may increase tissue injury and limit recovery. Initial recognition and rapid treatment of acute SCI are vital to limiting secondary injury, reducing morbidity, and providing the best chance of functional recovery. This article aims to review the pathophysiology of SCI and the most up-to-date management of the acute traumatic SCI, specifically examining the modern approaches to surgical treatments along with the ethical limitations of research in this field.
Collapse
Affiliation(s)
| | - Nicholle E. Lewis
- Doctor of Physical Therapy Division, Department of Orthopaedic Surgery, Duke University, Durham, NC, USA
| | | | - Nina Zhang
- Department of Psychology and Neuroscience, Trinity College of Arts and Sciences, Duke University, Durham, NC, USA
| | - Brianna R. Cellini
- Department of Psychology and Neuroscience, Trinity College of Arts and Sciences, Duke University, Durham, NC, USA
| | - Eric J. Marrotte
- Department of Neurology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Theresa Williamson
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA,Center for Bioethics, Harvard Medical School, Boston, MA, USA
| | - Haichen Wang
- Department of Neurology, Duke University, Durham, NC, USA
| | | | - Timothy D. Faw
- Doctor of Physical Therapy Division, Department of Orthopaedic Surgery, Duke University, Durham, NC, USA,Duke Institute for Brain Sciences, Duke University, Durham, NC, USA
| | - Muhammad M. Abd-El-Barr
- Department of Neurosurgery, Duke University, Durham, NC, USA,Corresponding Author Muhammad M. Abd-El-Barr Department of Neurosurgery, Duke University Medical Center 2840, Room 5335 5th Floor, Orange Zone, Duke South, Durham, NC 27710, USA
| |
Collapse
|
221
|
Spatiotemporal dynamics of the cellular components involved in glial scar formation following spinal cord injury. Biomed Pharmacother 2022; 153:113500. [DOI: 10.1016/j.biopha.2022.113500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 06/19/2022] [Accepted: 07/30/2022] [Indexed: 11/30/2022] Open
|
222
|
Freyermuth-Trujillo X, Segura-Uribe JJ, Salgado-Ceballos H, Orozco-Barrios CE, Coyoy-Salgado A. Inflammation: A Target for Treatment in Spinal Cord Injury. Cells 2022; 11:2692. [PMID: 36078099 PMCID: PMC9454769 DOI: 10.3390/cells11172692] [Citation(s) in RCA: 104] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/16/2022] [Accepted: 08/25/2022] [Indexed: 12/15/2022] Open
Abstract
Spinal cord injury (SCI) is a significant cause of disability, and treatment alternatives that generate beneficial outcomes and have no side effects are urgently needed. SCI may be treatable if intervention is initiated promptly. Therefore, several treatment proposals are currently being evaluated. Inflammation is part of a complex physiological response to injury or harmful stimuli induced by mechanical, chemical, or immunological agents. Neuroinflammation is one of the principal secondary changes following SCI and plays a crucial role in modulating the pathological progression of acute and chronic SCI. This review describes the main inflammatory events occurring after SCI and discusses recently proposed potential treatments and therapeutic agents that regulate inflammation after insult in animal models.
Collapse
Affiliation(s)
- Ximena Freyermuth-Trujillo
- Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades Dr. Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City CP 06720, Mexico
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Mexico City CP 04510, Mexico
| | - Julia J. Segura-Uribe
- Subdirección de Gestión de la Investigación, Hospital Infantil de México Federico Gómez, Secretaría de Salud, Mexico City CP 06720, Mexico
| | - Hermelinda Salgado-Ceballos
- Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades Dr. Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City CP 06720, Mexico
| | - Carlos E. Orozco-Barrios
- CONACyT-Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades Dr. Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City CP 06720, Mexico
| | - Angélica Coyoy-Salgado
- CONACyT-Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades Dr. Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City CP 06720, Mexico
| |
Collapse
|
223
|
Bijelić D, Adžić M, Perić M, Reiss G, Milošević M, Andjus PR, Jakovčevski I. Tenascin-C fibronectin D domain is involved in the fine-tuning of glial response to CNS injury in vitro. Front Cell Dev Biol 2022; 10:952208. [PMID: 36092707 PMCID: PMC9462431 DOI: 10.3389/fcell.2022.952208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/18/2022] [Indexed: 11/19/2022] Open
Abstract
Understanding processes that occur after injuries to the central nervous system is essential in order to gain insight into how the restoration of function can be improved. Extracellular glycoprotein tenascin-C (TnC) has numerous functions in wound healing process depending on the expression time, location, isoform and binding partners which makes it interesting to study in this context. We used an in vitro injury model, the mixed culture of cortical astrocytes and microglia, and observed that without TnC microglial cells tend to populate gap area in greater numbers and proliferate more, whereas astrocytes build up in the border region to promote faster gap closure. Alternatively spliced domain of TnC, fibronectin type III-like repeat D (FnD) strongly affected physiological properties and morphology of both astrocytes and microglia in this injury model. The rate of microglial proliferation in the injury region decreased significantly with the addition of FnD. Additionally, density of microglia also decreased, in part due to reduced proliferation, and possibly due to reduced migration and increased contact inhibition between enlarged FnD-treated cells. Overall morphology of FnD-treated microglia resembled the activated pro-inflammatory cells, and elevated expression of iNOS was in accordance with this phenotype. The effect of FnD on astrocytes was different, as it did not affect their proliferation, but stimulated migration of reactivated astrocytes into the scratched area 48 h after the lesion. Elevated expression and secretion of TNF-α and IL-1β upon FnD treatment indicated the onset of inflammation. Furthermore, on Western blots we observed increased intensity of precursor bands of β1 integrin and appearance of monomeric bands of P2Y12R after FnD treatment which substantiates and clarifies its role in cellular shape and motility changes. Our results show versatile functions of TnC and in particular FnD after injury, mostly contributing to ongoing inflammation in the injury region. Based on our findings, FnD might be instrumental in limiting immune cell infiltration, and promoting astrocyte migration within the injury region, thus influencing spaciotemporal organization of the wound and surrounding area.
Collapse
Affiliation(s)
- Dunja Bijelić
- Centre for Laser Microscopy, Institute of Physiology and Biochemistry “Ivan Djaja”, Faculty of Biology, University of Belgrade, Belgrade, Serbia
- *Correspondence: Dunja Bijelić, ; Igor Jakovčevski,
| | - Marija Adžić
- Centre for Laser Microscopy, Institute of Physiology and Biochemistry “Ivan Djaja”, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Mina Perić
- Centre for Laser Microscopy, Institute of Physiology and Biochemistry “Ivan Djaja”, Faculty of Biology, University of Belgrade, Belgrade, Serbia
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Gebhard Reiss
- Institute for Anatomy and Clinical Morphology, University Witten / Herdecke, Witten, Germany
| | - Milena Milošević
- Centre for Laser Microscopy, Institute of Physiology and Biochemistry “Ivan Djaja”, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Pavle R. Andjus
- Centre for Laser Microscopy, Institute of Physiology and Biochemistry “Ivan Djaja”, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Igor Jakovčevski
- Institute for Anatomy and Clinical Morphology, University Witten / Herdecke, Witten, Germany
| |
Collapse
|
224
|
Nakajima N, Ohnishi Y, Yamamoto M, Setoyama D, Imai H, Takenaka T, Matsumoto M, Hosomi K, Saitoh Y, Furue H, Kishima H. Excess intracellular ATP causes neuropathic pain following spinal cord injury. Cell Mol Life Sci 2022; 79:483. [PMID: 35972649 PMCID: PMC11072579 DOI: 10.1007/s00018-022-04510-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/16/2022] [Accepted: 08/01/2022] [Indexed: 11/03/2022]
Abstract
Intractable neuropathic pain following spinal cord injury (NP-SCI) reduces a patient's quality of life. Excessive release of ATP into the extracellular space evokes neuroinflammation via purinergic receptor. Neuroinflammation plays an important role in the initiation and maintenance of NP. However, little is known about whether or not extracellular ATP cause NP-SCI. We found in the present study that excess of intracellular ATP at the lesion site evokes at-level NP-SCI. No significant differences in the body weight, locomotor function, or motor behaviors were found in groups that were negative and positive for at-level allodynia. The intracellular ATP level at the lesion site was significantly higher in the allodynia-positive mice than in the allodynia-negative mice. A metabolome analysis revealed that there were no significant differences in the ATP production or degradation between allodynia-negative and allodynia-positive mice. Dorsal horn neurons in allodynia mice were found to be inactivated in the resting state, suggesting that decreased ATP consumption due to neural inactivity leads to a build-up of intracellular ATP. In contrast to the findings in the resting state, mechanical stimulation increased the neural activity of dorsal horn and extracellular ATP release at lesion site. The forced production of intracellular ATP at the lesion site in non-allodynia mice induced allodynia. The inhibition of P2X4 receptors in allodynia mice reduced allodynia. These results suggest that an excess buildup of intracellular ATP in the resting state causes at-level NP-SCI as a result of the extracellular release of ATP with mechanical stimulation.
Collapse
Affiliation(s)
- Nobuhiko Nakajima
- Department of Neurosurgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yuichiro Ohnishi
- Department of Research Promotion and Management, National Cerebral and Cardiovascular Center, 6-1 Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan.
- Department of Neurosurgery, Osaka Gyoumeikan Hospital, Osaka, Japan.
| | - Masamichi Yamamoto
- Department of Research Promotion and Management, National Cerebral and Cardiovascular Center, 6-1 Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan.
| | - Daiki Setoyama
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hirohiko Imai
- Department of Systems Science, Graduate School of Informatics, Kyoto University, Kyoto, Japan
| | - Tomofumi Takenaka
- Department of Neurosurgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Mari Matsumoto
- Department of Research Promotion and Management, National Cerebral and Cardiovascular Center, 6-1 Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan
| | - Koichi Hosomi
- Department of Neurosurgery, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Neuromodulation and Neurosurgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yoichi Saitoh
- Department of Neuromodulation and Neurosurgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hidemasa Furue
- Department of Neurophysiology, Hyogo College of Medicine, Hyogo, Japan
| | - Haruhiko Kishima
- Department of Neurosurgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
225
|
Fibrotic Scar in CNS Injuries: From the Cellular Origins of Fibroblasts to the Molecular Processes of Fibrotic Scar Formation. Cells 2022; 11:cells11152371. [PMID: 35954214 PMCID: PMC9367779 DOI: 10.3390/cells11152371] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/29/2022] [Accepted: 07/30/2022] [Indexed: 02/06/2023] Open
Abstract
Central nervous system (CNS) trauma activates a persistent repair response that leads to fibrotic scar formation within the lesion. This scarring is similar to other organ fibrosis in many ways; however, the unique features of the CNS differentiate it from other organs. In this review, we discuss fibrotic scar formation in CNS trauma, including the cellular origins of fibroblasts, the mechanism of fibrotic scar formation following an injury, as well as the implication of the fibrotic scar in CNS tissue remodeling and regeneration. While discussing the shared features of CNS fibrotic scar and fibrosis outside the CNS, we highlight their differences and discuss therapeutic targets that may enhance regeneration in the CNS.
Collapse
|
226
|
Corrigendum: Purinergic signaling systems across comparative models of spinal cord injury. Neural Regen Res 2022; 18:689-696. [PMID: 36018196 PMCID: PMC9727416 DOI: 10.4103/1673-5374.350234] [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] [Indexed: 12/15/2022] Open
Abstract
[This corrects the article DOI: 10.4103/1673-5374.338993].
Collapse
|
227
|
Cornelison C, Fadel S. Clickable Biomaterials for Modulating Neuroinflammation. Int J Mol Sci 2022; 23:8496. [PMID: 35955631 PMCID: PMC9369181 DOI: 10.3390/ijms23158496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/27/2022] [Accepted: 07/27/2022] [Indexed: 02/04/2023] Open
Abstract
Crosstalk between the nervous and immune systems in the context of trauma or disease can lead to a state of neuroinflammation or excessive recruitment and activation of peripheral and central immune cells. Neuroinflammation is an underlying and contributing factor to myriad neuropathologies including neurodegenerative diseases like Alzheimer's disease and Parkinson's disease; autoimmune diseases like multiple sclerosis; peripheral and central nervous system infections; and ischemic and traumatic neural injuries. Therapeutic modulation of immune cell function is an emerging strategy to quell neuroinflammation and promote tissue homeostasis and/or repair. One such branch of 'immunomodulation' leverages the versatility of biomaterials to regulate immune cell phenotypes through direct cell-material interactions or targeted release of therapeutic payloads. In this regard, a growing trend in biomaterial science is the functionalization of materials using chemistries that do not interfere with biological processes, so-called 'click' or bioorthogonal reactions. Bioorthogonal chemistries such as Michael-type additions, thiol-ene reactions, and Diels-Alder reactions are highly specific and can be used in the presence of live cells for material crosslinking, decoration, protein or cell targeting, and spatiotemporal modification. Hence, click-based biomaterials can be highly bioactive and instruct a variety of cellular functions, even within the context of neuroinflammation. This manuscript will review recent advances in the application of click-based biomaterials for treating neuroinflammation and promoting neural tissue repair.
Collapse
Affiliation(s)
- Chase Cornelison
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA 01003, USA;
| | | |
Collapse
|
228
|
Uppada S, Zou D, Scott EM, Ko G, Pflugfelder S, Kumar MNVR, Ganugula R. Paclitaxel and Urolithin A Prevent Histamine-Induced Neurovascular Breakdown Alike, in an Ex Vivo Rat Eye Model. ACS Chem Neurosci 2022; 13:2092-2098. [PMID: 35574761 PMCID: PMC9928511 DOI: 10.1021/acschemneuro.1c00692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Neurovascular eye problems are better prevented than managed or treated. Despite growing concern of occurrence in aging populations and development secondary to diseases such as diabetes and hypertension, we currently have very few options to tackle this global problem. Creating effective and high-throughput screening strategies is as important as the intervention itself. Here, we present for the first time a robust ex vivo rat eye model of histamine-induced vascular damage for investigating the therapeutic potential of paclitaxel (PTX) and urolithin A (UA) as alternatives to dexamethasone for preventing vascular damage in the retina. Extensive loss of vascularization and apoptosis were observed in the histamine-challenged group and successfully prevented in the intervention groups, more significantly in the PTX and UA. These important early results indicate that PTX and UA could be developed as potential preventive strategies for a wide variety of retinal diseases.
Collapse
Affiliation(s)
- Srijayaprakash Uppada
- The Center for Convergent Bioscience and Medicine (CCBM), The University of Alabama, Tuscaloosa, Alabama 35401, United States; College of Community Health Sciences, The University of Alabama, Tuscaloosa, Alabama 35401, United States
| | - Dianxiong Zou
- College of Community Health Sciences, The University of Alabama, Tuscaloosa, Alabama 35401, United States
| | - Erin M. Scott
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas 77843, United States
| | - Gladys Ko
- Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas 77843, United States
| | - Stephen Pflugfelder
- Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas 77030, United States
| | - M. N. V. Ravi Kumar
- The Center for Convergent Bioscience and Medicine (CCBM), The University of Alabama, Tuscaloosa, Alabama 35401, United States; College of Community Health Sciences and Alabama Life Research Institute, The University of Alabama, Tuscaloosa, Alabama 35401, United States; Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, College Station, Texas 77843, United States; Department of Biological Sciences, The University of Alabama, Tuscaloosa, Alabama 35401, United States; Chemical and Biological Engineering, University of Alabama, Tuscaloosa, Alabama 35401, United States
| | - Raghu Ganugula
- The Center for Convergent Bioscience and Medicine (CCBM), The University of Alabama, Tuscaloosa, Alabama 35401, United States; College of Community Health Sciences and Alabama Life Research Institute, The University of Alabama, Tuscaloosa, Alabama 35401, United States; Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, College Station, Texas 77843, United States; Department of Biological Sciences, The University of Alabama, Tuscaloosa, Alabama 35401, United States
| |
Collapse
|
229
|
Brennan FH, Li Y, Wang C, Ma A, Guo Q, Li Y, Pukos N, Campbell WA, Witcher KG, Guan Z, Kigerl KA, Hall JCE, Godbout JP, Fischer AJ, McTigue DM, He Z, Ma Q, Popovich PG. Microglia coordinate cellular interactions during spinal cord repair in mice. Nat Commun 2022; 13:4096. [PMID: 35835751 PMCID: PMC9283484 DOI: 10.1038/s41467-022-31797-0] [Citation(s) in RCA: 127] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 07/01/2022] [Indexed: 12/27/2022] Open
Abstract
Traumatic spinal cord injury (SCI) triggers a neuro-inflammatory response dominated by tissue-resident microglia and monocyte derived macrophages (MDMs). Since activated microglia and MDMs are morphologically identical and express similar phenotypic markers in vivo, identifying injury responses specifically coordinated by microglia has historically been challenging. Here, we pharmacologically depleted microglia and use anatomical, histopathological, tract tracing, bulk and single cell RNA sequencing to reveal the cellular and molecular responses to SCI controlled by microglia. We show that microglia are vital for SCI recovery and coordinate injury responses in CNS-resident glia and infiltrating leukocytes. Depleting microglia exacerbates tissue damage and worsens functional recovery. Conversely, restoring select microglia-dependent signaling axes, identified through sequencing data, in microglia depleted mice prevents secondary damage and promotes recovery. Additional bioinformatics analyses reveal that optimal repair after SCI might be achieved by co-opting key ligand-receptor interactions between microglia, astrocytes and MDMs.
Collapse
Affiliation(s)
- Faith H Brennan
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Belford Center for Spinal Cord Injury, Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Yang Li
- Department of Biomedical Informatics, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Cankun Wang
- Department of Biomedical Informatics, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Anjun Ma
- Department of Biomedical Informatics, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Qi Guo
- Department of Biomedical Informatics, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Yi Li
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Department of Neurology, Harvard Medical School, Boston, MA, USA
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Nicole Pukos
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Belford Center for Spinal Cord Injury, Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Warren A Campbell
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Kristina G Witcher
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Zhen Guan
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Belford Center for Spinal Cord Injury, Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Kristina A Kigerl
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Belford Center for Spinal Cord Injury, Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Jodie C E Hall
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Belford Center for Spinal Cord Injury, Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Jonathan P Godbout
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Andy J Fischer
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Dana M McTigue
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Belford Center for Spinal Cord Injury, Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Zhigang He
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Qin Ma
- Department of Biomedical Informatics, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Phillip G Popovich
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA.
- Belford Center for Spinal Cord Injury, Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA.
| |
Collapse
|
230
|
Shi W, Zhang J, Shang Z, Zhang Y, Xia Y, Fu H, Yu T. Restorative therapy using microglial depletion and repopulation for central nervous system injuries and diseases. Front Immunol 2022; 13:969127. [PMID: 35911768 PMCID: PMC9329909 DOI: 10.3389/fimmu.2022.969127] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 06/27/2022] [Indexed: 12/02/2022] Open
Abstract
Microglia are important resident immune cells in the central nervous system (CNS) and play an important role in its development, homeostasis, and disease treatments. Activated microglia perform diverse functions in mouse models of CNS neurodegenerative diseases or deficits. In humans, microglia have been linked to various neurodegenerative diseases. Following brain or spinal cord injury, microglia express pro- and anti-inflammatory phenotypes at different stages of recovery. With the development of pharmacological and genetic tools for microglial depletion, studies have demonstrated that microglial depletion exerts both positive and negative effects in the treatment of CNS diseases. Notably, microglial depletion provides an empty niche that stimulates production of new microglia. Microglial depletion and repopulation can not only treat diseases by eliminating dysfunctional microglia but can also provide an indication of the molecular mechanisms of diseases. Although this approach has shown impressive results, its use is still in its infancy. In this review, we summarize the current pharmacological and genetic tools for microglial depletion and highlight recent advances in microglial repopulation therapy for the treatment and functional recovery of neurological diseases and deficits. Finally, we briefly discuss the therapeutic challenges and prospective uses of microglial repopulation therapy.
Collapse
Affiliation(s)
- Weipeng Shi
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- Medical Department of Qingdao University, Qingdao, China
| | - Jing Zhang
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- Medical Department of Qingdao University, Qingdao, China
| | - Zhen Shang
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- Medical Department of Qingdao University, Qingdao, China
| | - Yingze Zhang
- Key Laboratory of Biomechanics of Hebei Province, Department of Trauma Emergency Center, The Third Hospital of Hebei Medical University, Orthopaedics Research Institution of Hebei Province, Shijiazhuang, China
| | - Yanzhi Xia
- State Key Laboratory of Bio-Fibers and Eco-Textiles, Collaborative Innovation Center for Marine Biomass Fibers, Materials and Textiles of Shandong Province, College of Materials Science and Engineering, Institute of Marine Biobased Materials, Qingdao University, Qingdao, China
| | - Haitao Fu
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- *Correspondence: Haitao Fu, ; Tengbo Yu,
| | - Tengbo Yu
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- *Correspondence: Haitao Fu, ; Tengbo Yu,
| |
Collapse
|
231
|
Pinelli F, Pizzetti F, Veneruso V, Petillo E, Raghunath M, Perale G, Veglianese P, Rossi F. Biomaterial-Mediated Factor Delivery for Spinal Cord Injury Treatment. Biomedicines 2022; 10:biomedicines10071673. [PMID: 35884981 PMCID: PMC9313204 DOI: 10.3390/biomedicines10071673] [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: 04/28/2022] [Revised: 05/24/2022] [Accepted: 07/05/2022] [Indexed: 11/19/2022] Open
Abstract
Spinal cord injury (SCI) is an injurious process that begins with immediate physical damage to the spinal cord and associated tissues during an acute traumatic event. However, the tissue damage expands in both intensity and volume in the subsequent subacute phase. At this stage, numerous events exacerbate the pathological condition, and therein lies the main cause of post-traumatic neural degeneration, which then ends with the chronic phase. In recent years, therapeutic interventions addressing different neurodegenerative mechanisms have been proposed, but have met with limited success when translated into clinical settings. The underlying reasons for this are that the pathogenesis of SCI is a continued multifactorial disease, and the treatment of only one factor is not sufficient to curb neural degeneration and resulting paralysis. Recent advances have led to the development of biomaterials aiming to promote in situ combinatorial strategies using drugs/biomolecules to achieve a maximized multitarget approach. This review provides an overview of single and combinatorial regenerative-factor-based treatments as well as potential delivery options to treat SCIs.
Collapse
Affiliation(s)
- Filippo Pinelli
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Via Mancinelli 7, 20131 Milan, Italy; (F.P.); (F.P.); (E.P.)
| | - Fabio Pizzetti
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Via Mancinelli 7, 20131 Milan, Italy; (F.P.); (F.P.); (E.P.)
| | - Valeria Veneruso
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy;
| | - Emilia Petillo
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Via Mancinelli 7, 20131 Milan, Italy; (F.P.); (F.P.); (E.P.)
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy;
| | - Michael Raghunath
- Center for Cell Biology and Tissue Engineering, Institute for Chemistry and Biotechnology (ICBT), Zurich University of Applied Sciences (ZHAW), 8820 Wädenswil, Switzerland;
| | - Giuseppe Perale
- Faculty of Biomedical Sciences, University of Southern Switzerland (USI), Via Buffi 13, 6900 Lugano, Switzerland;
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Donaueschingenstrasse 13, 1200 Vienna, Austria
| | - Pietro Veglianese
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy;
- Correspondence: (P.V.); (F.R.); Tel.: +39-02-3901-4205 (P.V.); +39-02-2399-3145 (F.R.)
| | - Filippo Rossi
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Via Mancinelli 7, 20131 Milan, Italy; (F.P.); (F.P.); (E.P.)
- Correspondence: (P.V.); (F.R.); Tel.: +39-02-3901-4205 (P.V.); +39-02-2399-3145 (F.R.)
| |
Collapse
|
232
|
Gong L, Gu Y, Han X, Luan C, Liu C, Wang X, Sun Y, Zheng M, Fang M, Yang S, Xu L, Sun H, Yu B, Gu X, Zhou S. Spatiotemporal Dynamics of the Molecular Expression Pattern and Intercellular Interactions in the Glial Scar Response to Spinal Cord Injury. Neurosci Bull 2022; 39:213-244. [PMID: 35788904 PMCID: PMC9905408 DOI: 10.1007/s12264-022-00897-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 04/28/2022] [Indexed: 12/22/2022] Open
Abstract
Nerve regeneration in adult mammalian spinal cord is poor because of the lack of intrinsic regeneration of neurons and extrinsic factors - the glial scar is triggered by injury and inhibits or promotes regeneration. Recent technological advances in spatial transcriptomics (ST) provide a unique opportunity to decipher most genes systematically throughout scar formation, which remains poorly understood. Here, we first constructed the tissue-wide gene expression patterns of mouse spinal cords over the course of scar formation using ST after spinal cord injury from 32 samples. Locally, we profiled gene expression gradients from the leading edge to the core of the scar areas to further understand the scar microenvironment, such as neurotransmitter disorders, activation of the pro-inflammatory response, neurotoxic saturated lipids, angiogenesis, obstructed axon extension, and extracellular structure re-organization. In addition, we described 21 cell transcriptional states during scar formation and delineated the origins, functional diversity, and possible trajectories of subpopulations of fibroblasts, glia, and immune cells. Specifically, we found some regulators in special cell types, such as Thbs1 and Col1a2 in macrophages, CD36 and Postn in fibroblasts, Plxnb2 and Nxpe3 in microglia, Clu in astrocytes, and CD74 in oligodendrocytes. Furthermore, salvianolic acid B, a blood-brain barrier permeation and CD36 inhibitor, was administered after surgery and found to remedy fibrosis. Subsequently, we described the extent of the scar boundary and profiled the bidirectional ligand-receptor interactions at the neighboring cluster boundary, contributing to maintain scar architecture during gliosis and fibrosis, and found that GPR37L1_PSAP, and GPR37_PSAP were the most significant gene-pairs among microglia, fibroblasts, and astrocytes. Last, we quantified the fraction of scar-resident cells and proposed four possible phases of scar formation: macrophage infiltration, proliferation and differentiation of scar-resident cells, scar emergence, and scar stationary. Together, these profiles delineated the spatial heterogeneity of the scar, confirmed the previous concepts about scar architecture, provided some new clues for scar formation, and served as a valuable resource for the treatment of central nervous system injury.
Collapse
Affiliation(s)
- Leilei Gong
- 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, 226001, China
| | - Yun Gu
- 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, 226001, China
| | - Xiaoxiao Han
- 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, 226001, China
| | - Chengcheng Luan
- 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, 226001, China
| | - Chang Liu
- 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, 226001, China
| | - Xinghui Wang
- 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, 226001, China
| | - Yufeng Sun
- 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, 226001, China
| | - Mengru Zheng
- 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, 226001, China
| | - Mengya Fang
- 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, 226001, China
| | - Shuhai Yang
- 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, 226001, China
| | - Lai Xu
- 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, 226001, China
| | - Hualin Sun
- 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, 226001, China
| | - Bin Yu
- 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, 226001, China
| | - Xiaosong Gu
- 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, 226001, China.
| | - Songlin Zhou
- 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, 226001, China.
| |
Collapse
|
233
|
Activin A is a novel chemoattractant for migration of microglial BV2 cells. J Neuroimmunol 2022; 371:577929. [DOI: 10.1016/j.jneuroim.2022.577929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/08/2022] [Accepted: 07/09/2022] [Indexed: 11/19/2022]
|
234
|
Chio JCT, Punjani N, Hejrati N, Zavvarian MM, Hong J, Fehlings MG. Extracellular Matrix and Oxidative Stress Following Traumatic Spinal Cord Injury: Physiological and Pathophysiological Roles and Opportunities for Therapeutic Intervention. Antioxid Redox Signal 2022; 37:184-207. [PMID: 34465134 DOI: 10.1089/ars.2021.0120] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Significance: Traumatic spinal cord injury (SCI) causes significant disruption to neuronal, glial, vascular, and extracellular elements. The spinal cord extracellular matrix (ECM) comprises structural and communication proteins that are involved in reparative and regenerative processes after SCI. In the healthy spinal cord, the ECM helps maintain spinal cord homeostasis. After SCI, the damaged ECM limits plasticity and contributes to inflammation through the expression of damage-associated molecules such as proteoglycans. Recent Advances: Considerable insights have been gained by characterizing the origins of the gliotic and fibrotic scars, which not only reduce the spread of injury but also limit neuroregeneration. These properties likely limit the success of therapies used to treat patients with SCI. The ECM, which is a major contributor to the scars and normal physiological functions of the spinal cord, represents an exciting therapeutic target to enhance recovery post-SCI. Critical Issue: Various ECM-based preclinical therapies have been developed. These include disrupting scar components, inhibiting activity of ECM metalloproteinases, and maintaining iron homeostasis. Biomaterials have also been explored. However, the majority of these treatments have not experienced successful clinical translation. This could be due to the ECM and scars' polarizing roles. Future Directions: This review surveys the complexity involved in spinal ECM modifications, discusses new ECM-based combinatorial strategies, and explores the biomaterials evaluated in clinical trials, which hope to introduce new treatments that enhance recovery after SCI. These topics will incorporate oxidative species, which are both beneficial and harmful in reparative and regenerative processes after SCI, and not often assessed in pertinent literature. Antioxid. Redox Signal. 37, 184-207.
Collapse
Affiliation(s)
- Jonathon Chon Teng Chio
- Department of Genetics and Development, Krembil Brain Institute, University Health Network, Toronto, Canada.,Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Nayaab Punjani
- Department of Genetics and Development, Krembil Brain Institute, University Health Network, Toronto, Canada.,Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Nader Hejrati
- Department of Genetics and Development, Krembil Brain Institute, University Health Network, Toronto, Canada
| | - Mohammad-Masoud Zavvarian
- Department of Genetics and Development, Krembil Brain Institute, University Health Network, Toronto, Canada.,Institute of Medical Science, University of Toronto, Toronto, Canada
| | - James Hong
- Department of Genetics and Development, Krembil Brain Institute, University Health Network, Toronto, Canada
| | - Michael G Fehlings
- Department of Genetics and Development, Krembil Brain Institute, University Health Network, Toronto, Canada.,Institute of Medical Science, University of Toronto, Toronto, Canada.,Department of Surgery and Spine Program, University of Toronto, Toronto, Canada
| |
Collapse
|
235
|
Hu X, Huang J, Li Y, Dong L, Chen Y, Ouyang F, Li J, Li Z, Jing J, Cheng L. TAZ Induces Migration of Microglia and Promotes Neurological Recovery After Spinal Cord Injury. Front Pharmacol 2022; 13:938416. [PMID: 35833021 PMCID: PMC9271831 DOI: 10.3389/fphar.2022.938416] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
Following spinal cord injury (SCI), microglia gradually migrate to the edge of the lesion, interweaving around the border of the lesion to form the microglial scar, which performs inflammatory limiting and neuroprotective functions. Recent reports showed that Yes-associated protein (YAP) was expressed in astrocytes and promoted the formation of astrocytic scars, while YAP was not expressed in microglia after SCI. YAP and its paralogue transcriptional coactivator with PDZ-binding motif (TAZ) are transcriptional coactivators, which have a similar functional role as both are negatively regulated by the Hippo signalling pathway. However, the expression and function of TAZ after SCI are unclear. Our research group previously found that Fascin-1 was highly expressed in microglia and promoted migration of microglia after SCI, and that, there was a close regulatory relationship between Fascin-1 and YAP/TAZ. In this study, we demonstrated that TAZ was significantly upregulated and mainly expressed in microglia after SCI, and accumulated in the nuclei of microglia in the spinal cord at 14 days post-SCI. Moreover, TAZ was upregulated and accumulated in the nuclei of anti-inflammatory M2-like (M2-L) polarized or myelin-treated microglia. Additionally, XMU-MP-1 (an inhibitor of the Hippo kinase MST1/2 to active TAZ) promoted the aggregation of microglia around the lesion core, resulting in the formation of microglial scars and the functional recovery of mice after SCI. Our findings also indicated that TAZ promoted microglial migration in vitro. Mechanistically, Fascin-1 interacted with TAZ, which upregulated TAZ expression and induced TAZ nuclear accumulation in microglia to promote microglial migration. These findings revealed that TAZ mediated microglial migration to the edge of the lesion core, promoting the formation of microglial scars and functional recovery after SCI. Moreover, TAZ was downstream of Fascin-1, which positively regulated microglial migration after SCI.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ziyu Li
- *Correspondence: Li Cheng, ; Juehua Jing, ; Ziyu Li,
| | - Juehua Jing
- *Correspondence: Li Cheng, ; Juehua Jing, ; Ziyu Li,
| | - Li Cheng
- *Correspondence: Li Cheng, ; Juehua Jing, ; Ziyu Li,
| |
Collapse
|
236
|
Xia L, Qi J, Tang M, Liu J, Zhang D, Zhu Y, Hu B. Continual Deletion of Spinal Microglia Reforms Astrocyte Scar Favoring Axonal Regeneration. Front Pharmacol 2022; 13:881195. [PMID: 35833026 PMCID: PMC9271995 DOI: 10.3389/fphar.2022.881195] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 05/19/2022] [Indexed: 11/16/2022] Open
Abstract
Astrocyte scar formation after spinal cord injury (SCI) efficiently limits the accurate damage but physically restricts the following axon regeneration. Lately, fine tuning scar formation is becoming a novel strategy to develop SCI treatment, yet how to leverage these opposite effects remains challenging. Here, utilizing an improved drug administration approach, we show that in a mouse model of spinal cord injury, continual deletion of microglia, especially upon scar formation, by pexidartinib decreases the amount of microglia-derived collagen I and reforms the astrocyte scar. The astrocytes become less compacted in the scar, which permits axon regeneration and extension. Although continual microglia deletion did not significantly improve the locomotive performance of the SCI mice, it did ameliorate their weight loss, possibly by improving their relevant health conditions. We thus identified a novel approach to regulate astrocyte scars for improved axon regeneration, which is indicative of the clinical treatment of SCI patients.
Collapse
Affiliation(s)
- Longkuo Xia
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences (CAS), Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Jianhuan Qi
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences (CAS), Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Mingming Tang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences (CAS), Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Jing Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences (CAS), Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Da Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Yanbing Zhu
- Beijing Clinical Research Institute, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- *Correspondence: Yanbing Zhu, ; Baoyang Hu,
| | - Baoyang Hu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences (CAS), Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- National Stem Cell Resource Center, Institute of Zoology (CAS), Beijing, China
- *Correspondence: Yanbing Zhu, ; Baoyang Hu,
| |
Collapse
|
237
|
The Inflammatory Response after Moderate Contusion Spinal Cord Injury: A Time Study. BIOLOGY 2022; 11:biology11060939. [PMID: 35741460 PMCID: PMC9220050 DOI: 10.3390/biology11060939] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/30/2022] [Accepted: 06/17/2022] [Indexed: 11/25/2022]
Abstract
Simple Summary The neuroinflammatory response is a rather complex event in spinal cord injury (SCI) and has the capacity to exacerbate cell damage but also to contribute to the repair of the injury. This complexity is thought to depend on a variety of inflammatory mediators, of which tumor necrosis factor (TNF) plays a key role. Evidence indicates that TNF can be both protective and detrimental in SCI. In the present study, we studied the temporal and cellular expression of TNF and its receptors after SCI in mice. We found TNF to be significantly increased in both the acute and the delayed phases after SCI, alongside a robust neuroinflammatory response. As we could verify some of our results in human postmortem tissue, our results imply that diminishing the detrimental immune signaling after SCI could also enhance recovery in humans. Abstract Spinal cord injury (SCI) initiates detrimental cellular and molecular events that lead to acute and delayed neuroinflammation. Understanding the role of the inflammatory response in SCI requires insight into the temporal and cellular synthesis of inflammatory mediators. We subjected C57BL/6J mice to SCI and investigated inflammatory reactions. We examined activation, recruitment, and polarization of microglia and infiltrating immune cells, focusing specifically on tumor necrosis factor (TNF) and its receptors TNFR1 and TNFR2. In the acute phase, TNF expression increased in glial cells and neuron-like cells, followed by infiltrating immune cells. TNFR1 and TNFR2 levels increased in the delayed phase and were found preferentially on neurons and glial cells, respectively. The acute phase was dominated by the infiltration of granulocytes and macrophages. Microglial/macrophage expression of Arg1 increased from 1–7 days after SCI, followed by an increase in Itgam, Cx3cr1, and P2ry12, which remained elevated throughout the study. By 21 and 28 days after SCI, the lesion core was populated by galectin-3+, CD68+, and CD11b+ microglia/macrophages, surrounded by a glial scar consisting of GFAP+ astrocytes. Findings were verified in postmortem tissue from individuals with SCI. Our findings support the consensus that future neuroprotective immunotherapies should aim to selectively neutralize detrimental immune signaling while sustaining pro-regenerative processes.
Collapse
|
238
|
Bourget C, Adams KV, Morshead CM. Reduced microglia activation following metformin administration or microglia ablation is sufficient to prevent functional deficits in a mouse model of neonatal stroke. J Neuroinflammation 2022; 19:146. [PMID: 35705953 PMCID: PMC9199194 DOI: 10.1186/s12974-022-02487-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/29/2022] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Neonatal stroke is a devastating insult that can lead to life-long impairments. In response to hypoxic-ischaemic injury, there is loss of neurons and glia as well as a neuroinflammatory response mediated by resident immune cells, including microglia and astrocytes, which can exacerbate damage. Administration of the antidiabetic drug metformin has been shown to improve functional outcomes in preclinical models of brain injury and the cellular basis for metformin-mediated recovery is unknown. Given metformin's demonstrated anti-inflammatory properties, we investigated its role in regulating the microglia activation and used a microglia ablation strategy to investigate the microglia-mediated outcomes in a mouse model of neonatal stroke. METHODS Hypoxia-ischaemia (H-I) was performed on post-natal day 8. Metformin was administered for one week, starting one day after injury. Immunohistochemistry was used to examine the spatiotemporal response of microglia and astrocytes after hypoxia-ischaemia, with or without metformin treatment. To evaluate the effects of microglia depletion after hypoxia-ischaemia, we delivered Plexxikon 5622 for 1 or 2 weeks post-injury. The regional pattern of microglia and astrocyte depletion was assessed through immunohistochemistry. Motor behaviour was assessed with the righting reflex, hindlimb suspension, grip strength and cylinder tests. RESULTS Herein, we revealed a spatiotemporally regulated response of microglia and astrocytes after hypoxia-ischaemia. Metformin treatment after hypoxia-ischaemia had no effect on microglia number and proliferation, but significantly reduced microglia activation in all regions examined, concomitant with improved behavioural outcomes in injured mice. Plexxikon 5622 treatment successfully ablated microglia, resulting in a > 90% depletion in microglia in the neonatal brain. Microglia rapidly repopulated upon treatment cessation of Plexxikon. Most interesting, microglia ablation was sufficient to reduce functional deficits after hypoxia-ischaemia, mimicking the effects of 1 week of metformin treatment post-injury. CONCLUSION These results highlight the importance of regulating the neuroinflammatory response after neonatal stroke to promote recovery.
Collapse
Affiliation(s)
- Clara Bourget
- Institute of Medical Sciences, University of Toronto, Toronto, M5S1A8, Canada
| | - Kelsey V Adams
- Institute of Medical Sciences, University of Toronto, Toronto, M5S1A8, Canada
| | - Cindi M Morshead
- Institute of Medical Sciences, University of Toronto, Toronto, M5S1A8, Canada.
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Room 1006, Toronto, ON, M5S3E1, Canada.
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, M5S1A8, Canada.
- Institute of Biomedical Engineering, University of Toronto, Toronto, M5S3E1, Canada.
| |
Collapse
|
239
|
Deng J, Meng F, Zhang K, Gao J, Liu Z, Li M, Liu X, Li J, Wang Y, Zhang L, Tang P. Emerging Roles of Microglia Depletion in the Treatment of Spinal Cord Injury. Cells 2022; 11:cells11121871. [PMID: 35741000 PMCID: PMC9221038 DOI: 10.3390/cells11121871] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/31/2022] [Accepted: 06/06/2022] [Indexed: 11/16/2022] Open
Abstract
Microglia, as the resident immune cells and first responder to neurological insults, play an extremely important role in the pathophysiological process of spinal cord injury. On the one hand, microglia respond rapidly and gather around the lesion in the early stage of injury to exert a protective role, but with the continuous stimulation of the injury, the excessive activated microglia secrete a large number of harmful substances, aggravate the injury of spinal cord tissue, and affect functional recovery. The effects of microglia depletion on the repair of spinal cord injury remain unclear, and there is no uniformly accepted paradigm for the removal methods and timing of microglia depletion, but different microglia depletion strategies greatly affect the outcomes after spinal cord injury. Therefore, this review summarizes the physiological and pathological roles of microglia, especially the effects of microglia depletion on spinal cord injury-sustained microglial depletion would aggravate injury and impair functional recovery, while the short-term depletion of microglial population in diseased conditions seems to improve tissue repair and promote functional improvement after spinal cord injury. Furthermore, we discuss the advantages and disadvantages of major strategies and timing of microglia depletion to provide potential strategy for the treatment of spinal cord injury.
Collapse
Affiliation(s)
- Junhao Deng
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing 100037, China;
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing 100853, China; (J.G.); (Z.L.); (M.L.); (X.L.); (J.L.)
| | - Fanqi Meng
- Department of Spine Surgery, Peking University People’s Hospital, Beijing 100044, China;
| | - Kexue Zhang
- Department of Pediatric Surgery, The Chinese PLA General Hospital, Beijing 100853, China;
| | - Jianpeng Gao
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing 100853, China; (J.G.); (Z.L.); (M.L.); (X.L.); (J.L.)
| | - Zhongyang Liu
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing 100853, China; (J.G.); (Z.L.); (M.L.); (X.L.); (J.L.)
| | - Ming Li
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing 100853, China; (J.G.); (Z.L.); (M.L.); (X.L.); (J.L.)
| | - Xiao Liu
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing 100853, China; (J.G.); (Z.L.); (M.L.); (X.L.); (J.L.)
| | - Jiantao Li
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing 100853, China; (J.G.); (Z.L.); (M.L.); (X.L.); (J.L.)
| | - Yu Wang
- Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopaedics, The Chinese PLA General Hospital, Beijing 100853, China;
| | - Licheng Zhang
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing 100037, China;
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing 100853, China; (J.G.); (Z.L.); (M.L.); (X.L.); (J.L.)
- Correspondence: (L.Z.); (P.T.)
| | - Peifu Tang
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing 100037, China;
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing 100853, China; (J.G.); (Z.L.); (M.L.); (X.L.); (J.L.)
- Correspondence: (L.Z.); (P.T.)
| |
Collapse
|
240
|
Tian F, Yang J, Xia R. Exosomes Secreted from circZFHX3-modified Mesenchymal Stem Cells Repaired Spinal Cord Injury Through mir-16-5p/IGF-1 in Mice. Neurochem Res 2022; 47:2076-2089. [PMID: 35657460 DOI: 10.1007/s11064-022-03607-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 03/28/2022] [Accepted: 04/12/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Spinal cord injury (SCI) is a devastating neurological event that leads to severe motor and sensory dysfunction. Exosome-mediated transfer of circular RNAs (circRNAs) was associated with SCI, and exosomes have been reported to be produced by mesenchymal stem cells (MSCs). This study is designed to explore the mechanism of exosomal circZFHX3 on LPS-induced MSCs injury in SCI. METHODS Exosomes were detected by transmission electron microscope and nanoparticle tracking analysis. CD9, CD63, CD81, and TSC101, B-cell lymphoma-2 (Bcl-2), Bcl-2 related X protein (Bax), Cleaved caspase 3, and Insulin-like growth factor 1 (IGF-1) protein levels were measured by western blot assay. CircZFHX3, microRNA-16-5p (miR-16-5p), and IGF-1 level were detected by real-time quantitative polymerase chain reaction (RT-qPCR). Cell viability and apoptosis were detected by Cell Counting Kit-8 (CCK-8) and flow cytometry assay. Levels of IL-1β, IL-6, and TNF-α were assessed using Enzyme-linked immunosorbent assays (ELISA). ROS, LDH, and SOD levels were measured by the special kits. The binding between miR-16-5p and circZFHX3 or IGF-1 was predicted by Starbase and DianaTools and then verified by a dual-luciferase reporter and RNA Immunoprecipitation (RIP) assays. The biological role of exosomal circZFHX3 on SCI mice was examined in vivo. RESULTS CircZFHX3 and IGF-1 were decreased, and miR-16-5p was increased in SCI mice. Also, exosomal circZFHX3 boosted cell viability and repress apoptosis, inflammation, and oxidative stress in LPS-treated BV-2 cells in vitro. Mechanically, circZFHX3 acted as a sponge of miR-16-5p to regulate IGF-1 expression. Exosomal circZFHX3 reduced cell injury of SCI in vivo. CONCLUSIONS Exosomal circZFHX3 inhibited LPS-induced BV-2 cell injury partly by regulating the miR-16-5p/ IGF-1 axis, hinting at a promising therapeutic strategy for the SCI treatment.
Collapse
Affiliation(s)
- Feng Tian
- Department of Traumatic orthopedics, The First Affiliated Hospital of USTC, No.17 Lujiang Road, Luyang District, 230001, Hefei City, Anhui Province, China
| | - Jiazhao Yang
- Department of Traumatic orthopedics, The First Affiliated Hospital of USTC, No.17 Lujiang Road, Luyang District, 230001, Hefei City, Anhui Province, China
| | - Rui Xia
- Department of Traumatic orthopedics, The First Affiliated Hospital of USTC, No.17 Lujiang Road, Luyang District, 230001, Hefei City, Anhui Province, China.
| |
Collapse
|
241
|
Wu YQ, Xiong J, He ZL, Yuan Y, Wang BN, Xu JY, Wu M, Zhang SS, Cai SF, Zhao JX, Xu K, Zhang HY, Xiao J. Metformin promotes microglial cells to facilitate myelin debris clearance and accelerate nerve repairment after spinal cord injury. Acta Pharmacol Sin 2022; 43:1360-1371. [PMID: 34480113 PMCID: PMC9160053 DOI: 10.1038/s41401-021-00759-5] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 08/08/2021] [Indexed: 02/07/2023]
Abstract
Spinal cord injury (SCI) is one kind of severe trauma for central nervous system. Myelin debris clearance and axon regeneration are essential for nerve regeneration after SCI. Metformin, a glucose-lowering drug, has been demonstrated to promote the locomotor functional recovery after SCI. In this study, we investigated the role and molecular mechanism of metformin on myelin preservation in a rat SCI model. SCI was induced in rats by compression at T9 level using a vascular clip. We showed that administration of metformin (50 mg·kg-1·d-1, ip) for 28 days significantly improved locomotor function in SCI rats. Metformin also ameliorated SCI-induced neuronal apoptosis and promoted axon regeneration in the spinal cord. Using co-immunofluorescence of IBa-1 and MBP, and luxol fasting blue (LFB) staining, we demonstrated that metformin promoted the transformation of M1 to M2 phenotype polarization of microglial cells, then greatly facilitated myelin debris clearance and protected the myelin in SCI rats. Furthermore, metformin ameliorated SCI-induced blockade of autophagic flux in the spinal cord, and enhanced the fusion of autophagosome and lysosome by inhibiting the AMPK-mTOR signaling pathway. Moreover, metformin significantly attenuated inflammatory responses in the spinal cord. In LPS-treated BV2 cells, pretreatment with metformin (2 mM) significantly enhanced autophagy level, suppressed inflammation and cell apoptosis. The protective effects were blocked in the presence of an autophagy inhibitor 3-methyladenine (3-MA, 5 mM), suggesting that the effect of metformin on autophagy in microglial cells is essential for the myelin preservation during nerve recovery. This study reveals a novel therapeutic effect of metformin in SCI recovery by regulating the activation of microglial cells and enhancing its autophagy level.
Collapse
Affiliation(s)
- Yan-Qing Wu
- The Institute of Life Sciences, Engineering Laboratory of Zhejiang Province for Pharmaceutical Development of Growth Factors, Biomedical Collaborative Innovation Center of Wenzhou, Wenzhou University, Wenzhou, 325035, China
| | - Jun Xiong
- Molecular Pharmacology Research Center, School of Pharmaceutical Science Wenzhou Medical University, Wenzhou, 325035, China
| | - Zi-Li He
- Molecular Pharmacology Research Center, School of Pharmaceutical Science Wenzhou Medical University, Wenzhou, 325035, China
| | - Yuan Yuan
- Department of Pharmacy, Hangzhou Red Cross Hospital, Zhejiang Province Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou, 310003, China
| | - Bei-Ni Wang
- Molecular Pharmacology Research Center, School of Pharmaceutical Science Wenzhou Medical University, Wenzhou, 325035, China
| | - Jing-Yu Xu
- The Institute of Life Sciences, Engineering Laboratory of Zhejiang Province for Pharmaceutical Development of Growth Factors, Biomedical Collaborative Innovation Center of Wenzhou, Wenzhou University, Wenzhou, 325035, China
| | - Man Wu
- Molecular Pharmacology Research Center, School of Pharmaceutical Science Wenzhou Medical University, Wenzhou, 325035, China
| | - Su-Su Zhang
- Molecular Pharmacology Research Center, School of Pharmaceutical Science Wenzhou Medical University, Wenzhou, 325035, China
| | - Shu-Fang Cai
- The Institute of Life Sciences, Engineering Laboratory of Zhejiang Province for Pharmaceutical Development of Growth Factors, Biomedical Collaborative Innovation Center of Wenzhou, Wenzhou University, Wenzhou, 325035, China
| | - Jia-Xin Zhao
- Molecular Pharmacology Research Center, School of Pharmaceutical Science Wenzhou Medical University, Wenzhou, 325035, China
| | - Ke Xu
- The Institute of Life Sciences, Engineering Laboratory of Zhejiang Province for Pharmaceutical Development of Growth Factors, Biomedical Collaborative Innovation Center of Wenzhou, Wenzhou University, Wenzhou, 325035, China
| | - Hong-Yu Zhang
- Molecular Pharmacology Research Center, School of Pharmaceutical Science Wenzhou Medical University, Wenzhou, 325035, China.
| | - Jian Xiao
- Molecular Pharmacology Research Center, School of Pharmaceutical Science Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
242
|
Dong Y, Jain RW, Lozinski BM, D'Mello C, Visser F, Ghorbani S, Zandee S, Brown DI, Prat A, Xue M, Yong VW. Single-cell and spatial RNA sequencing identify perturbators of microglial functions with aging. NATURE AGING 2022; 2:508-525. [PMID: 37118444 DOI: 10.1038/s43587-022-00205-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 03/11/2022] [Indexed: 04/30/2023]
Abstract
Microglia are the immune sentinels of the central nervous system with protective roles such as the removal of neurotoxic oxidized phosphatidylcholines (OxPCs). As aging alters microglial function and elevates neurological disability in diseases such as multiple sclerosis, defining aging-associated factors that cause microglia to lose their custodial properties or even become injurious can help to restore their homeostasis. We used single-cell and spatial RNA sequencing in the spinal cord of young (6-week-old) and middle-aged (52-week-old) mice to determine aging-driven microglial reprogramming at homeostasis or after OxPC injury. We identified numerous aging-associated microglial transcripts including osteopontin elevated in OxPC-treated 52-week-old mice, which correlated with greater neurodegeneration. Osteopontin delivery into the spinal cords of 6-week-old mice worsened OxPC lesions, while its knockdown in 52-week-old lesions attenuated microglial inflammation and axon loss. Thus, elevation of osteopontin and other transcripts in aging disorders including multiple sclerosis perturbs microglial functions contributing to aging-associated neurodegeneration.
Collapse
Affiliation(s)
- Yifei Dong
- Hotchkiss Brain Institute and the Department of Clinical Neuroscience, University of Calgary, Calgary, Alberta, Canada
| | - Rajiv W Jain
- Hotchkiss Brain Institute and the Department of Clinical Neuroscience, University of Calgary, Calgary, Alberta, Canada
| | - Brian M Lozinski
- Hotchkiss Brain Institute and the Department of Clinical Neuroscience, University of Calgary, Calgary, Alberta, Canada
| | - Charlotte D'Mello
- Hotchkiss Brain Institute and the Department of Clinical Neuroscience, University of Calgary, Calgary, Alberta, Canada
| | - Frank Visser
- Hotchkiss Brain Institute and the Department of Clinical Neuroscience, University of Calgary, Calgary, Alberta, Canada
| | - Samira Ghorbani
- Hotchkiss Brain Institute and the Department of Clinical Neuroscience, University of Calgary, Calgary, Alberta, Canada
| | - Stephanie Zandee
- Neuroimmunology Unit, The Research Center of the Centre Hospitalier de l'Université de Montréal (CRCHUM), Department of Neuroscience, Faculty of Medicine, Université de Montréal, Montréal, Quebec, Canada
| | - Dennis I Brown
- Hotchkiss Brain Institute and the Department of Clinical Neuroscience, University of Calgary, Calgary, Alberta, Canada
| | - Alexandre Prat
- Neuroimmunology Unit, The Research Center of the Centre Hospitalier de l'Université de Montréal (CRCHUM), Department of Neuroscience, Faculty of Medicine, Université de Montréal, Montréal, Quebec, Canada
| | - Mengzhou Xue
- Departments of Cerebrovascular Diseases and Anesthesiology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - V Wee Yong
- Hotchkiss Brain Institute and the Department of Clinical Neuroscience, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
243
|
Zheng J, Murugan M, Wang L, Wu LJ. Microglial voltage-gated proton channel Hv1 in spinal cord injury. Neural Regen Res 2022; 17:1183-1189. [PMID: 34782552 PMCID: PMC8643068 DOI: 10.4103/1673-5374.327325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/12/2020] [Accepted: 05/20/2021] [Indexed: 11/23/2022] Open
Abstract
After spinal cord injury, microglia as the first responders to the lesion display both beneficial and detrimental characteristics. Activated microglia phagocyte and eliminate cell debris, release cytokines to recruit peripheral immune cells to the injury site. Excessively activated microglia can aggravate the secondary damage by producing extravagant reactive oxygen species and pro-inflammatory cytokines. Recent studies demonstrated that the voltage-gated proton channel Hv1 is selectively expressed in microglia and regulates microglial activation upon injury. In mouse models of spinal cord injury, Hv1 deficiency ameliorates microglia activation, resulting in alleviated production of reactive oxygen species and pro-inflammatory cytokines. The reduced secondary damage subsequently decreases neuronal loss and correlates with improved locomotor recovery. This review provides a brief historical perspective of advances in investigating voltage-gated proton channel Hv1 and home in on microglial Hv1. We discuss recent studies on the roles of Hv1 activation in pathophysiological activities of microglia, such as production of NOX-dependent reactive oxygen species, microglia polarization, and tissue acidosis, particularly in the context of spinal cord injury. Further, we highlight the rationale for targeting Hv1 for the treatment of spinal cord injury and related disorders.
Collapse
Affiliation(s)
- Jiaying Zheng
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Madhuvika Murugan
- Department of Neurosurgery, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Lingxiao Wang
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
244
|
Characterization of Ex Vivo and In Vitro Wnt Transcriptome Induced by Spinal Cord Injury in Rat Microglial Cells. Brain Sci 2022; 12:brainsci12060708. [PMID: 35741593 PMCID: PMC9221341 DOI: 10.3390/brainsci12060708] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/18/2022] [Accepted: 05/27/2022] [Indexed: 12/10/2022] Open
Abstract
It is well known that inflammation is crucial in the onset and progression of neurodegenerative diseases and traumatic central nervous system (CNS) injuries, and that microglia and monocyte-derived macrophages (MDMs) play a pivotal role in neuroinflammation. Therefore, the exploration of molecular signaling pathways that are involved in the microglia/macrophage response might help us to shed light on their eventual therapeutic modulation. Interestingly, there is growing evidence showing that the Wnt family of proteins is involved in different neuropathologies that are characterized by a dysregulated neuroinflammatory response, including spinal cord injury (SCI). Here, we aimed to validate a methodology with competence to assess the physiologically relevant Wnt expression patterns of active microglia and MDMs in a rat model of SCI. For that purpose, we have selected and adapted an in vitro system of primary microglia culture that were stimulated with a lesioned spinal cord extract (SCE), together with an ex vivo protocol of flow cytometry sorting of rat microglia/MDMs at different time-points after contusive SCI. Our study demonstrates that the expression profile of Wnt-related genes in microglia/MDM cells exhibit important differences between these particular scenarios which would be in line with previous studies where similar discrepancies have been described for other molecules. Moreover, our results provide for a first experimental report of the Wnt transcriptome in rat microglia and MDMs after SCI which, together with the research platform that was used in the study, and considering its limitations, we expect might contribute to foster the research on Wnt-driven immunomodulatory therapies.
Collapse
|
245
|
Chondroitin sulfate proteoglycans prevent immune cell phenotypic conversion and inflammation resolution via TLR4 in rodent models of spinal cord injury. Nat Commun 2022; 13:2933. [PMID: 35614038 PMCID: PMC9133109 DOI: 10.1038/s41467-022-30467-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 05/03/2022] [Indexed: 12/13/2022] Open
Abstract
Chondroitin sulfate proteoglycans (CSPGs) act as potent inhibitors of axonal growth and neuroplasticity after spinal cord injury (SCI). Here we reveal that CSPGs also play a critical role in preventing inflammation resolution by blocking the conversion of pro-inflammatory immune cells to a pro-repair phenotype in rodent models of SCI. We demonstrate that enzymatic digestion of CSPG glycosaminoglycans enhances immune cell clearance and reduces pro-inflammatory protein and gene expression profiles at key resolution time points. Analysis of phenotypically distinct immune cell clusters revealed CSPG-mediated modulation of macrophage and microglial subtypes which, together with T lymphocyte infiltration and composition changes, suggests a role for CSPGs in modulating both innate and adaptive immune responses after SCI. Mechanistically, CSPG activation of a pro-inflammatory phenotype in pro-repair immune cells was found to be TLR4-dependent, identifying TLR4 signalling as a key driver of CSPG-mediated immune modulation. These findings establish CSPGs as critical mediators of inflammation resolution failure after SCI in rodents, which leads to prolonged inflammatory pathology and irreversible tissue destruction.
Collapse
|
246
|
Jiang Y, Guo J, Tang X, Wang X, Hao D, Yang H. The Immunological Roles of Olfactory Ensheathing Cells in the Treatment of Spinal Cord Injury. Front Immunol 2022; 13:881162. [PMID: 35669779 PMCID: PMC9163387 DOI: 10.3389/fimmu.2022.881162] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/22/2022] [Indexed: 01/16/2023] Open
Abstract
Spinal cord injury (SCI) is a devastating type of neurological disorder of the central nervous system (CNS) with high mortality and disability. The pathological processes of SCI can usually be described as two stages, namely, primary and acute secondary injuries. Secondary injury produces more significant exacerbations of the initial injury. Among all the mechanisms of secondary damage, infection and inflammatory responses, as the principle culprits in initiating the second phase of SCI, can greatly contribute to the severity of SCI and numerous sequelae after SCI. Therefore, effectively antagonizing pro-inflammatory responses may be a promising treatment strategy to facilitate functional recovery after SCI. Olfactory ensheathing cells (OECs), a unique type of glial cells, have increasingly become potential candidates for cell-based therapy in the injured CNS. Strikingly, there is growing evidence that the mechanisms underlying the anti-inflammatory role of OECs are associated with the immune properties and secretory functions of these cells responsible for anti-neuroinflammation and immunoregulatory effects, leading to maintenance of the internal microenvironment. Accordingly, a more profound understanding of the mechanism of OEC immunological functions in the treatment of SCI would be beneficial to improve the therapeutic clinical applications of OECs for SCI. In this review, we mainly summarize recent research on the cellular and molecular immune attributes of OECs. The unique biological functions of these cells in promoting neural regeneration are discussed in relation of the development of novel therapies for CNS injury.
Collapse
Affiliation(s)
- Yizhen Jiang
- Translational Medicine Center, Hong Hui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Jianbin Guo
- Department of Joint Surgery, Hong Hui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Xiangwen Tang
- Translational Medicine Center, Hong Hui Hospital, Xi’an Jiaotong University, Xi’an, China
- Basic Medical School Academy, Shaanxi University of Traditional Chinese Medicine, Xianyang, China
| | - Xiaohui Wang
- Department of Spine Surgery, Hong Hui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Dingjun Hao
- Department of Spine Surgery, Hong Hui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Hao Yang
- Translational Medicine Center, Hong Hui Hospital, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
247
|
Recent Advances in the Molecular and Cellular Mechanisms of gp120-Mediated Neurotoxicity. Cells 2022; 11:cells11101599. [PMID: 35626635 PMCID: PMC9139548 DOI: 10.3390/cells11101599] [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: 04/04/2022] [Revised: 05/05/2022] [Accepted: 05/07/2022] [Indexed: 02/01/2023] Open
Abstract
Axonal degeneration and loss of synapses are often seen in different brain areas of people living with human immunodeficiency virus (HIV). Nevertheless, the underlying causes of the pathological alterations observed in these individuals are poorly comprehended, considering that HIV does not infect neurons. Experimental data have shown that viral proteins, including the envelope protein gp120, cause synaptic pathology followed by neuronal cell death. These neurotoxic effects on synapses could be the result of a variety of mechanisms that decrease synaptic plasticity. In this paper, we will briefly present new emerging concepts connected with the ability of gp120 to promote the degeneration of synapses by either directly damaging the axonal cytoskeleton and/or the indirect activation of the p75 neurotrophin receptor death domain in dendrites.
Collapse
|
248
|
Wang J, Xu L, Lin W, Yao Y, Li H, Shen G, Cao X, He N, Chen J, Hu J, Zheng M, Song X, Ding Y, Shen Y, Zhong J, Wang LL, Chen YY, Zhu Y. Single-cell transcriptome analysis reveals the immune heterogeneity and the repopulation of microglia by Hif1α in mice after spinal cord injury. Cell Death Dis 2022; 13:432. [PMID: 35504882 PMCID: PMC9065023 DOI: 10.1038/s41419-022-04864-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/13/2022] [Accepted: 04/19/2022] [Indexed: 12/14/2022]
Abstract
Neuroinflammation is regarded as a vital pathological process in spinal cord injury (SCI), which removes damaged tissue, secretes cytokines, and facilitates regeneration. Repopulation of microglia has been shown to favor recovery from SCI. However, the origin and regulatory factors of microglia repopulation after SCI remain unknown. Here, we used single-cell RNA sequencing to portray the dynamic transcriptional landscape of immune cells during the early and late phases of SCI in mice. B cells and migDCs, located in the meninges under physiological conditions, are involved in immune surveillance. Microglia quickly reduced, and peripheral myeloid cells infiltrated three days-post-injury (dpi). At 14 dpi, microglia repopulated, myeloid cells were reduced, and lymphocytes infiltrated. Importantly, genetic lineage tracing of nestin+ and Cx3cr1+ cells in vivo showed that the repopulation of microglia was derived from residual microglia after SCI. We found that residual microglia regress to a developmental growth state in the early stages after SCI. Hif1α promotes microglial proliferation. Conditional ablation of Hif1α in microglia causes larger lesion sizes, fewer axon fibers, and impaired functional recovery in the late stages after SCI. Our results mapped the immune heterogeneity in SCI and raised the possibility that targeting Hif1α may help in axon regeneration and functional recovery after SCI.
Collapse
Affiliation(s)
- Jingyu Wang
- grid.412465.0Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, China ,Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Lintao Xu
- grid.412465.0Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, China ,Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Weiwei Lin
- grid.412465.0Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, China ,Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Yin Yao
- grid.412465.0Department of Neurointensive Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Heyangzi Li
- grid.13402.340000 0004 1759 700XDepartment of Basic Medicine Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Gerong Shen
- grid.13402.340000 0004 1759 700XDepartment of Basic Medicine Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Xi Cao
- grid.13402.340000 0004 1759 700XDepartment of Basic Medicine Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Ning He
- grid.13402.340000 0004 1759 700XDepartment of Basic Medicine Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Jun Chen
- grid.13402.340000 0004 1759 700XDepartment of Basic Medicine Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Jue Hu
- grid.506977.a0000 0004 1757 7957School of Basic Medical Sciences & Forensic Medicine of Hangzhou Medical College, Hangzhou, China
| | - Mingzhi Zheng
- grid.506977.a0000 0004 1757 7957School of Basic Medical Sciences & Forensic Medicine of Hangzhou Medical College, Hangzhou, China
| | - Xinghui Song
- grid.13402.340000 0004 1759 700XCore Facilities, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuemin Ding
- grid.13402.340000 0004 1759 700XSchool of Medicine, Zhejiang University City College, Hangzhou, China
| | - Yueliang Shen
- grid.13402.340000 0004 1759 700XDepartment of Basic Medicine Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinjie Zhong
- grid.13402.340000 0004 1759 700XDepartment of Basic Medicine Sciences, and Department of Obstetrics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lin-lin Wang
- grid.13402.340000 0004 1759 700XDepartment of Basic Medicine Sciences, and Department of Orthopaedics of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ying-ying Chen
- grid.13402.340000 0004 1759 700XDepartment of Basic Medicine Sciences, and Department of Obstetrics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yongjian Zhu
- grid.412465.0Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, China ,Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| |
Collapse
|
249
|
Plácido A, do Pais do Amaral C, Teixeira C, Nogueira A, Brango-Vanegas J, Alves Barbosa E, C Moreira D, Silva-Carvalho AÉ, da Silva MDG, do Nascimento Dias J, Albuquerque P, Saldanha-Araújo F, C D A Lima F, Batagin-Neto A, Kuckelhaus S, Bessa LJ, Freitas J, Dotto Brand G, C Santos N, B Relvas J, Gomes P, S A Leite JR, Eaton P. Neuroprotective effects on microglia and insights into the structure-activity relationship of an antioxidant peptide isolated from Pelophylax perezi. J Cell Mol Med 2022; 26:2793-2807. [PMID: 35460166 PMCID: PMC9097852 DOI: 10.1111/jcmm.17292] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 02/07/2022] [Accepted: 02/22/2022] [Indexed: 11/29/2022] Open
Abstract
Tryptophyllins constitute a heterogeneous group of peptides that are one of the first classes of peptides identified from amphibian’s skin secretions. Here, we report the structural characterization and antioxidant properties of a novel tryptophyllin‐like peptide, named PpT‐2, isolated from the Iberian green frog Pelophylax perezi. The skin secretion of P. perezi was obtained by electrical stimulation and fractionated using RP‐HPLC. De novo peptide sequencing was conducted using MALDI MS/MS. The primary structure of PpT‐2 (FPWLLS‐NH2) was confirmed by Edman degradation and subsequently investigated using in silico tools. PpT‐2 shared physicochemical properties with other well‐known antioxidants. To test PpT‐2 for antioxidant activity in vitro, the peptide was synthesized by solid phase and assessed in the chemical‐based ABTS and DPPH scavenging assays. Then, a flow cytometry experiment was conducted to assess PpT‐2 antioxidant activity in oxidatively challenged murine microglial cells. As predicted by the in silico analyses, PpT‐2 scavenged free radicals in vitro and suppressed the generation of reactive species in PMA‐stimulated BV‐2 microglia cells. We further explored possible bioactivities of PpT‐2 against prostate cancer cells and bacteria, against which the peptide exerted a moderate antiproliferative effect and negligible antimicrobial activity. The biocompatibility of PpT‐2 was evaluated in cytotoxicity assays and in vivo toxicity with Galleria mellonella. No toxicity was detected in cells treated with up to 512 µg/ml and in G. mellonella treated with up to 40 mg/kg PpT‐2. This novel peptide, PpT‐2, stands as a promising peptide with potential therapeutic and biotechnological applications, mainly for the treatment/prevention of neurodegenerative disorders.
Collapse
Affiliation(s)
- Alexandra Plácido
- Department of Chemistry and Biochemistry, LAQV/REQUIMTE, Faculty of Sciences, University of Porto, Porto, Portugal
| | | | - Cátia Teixeira
- Department of Chemistry and Biochemistry, LAQV/REQUIMTE, Faculty of Sciences, University of Porto, Porto, Portugal
| | - Ariane Nogueira
- Center for Research in Applied Morphology and Immunology (NuPMIA), University of Brasilia, Brasilia, Brazil
| | - José Brango-Vanegas
- Center for Research in Applied Morphology and Immunology (NuPMIA), University of Brasilia, Brasilia, Brazil
| | - Eder Alves Barbosa
- Center for Research in Applied Morphology and Immunology (NuPMIA), University of Brasilia, Brasilia, Brazil.,Laboratory of Synthesis and Analysis of Biomolecules (LSAB), Institute of Chemistry (IQ), University of Brasilia, Brasília, Brazil
| | - Daniel C Moreira
- Center for Research in Applied Morphology and Immunology (NuPMIA), University of Brasilia, Brasilia, Brazil
| | - Amandda É Silva-Carvalho
- Laboratory of Hematology and Stem Cells, Faculty of Health Sciences, University of Brasilia, Brasília, Brazil
| | - Maria da Gloria da Silva
- Center for Research in Applied Morphology and Immunology (NuPMIA), University of Brasilia, Brasilia, Brazil
| | - Jhones do Nascimento Dias
- Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil.,Biomedicine Course, Federal University of Delta do Parnaíba (UFDPar), Parnaíba, Brazil
| | - Patrícia Albuquerque
- Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil.,Faculty of Ceilândia, University of Brasilia, Brasilia, Brazil
| | - Felipe Saldanha-Araújo
- Laboratory of Hematology and Stem Cells, Faculty of Health Sciences, University of Brasilia, Brasília, Brazil
| | - Filipe C D A Lima
- Federal Institute of Education, Science and Technology of São Paulo, Matão, Brazil
| | | | - Selma Kuckelhaus
- Center for Research in Applied Morphology and Immunology (NuPMIA), University of Brasilia, Brasilia, Brazil
| | - Lucinda J Bessa
- Department of Chemistry and Biochemistry, LAQV/REQUIMTE, Faculty of Sciences, University of Porto, Porto, Portugal.,Egas Moniz Interdisciplinary Research Center (CiiEM), Egas Moniz - Cooperative for Higher Education, CRL, Almada, Portugal
| | - Jaime Freitas
- Institute for Research and Innovation in Health (i3S), National Institute of Biomedical Engineering (INEB), University of Porto, Porto, Portugal
| | - Guilherme Dotto Brand
- Laboratory of Synthesis and Analysis of Biomolecules (LSAB), Institute of Chemistry (IQ), University of Brasilia, Brasília, Brazil
| | - Nuno C Santos
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - João B Relvas
- Institute for Research and Innovation in Health (i3S), Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal
| | - Paula Gomes
- Department of Chemistry and Biochemistry, LAQV/REQUIMTE, Faculty of Sciences, University of Porto, Porto, Portugal
| | - José Roberto S A Leite
- Department of Chemistry and Biochemistry, LAQV/REQUIMTE, Faculty of Sciences, University of Porto, Porto, Portugal.,Center for Research in Applied Morphology and Immunology (NuPMIA), University of Brasilia, Brasilia, Brazil
| | - Peter Eaton
- Department of Chemistry and Biochemistry, LAQV/REQUIMTE, Faculty of Sciences, University of Porto, Porto, Portugal.,The Bridge, School of Chemistry, Joseph Banks Laboratories, University of Lincoln, Lincoln, UK
| |
Collapse
|
250
|
Cai L, Gong Q, Qi L, Xu T, Suo Q, Li X, Wang W, Jing Y, Yang D, Xu Z, Yuan F, Tang Y, Yang G, Ding J, Chen H, Tian H. ACT001 attenuates microglia-mediated neuroinflammation after traumatic brain injury via inhibiting AKT/NFκB/NLRP3 pathway. Cell Commun Signal 2022; 20:56. [PMID: 35461293 PMCID: PMC9035258 DOI: 10.1186/s12964-022-00862-y] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 03/12/2022] [Indexed: 12/20/2022] Open
Abstract
Abstract
Background
Microglia-mediated neuroinflammatory response following traumatic brain injury (TBI) is considered as a vital secondary injury factor, which drives trauma-induced neurodegeneration and is lack of efficient treatment. ACT001, a sesquiterpene lactone derivative, is reportedly involved in alleviation of inflammatory response. However, little is known regarding its function in regulating innate immune response of central nervous system (CNS) after TBI. This study aimed to investigate the role and underlying mechanism of ACT001 in TBI.
Methods
Controlled cortical impact (CCI) models were used to establish model of TBI. Cresyl violet staining, evans blue extravasation, neurobehavioral function assessments, immunofluorescence and transmission electron microscopy were used to evaluate therapeutic effects of ACT001 in vivo. Microglial depletion was induced by administering mice with colony stimulating factor 1 receptor (CSF1R) inhibitor, PLX5622. Cell-cell interaction models were established as co-culture system to simulate TBI conditions in vitro. Cytotoxic effect of ACT001 on cell viability was assessed by cell counting kit-8 and activation of microglia cells were induced by Lipopolysaccharides (LPS). Pro-inflammatory cytokines expression was determined by Real-time PCR and nitric oxide production. Apoptotic cells were detected by TUNEL and flow cytometry assays. Tube formation was performed to evaluate cellular angiogenic ability. ELISA and western blot experiments were used to determine proteins expression. Pull-down assay was used to analyze proteins that bound ACT001.
Results
ACT001 relieved the extent of blood-brain barrier integrity damage and alleviated motor function deficits after TBI via reducing trauma-induced activation of microglia cells. Delayed depletion of microglia with PLX5622 hindered therapeutic effect of ACT001. Furthermore, ACT001 alleviated LPS-induced activation in mouse and rat primary microglia cells. Besides, ACT001 was effective in suppressing LPS-induced pro-inflammatory cytokines production in BV2 cells, resulting in reduction of neuronal apoptosis in HT22 cells and improvement of tube formation in bEnd.3 cells. Mechanism by which ACT001 functioned was related to AKT/NFκB/NLRP3 pathway. ACT001 restrained NFκB nuclear translocation in microglia cells through inhibiting AKT phosphorylation, resulting in decrease of NLRP3 inflammasome activation, and finally down-regulated microglial neuroinflammatory response.
Conclusions
Our study indicated that ACT001 played critical role in microglia-mediated neuroinflammatory response and might be a novel potential chemotherapeutic drug for TBI.
Collapse
|