1
|
Qu W, Wu X, Wu W, Wang Y, Sun Y, Deng L, Walker M, Chen C, Dai H, Han Q, Ding Y, Xia Y, Smith G, Li R, Liu NK, Xu XM. Chondroitinase ABC combined with Schwann cell transplantation enhances restoration of neural connection and functional recovery following acute and chronic spinal cord injury. Neural Regen Res 2025; 20:1467-1482. [PMID: 39075913 PMCID: PMC11624882 DOI: 10.4103/nrr.nrr-d-23-01338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 12/20/2023] [Accepted: 05/16/2024] [Indexed: 07/31/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202505000-00029/figure1/v/2024-07-28T173839Z/r/image-tiff Schwann cell transplantation is considered one of the most promising cell-based therapy to repair injured spinal cord due to its unique growth-promoting and myelin-forming properties. A the Food and Drug Administration-approved Phase I clinical trial has been conducted to evaluate the safety of transplanted human autologous Schwann cells to treat patients with spinal cord injury. A major challenge for Schwann cell transplantation is that grafted Schwann cells are confined within the lesion cavity, and they do not migrate into the host environment due to the inhibitory barrier formed by injury-induced glial scar, thus limiting axonal reentry into the host spinal cord. Here we introduce a combinatorial strategy by suppressing the inhibitory extracellular environment with injection of lentivirus-mediated transfection of chondroitinase ABC gene at the rostral and caudal borders of the lesion site and simultaneously leveraging the repair capacity of transplanted Schwann cells in adult rats following a mid-thoracic contusive spinal cord injury. We report that when the glial scar was degraded by chondroitinase ABC at the rostral and caudal lesion borders, Schwann cells migrated for considerable distances in both rostral and caudal directions. Such Schwann cell migration led to enhanced axonal regrowth, including the serotonergic and dopaminergic axons originating from supraspinal regions, and promoted recovery of locomotor and urinary bladder functions. Importantly, the Schwann cell survival and axonal regrowth persisted up to 6 months after the injury, even when treatment was delayed for 3 months to mimic chronic spinal cord injury. These findings collectively show promising evidence for a combinatorial strategy with chondroitinase ABC and Schwann cells in promoting remodeling and recovery of function following spinal cord injury.
Collapse
Affiliation(s)
- Wenrui Qu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Hand Surgery, the Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Xiangbing Wu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Wei Wu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ying Wang
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yan Sun
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Lingxiao Deng
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Melissa Walker
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Chen Chen
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Heqiao Dai
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Qi Han
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ying Ding
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yongzhi Xia
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - George Smith
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, PA, USA
| | - Rui Li
- Department of Hand Surgery, the Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Nai-Kui Liu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Xiao-Ming Xu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
2
|
Zha X, Zheng G, Skutella T, Kiening K, Unterberg A, Younsi A. Microglia: a promising therapeutic target in spinal cord injury. Neural Regen Res 2025; 20:454-463. [PMID: 38819048 PMCID: PMC11317945 DOI: 10.4103/nrr.nrr-d-23-02044] [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: 12/18/2023] [Revised: 02/23/2024] [Accepted: 03/22/2024] [Indexed: 06/01/2024] Open
Abstract
Microglia are present throughout the central nervous system and are vital in neural repair, nutrition, phagocytosis, immunological regulation, and maintaining neuronal function. In a healthy spinal cord, microglia are accountable for immune surveillance, however, when a spinal cord injury occurs, the microenvironment drastically changes, leading to glial scars and failed axonal regeneration. In this context, microglia vary their gene and protein expression during activation, and proliferation in reaction to the injury, influencing injury responses both favorably and unfavorably. A dynamic and multifaceted injury response is mediated by microglia, which interact directly with neurons, astrocytes, oligodendrocytes, and neural stem/progenitor cells. Despite a clear understanding of their essential nature and origin, the mechanisms of action and new functions of microglia in spinal cord injury require extensive research. This review summarizes current studies on microglial genesis, physiological function, and pathological state, highlights their crucial roles in spinal cord injury, and proposes microglia as a therapeutic target.
Collapse
Affiliation(s)
- Xiaowei Zha
- Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Guoli Zheng
- Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Thomas Skutella
- Department of Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Karl Kiening
- Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Andreas Unterberg
- Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Alexander Younsi
- Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
3
|
Zhou Y, Zhang Y, Xu D, Yang C, Lin X, Jin K, Xia L, Zhuge Q, Yang S. Exosomes from polarized Microglia: Proteomic insights into potential mechanisms affecting intracerebral hemorrhage. Gene 2025; 935:149080. [PMID: 39510328 DOI: 10.1016/j.gene.2024.149080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 11/15/2024]
Abstract
Intracerebral hemorrhage (ICH) is a devastating form of stroke associated with significant morbidity and mortality. Microglia are intracranial innate immune cell that play critical roles in Intracerebral hemorrhage through direct or indirect means. Vesicle transport is a fundamental mechanism of intercellular communication. Recent studies have identified microglia in specific polarized states correlate with pathogenesis, material and signal transmission in ICH through derived extracellular vesicles. Diverse polarization states trigger distinct functions, however, the exosome proteomes across these states remain poorly characterized. Here, we hypothesized that microglia exosomal profiles vary with polarization states, impacting their functional repertoire and influencing outcomes in cerebral hemorrhage. In vitro model of cerebral hemorrhage, administration of 20 μg/ml LPS-induced M1 microglia derived exosomes (M1-Exo) with HT22 enhanced hemin-induced neuronal death, while IL-4-induced M2 microglia derived exosomes (M2-Exo) significantly reduced hemin-induced cell apoptosis and inflammation. Then we identified novel state-specific proteomic profiles of microglia-derived exosomes under these polarization conditions through label-free quantitative mass spectrometry (LFQ-MS). Analysis of protein content identified several exosomal signature proteins and hundreds of differentially expressed proteins across polarization states. Specifically, proteins including UMOD, NLRP3, ACOD1, IL1RN, heme oxygenase 1 (HMOX1), CCL4, and TNFRSF1B in M1-Exo were enriched in inflammatory pathways, while those in M2-Exo exhibited enrichment in autophagy, ubiquitination, and mitochondrial respiration. The analysis of those diverse exosomal proteins suggested unique proteomic profiles and possible intracellular signal transmission and regulation mechanisms. Together, these findings offer new insights and resources for studying microglia-derived exosome and pave the way for the development of novel therapeutic strategies targeting microglial exosome-mediated pathways.
Collapse
Affiliation(s)
- Yinan Zhou
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Ying Zhang
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Zhejiang-US Joint Laboratory for Aging and Neurological Disease Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Dongchen Xu
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Chenguang Yang
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Xiao Lin
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Kunlin Jin
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Lei Xia
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Qichuan Zhuge
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Zhejiang-US Joint Laboratory for Aging and Neurological Disease Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| | - Su Yang
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Zhejiang-US Joint Laboratory for Aging and Neurological Disease Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| |
Collapse
|
4
|
Yang ZF, Jiang XC, Gao JQ. Present insights into the progress in gene therapy delivery systems for central nervous system diseases. Int J Pharm 2025; 669:125069. [PMID: 39662855 DOI: 10.1016/j.ijpharm.2024.125069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 12/06/2024] [Accepted: 12/08/2024] [Indexed: 12/13/2024]
Abstract
Central nervous system (CNS) diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), spinal cord injury (SCI), and ischemic strokes and certain rare diseases, such as amyotrophic lateral sclerosis (ALS) and ataxia, present significant obstacles to treatment using conventional molecular pharmaceuticals. Gene therapy, with its ability to target previously "undruggable" proteins with high specificity and safety, is increasingly utilized in both preclinical and clinical research for CNS ailments. As our comprehension of the pathophysiology of these conditions deepens, gene therapy stands out as a versatile and promising strategy with the potential to both prevent and treat these diseases. Despite the remarkable progress in refining and enhancing the structural design of gene therapy agents, substantial obstacles persist in their effective and safe delivery within living systems. To surmount these obstacles, a diverse array of gene delivery systems has been devised and continuously improved. Notably, Adeno-Associated Virus (AAVs)-based viral gene vectors and lipid-based nanocarriers have each advanced the in vivo delivery of gene therapies to various extents. This review aims to concisely summarize the pathophysiological foundations of CNS diseases and to shed light on the latest advancements in gene delivery vector technologies. It discusses the primary categories of these vectors, their respective advantages and limitations, and their specialized uses in the context of gene therapy delivery.
Collapse
Affiliation(s)
- Ze-Feng Yang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xin-Chi Jiang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China..
| | - Jian-Qing Gao
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China..
| |
Collapse
|
5
|
Stewart AN, Bosse-Joseph CC, Kumari R, Bailey WM, Park KA, Slone VK, Gensel JC. Nonresolving Neuroinflammation Regulates Axon Regeneration in Chronic Spinal Cord Injury. J Neurosci 2025; 45:e1017242024. [PMID: 39510834 DOI: 10.1523/jneurosci.1017-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/17/2024] [Accepted: 10/11/2024] [Indexed: 11/15/2024] Open
Abstract
Chronic spinal cord injury (SCI) lesions retain increased densities of microglia and macrophages. In acute SCI, macrophages induce growth cone collapse and facilitate axon retraction away from lesion boundaries. Little is known about the role of sustained inflammation in chronic SCI or whether chronic inflammation affects regeneration. We used the colony-stimulating factor-1 receptor inhibitor, PLX-5622, to deplete microglia and macrophages months after complete crush SCI in female mice. Transcriptional analyses revealed a significant inflammatory depletion within chronic SCI lesions after PLX-5622 treatment. Both transcriptional analyses and immunohistochemistry revealed that Iba1+ cells repopulate to predepleted densities after treatment removal. Neuronal-enriched transcripts were significantly elevated in mice after inflammatory repopulation, but no significant effects were observed with inflammatory depletion alone. Axon densities also significantly increased within the lesion after PLX-5622 treatment and after repopulation. To better examine the effect of chronic inflammation on axon regeneration, we tested PLX-5622 treatment in neuronal-specific phosphatase and tensin homolog protein (PTEN) knock-out (PTEN-KO) mice. PTEN-KO was delivered using spinal injections of retrogradely transported adeno-associated viruses (AAVrg's). PTEN-KO did not further increase axon densities within the lesion beyond the effects induced by PLX-5622. Axons that grew within the lesion were histologically identified as 5-HT+ and CGRP+, both of which are not robustly transduced by AAVrg's. Our work identified that increased macrophage/microglial densities in the chronic SCI environment may be actively retained by homeostatic mechanisms likely affiliated with a sustained elevated expression of CSF1 and other chemokines. Finally, we identify a novel role of sustained inflammation as a prospective barrier to axon regeneration in chronic SCI.
Collapse
Affiliation(s)
- Andrew N Stewart
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky 40536
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky 40536
- College of Medicine, University of Kentucky, Lexington, Kentucky 40536
| | - Christopher C Bosse-Joseph
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky 40536
- College of Medicine, University of Kentucky, Lexington, Kentucky 40536
| | - Reena Kumari
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky 40536
- College of Medicine, University of Kentucky, Lexington, Kentucky 40536
- Department of Physiology, University of Kentucky, Lexington, Kentucky 40536
| | - William M Bailey
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky 40536
- College of Medicine, University of Kentucky, Lexington, Kentucky 40536
- Department of Physiology, University of Kentucky, Lexington, Kentucky 40536
| | - Kennedy A Park
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky 40536
- College of Medicine, University of Kentucky, Lexington, Kentucky 40536
| | - Victoria K Slone
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky 40536
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky 40536
- College of Medicine, University of Kentucky, Lexington, Kentucky 40536
| | - John C Gensel
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky 40536
- College of Medicine, University of Kentucky, Lexington, Kentucky 40536
- Department of Physiology, University of Kentucky, Lexington, Kentucky 40536
| |
Collapse
|
6
|
Verma H, Kaur S, Jeeth P, Kumar P, Kadhirvel S, Dhiman M, Mantha AK. Understanding Aβ 25-35 peptide altered exosomal proteome and associated pathways linked with the Alzheimer's disease pathogenesis using human neuroblastoma SH-SY5Y Cells. Metab Brain Dis 2024; 40:25. [PMID: 39565424 DOI: 10.1007/s11011-024-01469-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 09/11/2024] [Indexed: 11/21/2024]
Abstract
The central nervous system (CNS) involves a complex interplay of communications between the neurons and various glial cells, which is crucial for brain functions. The major interactomes are exosomes that transmit sundry molecules (DNA, miRNAs, and proteins) between the cells and thus alter cell physiology. Exosomes can act as neuroprotective or neurodegenerative agents depending on the microenvironment of cells secreting them. Therefore, revealing exosome proteome becomes important to understand donor cells' physiology and its effect on the recipient cell. In this study, oxidative stress was induced by Aβ25-35 in the human neuroblastoma SH-SY5Y cells and the protective effects of phytochemical ferulic acid (FA) were evaluated alone and in combination with Aβ25-35 (pre-treated for 3 h before Aβ25-35 exposure) and proteome of their secreted exosomes was analyzed, which was carried out via a high-resolution LC-MS Triple-ToF and further network-based analysis has been carried out using various bioinformatics tools. The proteomic profiling enlightened the multiple roles of exosomes as proteins associated with the various pathways advocate that exosomes can mediate a wide range of effects, from normal physiological processes like synaptic plasticity, neuronal metabolic support, nerve regeneration, DNA repair, axon guidance, and long-term potentiation (LTP) to abnormal pathological processes like inflammatory responses, oxidative stress, apoptosis, and formation of neutrophil extracellular traps (NETs). On comparison, treatment with Aβ25-35 resulted in a significant modulation of the exosomal proteome, promoting pathways associated with neurodegeneration. Conversely, the phytochemical FA displayed a protective effect by effectively countering Aβ25-35-induced oxidative stress responses linked with neurodegeneration, as seen in Alzheimer's disease (AD). Taken together, this study highlights the dual role of exosomes in physiological and pathophysiological neurodegenerative AD, which intricately depend on the particular cellular milieu.
Collapse
Affiliation(s)
- Harkomal Verma
- Department of Zoology, School of Basic Sciences, Central University of Punjab, VPO - Ghudda, Bathinda, 151 401, Punjab, India
| | - Sharanjot Kaur
- Department of Microbiology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| | - Priyanka Jeeth
- Department of Computational Sciences, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| | - Puneet Kumar
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| | - Saraboji Kadhirvel
- Department of Computational Sciences, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| | - Monisha Dhiman
- Department of Microbiology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| | - Anil Kumar Mantha
- Department of Zoology, School of Basic Sciences, Central University of Punjab, VPO - Ghudda, Bathinda, 151 401, Punjab, India.
| |
Collapse
|
7
|
Zhang K, Wen R, Ma W, Ji H, He X, Yang Z, Liu D, Li X. P2Y12-targeted modulation of microglial phenotypes: A novel therapeutic strategy for enhanced axonal regeneration post-spinal cord injury. Life Sci 2024; 357:123057. [PMID: 39277132 DOI: 10.1016/j.lfs.2024.123057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/08/2024] [Accepted: 09/11/2024] [Indexed: 09/17/2024]
Abstract
AIMS Microglia activation after spinal cord injury (SCI) is a double-edged sword, modulation of the activated microglia populations toward pro-regenerative phenotypes highlights the potential therapeutic implications. P2Y12, a microglia-specific marker, remains underexplored in its capacity to polarize microglial activation populations in SCI repair. We aimed to explore the effects of modulating P2Y12 on microglia function after spinal cord injury, and further on axonal regeneration and motor recovery after spinal cord injury. MATERIALS AND METHODS The study employed both in vitro and in vivo models, using BV2 cells and a mouse model of SCI, respectively. Ticagrelor, a P2Y12 antagonist, was administered via a collagen scaffold to ensure stable and sustained release. Transcriptome sequencing analysis, immunofluorescence staining, and Basso Mouse Scale (BMS) scores were used to assess microglial activation, axonal regeneration, and functional recovery. KEY FINDINGS Herein, we observed P2Y12+ microglia localized predominantly at the lesion periphery within 3 days post injury (dpi), manifesting a pro-inflammatory phenotype, but not anti-inflammatory phenotype. In vitro investigations revealed that P2Y12 inhibition of the activated microglia curtailed pro-inflammatory differentiation while augmenting anti-inflammatory differentiation. SIGNIFICANCE Leveraging this insight, we engineered a collagen scaffold-based delivery system for sustained release of the P2Y12 antagonist, ticagrelor, at the injury site in a mouse complete SCI model. Notably, P2Y12 suppression markedly enhanced axonal regeneration within the injured site and ameliorated lower limb motor functions in SCI mice. Collectively, our findings illuminate P2Y12-targeted microglial modulation as a promising therapeutic approach for SCI.
Collapse
Affiliation(s)
- Kai Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Runlin Wen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Wanrong Ma
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha 410078, Hunan Province, China
| | - Huaqing Ji
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha 410078, Hunan Province, China
| | - Xinghui He
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Zhiquan Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Dingyang Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Xing Li
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha 410078, Hunan Province, China.
| |
Collapse
|
8
|
Yuan Y, Xu M, Feng L, Zhong W, Zhang L, Du R, Sun J, Wang C, Du J. Nanozyme Hydrogels Promote Nerve Regeneration in Spinal Cord Injury by Reducing Oxidative Stress. ACS APPLIED MATERIALS & INTERFACES 2024; 16:59949-59961. [PMID: 39454206 DOI: 10.1021/acsami.4c13671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2024]
Abstract
Inhibiting secondary cell death and promoting neuronal regeneration are critical for nerve repair after spinal cord injury (SCI). The excessive accumulation of reactive oxygen species (ROS) after SCI causes cell death and induces apoptosis. These reactions further increase the level of ROS production, leading to a vicious cycle of spinal cord tissue damage. Therefore, intervention targeting ROS is a potential therapeutic approach to improve the recovery of locomotor function after SCI. In this study, we designed and synthesized a nanozyme hydrogel delivery system loaded with multiple drugs, LA/Me/Se NPs-h. LA/Me/Se NPs-h exhibited a satisfactory size distribution and excellent stability, enhancing the bioavailability of therapeutic drugs. Moreover, we explored the antioxidant and protective effects of LA/Me/Se NPs-h against oxidative stress-induced cell damage caused by ROS production after SCI in vitro. In the mice SCI model, the Basso mouse scale and gait analysis showed that LA/Me/Se NPs-h significantly promoted the recovery of locomotor function after SCI. The histological and immunofluorescence results of the injury site revealed that LA/Me/Se NPs-h upregulated the expression of GFAP, NF-200, and superoxide dismutase in spinal cord lesion, reduced caspase-3 expression, improved spinal cord continuity, reduced lesion cavity, and inhibited the axonal demyelination. Consequently, LA/Me/Se NPs-h increased the activity of antioxidant enzymes and reduced neuronal apoptosis by reducing oxidative stress and ultimately promoted nerve regeneration. Taken together, this study demonstrated promising nanozyme hydrogels and provided an effective therapeutic strategy for SCI and other ROS-related diseases.
Collapse
Affiliation(s)
- Yitong Yuan
- Department of Medical Imaging, Shanxi Key Laboratory of Intelligent Imaging and Nanomedicine, First Hospital of Shanxi Medical University, Taiyuan 030001, P. R. China
- Laboratory Animal Center, Shanxi Key Laboratory of Experimental Animal Science and Animal Model of Human Disease, Shanxi Medical University, Taiyuan 030001, P. R. China
- Department of Histology and Embryology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, P. R. China
| | - Miaomiao Xu
- Academy of Medical Sciences, Shanxi Medical University, Taiyuan 030001, P. R. China
| | - Limin Feng
- School of Stomatology, Shanxi Medical University, Taiyuan 030001, P. R. China
| | - Wanting Zhong
- Academy of Medical Sciences, Shanxi Medical University, Taiyuan 030001, P. R. China
| | - Longzhu Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, P. R. China
| | - Ruochen Du
- Laboratory Animal Center, Shanxi Key Laboratory of Experimental Animal Science and Animal Model of Human Disease, Shanxi Medical University, Taiyuan 030001, P. R. China
| | - Jingjing Sun
- Zhenjiang Mental Health Center, Zhenjiang 212000, P. R. China
| | - Chunfang Wang
- Laboratory Animal Center, Shanxi Key Laboratory of Experimental Animal Science and Animal Model of Human Disease, Shanxi Medical University, Taiyuan 030001, P. R. China
- School of Stomatology, Shanxi Medical University, Taiyuan 030001, P. R. China
| | - Jiangfeng Du
- Department of Medical Imaging, Shanxi Key Laboratory of Intelligent Imaging and Nanomedicine, First Hospital of Shanxi Medical University, Taiyuan 030001, P. R. China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan 030001, P. R. China
| |
Collapse
|
9
|
Yuan NJ, Zhu WJ, Ma QY, Huang MY, Huo RR, She KJ, Pan JP, Wang JG, Chen JX. Luteolin ameliorates chronic stress-induced depressive-like behaviors in mice by promoting the Arginase-1 + microglial phenotype via a PPARγ-dependent mechanism. Acta Pharmacol Sin 2024:10.1038/s41401-024-01402-9. [PMID: 39496862 DOI: 10.1038/s41401-024-01402-9] [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: 04/30/2024] [Accepted: 09/23/2024] [Indexed: 11/06/2024] Open
Abstract
Accumulating evidence shows that neuroinflammation substantially contributes to the pathology of depression, a severe psychiatric disease with an increasing prevalence worldwide. Although modulating microglial phenotypes is recognized as a promising therapeutic strategy, effective treatments are still lacking. Previous studies have shown that luteolin (LUT) has anti-inflammatory effects and confers benefits on chronic stress-induced depression. In this study, we investigated the molecular mechanisms by which LUT regulates the functional phenotypes of microglia in mice with depressive-like behaviors. Mice were exposed to chronic restraint stress (CRS) for 7 weeks, and were administered LUT (10, 30, 40 mg· kg-1 ·day-1, i.g.) in the last 4 weeks. We showed that LUT administration significantly ameliorated depressive-like behaviors and decreased hippocampal inflammation. LUT administration induced pro-inflammatory microglia to undergo anti-inflammatory arginase (Arg)-1+ phenotypic polarization, which was associated with its antidepressant effects. Furthermore, we showed that LUT concentration-dependently increased the expression of PPARγ in LPS + ATP-treated microglia and the hippocampus of CRS-exposed mice, promoting the subsequent inhibition of the NLRP3 inflammasome. Molecular dynamics (MD) simulation and microscale thermophoresis (MST) analysis confirmed a direct interaction between LUT and peroxisome proliferator-activated receptor gamma (PPARγ). By using the PPARγ antagonist GW9662, we demonstrated that LUT-driven protection, both in vivo and in vitro, resulted from targeting PPARγ. First, LUT-induced Arg-1+ microglia were no longer detected when PPARγ was blocked. Next, LUT-mediated inhibition of the NLRP3 inflammasome and downregulation of pro-inflammatory cytokine production were reversed by the inhibition of PPARγ. Finally, the protective effects of LUT, which attenuated the microglial engulfment of synapses and prevented apparent synapse loss in the hippocampus of CRS-exposed mice, were eliminated by blocking PPARγ. In conclusion, this study showed that LUT ameliorates CRS-induced depressive-like behaviors by promoting the Arg-1+ microglial phenotype through a PPARγ-dependent mechanism, thereby alleviating microglial pro-inflammatory responses and reversing microglial phagocytosis-mediated synapse loss.
Collapse
Affiliation(s)
- Nai-Jun Yuan
- Department of Critical Care Medicine, Shenzhen Clinical Research Center for Geriatric, and Guangdong Provincial Clinical Research Center for Geriatrics, Integrated Chinese and Western Medicine Postdoctoral Research Station, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, 518020, China
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Wen-Jun Zhu
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Qing-Yu Ma
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Min-Yi Huang
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Rou-Rou Huo
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Kai-Jie She
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Jun-Ping Pan
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, 1478, Lørenskog, Norway
- Guangdong Second Provincial General Hospital, Postdoctoral Research Station of Basic Medicine, Jinan University, Guangzhou, 510632, China
| | - Ji-Gang Wang
- Department of Critical Care Medicine, Shenzhen Clinical Research Center for Geriatric, and Guangdong Provincial Clinical Research Center for Geriatrics, Integrated Chinese and Western Medicine Postdoctoral Research Station, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, 518020, China.
- State Key Laboratory for Quality Assurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Jia-Xu Chen
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China.
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
10
|
Li H, Liu Y, Sun Y, Guo H, Lv S, Guo W, Ren J, Wang Y, Zu J, Yan J, Wang N. Targeting astrocytes polarization after spinal cord injury: a promising direction. Front Cell Neurosci 2024; 18:1478741. [PMID: 39479524 PMCID: PMC11521873 DOI: 10.3389/fncel.2024.1478741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 10/07/2024] [Indexed: 11/02/2024] Open
Abstract
Spinal cord injury (SCI) is a serious neurological injury that causes severe trauma to motor and sensory functions. Although long considered incurable, recent research has brought new hope for functional recovery from SCI. After SCI, astrocytes are activated into many polarization states. Here we discuss the two most important classical phenotypes: the 'A1' neurotoxic phenotype and the 'A2' neuroprotective phenotype, with A1 astrocytes being neurotoxic and impeding neurorecovery, and A2 astrocytes being neuroprotective. This paper discusses the changes in astrocyte responsiveness after SCI and the pros and cons of their polarization in SCI. It also elucidates the feasibility of astrocyte polarization as a therapeutic target for neuroprotection. In the future, multiple intervention strategies targeting astrocyte polarization are expected to gain wider clinical application, ultimately improving motor-sensory function and quality of life in SCI patients.
Collapse
Affiliation(s)
- Helin Li
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Ying Liu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yucao Sun
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China
| | - Hangyu Guo
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Shiyan Lv
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Wenhui Guo
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Jiyu Ren
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yufu Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Jianing Zu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Jinglong Yan
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Nanxiang Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| |
Collapse
|
11
|
Chen R, Hu J, Zhang Y, Liu Y, Cao L, He F, Wang Q, Chen Y, Zhang S, Tang S, Min B. Sodium aescinate alleviates neuropathic pain through suppressing OGT-mediated O-GlcNAc modification of TLR3 to inactivate MAPK signaling pathway. Brain Res Bull 2024; 217:111077. [PMID: 39265741 DOI: 10.1016/j.brainresbull.2024.111077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 09/08/2024] [Accepted: 09/10/2024] [Indexed: 09/14/2024]
Abstract
Neuropathic pain results from damage to nerves or the brain, and is characterized by symptoms such as allodynia, spontaneous pain, and hyperalgesia. The causes of this type of pain are intricate, which can make it difficult to treat. Sodium aescinate (SA), a natural extract from horse chestnut tree seeds, has been shown to act as a neuroprotector by inhibiting microglia activation. This study aims to explore the therapeutic potential of SA for neuropathic pain and the molecular mechanisms regulated by SA treatment. Through in vivo animal models and experiments, we found that SA treatment significantly reduced mechanical allodynia and heat hyperalgesia in neuropathic pain models. Additionally, SA inhibited O-GlcNAc-transferase (OGT)-induced O-GlcNAcylation (O-GlcNAc) modification in neuropathic pain mice. OGT overexpression could impede the therapeutic effects of SA on neuropathic pain. Further investigation revealed that Toll-like receptor 3 (TLR3), stabilized by OGT-induced O-GlcNAc modification, could activate the Mitogen activated protein kinase (MAPK) signaling pathway. Further in vivo experiments demonstrated that TLR3-mediated p38 mitogen-activated protein kinase (p38MAPK) activation is involved in SA-mediated relief of neuropathic pain. In conclusion, this study uncovers a novel molecular pathway deactivated by SA treatment in neuropathic pain.
Collapse
Affiliation(s)
- Rong Chen
- Department of Pain, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550001, China
| | - Jiantao Hu
- Department of Respiratory Medicine, Bijie City Qixingguan District People's Hospital, Bijie, Guizhou 551700, China
| | - Yang Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550001, China
| | - Yang Liu
- Department of Orthopedics, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550001, China
| | - Liujian Cao
- Department of Anorectal Surgery, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550001, China
| | - Fan He
- Department of Oncology, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550001, China
| | - Qin Wang
- Department of Rheumatology, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550001, China
| | - Ying Chen
- Department of Anesthesiology, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550001, China
| | - Shengwei Zhang
- Graduate School, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550001, China.
| | - Songjiang Tang
- Department of Anesthesiology, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550001, China
| | - Baojun Min
- Department of Anesthesiology, Qianxi People's Hospital, Bijie, Guizhou 551500, China.
| |
Collapse
|
12
|
Ganesan S, Dharmarajan A, Sudhir G, Perumalsamy LR. Unravelling the Road to Recovery: Mechanisms of Wnt Signalling in Spinal Cord Injury. Mol Neurobiol 2024; 61:7661-7679. [PMID: 38421469 DOI: 10.1007/s12035-024-04055-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 02/12/2024] [Indexed: 03/02/2024]
Abstract
Spinal cord injury (SCI) is a complex neurodegenerative pathology that consistently harbours a poor prognostic outcome. At present, there are few therapeutic strategies that can halt neuronal cell death and facilitate functional motor recovery. However, recent studies have highlighted the Wnt pathway as a key promoter of axon regeneration following central nervous system (CNS) injuries. Emerging evidence also suggests that the temporal dysregulation of Wnt may drive cell death post-SCI. A major challenge in SCI treatment resides in developing therapeutics that can effectively target inflammation and facilitate glial scar repair. Before Wnt signalling is exploited for SCI therapy, further research is needed to clarify the implications of Wnt on neuroinflammation during chronic stages of injury. In this review, an attempt is made to dissect the impact of canonical and non-canonical Wnt pathways in relation to individual aspects of glial and fibrotic scar formation. Furthermore, it is also highlighted how modulating Wnt activity at chronic time points may aid in limiting lesion expansion and promoting axonal repair.
Collapse
Affiliation(s)
- Suchita Ganesan
- Department of Biomedical Sciences, Sri Ramachandra Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | - Arun Dharmarajan
- Department of Biomedical Sciences, Sri Ramachandra Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
- Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA, 6102, Australia
- Curtin Medical School, Curtin University, Perth, WA, Australia
- School of Human Sciences, The University of Western Australia, Nedlands, WA, Australia
- Sri Ramachandra Faculty of Clinical Research, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | - G Sudhir
- Department of Orthopedics and Spine Surgery, Sri Ramachandra Medical College and Research Institute, Sri Ramachandra Institute of Higher Education and Research, Chennai, India.
| | - Lakshmi R Perumalsamy
- Department of Biomedical Sciences, Sri Ramachandra Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research, Chennai, India.
| |
Collapse
|
13
|
Zeng X, Ropper AE, Aljuboori Z, Yu D, Teng TW, Kabatas S, Usuga E, Anderson JE, Teng YD. Concurrent Oncolysis and Neurolesion Repair by Dual Gene-Engineered hNSCs in an Experimental Model of Intraspinal Cord Glioblastoma. Cells 2024; 13:1522. [PMID: 39329707 PMCID: PMC11429792 DOI: 10.3390/cells13181522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 09/28/2024] Open
Abstract
Intramedullary spinal cord glioblastoma (ISCG) is lethal due to lack of effective treatment. We previously established a rat C6-ISCG model and the antitumor effect of F3.CD-TK, an hNSC line expressing CD and TK, via producing cytocidal 5FU and GCV-TP. However, the neurotherapeutic potential of this hNSC approach has remained uninvestigated. Here for the first time, cultured F3.CD-TK cells were found to have a markedly higher oncolytic effect, which was GJIC-dependent, and BDNF expression but less VEGF secretion than F3.CD. In Rowett athymic rats, F3.CD-TK (1.5 × 106 cells/10 µL × 2), injected near C6-ISCG (G55 seeding 7 days earlier: 10 K/each) and followed by q.d. (×5/each repeat; i.p.) of 5FC (500 mg/kg/5 mL/day) and GCV (25 mg/kg/1 mL/day), robustly mitigated cardiorespiratory, locomotor, and sensory deficits to improve neurofunction and overall survival compared to animals receiving either F3.CD or F3.CD-TK+F3.CD debris formula. The F3.CD-TK regimen exerted greater tumor penetration and neural inflammation/immune modulation, reshaped C6-ISCG topology to increase the tumor's surface area/volume ratio to spare/repair host axons (e.g., vGlut1+ neurites), and had higher post-prodrug donor self-clearance. The multimodal data and mechanistic leads from this proof-of-principle study suggest that the overall stronger anti-ISCG benefit of our hNSC-based GDEPT is derived from its concurrent oncolytic and neurotherapeutic effects.
Collapse
Affiliation(s)
- Xiang Zeng
- Department of Physical Medicine and Rehabilitation, Harvard Medical School and Spaulding Rehabilitation Hospital, Boston, MA 02129, USA
- Department of Neurosurgery, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
- Laboratory of SCI, Stem Cell, and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical School, Boston, MA 02129, USA
| | - Alexander E Ropper
- Department of Neurosurgery, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
- Laboratory of SCI, Stem Cell, and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical School, Boston, MA 02129, USA
| | - Zaid Aljuboori
- Department of Neurosurgery, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
- Laboratory of SCI, Stem Cell, and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical School, Boston, MA 02129, USA
| | - Dou Yu
- Department of Physical Medicine and Rehabilitation, Harvard Medical School and Spaulding Rehabilitation Hospital, Boston, MA 02129, USA
- Department of Neurosurgery, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
- Laboratory of SCI, Stem Cell, and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical School, Boston, MA 02129, USA
| | - Theodore W Teng
- Harvard College, Harvard University, Cambridge, MA 02138, USA
| | - Serdar Kabatas
- Department of Neurosurgery, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
- Laboratory of SCI, Stem Cell, and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical School, Boston, MA 02129, USA
| | - Esteban Usuga
- Department of Physical Medicine and Rehabilitation, Harvard Medical School and Spaulding Rehabilitation Hospital, Boston, MA 02129, USA
- Laboratory of SCI, Stem Cell, and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical School, Boston, MA 02129, USA
| | - Jamie E Anderson
- Department of Physical Medicine and Rehabilitation, Harvard Medical School and Spaulding Rehabilitation Hospital, Boston, MA 02129, USA
- Department of Neurosurgery, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
- Laboratory of SCI, Stem Cell, and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical School, Boston, MA 02129, USA
| | - Yang D Teng
- Department of Physical Medicine and Rehabilitation, Harvard Medical School and Spaulding Rehabilitation Hospital, Boston, MA 02129, USA
- Department of Neurosurgery, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
- Laboratory of SCI, Stem Cell, and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical School, Boston, MA 02129, USA
| |
Collapse
|
14
|
Anwar MM, Pérez-Martínez L, Pedraza-Alva G. Exploring the Significance of Microglial Phenotypes and Morphological Diversity in Neuroinflammation and Neurodegenerative Diseases: From Mechanisms to Potential Therapeutic Targets. Immunol Invest 2024; 53:891-946. [PMID: 38836373 DOI: 10.1080/08820139.2024.2358446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
Studying various microglial phenotypes and their functions in neurodegenerative diseases is crucial due to the intricate nature of their phenomics and their vital immunological role. Microglia undergo substantial phenomic changes, encompassing morphological, transcriptional, and functional aspects, resulting in distinct cell types with diverse structures, functions, properties, and implications. The traditional classification of microglia as ramified, M1 (proinflammatory), or M2 (anti-inflammatory) phenotypes is overly simplistic, failing to capture the wide range of recently identified microglial phenotypes in various brain regions affected by neurodegenerative diseases. Altered and activated microglial phenotypes deviating from the typical ramified structure are significant features of many neurodegenerative conditions. Understanding the precise role of each microglial phenotype is intricate and sometimes contradictory. This review specifically focuses on elucidating recent modifications in microglial phenotypes within neurodegenerative diseases. Recognizing the heterogeneity of microglial phenotypes in diseased states can unveil novel therapeutic strategies for targeting microglia in neurodegenerative diseases. Moreover, the exploration of the use of healthy isolated microglia to mitigate disease progression has provided an innovative perspective. In conclusion, this review discusses the dynamic landscape of mysterious microglial phenotypes, emphasizing the need for a nuanced understanding to pave the way for innovative therapeutic strategies for neurodegenerative diseases.
Collapse
Affiliation(s)
- Mai M Anwar
- Department of Biochemistry, National Organization for Drug Control and Research (NODCAR)/Egyptian Drug Authority (EDA), Cairo, Egypt
| | - Leonor Pérez-Martínez
- Neuroimmunobiology Laboratory, Department of Molecular Medicine and Bioprocesses, Institute of Biotechnology, National Autonomous University of Mexico, Cuernavaca, Morelos, Mexico
| | - Gustavo Pedraza-Alva
- Neuroimmunobiology Laboratory, Department of Molecular Medicine and Bioprocesses, Institute of Biotechnology, National Autonomous University of Mexico, Cuernavaca, Morelos, Mexico
| |
Collapse
|
15
|
Li D, Liu C, Wang H, Li Y, Wang Y, An S, Sun S. The Role of Neuromodulation and Potential Mechanism in Regulating Heterotopic Ossification. Neurochem Res 2024; 49:1628-1642. [PMID: 38416374 DOI: 10.1007/s11064-024-04118-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/17/2024] [Accepted: 01/28/2024] [Indexed: 02/29/2024]
Abstract
Heterotopic ossification (HO) is a pathological process characterized by the aberrant formation of bone in muscles and soft tissues. It is commonly triggered by traumatic brain injury, spinal cord injury, and burns. Despite a wide range of evidence underscoring the significance of neurogenic signals in proper bone remodeling, a clear understanding of HO induced by nerve injury remains rudimentary. Recent studies suggest that injury to the nervous system can activate various signaling pathways, such as TGF-β, leading to neurogenic HO through the release of neurotrophins. These pathophysiological changes lay a robust groundwork for the prevention and treatment of HO. In this review, we collected evidence to elucidate the mechanisms underlying the pathogenesis of HO related to nerve injury, aiming to enhance our understanding of how neurological repair processes can culminate in HO.
Collapse
Affiliation(s)
- Dengju Li
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong First Medical University, Jinan, Shandong, China
| | - Changxing Liu
- Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Haojue Wang
- Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Yunfeng Li
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yaqi Wang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Senbo An
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- Shandong First Medical University, Jinan, Shandong, China.
| | - Shui Sun
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- Shandong First Medical University, Jinan, Shandong, China.
- Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
16
|
Hosen S, Ikeda-Yorifuji I, Yamashita T. Asporin and CD109, expressed in the injured neonatal spinal cord, attenuate axonal re-growth in vitro. Neurosci Lett 2024; 833:137832. [PMID: 38796094 DOI: 10.1016/j.neulet.2024.137832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 05/17/2024] [Accepted: 05/22/2024] [Indexed: 05/28/2024]
Abstract
Axonal regeneration is restricted in adults and causes irreversible motor dysfunction following spinal cord injury (SCI). In contrast, neonates have prominent regenerative potential and can restore their neural function. Although the distinct cellular responses in neonates have been studied, how they contribute to neural recovery remains unclear. To assess whether the secreted molecules in neonatal SCI can enhance neural regeneration, we re-analyzed the previously performed single-nucleus RNA-seq (snRNA-seq) and focused on Asporin and Cd109, the highly expressed genes in the injured neonatal spinal cord. In the present study, we showed that both these molecules were expressed in the injured spinal cords of adults and neonates. We treated the cortical neurons with recombinant Asporin or CD109 to observe their direct effects on neurons in vitro. We demonstrated that these molecules enhance neurite outgrowth in neurons. However, these molecules did not enhance re-growth of severed axons. Our results suggest that Asporin and CD109 influence neurites at the lesion site, rather than promoting axon regeneration, to restore neural function in neonates after SCI.
Collapse
Affiliation(s)
- Sakura Hosen
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Iyo Ikeda-Yorifuji
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Osaka, Japan.
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Osaka, Japan; WPI Immunology Frontier Research Center, Osaka University, Suita, Japan; Department of Molecular Neuroscience, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan; Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita, Japan.
| |
Collapse
|
17
|
Stewart AN, Bosse-Joseph CC, Kumari R, Bailey WM, Park KA, Slone VK, Gensel JC. Non-resolving neuroinflammation regulates axon regeneration in chronic spinal cord injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.19.590106. [PMID: 38712123 PMCID: PMC11071389 DOI: 10.1101/2024.04.19.590106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Chronic spinal cord injury (SCI) lesions retain increased densities of microglia and macrophages. In acute SCI, macrophages induce growth cone collapse, facilitate axon retraction away from lesion boundaries, as well as play a key role in orchestrating the growth-inhibitory glial scar. Little is known about the role of sustained inflammation in chronic SCI, or whether chronic inflammation affects repair and regeneration. We performed transcriptional analysis using the Nanostring Neuropathology panel to characterize the resolution of inflammation into chronic SCI, to characterize the chronic SCI microenvironment, as well as to identify spinal cord responses to macrophage depletion and repopulation using the CSF1R inhibitor, PLX-5622. We determined the ability for macrophage depletion and repopulation to augment axon growth into chronic lesions both with and without regenerative stimulation using neuronal-specific PTEN knockout (PTEN-KO). PTEN-KO was delivered with spinal injections of retrogradely transported adeno associated viruses (AAVrg's). Both transcriptional analyses and immunohistochemistry revealed the ability for PLX-5622 to significantly deplete inflammation around and within chronic SCI lesions, with a return to pre-depleted inflammatory densities after treatment removal. Neuronal-specific transcripts were significantly elevated in mice after inflammatory repopulation, but no significant effects were observed with macrophage depletion alone. Axon densities significantly increased within the lesion after PLX-5622 treatment with a more consistent effect observed in mice with inflammatory repopulation. PTEN-KO did not further increase axon densities within the lesion beyond effects induced by PLX-5622. We identified that PLX-5622 increased axon densities within the lesion that are histologically identified as 5-HT+and CGRP+, both of which are not robustly transduced by AAVrg's. Our work identified that increased macrophage/microglia densities in the chronic SCI environment may be actively retained by homeostatic mechanisms likely affiliated with a sustained elevated expression of CSF1 and other chemokines. Finally, we identify a novel role of sustained inflammation as a prospective barrier to axon regeneration in chronic SCI.
Collapse
Affiliation(s)
- Andrew N. Stewart
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky 40536, USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky 40536, USA
- College of Medicine, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Christopher C. Bosse-Joseph
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky 40536, USA
- College of Medicine, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Reena Kumari
- Department of Physiology, University of Kentucky, Lexington, Kentucky 40536, USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky 40536, USA
- College of Medicine, University of Kentucky, Lexington, Kentucky 40536, USA
| | - William M. Bailey
- Department of Physiology, University of Kentucky, Lexington, Kentucky 40536, USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky 40536, USA
- College of Medicine, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Kennedy A. Park
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky 40536, USA
- College of Medicine, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Victoria K. Slone
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky 40536, USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky 40536, USA
- College of Medicine, University of Kentucky, Lexington, Kentucky 40536, USA
| | - John C. Gensel
- Department of Physiology, University of Kentucky, Lexington, Kentucky 40536, USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky 40536, USA
- College of Medicine, University of Kentucky, Lexington, Kentucky 40536, USA
| |
Collapse
|
18
|
Wang XX, Li GS, Wang KH, Hu XS, Hu Y. Positive effect of microvascular proliferation on functional recovery in experimental cervical spondylotic myelopathy. Front Neurosci 2024; 18:1254600. [PMID: 38510463 PMCID: PMC10951064 DOI: 10.3389/fnins.2024.1254600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 02/09/2024] [Indexed: 03/22/2024] Open
Abstract
Background and purpose Cervical Spondylotic Myelopathy (CSM), the most common cause of spinal cord dysfunction globally, is a degenerative disease that results in non-violent, gradual, and long-lasting compression of the cervical spinal cord. The objective of this study was to investigate whether microvascular proliferation could positively affect neural function recovery in experimental cervical spondylotic myelopathy (CSM). Methods A total of 60 male adult Sprague-Dawley (SD) were randomly divided into four groups: Control (CON), Compression (COM), Angiostasis (AS), and Angiogenesis (A G),with 15 rats in each group. Rats in the AS group received SU5416 to inhibit angiogenesis, while rats in the AG group received Deferoxamine (DFO) to promote angiogenesis. Motor and sensory functions were assessed using the Basso Beattie Bresnahan (BBB) scale and somatosensory evoked potential (SEP) examination. Neuropathological degeneration was evaluated by the number of neurons, Nissl bodies (NB), and the de-myelination of white matter detected by Hematoxylin & Eosin(HE), Toluidine Blue (TB), and Luxol Fast Blue (LFB) staining. Immunohistochemical (IHC) staining was used to observe the Neurovascular Unit (NVU). Results Rats in the CON group exhibited normal locomotor function with full BBB score, normal SEP latency and amplitude. Among the other three groups, the AG group had the highest BBB score and the shortest SEP latency, while the AS group had the lowest BBB score and the most prolonged SEP latency. The SEP amplitude showed an opposite performance to the latency. Compared to the COM and AS groups, the AG group demonstrated significant neuronal restoration in gray matter and axonal remyelination in white matter. DFO promoted microvascular proliferation, especially in gray matter, and improved the survival of neuroglial cells. In contrast, SU-5416 inhibited the viability of neuroglial cells by reducing micro vessels. Conclusion The microvascular status was closely related to NVU remodeling an-d functional recovery. Therefore, proliferation of micro vessels contributed to function -al recovery in experimental CSM, which may be associated with NVU remodeling.
Collapse
Affiliation(s)
- Xu-xiang Wang
- Department of Minimally Invasive Spine Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Guang-sheng Li
- Department of Minimally Invasive Spine Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Kang-heng Wang
- Department of Minimally Invasive Spine Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Xiao-song Hu
- Department of Minimally Invasive Spine Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Orthopedics Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Yong Hu
- Department of Minimally Invasive Spine Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Orthopedics Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
19
|
Yang Y, Liu Q, Deng S, Shao Q, Peng L, Ling Y, Huang Y, Zheng S, Jiang Q, Nie D, Chen J. Human umbilical cord derived mesenchymal stem cells overexpressing HO-1 attenuate neural injury and enhance functional recovery by inhibiting inflammation in stroke mice. CNS Neurosci Ther 2024; 30:e14412. [PMID: 37592866 PMCID: PMC10848045 DOI: 10.1111/cns.14412] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/27/2023] [Accepted: 08/01/2023] [Indexed: 08/19/2023] Open
Abstract
AIMS The current evidence demonstrates that mesenchymal stem cells (MSCs) hold therapeutic potential for ischemic stroke. However, it remains unclear how changes in the secretion of MSC cytokines following the overexpression of heme oxygenase-1 (HO-1) impact excessive inflammatory activation in a mouse ischemic stroke model. This study investigated this aspect and provided further insights. METHODS The middle cerebral artery occlusion (MCAO) mouse model was established, and subsequent injections of MSC, MSCHO-1 , or PBS solutions of equal volume were administered via the mice's tail vein. Histopathological analysis was conducted on Days 3 and 28 post-MCAO to observe morphological changes in brain slices. mRNA expression levels of various factors, including IL-1β, IL-6, IL-17, TNF-α, IL-1Ra, IL-4, IL-10, TGF-β, were quantified. The effects of MSCHO-1 treatment on neurons, microglia, and astrocytes were observed using immunofluorescence after transplantation. The polarization direction of macrophages/microglia was also detected using flow cytometry. RESULTS The results showed that the expression of anti-inflammatory factors in the MSCHO-1 group increased while that of pro-inflammatory factors decreased. Small animal fluorescence studies and immunofluorescence assays showed that the homing function of MSCsHO-1 was unaffected, leading to a substantial accumulation of MSCsHO-1 in the cerebral ischemic region within 24 h. Neurons were less damaged, activation and proliferation of microglia were reduced, and polarization of microglia to the M2 type increased after MSCHO-1 transplantation. Furthermore, after transplantation of MSCsHO-1 , the mortality of mice decreased, and motor function improved significantly. CONCLUSION The findings indicate that MSCs overexpressing HO-1 exhibited significant therapeutic effects against hyper-inflammatory injury after stroke in mice, ultimately promoting recovery after ischemic stroke.
Collapse
Affiliation(s)
- Yu Yang
- Department of NeurosurgeryAffiliated Hospital of Nantong University, Medical School of Nantong UniversityNantongChina
| | - Qianqian Liu
- Department of NeurosurgeryAffiliated Hospital of Nantong University, Medical School of Nantong UniversityNantongChina
| | - Song Deng
- Department of NeurosurgeryAffiliated Hospital of Nantong University, Medical School of Nantong UniversityNantongChina
| | - Qian Shao
- Research Center of Clinical MedicineAffiliated Hospital of Nantong UniversityNantongChina
| | - Long Peng
- Department of NeurosurgeryAffiliated Hospital of Nantong University, Medical School of Nantong UniversityNantongChina
| | - Yuejuan Ling
- Research Center of Clinical MedicineAffiliated Hospital of Nantong UniversityNantongChina
| | - Yue Huang
- Department of NeurosurgeryAffiliated Hospital of Nantong University, Medical School of Nantong UniversityNantongChina
| | - Siqi Zheng
- Department of NeurosurgeryAffiliated Hospital of Nantong University, Medical School of Nantong UniversityNantongChina
| | - Qiaoji Jiang
- Department of Neurosurgery, The Yancheng Clinical College of Xuzhou Medical UniversityThe First People's Hospital of YanchengYanchengChina
| | - Dekang Nie
- Department of NeurosurgeryAffiliated Hospital of Nantong University, Medical School of Nantong UniversityNantongChina
- Department of Neurosurgery, The Yancheng Clinical College of Xuzhou Medical UniversityThe First People's Hospital of YanchengYanchengChina
| | - Jian Chen
- Department of NeurosurgeryAffiliated Hospital of Nantong University, Medical School of Nantong UniversityNantongChina
| |
Collapse
|
20
|
Czyżewski W, Mazurek M, Sakwa L, Szymoniuk M, Pham J, Pasierb B, Litak J, Czyżewska E, Turek M, Piotrowski B, Torres K, Rola R. Astroglial Cells: Emerging Therapeutic Targets in the Management of Traumatic Brain Injury. Cells 2024; 13:148. [PMID: 38247839 PMCID: PMC10813911 DOI: 10.3390/cells13020148] [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: 11/29/2023] [Revised: 12/20/2023] [Accepted: 12/20/2023] [Indexed: 01/23/2024] Open
Abstract
Traumatic Brain Injury (TBI) represents a significant health concern, necessitating advanced therapeutic interventions. This detailed review explores the critical roles of astrocytes, key cellular constituents of the central nervous system (CNS), in both the pathophysiology and possible rehabilitation of TBI. Following injury, astrocytes exhibit reactive transformations, differentiating into pro-inflammatory (A1) and neuroprotective (A2) phenotypes. This paper elucidates the interactions of astrocytes with neurons, their role in neuroinflammation, and the potential for their therapeutic exploitation. Emphasized strategies encompass the utilization of endocannabinoid and calcium signaling pathways, hormone-based treatments like 17β-estradiol, biological therapies employing anti-HBGB1 monoclonal antibodies, gene therapy targeting Connexin 43, and the innovative technique of astrocyte transplantation as a means to repair damaged neural tissues.
Collapse
Affiliation(s)
- Wojciech Czyżewski
- Department of Didactics and Medical Simulation, Medical University of Lublin, 20-954 Lublin, Poland;
- Department of Neurosurgery and Pediatric Neurosurgery, Medical University of Lublin, 20-954 Lublin, Poland; (M.M.); (R.R.)
| | - Marek Mazurek
- Department of Neurosurgery and Pediatric Neurosurgery, Medical University of Lublin, 20-954 Lublin, Poland; (M.M.); (R.R.)
| | - Leon Sakwa
- Student Scientific Society, Kazimierz Pulaski University of Radom, 26-600 Radom, Poland;
| | - Michał Szymoniuk
- Student Scientific Association, Department of Neurosurgery and Pediatric Neurosurgery, Medical University of Lublin, 20-954 Lublin, Poland;
| | - Jennifer Pham
- Student Scientific Society, Medical University of Lublin, 20-954 Lublin, Poland; (J.P.); (M.T.)
| | - Barbara Pasierb
- Department of Dermatology, Radom Specialist Hospital, 26-600 Radom, Poland;
| | - Jakub Litak
- Department of Clinical Immunology, Medical University of Lublin, 20-954 Lublin, Poland;
| | - Ewa Czyżewska
- Department of Otolaryngology, Mazovian Specialist Hospital, 26-617 Radom, Poland;
| | - Michał Turek
- Student Scientific Society, Medical University of Lublin, 20-954 Lublin, Poland; (J.P.); (M.T.)
| | - Bartłomiej Piotrowski
- Institute of Automatic Control and Robotics, Warsaw University of Technology, 00-661 Warsaw, Poland;
| | - Kamil Torres
- Department of Didactics and Medical Simulation, Medical University of Lublin, 20-954 Lublin, Poland;
| | - Radosław Rola
- Department of Neurosurgery and Pediatric Neurosurgery, Medical University of Lublin, 20-954 Lublin, Poland; (M.M.); (R.R.)
| |
Collapse
|
21
|
Tolentino MJ, Tolentino AJ, Tolentino EM, Krishnan A, Genead MA. Sialic Acid Mimetic Microglial Sialic Acid-Binding Immunoglobulin-like Lectin Agonism: Potential to Restore Retinal Homeostasis and Regain Visual Function in Age-Related Macular Degeneration. Pharmaceuticals (Basel) 2023; 16:1735. [PMID: 38139861 PMCID: PMC10747662 DOI: 10.3390/ph16121735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/29/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
Age-related macular degeneration (AMD), a leading cause of visual loss and dysfunction worldwide, is a disease initiated by genetic polymorphisms that impair the negative regulation of complement. Proteomic investigation points to altered glycosylation and loss of Siglec-mediated glyco-immune checkpoint parainflammatory and inflammatory homeostasis as the main determinant for the vision impairing complications of macular degeneration. The effect of altered glycosylation on microglial maintained retinal para-inflammatory homeostasis and eventual recruitment and polarization of peripheral blood monocyte-derived macrophages (PBMDMs) into the retina can explain the phenotypic variability seen in this clinically heterogenous disease. Restoring glyco-immune checkpoint control with a sialic acid mimetic agonist targeting microglial/macrophage Siglecs to regain retinal para-inflammatory and inflammatory homeostasis is a promising therapeutic that could halt the progression of and improve visual function in all stages of macular degeneration.
Collapse
Affiliation(s)
- Michael J. Tolentino
- Department of Ophthalmology, University of Central Florida College of Medicine, Orlando, FL 32827, USA
- Department of Ophthalmology, Orlando College of Osteopathic Medicine, Orlando, FL 34787, USA
- Aviceda Therapeutics, Cambridge, MA 02142, USA; (A.K.); (M.A.G.)
| | - Andrew J. Tolentino
- Department of Biology, University of California Berkeley, Berkeley, CA 94720, USA;
| | | | - Anitha Krishnan
- Aviceda Therapeutics, Cambridge, MA 02142, USA; (A.K.); (M.A.G.)
| | | |
Collapse
|
22
|
Yilihamu EEY, Fan X, Yang Z, Feng S. A novel mouse model of central cord syndrome. Neural Regen Res 2023; 18:2751-2756. [PMID: 37449640 DOI: 10.4103/1673-5374.373718] [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: 07/18/2023] Open
Abstract
Patients with potential spinal stenosis are susceptible to central cord syndrome induced by blunt trauma. Suitable animal models are helpful for studying the pathogenesis and treatment of such injuries. In this study, we established a mouse model of acute blunt traumatic spinal cord injury by compressing the C6 spinal cord with 5 and 10 g/mm2 compression weights to simulate cervical central cord syndrome. Behavioral testing confirmed that this model exhibited the characteristics of central cord syndrome because motor function in the front paws was impaired, whereas basic motor and sensory functions of the lower extremities were retained. Hematoxylin-eosin staining showed that the diseased region of the spinal cord in this mouse model was restricted to the gray matter of the central cord, whereas the white matter was rarely affected. Magnetic resonance imaging showed a hypointense signal in the lesion after mild and severe injury. In addition, immunofluorescence staining showed that the degree of nerve tract injury in the spinal cord white matter was mild, and that there was a chronic inflammation reaction. These findings suggest that this mouse model of central cord syndrome can be used as a model for preclinical research, and that gray matter is most vulnerable to injury in central cord syndrome, leading to impaired motor function.
Collapse
Affiliation(s)
| | - Xiangchuang Fan
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Zimeng Yang
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong Province; Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Shiqing Feng
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| |
Collapse
|
23
|
St-Pierre MK, González Ibáñez F, Kroner A, Tremblay MÈ. Microglia/macrophages are ultrastructurally altered by their proximity to spinal cord injury in adult female mice. J Neuroinflammation 2023; 20:273. [PMID: 37990235 PMCID: PMC10664529 DOI: 10.1186/s12974-023-02953-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 11/08/2023] [Indexed: 11/23/2023] Open
Abstract
Traumatic spinal cord injury can cause immediate physical damage to the spinal cord and result in severe neurological deficits. The primary, mechanical tissue damage triggers a variety of secondary damage mechanisms at the injury site which significantly contribute to a larger lesion size and increased functional damage. Inflammatory mechanisms which directly involve both microglia (MG) and monocyte-derived macrophages (MDM) play important roles in the post-injury processes, including inflammation and debris clearing. In the current study, we investigated changes in the structure and function of MG/MDM in the injured spinal cord of adult female mice, 7 days after a thoracic contusion SCI. With the use of chip mapping scanning electron microscopy, which allows to image large samples at the nanoscale, we performed an ultrastructural comparison of MG/MDM located near the lesion vs adjacent regions to provide novel insights into the mechanisms at play post-injury. We found that MG/MDM located near the lesion had more mitochondria overall, including mitochondria with and without morphological alterations, and had a higher proportion of altered mitochondria. MG/MDM near the lesion also showed an increased number of phagosomes, including phagosomes containing myelin and partiallydigested materials. MG/MDM near the injury interacted differently with the spinal cord parenchyma, as shown by their reduced number of direct contacts with synaptic elements, axon terminals and dendritic spines. In this study, we characterized the ultrastructural changes of MG/MDM in response to spinal cord tissue damage in mice, uncovering changes in phagocytic activity, mitochondrial ultrastructure, and inter-cellular interactions within the spinal cord parenchyma.
Collapse
Affiliation(s)
- Marie-Kim St-Pierre
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
- Department of Molecular Medicine, Université Laval, Québec City, QC, Canada
- Division of Medical Sciences, University of Victoria, 3800 Finnerty Road, Victoria, BC, V8P 5C2, Canada
| | - Fernando González Ibáñez
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
- Department of Molecular Medicine, Université Laval, Québec City, QC, Canada
- Division of Medical Sciences, University of Victoria, 3800 Finnerty Road, Victoria, BC, V8P 5C2, Canada
| | - Antje Kroner
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, USA.
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, USA.
- Clement J. Zablocki Veterans Affairs Medical Center, 5000 W. National Ave, Milwaukee, WI, 53295, USA.
| | - Marie-Ève Tremblay
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada.
- Department of Molecular Medicine, Université Laval, Québec City, QC, Canada.
- Division of Medical Sciences, University of Victoria, 3800 Finnerty Road, Victoria, BC, V8P 5C2, Canada.
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada.
- Centre for Advanced Materials and Related Technology (CAMTEC) and Institute on Aging and Lifelong Health (IALH), University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
24
|
Denes V, Lukats A, Szarka G, Subicz R, Mester A, Kovacs-Valasek A, Geck P, Berta G, Herczeg R, Postyeni E, Gyenesei A, Gabriel R. Long-term Effects of the pituitary-adenylate cyclase-activating Polypeptide (PACAP38) in the Adult Mouse Retina: Microglial Activation and Induction of Neural Proliferation. Neurochem Res 2023; 48:3430-3446. [PMID: 37466802 PMCID: PMC10514177 DOI: 10.1007/s11064-023-03989-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 07/01/2023] [Accepted: 07/07/2023] [Indexed: 07/20/2023]
Abstract
The degenerative retinal disorders characterized by progressive cell death and exacerbating inflammation lead ultimately to blindness. The ubiquitous neuropeptide, PACAP38 is a promising therapeutic agent as its proliferative potential and suppressive effect on microglia might enable cell replacement and attenuate inflammation, respectively. Our previous finding that PACAP38 caused a marked increase of the amacrine cells in the adult (1-year-old) mouse retina, served as a rationale of the current study. We aimed to determine the proliferating elements and the inflammatory status of the PACAP38-treated retina. Three months old mice were intravitreally injected with 100 pmol PACAP38 at 3 months intervals (3X). Retinas of 1-year-old animals were dissected and effects on cell proliferation, and expression of inflammatory regulators were analyzed. Interestingly, both mitogenic and anti-mitogenic actions were detected after PACAP38-treatment. Further analysis of the mitogenic effect revealed that proliferating cells include microglia, endothelial cells, and neurons of the ganglion cell layer but not amacrine cells. Furthermore, PACAP38 stimulated retinal microglia to polarize dominantly into M2-phenotype but also might cause subsequent angiogenesis. According to our results, PACAP38 might dampen pro-inflammatory responses and help tissue repair by reprogramming microglia into an M2 phenotype, nonetheless, with angiogenesis as a warning side effect.
Collapse
Affiliation(s)
- Viktoria Denes
- Department of Neurobiology, University of Pécs, 6 Ifjúság str, Pécs, H-7624, Hungary.
| | - Akos Lukats
- Institute of Translational Medicine, Translational Retina Research Group, Semmelweis University, Budapest, Hungary
| | - Gergely Szarka
- Department of Neurobiology, University of Pécs, 6 Ifjúság str, Pécs, H-7624, Hungary
| | - Rovena Subicz
- Department of Neurobiology, University of Pécs, 6 Ifjúság str, Pécs, H-7624, Hungary
| | - Adrienn Mester
- Department of Neurobiology, University of Pécs, 6 Ifjúság str, Pécs, H-7624, Hungary
| | - Andrea Kovacs-Valasek
- Department of Neurobiology, University of Pécs, 6 Ifjúság str, Pécs, H-7624, Hungary
| | - Peter Geck
- Department of Medical Education, School of Medicine, Tufts University, 136 Harrison Ave, Boston, MA, 02111, USA
| | - Gergely Berta
- Department of Medical Biology and Central Electron Microscope Laboratory, Medical School, University of Pécs, Pécs, Hungary
| | - Robert Herczeg
- János Szentágothai Research Centre, Bioinformatics Research Group, University of Pécs, 20 Ifjúság str, Pécs, H-7624, Hungary
| | - Etelka Postyeni
- Department of Neurobiology, University of Pécs, 6 Ifjúság str, Pécs, H-7624, Hungary
| | - Attila Gyenesei
- János Szentágothai Research Centre, Bioinformatics Research Group, University of Pécs, 20 Ifjúság str, Pécs, H-7624, Hungary
| | - Robert Gabriel
- Department of Neurobiology, University of Pécs, 6 Ifjúság str, Pécs, H-7624, Hungary
| |
Collapse
|
25
|
Guha L, Kumar H. Drug Repurposing for Spinal Cord Injury: Progress Towards Therapeutic Intervention for Primary Factors and Secondary Complications. Pharmaceut Med 2023; 37:463-490. [PMID: 37698762 DOI: 10.1007/s40290-023-00499-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/10/2023] [Indexed: 09/13/2023]
Abstract
Spinal cord injury (SCI) encompasses a plethora of complex mechanisms like the involvement of major cell death pathways, neurodegeneration of spinal cord neurons, overexpression of glutaminergic transmission and inflammation cascade, along with different co-morbidities like neuropathic pain, urinary and sexual dysfunction, respiratory and cardiac failures, making it one of the leading causes of morbidity and mortality globally. Corticosteroids such as methylprednisolone and dexamethasone, and non-steroidal anti-inflammatory drugs such as naproxen, aspirin and ibuprofen are the first-line treatment options for SCI, inhibiting primary and secondary progression by preventing inflammation and action of reactive oxygen species. However, they are constrained by a short effective drug administration window and their pharmacological action being limited to symptomatic relief of the secondary effects related to spinal cord injury only. Although post-injury rehabilitation treatments may enable functional recovery, they take a long time to show results. Drug repurposing might be an innovative method for expanding therapy alternatives, utilising drugs that are already approved by various esteemed federal agencies throughout the world. Reutilising a drug molecule to treat SCI can eliminate the need for expensive and lengthy drug discovery processes and pave the way for new therapeutic approaches in SCI. This review summarises marketed drugs that could be repurposed based on their safety and efficacy data. We also discuss their mechanisms of action and provide a list of repurposed drugs under clinical trials for SCI therapy.
Collapse
Affiliation(s)
- Lahanya Guha
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Opposite Air Force Station, Palaj, P.O-382355, Gandhinagar, Gujarat, India
| | - Hemant Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Opposite Air Force Station, Palaj, P.O-382355, Gandhinagar, Gujarat, India.
| |
Collapse
|
26
|
Fang S, Wu Z, Guo Y, Zhu W, Wan C, Yuan N, Chen J, Hao W, Mo X, Guo X, Fan L, Li X, Chen J. Roles of microglia in adult hippocampal neurogenesis in depression and their therapeutics. Front Immunol 2023; 14:1193053. [PMID: 37881439 PMCID: PMC10597707 DOI: 10.3389/fimmu.2023.1193053] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 09/20/2023] [Indexed: 10/27/2023] Open
Abstract
Adult hippocampal neurogenesis generates functional neurons from neural progenitor cells in the hippocampal dentate gyrus (DG) to complement and repair neurons and neural circuits, thus benefiting the treatment of depression. Increasing evidence has shown that aberrant microglial activity can disrupt the appropriate formation and development of functional properties of neurogenesis, which will play a crucial role in the occurrence and development of depression. However, the mechanisms of the crosstalk between microglia and adult hippocampal neurogenesis in depression are not yet fully understood. Therefore, in this review, we first introduce recent discoveries regarding the roles of microglia and adult hippocampal neurogenesis in the etiology of depression. Then, we systematically discuss the possible mechanisms of how microglia regulate adult hippocampal neurogenesis in depression according to recent studies, which involve toll-like receptors, microglial polarization, fractalkine-C-X3-C motif chemokine receptor 1, hypothalamic-pituitary-adrenal axis, cytokines, brain-derived neurotrophic factor, and the microbiota-gut-brain axis, etc. In addition, we summarize the promising drugs that could improve the adult hippocampal neurogenesis by regulating the microglia. These findings will help us understand the complicated pathological mechanisms of depression and shed light on the development of new treatment strategies for this disease.
Collapse
Affiliation(s)
- Shaoyi Fang
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Zhibin Wu
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Yali Guo
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Wenjun Zhu
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Chunmiao Wan
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Naijun Yuan
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
- Shenzhen People’s Hospital, 2Clinical Medical College, Jinan University, Shenzhen, China
| | - Jianbei Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Wenzhi Hao
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Xiaowei Mo
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Xiaofang Guo
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Lili Fan
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Xiaojuan Li
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Jiaxu Chen
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
27
|
Sharma A, Feng L, Muresanu DF, Tian ZR, Lafuente JV, Buzoianu AD, Nozari A, Wiklund L, Sharma HS. Spinal cord injury induced exacerbation of Alzheimer's disease like pathophysiology is reduced by topical application of nanowired cerebrolysin with monoclonal antibodies to amyloid beta peptide, p-tau and tumor necrosis factor alpha. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 172:3-35. [PMID: 37833015 DOI: 10.1016/bs.irn.2023.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2023]
Abstract
Hallmark of Alzheimer's disease include amyloid beta peptide and phosphorylated tau deposition in brain that could be aggravated following traumatic of concussive head injury. However, amyloid beta peptide or p-tau in spinal cord following injury is not well known. In this investigation we measured amyloid beta peptide and p-tau together with tumor necrosis factor-alpha (TNF-α) in spinal cord and brain following 48 h after spinal cord injury in relation to the blood-spinal cord and blood-brain barrier, edema formation, blood flow changes and cell injury in perifocal regions of the spinal cord and brain areas. A focal spinal cord injury was inflicted over the right dorsal horn of the T10-11 segment (4 mm long and 2 mm deep) and amyloid beta peptide and p-tau was measured in perifocal rostral (T9) and caudal (T12) spinal cord segments as well as in the brain areas. Our observations showed a significant increase in amyloid beta peptide in the T9 and T12 segments as well as in remote areas of brain and spinal cord after 24 and 48 h injury. This is associated with breakdown of the blood-spinal cord (BSCB) and brain barriers (BBB), edema formation, reduction in blood flow and cell injury. After 48 h of spinal cord injury elevation of amyloid beta peptide, phosphorylated tau (p-tau) and tumor necrosis factor-alpha (TNF-α) was seen in T9 and T12 segments of spinal cord in cerebral cortex, hippocampus and brain stem regions associated with microglial activation as seen by upregulation of Iba1 and CD86. Repeated nanowired delivery of cerebrolysin topically over the traumatized segment repeatedly together with monoclonal antibodies (mAb) to amyloid beta peptide (AβP), p-tau and TNF-α significantly attenuated amyloid beta peptide, p-tau deposition and reduces Iba1, CD68 and TNF-α levels in the brain and spinal cord along with blockade of BBB and BSCB, reduction in blood flow, edema formation and cell injury. These observations are the first to show that spinal cord injury induces Alzheimer's disease like symptoms in the CNS, not reported earlier.
Collapse
Affiliation(s)
- Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Dept. of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| | - Lianyuan Feng
- Department of Neurology, Bethune International Peace Hospital, Zhongshan Road (West), Shijiazhuang, Hebei Province, P.R. China
| | - Dafin F Muresanu
- Dept. Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania; ''RoNeuro'' Institute for Neurological Research and Diagnostic, Mircea Eliade Street, Cluj-Napoca, Romania
| | - Z Ryan Tian
- Dept. Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - José Vicente Lafuente
- LaNCE, Dept. Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ala Nozari
- Department of Anesthesiology, Boston University, Albany str, Boston MA, USA
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Dept. of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Dept. of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden; LaNCE, Dept. Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| |
Collapse
|
28
|
Wang J, Zhao X, Zhou R, Wang M, Xiang W, You Z, Li M, Tang R, Zheng J, Li J, Zhu L, Gao J, Li H, Pang R, Zhang A. Gut microbiota and transcriptome dynamics in every-other-day fasting are associated with neuroprotection in rats with spinal cord injury. Front Microbiol 2023; 14:1206909. [PMID: 37577426 PMCID: PMC10417830 DOI: 10.3389/fmicb.2023.1206909] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 07/04/2023] [Indexed: 08/15/2023] Open
Abstract
Introduction Every-other-day fasting (EODF) is a classical intermittent fasting (IF) mode with neuroprotective effects that promotes motor function recovery after spinal cord injury (SCI) in rats. However, its dynamic effects on the gut microbiota and spinal cord transcriptome remain unknown. Methods In this study, 16S rRNA sequencing and RNA-seq analysis were used to investigate the effects of ad libitum (AL) and EODF dietary modes on the structural characteristics of rat gut microbiota in rats and the spinal cord transcriptome at various time points after SCI induction. Results Our results showed that both dietary modes affected the bacterial community composition in SCI rats, with EODF treatment inducing and suppressing dynamic changes in the abundances of potentially anti-inflammatory and pro-inflammatory bacteria. Furthermore, the differentially expressed genes (DEGs) enriched after EODF intervention in SCI rats were associated with various biological events, including immune inflammatory response, cell differentiation, protein modification, neural growth, and apoptosis. In particular, significant spatiotemporal differences were apparent in the DEGs associated with neuroprotection between the EODF and AL interventions. These DGEs were mainly focused on days 1, 3, and 7 after SCI. The relative abundance of certain genera was significantly correlated with DEGs associated with neuroprotective effects in the EODF-SCI group. Discussion Our results showed that EODF treatment may exert neuroprotective effects by modulating the transcriptome expression profile following SCI in rats. Furthermore, gut microbiota may be partially involved in mediating these effects.
Collapse
Affiliation(s)
- Junyu Wang
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaohua Zhao
- Department of Rehabilitation Medicine, General Hospital of Western Theater Command, Chengdu, China
- Department of Rehabilitation Medicine, The People’s Hospital of Tongliang District, Chongqing, China
| | - Ruihan Zhou
- Department of Rehabilitation Medicine, General Hospital of Western Theater Command, Chengdu, China
| | - Meiyu Wang
- Rehabilitation and Wellness Care Centre, Tian Fu College of Swufe, Chengdu, China
| | - Wu Xiang
- Department of Rehabilitation Medicine, General Hospital of Western Theater Command, Chengdu, China
| | - Zilong You
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Min Li
- Department of Rehabilitation Medicine, Shanghai Fourth People’s Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Ruiling Tang
- Department of Rehabilitation Medicine, General Hospital of Western Theater Command, Chengdu, China
| | - Jingqi Zheng
- Department of Rehabilitation Medicine, General Hospital of Western Theater Command, Chengdu, China
| | - Jiayu Li
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Li Zhu
- Department of Rehabilitation Medicine, Shanghai Fourth People’s Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Jiaxin Gao
- Department of Rehabilitation Medicine, Shanghai Fourth People’s Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Huaqiang Li
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rizhao Pang
- Department of Rehabilitation Medicine, The People’s Hospital of Tongliang District, Chongqing, China
| | - Anren Zhang
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
29
|
Hu X, Xu W, Ren Y, Wang Z, He X, Huang R, Ma B, Zhao J, Zhu R, Cheng L. Spinal cord injury: molecular mechanisms and therapeutic interventions. Signal Transduct Target Ther 2023; 8:245. [PMID: 37357239 DOI: 10.1038/s41392-023-01477-6] [Citation(s) in RCA: 119] [Impact Index Per Article: 59.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 03/22/2023] [Accepted: 05/07/2023] [Indexed: 06/27/2023] Open
Abstract
Spinal cord injury (SCI) remains a severe condition with an extremely high disability rate. The challenges of SCI repair include its complex pathological mechanisms and the difficulties of neural regeneration in the central nervous system. In the past few decades, researchers have attempted to completely elucidate the pathological mechanism of SCI and identify effective strategies to promote axon regeneration and neural circuit remodeling, but the results have not been ideal. Recently, new pathological mechanisms of SCI, especially the interactions between immune and neural cell responses, have been revealed by single-cell sequencing and spatial transcriptome analysis. With the development of bioactive materials and stem cells, more attention has been focused on forming intermediate neural networks to promote neural regeneration and neural circuit reconstruction than on promoting axonal regeneration in the corticospinal tract. Furthermore, technologies to control physical parameters such as electricity, magnetism and ultrasound have been constantly innovated and applied in neural cell fate regulation. Among these advanced novel strategies and technologies, stem cell therapy, biomaterial transplantation, and electromagnetic stimulation have entered into the stage of clinical trials, and some of them have already been applied in clinical treatment. In this review, we outline the overall epidemiology and pathophysiology of SCI, expound on the latest research progress related to neural regeneration and circuit reconstruction in detail, and propose future directions for SCI repair and clinical applications.
Collapse
Affiliation(s)
- Xiao Hu
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China
| | - Wei Xu
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China
| | - Yilong Ren
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China
| | - Zhaojie Wang
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China
| | - Xiaolie He
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China
| | - Runzhi Huang
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China
| | - Bei Ma
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China
| | - Jingwei Zhao
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China
| | - Rongrong Zhu
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China.
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China.
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China.
| | - Liming Cheng
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China.
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China.
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China.
| |
Collapse
|
30
|
Lee-Gosselin A, Jury-Garfe N, You Y, Dabin L, Soni D, Dutta S, Rochet JC, Kim J, Oblak AL, Lasagna-Reeves CA. TREM2-Deficient Microglia Attenuate Tau Spreading In Vivo. Cells 2023; 12:1597. [PMID: 37371067 PMCID: PMC10296847 DOI: 10.3390/cells12121597] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/05/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
The role of TREM2 in Alzheimer's disease (AD) is not fully understood. Previous studies investigating the effect of TREM2 deletion on tauopathy mouse models without the contribution of b-amyloid have focused only on tau overexpression models. Herein, we investigated the effects of TREM2 deficiency on tau spreading using a mouse model in which endogenous tau is seeded to produce AD-like tau features. We found that Trem2-/- mice exhibit attenuated tau pathology in multiple brain regions concomitant with a decreased microglial density. The neuroinflammatory profile in TREM2-deficient mice did not induce an activated inflammatory response to tau pathology. These findings suggest that reduced TREM2 signaling may alter the response of microglia to pathological tau aggregates, impairing their activation and decreasing their capacity to contribute to tau spreading. However, caution should be exercised when targeting TREM2 as a therapeutic entry point for AD until its involvement in tau aggregation and propagation is better understood.
Collapse
Affiliation(s)
- Audrey Lee-Gosselin
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.L.-G.); (N.J.-G.)
| | - Nur Jury-Garfe
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.L.-G.); (N.J.-G.)
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yanwen You
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.L.-G.); (N.J.-G.)
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Luke Dabin
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.L.-G.); (N.J.-G.)
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Disha Soni
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.L.-G.); (N.J.-G.)
- Department of Radiology & Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sayan Dutta
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA
| | - Jean-Christophe Rochet
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA
| | - Jungsu Kim
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.L.-G.); (N.J.-G.)
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Adrian L. Oblak
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.L.-G.); (N.J.-G.)
- Department of Radiology & Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Cristian A. Lasagna-Reeves
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.L.-G.); (N.J.-G.)
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
31
|
Kiss Bimbova K, Bacova M, Kisucka A, Gálik J, Ileninova M, Kuruc T, Magurova M, Lukacova N. Impact of Endurance Training on Regeneration of Axons, Glial Cells, and Inhibitory Neurons after Spinal Cord Injury: A Link between Functional Outcome and Regeneration Potential within the Lesion Site and in Adjacent Spinal Cord Tissue. Int J Mol Sci 2023; 24:ijms24108616. [PMID: 37239968 DOI: 10.3390/ijms24108616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/04/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Endurance training prior to spinal cord injury (SCI) has a beneficial effect on the activation of signaling pathways responsible for survival, neuroplasticity, and neuroregeneration. It is, however, unclear which training-induced cell populations are essential for the functional outcome after SCI. Adult Wistar rats were divided into four groups: control, six weeks of endurance training, Th9 compression (40 g/15 min), and pretraining + Th9 compression. The animals survived six weeks. Training alone increased the gene expression and protein level of immature CNP-ase oligodendrocytes (~16%) at Th10, and caused rearrangements in neurotrophic regulation of inhibitory GABA/glycinergic neurons at the Th10 and L2 levels, known to contain the interneurons with rhythmogenic potential. Training + SCI upregulated markers for immature and mature (CNP-ase, PLP1) oligodendrocytes by ~13% at the lesion site and caudally, and increased the number of GABA/glycinergic neurons in specific spinal cord regions. In the pretrained SCI group, the functional outcome of hindlimbs positively correlated with the protein levels of CNP-ase, PLP1, and neurofilaments (NF-l), but not with the outgrowing axons (Gap-43) at the lesion site and caudally. These results indicate that endurance training applied before SCI potentiates the repair in damaged spinal cord, and creates a suitable environment for neurological outcome.
Collapse
Affiliation(s)
- Katarina Kiss Bimbova
- Department of Neurodegeneration, Plasticity and Repair, Institute of Neurobiology, Biomedical Research Centre of Slovak Academy of Sciences, Soltesovej 4-6, 040 01 Kosice, Slovakia
| | - Maria Bacova
- Department of Neurodegeneration, Plasticity and Repair, Institute of Neurobiology, Biomedical Research Centre of Slovak Academy of Sciences, Soltesovej 4-6, 040 01 Kosice, Slovakia
| | - Alexandra Kisucka
- Department of Neurodegeneration, Plasticity and Repair, Institute of Neurobiology, Biomedical Research Centre of Slovak Academy of Sciences, Soltesovej 4-6, 040 01 Kosice, Slovakia
| | - Ján Gálik
- Department of Neurodegeneration, Plasticity and Repair, Institute of Neurobiology, Biomedical Research Centre of Slovak Academy of Sciences, Soltesovej 4-6, 040 01 Kosice, Slovakia
| | - Maria Ileninova
- Department of Neurodegeneration, Plasticity and Repair, Institute of Neurobiology, Biomedical Research Centre of Slovak Academy of Sciences, Soltesovej 4-6, 040 01 Kosice, Slovakia
| | - Tomas Kuruc
- Department of Neurodegeneration, Plasticity and Repair, Institute of Neurobiology, Biomedical Research Centre of Slovak Academy of Sciences, Soltesovej 4-6, 040 01 Kosice, Slovakia
| | - Martina Magurova
- Department of Neurodegeneration, Plasticity and Repair, Institute of Neurobiology, Biomedical Research Centre of Slovak Academy of Sciences, Soltesovej 4-6, 040 01 Kosice, Slovakia
| | - Nadezda Lukacova
- Department of Neurodegeneration, Plasticity and Repair, Institute of Neurobiology, Biomedical Research Centre of Slovak Academy of Sciences, Soltesovej 4-6, 040 01 Kosice, Slovakia
| |
Collapse
|
32
|
Akhmetzyanova ER, Zhuravleva MN, Timofeeva AV, Tazetdinova LG, Garanina EE, Rizvanov AA, Mukhamedshina YO. Severity- and Time-Dependent Activation of Microglia in Spinal Cord Injury. Int J Mol Sci 2023; 24:ijms24098294. [PMID: 37176001 PMCID: PMC10179339 DOI: 10.3390/ijms24098294] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/20/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
A spinal cord injury (SCI) initiates a number of cascades of biochemical reactions and intercellular interactions, the outcome of which determines the regenerative potential of the nervous tissue and opens up capacities for preserving its functions. The key elements of the above-mentioned processes are microglia. Many assumptions have been put forward, and the first evidence has been obtained, suggesting that, depending on the severity of SCI and the post-traumatic period, microglia behave differently. In this regard, we conducted a study to assess the microglia behavior in the model of mild, moderate and severe SCI in vitro for various post-traumatic periods. We reported for the first time that microglia make a significant contribution to both anti- and pro-inflammatory patterns for a prolonged period after severe SCI (60 dpi), while reduced severities of SCI do not lead to prolonged activation of microglia. The study also revealed the following trend: the greater the severity of the SCI, the lower the proliferative and phagocytic activity of microglia, which is true for all post-traumatic periods of SCI.
Collapse
Affiliation(s)
- Elvira Ruslanovna Akhmetzyanova
- OpenLab Gene and Cell Technology, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Margarita Nikolaevna Zhuravleva
- OpenLab Gene and Cell Technology, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Anna Viktorovna Timofeeva
- OpenLab Gene and Cell Technology, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Leisan Gazinurovna Tazetdinova
- Department of Morphology and General Pathology, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Ekaterina Evgenevna Garanina
- OpenLab Gene and Cell Technology, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Albert Anatolevich Rizvanov
- OpenLab Gene and Cell Technology, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Yana Olegovna Mukhamedshina
- OpenLab Gene and Cell Technology, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
- Department of Histology, Cytology, and Embryology, Kazan State Medical University, 420012 Kazan, Russia
| |
Collapse
|
33
|
Wang SM, Hsu JYC, Ko CY, Wu HE, Hsiao YW, Wang JM. Astrocytic Cebpd Regulates Pentraxin 3 Expression to Promote Fibrotic Scar Formation After Spinal Cord Injury. Mol Neurobiol 2023; 60:2200-2208. [PMID: 36633805 PMCID: PMC9984521 DOI: 10.1007/s12035-023-03207-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 12/29/2022] [Indexed: 01/13/2023]
Abstract
Astroglial-fibrotic scars resulted from spinal cord injury affect motor and sensory function, leading to paralysis. In particular, the fibrotic scar is a main barrier that disrupts neuronal regeneration after spinal cord injury. However, the association between astrocytes and fibrotic scar formation is not yet understood. We have previously demonstrated that the transcriptional factor Cebpd contributes to astrogliosis, which promotes glial scar formation after spinal cord injury. Herein, we show that fibrotic scar formation was decreased in the epicenter region in Cebpd-/- mice after contusive spinal cord injury and astrocytic Cebpd promoted fibroblast migration through secretion of Ptx3. Furthermore, the expression of Mmp3 was increased under recombinant protein Ptx3 treatment in fibroblasts by observing microarray data, resulting in fibroblast migration. In addition, regulation of Mmp3 occurs through the NFκB signaling pathway by using an irreversible inhibitor of IκBα phosphorylation in pretreated fibroblasts. Of note, we used the synthetic peptide RI37, which blocks fibroblast migration and decreases fibroblast Mmp3 expression in IL-1β-treated astrocyte conditioned media. Collectively, our data suggest that fibroblast migration can be affected by astrocytic Cebpd through the Ptx3/NFκB/Mmp3 axis pathway and that the RI37 peptide may act as a therapeutic medicine to inhibit fibrotic scar formation after spinal cord injury.
Collapse
Affiliation(s)
- Shao-Ming Wang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 404333, Taiwan. .,Neuroscience and Brain Disease Center, China Medical University, Taichung, Taiwan. .,Department of Neurology, China Medical University Hospital, Taichung, Taiwan.
| | - Jung-Yu C Hsu
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chiung-Yuan Ko
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,TMU Research Center of Neuroscience, Taipei Medical University, Taipei, Taiwan.,TMU Research Center of Cancer Translational Medicine, Taipei, Taiwan
| | - Hsiang-En Wu
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH/DHHS, Suite 3512, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Yu-Wei Hsiao
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Ju-Ming Wang
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan. .,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan. .,International Research Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan. .,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan. .,Institute of Bioinformatics and Biosignal Transduction, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 701, Taiwan.
| |
Collapse
|
34
|
Wang J, He W, Zhang J. A richer and more diverse future for microglia phenotypes. Heliyon 2023; 9:e14713. [PMID: 37025898 PMCID: PMC10070543 DOI: 10.1016/j.heliyon.2023.e14713] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 03/01/2023] [Accepted: 03/15/2023] [Indexed: 03/29/2023] Open
Abstract
Microglia are the only resident innate immune cells derived from the mesoderm in the nerve tissue. They play a role in the development and maturation of the central nervous system (CNS). Microglia mediate the repair of CNS injury and participate in endogenous immune response induced by various diseases by exerting neuroprotective or neurotoxic effects. Traditionally, microglia are considered to be in a resting state, the M0 type, under physiological conditions. In this state, they perform immune surveillance by constantly monitoring pathological responses in the CNS. In the pathological state, microglia undergo a series of morphological and functional changes from the M0 state and eventually polarize into classically activated microglia (M1) and alternatively activated microglia (M2). M1 microglia release inflammatory factors and toxic substances to inhibit pathogens, while M2 microglia exert neuroprotective effects by promoting nerve repair and regeneration. However, in recent years, the view regarding M1/M2 polarization of microglia has gradually changed. According to some researchers, the phenomenon of microglia polarization is not yet confirmed. The M1/M2 polarization term is used for a simplified description of its phenotype and function. Other researchers believe that the microglia polarization process is rich and diverse, and consequently, the classification method of M1/M2 has limitations. This conflict hinders the academic community from establishing more meaningful microglia polarization pathways and terms, and therefore, a careful revision of the concept of microglia polarization is required. The present article briefly reviews the current consensus and controversy regarding microglial polarization typing to provide supporting materials for a more objective understanding of the functional phenotype of microglia.
Collapse
|
35
|
Lawrence JM, Schardien K, Wigdahl B, Nonnemacher MR. Roles of neuropathology-associated reactive astrocytes: a systematic review. Acta Neuropathol Commun 2023; 11:42. [PMID: 36915214 PMCID: PMC10009953 DOI: 10.1186/s40478-023-01526-9] [Citation(s) in RCA: 99] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 02/08/2023] [Indexed: 03/16/2023] Open
Abstract
In the contexts of aging, injury, or neuroinflammation, activated microglia signaling with TNF-α, IL-1α, and C1q induces a neurotoxic astrocytic phenotype, classified as A1, A1-like, or neuroinflammatory reactive astrocytes. In contrast to typical astrocytes, which promote neuronal survival, support synapses, and maintain blood-brain barrier integrity, these reactive astrocytes downregulate supportive functions and begin to secrete neurotoxic factors, complement components like C3, and chemokines like CXCL10, which may facilitate recruitment of immune cells across the BBB into the CNS. The proportion of pro-inflammatory reactive astrocytes increases with age through associated microglia activation, and these pro-inflammatory reactive astrocytes are particularly abundant in neurodegenerative disorders. As the identification of astrocyte phenotypes progress, their molecular and cellular effects are characterized in a growing array of neuropathologies.
Collapse
Affiliation(s)
- Jill M Lawrence
- Molecular and Cell Biology and Genetics Graduate Program, Drexel University College of Medicine, Philadelphia, PA, USA
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
- Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Kayla Schardien
- Molecular and Cell Biology and Genetics Graduate Program, Drexel University College of Medicine, Philadelphia, PA, USA
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Brian Wigdahl
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
- Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Michael R Nonnemacher
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA.
- Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA.
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
36
|
Sex Differences in the Expression of Neuroimmune Molecules in the Spinal Cord of a Mouse Model of Antiretroviral-Induced Neuropathic Pain. Biomedicines 2023; 11:biomedicines11030875. [PMID: 36979854 PMCID: PMC10045154 DOI: 10.3390/biomedicines11030875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 03/07/2023] [Accepted: 03/11/2023] [Indexed: 03/16/2023] Open
Abstract
Nucleoside reverse transcriptase inhibitors (NRTIs), drugs used to treat HIV infection, can cause neuropathic pain (NP) and neuroinflammation. An NRTI, 2′-3′-dideoxycytidine (ddC), was reported to induce mechanical allodynia and increase proinflammatory cytokines in the brains of female mice. In some models of NP, microglia activation is important for NP pathophysiology in male mice, while T cells are important in female mice. Age-matched female and male mice (BALB/c strain) treated intraperitoneally once daily with ddC for 5 days developed mechanical allodynia. Treatment with ddC increased Cd11b, H2-Aa, Cd3e, Mapk1, Il1b, Tnf, and Il10 mRNA levels in the spinal cords of female, but not male, mice, whereas there was no alteration found in Gfap and Mapk14 transcripts in both sexes on day 7 after ddC administration. The protein expression of CD11b and phospho-p38 MAPK was significantly increased in the spinal cords of ddC-treated female, but not male, mice, whereas Iba1 protein was elevated in ddC-treated male mice. There was no change in GFAP, CD3e, and phospho-p44/42 MAPK protein levels in both sexes. Thus, changes in neuroimmune cells and molecules in the spinal cords during ddC-induced neuroinflammation were sex-dependent, with female mice being more prone to neuroimmune changes than male mice.
Collapse
|
37
|
Implications of microglial heterogeneity in spinal cord injury progression and therapy. Exp Neurol 2023; 359:114239. [PMID: 36216123 DOI: 10.1016/j.expneurol.2022.114239] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 09/21/2022] [Accepted: 10/03/2022] [Indexed: 11/07/2022]
Abstract
Microglia are widely distributed in the central nervous system (CNS), where they aid in the maintenance of neuronal function and perform key auxiliary roles in phagocytosis, neural repair, immunological control, and nutrition delivery. Microglia in the undamaged spinal cord is in a stable state and serve as immune monitors. In the event of spinal cord injury (SCI), severe changes in the microenvironment and glial scar formation lead to axonal regeneration failure. Microglia participates in a series of pathophysiological processes and behave both positive and negative consequences during this period. A deep understanding of the characteristics and functions of microglia can better identify therapeutic targets for SCI. Technological innovations such as single-cell RNA sequencing (Sc-RNAseq) have led to new advances in the study of microglia heterogeneity throughout the lifespan. Here,We review the updated studies searching for heterogeneity of microglia from the developmental and pathological state, survey the activity and function of microglia in SCI and explore the recent therapeutic strategies targeting microglia in the CNS injury.
Collapse
|
38
|
Xia Y, Yang R, Wang H, Hou Y, Li Y, Zhu J, Xu F, Fu C. Biomaterials delivery strategies to repair spinal cord injury by modulating macrophage phenotypes. J Tissue Eng 2022; 13:20417314221143059. [PMID: 36600997 PMCID: PMC9806413 DOI: 10.1177/20417314221143059] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 11/17/2022] [Indexed: 12/28/2022] Open
Abstract
Spinal cord injury (SCI) causes tremendous harm to a patient's physical, mental, and financial health. Moreover, recovery of SCI is affected by many factors, inflammation is one of the most important as it engulfs necrotic tissue and cells during the early stages of injury. However, excessive inflammation is not conducive to damage repair. Macrophages are classified into either blood-derived macrophages or resident microglia based on their origin, their effects on SCI being two-sided. Microglia first activate and recruit blood-derived macrophages at the site of injury-blood-borne macrophages being divided into pro-inflammatory M1 phenotypes and anti-inflammatory M2 phenotypes. Among them, M1 macrophages secrete inflammatory factors such as interleukin-β (IL-β), tumor necrosis factor-α (TNF-α), IL-6, and interferon-γ (IFN-γ) at the injury site, which aggravates SCIs. M2 macrophages secrete IL-4, IL-10, IL-13, and neurotrophic factors to inhibit the inflammatory response and inhibit neuronal apoptosis. Consequently, modulating phenotypic differentiation of macrophages appears to be a meaningful therapeutic target for the treatment of SCI. Biomaterials are widely used in regenerative medicine and tissue engineering due to their targeting and bio-histocompatibility. In this review, we describe the effects of biomaterials applied to modulate macrophage phenotypes on SCI recovery and provide an outlook.
Collapse
Affiliation(s)
- Yuanliang Xia
- Department of Spine Surgery, The First
Hospital of Jilin University, Changchun, PR China
| | - Ruohan Yang
- Cancer Center, The First Hospital of
Jilin University, Changchun, PR China
| | - Hengyi Wang
- Department of Spine Surgery, The First
Hospital of Jilin University, Changchun, PR China
| | - Yulin Hou
- Depattment of Cardiology, Guangyuan
Central Hospital, Guangyuan, PR China
| | - Yuehong Li
- Department of Spine Surgery, The First
Hospital of Jilin University, Changchun, PR China
| | - Jianshu Zhu
- Department of Spine Surgery, The First
Hospital of Jilin University, Changchun, PR China
| | - Feng Xu
- Department of Spine Surgery, The First
Hospital of Jilin University, Changchun, PR China
| | - Changfeng Fu
- Department of Spine Surgery, The First
Hospital of Jilin University, Changchun, PR China,Changfeng Fu, Department of Spine Surgery,
The First Hospital of Jilin University, 1 Xinmin Street, Changchun 130021, PR
China.
| |
Collapse
|
39
|
Wang J, Zhang G, Yang Y, Zhang X, Shi K, Zhang X, Yan T, Jia Y. Schisandra chinensis Lignans Exert Antidepressant Effects by Promoting BV2 Microglia Polarization toward the M2 Phenotype through the Activation of the Cannabinoid Receptor Type-2-Signal Transducer and Activator of Transcription 6 Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:14157-14169. [PMID: 36349542 DOI: 10.1021/acs.jafc.2c04565] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Based on the current results, they showed that Schisandra chinensis lignans (SCL) ameliorated depressive-like behaviors in chronic unpredictable mild stress (CUMS) mice, alleviated neuroinflammation, and improved neuronal injury. This study aimed to explore whether SCL exerted antidepressant effects through inhibiting neuroinflammation, in turn improving neuronal injury. In vitro studies revealed that SCL blocked lipopolysaccharide-increased BV2 microglial M1 but promoted the M2 phenotype. The BV2-N2a interaction model suggested that increasing the M2 phenotype of BV2 played neuroprotective effects. The current studies demonstrated that SCL up-regulated the expression of CUMS- and LPS-decreased cannabinoid receptor type-2 (CB2R) mRNA. In vitro studies showed that the transfection of BV2 with siCrn2 blocked the SCL-increased M2 phenotype via the inactivating signal transducer and activator of transcription 6 (STAT6) pathway, further decreasing the viability of N2a cells. Finally, the possible pharmacodynamic compounds, γ-schisandrin and schisantherin A, were indicated by AutoDuck analysis. Overall, our study showed that SCL promoted microglia polarization toward the M2 phenotype, in turn exerting neuroprotective effects by activating CB2R-STAT6 signaling further to play antidepressant roles.
Collapse
Affiliation(s)
- Jinyu Wang
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, P.R. China
| | - Guanglin Zhang
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, P.R. China
| | - Yunfang Yang
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, P.R. China
| | - Xiaoying Zhang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Kaifang Shi
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Xiaozhuo Zhang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, China
| | - Tingxu Yan
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, P.R. China
| | - Ying Jia
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, P.R. China
| |
Collapse
|
40
|
Zhang Y, Wang X, Jiang C, Chen Z, Ni S, Fan H, Wang Z, Tian F, An J, Yang H, Hao D. Rho Kinase Inhibitor Y27632 Improves Recovery After Spinal Cord Injury by Shifting Astrocyte Phenotype and Morphology via the ROCK/NF-κB/C3 Pathway. Neurochem Res 2022; 47:3733-3744. [DOI: 10.1007/s11064-022-03756-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 11/28/2022]
Abstract
AbstractSpinal cord injury (SCI) usually results in loss or reduction in motor and sensory functions. Despite extensive research, no available therapy can restore the lost functions after SCI. Reactive astrocytes play a pivotal role in SCI. Rho kinase inhibitors have also been shown to promote functional recovery of SCI. However, the role of Rho kinase inhibitors in reactive astrocytic phenotype switch within SCI remains largely unexplored. In this study, astrocytes were treated with proinflammatory cytokines and/or the Rho kinase inhibitor Y27632. Concomitantly the phenotype and morphology of astrocytes were examined. Meanwhile, the SCI model of SD rats was established, and nerve functions were evaluated following treatment with Y27632. Subsequently, the number of A1 astrocytes in the injured area was observed and analyzed. Eventually, the expression levels of nuclear factor kappa B (NF-κB), C3, and S100A10 were measured. The present study showed that the Rho kinase inhibitor Y27632 improved functional recovery of SCI and elevated the proliferation and migration abilities of the astrocytes. In addition, Y27632 treatment initiated the switch of astrocytes morphology from a flattened shape to a process-bearing shape and transformed the reactive astrocytes A1 phenotype to an A2 phenotype. More importantly, further investigation suggested that Y27632 was actively involved in promoting the functional recovery of SCI in rats by inhabiting the ROCK/NF-κB/C3 signaling pathway. Together, Rho kinase inhibitor Y27632 effectively promotes the functional recovery of SCI by shifting astrocyte phenotype and morphology. Furthermore, the pro-regeneration event is strongly associated with the ROCK/NF-κB/C3 signal pathway.
Collapse
|
41
|
Zhang Q, Liu M, Nong H, Zhang Y, Bai Y, Liu P, Zong S, Zeng G. Total flavonoids of hawthorn leaves protect spinal motor neurons via promotion of autophagy after spinal cord injury. Front Pharmacol 2022; 13:925568. [PMID: 36071834 PMCID: PMC9441667 DOI: 10.3389/fphar.2022.925568] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 07/29/2022] [Indexed: 12/03/2022] Open
Abstract
The death of spinal motor neurons (SMNs) after spinal cord injury (SCI) is a crucial cause, contributing to a permanent neurological deficit. Total flavonoids of hawthorn leaves (TFHL) have been confirmed to have potentially therapeutic for SCI. Nonetheless, the roles and mechanisms of TFHL in recovering neuromotor function and regenerating axons of SMNs have not been fully elucidated. In this study, TFHL was applied to treat rats with SCI and injured SMNs for 7 days. In vivo experiment, rats with SCI were evaluated by a BBB (Basso-Beattie-Bresnahan) score to assess their motor functional recovery. The morphology, microstructure, apoptosis, Nissl bodies, and autophagy of SMNs in spinal cord tissue were detected by Hematoxylin-eosin (HE) staining, transmission electron microscopy, TUNEL staining, Nissl staining, and immunohistochemistry respectively. In vitro experiment, the co-culture model of SMNs and astrocytes was constructed to simulate the internal environment around SMNs in the spinal cord tissue. The cell morphology, microstructure, axonal regeneration, and autophagy were observed via optical microscope, transmission electron microscopy, and immunofluorescence. The content of neurotrophic factors in the cell culture medium of the co-culture model was detected by ELISA. Moreover, the expression of axon-related and autophagy-related proteins in the spinal cord tissue and SMNs was measured by Western Blot. We demonstrated that TFHL improved the neuromotor function recovery in rats after SCI. We then found that TFHL significantly promoted injured spinal cord tissue repair, reduced apoptosis, and improved the functional status of neurons in spinal cord tissue in vivo. Meanwhile, the cell morphology, microstructure, and axonal regeneration of damaged SMNs also obviously were improved, and the secretion of neurotrophic factors was facilitated after treatment with TFHL in vitro. Further, we revealed that TFHL promoted autophagy and related protein expression in vivo and vitro. Taken together, our study suggested that TFHL might facilitate autophagy and have neuroprotective properties in SMNs to enhance the recovery of neuromotor function of rats with SCI.
Collapse
Affiliation(s)
- Qiong Zhang
- School of Public Health of Guangxi Medical University, Nanning, China
| | - Mingfu Liu
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Haibin Nong
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yanan Zhang
- Collaborative Innovation Center of Guangxi Biological Medicine, Guangxi Medical University, Nanning, China
| | - Yiguang Bai
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Pan Liu
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shaohui Zong
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Research Centre for Regenerative Medicine and Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China
- *Correspondence: Shaohui Zong, ; Gaofeng Zeng,
| | - Gaofeng Zeng
- School of Public Health of Guangxi Medical University, Nanning, China
- *Correspondence: Shaohui Zong, ; Gaofeng Zeng,
| |
Collapse
|
42
|
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
|
43
|
Xue MT, Sheng WJ, Song X, Shi YJ, Geng ZJ, Shen L, Wang R, Lü HZ, Hu JG. Atractylenolide III ameliorates spinal cord injury in rats by modulating microglial/macrophage polarization. CNS Neurosci Ther 2022; 28:1059-1071. [PMID: 35403332 PMCID: PMC9160450 DOI: 10.1111/cns.13839] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 01/04/2023] Open
Abstract
Background Inflammatory reactions induced by spinal cord injury (SCI) are essential for recovery after SCI. Atractylenolide III (ATL‐III) is a natural monomeric herbal bioactive compound that is mainly derived in Atractylodes macrocephala Koidz and has anti‐inflammatory and neuroprotective effects. Objective Here, we speculated that ATL‐III may ameliorate SCI by modulating microglial/macrophage polarization. In the present research, we focused on investigating the role of ATL‐III on SCI in rats and explored the potential mechanism. Methods The protective and anti‐inflammatory effects of ATL‐III on neuronal cells were examined in a rat SCI model and lipopolysaccharide (LPS)‐stimulated BV2 microglial line. The spinal cord lesion area, myelin integrity, and surviving neurons were assessed by specific staining. Locomotor function was evaluated by the Basso, Beattie, and Bresnahan (BBB) scale, grid walk test, and footprint test. The activation and polarization of microglia/macrophages were assessed by immunohistofluorescence and flow cytometry. The expression of corresponding inflammatory factors from M1/M2 and the activation of relevant signaling pathways were assessed by Western blotting. Results ATL‐III effectively improved histological and functional recovery in SCI rats. Furthermore, ATL‐III promoted the transformation of M1 into M2 and attenuated the activation of microglia/macrophages, further suppressing the expression of corresponding inflammatory mediators. This effect may be partly mediated by inhibition of neuroinflammation through the NF‐κB, JNK MAPK, p38 MAPK, and Akt pathways. Conclusion This study reveals a novel effect of ATL‐III in the regulation of microglial/macrophage polarization and provides initial evidence that ATL‐III has potential therapeutic benefits in SCI rats.
Collapse
Affiliation(s)
- Meng-Tong Xue
- Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, P.R. China.,Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, P.R. China
| | - Wen-Jie Sheng
- Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, P.R. China.,Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, P.R. China
| | - Xue Song
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, P.R. China
| | - Yu-Jiao Shi
- Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, P.R. China.,Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, P.R. China
| | - Zhi-Jun Geng
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, P.R. China
| | - Lin Shen
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, P.R. China
| | - Rui Wang
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, P.R. China
| | - He-Zuo Lü
- Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, P.R. China.,Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, P.R. China.,Department of Immunology, Bengbu Medical College, Bengbu, P.R. China
| | - Jian-Guo Hu
- Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, P.R. China.,Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, P.R. China
| |
Collapse
|
44
|
He X, Li Y, Deng B, Lin A, Zhang G, Ma M, Wang Y, Yang Y, Kang X. The PI3K/AKT signalling pathway in inflammation, cell death and glial scar formation after traumatic spinal cord injury: Mechanisms and therapeutic opportunities. Cell Prolif 2022; 55:e13275. [PMID: 35754255 PMCID: PMC9436900 DOI: 10.1111/cpr.13275] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 04/17/2022] [Accepted: 05/24/2022] [Indexed: 02/06/2023] Open
Abstract
Objects Traumatic spinal cord injury (TSCI) causes neurological dysfunction below the injured segment of the spinal cord, which significantly impacts the quality of life in affected patients. The phosphoinositide 3kinase/serine‐threonine kinase (PI3K/AKT) signaling pathway offers a potential therapeutic target for the inhibition of secondary TSCI. This review summarizes updates concerning the role of the PI3K/AKT pathway in TSCI. Materials and Methods By searching articles related to the TSCI field and the PI3K/AKT signaling pathway, we summarized the mechanisms of secondary TSCI and the PI3K/AKT signaling pathway; we also discuss current and potential future treatment methods for TSCI based on the PI3K/AKT signaling pathway. Results Early apoptosis and autophagy after TSCI protect the body against injury; a prolonged inflammatory response leads to the accumulation of pro‐inflammatory factors and excessive apoptosis, as well as excessive autophagy in the surrounding normal nerve cells, thus aggravating TSCI in the subacute stage of secondary injury. Initial glial scar formation in the subacute phase is a protective mechanism for TSCI, which limits the spread of damage and inflammation. However, mature scar tissue in the chronic phase hinders axon regeneration and prevents the recovery of nerve function. Activation of PI3K/AKT signaling pathway can inhibit the inflammatory response and apoptosis in the subacute phase after secondary TSCI; inhibiting this pathway in the chronic phase can reduce the formation of glial scar. Conclusion The PI3K/AKT signaling pathway has an important role in the recovery of spinal cord function after secondary injury. Inducing the activation of PI3K/AKT signaling pathway in the subacute phase of secondary injury and inhibiting this pathway in the chronic phase may be one of the potential strategies for the treatment of TSCI.
Collapse
Affiliation(s)
- Xuegang He
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, China.,The Second Clinical Medical College, Lanzhou University, Lanzhou, China.,The International Cooperation Base of Gansu Province for the Pain Research in Spinal Disorders, Lanzhou, China
| | - Ying Li
- Medical School of Yan'an University, Yan'an University, Yan'an, China
| | - Bo Deng
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, China.,The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Aixin Lin
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, China.,The Second Clinical Medical College, Lanzhou University, Lanzhou, China.,The International Cooperation Base of Gansu Province for the Pain Research in Spinal Disorders, Lanzhou, China
| | - Guangzhi Zhang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, China.,The Second Clinical Medical College, Lanzhou University, Lanzhou, China.,The International Cooperation Base of Gansu Province for the Pain Research in Spinal Disorders, Lanzhou, China
| | - Miao Ma
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, China.,The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Yonggang Wang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, China.,The Second Clinical Medical College, Lanzhou University, Lanzhou, China.,The International Cooperation Base of Gansu Province for the Pain Research in Spinal Disorders, Lanzhou, China
| | - Yong Yang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, China.,The International Cooperation Base of Gansu Province for the Pain Research in Spinal Disorders, Lanzhou, China
| | - Xuewen Kang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, China.,The Second Clinical Medical College, Lanzhou University, Lanzhou, China.,The International Cooperation Base of Gansu Province for the Pain Research in Spinal Disorders, Lanzhou, China
| |
Collapse
|
45
|
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
|
46
|
Scott EP, Breyak E, Nishinakamura R, Nakagawa Y. The zinc finger transcription factor Sall1 is required for the early developmental transition of microglia in mouse embryos. Glia 2022; 70:1720-1733. [PMID: 35567352 PMCID: PMC9276639 DOI: 10.1002/glia.24192] [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: 02/13/2022] [Revised: 05/02/2022] [Accepted: 05/03/2022] [Indexed: 11/06/2022]
Abstract
Microglia play many critical roles in neural development. Recent single-cell RNA-sequencing studies have found diversity of microglia both across different stages and within the same stage in the developing brain. However, how such diversity is controlled during development is poorly understood. In this study, we first found the expression of the macrophage mannose receptor CD206 in early-stage embryonic microglia on mouse brain sections. This expression showed a sharp decline between E12.5 and E13.5 across the central nervous system. We next tested the roles of the microglia-expressed zinc finger transcription factor SALL1 in this early transition of gene expression. By deleting Sall1 specifically in microglia, we found that many microglia continued to express CD206 when it is normally downregulated. In addition, the mutant microglia continued to show less ramified morphology in comparison with controls even into postnatal stages. Thus, SALL1 is required for early microglia to transition into a more mature status during development.
Collapse
Affiliation(s)
- Earl Parker Scott
- Department of Neuroscience, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Emma Breyak
- Department of Neuroscience, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Ryuichi Nishinakamura
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Yasushi Nakagawa
- Department of Neuroscience, University of Minnesota Medical School, Minneapolis, Minnesota, USA.,Developmental Biology Center, Masonic Institute for the Developing Brain, Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
47
|
Tian J, Yao H, Liu Y, Wang X, Wu J, Zhu Y, Yang C. Extracellular vesicles from bone marrow stromal cells reduce the impact of stroke on glial cell activation and blood brain-barrier permeability via a putative miR-124/PRX1 signaling pathway. Eur J Neurosci 2022; 56:3786-3805. [PMID: 35441400 DOI: 10.1111/ejn.15669] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 03/22/2022] [Accepted: 03/27/2022] [Indexed: 11/30/2022]
Abstract
Ischemic stroke (IS) is a cerebrovascular disease caused by cerebral infarction and cerebral artery occlusion. In this study we proposed that EVs from bone marrow stromal cells (BMSCs) could reduce the impact of stroke by reducing the resultant glial cell activation and blood-brain barrier (BBB) leak. We furthermore investigated some of the signaling mechanisms. The transient middle cerebral artery occlusion (t-MCAO) mouse model was established. The behavioral deficits and neuronal damage were verified using Bederson's scale and the 28-point neurological score. The area of cerebral infarction was detected. The expressions of astrocytes/microglia markers and BBB permeability were evaluated by 2, 3,5-triphenyltetrazolium chloride (TTC) staining. The internalization of EVs by astrocytes/microglia in the peripheral area was detected by fluorescence labeling. The expressions of astrocyte/microglia markers were measured by RT-qPCR. Levels of TNF-α and IL-1β in microglia were detected by ELISA. BBB permeability was evaluated. The downstream target genes and pathway of miR-124 were analyzed. Microglia/astrocytes were treated by oxygen-glucose deprivation reoxygenation (OGD/R). OGD/R microglia/astrocyte conditioned medium was used to culture bEnd.3 cells. The transendothelial electric resistance (TEER) of bEnd.3 cells was measured and BBB permeability was characterized. Our results suggested that EVs from BMSCs can indeed reduce the extent of stroke-mediated damage, and evidenced that these effects are mediated via expression of the non-coding RNA, miR-124 that may act via the peroxiredoxin 1 (PRX1). Our results provided further motivation to pursue the use of modified EVs as a treatment option for neurological diseases.
Collapse
Affiliation(s)
- Jianan Tian
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University
| | - Haiqian Yao
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University
| | - Yihang Liu
- Department of Cardiology, The Second Affiliated Hospital of Jilin University
| | - Xiaokun Wang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University
| | - Jiarong Wu
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University
| | - Yulan Zhu
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University
| | - Chunxiao Yang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University
| |
Collapse
|
48
|
Pang QM, Chen SY, Xu QJ, Zhang M, Liang DF, Fu SP, Yu J, Liu ZL, Zhang Q, Zhang T. Effects of astrocytes and microglia on neuroinflammation after spinal cord injury and related immunomodulatory strategies. Int Immunopharmacol 2022; 108:108754. [PMID: 35397392 DOI: 10.1016/j.intimp.2022.108754] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/14/2022] [Accepted: 03/31/2022] [Indexed: 12/12/2022]
Abstract
Spinal cord injury (SCI) is a catastrophic event which is still without adequate therapies. Neuroinflammation is the main pathogenesis of secondary damage post-SCI, leading to tissue loss and neurological dysfunction. Previous studies have shown that microglia and astrocytes are the major immune cells in the central nervous system (CNS) and play a crucial role in modulating neuroinflammatory responses. In this study, we mainly review the effects of neuroinflammation in SCI, focusing on the contributions of microglia and astrocytes and their cross-talk. Furthermore, we will also discuss therapeutic strategies on how to regulate their immunophenotype to suppress robust inflammation and facilitate injury prognosis.
Collapse
Affiliation(s)
- Qi-Ming Pang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China; Department of Orthopedics, 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-Jing Xu
- Department of Human Anatomy, 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
| | - Da-Fei Liang
- Department of Human Anatomy, Zunyi Medical University, Zunyi, Guizhou, China
| | - Sheng-Ping Fu
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China; Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Jiang Yu
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Zu-Lin Liu
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Qian Zhang
- Department of Human Anatomy, 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 Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China.
| |
Collapse
|
49
|
Icariin alleviates uveitis by targeting peroxiredoxin 3 to modulate retinal microglia M1/M2 phenotypic polarization. Redox Biol 2022; 52:102297. [PMID: 35334248 PMCID: PMC8956882 DOI: 10.1016/j.redox.2022.102297] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/10/2022] [Accepted: 03/16/2022] [Indexed: 11/22/2022] Open
Abstract
Uveitis causes blindness and critical visual impairment in people of all ages, and retinal microglia participate in uveitis progression. Unfortunately, effective treatment is deficient. Icariin (ICA) is a bioactive monomer derived from Epimedium. However, the role of ICA in uveitis remains elusive. Our study indicated that ICA alleviated intraocular inflammation in vivo. Further results showed the proinflammatory M1 microglia could be transferred to anti-inflammatory M2 microglia by ICA in the retina and HMC3 cells. However, the direct pharmacological target of ICA is unknown, to this end, proteome microarrays and molecular simulations were used to identify the molecular targets of ICA. Data showed that ICA binds to peroxiredoxin-3 (PRDX3), increasing PRDX3 protein expression in both a time- and a concentration-dependent manner and promoting the subsequent elimination of H2O2. In addition, GPX4/SLC7A11/ACSL4 pathways were activated accompanied by PRDX3 activation. Functional tests demonstrated that ICA-derived protection is afforded through targeting PRDX3. First, ICA-shifted microglial M1/M2 phenotypic polarization was no longer detected by blocking PRDX3 both in vivo and in vitro. Next, ICA-activated GPX4/SLC7A11/ACSL4 pathways and downregulated H2O2 production were also reversed via inhibiting PRDX3 both in vivo and in vitro. Finally, ICA-elicited positive effects on intraocular inflammation were eliminated in PRDX3-deficient retina from experimental autoimmune uveitis (EAU) mice. Taking together, ICA-derived PRDX3 activation has therapeutic potential for uveitis, which might be associated with modulating microglial M1/M2 phenotypic polarization.
Collapse
|
50
|
Pang QM, Chen SY, Fu SP, Zhou H, Zhang Q, Ao J, Luo XP, Zhang T. Regulatory Role of Mesenchymal Stem Cells on Secondary Inflammation in Spinal Cord Injury. J Inflamm Res 2022; 15:573-593. [PMID: 35115806 PMCID: PMC8802142 DOI: 10.2147/jir.s349572] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 12/24/2021] [Indexed: 12/13/2022] Open
Affiliation(s)
- Qi-Ming Pang
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
| | - Si-Yu Chen
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
| | - Sheng-Ping Fu
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
| | - Hui Zhou
- The First School of Clinical Medicine, Zunyi Medical University, Zunyi, People’s Republic of China
| | - Qian Zhang
- Department of Human Anatomy, Zunyi Medical University, Zunyi, People’s Republic of China
| | - Jun Ao
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
| | - Xiao-Ping Luo
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
| | - Tao Zhang
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
- Correspondence: Tao Zhang; Qian Zhang, Email ;
| |
Collapse
|