1
|
Zhou M, Jiang Z, Zhang M, Feng S, Ma B, Kan S, Fu X, Zhu R. Exploring the molecular mechanism of icariin improving spinal cord injury through network pharmacology combined with experimental verification. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03904-7. [PMID: 40014127 DOI: 10.1007/s00210-025-03904-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 02/08/2025] [Indexed: 02/28/2025]
Abstract
This study aimed to investigate the potential pharmacological effects of icariin (ICA) in the treatment of spinal cord injury (SCI). Network pharmacology was used to focus on the potential targets and biological processes of ICA in SCI. Molecular docking was used to verify the ability of ICA to bind to its core targets. Finally, valuate the efficacy and potential mechanisms of ICA in treating spinal cord injury through in vitro and in vivo experiments. A total of 37 targets were screened out, and core genes were screened out from the protein‒protein interaction network. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses revealed that these targets are enriched mainly in response to hypoxia, regulation of the cellular response to stress, and the TGF-beta signaling pathway. Molecular docking analysis showed that ICA has good docking ability with core targets. In animal experiments, Basso, Beattie and Bresnahan scores, catwalk gait analysis, hematoxylin and eosin staining, and RT-qPCR showed that ICA can inhibit spinal cord inflammation and effectively improve the behavioral and histological recovery after SCI rats. Western blot and immunofluorescence showed that ICA can reduce astrocyte activation and downregulate the TGF-beta signaling pathway after SCI. In addition, ICA can promote axonal nerve elongation and promotes angiogenesis after spinal cord injury in rats. In vitro experiments revealed that ICA can inhibit TGFβ1-induced activation of the TGF-beta signaling pathway and astrocyte activation. ICA treats SCI through multiple targets and pathways. ICA plays a major role in protecting nerves, promoting angiogenesis, and inhibiting reactive astrocyte activation in the treatment of SCI.
Collapse
Grants
- 2022JZXK05, 2022JZXK02, 2023YJZD002, 2022JZXK06 Tianjin Union Medical Center
- 2022JZXK05, 2022JZXK02, 2023YJZD002, 2022JZXK06 Tianjin Union Medical Center
- 2022JZXK05, 2022JZXK02, 2023YJZD002, 2022JZXK06 Tianjin Union Medical Center
- 2022JZXK05, 2022JZXK02, 2023YJZD002, 2022JZXK06 Tianjin Union Medical Center
- TJYXZDXK-064B Tianjin Key Medical Discipline (Specialty) Construction Project
- 2021125 Tianjin Municipal Health Commission's Integrated Traditional Chinese Medicine and Western Medicine Project
- 202305 Tianjin Health Commission Science and Technology Project
- TJWJ2024QN045 Tianjin Health Science and Technology Project
- 2023RC006 Yunnan Province talented Xing border plan
Collapse
Affiliation(s)
- Mengmeng Zhou
- Department of Spine Surgery, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, China
- Tianjin Key Specialty of Integrated Traditional Chinese and Western Medicine, Department of Spinal Rehabilitation with Integrated Traditional Chinese and Western Medicine), Tianjin, China
- Tianjin Institute of Rehabilitation, Tianjin, China
| | - Zehua Jiang
- Department of Spine Surgery, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, China
- Tianjin Key Specialty of Integrated Traditional Chinese and Western Medicine, Department of Spinal Rehabilitation with Integrated Traditional Chinese and Western Medicine), Tianjin, China
- Tianjin Institute of Rehabilitation, Tianjin, China
| | - Maosen Zhang
- Department of Spine Surgery, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, China
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin Medical University, Tianjin, China
| | - Sa Feng
- Department of Spine Surgery, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, China
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin Medical University, Tianjin, China
| | - Boyuan Ma
- Department of Spine Surgery, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, China
| | - Shunli Kan
- Department of Spine Surgery, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, China
- Tianjin Key Specialty of Integrated Traditional Chinese and Western Medicine, Department of Spinal Rehabilitation with Integrated Traditional Chinese and Western Medicine), Tianjin, China
- Tianjin Institute of Rehabilitation, Tianjin, China
| | - Xuanhao Fu
- Department of Spine Surgery, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, China
- Tianjin Key Specialty of Integrated Traditional Chinese and Western Medicine, Department of Spinal Rehabilitation with Integrated Traditional Chinese and Western Medicine), Tianjin, China
- Tianjin Institute of Rehabilitation, Tianjin, China
| | - Rusen Zhu
- Department of Spine Surgery, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, China.
- Tianjin Key Specialty of Integrated Traditional Chinese and Western Medicine, Department of Spinal Rehabilitation with Integrated Traditional Chinese and Western Medicine), Tianjin, China.
- Tianjin Institute of Rehabilitation, Tianjin, China.
| |
Collapse
|
2
|
Chen H, Feng Z, Min L, Tan M, Zhang D, Gong Q, Liu H, Hou J. Vagus Nerve Stimulation Prevents Endothelial Necroptosis to Alleviate Blood-Spinal Cord Barrier Disruption After Spinal Cord Injury. Mol Neurobiol 2023; 60:6466-6475. [PMID: 37460917 DOI: 10.1007/s12035-023-03477-7] [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: 04/24/2023] [Accepted: 06/30/2023] [Indexed: 09/28/2023]
Abstract
Vagus nerve stimulation (VNS) is a promising neuromodulation technique, which has been demonstrated to promote functional recovery after spinal cord injury (SCI) in our previous study. But the underlying mechanism remains to be explored. Using a compressed SCI model, our present study first demonstrated that activated microglia produce abundant tumor necrosis factor-α (TNF-α) to induce endothelial necroptosis via receptor-interacting protein kinase 1 (RIP1)/RIP3/mixed lineage kinase domain-like protein (MLKL) pathway, thus destroying the blood-spinal cord barrier (BSCB) after SCI. While both TNF-α specifical antibody (infliximab) and necroptosis inhibitor (necrostatin-1) alleviate BSCB disruption. Then our study found that VNS significantly inhibits microglia-derived TNF-α production and reduces expression of p-RIP3 and p-MLKL in endothelial cells. As expected, further results indicated that VNS mitigates the BSCB disruption, thus reducing inflammatory cells infiltration and neural damage. Finally, both electrophysiological evaluation and locomotor test demonstrated that VNS promotes motor function recovery after SCI. In conclusion, our data demonstrated VNS restricts microglia-derived TNF-α to prevent RIP1/RIP3/MLKL mediated endothelial necroptosis, thus alleviating the decisive pathophysiological BSCB disruption to reduce neuroinflammation and neural damage, which ultimately promotes motor function recovery after SCI. Therefore, these results further elaborate that VNS might be a promising therapeutic strategy for SCI. Vagus nerve stimulation prevents microglia-derived TNF-α induced endothelial necroptosis to alleviate blood-spinal cord barrier disruption after spinal cord injury.
Collapse
Affiliation(s)
- Hui Chen
- Department of Rehabilitation, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Department of Rehabilitation, Wusheng Hospital of Traditional Chinese Medicine, Sichuan, China
| | - Zhou Feng
- Department of Rehabilitation, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Lingxia Min
- Department of Rehabilitation, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Mingliang Tan
- Department of Rehabilitation, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Dongyun Zhang
- Department of Rehabilitation, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Qiuwen Gong
- Department of Rehabilitation, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hongliang Liu
- Department of Rehabilitation, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jingming Hou
- Department of Rehabilitation, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
| |
Collapse
|
3
|
Monaco CF, Davis JS. Mechanisms of angioregression of the corpus luteum. Front Physiol 2023; 14:1254943. [PMID: 37841308 PMCID: PMC10568036 DOI: 10.3389/fphys.2023.1254943] [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: 07/17/2023] [Accepted: 09/18/2023] [Indexed: 10/17/2023] Open
Abstract
The corpus luteum is a transient ovarian endocrine gland that produces the progesterone necessary for the establishment and maintenance of pregnancy. The formation and function of this gland involves angiogenesis, establishing the tissue with a robust blood flow and vast microvasculature required to support production of progesterone. Every steroidogenic cell within the corpus luteum is in direct contact with a capillary, and disruption of angiogenesis impairs luteal development and function. At the end of a reproductive cycle, the corpus luteum ceases progesterone production and undergoes rapid structural regression into a nonfunctional corpus albicans in a process initiated and exacerbated by the luteolysin prostaglandin F2α (PGF2α). Structural regression is accompanied by complete regression of the luteal microvasculature in which endothelial cells die and are sloughed off into capillaries and lymphatic vessels. During luteal regression, changes in nitric oxide transiently increase blood flow, followed by a reduction in blood flow and progesterone secretion. Early luteal regression is marked by an increased production of cytokines and chemokines and influx of immune cells. Microvascular endothelial cells are sensitive to released factors during luteolysis, including thrombospondin, endothelin, and cytokines like tumor necrosis factor alpha (TNF) and transforming growth factor β 1 (TGFB1). Although PGF2α is known to be a vasoconstrictor, endothelial cells do not express receptors for PGF2α, therefore it is believed that the angioregression occurring during luteolysis is mediated by factors downstream of PGF2α signaling. Yet, the exact mechanisms responsible for angioregression in the corpus luteum remain unknown. This review describes the current knowledge on angioregression of the corpus luteum and the roles of vasoactive factors released during luteolysis on luteal vasculature and endothelial cells of the microvasculature.
Collapse
Affiliation(s)
- Corrine F. Monaco
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE, United States
| | - John S. Davis
- Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE, United States
- US Department of Veterans Affairs Nebraska-Western Iowa Healthcare System, Omaha, NE, United States
| |
Collapse
|
4
|
Chopra M, Bhagwani A, Kumar H. The Provenance, Providence, and Position of Endothelial Cells in Injured Spinal Cord Vascular Pathology. Cell Mol Neurobiol 2023; 43:1519-1535. [PMID: 35945301 PMCID: PMC11412425 DOI: 10.1007/s10571-022-01266-9] [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: 01/06/2022] [Accepted: 07/21/2022] [Indexed: 11/03/2022]
Abstract
Endothelial cells (ECs) and pericytes are present in all blood vessels. Their position confers an important role in controlling oxygen and nutrient transportation to the different organs. ECs can adopt different morphologies based on their need and functions. Both ECs and pericytes express different surface markers that help in their identification, but heterogeneity and overlapping between markers among different cells pose a challenge for their precise identification. Spatiotemporal association of ECs and pericytes have great importance in sprout formation and vessel stabilization. Any traumatic injury in CNS may lead to vascular damage along with neuronal damage. Hence, ECs-pericyte interaction by physical contact and paracrine molecules is crucial in recovering the epicenter region by promoting angiogenesis. ECs can transform into other types of cells through endothelial-mesenchymal transition (EndMT), promoting wound healing in the epicenter region. Various signaling pathways mediate the interaction of ECs with pericytes that have an extensive role in angiogenesis. In this review, we discussed ECs and pericytes surface markers, the spatiotemporal association and interaction of ECs-pericytes, and signaling associated with the pathology of traumatic SCI. Linking the brain or spinal cord-specific pathologies and human vascular pathology will pave the way toward identifying new therapeutic targets and developing innovative preventive strategies. Endothelial-pericyte interaction strategic for formation of functional neo-vessels that are crucial for neurological recovery.
Collapse
Affiliation(s)
- Manjeet Chopra
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Ahmedabad, Opposite Air Force Station, Palaj, Gandhinagar, Gujarat, 382355, India
| | - Ankita Bhagwani
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Ahmedabad, Opposite Air Force Station, Palaj, Gandhinagar, Gujarat, 382355, India
| | - Hemant Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Ahmedabad, Opposite Air Force Station, Palaj, Gandhinagar, Gujarat, 382355, India.
| |
Collapse
|
5
|
Zhang H, Zhu L, Gao DS, Liu Y, Zhang J, Yan M, Qian J, Xi W. Imaging the Deep Spinal Cord Microvascular Structure and Function with High-Speed NIR-II Fluorescence Microscopy. SMALL METHODS 2022; 6:e2200155. [PMID: 35599368 DOI: 10.1002/smtd.202200155] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/23/2022] [Indexed: 06/15/2023]
Abstract
The spinal cord (SC) is crucial for a myriad of somatosensory, autonomic signal processing, and transductions. Understanding the SC vascular structure and function thus plays an integral part in neuroscience and clinical research. However, the dense layers of myelinated ascending axons on the dorsal side inconveniently grant the SC tissue with high optical scattering property, which significantly hinders the imaging depth of the SC vasculature in vivo. Commonly used antiscattering techniques such as multiphoton fluorescence microscopy have low imaging speed and cannot capture the rapid vascular particle flow without significant motion blur. Here, advantage of the high penetration of near-infrared (NIR)-II fluorescence is taken to demonstrate a deep SC vascular structural image stack up to 350 µm, comparable to two-photon microscopy. Furthermore, the red blood cells are labelled with the clinically approved NIR dye indocyanine. The combination of a fast NIR camera and indocyanine green-red blood cells (RBCs) makes it possible to attain high-speed 100 frame-per-second NIR-II imaging to identify the corresponding changes in RBC velocity during the external hind leg stimulus. For the first time, it is established that the NIR-II region would be a promising spectral window for SC imaging. NIR-II fluorescence microscopy has excellent potential for clinical and basic science research on SC.
Collapse
Affiliation(s)
- Hequn Zhang
- Interdisciplinary Institute of Neuroscience and Technology (ZIINT), Department of Anesthesiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, China
- MOE Frontier Science Center for Brain Research and Brain Machine Integration, Zhejiang University, Hangzhou, 310058, China
| | - Liang Zhu
- Interdisciplinary Institute of Neuroscience and Technology (ZIINT), Department of Anesthesiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, China
- MOE Frontier Science Center for Brain Research and Brain Machine Integration, Zhejiang University, Hangzhou, 310058, China
- Interdisciplinary Institute of Neuroscience and Technology (ZIINT), College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, 310027, China
| | - Dave Schwinn Gao
- Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Department of Anesthesiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Yin Liu
- Interdisciplinary Institute of Neuroscience and Technology (ZIINT), Department of Anesthesiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, China
- MOE Frontier Science Center for Brain Research and Brain Machine Integration, Zhejiang University, Hangzhou, 310058, China
| | - Jun Zhang
- Department of Spine Surgery, Zhejiang Provincial People's Hospital, Hangzhou Medical School People's Hospital, Shangtang Road 158th, Hangzhou, Zhejiang Province, 310014, China
| | - Min Yan
- Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Department of Anesthesiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Jun Qian
- State Key Laboratory of Modern Optical Instrumentations, Centre for Optical and Electromagnetic Research, College of Optical Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Wang Xi
- Interdisciplinary Institute of Neuroscience and Technology (ZIINT), Department of Anesthesiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, China
- MOE Frontier Science Center for Brain Research and Brain Machine Integration, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and instrument Science, Zhejiang University, Hangzhou, 310027, China
| |
Collapse
|
6
|
Gong L, Gu Y, Han X, Luan C, Liu C, Wang X, Sun Y, Zheng M, Fang M, Yang S, Xu L, Sun H, Yu B, Gu X, Zhou S. Spatiotemporal Dynamics of the Molecular Expression Pattern and Intercellular Interactions in the Glial Scar Response to Spinal Cord Injury. Neurosci Bull 2022; 39:213-244. [PMID: 35788904 PMCID: PMC9905408 DOI: 10.1007/s12264-022-00897-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 04/28/2022] [Indexed: 12/22/2022] Open
Abstract
Nerve regeneration in adult mammalian spinal cord is poor because of the lack of intrinsic regeneration of neurons and extrinsic factors - the glial scar is triggered by injury and inhibits or promotes regeneration. Recent technological advances in spatial transcriptomics (ST) provide a unique opportunity to decipher most genes systematically throughout scar formation, which remains poorly understood. Here, we first constructed the tissue-wide gene expression patterns of mouse spinal cords over the course of scar formation using ST after spinal cord injury from 32 samples. Locally, we profiled gene expression gradients from the leading edge to the core of the scar areas to further understand the scar microenvironment, such as neurotransmitter disorders, activation of the pro-inflammatory response, neurotoxic saturated lipids, angiogenesis, obstructed axon extension, and extracellular structure re-organization. In addition, we described 21 cell transcriptional states during scar formation and delineated the origins, functional diversity, and possible trajectories of subpopulations of fibroblasts, glia, and immune cells. Specifically, we found some regulators in special cell types, such as Thbs1 and Col1a2 in macrophages, CD36 and Postn in fibroblasts, Plxnb2 and Nxpe3 in microglia, Clu in astrocytes, and CD74 in oligodendrocytes. Furthermore, salvianolic acid B, a blood-brain barrier permeation and CD36 inhibitor, was administered after surgery and found to remedy fibrosis. Subsequently, we described the extent of the scar boundary and profiled the bidirectional ligand-receptor interactions at the neighboring cluster boundary, contributing to maintain scar architecture during gliosis and fibrosis, and found that GPR37L1_PSAP, and GPR37_PSAP were the most significant gene-pairs among microglia, fibroblasts, and astrocytes. Last, we quantified the fraction of scar-resident cells and proposed four possible phases of scar formation: macrophage infiltration, proliferation and differentiation of scar-resident cells, scar emergence, and scar stationary. Together, these profiles delineated the spatial heterogeneity of the scar, confirmed the previous concepts about scar architecture, provided some new clues for scar formation, and served as a valuable resource for the treatment of central nervous system injury.
Collapse
Affiliation(s)
- Leilei Gong
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Yun Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Xiaoxiao Han
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Chengcheng Luan
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Chang Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Xinghui Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Yufeng Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Mengru Zheng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Mengya Fang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Shuhai Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Lai Xu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Hualin Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Bin Yu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Xiaosong Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
| | - Songlin Zhou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
| |
Collapse
|
7
|
Abstract
This review provides a concise outline of the advances made in the care of patients and to the quality of life after a traumatic spinal cord injury (SCI) over the last century. Despite these improvements reversal of the neurological injury is not yet possible. Instead, current treatment is limited to providing symptomatic relief, avoiding secondary insults and preventing additional sequelae. However, with an ever-advancing technology and deeper understanding of the damaged spinal cord, this appears increasingly conceivable. A brief synopsis of the most prominent challenges facing both clinicians and research scientists in developing functional treatments for a progressively complex injury are presented. Moreover, the multiple mechanisms by which damage propagates many months after the original injury requires a multifaceted approach to ameliorate the human spinal cord. We discuss potential methods to protect the spinal cord from damage, and to manipulate the inherent inhibition of the spinal cord to regeneration and repair. Although acute and chronic SCI share common final pathways resulting in cell death and neurological deficits, the underlying putative mechanisms of chronic SCI and the treatments are not covered in this review.
Collapse
Affiliation(s)
- Stuart Stokes
- Spinal Unit, Department of Neurosurgery, Hull Royal Infirmary, Hull, UK
| | - Martin Drozda
- Spinal Unit, Department of Neurosurgery, Hull Royal Infirmary, Hull, UK
| | - Christopher Lee
- Spinal Unit, Department of Neurosurgery, Hull Royal Infirmary, Hull, UK
| |
Collapse
|
8
|
Wang J, Li Y, Yu H, Li G, Bai S, Chen S, Zhang P, Tang Z. Dl-3-N-Butylphthalide Promotes Angiogenesis in an Optimized Model of Transient Ischemic Attack in C57BL/6 Mice. Front Pharmacol 2021; 12:751397. [PMID: 34658892 PMCID: PMC8513739 DOI: 10.3389/fphar.2021.751397] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 09/15/2021] [Indexed: 11/16/2022] Open
Abstract
Transient ischemic attack (TIA) has been widely regarded as a clinical entity. Even though magnetic resonance imaging (MRI) results of TIA patients are negative, potential neurovascular damage might be present, and may account for long-term cognitive impairment. Animal models that simulate human diseases are essential tools for in-depth study of TIA. Previous studies have clarified that Dl-3-N-butylphthalide (NBP) promotes angiogenesis after stroke. However, the effects of NBP on TIA remain unknown. This study aims to develop an optimized TIA model in C57BL/6 mice to explore the microscopic evidence of ischemic injury after TIA, and investigate the therapeutic effects of NBP on TIA. C57BL/6 mice underwent varying durations (7, 8, 9 or 10 min) of middle cerebral artery occlusion (MCAO). Cerebral artery occlusion and reperfusion were assessed by laser speckle contrast imaging. TIA and ischemic stroke were distinguished by neurological testing and MRI examination at 24 h post-operation. Neuronal apoptosis was examined by TUNEL staining. Images of submicron cerebrovascular networks were obtained via micro-optical sectioning tomography. Subsequently, the mice were randomly assigned to a sham-operated group, a vehicle-treated TIA group or an NBP-treated TIA group. Vascular density was determined by immunofluorescent staining and fluorescein isothiocyanate method, and the expression of angiogenic growth factors were detected by western blot analysis. We found that an 8-min or shorter period of ischemia induced neither permanent neurological deficits nor MRI detectable brain lesions in C57BL/6 mice, but histologically caused neuronal apoptosis and cerebral vasculature abnormalities. NBP treatment increased the number of CD31+ microvessels and perfused microvessels after TIA. NBP also up-regulated the expression of VEGF, Ang-1 and Ang-2 and improved the cerebrovascular network. In conclusion, 8 min or shorter cerebral ischemia induced by the suture MCAO method is an appropriate TIA model in C57BL/6 mice, which conforms to the definition of human TIA, but causes microscopic neurovascular impairment. NBP treatment increased the expression of angiogenic growth factors, promoted angiogenesis and improved cerebral microvessels after TIA. Our study provides new insights on the pathogenesis and potential treatments of TIA.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ping Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhouping Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
9
|
Development of a skin temperature map for dermatomes in individuals with spinal cord injury: a cross-sectional study. Spinal Cord 2020; 58:1090-1095. [PMID: 32371938 DOI: 10.1038/s41393-020-0471-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 04/07/2020] [Accepted: 04/08/2020] [Indexed: 11/09/2022]
Abstract
STUDY DESIGN Cross-sectional study. OBJECTIVE The aim of this study was to map the skin temperature (Tsk) of individuals with SCI and compare able-bodied individuals, and among the groups to demonstrate the effects of differences in the levels of injury (paraplegia and tetraplegia with high and low injuries). SETTING Outpatient clinic, Brazil. METHODS Individuals with tetraplegia (n = 20), paraplegia (n = 21), and able-bodied (n = 11) individuals were recruited. A noncontact infrared thermometer (IRT) was used to measure three times the Tsk at the forehead, and at the C2 to S2 dermatomes. Core body temperature was measured at the axilla using the IRT and three other clinical thermometers. RESULTS Autonomic regulation is impaired by the injury. A Tsk map was constructed for the three groups. Significant differences in the Tsk of dermatomes were observed when comparing individuals with SCI and the able-bodied at the following dermatomes: C3, C7, T2, T3, T8, T9, L1, L2, L4, and S2. When comparing individuals with tetraplegia and able-bodied individuals, the dermatomes that showed significant differences were C5, C6, C8, T1, T10, L3, and S1. Dermatomes C5-C7, and T5 showed significant differences between individuals with tetraplegia and those with paraplegia. For L5 and S1 in paraplegia significant differences were found when comparing high with low injury. CONCLUSION A Tsk map on dermatomes in individuals with SCI was implemented, and showed a significant difference between able-bodied. As temperature is a parameter for analyzing autonomic function, the study could benefit rehabilitation by providing baseline values when constructing clinical protocols.
Collapse
|
10
|
Kumar H, Lim CS, Choi H, Joshi HP, Kim KT, Kim YH, Park CK, Kim HM, Han IB. Elevated TRPV4 Levels Contribute to Endothelial Damage and Scarring in Experimental Spinal Cord Injury. J Neurosci 2020; 40:1943-1955. [PMID: 31974206 PMCID: PMC7046444 DOI: 10.1523/jneurosci.2035-19.2020] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 12/10/2019] [Accepted: 01/04/2020] [Indexed: 11/21/2022] Open
Abstract
Currently, the role of transient receptor potential vanilloid type 4 (TRPV4), a nonselective cation channel in the pathology of spinal cord injury (SCI), is not recognized. Herein, we report the expression and contribution of TRPV4 in the pathology of scarring and endothelial and secondary damage after SCI. TRPV4 expression increased during the inflammatory phase in female rats after SCI and was expressed primarily by cells at endothelial-microglial junctions. Two-photon microscopy of intracellular-free Ca2+ levels revealed a biphasic increase at similar time points after SCI. Expression of TRPV4 at the injury epicenter, but not intracellular-free Ca2+, progressively increases with the severity of the injury. Activation of TRPV4 with specific agonist altered the organization of endothelial cells, affected tight junctions in the hCMEC/D3 BBB cell line in vitro, and increases the scarring in rat spinal cord as well as induced endothelial damage. By contrast, suppression of TRPV4 with a specific antagonist or in female Trpv4 KO mouse attenuated inflammatory cytokines and chemokines, prevented the degradation of tight junction proteins, and preserve blood-spinal cord barrier integrity, thereby attenuate the scarring after SCI. Likewise, secondary damage was reduced, and behavioral outcomes were improved in Trpv4 KO mice after SCI. These results suggest that increased TRPV4 expression disrupts endothelial cell organization during the early inflammatory phase of SCI, resulting in tissue damage, vascular destabilization, blood-spinal cord barrier breakdown, and scarring. Thus, TRPV4 inhibition/knockdown represents a promising therapeutic strategy to stabilize/protect endothelial cells, attenuate nociception and secondary damage, and reduce scarring after SCI.SIGNIFICANCE STATEMENT TRPV4, a calcium-permeable nonselective cation channel, is widely expressed in both excitable and nonexcitable cells. Spinal cord injury (SCI) majorly caused by trauma/accidents is associated with changes in osmolarity, mechanical injury, and shear stress. After SCI, TRPV4 was increased and were found to be linked with the severity of injury at the epicenter at the time points that were reported to be critical for repair/treatment. Activation of TRPV4 was damaging to endothelial cells that form the blood-spinal cord barrier and thus contributes to scarring (glial and fibrotic). Importantly, inhibition/knockdown of TRPV4 prevented these effects. Thus, the manipulation of TRPV4 signaling might lead to new therapeutic strategies or combinatorial therapies to protect endothelial cells and enhance repair after SCI.
Collapse
Affiliation(s)
- Hemant Kumar
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, Republic of Korea, 13488
| | - Chang Su Lim
- Department of Energy System Research and Department of Chemistry, Ajou University, Suwon, Gyeonggi-do, Republic of Korea, 16499
| | - Hyemin Choi
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, Republic of Korea, 13488
| | - Hari Prasad Joshi
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, Republic of Korea, 13488
| | - Kyoung-Tae Kim
- Department of Neurosurgery, School of Medicine, Kyungpook National University, Daegu, Republic of Korea, 41944
- Department of Neurosurgery, Kyungpook National University Hospital, Daegu, Republic of Korea, 41944, and
| | - Yong Ho Kim
- Gachon Pain Center and Department of Physiology, College of Medicine, Gachon University, Incheon, Republic of Korea, 13120
| | - Chul-Kyu Park
- Gachon Pain Center and Department of Physiology, College of Medicine, Gachon University, Incheon, Republic of Korea, 13120
| | - Hwan Myung Kim
- Department of Energy System Research and Department of Chemistry, Ajou University, Suwon, Gyeonggi-do, Republic of Korea, 16499
| | - In-Bo Han
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, Republic of Korea, 13488,
| |
Collapse
|
11
|
Krishnaswamy VR, Benbenishty A, Blinder P, Sagi I. Demystifying the extracellular matrix and its proteolytic remodeling in the brain: structural and functional insights. Cell Mol Life Sci 2019; 76:3229-3248. [PMID: 31197404 PMCID: PMC11105229 DOI: 10.1007/s00018-019-03182-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 05/29/2019] [Accepted: 05/31/2019] [Indexed: 12/29/2022]
Abstract
The extracellular matrix (ECM) plays diverse roles in several physiological and pathological conditions. In the brain, the ECM is unique both in its composition and in functions. Furthermore, almost all the cells in the central nervous system contribute to different aspects of this intricate structure. Brain ECM, enriched with proteoglycans and other small proteins, aggregate into distinct structures around neurons and oligodendrocytes. These special structures have cardinal functions in the normal functioning of the brain, such as learning, memory, and synapse regulation. In this review, we have compiled the current knowledge about the structure and function of important ECM molecules in the brain and their proteolytic remodeling by matrix metalloproteinases and other enzymes, highlighting the special structures they form. In particular, the proteoglycans in brain ECM, which are essential for several vital functions, are emphasized in detail.
Collapse
Affiliation(s)
| | - Amit Benbenishty
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Pablo Blinder
- Neurobiology, Biochemistry and Biophysics School, Tel Aviv University, Tel Aviv, Israel
- Sagol School for Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Irit Sagi
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
12
|
Synchrotron Radiation Imaging Reveals the Role of Estrogen in Promoting Angiogenesis After Acute Spinal Cord Injury in Rats. Spine (Phila Pa 1976) 2018. [PMID: 29529001 DOI: 10.1097/brs.0000000000002629] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN The efficacy of estrogen on vessel angiogenesis in acute spinal cord injury (SCI) in a rat model was evaluated by synchrotron radiation. OBJECTIVE Here, we investigate the change in injured spinal cord vessels and used the synchrotron radiation to investigate the effect of estrogen on vessel angiogenesis and functional recovery in a rat model of SCI. SUMMARY OF BACKGROUND DATA The promotion of angiogenesis after SCI may be a therapeutic target in the treatment of SCI. Estrogen has been reported to improve locomotor recovery after SCI. However, how estrogen regulates angiogenesis in acute SCI and enhances neurological functional recovery has not been fully characterized. METHODS Synchrotron radiation imaging combined with histological methods was used to image angiogenesis in acute spinal cord treatment with estrogen in rats. RESULTS Synchrotron radiation imaging vividly demonstrated three-dimensional vessel changes in the spinal cord after injury. The imaging showed that vessel number, vessel volume fraction, and vessel connectivity value in the groups treated with estrogen after SCI were significantly increased compared to control groups (P < 0.05). Vessel angiogenesis increased in groups treated with estrogen compared with control rats, which was confirmed with histological staining. Estrogen treatment also attenuated the injury-induced lesion area compared with control groups and improved locomotor functional recovery after SCI. CONCLUSION The results indicated that synchrotron radiation is a powerful imaging tool for visualizing angiogenesis after acute SCI. Estrogen treatment exerted a neuroprotective effect on acute SCI treatment by promoting angiogenesis and reducing the injury-induced lesion area could be recommended as a potential preclinical treatment approach for acute SCI. LEVEL OF EVIDENCE N/A.
Collapse
|
13
|
Kumar H, Choi H, Jo MJ, Joshi HP, Muttigi M, Bonanomi D, Kim SB, Ban E, Kim A, Lee SH, Kim KT, Sohn S, Zeng X, Han I. Neutrophil elastase inhibition effectively rescued angiopoietin-1 decrease and inhibits glial scar after spinal cord injury. Acta Neuropathol Commun 2018; 6:73. [PMID: 30086801 PMCID: PMC6080383 DOI: 10.1186/s40478-018-0576-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 07/23/2018] [Indexed: 01/13/2023] Open
Abstract
After spinal cord injury (SCI), neutrophil elastase (NE) released at injury site disrupts vascular endothelium integrity and stabilization. Angiopoietins (ANGPTs) are vascular growth factors that play an important role in vascular stabilization. We hypothesized that neutrophil elastase is one of the key determinants of vascular endothelium disruption/destabilization and affects angiopoietins expression after spinal cord injury. To test this, tubule formation and angiopoietins expression were assessed in endothelial cells exposed to different concentrations of recombinant neutropil elastase. Then, the expression of angiopoietin-1, angiopoietin-2, and neutrophil elastase was determined at 3 h and at 1, 3, 5, 7, 14, 21, and 28 days in a clinically relevant model of moderate compression (35 g for 5 min at T10) spinal cord injury. A dichotomy between the levels of angiopoietin-1 and angiopoietin-2 was observed; thus, we utilized a specific neutrophil elastase inhibitor (sivelestat sodium; 30 mg/kg, i.p., b.i.d.) after spinal cord injury. The expression levels of neutropil elastase and angiopoietin-2 increased, and that of angiopoietin-1 decreased after spinal cord injury in rats. The sivelestat regimen, optimized via a pharmacokinetics study, had potent effects on vascular stabilization by upregulating angiopoietin-1 via the AKT pathway and preventing tight junction protein degradation. Moreover, sivelestat attenuated the levels of inflammatory cytokines and chemokines after spinal cord injury and hence subsequently alleviated secondary damage observed as a reduction in glial scar formation and the promotion of blood vessel formation and stabilization. As a result, hindlimb locomotor function significantly recovered in the sivelestat-treated animals as determined by the Basso, Beattie, and Bresnahan scale and footprint analyses. Furthermore, sivelestat treatment attenuated neuropathic pain as assessed by responses to von Frey filaments after spinal cord injury. Thus, our result suggests that inhibiting neutropil elastase by administration of sivelestat is a promising therapeutic strategy to inhibit glial scar and promote functional recovery by upregulating angiopoietin-1 after spinal cord injury.
Collapse
Affiliation(s)
- Hemant Kumar
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, 13496, Republic of Korea
| | - Hyemin Choi
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, 13496, Republic of Korea
| | - Min-Jae Jo
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, 13496, Republic of Korea
| | - Hari Prasad Joshi
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, 13496, Republic of Korea
| | - Manjunatha Muttigi
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Dario Bonanomi
- Molecular Neurobiology Laboratory, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Sung Bum Kim
- Department of Neurosurgery, Kyung Hee University, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Eunmi Ban
- College of Pharmacy, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Aeri Kim
- College of Pharmacy, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Soo-Hong Lee
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Kyoung-Tae Kim
- Department of Neurosurgery, Kyungpook National University Hospital, Kyungpook National University, 130, Dongdeok-ro, Jung-gu, Daegu, 41944, Republic of Korea
- Department of Neurosurgery, School of Medicine,Kyungpook National University, 130, Dongdeok-ro, Jung-gu, Daegu, 41944, Republic of Korea
| | - Seil Sohn
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, 13496, Republic of Korea
| | - Xiang Zeng
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong Province, China.
| | - Inbo Han
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, 13496, Republic of Korea.
| |
Collapse
|
14
|
Ferrer-Ferrer M, Dityatev A. Shaping Synapses by the Neural Extracellular Matrix. Front Neuroanat 2018; 12:40. [PMID: 29867379 PMCID: PMC5962695 DOI: 10.3389/fnana.2018.00040] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 04/25/2018] [Indexed: 11/13/2022] Open
Abstract
Accumulating data support the importance of interactions between pre- and postsynaptic neuronal elements with astroglial processes and extracellular matrix (ECM) for formation and plasticity of chemical synapses, and thus validate the concept of a tetrapartite synapse. Here we outline the major mechanisms driving: (i) synaptogenesis by secreted extracellular scaffolding molecules, like thrombospondins (TSPs), neuronal pentraxins (NPs) and cerebellins, which respectively promote presynaptic, postsynaptic differentiation or both; (ii) maturation of synapses via reelin and integrin ligands-mediated signaling; and (iii) regulation of synaptic plasticity by ECM-dependent control of induction and consolidation of new synaptic configurations. Particularly, we focused on potential importance of activity-dependent concerted activation of multiple extracellular proteases, such as ADAMTS4/5/15, MMP9 and neurotrypsin, for permissive and instructive events in synaptic remodeling through localized degradation of perisynaptic ECM and generation of proteolytic fragments as inducers of synaptic plasticity.
Collapse
Affiliation(s)
- Maura Ferrer-Ferrer
- Molecular Neuroplasticity German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Alexander Dityatev
- Molecular Neuroplasticity German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.,Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany.,Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
15
|
Sugar-Binding Profiles of Chitin-Binding Lectins from the Hevein Family: A Comprehensive Study. Int J Mol Sci 2017; 18:ijms18061160. [PMID: 28556796 PMCID: PMC5485984 DOI: 10.3390/ijms18061160] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 04/28/2017] [Accepted: 05/21/2017] [Indexed: 01/16/2023] Open
Abstract
Chitin-binding lectins form the hevein family in plants, which are defined by the presence of single or multiple structurally conserved GlcNAc (N-acetylglucosamine)-binding domains. Although they have been used as probes for chito-oligosaccharides, their detailed specificities remain to be investigated. In this study, we analyzed six chitin-binding lectins, DSA, LEL, PWM, STL, UDA, and WGA, by quantitative frontal affinity chromatography. Some novel features were evident: WGA showed almost comparable affinity for pyridylaminated chitotriose and chitotetraose, while LEL and UDA showed much weaker affinity, and DSA, PWM, and STL had no substantial affinity for the former. WGA showed selective affinity for hybrid-type N-glycans harboring a bisecting GlcNAc residue. UDA showed extensive binding to high-mannose type N-glycans, with affinity increasing with the number of Man residues. DSA showed the highest affinity for highly branched N-glycans consisting of type II LacNAc (N-acetyllactosamine). Further, multivalent features of these lectins were investigated by using glycoconjugate and lectin microarrays. The lectins showed substantial binding to immobilized LacNAc as well as chito-oligosaccharides, although the extents to which they bound varied among them. WGA showed strong binding to heavily sialylated glycoproteins. The above observations will help interpret lectin-glycoprotein interactions in histochemical studies and glyco-biomarker investigations.
Collapse
|
16
|
Lee JY, Choi HY, Baik HH, Ju BG, Kim WK, Yune TY. Cordycepin-enriched WIB-801C from Cordyceps militaris improves functional recovery by attenuating blood-spinal cord barrier disruption after spinal cord injury. JOURNAL OF ETHNOPHARMACOLOGY 2017; 203:90-100. [PMID: 28363523 DOI: 10.1016/j.jep.2017.03.047] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 03/07/2017] [Accepted: 03/25/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cordyceps militaris is an ingredient of traditional Chinese medicine and have been widely used for inflammatory diseases and cancer. Cordycepin is one of the major bioactive components of Cordyceps militaris, and has been known to have anti-inflammatory and anti-oxidant effects. AIM OF THIS STUDY In the present study, we examined whether WIB-801C, a standardized and cordycepin-enriched extract of caterpillar fungus (Cordyceps militaris), would attenuate blood-spinal cord barrier (BSCB) disruption by inhibiting matrix metalloprotease (MMP)-9 activity, leading to improvement of functional outcomes after spinal cord injury (SCI). MATERIALS AND METHODS Male Sprague-Dawley rats were subjected to contusive SCI using a New York University (NYU) impactor, and WIB-801C (50mg/kg) was administered at 2h and 8h after injury orally and further treated once a day for indicated time points. BSCB disruption, MMP-9 activity, blood infiltration, inflammation, neuronal apoptosis, axonal loss, demyelination, and neurological deficit were evaluated. RESULTS We found that WIB-801C significantly attenuated BSCB disruption by inhibiting MMP-9 expression and activation after injury. The infiltration of neutrophils at 1 d and macrophage at 5 d after SCI was also ameliorated by WIB-801C as compared with vehicle control. In addition, the expression of inflammatory cytokines and mediators such as Tnf-α, IL-1β, IL-6, Cox-2, and inos as well as chemokines such as Gro-α and Mip-2α was significantly inhibited by WIB-801C. Furthermore, WIB-801C inhibits p38MAPK activation and proNGF production in microglia after injury. These events eventually led to the inhibition of apoptotic cell death of neurons and oligodendrocytes, improved functional recovery and attenuated demyelination and axon loss after SCI. CONCLUSION Our results suggest that WIB-801C can be used as a therapeutic agent after SCI by attenuating BSCB disruption followed inflammation.
Collapse
Affiliation(s)
- Jee Youn Lee
- Age-Related and Brain Diseases Research Center, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea.
| | - Hye Young Choi
- Age-Related and Brain Diseases Research Center, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea.
| | - Hyung Hwan Baik
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea.
| | - Bong G Ju
- Department of Life Science, Sogang University, Seoul 04107, Republic of Korea.
| | - Won-Ki Kim
- Department of Neuroscience, College of Medicine, Korea University, Seoul 02841, Republic of Korea.
| | - Tae Young Yune
- Age-Related and Brain Diseases Research Center, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea.
| |
Collapse
|
17
|
Olfert IM. Physiological Capillary Regression is not Dependent on Reducing VEGF Expression. Microcirculation 2016; 23:146-56. [PMID: 26660949 PMCID: PMC4744091 DOI: 10.1111/micc.12263] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 12/03/2015] [Indexed: 01/04/2023]
Abstract
Investigations into physiologically controlled capillary regression report the provocative finding that microvessel regression occurs in the face of persistent elevation of skeletal muscle VEGF expression. TSP-1, a negative angiogenic regulator, is increasingly being observed to temporally correlate with capillary regression, suggesting that increased TSP-1 (and not reduction in VEGF per se) is needed to initiate, and likely regulate, capillary regression. Based on evidence being gleaned from physiologically mediated regression of capillaries, it needs to be recognized that capillary regression (and perhaps capillary rarefaction with disease) is not simply the reversal of factors used to stimulate angiogenesis. Rather, the conceptual understanding that angiogenesis and capillary regression each have specific and unique requirements that are biologically constrained to opposite sides of the balance between positive and negative angioregulatory factors may shed light on why anti-VEGF therapies have not lived up to the promise in reversing angiogenesis and providing the cure that many had hoped toward fighting cancer. Emerging evidence from physiological controlled angiogenesis suggest that cases involving excessive or uncontrolled capillary expansion may be best treated by therapies designed to increase expression of negative angiogenic regulators, whereas those involving capillary rarefaction may benefit from inhibiting negative regulators (like TSP-1).
Collapse
Affiliation(s)
- I Mark Olfert
- Division of Exercise Physiology, Center for Cardiovascular and Respiratory Sciences, Mary Babb Randolph Cancer Center, West Virginia Clinical and Translational Science Institute, West Virginia University School of Medicine, Morgantown, West Virginia, USA
| |
Collapse
|
18
|
Jiang DX, Lu ZS, Li GB, Sun SY, Mu X, Lee P, Chen W. Electroacupuncture improves microcirculation and neuronal morphology in the spinal cord of a rat model of intervertebral disc extrusion. Neural Regen Res 2015; 10:237-43. [PMID: 25883622 PMCID: PMC4392671 DOI: 10.4103/1673-5374.152377] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/22/2014] [Indexed: 12/03/2022] Open
Abstract
Most studies on spinal cord neuronal injury have focused on spinal cord tissue histology and the expression of nerve cell damage and repair-related genes. The importance of the microcirculation is often ignored in spinal cord injury and repair research. Therefore, in this study, we established a rat model of intervertebral disc extrusion by inserting a silica gel pad into the left ventral surface of T13. Electroacupuncture was used to stimulate the bilateral Zusanli point (ST36) and Neiting point (ST44) for 14 days. Compared with control animals, blood flow in the first lumbar vertebra (L1) was noticeably increased in rats given electroacupuncture. Microvessel density in the T13 segment of the spinal cord was increased significantly as well. The number of normal neurons was higher in the ventral horn of the spinal cord. In addition, vacuolation in the white matter was lessened. No obvious glial cell proliferation was visible. Furthermore, hindlimb motor function was improved significantly. Collectively, our results suggest that electroacupuncture can improve neuronal morphology and microcirculation, and promote the recovery of neurological functions in a rat model of intervertebral disc extrusion.
Collapse
Affiliation(s)
- Dai-Xun Jiang
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, Beijing, China
| | - Zhi-Song Lu
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, Beijing, China
| | - Ge-Bin Li
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, Beijing, China ; Department of Veterinary Biochemistry, Nippon Veterinary and Life Science University, Musashino, Tokyo, Japan
| | - Sheng-Yong Sun
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, Beijing, China
| | - Xiang Mu
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, Beijing, China
| | - Peter Lee
- Department of Veterinary Biochemistry, Nippon Veterinary and Life Science University, Musashino, Tokyo, Japan
| | - Wu Chen
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, Beijing, China
| |
Collapse
|
19
|
Zhang H, Li F, Yang Y, Chen J, Hu X. SIRP/CD47 signaling in neurological disorders. Brain Res 2015; 1623:74-80. [PMID: 25795378 DOI: 10.1016/j.brainres.2015.03.012] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 03/02/2015] [Accepted: 03/03/2015] [Indexed: 11/26/2022]
Abstract
Microglia play important roles in the process of neuronal injury and recovery. Numeous surface receptors have been described to regulate microglial activation. These receptors tightly mediate normal microglial functions including cell mobility, phagocytosis, and production of inflammatory mediators or trophic factors. In recent years, significant progresses have been achieved for understanding the signaling mechanisms underlying these receptors. Their specific roles in neurological diseases have been documented. This review will focus on the signal regulatory protein (SIRP) and its ligand CD47, two surface receptors expressed on microglia and other cells in the central nervous system (CNS) such as neurons. We will discuss the involvement of SIRP/CD47 signaling in microglial activation and in the interplay between microglia and other CNS cells. Current studies reveal the importance of CD47 and SIRPα in the process of neuroinflammation in the CNS disorders. The dual and contradictory role of CD47 suggests that targeting the SIRPα/CD47 signaling may achieve different effects depending on disease stage. This article is part of a Special Issue entitled SI: Cell Interactions In Stroke.
Collapse
Affiliation(s)
- Haiyue Zhang
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Xianaya School of Medicine, Central South University, Changsha, Hunan, China
| | - Fengwu Li
- China-America Institute of Neuroscience, Luhe Teaching Hospital, Capital Medical University, Beijing, China
| | - Yuanyuan Yang
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Xianaya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jun Chen
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| | - Xiaoming Hu
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; China-America Institute of Neuroscience, Luhe Teaching Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
20
|
Phamduong E, Rathore MK, Crews NR, D’Angelo AS, Leinweber AL, Kappera P, Krenning TM, Rendell VR, Belcheva MM, Coscia CJ. Acute and chronic mu opioids differentially regulate thrombospondins 1 and 2 isoforms in astrocytes. ACS Chem Neurosci 2014; 5:106-14. [PMID: 24304333 DOI: 10.1021/cn400172n] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Chronic opioids induce synaptic plasticity, a major neuronal adaptation. Astrocyte activation in synaptogenesis may play a critical role in opioid tolerance, withdrawal, and dependence. Thrombospondins 1 and 2 (TSP1/2) are astrocyte-secreted matricellular glycoproteins that promote neurite outgrowth as well as dendritic spine and synapse formation, all of which are inhibited by chronic μ opioids. In prior studies, we discovered that the mechanism of TSP1 regulation by μ opioids in astrocytes involves crosstalk between three different classes of receptors, μ opioid receptor, EGFR and TGFβR. Moreover, TGFβ1 stimulated TSP1 expression via EGFR and ERK/MAPK activation, indicating that EGFR is a signaling hub for opioid and TGFβ1 actions. Using various selective antagonists, and inhibitors, here we compared the mechanisms of chronic opioid regulation of TSP1/2 isoform expression in vivo and in immortalized rat cortical astrocytes. TSP1/2 release from astrocytes was also monitored. Acute and chronic μ opioids, morphine, and the prototypic μ ligand, DAMGO, modulated TSP2 protein levels. TSP2 but not TSP1 protein content was up-regulated by acute (3 h) morphine or DAMGO by an ERK/MAPK dependent mechanism. Paradoxically, TSP2 protein levels were altered neither by TGFβ1 nor by astrocytic neurotrophic factors, EGF, CNTF, and BMP4. TSP1/2 immunofluorescence was increased in astrocytes subjected to scratch-wounding, suggesting TSPs may be useful markers for the "reactive" state of these cells and potentially for different types of injury. Previously, we determined that chronic morphine attenuated both neurite outgrowth and synapse formation in cocultures of primary astrocytes and neurons under similar temporal conditions that μ opioids reduced TSP1 protein levels in astrocytes. Here we found that, after the same 8 day treatment, morphine or DAMGO diminished TSP2 protein levels in astrocytes. Therefore, μ opioids may deter synaptogenesis via both TSP1/2 isoforms, but by distinct mechanisms.
Collapse
Affiliation(s)
- Ellen Phamduong
- E. A. Doisy Department of
Biochemistry and Molecular Biology, St. Louis University of Medicine, 1100 S. Grand Blvd., St. Louis, Missouri, 63104, United States
| | - Maanjot K. Rathore
- E. A. Doisy Department of
Biochemistry and Molecular Biology, St. Louis University of Medicine, 1100 S. Grand Blvd., St. Louis, Missouri, 63104, United States
| | - Nicholas R. Crews
- E. A. Doisy Department of
Biochemistry and Molecular Biology, St. Louis University of Medicine, 1100 S. Grand Blvd., St. Louis, Missouri, 63104, United States
| | - Alexander S. D’Angelo
- E. A. Doisy Department of
Biochemistry and Molecular Biology, St. Louis University of Medicine, 1100 S. Grand Blvd., St. Louis, Missouri, 63104, United States
| | - Andrew L. Leinweber
- E. A. Doisy Department of
Biochemistry and Molecular Biology, St. Louis University of Medicine, 1100 S. Grand Blvd., St. Louis, Missouri, 63104, United States
| | - Pranay Kappera
- E. A. Doisy Department of
Biochemistry and Molecular Biology, St. Louis University of Medicine, 1100 S. Grand Blvd., St. Louis, Missouri, 63104, United States
| | - Thomas M. Krenning
- E. A. Doisy Department of
Biochemistry and Molecular Biology, St. Louis University of Medicine, 1100 S. Grand Blvd., St. Louis, Missouri, 63104, United States
| | - Victoria R. Rendell
- E. A. Doisy Department of
Biochemistry and Molecular Biology, St. Louis University of Medicine, 1100 S. Grand Blvd., St. Louis, Missouri, 63104, United States
| | - Mariana M. Belcheva
- E. A. Doisy Department of
Biochemistry and Molecular Biology, St. Louis University of Medicine, 1100 S. Grand Blvd., St. Louis, Missouri, 63104, United States
| | - Carmine J. Coscia
- E. A. Doisy Department of
Biochemistry and Molecular Biology, St. Louis University of Medicine, 1100 S. Grand Blvd., St. Louis, Missouri, 63104, United States
| |
Collapse
|
21
|
Zhou L, Wang H, Luo J, Xiong K, Zeng L, Chen D, Huang J. Regulatory effects of inhibiting the activation of glial cells on retinal synaptic plasticity. Neural Regen Res 2014; 9:385-393. [PMID: 25206825 PMCID: PMC4146193 DOI: 10.4103/1673-5374.128240] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2014] [Indexed: 01/09/2023] Open
Abstract
Various retinal injuries induced by ocular hypertension have been shown to induce plastic changes in retinal synapses, but the potential regulatory mechanism of synaptic plasticity after retinal injury was still unclear. A rat model of acute ocular hypertension was established by injecting saline intravitreally for an hour, and elevating the intraocular pressure to 14.63 kPa (110 mmHg). Western blot assay and immunofluorescence results showed that synaptophysin expression had a distinct spatiotemporal change that increased in the inner plexiform layer within 1 day and spread across the outer plexiform layer after 3 days. Glial fibrillary acidic protein expression in retinae was greatly increased after 3 days, and reached a peak at 7 days, which was also consistent with the peak time of synaptophysin expression in the outer plexiform layer following the increased intraocular pressure. Fluorocitrate, a glial metabolic inhibitor, was intravitreally injected to inhibit glial cell activation following high intraocular pressure. This significantly inhibited the enhanced glial fibrillary acidic protein expression induced by high intraocular pressure injury. Synaptophysin expression also decreased in the inner plexiform layer within a day and the widened distribution in the outer plexiform layer had disappeared by 3 days. The results suggested that retinal glial cell activation might play an important role in the process of retinal synaptic plasticity induced by acute high intraocular pressure through affecting the expression and distribution of synaptic functional proteins, such as synaptophysin.
Collapse
Affiliation(s)
- Lihong Zhou
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Hui Wang
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Jia Luo
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Kun Xiong
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Leping Zeng
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Dan Chen
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| | - Jufang Huang
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, China
| |
Collapse
|
22
|
Kundi S, Bicknell R, Ahmed Z. The role of angiogenic and wound-healing factors after spinal cord injury in mammals. Neurosci Res 2013; 76:1-9. [PMID: 23562792 DOI: 10.1016/j.neures.2013.03.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 03/18/2013] [Accepted: 03/20/2013] [Indexed: 12/17/2022]
Abstract
Patients with spinal cord injury (SCI) are permanently paralysed and anaesthetic below the lesion. This morbidity is attributed to the deposition of a dense scar at the injury site, the cellular components of which secrete axon growth inhibitory ligands that prevent severed axons reconnecting with denervated targets. Another complication of SCI is wound cavitation where a fluid filled cyst forms in the peri-lesion neuropil, enlarging over the first few months after injury and causes secondary axonal damage. Wound healing after SCI is accompanied by angiogenesis, which is regulated by angiogenic proteins, produced in response to oxygen deprivation. Necrosis in and about the SCI lesion sites may be suppressed by promoting angiogenesis and the resulting neuropil protection will enhance recovery after SCI. This review addresses the use of angiogenic/wound-healing related proteins including vascular endothelial growth factor, fibroblast growth factor, angiopoietin-1, angiopoietin-2 and transforming growth factor-β to moderate necrosis and axon sparing after SCI, providing a conducive environment for growth essential to functional recovery.
Collapse
Affiliation(s)
- Sarina Kundi
- Neurotrauma and Neurodegeneration, School of Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, UK
| | | | | |
Collapse
|
23
|
Lee K, Na W, Lee JY, Na J, Cho H, Wu H, Yune TY, Kim WS, Ju BG. Molecular mechanism of Jmjd3-mediated interleukin-6 gene regulation in endothelial cells underlying spinal cord injury. J Neurochem 2012; 122:272-82. [PMID: 22578249 DOI: 10.1111/j.1471-4159.2012.07786.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The inflammatory response contributes substantially to secondary injury cascades after spinal cord injury, with both neurotoxic and protective effects. However, epigenetic regulations of inflammatory genes following spinal cord injury have yet to be characterized thoroughly. In this study, we found that histone H3K27me3 demethylase Jmjd3 expression is acutely up-regulated in blood vessels of the injured spinal cord. We also observed up-regulation of Jmjd3 gene expression in bEnd.3 endothelial cells that were subjected to oxygen-glucose deprivation/reperfusion injury. When Jmjd3 was depleted by siRNA, oxygen-glucose deprivation/reperfusion injury-induced up-regulation of IL-6 was significantly inhibited. In addition, Jmjd3 associated with NF-κB (p65/p50) and CCAAT-enhancer-binding protein β at the IL-6 gene promoter. The recruitment of Jmjd3 coincided with decreased levels of tri-methylated H3K27 as well as increased levels of mono-methylated H3K27 at the IL-6 gene promoter. Furthermore, Jmjd3 depletion did not result in significant changes of methylation level of H3K27 at the IL-6 gene promoter. Collectively, our findings imply that Jmjd3-mediated H3K27me3 demethylation is crucial for IL-6 gene activation in endothelial cells, and this molecular event may regulate acute inflammatory response and integrity of the blood-spinal cord barrier following spinal cord injury.
Collapse
Affiliation(s)
- Kwanghyun Lee
- Department of Life Science, Sogang University, Seoul, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Jaerve A, Müller HW. Chemokines in CNS injury and repair. Cell Tissue Res 2012; 349:229-48. [PMID: 22700007 DOI: 10.1007/s00441-012-1427-3] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Accepted: 04/05/2012] [Indexed: 12/17/2022]
Abstract
Recruitment of inflammatory cells is known to drive the secondary damage cascades that are common to injuries of the central nervous system (CNS). Cell activation and infiltration to the injury site is orchestrated by changes in the expression of chemokines, the chemoattractive cytokines. Reducing the numbers of recruited inflammatory cells by the blocking of the action of chemokines has turned out be a promising approach to diminish neuroinflammation and to improve tissue preservation and neovascularization. In addition, several chemokines have been shown to be essential for stem/progenitor cell attraction, their survival, differentiation and cytokine production. Thus, chemokines might indirectly participate in remyelination, neovascularization and neuroprotection, which are important prerequisites for CNS repair after trauma. Moreover, CXCL12 promotes neurite outgrowth in the presence of growth inhibitory CNS myelin and enhances axonal sprouting after spinal cord injury (SCI). Here, we review current knowledge about the exciting functions of chemokines in CNS trauma, including SCI, traumatic brain injury and stroke. We identify common principles of chemokine action and discuss the potentials and challenges of therapeutic interventions with chemokines.
Collapse
Affiliation(s)
- Anne Jaerve
- Molecular Neurobiology Laboratory, Department of Neurology, Medical Faculty Heinrich Heine University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | | |
Collapse
|
25
|
Risher WC, Eroglu C. Thrombospondins as key regulators of synaptogenesis in the central nervous system. Matrix Biol 2012; 31:170-7. [PMID: 22285841 DOI: 10.1016/j.matbio.2012.01.004] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2011] [Revised: 01/04/2012] [Accepted: 01/04/2012] [Indexed: 01/07/2023]
Abstract
Thrombospondins (TSPs) are a family of large, oligomeric multidomain glycoproteins that participate in a variety of biological functions as part of the extracellular matrix (ECM). Through their associations with a number of binding partners, TSPs mediate complex cell-cell and cell-matrix interactions in such diverse processes as angiogenesis, inflammation, osteogenesis, cell proliferation, and apoptosis. It was recently shown in the developing central nervous system (CNS) that TSPs promote the formation of new synapses, which are the unique cell-cell adhesions between neurons in the brain. This increase in synaptogenesis is mediated by the interaction between astrocyte-secreted TSPs and their neuronal receptor, calcium channel subunit α2δ-1. The cellular and molecular mechanisms that underlie induction of synaptogenesis via this interaction are yet to be fully elucidated. This review will focus on what is known about TSP and synapse formation during development, possible roles for TSP following brain injury, and what the previously established actions of TSP in other biological tissues may tell us about the mechanisms underlying TSP's functions in CNS synaptogenesis.
Collapse
Affiliation(s)
- W Christopher Risher
- Cell Biology Department, Duke University Medical Center, Durham, NC 27710, United States
| | | |
Collapse
|
26
|
Chew DJ, Fawcett JW, Andrews MR. The challenges of long-distance axon regeneration in the injured CNS. PROGRESS IN BRAIN RESEARCH 2012. [PMID: 23186719 DOI: 10.1016/b978-0-444-59544-7.00013-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Injury to the central nervous system (CNS) that results in long-tract axonal damage typically leads to permanent functional deficits in areas innervated at, and below, the level of the lesion. The initial ischemia, inflammation, and neurodegeneration are followed by a progressive generation of scar tissue, dieback of transected axons, and demyelination, creating an area inhibitory to regrowth and recovery. Two ways to combat this inhibition is to therapeutically target the extrinsic and intrinsic properties of the axon-scar environment. Scar tissue within and surrounding the lesion site can be broken down using an enzyme known as chondroitinase. Negative regulators of adult neuronal growth, such as Nogo, can be neutralized with antibodies. Both therapies greatly improve functional recovery in animal models. Alternatively, modifying the intrinsic growth properties of CNS neurons through gene therapy or pharmacotherapy has also shown promising axonal regeneration after injury. Despite these promising therapies, the main challenge of long-distance axon regeneration still remains; achieving a level of functional and organized connectivity below the level of the lesion that mimics the intact CNS.
Collapse
Affiliation(s)
- Daniel J Chew
- Centre for Brain Repair, University of Cambridge, Forvie Site, Robinson Way, Cambridge, UK
| | | | | |
Collapse
|
27
|
Myers SA, DeVries WH, Gruenthal MJ, Andres KR, Hagg T, Whittemore SR. Sildenafil improves epicenter vascular perfusion but not hindlimb functional recovery after contusive spinal cord injury in mice. J Neurotrauma 2011; 29:528-38. [PMID: 21970599 DOI: 10.1089/neu.2011.2036] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Nitric oxide (NO) is an important regulator of vasodilation and angiogenesis in the central nervous system (CNS). Signaling initiated by the membrane receptor CD47 antagonizes vasodilation and angiogenesis by inhibiting synthesis of cyclic guanosine monophosphate (cGMP). We recently found that deletion of CD47 led to significant functional locomotor improvements, enhanced angiogenesis, and increased epicenter microvascular perfusion in mice after moderate contusive spinal cord injury (SCI). We tested the hypothesis that improving NO/cGMP signaling within the spinal cord immediately after injury would increase microvascular perfusion, angiogenesis, and functional recovery, with an acute, 7-day administration of the cGMP phosphodiesterase 5 (PDE5) inhibitor sildenafil. PDE5 expression is localized within spinal cord microvascular endothelial cells and smooth muscle cells. While PDE5 antagonism has been shown to increase angiogenesis in a rat embolic stroke model, sildenafil had no significant effect on angiogenesis at 7 days post-injury after murine contusive SCI. Sildenafil treatment increased cGMP concentrations within the spinal cord and improved epicenter microvascular perfusion. Basso Mouse Scale (BMS) and Treadscan analyses revealed that sildenafil treatment had no functional consequence on hindlimb locomotor recovery. These data support the hypothesis that acutely improving microvascular perfusion within the injury epicenter by itself is an insufficient strategy for improving functional deficits following contusive SCI.
Collapse
Affiliation(s)
- Scott A Myers
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | | | | | | | | | | |
Collapse
|
28
|
Vascular Pathology as a Potential Therapeutic Target in SCI. Transl Stroke Res 2011; 2:556-74. [PMID: 24323683 DOI: 10.1007/s12975-011-0128-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Revised: 10/21/2011] [Accepted: 10/25/2011] [Indexed: 10/15/2022]
Abstract
Acute traumatic spinal cord injury (SCI) is characterized by a progressive secondary degeneration which exacerbates the loss of penumbral tissue and neurological function. Here, we first provide an overview of the known pathophysiological mechanisms involving injured microvasculature and molecular regulators that contribute to the loss and dysfunction of existing and new blood vessels. We also highlight the differences between traumatic and ischemic injuries which may yield clues as to the more devastating nature of traumatic injuries, possibly involving toxicity associated with hemorrhage. We also discuss known species differences with implications for choosing models, their relevance and utility to translate new treatments towards the clinic. Throughout this review, we highlight the potential opportunities and proof-of-concept experimental studies for targeting therapies to endothelial cell-specific responses. Lastly, we comment on the need for vascular mechanisms to be included in drug development and non-invasive diagnostics such as serum and cerebrospinal fluid biomarkers and imaging of spinal cord pathology.
Collapse
|
29
|
Durham-Lee JC, Wu Y, Mokkapati VUL, Paulucci-Holthauzen AA, Nesic O. Induction of angiopoietin-2 after spinal cord injury. Neuroscience 2011; 202:454-64. [PMID: 22020092 DOI: 10.1016/j.neuroscience.2011.09.058] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Revised: 09/22/2011] [Accepted: 09/22/2011] [Indexed: 01/27/2023]
Abstract
Angiopoietin-1 (Ang-1) and angiopoietin-2 (Ang-2) have opposing effects on blood vessels, with Ang-2 being mainly induced during the endothelial barrier breakdown. It is known that spinal cord injury (SCI) induces lasting decreases in Ang-1 levels, underlying endothelial barrier disruption, but the expression of Ang-2 in spinal cord injury has not been studied. We characterized Ang-2 after SCI using a clinically relevant rat model of contusion SCI. We found that SCI induces marked and persistent upregulation of Ang-2 (up to 10 weeks after SCI), which does not reflect well-characterized temporal profile of the blood-spinal cord barrier (BSCB) breakdown after SCI, and thus suggests other role(s) for Ang-2 in injured spinal cords. Furthermore, we also found that higher Ang-2 levels were associated with more successful locomotor recovery after SCI, both in SCI rats with markedly better spontaneous motor recovery and in SCI rats receiving a neuroprotective pharmacological intervention (amiloride), suggesting a beneficial role for Ang-2 in injured spinal cords. Immunocytochemical analyses revealed that Ang-2 was not induced in endothelial cells, but in perivascular and non-vascular cells labeled with glial fibrillary acidic protein (GFAP) or with chondroitin sulfate proteoglycan (NG2). Therefore, it is unlikely that induction of Ang-2 contributes to vascular dysfunction underlying functional impairment after SCI, but rather that it contributes to the beneficial pro-angiogenic and/or gliogenic processes underlying recovery processes after SCI.
Collapse
Affiliation(s)
- J C Durham-Lee
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555-1072, USA
| | | | | | | | | |
Collapse
|
30
|
Myers SA, DeVries WH, Andres KR, Gruenthal MJ, Benton RL, Hoying JB, Hagg T, Whittemore SR. CD47 knockout mice exhibit improved recovery from spinal cord injury. Neurobiol Dis 2010; 42:21-34. [PMID: 21168495 DOI: 10.1016/j.nbd.2010.12.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Revised: 12/01/2010] [Accepted: 12/10/2010] [Indexed: 11/15/2022] Open
Abstract
Recent data have implicated thrombospondin-1 (TSP-1) signaling in the acute neuropathological events that occur in microvascular endothelial cells (ECs) following spinal cord injury (SCI) (Benton et al., 2008b). We hypothesized that deletion of TSP-1 or its receptor CD47 would reduce these pathological events following SCI. CD47 is expressed in a variety of tissues, including vascular ECs and neutrophils. CD47 binds to TSP-1 and inhibits angiogenesis. CD47 also binds to the signal regulatory protein (SIRP)α and facilitates neutrophil diapedesis across ECs to sites of injury. After contusive SCI, TSP-1(-/-) mice did not show functional improvement compared to wildtype (WT) mice. CD47(-/-) mice, however, exhibited functional locomotor improvements and greater white matter sparing. Whereas targeted deletion of either CD47 or TSP-1 improved acute epicenter vascularity in contused mice, only CD47 deletion reduced neutrophil diapedesis and increased microvascular perfusion. An ex vivo model of the CNS microvasculature revealed that CD47(-/-)-derived microvessels (MVs) prominently exhibit adherent WT or CD47(-/-) neutrophils on the endothelial lumen, whereas WT-derived MVs do not. This implicates a defect in diapedesis mediated by the loss of CD47 expression on ECs. In vitro transmigration assays confirmed the role of SIRPα in neutrophil diapedesis through EC monolayers. We conclude that CD47 deletion modestly, but significantly, improves functional recovery from SCI via an increase in vascular patency and a reduction of SIRPα-mediated neutrophil diapedesis, rather than the abrogation of TSP-1-mediated anti-angiogenic signaling.
Collapse
Affiliation(s)
- Scott A Myers
- Kentucky Spinal Cord Injury Research Center, 511 S. Floyd St., Rm. 616A, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Huppert J, Closhen D, Croxford A, White R, Kulig P, Pietrowski E, Bechmann I, Becher B, Luhmann HJ, Waisman A, Kuhlmann CRW. Cellular mechanisms of IL-17-induced blood-brain barrier disruption. FASEB J 2009; 24:1023-34. [PMID: 19940258 DOI: 10.1096/fj.09-141978] [Citation(s) in RCA: 371] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Recently T-helper 17 (Th17) cells were demonstrated to disrupt the blood-brain barrier (BBB) by the action of IL-17A. The aim of the present study was to examine the mechanisms that underlie IL-17A-induced BBB breakdown. Barrier integrity was analyzed in the murine brain endothelial cell line bEnd.3 by measuring the electrical resistance values using electrical call impedance sensing technology. Furthermore, in-cell Western blots, fluorescence imaging, and monocyte adhesion and transendothelial migration assays were performed. Experimental autoimmune encephalomyelitis (EAE) was induced in C57BL/6 mice. IL-17A induced NADPH oxidase- or xanthine oxidase-dependent reactive oxygen species (ROS) production. The resulting oxidative stress activated the endothelial contractile machinery, which was accompanied by a down-regulation of the tight junction molecule occludin. Blocking either ROS formation or myosin light chain phosphorylation or applying IL-17A-neutralizing antibodies prevented IL-17A-induced BBB disruption. Treatment of mice with EAE using ML-7, an inhibitor of the myosin light chain kinase, resulted in less BBB disruption at the spinal cord and less infiltration of lymphocytes via the BBB and subsequently reduced the clinical characteristics of EAE. These observations indicate that IL-17A accounts for a crucial step in the development of EAE by impairing the integrity of the BBB, involving augmented production of ROS.-Huppert, J., Closhen, D., Croxford, A., White, R., Kulig, P., Pietrowski, E., Bechmann, I., Becher, B., Luhmann, H. J., Waisman, A., Kuhlmann, C. R. W. Cellular mechanisms of IL-17-induced blood-brain barrier disruption.
Collapse
Affiliation(s)
- Jula Huppert
- University Medical Center of the Johannes Gutenberg University Mainz, Institute of Physiology and Pathophysiology, Duesbergweg 6, 55131 Mainz, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Benton RL, Maddie MA, Dincman TA, Hagg T, Whittemore SR. Transcriptional activation of endothelial cells by TGFβ coincides with acute microvascular plasticity following focal spinal cord ischaemia/reperfusion injury. ASN Neuro 2009; 1:e00015. [PMID: 19663807 PMCID: PMC2810814 DOI: 10.1042/an20090008] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2009] [Revised: 08/07/2009] [Accepted: 08/10/2009] [Indexed: 11/17/2022] Open
Abstract
Microvascular dysfunction, loss of vascular support, ischaemia and sub-acute vascular instability in surviving blood vessels contribute to secondary injury following SCI (spinal cord injury). Neither the precise temporal profile of the cellular dynamics of spinal microvasculature nor the potential molecular effectors regulating this plasticity are well understood. TGFβ (transforming growth factor β) isoforms have been shown to be rapidly increased in response to SCI and CNS (central nervous system) ischaemia, but no data exist regarding their contribution to microvascular dysfunction following SCI. To examine these issues, in the present study we used a model of focal spinal cord ischaemia/reperfusion SCI to examine the cellular response(s) of affected microvessels from 30 min to 14 days post-ischaemia. Spinal endothelial cells were isolated from affected tissue and subjected to focused microarray analysis of TGFβ-responsive/related mRNAs 6 and 24 h post-SCI. Immunohistochemical analyses of histopathology show neuronal disruption/loss and astroglial regression from spinal microvessels by 3 h post-ischaemia, with complete dissolution of functional endfeet (loss of aquaporin-4) by 12 h post-ischaemia. Coincident with this microvascular plasticity, results from microarray analyses show 9 out of 22 TGFβ-responsive mRNAs significantly up-regulated by 6 h post-ischaemia. Of these, serpine 1/PAI-1 (plasminogen-activator inhibitor 1) demonstrated the greatest increase (>40-fold). Furthermore, uPA (urokinase-type plasminogen activator), another member of the PAS (plasminogen activator system), was also significantly increased (>7.5-fold). These results, along with other select up-regulated mRNAs, were confirmed biochemically or immunohistochemically. Taken together, these results implicate TGFβ as a potential molecular effector of the anatomical and functional plasticity of microvessels following SCI.
Collapse
Key Words
- endothelin
- insulin-like growth factor binding protein 3 (igfbp-3)
- interleukin-6 (il-6)
- matrix metalloproteinase 9 (mmp-9)
- plasminogen-activator inhibitor 1 (pai-1)
- urokinase-type plasminogen activator (upa)
- aqp-4, aquaporin-4
- bmp, bone morphogenetic protein
- bscb, blood-spinal cord-barrier
- cns, central nervous system
- ec, endothelial cell
- et, endothelin
- gfap, glial fibrillary acidic protein
- huvec, human umbilical vein endothelial cell
- igf, insulin-like growth factor
- igfbp-3, igf-binding protein 3
- il, interleukin
- lea, lycopersicon esculentum agglutinin
- llc, large latent complex
- map2, microtubule-associated protein 2
- mcao, middle cerebral artery occlusion
- mmp, matrix metalloproteinase
- nvu, neurovascular unit
- pa, plasminogen activator
- pai, pa inhibitor
- pas, pa system
- sci, spinal cord injury
- smvec, spinal microvascular ec
- tbs, tris-buffered saline
- tgfβ, transforming growth factor β
- tpa, tissue-type pa
- tsp-1, thrombospondin-1
- upa, urokinase-type pa
- upar, upa receptor
- vegf, vascular endothelial growth factor
Collapse
Affiliation(s)
- Richard L Benton
- daggerKentucky Spinal Cord Injury Research Center, Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, KY 40292, USA.
| | | | | | | | | |
Collapse
|
33
|
Benton RL, Maddie MA, Gruenthal MJ, Hagg T, Whittemore SR. Neutralizing endogenous VEGF following traumatic spinal cord injury modulates microvascular plasticity but not tissue sparing or functional recovery. Curr Neurovasc Res 2009; 6:124-31. [PMID: 19442162 DOI: 10.2174/156720209788185678] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Acute loss of spinal cord vascularity followed by an endogenous adaptive angiogenic response with concomitant microvascular dysfunction is a hallmark of traumatic spinal cord injury (SCI). Recently, the potent vasoactive factor vascular endothelial growth factor (VEGF) has received much attention as a putative therapeutic for the treatment of various neurodegenerative disorders, including SCI. Exogenous VEGF exerts both protective and destabilizing effects on microvascular elements and tissue following SCI but the role of endogenous VEGF is unclear. In the present study, we systemically applied a potent and well characterized soluble VEGF antagonist to adult C57Bl/6 mice post-SCI to elucidate the relative contribution of VEGF on the acute evolving microvascular response and its impact on functional recovery. While the VEGF Trap did not alter vascular density in the injury epicenter or penumbra, an overall increase in the number of Griffonia simplicifolia isolectin-B4 bound microvessels was observed, suggesting a VEGF-dependency to more subtle aspects of endothelial plasticity post-SCI. Neutralizing endogenous VEGF neither attenuated nor exacerbated chronic histopathology or functional recovery. These results support the idea that overall, endogenous VEGF is not neuroprotective or detrimental following traumatic SCI. Furthermore, they suggest that angiogenesis in traumatically injured spinal tissue is regulated by multiple effectors and is not limited by endogenous VEGF activation of affected spinal microvessels.
Collapse
Affiliation(s)
- Richard L Benton
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, KY 40292, USA.
| | | | | | | | | |
Collapse
|
34
|
Mahoney ET, Benton RL, Maddie MA, Whittemore SR, Hagg T. ADAM8 is selectively up-regulated in endothelial cells and is associated with angiogenesis after spinal cord injury in adult mice. J Comp Neurol 2009; 512:243-55. [PMID: 19003792 DOI: 10.1002/cne.21902] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Endothelial cell (EC) loss and subsequent angiogenesis occur over the first week after spinal cord injury (SCI). To identify molecular mechanisms that could be targeted with intravenous (i.v.) treatments, we determined whether transmembrane "a disintegrin and metalloprotease" (ADAM) proteins are expressed in ECs of the injured spinal cord. ADAMs bind to integrins, which are important for EC survival and angiogenesis. Female adult C57Bl/6 mice with a spinal cord contusion had progressively more ADAM8 (CD156) immunostaining in blood vessels and individual ECs between 1 and 28 days following injury. Uninjured spinal cords had little ADAM8 staining. The increase in ADAM8 mRNA and protein was confirmed in spinal cord lysates, and ADAM8 mRNA was present in FACS-enriched ECs. ADAM8 colocalized extensively and exclusively with the EC marker PECAM and also with i.v.-injected lectins. Intravenous isolectin B4 (IB4) labels a subpopulation of blood vessels at and within the injury epicenter 3-7 days after injury, coincident with angiogenesis. Both ADAM8 and the proliferation marker Ki-67 were present in IB4-positive microvessels. ADAM8-positive proliferating cells were seen at the leading end of IB4-positive blood vessels. Angiogenesis was confirmed by BrdU incorporation, binding of i.v.-injected nucleolin antibodies, and MT1-MMP immunostaining in a subset of blood vessels. These data suggest that ADAM8 is vascular selective and plays a role in proliferation and/or migration of ECs during angiogenesis following SCI.
Collapse
Affiliation(s)
- Edward T Mahoney
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, Kentucky 40292, USA
| | | | | | | | | |
Collapse
|