1
|
Ganguly K, Adhikary K, Acharjee A, Acharjee P, Trigun SK, Mutlaq AS, Ashique S, Yasmin S, Alshahrani AM, Ansari MY. Biological significance and pathophysiological role of Matrix Metalloproteinases in the Central Nervous System. Int J Biol Macromol 2024; 280:135967. [PMID: 39322129 DOI: 10.1016/j.ijbiomac.2024.135967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 09/21/2024] [Accepted: 09/21/2024] [Indexed: 09/27/2024]
Abstract
Matrix Metalloproteinases (MMPs), which are endopeptidase reliant on zinc, are low in embryonic tissues but increases in response to a variety of physiological stimulus and pathological stresses. Neuro-glial cells, endothelial cells, fibroblasts, and leucocytes secrete MMPs, which cleave extracellular matrix proteins in a time-dependent manner. MMPs affect synaptic plasticity and the development of short-term memory by controlling the size, shape, and excitatory synapses' function through the lateral diffusion of receptors. In addition, MMPs influence the Extracellular Matrix proteins in the Peri-Neuronal Net at the Neuro-glial interface, which aids in the establishment of long-term memory. Through modulating neuronal, and glial cells migration, differentiation, Neurogenesis, and survival, MMPs impact brain development in mammals. In adult brains, MMPs play a beneficial role in physiological plasticity, which includes learning, memory consolidation, social interaction, and complex behaviors, by proteolytically altering a wide variety of factors, including growth factors, cytokines, receptors, DNA repair enzymes, and matrix proteins. Additionally, stress, depression, addiction, hepatic encephalopathy, and stroke may all have negative effects on MMPs. In addition to their role in glioblastoma development, MMPs influence neurological diseases such as epilepsy, schizophrenia, autism spectrum disorder, brain damage, pain, neurodegeneration, and Alzheimer's and Parkinson's. To help shed light on the potential of MMPs as a therapeutic target for neurodegenerative diseases, this review summarizes their regulation, mode of action, and participation in brain physiological plasticity and pathological damage. Finally, by employing different MMP-based nanotools and inhibitors, MMPs may also be utilized to map the anatomical and functional connectome of the brain, analyze its secretome, and treat neurodegenerative illnesses.
Collapse
Affiliation(s)
- Krishnendu Ganguly
- Department of Medical Lab Technology, Paramedical College Durgapur, Helen Keller Sarani, Durgapur 713212, West Bengal, India.
| | - Krishnendu Adhikary
- Department of Medical Lab Technology, Paramedical College Durgapur, Helen Keller Sarani, Durgapur 713212, West Bengal, India.
| | - Arup Acharjee
- Molecular Omics Laboratory, Department of Zoology, University of Allahabad, Allahabad, Uttar Pradesh, India.
| | - Papia Acharjee
- Biochemistry Section, Department of Zoology, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India.
| | - Surendra Kumar Trigun
- Biochemistry Section, Department of Zoology, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India.
| | | | - Sumel Ashique
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Department of Pharmaceutics, Bengal College of Pharmaceutical Sciences & Research, Durgapur 713212, West Bengal, India.
| | - Sabina Yasmin
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia.
| | - Asma M Alshahrani
- Department of Clinical Pharmacy, Faculty of Pharmacy, King Khalid University, Abha, Saudi Arabia; Department of Clinical Pharmacy, Shaqra University, Saudi Arabia.
| | - Mohammad Yousuf Ansari
- MM college of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana 133207, India.
| |
Collapse
|
2
|
Almarghalani DA, Shah ZA. Progress on siRNA-based gene therapy targeting secondary injury after intracerebral hemorrhage. Gene Ther 2023; 30:1-7. [PMID: 34754099 PMCID: PMC10927018 DOI: 10.1038/s41434-021-00304-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/25/2021] [Accepted: 10/27/2021] [Indexed: 12/13/2022]
Abstract
Intracerebral hemorrhage (ICH) is a life-threatening condition with a high mortality rate. For survivors, quality of life is determined by primary and secondary phases of injury. The prospects for injury repair and recovery after ICH are highly dependent on the extent of secondary injury. Currently, no effective treatments are available to prevent secondary injury or its long-term effects. One promising strategy that has recently garnered attention is gene therapy, in particular, small interfering RNAs (siRNA), which silence specific genes responsible for destructive effects after hemorrhage. Gene therapy as a potential treatment for ICH is being actively researched in animal studies. However, there are many barriers to the systemic delivery of siRNA-based therapy, as the use of naked siRNA has limitations. Recently, the Food and Drug Administration approved two siRNA-based therapies, and several are undergoing Phase 3 clinical trials. In this review, we describe the advancements in siRNA-based gene therapy for ICH and also summarize its advantages and disadvantages.
Collapse
Affiliation(s)
- Daniyah A Almarghalani
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH, 43614, USA
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, 43614, USA
| | - Zahoor A Shah
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, 43614, USA.
| |
Collapse
|
3
|
Yang C, Wu J, Lu X, Xiong S, Xu X. Identification of novel biomarkers for intracerebral hemorrhage via long noncoding RNA-associated competing endogenous RNA network. Mol Omics 2021; 18:71-82. [PMID: 34807207 DOI: 10.1039/d1mo00298h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Intracerebral hemorrhage (ICH) is a leading cause of death and disability worldwide. This study aimed to examine the involvement of long non-coding RNAs (lncRNAs), a group of non-coding transcripts, in ICH as potential biomarkers. An expression profile of patients with ICH using four contralateral grey matter controls (GM) and four contralateral white matter controls (WM) was downloaded from the Gene Expression Omnibus (GEO) database. Co-expressed lncRNAs and mRNAs were selected to create competing endogenous RNA (ceRNA) networks. Key lncRNAs were identified in ceRNA networks, which were validated through Real-time qPCR (RT-qPCR) with peripheral blood samples from patients with ICH. A total of 49 differentially expressed lncRNAs were discovered in different brain regions. The ceRNA network in GM included 9 lncRNAs, 40 mRNAs, and 20 microRNAs (miRNAs), while the one in WM covered 6 lncRNAs, 25 mRNAs, and 14 miRNAs. Six hub lncRNAs were observed and RT-qPCR results showed that LY86-AS1, DLX6-AS1, RRN3P2, and CRNDE were down-regulated, while HCP5 and MIAT were up-regulated in patients with ICH. Receiver Operating Characteristic (ROC) assessments demonstrated the diagnostic value of these lncRNAs. Our findings highlight the potential roles of lncRNA in ICH pathogenesis. Moreover, the hub lncRNAs discovered here might become novel biomarkers and promising targets for ICH drug development.
Collapse
Affiliation(s)
- Chunyu Yang
- Department of Neurology, the First Hospital of China Medical University, No 155, Nanjing Street, Heping District, Shenyang, Liaoning, 110001, China. .,Department of Pharmacy, The Fourth Hospital of China Medical University, Shenyang, China
| | - Jiao Wu
- Department of Neurology, The People's Hospital of Liaoning Province, Shenyang, China
| | - Xi Lu
- Department of Public Health, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Shuang Xiong
- Liaoning Academy of Analytic Science, Construction Engineering Center of Important Technology Innovation and Research and Development Base in Liaoning Province, Shenyang, China
| | - Xiaoxue Xu
- Department of Neurology, the First Hospital of China Medical University, No 155, Nanjing Street, Heping District, Shenyang, Liaoning, 110001, China.
| |
Collapse
|
4
|
Bi R, Fang Z, You M, He Q, Hu B. Microglia Phenotype and Intracerebral Hemorrhage: A Balance of Yin and Yang. Front Cell Neurosci 2021; 15:765205. [PMID: 34720885 PMCID: PMC8549831 DOI: 10.3389/fncel.2021.765205] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 09/23/2021] [Indexed: 11/15/2022] Open
Abstract
Intracerebral hemorrhage (ICH) features extremely high rates of morbidity and mortality, with no specific and effective therapy. And local inflammation caused by the over-activated immune cells seriously damages the recovery of neurological function after ICH. Fortunately, immune intervention to microglia has provided new methods and ideas for ICH treatment. Microglia, as the resident immune cells in the brain, play vital roles in both tissue damage and repair processes after ICH. The perihematomal activated microglia not only arouse acute inflammatory responses, oxidative stress, excitotoxicity, and cytotoxicity to cause neuron death, but also show another phenotype that inhibit inflammation, clear hematoma and promote tissue regeneration. The proportion of microglia phenotypes determines the progression of brain tissue damage or repair after ICH. Therefore, microglia may be a promising and imperative therapeutic target for ICH. In this review, we discuss the dual functions of microglia in the brain after an ICH from immunological perspective, elaborate on the activation mechanism of perihematomal microglia, and summarize related therapeutic drugs researches.
Collapse
Affiliation(s)
- Rentang Bi
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhi Fang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mingfeng You
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Quanwei He
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
5
|
Central Nervous System Tissue Regeneration after Intracerebral Hemorrhage: The Next Frontier. Cells 2021; 10:cells10102513. [PMID: 34685493 PMCID: PMC8534252 DOI: 10.3390/cells10102513] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/13/2021] [Accepted: 09/17/2021] [Indexed: 12/11/2022] Open
Abstract
Despite marked advances in surgical techniques and understanding of secondary brain injury mechanisms, the prognosis of intracerebral hemorrhage (ICH) remains devastating. Harnessing and promoting the regenerative potential of the central nervous system may improve the outcomes of patients with hemorrhagic stroke, but approaches are still in their infancy. In this review, we discuss the regenerative phenomena occurring in animal models and human ICH, provide results related to cellular and molecular mechanisms of the repair process including by microglia, and review potential methods to promote tissue regeneration in ICH. We aim to stimulate research involving tissue restoration after ICH.
Collapse
|
6
|
Neuroprotective Therapies for Spontaneous Intracerebral Hemorrhage. Neurocrit Care 2021; 35:862-886. [PMID: 34341912 DOI: 10.1007/s12028-021-01311-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 06/25/2021] [Indexed: 12/15/2022]
Abstract
Patients who survive the initial ictus of spontaneous intracerebral hemorrhage (ICH) remain vulnerable to subsequent injury of the perilesional parenchyma by molecular and cellular responses to the hematoma. Secondary brain injury after ICH, which contributes to long-term functional impairment and mortality, has emerged as an attractive therapeutic target. This review summarizes preclinical and clinical evidence for neuroprotective therapies targeting secondary injury pathways following ICH. A focus on therapies with pleiotropic antiinflammatory effects that target thrombin-mediated chemotaxis and inflammatory cell migration has led to studies investigating statins, anticholinergics, sphingosine-1-phosphate receptor modulators, peroxisome proliferator activated receptor gamma agonists, and magnesium. Attempts to modulate ICH-induced blood-brain barrier breakdown and perihematomal edema formation has prompted studies of nonsteroidal antiinflammatory agents, matrix metalloproteinase inhibitors, and complement inhibitors. Iron chelators, such as deferoxamine and albumin, have been used to reduce the free radical injury that ensues from erythrocyte lysis. Stem cell transplantation has been assessed for its potential to enhance subacute neurogenesis and functional recovery. Despite promising preclinical results of numerous agents, their outcomes have not yet translated into positive clinical trials in patients with ICH. Further studies are necessary to improve our understanding of the molecular events that promote damage and inflammation of the perihematomal parenchyma after ICH. Elucidating the temporal and pathophysiologic features of this secondary brain injury could enhance the clinical efficacy of neuroprotective therapies for ICH.
Collapse
|
7
|
Sun Q, Xu X, Wang T, Xu Z, Lu X, Li X, Chen G. Neurovascular Units and Neural-Glia Networks in Intracerebral Hemorrhage: from Mechanisms to Translation. Transl Stroke Res 2021; 12:447-460. [PMID: 33629275 DOI: 10.1007/s12975-021-00897-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 02/07/2021] [Accepted: 02/09/2021] [Indexed: 12/20/2022]
Abstract
Intracerebral hemorrhage (ICH), the most lethal type of stroke, often leads to poor outcomes in the clinic. Due to the complex mechanisms and cell-cell crosstalk during ICH, the neurovascular unit (NVU) was proposed to serve as a promising therapeutic target for ICH research. This review aims to summarize the development of pathophysiological shifts in the NVU and neural-glia networks after ICH. In addition, potential targets for ICH therapy are discussed in this review. Beyond cerebral blood flow, the NVU also plays an important role in protecting neurons, maintaining central nervous system (CNS) homeostasis, coordinating neuronal activity among supporting cells, forming and maintaining the blood-brain barrier (BBB), and regulating neuroimmune responses. During ICH, NVU dysfunction is induced, along with neuronal cell death, microglia and astrocyte activation, endothelial cell (EC) and tight junction (TJ) protein damage, and BBB disruption. In addition, it has been shown that certain targets and candidates can improve ICH-induced secondary brain injury based on an NVU and neural-glia framework. Moreover, therapeutic approaches and strategies for ICH are discussed.
Collapse
Affiliation(s)
- Qing Sun
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, China
| | - Xiang Xu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, China
| | - Tianyi Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, China
| | - Zhongmou Xu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, China
| | - Xiaocheng Lu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, China.
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, China.
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, China
| |
Collapse
|
8
|
Cabral-Pacheco GA, Garza-Veloz I, Castruita-De la Rosa C, Ramirez-Acuña JM, Perez-Romero BA, Guerrero-Rodriguez JF, Martinez-Avila N, Martinez-Fierro ML. The Roles of Matrix Metalloproteinases and Their Inhibitors in Human Diseases. Int J Mol Sci 2020; 21:E9739. [PMID: 33419373 PMCID: PMC7767220 DOI: 10.3390/ijms21249739] [Citation(s) in RCA: 866] [Impact Index Per Article: 173.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/10/2020] [Accepted: 12/18/2020] [Indexed: 02/07/2023] Open
Abstract
Matrix metalloproteinases (MMPs) are a family of zinc-dependent extracellular matrix (ECM) remodeling endopeptidases that have the capacity to degrade almost every component of the ECM. The degradation of the ECM is of great importance, since it is related to embryonic development and angiogenesis. It is also involved in cell repair and the remodeling of tissues. When the expression of MMPs is altered, it can generate the abnormal degradation of the ECM. This is the initial cause of the development of chronic degenerative diseases and vascular complications generated by diabetes. In addition, this process has an association with neurodegeneration and cancer progression. Within the ECM, the tissue inhibitors of MMPs (TIMPs) inhibit the proteolytic activity of MMPs. TIMPs are important regulators of ECM turnover, tissue remodeling, and cellular behavior. Therefore, TIMPs (similar to MMPs) modulate angiogenesis, cell proliferation, and apoptosis. An interruption in the balance between MMPs and TIMPs has been implicated in the pathophysiology and progression of several diseases. This review focuses on the participation of both MMPs (e.g., MMP-2 and MMP-9) and TIMPs (e.g., TIMP-1 and TIMP-3) in physiological processes and on how their abnormal regulation is associated with human diseases. The inclusion of current strategies and mechanisms of MMP inhibition in the development of new therapies targeting MMPs was also considered.
Collapse
Affiliation(s)
| | - Idalia Garza-Veloz
- Molecular Medicine Laboratory, Unidad Académica de Medicina Humana y Ciencias de la Salud, Carretera Zacatecas-Guadalajara Km.6. Ejido la Escondida, Zacatecas 98160, Mexico; (G.AC.-P.); (C.C.-D.l.R.); (J.MR.-A.); (B.AP.-R.); (J.FG.-R.); (N.M.-A.)
| | | | | | | | | | | | - Margarita L Martinez-Fierro
- Molecular Medicine Laboratory, Unidad Académica de Medicina Humana y Ciencias de la Salud, Carretera Zacatecas-Guadalajara Km.6. Ejido la Escondida, Zacatecas 98160, Mexico; (G.AC.-P.); (C.C.-D.l.R.); (J.MR.-A.); (B.AP.-R.); (J.FG.-R.); (N.M.-A.)
| |
Collapse
|
9
|
Gao C, Meng Y, Chen G, Chen W, Chen XS, Luo CL, Zhang MY, Wang ZF, Wang T, Tao LY. Chronic restraint stress exacerbates neurological deficits and disrupts the remodeling of the neurovascular unit in a mouse intracerebral hemorrhage model. Stress 2020; 23:338-348. [PMID: 31591949 DOI: 10.1080/10253890.2019.1678023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Growing evidences have shown that patients recovering from stroke experience high and unremitting stress. Chronic restraint stress (CRS) has been found to exacerbate neurological impairments in an experimental focal cortical ischemia model. However, there have been no studies reporting the effect and mechanism of CRS on intracerebral hemorrhage (ICH). This study aimed to evaluate the effect of CRS on a mouse ICH model. Adult male C57BL mice were subjected to infusion of collagenase IV (to induce ICH) or saline (for sham) into the left striatum. After ICH, animals were stressed with application of CRS protocol for 21 days. Our results showed that CRS significantly exacerbated neurological deficits (Garcia test, corner turn test, and wire grip test) and the ipsilateral brain atrophy and reduced body weight gain after ICH. Immunofluorescence staining indicated that CRS exerted significant suppressive effects on neuron, astrocyte, vascular endothelial cell and pericyte and excessively activated microglia post ICH. All of the key cellular components mentioned above are involved in the neurovascular unit (NVU) remodeling in the peri-hemorrhagic region after ICH. Western blot results showed that matrix metalloproteinase (MMP)-9 and tight junction (TJ) proteins including zonula occludens-1, occludin and claudin-5 were increased after ICH, but MMP-9 protein was further up-regulated and TJ-related proteins were down-regulated by CRS. In addition, ICH-induced activation of endoplasmic reticulum stress and apoptosis were further strengthened by CRS. Collectively, CRS exacerbates neurological deficits and disrupts the remodeling of the peri-hemorrhagic NVU after ICH, which may be associated with TJ proteins degradation and excessive activation of MMP-9 and endoplasmic reticulum stress-apoptosis.LAY SUMMARYCRS exacerbates neurological deficits and disrupts the remodeling of the NVU in the recovery stage after ICH, which suggest that monitoring chronic stress levels in patients recovering from ICH may merit consideration in the future.
Collapse
Affiliation(s)
- Cheng Gao
- Shanghai Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Academy of Forensic Science, Shanghai, China
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou, China
| | - Ying Meng
- Community Health Center, Suzhou Western Eco-City, Suzhou, China
| | - Guang Chen
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou, China
| | - Wei Chen
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou, China
| | - Xue-Shi Chen
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou, China
| | - Cheng-Liang Luo
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou, China
| | - Ming-Yang Zhang
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou, China
| | - Zu-Feng Wang
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou, China
| | - Tao Wang
- Shanghai Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Academy of Forensic Science, Shanghai, China
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou, China
- School of Pharmacy, Soochow University, Suzhou, China
| | - Lu-Yang Tao
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou, China
| |
Collapse
|
10
|
Lattanzi S, Di Napoli M, Ricci S, Divani AA. Matrix Metalloproteinases in Acute Intracerebral Hemorrhage. Neurotherapeutics 2020; 17:484-496. [PMID: 31975152 PMCID: PMC7283398 DOI: 10.1007/s13311-020-00839-0] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Spontaneous intracerebral hemorrhage (ICH) accounts for 10-30% of all strokes and affects more than one million people every year worldwide, and it is the stroke subtype associated with the highest rates of mortality and residual disability. So far, clinical trials have mainly targeted primary cerebral injury and have substantially failed to improve clinical outcomes. The understanding of the pathophysiology of early and delayed injury after ICH is, hence, of paramount importance to identify potential targets of intervention and develop effective therapeutic strategies. Matrix metalloproteinases (MMPs) represent a ubiquitous superfamily of structurally related zinc-dependent endopeptidases able to degrade any component of the extracellular matrix. They are upregulated after ICH, in which different cell types, including leukocytes, activated microglia, neurons, and endothelial cells, are involved in their synthesis and secretion. The aim of this review is to summarize the available experimental and clinical evidence about the role of MMPs in brain injury following spontaneous ICH and provide critical insights into the underlying mechanisms.
Collapse
Affiliation(s)
- Simona Lattanzi
- Neurological Clinic, Department of Experimental and Clinical Medicine, Marche Polytechnic University, Ancona, Italy
| | - Mario Di Napoli
- Department of Neurology and Stroke Unit, San Camillo de' Lellis District General Hospital, Rieti, Italy
| | - Silvia Ricci
- Department of Neurology and Stroke Unit, San Camillo de' Lellis District General Hospital, Rieti, Italy
| | - Afshin A Divani
- Department of Neurology, University of New Mexico, Albuquerque, New Mexico, USA.
| |
Collapse
|
11
|
Zhu H, Wang Z, Yu J, Yang X, He F, Liu Z, Che F, Chen X, Ren H, Hong M, Wang J. Role and mechanisms of cytokines in the secondary brain injury after intracerebral hemorrhage. Prog Neurobiol 2019; 178:101610. [PMID: 30923023 DOI: 10.1016/j.pneurobio.2019.03.003] [Citation(s) in RCA: 219] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 03/07/2019] [Accepted: 03/16/2019] [Indexed: 12/18/2022]
Abstract
Intracerebral hemorrhage (ICH) is a common and severe cerebrovascular disease that has high mortality. Few survivors achieve self-care. Currently, patients receive only symptomatic treatment for ICH and benefit poorly from this regimen. Inflammatory cytokines are important participants in secondary injury after ICH. Increases in proinflammatory cytokines may aggravate the tissue injury, whereas increases in anti-inflammatory cytokines might be protective in the ICH brain. Inflammatory cytokines have been studied as therapeutic targets in a variety of acute and chronic brain diseases; however, studies on ICH are limited. This review summarizes the roles and functions of various pro- and anti-inflammatory cytokines in secondary brain injury after ICH and discusses pathogenic mechanisms and emerging therapeutic strategies and directions for treatment of ICH.
Collapse
Affiliation(s)
- Huimin Zhu
- Department of Neurology, Linyi People's Hospital, Linyi, Shandong 276003, China
| | - Zhiqiang Wang
- Central laboratory, Linyi People's Hospital, Linyi, Shandong 276003, China
| | - Jixu Yu
- Department of Neurology, Linyi People's Hospital, Linyi, Shandong 276003, China; Central laboratory, Linyi People's Hospital, Linyi, Shandong 276003, China; Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Xiuli Yang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Feng He
- Department of Neurology, Linyi People's Hospital, Linyi, Shandong 276003, China
| | - Zhenchuan Liu
- Department of Neurology, Linyi People's Hospital, Linyi, Shandong 276003, China.
| | - Fengyuan Che
- Department of Neurology, Linyi People's Hospital, Linyi, Shandong 276003, China; Central laboratory, Linyi People's Hospital, Linyi, Shandong 276003, China.
| | - Xuemei Chen
- Department of Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, Henan, China
| | - Honglei Ren
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Michael Hong
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jian Wang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
12
|
Li X, Feng D, Chen G. An Update On Medical Treatment for Intracerebral Hemorrhage. Transl Stroke Res 2018; 9:10.1007/s12975-018-0664-5. [PMID: 30206812 DOI: 10.1007/s12975-018-0664-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 09/04/2018] [Indexed: 12/26/2022]
Affiliation(s)
- Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, China
| | - Dongxia Feng
- Department of Neurosurgery, Baylor Scott & White Medical Center, Texas A&M University College of Medicine, Temple, 76508, USA
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, China.
| |
Collapse
|
13
|
Hsu WC, Yu CH, Kung WM, Huang KF. Enhancement of matrix metalloproteinases 2 and 9 accompanied with neurogenesis following collagen glycosaminoglycan matrix implantation after surgical brain injury. Neural Regen Res 2018; 13:1007-1012. [PMID: 29926827 PMCID: PMC6022476 DOI: 10.4103/1673-5374.233443] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/02/2018] [Indexed: 02/05/2023] Open
Abstract
Surgical brain injury may result in irreversible neurological deficits. Our previous report showed that partial regeneration of a traumatic brain lesion is achieved by implantation of collagen glycosaminoglycan (CGM). Matrix metalloproteinases (MMPs) may play an important role in neurogenesis but there is currently a lack of studies displaying the relationship between the stimulation of MMPs and neurogenesis after collagen glycosaminoglycan implantation following surgical brain trauma. The present study was carried out to further examine the expression of MMP2 and MMP9 after implantation of collagen glycosaminoglycan (CGM) following surgical brain trauma. Using the animal model of surgically induced brain lesion, we implanted CGM into the surgical trauma. Rats were thus divided into three groups: (1) sham operation group: craniotomy only; (2) lesion (L) group: craniotomy + surgical trauma lesion; (3) lesion + CGM (L + CGM) group: CGM implanted following craniotomy and surgical trauma lesion. Cells positive for SOX2 (marker of proliferating neural progenitor cells) and matrix metalloproteinases (MMP2 and MMP9) in the lesion boundary zone were assayed and analyzed by immunofluorescence and ELISA commercial kits, respectively. Our results demonstrated that following implantation of CGM after surgical brain trauma, significant increases in MMP2+/SOX2+ cells and MMP9+/SOX2+ cells were seen within the lesion boundary zone in the L + CGM group. Tissue protein concentrations of MMP2 and MMP9 also increased after CGM scaffold implantation. These findings suggest that implantation of a CGM scaffold alone after surgical brain trauma can enhance the expression of MMP2 and MMP9 accompanied by neurogenesis.
Collapse
Affiliation(s)
- Wei-Cherng Hsu
- Department of Ophthalmology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan, China
- School of Medicine, Buddhist Tzu Chi University, Hualien, Taiwan, China
| | - Chun-Hsien Yu
- School of Medicine, Buddhist Tzu Chi University, Hualien, Taiwan, China
- Department of Pediatrics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan, China
| | - Woon-Man Kung
- Division of Neurosurgery, Department of Surgery, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan, China
- Department of Exercise and Health Promotion, College of Education, Chinese Culture University, Taipei, Taiwan, China
| | - Kuo-Feng Huang
- School of Medicine, Buddhist Tzu Chi University, Hualien, Taiwan, China
- Division of Neurosurgery, Department of Surgery, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan, China
| |
Collapse
|
14
|
Xie Y, Yu N, Chen Y, Zhang K, Ma HY, Di Q. HMGB1 regulates P-glycoprotein expression in status epilepticus rat brains via the RAGE/NF-κB signaling pathway. Mol Med Rep 2017. [PMID: 28627626 PMCID: PMC5562060 DOI: 10.3892/mmr.2017.6772] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Overexpression of P-glycoprotein (P-gp) in the brain is an important mechanism involved in drug-resistant epilepsy (DRE). High-mobility group box 1 (HMGB1), an inflammatory cytokine, significantly increases following seizures and may be involved in upregulation of P-gp. However, the underlying mechanisms remain elusive. The aim of the present study was to evaluate the role of HMGB1 and its downstream signaling components, receptor for advanced glycation end-product (RAGE) and nuclear factor-κB (NF-κB), on P-gp expression in rat brains during status epilepticus (SE). Small interfering RNA (siRNA) was administered to rats prior to induction of SE by pilocarpine, to block transcription of the genes encoding HMGB1 and RAGE, respectively. An inhibitor of NF-κB, pyrrolidinedithiocarbamic acid (PDTC), was utilized to inhibit activation of NF-κB. The expression levels of HMGB1, RAGE, phosphorylated-NF-κB p65 (p-p65) and P-gp were detected by western blotting. The relative mRNA expression levels of the genes encoding these proteins were measured using reverse transcription-quantitative polymerase chain reaction and the cellular localization of the proteins was determined by immunofluorescence. Pre-treatment with HMGB1 siRNA reduced the expression levels of RAGE, p-p65 and P-gp. PDTC reduced the expression levels of P-gp. These findings suggested that overexpression of P-gp during seizures may be regulated by HMGB1 via the RAGE/NF-κB signaling pathway, and may be a novel target for treating DRE.
Collapse
Affiliation(s)
- Yuan Xie
- Department of Neurology, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Nian Yu
- Department of Neurology, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yan Chen
- Department of Neurology, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Kang Zhang
- Department of Neurology, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Hai-Yan Ma
- Department of Neurology, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Qing Di
- Department of Neurology, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
15
|
Wang S, Zhou Y, Yang B, Li L, Yu S, Chen Y, Zhu J, Zhao Y. C1q/Tumor Necrosis Factor-Related Protein-3 Attenuates Brain Injury after Intracerebral Hemorrhage via AMPK-Dependent Pathway in Rat. Front Cell Neurosci 2016; 10:237. [PMID: 27807406 PMCID: PMC5069420 DOI: 10.3389/fncel.2016.00237] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 09/29/2016] [Indexed: 12/03/2022] Open
Abstract
C1q/tumor necrosis factor (TNF)-related protein-3 (CTRP3) is a recently discovered adiponectin paralog with established metabolic regulatory properties. However, the role of CTRP3 in intracerebral hemorrhage (ICH) is still mostly unresolved. The aim of the present report was to explore the possible neuroprotective effect of CTRP3 in an ICH rat model and to elucidate the fundamental mechanisms. ICH was induced in rats by intracerebral infusion of autologous arterial blood. The effects of exogenous CTRP3 (recombinant or lentivirus CTRP3) on brain injury were explored on day 7. Treatment with CTRP3 reduced brain edema, protected against disruption of the blood-brain barrier (BBB), improved neurological functions and promoted angiogenesis. Furthermore, CTRP3 greatly intensified phosphorylation of AMP-activated protein kinase (AMPK) in addition to expression of hypoxia inducing factor-1α (HIF-1α) and vascular endothelial growth factor (VEGF). Finally, the protective effects of CTRP3 could be blocked by either AMPK or VEGF inhibitors. Our findings give the first evidence that CTRP3 is a new proangiogenic and neuroprotective adipokine, which may exert its protective effects at least partly through an AMPK/HIF-1α/ VEGF-dependent pathway, and suggest that CTRP3 may provide a new therapeutic strategy for ICH.
Collapse
Affiliation(s)
- Shaohua Wang
- Department of Pathology, Chongqing Medical UniversityChongqing, China; Key Laboratory of Neurobiology, Chongqing Medical UniversityChongqing, China
| | - Yang Zhou
- Department of Pathology, Chongqing Medical UniversityChongqing, China; Key Laboratory of Neurobiology, Chongqing Medical UniversityChongqing, China
| | - Bo Yang
- Department of Pathology, Chongqing Medical UniversityChongqing, China; Key Laboratory of Neurobiology, Chongqing Medical UniversityChongqing, China
| | - Lingyu Li
- Department of Pathology, Chongqing Medical UniversityChongqing, China; Key Laboratory of Neurobiology, Chongqing Medical UniversityChongqing, China
| | - Shanshan Yu
- Department of Pathology, Chongqing Medical UniversityChongqing, China; Key Laboratory of Neurobiology, Chongqing Medical UniversityChongqing, China
| | - Yanlin Chen
- Department of Pathology, Chongqing Medical UniversityChongqing, China; Key Laboratory of Neurobiology, Chongqing Medical UniversityChongqing, China
| | - Jin Zhu
- Department of Pathology, Chongqing Medical UniversityChongqing, China; Key Laboratory of Neurobiology, Chongqing Medical UniversityChongqing, China
| | - Yong Zhao
- Department of Pathology, Chongqing Medical UniversityChongqing, China; Key Laboratory of Neurobiology, Chongqing Medical UniversityChongqing, China
| |
Collapse
|
16
|
Xiong XY, Liu L, Yang QW. Functions and mechanisms of microglia/macrophages in neuroinflammation and neurogenesis after stroke. Prog Neurobiol 2016; 142:23-44. [PMID: 27166859 DOI: 10.1016/j.pneurobio.2016.05.001] [Citation(s) in RCA: 506] [Impact Index Per Article: 56.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 04/20/2016] [Accepted: 05/01/2016] [Indexed: 02/08/2023]
Abstract
Microglia/macrophages are the major immune cells involved in the defence against brain damage. Their morphology and functional changes are correlated with the release of danger signals induced by stroke. These cells are normally responsible for clearing away dead neural cells and restoring neuronal functions. However, when excessively activated by the damage-associated molecular patterns following stroke, they can produce a large number of proinflammatory cytokines that can disrupt neural cells and the blood-brain barrier and influence neurogenesis. These effects indicate the important roles of microglia/macrophages in the pathophysiological processes of stroke. However, the modifiable and adaptable nature of microglia/macrophages may also be beneficial for brain repair and not just result in damage. These distinct roles may be attributed to the different microglia/macrophage phenotypes because the M1 population is mainly destructive, while the M2 population is neuroprotective. Additionally, different gene expression signature changes in microglia/macrophages have been found in diverse inflammatory milieus. These biofunctional features enable dual roles for microglia/macrophages in brain damage and repair. Currently, it is thought that the proper inflammatory milieu may provide a suitable microenvironment for neurogenesis; however, detailed mechanisms underlying the inflammatory responses that initiate or inhibit neurogenesis remain unknown. This review summarizes recent progress concerning the mechanisms involved in brain damage, repair and regeneration related to microglia/macrophage activation and phenotype transition after stroke. We also argue that future translational studies should be targeting multiple key regulating molecules to improve brain repair, which should be accompanied by the concept of a "therapeutic time window" for sequential therapies.
Collapse
Affiliation(s)
- Xiao-Yi Xiong
- Department of Neurology, Xinqiao Hospital & The Second Affiliated Hospital, The Third Military Medical University, Xinqiao zhengjie No.183, Shapingba District Chongqing, 400037, China
| | - Liang Liu
- Department of Neurology, Xinqiao Hospital & The Second Affiliated Hospital, The Third Military Medical University, Xinqiao zhengjie No.183, Shapingba District Chongqing, 400037, China
| | - Qing-Wu Yang
- Department of Neurology, Xinqiao Hospital & The Second Affiliated Hospital, The Third Military Medical University, Xinqiao zhengjie No.183, Shapingba District Chongqing, 400037, China.
| |
Collapse
|
17
|
Strecker JK, Olk J, Hoppen M, Gess B, Diederich K, Schmidt A, Schäbitz WR, Schilling M, Minnerup J. Combining Growth Factor and Bone Marrow Cell Therapy Induces Bleeding and Alters Immune Response After Stroke in Mice. Stroke 2016; 47:852-62. [PMID: 26839353 DOI: 10.1161/strokeaha.115.011230] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 12/31/2015] [Indexed: 12/27/2022]
Abstract
BACKGROUND AND PURPOSE Bone marrow cell (BMC)-based therapies, either the transplantation of exogenous cells or stimulation of endogenous cells by growth factors like the granulocyte colony-stimulating factor (G-CSF), are considered a promising means of treating stroke. In contrast to large preclinical evidence, however, a recent clinical stroke trial on G-CSF was neutral. We, therefore, aimed to investigate possible synergistic effects of co-administration of G-CSF and BMCs after experimental stroke in mice to enhance the efficacy compared with single treatments. METHODS We used an animal model for experimental stroke as paradigm to study possible synergistic effects of co-administration of G-CSF and BMCs on the functional outcome and the pathophysiological mechanism. RESULTS G-CSF treatment alone led to an improved functional outcome, a reduced infarct volume, increased blood vessel stabilization, and decreased overall inflammation. Surprisingly, the combination of G-CSF and BMCs abrogated G-CSFs' beneficial effects and resulted in increased hemorrhagic infarct transformation, altered blood-brain barrier, excessive astrogliosis, and altered immune cell polarization. These increased rates of infarct bleeding were mainly mediated by elevated matrix metalloproteinase-9-mediated blood-brain barrier breakdown in G-CSF- and BMCs-treated animals combined with an increased number of dilated and thus likely more fragile vessels in the subacute phase after cerebral ischemia. CONCLUSIONS Our results provide new insights into both BMC-based therapies and immune cell biology and help to understand potential adverse and unexpected side effects.
Collapse
Affiliation(s)
- Jan-Kolja Strecker
- From the Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany (J.-K.S., J.O., M.H., B.G., K.D., A.S., M.S., J.M.); and EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.).
| | - Joanna Olk
- From the Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany (J.-K.S., J.O., M.H., B.G., K.D., A.S., M.S., J.M.); and EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.)
| | - Maike Hoppen
- From the Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany (J.-K.S., J.O., M.H., B.G., K.D., A.S., M.S., J.M.); and EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.)
| | - Burkhard Gess
- From the Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany (J.-K.S., J.O., M.H., B.G., K.D., A.S., M.S., J.M.); and EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.)
| | - Kai Diederich
- From the Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany (J.-K.S., J.O., M.H., B.G., K.D., A.S., M.S., J.M.); and EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.)
| | - Antje Schmidt
- From the Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany (J.-K.S., J.O., M.H., B.G., K.D., A.S., M.S., J.M.); and EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.)
| | - Wolf-Rüdiger Schäbitz
- From the Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany (J.-K.S., J.O., M.H., B.G., K.D., A.S., M.S., J.M.); and EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.)
| | - Matthias Schilling
- From the Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany (J.-K.S., J.O., M.H., B.G., K.D., A.S., M.S., J.M.); and EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.)
| | - Jens Minnerup
- From the Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany (J.-K.S., J.O., M.H., B.G., K.D., A.S., M.S., J.M.); and EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.)
| |
Collapse
|
18
|
The MMP-1/PAR-1 Axis Enhances Proliferation and Neuronal Differentiation of Adult Hippocampal Neural Progenitor Cells. Neural Plast 2015; 2015:646595. [PMID: 26783471 PMCID: PMC4691474 DOI: 10.1155/2015/646595] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 08/13/2015] [Accepted: 09/06/2015] [Indexed: 12/26/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are zinc-dependent endopeptidases that play a role in varied forms of developmental and postnatal neuroplasticity. MMP substrates include protease-activated receptor-1 (PAR-1), a G-protein coupled receptor expressed in hippocampus. We examined proliferation and differentiation of adult neural progenitor cells (aNPCs) from hippocampi of mice that overexpress the potent PAR-1 agonist MMP-1. We found that, as compared to aNPCs from littermate controls, MMP-1 tg aNPCs display enhanced proliferation. Under differentiating conditions, these cells give rise to a higher percentage of MAP-2(+) neurons and a reduced number of oligodendrocyte precursors, and no change in the number of astrocytes. The fact that these results are MMP and PAR-1 dependent is supported by studies with distinct antagonists. Moreover, JSH-23, an inhibitor of NF-κB p65 nuclear translocation, counteracted both the proliferation and differentiation changes seen in MMP-1 tg-derived NPCs. In complementary studies, we found that the percentage of Sox2(+) undifferentiated progenitor cells is increased in hippocampi of MMP-1 tg animals, compared to wt mice. Together, these results add to a growing body of data suggesting that MMPs are effectors of hippocampal neuroplasticity in the adult CNS and that the MMP-1/PAR-1 axis may play a role in neurogenesis following physiological and/or pathological stimuli.
Collapse
|
19
|
Lei C, Wu B, Cao T, Liu M, Hao Z. Brain recovery mediated by toll-like receptor 4 in rats after intracerebral hemorrhage. Brain Res 2015; 1632:1-8. [PMID: 26657742 DOI: 10.1016/j.brainres.2015.11.045] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 11/07/2015] [Accepted: 11/27/2015] [Indexed: 02/05/2023]
Abstract
Activation of the immune system via toll-like receptor 4 (TLR4) is implicated in both negative and positive processes in the central nervous system, including inflammation and angiogenesis. Whether TLR4 also participates in brain recovery following intracerebral hemorrhage (ICH) has not been investigated. We used the rat model of collagenase-induced ICH to determine whether TLR4 acts as a key regulator of brain recovery in the late phase of injury. After ICH, TLR4 levels in the ipsilateral striatum were significantly higher in the ICH group than in the Sham group on days 1, 3, 7 and 14 after ICH induction. By 14 d, the ICH group showed significantly higher levels of vascular endothelial growth factor, brain-derived neurotrophic factor, and MMP-9 than the Sham group, as well as greater numbers of vessels and BrdU- and DCX-positive cells. All these ICH-induced increases were significantly smaller in the TAK-242 group. The TLR4 antagonist also inhibited the recovery of neurological function after ICH. A TLR4 antagonist reduced ICH-induced neurogenesis and angiogenesis in a rat. These findings suggest that TLR4 may promote brain repair in the late phase of ICH.
Collapse
Affiliation(s)
- Chunyan Lei
- Stroke Clinical Research Unit, Department of Neurology, West China Hospital, Sichuan University, PR China
| | - Bo Wu
- Stroke Clinical Research Unit, Department of Neurology, West China Hospital, Sichuan University, PR China; State Key Laboratory of Human Disease Biotherapy and Ministry of Education, West China Hospital, Sichuan University, PR China.
| | - Tian Cao
- Stroke Clinical Research Unit, Department of Neurology, West China Hospital, Sichuan University, PR China
| | - Ming Liu
- Stroke Clinical Research Unit, Department of Neurology, West China Hospital, Sichuan University, PR China; State Key Laboratory of Human Disease Biotherapy and Ministry of Education, West China Hospital, Sichuan University, PR China
| | - Zilong Hao
- Stroke Clinical Research Unit, Department of Neurology, West China Hospital, Sichuan University, PR China
| |
Collapse
|
20
|
Jablonska A, Drela K, Wojcik-Stanaszek L, Janowski M, Zalewska T, Lukomska B. Short-Lived Human Umbilical Cord-Blood-Derived Neural Stem Cells Influence the Endogenous Secretome and Increase the Number of Endogenous Neural Progenitors in a Rat Model of Lacunar Stroke. Mol Neurobiol 2015; 53:6413-6425. [PMID: 26607630 PMCID: PMC5085993 DOI: 10.1007/s12035-015-9530-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 11/08/2015] [Indexed: 12/12/2022]
Abstract
Stroke is the leading cause of severe disability, and lacunar stroke is related to cognitive decline and hemiparesis. There is no effective treatment for the majority of patients with stroke. Thus, stem cell-based regenerative medicine has drawn a growing body of attention due to the capabilities for trophic factor expression and neurogenesis enhancement. Moreover, it was shown in an experimental autoimmune encephalomyelitis (EAE) model that even short-lived stem cells can be therapeutic, and we have previously observed that phenomenon indirectly. Here, in a rat model of lacunar stroke, we investigated the molecular mechanisms underlying the positive therapeutic effects of short-lived human umbilical cord-blood-derived neural stem cells (HUCB-NSCs) through the distinct measurement of exogenous human and endogenous rat trophic factors. We have also evaluated neurogenesis and metalloproteinase activity as cellular components of therapeutic activity. As expected, we observed an increased proliferation and migration of progenitors, as well as metalloproteinase activity up to 14 days post transplantation. These changes were most prominent at the 7-day time point when we observed 30 % increases in the number of bromodeoxyuridine (BrdU)-positive cells in HUCB-NSC transplanted animals. The expression of human trophic factors was present until 7 days post transplantation, which correlated well with the survival of the human graft. For these 7 days, the level of messenger RNA (mRNA) in the analyzed trophic factors was from 300-fold for CNTF to 10,000-fold for IGF, much higher compared to constitutive expression in HUCB-NSCs in vitro. What is interesting is that there was no increase in the expression of rat trophic factors during the human graft survival, compared to that in non-transplanted animals. However, there was a prolongation of a period of increased trophic expression until 14 days post transplantation, while, in non-transplanted animals, there was a significant drop in rat trophic expression at that time point. We conclude that the positive therapeutic effect of short-lived stem cells may be related to the net increase in the amount of trophic factors (rat + human) until graft death and to the prolonged increase in rat trophic factor expression subsequently.
Collapse
Affiliation(s)
- Anna Jablonska
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Katarzyna Drela
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Luiza Wojcik-Stanaszek
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Miroslaw Janowski
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland.,Department of Radiology and Radiological Science, School of Medicine, Johns Hopkins University, Baltimore, MD, USA.,Cellular Imaging Section, Institute for Cell Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Teresa Zalewska
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Barbara Lukomska
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland.
| |
Collapse
|
21
|
Ding R, Feng L, He L, Chen Y, Wen P, Fu Z, Lin C, Yang S, Deng X, Zeng J, Sun G. Peroxynitrite decomposition catalyst prevents matrix metalloproteinase-9 activation and neurovascular injury after hemoglobin injection into the caudate nucleus of rats. Neuroscience 2015; 297:182-93. [PMID: 25849612 DOI: 10.1016/j.neuroscience.2015.03.065] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Revised: 03/19/2015] [Accepted: 03/29/2015] [Indexed: 10/23/2022]
Abstract
Hemoglobin (Hb) is a major constituent of blood and a potent mediator of oxidative or nitrative stress after intracerebral hemorrhage (ICH). Our previous study demonstrated that Hb could induce abundant peroxynitrite (ONOO(-)) formation in vivo, which may be involved in the blood-brain barrier (BBB) disruption, however, the drug intervention is absent and also the underlying mechanism. Using an experimental stroke model by injecting Hb into the caudate nucleus of male Sprague-Dawley rats, we assessed the role of ONOO(-) decomposition catalyst, 5,10,15,20-tetrakis (4-sulfonatophenyl) porphyrinato iron(III) [FeTPPS] in the activation of MMP-9 and Hb-induced neurovascular injuries. 3-Nitrotyrosine (3-NT, as an index of ONOO(-) formation) and NF-κB expression was measured by western blot (WB) and immunohistochemistry (IHC)/immunofluorescence (IF). Activity of MMP was evaluated by in situ zymography. Neurovascular injury was assessed using zonula occludens-1 (ZO-1) by WB and IF, fibronectin (FN) and neuron-specific nuclear protein (NeuN) IHC. Perihematomal cell death was determined by TUNEL assay. Behavioral outcome was measured by modified neurological severity score (mNSS) test. At the injured striata, profuse 3-NT was produced and mainly expressed in neutrophils and microglia/macrophages. 3-NT formation significantly colocalized with nuclear factor-κB (NF-κB) expression. In situ zymography showed that gelatinase activity was mostly co-localized with neurons and blood vessel walls and partly with neutrophils and microglia/macrophages. Enhanced 3-NT production, NF-κB induction and MMP-9 activation were obviously reduced after FeTPPS treatment. Hb-induced injury to tight junction protein (ZO-1), basal lamina of FN-immunopositive microvasculature and neural cells was evidently ameliorated by FeTPPS. In addition, apoptotic cell numbers as well as behavioral deficits were also improved. The present study shows that the administration of the ONOO(-) decomposition catalyst FeTPPS protects against Hb-induced neurovascular injuries and improves neurological function, which possibly in part by suppressing MMP-9 activation.
Collapse
Affiliation(s)
- R Ding
- The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - L Feng
- The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - L He
- Department of Endocrinology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Y Chen
- The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China.
| | - P Wen
- The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Z Fu
- The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - C Lin
- The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - S Yang
- Department of Neurosurgery, Gaoqing Campus of Central Hospital of Zibo, Gaoqing People's Hospital, Gaoqing, Zibo 256300, Shandong, China
| | - X Deng
- Department of Neurosurgery, 999 Brain Hospital, Jinan University, Guangzhou 510510, China
| | - J Zeng
- The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - G Sun
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
22
|
Prakash A, Bharti K, Majeed ABA. Zinc: indications in brain disorders. Fundam Clin Pharmacol 2015; 29:131-49. [PMID: 25659970 DOI: 10.1111/fcp.12110] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 07/08/2014] [Accepted: 12/24/2014] [Indexed: 12/14/2022]
Abstract
Zinc is the authoritative metal which is present in our body, and reactive zinc metal is crucial for neuronal signaling and is largely distributed within presynaptic vesicles. Zinc also plays an important role in synaptic function. At cellular level, zinc is a modulator of synaptic activity and neuronal plasticity in both development and adulthood. Different importers and transporters are involved in zinc homeostasis. ZnT-3 is a main transporter involved in zinc homeostasis in the brain. It has been found that alterations in brain zinc status have been implicated in a wide range of neurological disorders including impaired brain development and many neurodegenerative disorders such as Alzheimer's disease, and mood disorders including depression, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, and prion disease. Furthermore, zinc has also been implicated in neuronal damage associated with traumatic brain injury, stroke, and seizure. Understanding the mechanisms that control brain zinc homeostasis is thus critical to the development of preventive and treatment strategies for these and other neurological disorders.
Collapse
Affiliation(s)
- Atish Prakash
- Brain Degeneration and Therapeutics Group, Brain and Neuroscience Communities of Research, Universiti Teknologi MARA (UiTM), Shah Alam, 40450, Malaysia; Department of Pharmacology, ISF college of Pharmacy, Ghal kalan, Moga, 142-001, India; Brain Research Laboratory, Faculty of Pharmacy, Campus Puncak Alam, Universiti Teknologi MARA (UiTM), Bandar Puncak Alam, 42300, Malaysia
| | | | | |
Collapse
|
23
|
Lei C, Wu B, Cao T, Zhang S, Liu M. Activation of the High-Mobility Group Box 1 Protein-Receptor for Advanced Glycation End-Products Signaling Pathway in Rats During Neurogenesis After Intracerebral Hemorrhage. Stroke 2015; 46:500-6. [PMID: 25538203 DOI: 10.1161/strokeaha.114.006825] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Background and Purpose—
Following intracerebral hemorrhage (ICH), high-mobility group box 1 protein (HMGB1) may promote neurogenesis that supports functional recovery. How HMGB1 regulates or participates in this process is unclear, as are the pattern recognition receptors and signaling pathways involved.
Methods—
ICH was induced by injection of collagenase in Sprague–Dawley rats, which were treated 3 days later with saline, with the HMGB1 inhibitor ethyl pyruvate or with FPS-ZM1, an antagonist of the receptor for advanced glycation end-products. A Sham group was treated with saline solution instead of collagenase and then treated 3 days later with saline again or with ethyl pyruvate or N-benzyl-4-chloro-N-cyclohexylbenzamide (FPS-ZM1). Expression of the following proteins was measured by Western blot, immunohistochemistry, or immunofluorescence: HMGB1, receptor for advanced glycation end-products, toll-like receptor (TLR)-2, TLR4, brain-derived neurotrophic factor, and matrix metalloproteinase-9. The number of cells positive for 5-bromo-2-deoxyuridine or doublecortin was determined by immunohistochemistry and immunofluorescence.
Results—
Levels of HMGB1, receptor for advanced glycation end-products, TLR4, TLR2, brain-derived neurotrophic factor, and matrix metalloproteinase-9 were significantly higher 14 days after ICH than at baseline, as were the numbers of 5-bromo-2-deoxyuridine- or doublecortin-positive cells. At the same time, HMGB1 moved from the nucleus into the cytoplasm. Administering ethyl pyruvate significantly reduced all these ICH-induced increases, except the increase in TLR4 and TLR2. Administering FPS-ZM1 reduced the ICH-induced increases in the expression of brain-derived neurotrophic factor and matrix metalloproteinase-9 and in the numbers of 5-bromo-2-deoxyuridine- or doublecortin-positive cells.
Conclusions—
These findings suggest that HMGB1 acts via the receptor for advanced glycation end-products signaling pathway to promote neurogenesis in later phases of ICH.
Collapse
Affiliation(s)
- Chunyan Lei
- From the Stroke Clinical Research Unit, Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, People’s Republic of China
| | - Bo Wu
- From the Stroke Clinical Research Unit, Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, People’s Republic of China
| | - Tian Cao
- From the Stroke Clinical Research Unit, Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, People’s Republic of China
| | - Shuting Zhang
- From the Stroke Clinical Research Unit, Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, People’s Republic of China
| | - Ming Liu
- From the Stroke Clinical Research Unit, Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, People’s Republic of China
| |
Collapse
|
24
|
Ito A, Fujimura M, Niizuma K, Kanoke A, Sakata H, Morita-Fujimura Y, Kikuchi A, Kure S, Tominaga T. Enhanced post-ischemic angiogenesis in mice lacking RNF213; a susceptibility gene for moyamoya disease. Brain Res 2014; 1594:310-20. [PMID: 25446450 DOI: 10.1016/j.brainres.2014.11.014] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2014] [Revised: 10/07/2014] [Accepted: 11/07/2014] [Indexed: 11/17/2022]
Abstract
Moyamoya disease (MMD) is a chronic occlusive cerebrovascular disease with unknown etiology that is characterized by the development of abnormal vascular networks at the base of the brain. Recent genome-wide studies identified RNF213 as an important MMD susceptibility gene. However, the exact mechanism by which the RNF213 abnormality leads to MMD remains unknown. Thus, we sought to clarify the role of RNF213 in angiogenesis under ischemic conditions using conventional RNF213 knockout mice. We assessed the infarction volume, cerebral edema, and vascular density in the ischemic brain after transient middle cerebral artery occlusion (tMCAO). To further evaluate systemic angiogenesis following chronic ischemia, we investigated blood flow recovery using laser speckle flowmetry, the severity of ambulatory impairments, and vascular density in the hind-limb after permanent femoral artery ligation. Results were compared between homozygous RNF213 knockout mice (RNF213 -/-) and wild-type littermates (Wt). No significant differences were observed in infarction volume or the formation of edema following tMCAO, or in vascular density 28 days after tMCAO between RNF213 -/- and Wt. Blood flow recovery was significantly improved in RNF213 -/- from 3 to 28 days after femoral artery ligation, and angiogenesis as shown by vascular density in the hind-limb was significantly enhanced in RNF213 -/- at 28 days. The amelioration of ambulatory impairments was also evident in RNF213 -/-. Angiogenesis was enhanced in mice lacking RNF213 after chronic hind-limb ischemia, which suggested the potential role of the RNF213 abnormality in the development of pathological vascular networks in chronic ischemia.
Collapse
Affiliation(s)
- Akira Ito
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Miki Fujimura
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Kuniyasu Niizuma
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Atsushi Kanoke
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroyuki Sakata
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yuiko Morita-Fujimura
- Department of Molecular Biology, Tohoku University Institute of Aging and Cancer, Sendai, Japan
| | - Atsuo Kikuchi
- Department of Pediatrics, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shigeo Kure
- Department of Pediatrics, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Teiji Tominaga
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
25
|
Chang JJ, Emanuel BA, Mack WJ, Tsivgoulis G, Alexandrov AV. Matrix metalloproteinase-9: dual role and temporal profile in intracerebral hemorrhage. J Stroke Cerebrovasc Dis 2014; 23:2498-2505. [PMID: 25306400 DOI: 10.1016/j.jstrokecerebrovasdis.2014.07.005] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 06/20/2014] [Accepted: 07/02/2014] [Indexed: 10/24/2022] Open
Abstract
BACKGROUND Clinical outcome after intracerebral hemorrhage (ICH) remains poor. Recent trials in ICH, focusing on hematoma reduction, have not yielded significant clinical improvement. The modulation of matrix metalloproteinase (MMP)-9 may represent a potential therapeutic target for reducing perihematomal edema (PHE) and improving clinical outcome. METHODS We searched Cochrane Library, Ovid/Medline, and PubMed databases using combinations of the following MeSH search terms: "intracerebral hemorrhage," "matrix metalloproteinase," "minocycline," "inhibition," and "neuroprotection". RESULTS MMP-9 levels in animal models have largely shown detrimental correlations with mortality, clinical outcome, hematoma volume, and PHE. Animal models and clinical studies have established a timeline for MMP-9 expression and corresponding PHE that include an initial peak on days 1-3 and a secondary peak on day 7. Clinical studies evaluating MMP-9 levels in the acute phase (days 1-3) and subacute phase (day 7) of ICH suggest that MMP-9 may be detrimental in the acute phase through destruction of basal lamina, activation of vascular endothelial growth factor, and activation of apoptosis but assist in recovery in the subacute phase through angiogenesis. CONCLUSIONS MMP-9 inhibition represents a potentially effective target for neuroprotection in ICH. However, as a ubiquitous protein, the inhibition of pathologic processes must be balanced against the preservation of neuroprotective angiogenesis. As the opposing roles of MMP-9 may have similar mechanisms, the most important factor may be the timing of MMP-9 inhibition. Further studies are necessary to delineate these mechanisms and their temporal relationship.
Collapse
Affiliation(s)
- Jason J Chang
- Department of Neurology, University of Southern California, Los Angeles, California.
| | - Benjamin A Emanuel
- Department of Neurology, University of Southern California, Los Angeles, California
| | - William J Mack
- Department of Neurosurgery, University of Southern California, Los Angeles, California
| | - Giorgios Tsivgoulis
- Department of Neurology, University of Athens School of Medicine, Athens, Greece; International Clinical Research Center, St. Anne's University, Brno, Czech Republic
| | - Andrei V Alexandrov
- Department of Neurology, The University of Tennessee Health Sciences Center, Memphis, TN
| |
Collapse
|