1
|
Cai Y, Zhang Y, Leng S, Ma Y, Jiang Q, Wen Q, Ju S, Hu J. The relationship between inflammation, impaired glymphatic system, and neurodegenerative disorders: A vicious cycle. Neurobiol Dis 2024; 192:106426. [PMID: 38331353 DOI: 10.1016/j.nbd.2024.106426] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 01/16/2024] [Accepted: 01/28/2024] [Indexed: 02/10/2024] Open
Abstract
The term "glymphatic" emerged roughly a decade ago, marking a pivotal point in neuroscience research. The glymphatic system, a glial-dependent perivascular network distributed throughout the brain, has since become a focal point of investigation. There is increasing evidence suggesting that impairment of the glymphatic system appears to be a common feature of neurodegenerative disorders, and this impairment exacerbates as disease progression. Nevertheless, the common factors contributing to glymphatic system dysfunction across most neurodegenerative disorders remain unclear. Inflammation, however, is suspected to play a pivotal role. Dysfunction of the glymphatic system can lead to a significant accumulation of protein and waste products, which can trigger inflammation. The interaction between the glymphatic system and inflammation appears to be cyclical and potentially synergistic. Yet, current research is limited, and there is a lack of comprehensive models explaining this association. In this perspective review, we propose a novel model suggesting that inflammation, impaired glymphatic function, and neurodegenerative disorders interconnected in a vicious cycle. By presenting experimental evidence from the existing literature, we aim to demonstrate that: (1) inflammation aggravates glymphatic system dysfunction, (2) the impaired glymphatic system exacerbated neurodegenerative disorders progression, (3) neurodegenerative disorders progression promotes inflammation. Finally, the implication of proposed model is discussed.
Collapse
Affiliation(s)
- Yu Cai
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing 210009, China
| | - Yangqiqi Zhang
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Shuo Leng
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing 210009, China
| | - Yuanyuan Ma
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing 210009, China
| | - Quan Jiang
- Department of Neurology, Henry Ford Health System, 2799 W Grand Blvd, Detroit, MI 48202, USA
| | - Qiuting Wen
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W.16th Street, Indianapolis, IN 46202-5188, USA
| | - Shenghong Ju
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing 210009, China.
| | - Jiani Hu
- Department of Radiology, School of Medicine, Wayne State University, Detroit, MI 48201, USA.
| |
Collapse
|
2
|
He Q, Wang Y, Fang C, Feng Z, Yin M, Huang J, Ma Y, Mo Z. Advancing stroke therapy: A deep dive into early phase of ischemic stroke and recanalization. CNS Neurosci Ther 2024; 30:e14634. [PMID: 38379112 PMCID: PMC10879038 DOI: 10.1111/cns.14634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 01/16/2024] [Accepted: 01/23/2024] [Indexed: 02/22/2024] Open
Abstract
Ischemic stroke, accounting for the majority of stroke events, significantly contributes to global morbidity and mortality. Vascular recanalization therapies, namely intravenous thrombolysis and mechanical thrombectomy, have emerged as critical interventions, yet their success hinges on timely application and patient-specific factors. This review focuses on the early phase pathophysiological mechanisms of ischemic stroke and the nuances of recanalization. It highlights the dual role of neutrophils in tissue damage and repair, and the critical involvement of the blood-brain barrier (BBB) in stroke outcomes. Special emphasis is placed on ischemia-reperfusion injury, characterized by oxidative stress, inflammation, and endothelial dysfunction, which paradoxically exacerbates cerebral damage post-revascularization. The review also explores the potential of targeting molecular pathways involved in BBB integrity and inflammation to enhance the efficacy of recanalization therapies. By synthesizing current research, this paper aims to provide insights into optimizing treatment protocols and developing adjuvant neuroprotective strategies, thereby advancing stroke therapy and improving patient outcomes.
Collapse
Affiliation(s)
- Qianyan He
- Department of Neurology, Stroke CenterThe First Hospital of Jilin UniversityJilinChina
- Institute of Biomedicine and BiotechnologyShenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenGuangdongChina
| | - Yueqing Wang
- Institute of Biomedicine and BiotechnologyShenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenGuangdongChina
| | - Cheng Fang
- Institute of Biomedicine and BiotechnologyShenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenGuangdongChina
| | - Ziying Feng
- Institute of Biomedicine and BiotechnologyShenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenGuangdongChina
| | - Meifang Yin
- Institute of Biomedicine and BiotechnologyShenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenGuangdongChina
| | - Juyang Huang
- School of Pharmaceutical Sciences (Shenzhen)Sun Yat‐sen UniversityShenzhenGuangdongChina
| | - Yinzhong Ma
- Institute of Biomedicine and BiotechnologyShenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenGuangdongChina
| | - Zhizhun Mo
- Emergency Department, Shenzhen Traditional Chinese Medicine HospitalThe Fourth Clinical Medical College of Guangzhou University of Chinese MedicineShenzhenGuangdongChina
| |
Collapse
|
3
|
Bani-Sadr A, Mechtouff L, De Bourguignon C, Mauffrey A, Boutelier T, Cho TH, Cappucci M, Ameli R, Hermier M, Derex L, Nighoghossian N, Berthezene Y. Blood-Brain Barrier Permeability and Kinetics of Inflammatory Markers in Acute Stroke Patients Treated With Thrombectomy. Neurology 2023; 101:e502-e511. [PMID: 37290975 PMCID: PMC10401692 DOI: 10.1212/wnl.0000000000207460] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 04/13/2023] [Indexed: 06/10/2023] Open
Abstract
BACKGROUND AND OBJECTIVES The aim of this study was to investigate the relationship between baseline blood-brain barrier (BBB) permeability and the kinetics of circulating inflammatory markers in a cohort of acute ischemic stroke (AIS) patients treated with mechanical thrombectomy. METHODS The CoHort of Patients to Identify Biological and Imaging markerS of CardiovascUlar Outcomes in Stroke includes AIS patients treated with mechanical thrombectomy after admission MRI and undergoing a sequential assessment of circulating inflammatory markers. Baseline dynamic susceptibility perfusion MRI was postprocessed with arrival time correction to provide K2 maps reflecting BBB permeability. After coregistration of apparent diffusion coefficient and K2 maps, the 90th percentile of K2 value was extracted within baseline ischemic core and expressed as a percentage change compared with contralateral normal-appearing white matter. Population was dichotomized according to the median K2 value. Univariable and multiple variable logistic regression analyses were performed to investigate factors associated with increased pretreatment BBB permeability in the whole population and in patients with symptom onset <6 hours. RESULTS In the whole population (n = 105 patients, median K2 = 1.59), patients with an increased BBB permeability had higher serum levels of matrix metalloproteinase (MMP)-9 at H48 (p = 0.02), a higher C-reactive protein (CRP) serum level at H48 (p = 0.01), poorer collateral status (p = 0.01), and a larger baseline ischemic core (p < 0.001). They were more likely to have hemorrhagic transformation (p = 0.008), larger final lesion volume (p = 0.02), and worst neurologic outcome at 3 months (p = 0.04). The multiple variable logistic regression indicated that an increased BBB permeability was associated only with ischemic core volume (odds ratio [OR] 1.04, 95% CI 1.01-1.06, p < 0.0001). Restricting analysis to patients with symptom onset <6 hours (n = 72, median K2 = 1.27), participants with an increased BBB permeability had higher serum levels of MMP-9 at H0 (p = 0.005), H6 (p = 0.004), H24 (p = 0.02), and H48 (p = 0.01), higher CRP levels at H48 (p = 0.02), and a larger baseline ischemic core (p < 0.0001). The multiple variable logistic analysis showed that increased BBB permeability was independently associated with higher H0 MMP-9 levels (OR 1.33, 95% CI 1.12-1.65, p = 0.01) and a larger ischemic core (OR 1.27, 95% CI 1.08-1.59, p = 0.04). DISCUSSION In AIS patients, increased BBB permeability is associated with a larger ischemic core. In the subgroup of patients with symptom onset <6 hours, increased BBB permeability is independently associated with higher H0 MMP-9 levels and a larger ischemic core.
Collapse
Affiliation(s)
- Alexandre Bani-Sadr
- From the Hospices Civils de Lyon (A.B.-S., L.M., C.D.B., T.-H.C., M.C., R.A., M.H., L.D., N.N., Y.B.); and Olea Medical (A.M., T.B.), La Ciotat, France.
| | - Laura Mechtouff
- From the Hospices Civils de Lyon (A.B.-S., L.M., C.D.B., T.-H.C., M.C., R.A., M.H., L.D., N.N., Y.B.); and Olea Medical (A.M., T.B.), La Ciotat, France
| | - Charles De Bourguignon
- From the Hospices Civils de Lyon (A.B.-S., L.M., C.D.B., T.-H.C., M.C., R.A., M.H., L.D., N.N., Y.B.); and Olea Medical (A.M., T.B.), La Ciotat, France
| | - Aela Mauffrey
- From the Hospices Civils de Lyon (A.B.-S., L.M., C.D.B., T.-H.C., M.C., R.A., M.H., L.D., N.N., Y.B.); and Olea Medical (A.M., T.B.), La Ciotat, France
| | - Timothe Boutelier
- From the Hospices Civils de Lyon (A.B.-S., L.M., C.D.B., T.-H.C., M.C., R.A., M.H., L.D., N.N., Y.B.); and Olea Medical (A.M., T.B.), La Ciotat, France
| | - Tae-Hee Cho
- From the Hospices Civils de Lyon (A.B.-S., L.M., C.D.B., T.-H.C., M.C., R.A., M.H., L.D., N.N., Y.B.); and Olea Medical (A.M., T.B.), La Ciotat, France
| | - Matteo Cappucci
- From the Hospices Civils de Lyon (A.B.-S., L.M., C.D.B., T.-H.C., M.C., R.A., M.H., L.D., N.N., Y.B.); and Olea Medical (A.M., T.B.), La Ciotat, France
| | - Roxana Ameli
- From the Hospices Civils de Lyon (A.B.-S., L.M., C.D.B., T.-H.C., M.C., R.A., M.H., L.D., N.N., Y.B.); and Olea Medical (A.M., T.B.), La Ciotat, France
| | - Marc Hermier
- From the Hospices Civils de Lyon (A.B.-S., L.M., C.D.B., T.-H.C., M.C., R.A., M.H., L.D., N.N., Y.B.); and Olea Medical (A.M., T.B.), La Ciotat, France
| | - Laurent Derex
- From the Hospices Civils de Lyon (A.B.-S., L.M., C.D.B., T.-H.C., M.C., R.A., M.H., L.D., N.N., Y.B.); and Olea Medical (A.M., T.B.), La Ciotat, France
| | - Norbert Nighoghossian
- From the Hospices Civils de Lyon (A.B.-S., L.M., C.D.B., T.-H.C., M.C., R.A., M.H., L.D., N.N., Y.B.); and Olea Medical (A.M., T.B.), La Ciotat, France
| | - Yves Berthezene
- From the Hospices Civils de Lyon (A.B.-S., L.M., C.D.B., T.-H.C., M.C., R.A., M.H., L.D., N.N., Y.B.); and Olea Medical (A.M., T.B.), La Ciotat, France
| |
Collapse
|
4
|
Thapa K, Shivam K, Khan H, Kaur A, Dua K, Singh S, Singh TG. Emerging Targets for Modulation of Immune Response and Inflammation in Stroke. Neurochem Res 2023; 48:1663-1690. [PMID: 36763312 DOI: 10.1007/s11064-023-03875-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 01/18/2023] [Accepted: 01/24/2023] [Indexed: 02/11/2023]
Abstract
The inflammatory and immunological responses play a significant role after stroke. The innate immune activation stimulated by microglia during stroke results in the migration of macrophages and lymphocytes into the brain and are responsible for tissue damage. The immune response and inflammation following stroke have no defined targets, and the intricacies of the immunological and inflammatory processes are only partially understood. Innate immune cells enter the brain and meninges during the acute phase, which can cause ischemia damage. Activation of systemic immunity is caused by danger signals sent into the bloodstream by injured brain cells, which is followed by a significant immunodepression that encourages life-threatening infections. Neuropsychiatric sequelae, a major source of post-stroke morbidity, may be induced by an adaptive immune response that is initiated by antigen presentation during the chronic period and is directed against the brain. Thus, the current review discusses the role of immune response and inflammation in stroke pathogenesis, their role in the progression of injury during the stroke, and the emerging targets for the modulation of the mechanism of immune response and inflammation that may have possible therapeutic benefits against stroke.
Collapse
Affiliation(s)
- Komal Thapa
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.,School of Pharmacy, Chitkara University, Rajpura, Himachal Pradesh, 174103, India
| | - Kumar Shivam
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Heena Khan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Amarjot Kaur
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW, 2007, Australia.,Faculty of Health, Australian Research Centre in Complementary & Integrative Medicine, University of Technology Sydney, Ultimo, 2007, Australia
| | - Sachin Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar - Delhi G.T. Road, Phagwara, Punjab, 144411, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
| |
Collapse
|
5
|
Anwar MM, Özkan E, Gürsoy-Özdemir Y. The role of extracellular matrix alterations in mediating astrocyte damage and pericyte dysfunction in Alzheimer's disease: A comprehensive review. Eur J Neurosci 2022; 56:5453-5475. [PMID: 34182602 DOI: 10.1111/ejn.15372] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/19/2021] [Accepted: 06/21/2021] [Indexed: 12/14/2022]
Abstract
The brain is a highly vascularized tissue protected by the blood-brain barrier (BBB), a complex structure allowing only necessary substances to pass through into the brain while limiting the entrance of harmful toxins. The BBB comprises several components, and the most prominent features are tight junctions between endothelial cells (ECs), which are further wrapped in a layer of pericytes. Pericytes are multitasked cells embedded in a thick basement membrane (BM) that consists of a fibrous extracellular matrix (ECM) and are surrounded by astrocytic endfeet. The primary function of astrocytes and pericytes is to provide essential blood supply and vital nutrients to the brain. In Alzheimer's disease (AD), long-term neuroinflammatory cascades associated with infiltration of harmful neurotoxic proteins may lead to BBB dysfunction and altered ECM components resulting in brain homeostatic imbalance, synaptic damage, and declined cognitive functions. Moreover, BBB structure and functional integrity may be lost due to induced ECM alterations, astrocyte damage, and pericytes dysfunction, leading to amyloid-beta (Aβ) hallmarks deposition in different brain regions. Herein, we highlight how BBB, ECM, astrocytes, and pericytes dysfunction can play a leading role in AD's pathogenesis and discuss their impact on brain functions.
Collapse
Affiliation(s)
- Mai M Anwar
- Neuroscience Research Lab, Research Center for Translational Medicine (KUTTAM), Koç University, Istanbul, Turkey.,Department of Biochemistry, National Organization for Drug Control and Research (NODCAR)/Egyptian Drug Authority, Cairo, Egypt
| | - Esra Özkan
- Neuroscience Research Lab, Research Center for Translational Medicine (KUTTAM), Koç University, Istanbul, Turkey
| | - Yasemin Gürsoy-Özdemir
- Neuroscience Research Lab, Research Center for Translational Medicine (KUTTAM), Koç University, Istanbul, Turkey.,Department of Neurology, School of Medicine, Koç University, Istanbul, Turkey
| |
Collapse
|
6
|
Zhang Y, Gao X, Bai X, Yao S, Chang YZ, Gao G. The emerging role of furin in neurodegenerative and neuropsychiatric diseases. Transl Neurodegener 2022; 11:39. [PMID: 35996194 PMCID: PMC9395820 DOI: 10.1186/s40035-022-00313-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 08/10/2022] [Indexed: 12/02/2022] Open
Abstract
Furin is an important mammalian proprotein convertase that catalyzes the proteolytic maturation of a variety of prohormones and proproteins in the secretory pathway. In the brain, the substrates of furin include the proproteins of growth factors, receptors and enzymes. Emerging evidence, such as reduced FURIN mRNA expression in the brains of Alzheimer's disease patients or schizophrenia patients, has implicated a crucial role of furin in the pathophysiology of neurodegenerative and neuropsychiatric diseases. Currently, compared to cancer and infectious diseases, the aberrant expression of furin and its pharmaceutical potentials in neurological diseases remain poorly understood. In this article, we provide an overview on the physiological roles of furin and its substrates in the brain, summarize the deregulation of furin expression and its effects in neurodegenerative and neuropsychiatric disorders, and discuss the implications and current approaches that target furin for therapeutic interventions. This review may expedite future studies to clarify the molecular mechanisms of furin deregulation and involvement in the pathogenesis of neurodegenerative and neuropsychiatric diseases, and to develop new diagnosis and treatment strategies for these diseases.
Collapse
Affiliation(s)
- Yi Zhang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China
| | - Xiaoqin Gao
- Shijiazhuang People's Hospital, Hebei Medical University, Shijiazhuang, 050027, China
| | - Xue Bai
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China
| | - Shanshan Yao
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China
| | - Yan-Zhong Chang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China.
| | - Guofen Gao
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, China.
| |
Collapse
|
7
|
Jurcau A, Ardelean AI. Oxidative Stress in Ischemia/Reperfusion Injuries following Acute Ischemic Stroke. Biomedicines 2022; 10:biomedicines10030574. [PMID: 35327376 PMCID: PMC8945353 DOI: 10.3390/biomedicines10030574] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/25/2022] [Accepted: 02/28/2022] [Indexed: 02/04/2023] Open
Abstract
Recanalization therapy is increasingly used in the treatment of acute ischemic stroke. However, in about one third of these patients, recanalization is followed by ischemia/reperfusion injuries, and clinically to worsening of the neurological status. Much research has focused on unraveling the involved mechanisms in order to prevent or efficiently treat these injuries. What we know so far is that oxidative stress and mitochondrial dysfunction are significantly involved in the pathogenesis of ischemia/reperfusion injury. However, despite promising results obtained in experimental research, clinical studies trying to interfere with the oxidative pathways have mostly failed. The current article discusses the main mechanisms leading to ischemia/reperfusion injuries, such as mitochondrial dysfunction, excitotoxicity, and oxidative stress, and reviews the clinical trials with antioxidant molecules highlighting recent developments and future strategies.
Collapse
Affiliation(s)
- Anamaria Jurcau
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
- Department of Neurology, Clinical Municipal Hospital Oradea, Louis Pasteur Street nr 26, 410054 Oradea, Romania
- Correspondence: ; Tel.: +40-744-600-833
| | - Adriana Ioana Ardelean
- Department of Preclinical Sciences, Faculty of Medicine and Pharmacy, University of Oradea, Universitatii Street nr 1, 410087 Oradea, Romania;
- Department of Cardiology, Clinical Emergency County Hospital Oradea, Gh. Doja Street nr 65, 410169 Oradea, Romania
| |
Collapse
|
8
|
Mechanisms of Thrombosis and Thrombolysis. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00002-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
9
|
Lai AKW, Ng TC, Hung VKL, Tam KC, Cheung CW, Chung SK, Lo ACY. Exacerbated VEGF up-regulation accompanies diabetes-aggravated hemorrhage in mice after experimental cerebral ischemia and delayed reperfusion. Neural Regen Res 2021; 17:1566-1575. [PMID: 34916442 PMCID: PMC8771109 DOI: 10.4103/1673-5374.330612] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Reperfusion therapy is the preferred treatment for ischemic stroke, but is hindered by its short treatment window, especially in patients with diabetes whose reperfusion after prolonged ischemia is often accompanied by exacerbated hemorrhage. The mechanisms underlying exacerbated hemorrhage are not fully understood. This study aimed to identify this mechanism by inducing prolonged 2-hour transient intraluminal middle cerebral artery occlusion in diabetic Ins2Akita/+ mice to mimic patients with diabetes undergoing delayed mechanical thrombectomy. The results showed that at as early as 2 hours after reperfusion, Ins2Akita/+ mice exhibited rapid development of neurological deficits, increased infarct and hemorrhagic transformation, together with exacerbated down-regulation of tight-junction protein ZO-1 and up-regulation of blood-brain barrier-disrupting matrix metallopeptidase 2 and matrix metallopeptidase 9 when compared with normoglycemic Ins2+/+ mice. This indicated that diabetes led to the rapid compromise of vessel integrity immediately after reperfusion, and consequently earlier death and further aggravation of hemorrhagic transformation 22 hours after reperfusion. This observation was associated with earlier and stronger up-regulation of pro-angiogenic vascular endothelial growth factor (VEGF) and its downstream phospho-Erk1/2 at 2 hours after reperfusion, which was suggestive of premature angiogenesis induced by early VEGF up-regulation, resulting in rapid vessel disintegration in diabetic stroke. Endoplasmic reticulum stress-related pro-apoptotic C/EBP homologous protein was overexpressed in challenged Ins2Akita/+ mice, which suggests that the exacerbated VEGF up-regulation may be caused by overwhelming endoplasmic reticulum stress under diabetic conditions. In conclusion, the results mimicked complications in patients with diabetes undergoing delayed mechanical thrombectomy, and diabetes-induced accelerated VEGF up-regulation is likely to underlie exacerbated hemorrhagic transformation. Thus, suppression of the VEGF pathway could be a potential approach to allow reperfusion therapy in patients with diabetic stroke beyond the current treatment window. Experiments were approved by the Committee on the Use of Live Animals in Teaching and Research of the University of Hong Kong [CULATR 3834-15 (approval date January 5, 2016); 3977-16 (approval date April 13, 2016); and 4666-18 (approval date March 29, 2018)].
Collapse
Affiliation(s)
- Angela Ka Wai Lai
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administration Region, China
| | - Tsz Chung Ng
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administration Region, China
| | - Victor Ka Lok Hung
- Department of Anesthesiology, Laboratory and Clinical Research Institute for Pain, The University of Hong Kong, Hong Kong Special Administration Region, China
| | - Ka Cheung Tam
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administration Region, China
| | - Chi Wai Cheung
- Department of Anesthesiology, Laboratory and Clinical Research Institute for Pain, The University of Hong Kong, Hong Kong Special Administration Region, China
| | - Sookja Kim Chung
- Macau University of Science and Technology, Taipa, Macau Special Administration Region; School of Biomedical Sciences, The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong Special Administration Region, China
| | - Amy Cheuk Yin Lo
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administration Region, China
| |
Collapse
|
10
|
Hamblin MH, Lee JP. Neural Stem Cells for Early Ischemic Stroke. Int J Mol Sci 2021; 22:ijms22147703. [PMID: 34299322 PMCID: PMC8306669 DOI: 10.3390/ijms22147703] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 12/11/2022] Open
Abstract
Clinical treatments for ischemic stroke are limited. Neural stem cell (NSC) transplantation can be a promising therapy. Clinically, ischemia and subsequent reperfusion lead to extensive neurovascular injury that involves inflammation, disruption of the blood-brain barrier, and brain cell death. NSCs exhibit multiple potentially therapeutic actions against neurovascular injury. Currently, tissue plasminogen activator (tPA) is the only FDA-approved clot-dissolving agent. While tPA’s thrombolytic role within the vasculature is beneficial, tPA’s non-thrombolytic deleterious effects aggravates neurovascular injury, restricting the treatment time window (time-sensitive) and tPA eligibility. Thus, new strategies are needed to mitigate tPA’s detrimental effects and quickly mediate vascular repair after stroke. Up to date, clinical trials focus on the impact of stem cell therapy on neuro-restoration by delivering cells during the chronic stroke stage. Also, NSCs secrete factors that stimulate endogenous repair mechanisms for early-stage ischemic stroke. This review will present an integrated view of the preclinical perspectives of NSC transplantation as a promising treatment for neurovascular injury, with an emphasis on early-stage ischemic stroke. Further, this will highlight the impact of early sub-acute NSC delivery on improving short-term and long-term stroke outcomes.
Collapse
Affiliation(s)
- Milton H. Hamblin
- Department of Pharmacology, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA 70112, USA
- Correspondence: (M.H.H.); (J.-P.L.)
| | - Jean-Pyo Lee
- Department of Physiology, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA 70112, USA
- Tulane Brain Institute, Tulane University, 1430 Tulane Ave, New Orleans, LA 70112, USA
- Correspondence: (M.H.H.); (J.-P.L.)
| |
Collapse
|
11
|
Islam Y, Leach AG, Smith J, Pluchino S, Coxon CR, Sivakumaran M, Downing J, Fatokun AA, Teixidò M, Ehtezazi T. Physiological and Pathological Factors Affecting Drug Delivery to the Brain by Nanoparticles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2002085. [PMID: 34105297 PMCID: PMC8188209 DOI: 10.1002/advs.202002085] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 01/06/2021] [Indexed: 05/04/2023]
Abstract
The prevalence of neurological/neurodegenerative diseases, such as Alzheimer's disease is known to be increasing due to an aging population and is anticipated to further grow in the decades ahead. The treatment of brain diseases is challenging partly due to the inaccessibility of therapeutic agents to the brain. An increasingly important observation is that the physiology of the brain alters during many brain diseases, and aging adds even more to the complexity of the disease. There is a notion that the permeability of the blood-brain barrier (BBB) increases with aging or disease, however, the body has a defense mechanism that still retains the separation of the brain from harmful chemicals in the blood. This makes drug delivery to the diseased brain, even more challenging and complex task. Here, the physiological changes to the diseased brain and aged brain are covered in the context of drug delivery to the brain using nanoparticles. Also, recent and novel approaches are discussed for the delivery of therapeutic agents to the diseased brain using nanoparticle based or magnetic resonance imaging guided systems. Furthermore, the complement activation, toxicity, and immunogenicity of brain targeting nanoparticles as well as novel in vitro BBB models are discussed.
Collapse
Affiliation(s)
- Yamir Islam
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| | - Andrew G. Leach
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
- Division of Pharmacy and OptometryThe University of ManchesterStopford Building, Oxford RoadManchesterM13 9PTUK
| | - Jayden Smith
- Cambridge Innovation Technologies Consulting (CITC) LimitedSt. John's Innovation CentreCowley RoadCambridgeCB4 0WSUK
| | - Stefano Pluchino
- Department of Clinical NeurosciencesClifford Allbutt Building – Cambridge Biosciences Campus and NIHR Biomedical Research CentreUniversity of CambridgeHills RoadCambridgeCB2 0HAUK
| | - Christopher R. Coxon
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
- School of Engineering and Physical SciencesHeriot‐Watt UniversityWilliam Perkin BuildingEdinburghEH14 4ASUK
| | - Muttuswamy Sivakumaran
- Department of HaematologyPeterborough City HospitalEdith Cavell CampusBretton Gate PeterboroughPeterboroughPE3 9GZUK
| | - James Downing
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| | - Amos A. Fatokun
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| | - Meritxell Teixidò
- Institute for Research in Biomedicine (IRB Barcelona)Barcelona Institute of Science and Technology (BIST)Baldiri Reixac 10Barcelona08028Spain
| | - Touraj Ehtezazi
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| |
Collapse
|
12
|
Chico TJA, Kugler EC. Cerebrovascular development: mechanisms and experimental approaches. Cell Mol Life Sci 2021; 78:4377-4398. [PMID: 33688979 PMCID: PMC8164590 DOI: 10.1007/s00018-021-03790-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 02/04/2021] [Accepted: 02/12/2021] [Indexed: 12/13/2022]
Abstract
The cerebral vasculature plays a central role in human health and disease and possesses several unique anatomic, functional and molecular characteristics. Despite their importance, the mechanisms that determine cerebrovascular development are less well studied than other vascular territories. This is in part due to limitations of existing models and techniques for visualisation and manipulation of the cerebral vasculature. In this review we summarise the experimental approaches used to study the cerebral vessels and the mechanisms that contribute to their development.
Collapse
Affiliation(s)
- Timothy J A Chico
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK.
- The Bateson Centre, Firth Court, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK.
- Insigneo Institute for in Silico Medicine, The Pam Liversidge Building, Sheffield, S1 3JD, UK.
| | - Elisabeth C Kugler
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK.
- The Bateson Centre, Firth Court, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK.
- Insigneo Institute for in Silico Medicine, The Pam Liversidge Building, Sheffield, S1 3JD, UK.
| |
Collapse
|
13
|
Bernardo-Castro S, Sousa JA, Brás A, Cecília C, Rodrigues B, Almendra L, Machado C, Santo G, Silva F, Ferreira L, Santana I, Sargento-Freitas J. Pathophysiology of Blood-Brain Barrier Permeability Throughout the Different Stages of Ischemic Stroke and Its Implication on Hemorrhagic Transformation and Recovery. Front Neurol 2020; 11:594672. [PMID: 33362697 PMCID: PMC7756029 DOI: 10.3389/fneur.2020.594672] [Citation(s) in RCA: 224] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 11/09/2020] [Indexed: 12/25/2022] Open
Abstract
The blood-brain barrier (BBB) is a dynamic interface responsible for maintaining the central nervous system homeostasis. Its unique characteristics allow protecting the brain from unwanted compounds, but its impairment is involved in a vast number of pathological conditions. Disruption of the BBB and increase in its permeability are key in the development of several neurological diseases and have been extensively studied in stroke. Ischemic stroke is the most prevalent type of stroke and is characterized by a myriad of pathological events triggered by an arterial occlusion that can eventually lead to fatal outcomes such as hemorrhagic transformation (HT). BBB permeability seems to follow a multiphasic pattern throughout the different stroke stages that have been associated with distinct biological substrates. In the hyperacute stage, sudden hypoxia damages the BBB, leading to cytotoxic edema and increased permeability; in the acute stage, the neuroinflammatory response aggravates the BBB injury, leading to higher permeability and a consequent risk of HT that can be motivated by reperfusion therapy; in the subacute stage (1-3 weeks), repair mechanisms take place, especially neoangiogenesis. Immature vessels show leaky BBB, but this permeability has been associated with improved clinical recovery. In the chronic stage (>6 weeks), an increase of BBB restoration factors leads the barrier to start decreasing its permeability. Nonetheless, permeability will persist to some degree several weeks after injury. Understanding the mechanisms behind BBB dysregulation and HT pathophysiology could potentially help guide acute stroke care decisions and the development of new therapeutic targets; however, effective translation into clinical practice is still lacking. In this review, we will address the different pathological and physiological repair mechanisms involved in BBB permeability through the different stages of ischemic stroke and their role in the development of HT and stroke recovery.
Collapse
Affiliation(s)
| | - João André Sousa
- Stroke Unit, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Ana Brás
- Stroke Unit, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Carla Cecília
- Stroke Unit, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Bruno Rodrigues
- Stroke Unit, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Luciano Almendra
- Stroke Unit, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Cristina Machado
- Stroke Unit, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Gustavo Santo
- Stroke Unit, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Fernando Silva
- Stroke Unit, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Lino Ferreira
- Faculdade de Medicina da Universidade de Coimbra, Coimbra, Portugal
| | - Isabel Santana
- Stroke Unit, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Faculdade de Medicina da Universidade de Coimbra, Coimbra, Portugal
| | - João Sargento-Freitas
- Stroke Unit, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Faculdade de Medicina da Universidade de Coimbra, Coimbra, Portugal
| |
Collapse
|
14
|
Ying X, Xie Q, Li S, Yu X, Zhou K, Yue J, Chen X, Tu W, Yang G, Jiang S. Water treadmill training attenuates blood-spinal cord barrier disruption in rats by promoting angiogenesis and inhibiting matrix metalloproteinase-2/9 expression following spinal cord injury. Fluids Barriers CNS 2020; 17:70. [PMID: 33292360 PMCID: PMC7722327 DOI: 10.1186/s12987-020-00232-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 11/17/2020] [Indexed: 01/05/2023] Open
Abstract
Background The permeability of the blood-spinal cord barrier (BSCB) is mainly determined by junction complexes between adjacent endothelial cells (ECs), including tight junctions (TJs) and adherens junctions (AJs), which can be severely damaged after spinal cord injury (SCI). Exercise training is a recognized method for the treatment of SCI. The destruction of the BSCB mediated by matrix metalloproteinases (MMPs) leads to inflammation, neurotoxin production, and neuronal apoptosis. The failure of new blood vessels to effectively regenerate is also an important cause of delayed recovery after SCI. For the first time, we introduced water treadmill training (TT) to help SCI rats successfully exercise and measured the effects of TT in promoting recovery after SCI and the possible mechanisms involved. Methods Sprague-Dawley (200–250 g) rats were randomly divided into the following three groups: sham operated, SCI, and SCI + TT. Animals were sacrificed at 7 or 14 days post-surgery. The degree of neurological deficit, tissue morphology and BSCB permeability were assessed by the Basso-Beattie-Bresnahan (BBB) motor function scale and appropriate staining protocols, and apoptosis, protein expression and vascular EC ultrastructure were assessed by TUNEL staining, Western blotting, immunofluorescence and transmission electron microscopy (TEM). Results Our experiments showed that TT reduced permeability of the BSCB and decreased structural tissue damage. TT significantly improved functional recovery when compared with that in the SCI group; TJ and AJ proteins expression increased significantly after TT, and training reduced apoptosis induced by SCI. TT could promote angiogenesis, and MMP-2 and MMP-9 expression was significantly inhibited by TT. Conclusions The results of this study indicate that TT promotes functional recovery for the following reasons: TT (1) protects residual BSCB structure from further damage, (2) promotes vascular regeneration, and (3) inhibits MMP-2/9 expression to mitigate BSCB damage.
Collapse
Affiliation(s)
- Xinwang Ying
- Department of Physical Medicine and Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, Wenzhou, 325000, China
| | - Qingfeng Xie
- Department of Physical Medicine and Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, Wenzhou, 325000, China
| | - Shengcun Li
- Department of Physical Medicine and Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, Wenzhou, 325000, China
| | - Xiaolan Yu
- Department of Physical Medicine and Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, Wenzhou, 325000, China
| | - Kecheng Zhou
- Department of Physical Medicine and Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, Wenzhou, 325000, China
| | - Jingjing Yue
- Department of Physical Medicine and Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, Wenzhou, 325000, China
| | - Xiaolong Chen
- Department of Physical Medicine and Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, Wenzhou, 325000, China
| | - Wenzhan Tu
- Department of Physical Medicine and Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, Wenzhou, 325000, China
| | - Guanhu Yang
- Department of Physical Medicine and Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, Wenzhou, 325000, China
| | - Songhe Jiang
- Department of Physical Medicine and Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, Wenzhou, 325000, China. .,Department of Intelligent Rehabilitation International (Cross-Strait), Alliance of Wenzhou Medical University, Zhejiang, Wenzhou, 325000, China.
| |
Collapse
|
15
|
Douglas AS, Shearer JA, Okolo A, Pandit A, Gilvarry M, Doyle KM. The Relationship Between Cerebral Reperfusion And Regional Expression Of Matrix Metalloproteinase-9 In Rat Brain Following Focal Cerebral Ischemia. Neuroscience 2020; 453:256-265. [PMID: 33220187 DOI: 10.1016/j.neuroscience.2020.10.034] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 10/23/2020] [Accepted: 10/27/2020] [Indexed: 12/29/2022]
Abstract
We investigated the effect of full and partial mechanical reperfusion on MMP-9 expression in rat brain following middle cerebral artery occlusion, mimicking mechanical thrombectomy. Using percentage hemispheric lesion volume and oedema as measures, partial reperfusion reduced extent of brain damage caused by MCA occlusion, but the protective effect was less pronounced than with complete reperfusion. Using ELISA quantification in fresh frozen tissue, confirmed by immunofluorescence in perfusion fixed tissue, increased MMP-9 expression was observed in infarcted tissue. MMP-9 was increased in lesioned tissue of the anterior and posterior temporal cortex and underlying striatal tissue, but also the normal appearing frontal cortex. No significant increase in MMP-9 in the hippocampus was observed, nor in the unlesioned contralateral hemisphere. Both partial reperfusion and full reperfusion reduced the regional MMP expression significantly. The highest levels of MMP-9 were observed in lesioned brain regions in the non-reperfused group. MMP-9 expression was evident in microvessels and in neuronal cell bodies of affected tissue. This study shows that MMP-9 brain levels are reduced relative to the extent of reperfusion. These observations suggest targeting early increases in MMP-9 expression as a possible neuroprotective therapeutic strategy and highlight the rat MCA occlusion model as an ideal model in which to study candidate therapeutics.
Collapse
Affiliation(s)
- A S Douglas
- Department of Physiology and Galway Neuroscience Centre, School of Medicine, National University of Ireland, Galway, Ireland; CÚRAM-Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland.
| | - J A Shearer
- Department of Physiology and Galway Neuroscience Centre, School of Medicine, National University of Ireland, Galway, Ireland; CÚRAM-Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - A Okolo
- Department of Physiology and Galway Neuroscience Centre, School of Medicine, National University of Ireland, Galway, Ireland; CÚRAM-Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - A Pandit
- CÚRAM-Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | | | - K M Doyle
- Department of Physiology and Galway Neuroscience Centre, School of Medicine, National University of Ireland, Galway, Ireland; CÚRAM-Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
16
|
Abstract
Ischemic stroke, which is caused by a sudden clot in the blood vessels, may cause severe brain tissue damage and has become a leading cause of death globally. Currently, thrombolysis is the gold standard primary treatment of ischemic stroke in clinics. However, the short therapeutic window of opportunity limits thrombolysis utility. Secondary cerebral damage caused by stroke is also an urgent problem. In this review, we discuss the present methods of treating ischemic stroke in clinics and their limitations. Various new drug delivery strategies targeting ischemic stroke lesions have also been summarized, including pharmaceutical methods, diagnostic approaches and other routes. These strategies could change the pharmacokinetic behavior, improve targeted delivery or minimize side effects. A better understanding of the novel approaches utilized to facilitate drug delivery in ischemic stroke would improve outcomes.
Collapse
Affiliation(s)
- Qiong Wu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, P. R. China
| | - Rong Yan
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, P. R. China
| | - Jingjing Sun
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, P. R. China
| |
Collapse
|
17
|
Wang ZM, Liu C, Wang YY, Deng YS, He XC, Du HZ, Liu CM, Teng ZQ. SerpinA3N deficiency deteriorates impairments of learning and memory in mice following hippocampal stab injury. Cell Death Discov 2020; 6:88. [PMID: 33014432 PMCID: PMC7501238 DOI: 10.1038/s41420-020-00325-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/22/2020] [Accepted: 09/01/2020] [Indexed: 12/19/2022] Open
Abstract
Traumatic brain injury is a global leading cause of disability and death, which puts patients at high risk for developing dementia. Early intervention is believed as the key to minimize the development of brain damages that could aggravate the symptoms. Here, we report that the serine protease inhibitor SerpinA3N is upregulated in hippocampal neurons in the early stage of hippocampal stab injury (HSI), while its deficiency causes a greater degree of neuronal apoptosis and severer impairments of spatial learning and memory in mice after HSI. We further show that MMP2 is a key substrate of SerpinA3N, and MMP2 specific inhibitor (ARP100) can protect against neuronal apoptosis and cognitive dysfunction in mice after HSI. These findings demonstrate a critical role for SerpinA3N in neuroprotection, suggesting that SerpinA3N and MMP2 inhibitors might be a novel therapeutic agents for neurotrauma.
Collapse
Affiliation(s)
- Zhi-Meng Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101 Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, 100408 Beijing, China
| | - Cong Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101 Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, 100408 Beijing, China
| | - Ying-Ying Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101 Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, 100408 Beijing, China
| | - Yu-Sen Deng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101 Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, 100408 Beijing, China
| | - Xuan-Cheng He
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101 Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101 Beijing, China
| | - Hong-Zhen Du
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101 Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101 Beijing, China
| | - Chang-Mei Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101 Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, 100408 Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101 Beijing, China
| | - Zhao-Qian Teng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101 Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, 100408 Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101 Beijing, China
| |
Collapse
|
18
|
Funel N, Dini V, Janowska A, Loggini B, Minale M, Grieco F, Riccio S, Romanelli M. Triticum vulgare Extract Modulates Protein-Kinase B and Matrix Metalloproteinases 9 Protein Expression in BV-2 Cells: Bioactivity on Inflammatory Pathway Associated with Molecular Mechanism Wound Healing. Mediators Inflamm 2020; 2020:2851949. [PMID: 32189993 PMCID: PMC7063223 DOI: 10.1155/2020/2851949] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 01/04/2020] [Accepted: 02/03/2020] [Indexed: 12/12/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are a large family of ubiquitously expressed zinc-dependent enzymes with proteolitic activities. They are expressed in physiological situations and pathological conditions involving inflammatory processes including epithelial to mesenchymal transition (EMT), neuronal injury, and cancer. There is also evidence that MMPs regulate inflammation in tumor microenvironment, which plays an important role in healing tissue processes. Looking at both inflammatory and neuronal damages, MMP9 is involved in both processes and their modulation seems to be regulated by two proteins: tumor necrosis factor-alpha (TNF-alpha) and interleukin 6 (IL-6). However other important genes are involved in molecular regulation of transcription factors, protein-kinase B (AKT), and p65. In addition, Triticum vulgare extract (TVE) modulated the biological markers associated with inflammatory processes, including p65 protein. While there are no evidence that TVE might be involved in the biological modulation of other inflammatory marker as AKT, we would like to assess whether TVE is able to (1) modulate phosphorylation of AKT (pAKT) as an early marker of inflammatory process in vitro and (2) affect MMP9 protein expression in an in vitro model. The BV-2 cells (microglial of mouse) have been used as an in vitro model to simulate both inflammatory and neuronal injury pathologies. Here, MMP9 seems to be involved in cellular migration through inflammatory marker activation. We simulate an inflammatory preclinical model treating BV-2 cells with lipopolysaccharide (LPS) to induce proinflammatory activation affecting pAKT and p65 proteins. TVE is revealed to restore the native expression of AKT and p65. Additionally, TVE extract modulates also the protein concentration of MMP9. Nevertheless, immunofluorescence confocal analyses revealed that both AKT and MMP9 are regulated together, synchronously. This work seems to demonstrate that two important genes can be used to monitor the beginning of an inflammatory process, AKT and MMP9, in which TVE seems able to modulate their expression of inflammation-associated molecules.
Collapse
Affiliation(s)
- Niccola Funel
- Department of Dermatology, University of Pisa, Italy
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Lind L, Eriksson K, Grahn A. Chemokines and matrix metalloproteinases in cerebrospinal fluid of patients with central nervous system complications caused by varicella-zoster virus. J Neuroinflammation 2019; 16:42. [PMID: 30777092 PMCID: PMC6378740 DOI: 10.1186/s12974-019-1428-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 02/03/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Varicella-zoster virus (VZV) is a common viral agent causing central nervous system (CNS) infections including encephalitis, meningitis, and Ramsay Hunt syndrome. Neurological complications occur frequently despite antiviral treatment. Matrix metalloproteinases (MMPs) and cytokines are involved in the neuroinflammatory response during CNS infection. Their role in VZV CNS infections and how they differ between different CNS entities caused by VZV are poorly investigated. METHODS We analyzed the levels of 30 chemokines and 9 MMPs in cerebrospinal fluid (CSF) and serum from 66 patients with VZV CNS infections diagnosed by detection of VZV DNA in CSF and concomitant neurological symptoms and compared with a control group (n = 24). RESULTS Levels of CCL19, CXCL8, CXCL9, and CXCL10 were significantly increased and surpassing the levels in serum when analyzing all patients with VZV CNS infections whereas CXCL11 was only increased in CSF of patients with VZV meningitis. MMP-2-levels were highly elevated in CSF of all 66 VZV patients. The patients with encephalitis had the most significantly increased levels of MMPs in CSF, and MMP-3, MMP-8, and MMP-12 were exclusively increased in this group, whereas MMP-9 in CSF was increased in the patients with VZV meningitis. CONCLUSIONS We show that both chemokines and MMPs are elevated in the CSF of patients with VZV CNS infections. Encephalitis and meningitis patients differed with respect to other chemokines (CXCL11) and MMPs (MMP-3, MMP-8, MMP-9, and MMP-12), indicating that different location of the virus gives rise to qualitative differences in the ensuing inflammatory response. In addition, the pronounced increase of MMPs in CSF of the patients with encephalitis suggests an association to the severity of this manifestation, compared to VZV meningitis and Ramsay Hunt syndrome. The role of MMPs in association to chemokines should be further investigated to evaluate their significance in the neuropathogenesis of VZV CNS infections and as a potential target for new treatment alternatives.
Collapse
Affiliation(s)
- Liza Lind
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kristina Eriksson
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anna Grahn
- Department of Infectious Diseases, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
20
|
Guo X, Deng G, Liu J, Zou P, Du F, Liu F, Chen AT, Hu R, Li M, Zhang S, Tang Z, Han L, Liu J, Sheth KN, Chen Q, Gou X, Zhou J. Thrombin-Responsive, Brain-Targeting Nanoparticles for Improved Stroke Therapy. ACS NANO 2018; 12:8723-8732. [PMID: 30107729 DOI: 10.1021/acsnano.8b04787] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Current treatments for ischemic stroke are insufficient. The lack of effective pharmacological approaches can be mainly attributed to the difficulty in overcoming the blood-brain barrier. Here, we report a simple strategy to synthesize protease-responsive, brain-targeting nanoparticles for the improved treatment of stroke. The resulting nanoparticles respond to proteases enriched in the ischemic microenvironment, including thrombin or matrix metalloproteinase-9, by shrinking or expanding their size. Targeted delivery was achieved using surface conjugation of ligands that bind to proteins that were identified to enrich in the ischemic brain using protein arrays. By screening a variety of formulations, we found that AMD3100-conjugated, size-shrinkable nanoparticles (ASNPs) exhibited the greatest delivery efficiency. The brain targeting effect is mainly mediated by AMD3100, which interacts with CXCR4 that is enriched in the ischemic brain tissue. We showed that ASNPs significantly enhanced the efficacy of glyburide, a promising stroke therapeutic drug whose efficacy is limited by its toxicity. Due to their high efficiency in penetrating the ischemic brain and low toxicity, we anticipate that ASNPs have the potential to be translated into clinical applications for the improved treatment of stroke patients.
Collapse
Affiliation(s)
| | - Gang Deng
- Department of Neurosurgery , Renmin Hospital of Wuhan University , Wuhan , Hubei 430060 , China
| | | | | | | | | | | | | | | | - Shenqi Zhang
- Department of Neurosurgery , Renmin Hospital of Wuhan University , Wuhan , Hubei 430060 , China
| | - Zhishu Tang
- Shaanxi Key Laboratory of Brain Disorders & Institute of Basic and Translational Medicine , Xi'an Medical University , Xi'an , Shannxi 710021 , China
| | | | - Jie Liu
- Department of Biomedical Engineering, School of Engineering , Sun Yat-sen University , Guangzhou , Guangdong 510006 , China
| | | | - Qianxue Chen
- Department of Neurosurgery , Renmin Hospital of Wuhan University , Wuhan , Hubei 430060 , China
| | - Xingchun Gou
- Shaanxi Key Laboratory of Brain Disorders & Institute of Basic and Translational Medicine , Xi'an Medical University , Xi'an , Shannxi 710021 , China
| | | |
Collapse
|
21
|
Yao Y, Xu J, Yu T, Chen Z, Xiao Z, Wang J, Hu Y, Wu Y, Zhu D. Flufenamic acid inhibits secondary hemorrhage and BSCB disruption after spinal cord injury. Am J Cancer Res 2018; 8:4181-4198. [PMID: 30128046 PMCID: PMC6096396 DOI: 10.7150/thno.25707] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Accepted: 06/19/2018] [Indexed: 12/22/2022] Open
Abstract
Acute spinal cord injury (SCI) induces secondary hemorrhage and initial blood-spinal cord barrier (BSCB) disruption. The transient receptor potential melastatin 4 (Trpm4) together with sulfonylurea receptor 1 (Sur1) forms the Sur1-Trpm4 channel complex. The up-regulation of Sur1-Trpm4 after injury plays a crucial role in secondary hemorrhage, which is the most destructive mechanism in secondary injuries of the central nervous system (CNS). The matrix metalloprotease (MMP)-mediated disruption of the BSCB leads to an inflammatory response, neurotoxin production and neuronal cell apoptosis. Thus, preventing secondary hemorrhage and BSCB disruption should be an important goal of therapeutic interventions in SCI. Methods: Using a moderate contusion injury model at T10 of the spinal cord, flufenamic acid (FFA) was injected intraperitoneally 1 h after SCI and then continuously once per day for one week. Results: Trpm4 expression is highly up-regulated in capillaries 1 d after SCI. Treatment with flufenamic acid (FFA) inhibited Trpm4 expression, secondary hemorrhage, and capillary fragmentation and promoted angiogenesis. In addition, FFA significantly inhibited the expression of MMP-2 and MMP-9 at 1 d after SCI and significantly attenuated BSCB disruption at 1 d and 3 d after injury. Furthermore, we found that FFA decreased the hemorrhage- and BSCB disruption-induced activation of microglia/macrophages and was associated with smaller lesions, decreased cavity formation, better myelin preservation and less reactive gliosis. Finally, FFA protected motor neurons and improved locomotor functions after SCI. Conclusion: This study indicates that FFA improves functional recovery, in part, due to the following reasons: (1) it inhibits the expression of Trpm4 to reduce the secondary hemorrhage; and (2) it inhibits the expression of MMP-2 and MMP-9 to block BSCB disruption. Thus, the results of our study suggest that FFA may represent a potential therapeutic agent for promoting functional recovery.
Collapse
|
22
|
Autophagy- and MMP-2/9-mediated Reduction and Redistribution of ZO-1 Contribute to Hyperglycemia-increased Blood–Brain Barrier Permeability During Early Reperfusion in Stroke. Neuroscience 2018. [DOI: 10.1016/j.neuroscience.2018.02.035] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
23
|
Thomsen MS, Routhe LJ, Moos T. The vascular basement membrane in the healthy and pathological brain. J Cereb Blood Flow Metab 2017; 37:3300-3317. [PMID: 28753105 PMCID: PMC5624399 DOI: 10.1177/0271678x17722436] [Citation(s) in RCA: 317] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 06/21/2017] [Accepted: 06/28/2017] [Indexed: 12/24/2022]
Abstract
The vascular basement membrane contributes to the integrity of the blood-brain barrier (BBB), which is formed by brain capillary endothelial cells (BCECs). The BCECs receive support from pericytes embedded in the vascular basement membrane and from astrocyte endfeet. The vascular basement membrane forms a three-dimensional protein network predominantly composed of laminin, collagen IV, nidogen, and heparan sulfate proteoglycans that mutually support interactions between BCECs, pericytes, and astrocytes. Major changes in the molecular composition of the vascular basement membrane are observed in acute and chronic neuropathological settings. In the present review, we cover the significance of the vascular basement membrane in the healthy and pathological brain. In stroke, loss of BBB integrity is accompanied by upregulation of proteolytic enzymes and degradation of vascular basement membrane proteins. There is yet no causal relationship between expression or activity of matrix proteases and the degradation of vascular matrix proteins in vivo. In Alzheimer's disease, changes in the vascular basement membrane include accumulation of Aβ, composite changes, and thickening. The physical properties of the vascular basement membrane carry the potential of obstructing drug delivery to the brain, e.g. thickening of the basement membrane can affect drug delivery to the brain, especially the delivery of nanoparticles.
Collapse
Affiliation(s)
- Maj S Thomsen
- Laboratory of Neurobiology, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Lisa J Routhe
- Laboratory of Neurobiology, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Torben Moos
- Laboratory of Neurobiology, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| |
Collapse
|
24
|
Chen XJ, Zhang JG, Wu L. Plumbagin inhibits neuronal apoptosis, intimal hyperplasia and also suppresses TNF-α/NF-κB pathway induced inflammation and matrix metalloproteinase-2/9 expression in rat cerebral ischemia. Saudi J Biol Sci 2017; 25:1033-1039. [PMID: 30174499 PMCID: PMC6116857 DOI: 10.1016/j.sjbs.2017.03.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Revised: 02/24/2017] [Accepted: 03/13/2017] [Indexed: 12/13/2022] Open
Abstract
Cerebral ischemic damage and infarction are well documented in stroke, which is presenting a foremost health concern globally with very high mortality and morbidity rates. Mechanisms that are associated with excitotoxicity, inflammation and oxidative stress are found to be critically involved in ischemic damage. Adverse effects of current therapies are imposing the need in development of neuroprotective agents that are very effective. To explore this we experimentally induced ischemic brain injury and investigated the effects of plumbagin. Induction of cerebral infarction and ischemia-reperfusion (I/R) was done by middle cerebral artery occlusion (MCAO) in Sprague-Dawley rats. Plumbagin (50, 100 or 200 mg/kg b.wt) was intragastrically administered for 9 days before ischemia induction and an hour prior on the day of ischemic insult. Plumbagin treatment attenuated pulmonary edema, neuronal apoptosis and reduced cerebral infarct volume. Cleaved caspase-3 and apoptotic cascade protein expressions were regulated. Overproduction of pro-inflammatory cytokines (TNF-α, IL-1β and IL-6) and nitric oxide (NO) following I/R were reduced. Prior plumbagin administration had down-regulated NF-κB signalling and MMP-2 and MMP-9 expression. Overall, the results reveal the potent neuroprotective efficacy of plumbagin against I/R-induced brain injury via effectively modulating apoptotic pathways, MMPs and neuro-inflammatory cascades.
Collapse
Affiliation(s)
- Xiao-Juan Chen
- Department of Neurology, Linyi People's Hospital, Linyi 276003, Shandong, China
| | - Jian-Guo Zhang
- Department of Critical Care Medicine, Linyi People's Hospital, Linyi 276003, Shandong, China
| | - Lan Wu
- Department of Neurology, Linyi People's Hospital, Linyi 276003, Shandong, China
| |
Collapse
|
25
|
Sun Q, Liao Y, Wang T, Tang H, Wang G, Zhao F, Jin Y. 2-Chloroethanol Induced Upregulation of Matrix Metalloproteinase-2 in Primary Cultured Rat Astrocytes Via MAPK Signal Pathways. Front Neurosci 2017; 10:593. [PMID: 28101000 PMCID: PMC5209348 DOI: 10.3389/fnins.2016.00593] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Accepted: 12/12/2016] [Indexed: 01/10/2023] Open
Abstract
This study was to explore the mechanisms underlying 1,2-dichloroethane (1,2-DCE) induced brain edema by focusing on alteration of matrix metalloproteinase-2 (MMP-2) in rat astrocytes induced by 2-chloroethanol (2-CE), an intermediate metabolite of 1,2-DCE in vivo. Protein and mRNA levels of MMP-2, and the phosphorylated protein levels of p38 MAPK (p-p38), extracellular signal regulated protein kinase (p-ERK1/2) and c-Jun N-terminal kinase (p-JNK1/2) in astrocytes were examined by immunostaining, western blot or real-time RT-PCR analysis. Findings from this study disclosed that protein levels of MMP-2 were upregulated by 2-CE in astrocytes. Meanwhile, protein levels of p-p38, p-ERK1/2 and p-JNK1/2 were also increased apparently in the cells treated with 2-CE. Moreover, pretreatment of astrocytes with SB202190 (inhibitor of p38 MAPK), U0126 (inhibitor of ERK1/2) or SP600125 (inhibitor of JNK1/2) could suppress the upregulated expression of p-p38, p-ERK1/2, and p-JNK1/2. In response to suppressed protein levels of p-p38 and p-JNK1/2, the protein levels of MMP-2 also decreased significantly, indicating that activation of MAPK signal pathways were involved in the mechanisms underlying 2-CE-induced upregulation of MMP-2 expression.
Collapse
Affiliation(s)
- Qi Sun
- Department of Occupational and Environmental Health, School of Public Health, China Medical University Shenyang, China
| | - Yingjun Liao
- Department of Physiology, China Medical University Shenyang, China
| | - Tong Wang
- Department of Occupational and Environmental Health, School of Public Health, China Medical University Shenyang, China
| | - Hongge Tang
- Department of Occupational and Environmental Health, School of Public Health, China Medical University Shenyang, China
| | - Gaoyang Wang
- Department of Occupational and Environmental Health, School of Public Health, China Medical University Shenyang, China
| | - Fenghong Zhao
- Department of Occupational and Environmental Health, School of Public Health, China Medical University Shenyang, China
| | - Yaping Jin
- Department of Occupational and Environmental Health, School of Public Health, China Medical University Shenyang, China
| |
Collapse
|
26
|
Chen S, Cui J, Jiang T, Olson ES, Cai QY, Yang M, Wu W, Guthrie JM, Robertson JD, Lipton SA, Ma L, Tsien RY, Gu Z. Gelatinase activity imaged by activatable cell-penetrating peptides in cell-based and in vivo models of stroke. J Cereb Blood Flow Metab 2017; 37:188-200. [PMID: 26681768 PMCID: PMC5363737 DOI: 10.1177/0271678x15621573] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 10/09/2015] [Accepted: 11/04/2015] [Indexed: 12/27/2022]
Abstract
Matrix metalloproteinases (MMPs), particularly gelatinases (MMP-2/-9), are involved in neurovascular impairment after stroke. Detection of gelatinase activity in vivo can provide insight into blood-brain barrier disruption, hemorrhage, and nerve cell injury or death. We applied gelatinase-activatable cell-penetrating peptides (ACPP) with a cleavable l-amino acid linker to examine gelatinase activity in primary neurons in culture and ischemic mouse brain in vivo We found uptake of Cy5-conjugated ACPP (ACPP-Cy5) due to gelatinase activation both in cultured neurons exposed to n-methyl-d-aspartate and in mice after cerebral ischemia. Fluorescence intensity was significantly reduced when cells or mice were treated with MMP inhibitors or when a cleavage-resistant ACPP-Cy5 was substituted. We also applied an ACPP dendrimer (ACPPD) conjugated with multiple Cy5 and/or gadolinium moieties for fluorescence and magnetic resonance imaging (MRI) in intact animals. Fluorescence analysis showed that ACPPD was detected in sub-femtomole range in ischemic tissues. Moreover, MRI and inductively coupled plasma mass spectrometry revealed that ACPPD produced quantitative measures of gelatinase activity in the ischemic region. The resulting spatial pattern of gelatinase activity and neurodegeneration were very similar. We conclude that ACPPs are capable of tracing spatiotemporal gelatinase activity in vivo, and will therefore be useful in elucidating mechanisms of gelatinase-mediated neurodegeneration after stroke.
Collapse
Affiliation(s)
- Shanyan Chen
- Department of Pathology and Anatomical Sciences, University of Missouri at Columbia, USA.,Center for Translational Neuroscience, University of Missouri at Columbia, USA
| | - Jiankun Cui
- Department of Pathology and Anatomical Sciences, University of Missouri at Columbia, USA.,Center for Translational Neuroscience, University of Missouri at Columbia, USA.,Truman VA Hospital Research Service
| | - Tao Jiang
- Department of Pharmacology, University of California San Diego School of Medicine, USA
| | - Emilia S Olson
- Department of Pharmacology, University of California San Diego School of Medicine, USA
| | - Quan-Yu Cai
- Truman VA Hospital Research Service.,Department of Radiology, University of Missouri at Columbia, USA
| | - Ming Yang
- Truman VA Hospital Research Service.,Department of Radiology, University of Missouri at Columbia, USA
| | - Wei Wu
- Department of Pathology and Anatomical Sciences, University of Missouri at Columbia, USA.,Center for Translational Neuroscience, University of Missouri at Columbia, USA
| | - James M Guthrie
- Research Reactor Center, University of Missouri at Columbia, USA
| | - J D Robertson
- Research Reactor Center, University of Missouri at Columbia, USA
| | - Stuart A Lipton
- Department of Neurosciences, University of California San Diego School of Medicine, USA.,Scintillon Institute Neurodegenerative Disease Center, USA
| | - Lixin Ma
- Truman VA Hospital Research Service.,Department of Radiology, University of Missouri at Columbia, USA
| | - Roger Y Tsien
- Department of Pharmacology, University of California San Diego School of Medicine, USA.,Howard Hughes Medical Institute, University of California San Diego School of Medicine, USA
| | - Zezong Gu
- Department of Pathology and Anatomical Sciences, University of Missouri at Columbia, USA .,Center for Translational Neuroscience, University of Missouri at Columbia, USA.,Truman VA Hospital Research Service
| |
Collapse
|
27
|
The multifaceted role of metalloproteinases in physiological and pathological conditions in embryonic and adult brains. Prog Neurobiol 2016; 155:36-56. [PMID: 27530222 DOI: 10.1016/j.pneurobio.2016.08.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 07/10/2016] [Accepted: 08/08/2016] [Indexed: 02/07/2023]
Abstract
Matrix metalloproteinases (MMPs) are a large family of ubiquitous extracellular endopeptidases, which play important roles in a variety of physiological and pathological conditions, from the embryonic stages throughout adult life. Their extraordinary physiological "success" is due to concomitant broad substrate specificities and strict regulation of their expression, activation and inhibition levels. In recent years, MMPs have gained increasing attention as significant effectors in various aspects of central nervous system (CNS) physiology. Most importantly, they have been recognized as main players in a variety of brain disorders having different etiologies and evolution. A common aspect of these pathologies is the development of acute or chronic neuroinflammation. MMPs play an integral part in determining the result of neuroinflammation, in some cases turning its beneficial outcome into a harmful one. This review summarizes the most relevant studies concerning the physiology of MMPs, highlighting their involvement in both the developing and mature CNS, in long-lasting and acute brain diseases and, finally, in nervous system repair. Recently, a concerted effort has been made in identifying therapeutic strategies for major brain diseases by targeting MMP activities. However, from this revision of the literature appears clear that MMPs have multifaceted functional characteristics, which modulate physiological processes in multiple ways and with multiple consequences. Therefore, when choosing MMPs as possible targets, great care must be taken to evaluate the delicate balance between their activation and inhibition and to determine at which stage of the disease and at what level they become active in order maximize chances of success.
Collapse
|
28
|
Han L, Cai Q, Tian D, Kong DK, Gou X, Chen Z, Strittmatter SM, Wang Z, Sheth KN, Zhou J. Targeted drug delivery to ischemic stroke via chlorotoxin-anchored, lexiscan-loaded nanoparticles. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 12:1833-1842. [PMID: 27039220 DOI: 10.1016/j.nano.2016.03.005] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 02/25/2016] [Accepted: 03/15/2016] [Indexed: 12/24/2022]
Abstract
Ischemic stroke is a leading cause of disability and death worldwide. Current drug treatment for stroke remains inadequate due to the existence of the blood-brain barrier. We proposed an innovative nanotechnology-based autocatalytic targeting approach, in which the blood-brain barrier modulator lexiscan is encapsulated in nanoparticles to enhance blood-brain barrier permeability and autocatalytically augment the brain stroke-targeting delivery efficiency of chlorotoxin-anchored nanoparticles. The nanoparticles efficiently and specifically accumulated in the brain ischemic microenvironment and the targeting efficiency autocatalytically increased with subsequent administrations. When Nogo-66 receptor antagonist peptide NEP1-40, a potential therapeutic agent for ischemic stroke, was loaded, nanoparticles significantly reduced infarct volumes and enhanced survival. Our findings suggest that the autocatalytic targeting approach is a promising strategy for drug delivery to the ischemic microenvironment inside the brain. Nanoparticles developed in this study may serve as a new approach for the clinical management of stroke.
Collapse
Affiliation(s)
- Liang Han
- Department of Neurosurgery, Yale University, New Haven, CT, USA; School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Qiang Cai
- Department of Neurosurgery, Yale University, New Haven, CT, USA; Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Daofeng Tian
- Department of Neurosurgery, Yale University, New Haven, CT, USA; Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Derek K Kong
- Department of Neurosurgery, Yale University, New Haven, CT, USA
| | - Xingchun Gou
- Department of Neurosurgery, Yale University, New Haven, CT, USA; The laboratory of Cell Biology and Translational Medicine, Xi'an Medical University, Xi'an, China
| | - Zeming Chen
- Department of Neurosurgery, Yale University, New Haven, CT, USA
| | | | - Zuoheng Wang
- Division of Biostatistics, School of Public Health, Yale University, New Haven, CT, USA
| | - Kevin N Sheth
- Department of Neurosurgery, Yale University, New Haven, CT, USA; Department of Neurology, Yale University, New Haven, CT, USA
| | - Jiangbing Zhou
- Department of Neurosurgery, Yale University, New Haven, CT, USA; Department of Biomedical Engineering, Yale University, New Haven, CT, USA.
| |
Collapse
|
29
|
Turner RJ, Sharp FR. Implications of MMP9 for Blood Brain Barrier Disruption and Hemorrhagic Transformation Following Ischemic Stroke. Front Cell Neurosci 2016; 10:56. [PMID: 26973468 PMCID: PMC4777722 DOI: 10.3389/fncel.2016.00056] [Citation(s) in RCA: 341] [Impact Index Per Article: 37.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 02/22/2016] [Indexed: 02/03/2023] Open
Abstract
Numerous studies have documented increases in matrix metalloproteinases (MMPs), specifically MMP-9 levels following stroke, with such perturbations associated with disruption of the blood brain barrier (BBB), increased risk of hemorrhagic complications, and worsened outcome. Despite this, controversy remains as to which cells release MMP-9 at the normal and pathological BBB, with even less clarity in the context of stroke. This may be further complicated by the influence of tissue plasminogen activator (tPA) treatment. The aim of the present review is to examine the relationship between neutrophils, MMP-9 and tPA following ischemic stroke to elucidate which cells are responsible for the increases in MMP-9 and resultant barrier changes and hemorrhage observed following stroke.
Collapse
Affiliation(s)
- Renée J Turner
- Discipline of Anatomy and Pathology, Adelaide Centre for Neuroscience Research, School of Medicine, The University of Adelaide Adelaide, SA, Australia
| | - Frank R Sharp
- Department of Neurology, MIND Institute, University of California at Davis Medical Center Sacramento, CA, USA
| |
Collapse
|
30
|
Mechanisms of Thrombosis and Thrombolysis. Stroke 2016. [DOI: 10.1016/b978-0-323-29544-4.00002-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
31
|
Abstract
In this issue of Blood, Hijazi et al challenge the view that consumptive coagulopathy that accompanies traumatic brain injury (TBI) results in a sequence of events that lead to intracranial hemorrhage (ICH).
Collapse
|
32
|
Chen W, Guo Y, Yang W, Zheng P, Zeng J, Tong W. Protective effect of ginsenoside Rb1 on integrity of blood-brain barrier following cerebral ischemia. Exp Brain Res 2015; 233:2823-31. [PMID: 26070903 DOI: 10.1007/s00221-015-4352-3] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 06/01/2015] [Indexed: 11/30/2022]
Abstract
Ginsenosides, the major bioactive compounds in ginseng root, have been found to have antioxidant, immunomodulatory, and anti-inflammatory activities. In the present study, we sought to investigate whether and how ginsenoside Rb1 (GS-Rb1), the most abundant ginsenoside, can protect blood-brain barrier (BBB) integrity following cerebral ischemia in middle cerebral artery occlusion (MCAO) animal model. ICR mice underwent MCAO and received GS-Rb1 by intraperitoneal injection at 3 h after reperfusion. We evaluated infarction, neurological scores, brain edema, Evans blue (EB) extravasation, and tight junction protein expression at 48 h after MCAO. We further examined whether GS-Rb1 protected BBB integrity by suppressing post-ischemic inflammation-induced activity of matrix metalloproteinase-9 (MMP-9) and nicotinamide adenine dinucleotide phosphate oxidase (NOX). First, GS-Rb1 decreased infarction and improved neurological deficits in MCAO animals. In addition, GS-Rb1 reduced EB extravasation and brain edema and preserved expression of tight junction proteins in the ischemic brain. Moreover, GS-Rb1 inhibited expression of pro-inflammatory factors including nitric oxide synthase and IL-1β, but increased expression of anti-inflammatory markers arginase 1 and IL-10 in the ischemic brain. Consistently, GS-Rb1 attenuated ischemia-induced expression and activity of MMP9. Finally, GS-Rb1 reduced NOX-4 mRNA expression and NOX activity in ischemic brain. These results suggest that GS-Rb1 protects loss of BBB integrity in ischemic stroke by suppressing neuroinflammation induction of MMP-9 and NOX4-derived free radicals, and indicate its potential for treating brain injuries, such as ischemia and stroke.
Collapse
Affiliation(s)
- Wei Chen
- The People's Hospital of Pu Dong New Area, 490 South Chuanhuan Road, Chuansha New Town, Shanghai, 201299, People's Republic of China
| | - Yijun Guo
- The People's Hospital of Pu Dong New Area, 490 South Chuanhuan Road, Chuansha New Town, Shanghai, 201299, People's Republic of China
| | - Wenjin Yang
- The People's Hospital of Pu Dong New Area, 490 South Chuanhuan Road, Chuansha New Town, Shanghai, 201299, People's Republic of China
| | - Ping Zheng
- The People's Hospital of Pu Dong New Area, 490 South Chuanhuan Road, Chuansha New Town, Shanghai, 201299, People's Republic of China
| | - Jinsong Zeng
- The People's Hospital of Pu Dong New Area, 490 South Chuanhuan Road, Chuansha New Town, Shanghai, 201299, People's Republic of China
| | - Wusong Tong
- The People's Hospital of Pu Dong New Area, 490 South Chuanhuan Road, Chuansha New Town, Shanghai, 201299, People's Republic of China.
| |
Collapse
|
33
|
Schomberg D, Miranpuri G, Duellman T, Crowell A, Vemuganti R, Resnick D. Spinal cord injury induced neuropathic pain: Molecular targets and therapeutic approaches. Metab Brain Dis 2015; 30:645-58. [PMID: 25588751 DOI: 10.1007/s11011-014-9642-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 12/05/2014] [Indexed: 10/24/2022]
Abstract
Neuropathic pain, especially that resulting from spinal cord injury, is a tremendous clinical challenge. A myriad of biological changes have been implicated in producing these pain states including cellular interactions, extracellular proteins, ion channel expression, and epigenetic influences. Physiological consequences of these changes are varied and include functional deficits and pain responses. Developing therapies that effectively address the cause of these symptoms require a deeper knowledge of alterations in the molecular pathways. Matrix metalloproteinases and tissue inhibitors of metalloproteinases are two promising therapeutic targets. Matrix metalloproteinases interact with and influence many of the studied pain pathways. Gene expression of ion channels and inflammatory mediators clearly contributes to neuropathic pain. Localized and time dependent targeting of these proteins could alleviate and even prevent neuropathic pain from developing. Current therapeutic options for neuropathic pain are limited primarily to analgesics targeting the opioid pathway. Therapies directed at molecular targets are highly desirable and in early stages of development. These include transplantation of exogenously engineered cell populations and targeted gene manipulation. This review describes specific molecular targets amenable to therapeutic intervention using currently available delivery systems.
Collapse
Affiliation(s)
- Dominic Schomberg
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, 600 Highland Ave, Madison, WI, 53792, USA
| | | | | | | | | | | |
Collapse
|
34
|
Eckert A, Huang L, Gonzalez R, Kim HS, Hamblin MH, Lee JP. Bystander Effect Fuels Human Induced Pluripotent Stem Cell-Derived Neural Stem Cells to Quickly Attenuate Early Stage Neurological Deficits After Stroke. Stem Cells Transl Med 2015; 4:841-51. [PMID: 26025980 DOI: 10.5966/sctm.2014-0184] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 04/06/2015] [Indexed: 12/16/2022] Open
Abstract
UNLABELLED : Present therapies for stroke rest with tissue plasminogen activator (tPA), the sole licensed antithrombotic on the market; however, tPA's effectiveness is limited in that the drug not only must be administered less than 3-5 hours after stroke but often exacerbates blood-brain barrier (BBB) leakage and increases hemorrhagic incidence. A potentially promising therapy for stroke is transplantation of human induced pluripotent stem cell-derived neural stem cells (hiPSC-NSCs). To date, the effects of iPSCs on injuries that take place during early stage ischemic stroke have not been well studied. Consequently, we engrafted iPSC-NSCs into the ipsilesional hippocampus, a natural niche of NSCs, at 24 hours after stroke (prior to secondary BBB opening and when inflammatory signature is abundant). At 48 hours after stroke (24 hours after transplant), hiPSC-NSCs had migrated to the stroke lesion and quickly improved neurological function. Transplanted mice showed reduced expression of proinflammatory factors (tumor necrosis factor-α, interleukin 6 [IL-6], IL-1β, monocyte chemotactic protein 1, macrophage inflammatory protein 1α), microglial activation, and adhesion molecules (intercellular adhesion molecule 1, vascular cell adhesion molecule 1) and attenuated BBB damage. We are the first to report that engrafted hiPSC-NSCs rapidly improved neurological function (less than 24 hours after transplant). Rapid hiPSC-NSC therapeutic activity is mainly due to a bystander effect that elicits reduced inflammation and BBB damage. SIGNIFICANCE Clinically, cerebral vessel occlusion is rarely permanent because of spontaneous or thrombolytic therapy-mediated reperfusion. These results have clinical implications indicating a much extended therapeutic window for transplantation of human induced pluripotent stem cell-derived neural stem cells (hiPSC-NSCs; 24 hours after stroke as opposed to the 5-hour window with tissue plasminogen activator [tPA]). In addition, there is potential for a synergistic effect by combining hiPSC-NSC transplantation with tPA to attenuate stroke's adverse effects.
Collapse
Affiliation(s)
- Auston Eckert
- Center for Stem Cell Research and Regenerative Medicine, Department of Neurology, Tulane University School of Medicine, New Orleans, Louisiana, USA; Sanford-Burnham Institute for Medical Research, Neuroscience, Aging and Stem Cell Research, La Jolla, California, USA; Seoul National University, College of Medicine, Department of Pharmacology, Seoul, Republic of Korea
| | - Lei Huang
- Center for Stem Cell Research and Regenerative Medicine, Department of Neurology, Tulane University School of Medicine, New Orleans, Louisiana, USA; Sanford-Burnham Institute for Medical Research, Neuroscience, Aging and Stem Cell Research, La Jolla, California, USA; Seoul National University, College of Medicine, Department of Pharmacology, Seoul, Republic of Korea
| | - Rodolfo Gonzalez
- Center for Stem Cell Research and Regenerative Medicine, Department of Neurology, Tulane University School of Medicine, New Orleans, Louisiana, USA; Sanford-Burnham Institute for Medical Research, Neuroscience, Aging and Stem Cell Research, La Jolla, California, USA; Seoul National University, College of Medicine, Department of Pharmacology, Seoul, Republic of Korea
| | - Hye-Sun Kim
- Center for Stem Cell Research and Regenerative Medicine, Department of Neurology, Tulane University School of Medicine, New Orleans, Louisiana, USA; Sanford-Burnham Institute for Medical Research, Neuroscience, Aging and Stem Cell Research, La Jolla, California, USA; Seoul National University, College of Medicine, Department of Pharmacology, Seoul, Republic of Korea
| | - Milton H Hamblin
- Center for Stem Cell Research and Regenerative Medicine, Department of Neurology, Tulane University School of Medicine, New Orleans, Louisiana, USA; Sanford-Burnham Institute for Medical Research, Neuroscience, Aging and Stem Cell Research, La Jolla, California, USA; Seoul National University, College of Medicine, Department of Pharmacology, Seoul, Republic of Korea
| | - Jean-Pyo Lee
- Center for Stem Cell Research and Regenerative Medicine, Department of Neurology, Tulane University School of Medicine, New Orleans, Louisiana, USA; Sanford-Burnham Institute for Medical Research, Neuroscience, Aging and Stem Cell Research, La Jolla, California, USA; Seoul National University, College of Medicine, Department of Pharmacology, Seoul, Republic of Korea
| |
Collapse
|
35
|
Yang Y, Rosenberg GA. Matrix metalloproteinases as therapeutic targets for stroke. Brain Res 2015; 1623:30-8. [PMID: 25916577 DOI: 10.1016/j.brainres.2015.04.024] [Citation(s) in RCA: 178] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 04/11/2015] [Accepted: 04/13/2015] [Indexed: 01/14/2023]
Abstract
Matrix metalloproteinases (MMPs) are important in injury and recovery in ischemic injury. They are proteolytic enzymes that degrade all components of the extracellular matrix (ECM). They are secreted in a latent form, protecting the cell from damage, but once activated induce injury prior to rapid inactivation by four tissue inhibitors to metalloproteinases (TIMPs). Normally the constitutive enzymes, MMP-2 and membrane type MMP (MMP-14), are activated in a spatially specific manner and act close to the site of activation, while the inducible enzymes, MMP-3 and MMP-9, become active through the action of free radicals and other enzymes during neuroinflammation. Because of the complex nature of the interactions with tissues during development, injury and repair, the MMPs have multiple roles, participating in the injury process in the early stages and contributing to recovery during the later stages. This dual role complicates the planning of treatment strategies. This article is part of a Special Issue entitled SI: Cell Interactions In Stroke.
Collapse
Affiliation(s)
- Yi Yang
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - Gary A Rosenberg
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
| |
Collapse
|
36
|
Neuropathic pain: role of inflammation, immune response, and ion channel activity in central injury mechanisms. Ann Neurosci 2014; 19:125-32. [PMID: 25205985 PMCID: PMC4117080 DOI: 10.5214/ans.0972.7531.190309] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2012] [Revised: 06/30/2012] [Accepted: 07/27/2012] [Indexed: 01/11/2023] Open
Abstract
Neuropathic pain (NP) is a significant and disabling clinical problem with very few therapeutic treatment options available. A major priority is to identify the molecular mechanisms responsible for NP. Although many seemingly relevant pathways have been identified, more research is needed before effective clinical interventions can be produced. Initial insults to the nervous system, such as spinal cord injury (SCI), are often compounded by secondary mechanisms such as inflammation, the immune response, and the changing expression of receptors and ion channels. The consequences of these secondary effects myriad and compound those elicited by the primary injury. Chronic NP syndromes following SCI can greatly complicate the clinical treatment of the primary injury and result in high comorbidity. In this review, we will describe physiological outcomes associated with SCI along with some of the mechanisms known to contribute to chronic NP development.
Collapse
|
37
|
Amantea D, Certo M, Russo R, Bagetta G, Corasaniti MT, Tassorelli C. Early reperfusion injury is associated to MMP2 and IL-1β elevation in cortical neurons of rats subjected to middle cerebral artery occlusion. Neuroscience 2014; 277:755-63. [PMID: 25108165 DOI: 10.1016/j.neuroscience.2014.07.064] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 07/14/2014] [Accepted: 07/29/2014] [Indexed: 12/27/2022]
Abstract
The pathophysiological processes implicated in ischemic brain damage are strongly affected by an inflammatory reaction characterized by activation of immune cells and release of soluble mediators, including cytokines and chemokines. The pro-inflammatory cytokine interleukin (IL)-1β has been implicated in ischemic brain injury, however, to date, the mechanisms involved in the maturation of this cytokine in the ischemic brain have not been completely elucidated. We have previously suggested that matrix metalloproteinases (MMPs) may be implicated in cytokine production under pathological conditions. Here, we demonstrate that significant elevation of IL-1β occurs in the cortex as early as 1h after the beginning of reperfusion in rats subjected to 2-h middle cerebral artery occlusion (MCAo). At this early stage, we observe increased expression of IL-1β in pericallosal astroglial cells and in cortical neurons and this latter signal colocalizes with elevated gelatinolytic activity. By gel zymography, we demonstrate that the increased gelatinolytic signal at 1-h reperfusion is mainly ascribed to MMP2. Thus, MMP2 seems to contribute to early brain elevation of IL-β after transient ischemia and this mechanism may promote damage since pharmacological inhibition of gelatinases by the selective MMP2/MMP9 inhibitor V provides neuroprotection in rats subjected to transient MCAo.
Collapse
Affiliation(s)
- D Amantea
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Italy.
| | - M Certo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Italy
| | - R Russo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Italy
| | - G Bagetta
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Italy
| | - M T Corasaniti
- Department of Health Sciences, University Magna Graecia of Catanzaro, Italy
| | - C Tassorelli
- IRCCS National Neurological Institute C. Mondino Foundation, Pavia, Italy
| |
Collapse
|
38
|
Plasmin-dependent modulation of the blood-brain barrier: a major consideration during tPA-induced thrombolysis? J Cereb Blood Flow Metab 2014; 34:1283-96. [PMID: 24896566 PMCID: PMC4126105 DOI: 10.1038/jcbfm.2014.99] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Revised: 05/09/2014] [Accepted: 05/09/2014] [Indexed: 01/16/2023]
Abstract
Plasmin, the principal downstream product of tissue-type plasminogen activator (tPA), is known for its potent fibrin-degrading capacity but is also recognized for many non-fibrinolytic activities. Curiously, plasmin has not been conclusively linked to blood-brain barrier (BBB) disruption during recombinant tPA (rtPA)-induced thrombolysis in ischemic stroke. This is surprising given the substantial involvement of tPA in the modulation of BBB permeability and the co-existence of tPA and plasminogen in both blood and brain throughout the ischemic event. Here, we review the work that argues a role for plasmin together with endogenous tPA or rtPA in BBB alteration, presenting the overall controversy around the topic yet creating a rational case for an involvement of plasmin in this process.
Collapse
|
39
|
Kim YS, Joh TH. Matrix metalloproteinases, new insights into the understanding of neurodegenerative disorders. Biomol Ther (Seoul) 2014; 20:133-43. [PMID: 24116286 PMCID: PMC3792209 DOI: 10.4062/biomolther.2012.20.2.133] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Revised: 03/15/2012] [Accepted: 03/15/2012] [Indexed: 12/01/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are a subfamily of zinc-dependent proteases that are responsible for degradation and remodeling of extracellular matrix proteins. The activity of MMPs is tightly regulated at several levels including cleavage of prodomain, allosteric activation, compartmentalization and complex formation with tissue inhibitor of metalloproteinases (TIMPs). In the central nervous system (CNS), MMPs play a wide variety of roles ranging from brain development, synaptic plasticity and repair after injury to the pathogenesis of various brain disorders. Following general discussion on the domain structure and the regulation of activity of MMPs, we emphasize their implication in various brain disorder conditions such as Alzheimer’s disease, multiple sclerosis, ischemia/reperfusion and Parkinson’s disease. We further highlight accumulating evidence that MMPs might be the culprit in Parkinson’s disease (PD). Among them, MMP-3 appears to be involved in a range of pathogenesis processes in PD including neuroinflammation, apoptosis and degradation of α-synuclein and DJ-1. MMP inhibitors could represent potential novel therapeutic strategies for treatments of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yoon-Seong Kim
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, 32827
| | | |
Collapse
|
40
|
Sung M, Wei E, Chavez E, Jain N, Levano S, Binkert L, Ramseier A, Setz C, Bodmer D, Ryan AF, Brand Y. Inhibition of MMP-2 but not MMP-9 influences inner ear spiral ganglion neurons in vitro. Cell Mol Neurobiol 2014; 34:1011-21. [PMID: 24935409 DOI: 10.1007/s10571-014-0077-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Accepted: 05/31/2014] [Indexed: 10/25/2022]
Abstract
Matrix metalloproteinases (MMPs) play an important role in modeling of the extracellular matrix. There is increasing evidence that these proteases are important in neurite elongation and axonal guidance during development in the central nervous system and retina. Moreover, they are also expressed after acute injury and can be the key mediators of pathogenesis. However, the role of MMPs in the inner ear is largely unknown. Our group recently demonstrated that general inhibition of MMPs resulted in auditory hair cell loss in vitro. In the present study, we investigated the role of MMPs in inner ear spiral ganglion neuron (SGN) survival, neuritogenesis and neurite extension by blocking MMPs known to be involved in axonal guidance, neurite elongation, and apoptosis in other neuronal systems. Spiral ganglion (SG) explants from 5-day-old Wistar rats were treated with different concentrations of the general MMP inhibitor GM6001, a specific MMP-2 inhibitor, and a specific MMP-9 inhibitor, in vitro. The general inhibitor of MMPs and the specific inhibition of MMP-2 significantly reduced both the number of neurites that extended from SG explants, as well as the length of individual neurites. However, neither the general inhibitor of MMPs nor the specific inhibition of MMP-2 influenced SGN survival. Inhibition of MMP-9 had no influence on SGNs. The data suggest that MMPs, and more specifically MMP-2, influence the growth of developing afferent neurites in the mammalian inner ear in vivo.
Collapse
Affiliation(s)
- Michael Sung
- Department of Biomedicine, University Hospital Basel, Hebelstrasse 20, 4031, Basel, Switzerland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
Modulation of coagulation has been successfully applied to ischemic disorders of the central nervous system (CNS). Some components of the coagulation system have been identified in the CNS, yet with limited exception their functions have not been clearly defined. Little is known about how events within the cerebral tissues affect hemostasis. Nonetheless, the interaction between cerebral cells and vascular hemostasis and the possibility that endogenous coagulation factors can participate in functions within the neurovascular unit provide intriguing possibilities for deeper insight into CNS functions and the potential for treatment of CNS injuries. Here, we consider the expression of coagulation factors in the CNS, the coagulopathy associated with focal cerebral ischemia (and its relationship to hemorrhagic transformation), the use of recombinant tissue plasminogen activator (rt-PA) in ischemic stroke and its study in animal models, the impact of rt-PA on neuron and CNS structure and function, and matrix protease generation and matrix degradation and hemostasis. Interwoven among these topics is evidence for interactions of coagulation factors with and within the CNS. How activation of hemostasis occurs in the cerebral tissues and how the brain responds are difficult questions that offer many research possibilities.
Collapse
Affiliation(s)
- Gregory J. del Zoppo
- Division of Hematology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington
- Department of Neurology, University of Washington School of Medicine, Seattle, Washington
| | - Yoshikane Izawa
- Division of Hematology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington
| | - Brian T. Hawkins
- Division of Hematology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington
| |
Collapse
|
42
|
Dasuri K, Zhang L, Keller JN. Oxidative stress, neurodegeneration, and the balance of protein degradation and protein synthesis. Free Radic Biol Med 2013; 62:170-185. [PMID: 23000246 DOI: 10.1016/j.freeradbiomed.2012.09.016] [Citation(s) in RCA: 262] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 09/05/2012] [Accepted: 09/11/2012] [Indexed: 12/12/2022]
Abstract
Oxidative stress occurs in a variety of disease settings and is strongly linked to the development of neuron death and neuronal dysfunction. Cells are equipped with numerous pathways to prevent the genesis, as well as the consequences, of oxidative stress in the brain. In this review we discuss the various forms and sources of oxidative stress in the brain and briefly discuss some of the complexities in detecting the presence of oxidative stress. We then focus the review on the interplay between the diverse cellular proteolytic pathways and their roles in regulating oxidative stress in the brain. Additionally, we discuss the involvement of protein synthesis in regulating the downstream effects of oxidative stress. Together, these components of the review demonstrate that the removal of damaged proteins by effective proteolysis and the synthesis of new and protective proteins are vital in the preservation of brain homeostasis during periods of increased levels of reactive oxygen species. Last, studies from our laboratory and others have demonstrated that protein synthesis is intricately linked to the rates of protein degradation, with impairment of protein degradation sufficient to decrease the rates of protein synthesis, which has important implications for successfully responding to periods of oxidative stress. Specific neurodegenerative diseases, including Alzheimer disease, Parkinson disease, amyotrophic lateral sclerosis, and stroke, are discussed in this context. Taken together, these findings add to our understanding of how oxidative stress is effectively managed in the healthy brain and help elucidate how impairments in proteolysis and/or protein synthesis contribute to the development of neurodegeneration and neuronal dysfunction in a variety of clinical settings.
Collapse
Affiliation(s)
- Kalavathi Dasuri
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | - Le Zhang
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | - Jeffrey N Keller
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA.
| |
Collapse
|
43
|
Omouendze PL, Henry VJ, Porte B, Dupré N, Carmeliet P, Gonzalez BJ, Marret S, Leroux P. Hypoxia-ischemia or excitotoxin-induced tissue plasminogen activator- dependent gelatinase activation in mice neonate brain microvessels. PLoS One 2013; 8:e71263. [PMID: 23940734 PMCID: PMC3735506 DOI: 10.1371/journal.pone.0071263] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Accepted: 06/26/2013] [Indexed: 12/02/2022] Open
Abstract
Hypoxia-ischemia (HI) and excitotoxicity are validated causes of neonatal brain injuries and tissue plasminogen activator (t-PA) participates in the processes through proteolytic and receptor-mediated pathways. Brain microvascular endothelial cells from neonates in culture, contain and release more t-PA and gelatinases upon glutamate challenge than adult cells. We have studied t-PA to gelatinase (MMP-2 and MMP-9) activity links in HI and excitotoxicity lesion models in 5 day–old pups in wild type and in t-PA or its inhibitor (PAI-1) genes inactivated mice. Gelatinolytic activities were detected in SDS-PAGE zymograms and by in situ fluorescent DQ-gelatin microscopic zymographies. HI was achieved by unilateral carotid ligature followed by a 40 min hypoxia (8%O2). Excitotoxic lesions were produced by intra parenchymal cortical (i.c.) injections of 10 µg ibotenate (Ibo). Gel zymograms in WT cortex revealed progressive extinction of MMP-2 and MMP-9 activities near day 15 or day 8 respectively. MMP-2 expression was the same in all strains while MMP-9 activity was barely detectable in t-PA−/− and enhanced in PAI-1−/− mice. HI or Ibo produced activation of MMP-2 activities 6 hours post-insult, in cortices of WT mice but not in t-PA−/− mice. In PAI-1−/− mice, HI or vehicle i.c. injection increased MMP-2 and MMP-9 activities. In situ zymograms using DQ-gelatin revealed vessel associated gelatinolytic activity in lesioned areas in PAI-1−/− and in WT mice. In WT brain slices incubated ex vivo, glutamate (200 µM) induced DQ-gelatin activation in vessels. The effect was not detected in t-PA−/−mice, but was restored by concomitant exposure to recombinant t-PA (20 µg/mL). In summary, neonatal brain lesion paradigms and ex vivo excitotoxic glutamate evoked t-PA-dependent gelatinases activation in vessels. Both MMP-2 and MMP-9 activities appeared t-PA-dependent. The data suggest that vascular directed protease inhibition may have neuroprotection potential against neonatal brain injuries.
Collapse
Affiliation(s)
- Priscilla L. Omouendze
- Haute-Normandie-INSERM ERI-28, Institute for Research and Innovation in Biomedicine of Rouen University, Rouen, France
| | - Vincent J. Henry
- Haute-Normandie-INSERM ERI-28, Institute for Research and Innovation in Biomedicine of Rouen University, Rouen, France
| | - Baptiste Porte
- Haute-Normandie-INSERM ERI-28, Institute for Research and Innovation in Biomedicine of Rouen University, Rouen, France
| | - Nicolas Dupré
- Haute-Normandie-INSERM ERI-28, Institute for Research and Innovation in Biomedicine of Rouen University, Rouen, France
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Leuven, Belgium
| | - Bruno J. Gonzalez
- Haute-Normandie-INSERM ERI-28, Institute for Research and Innovation in Biomedicine of Rouen University, Rouen, France
| | - Stéphane Marret
- Haute-Normandie-INSERM ERI-28, Institute for Research and Innovation in Biomedicine of Rouen University, Rouen, France
- Department of Neonatal Paediatrics and Intensive Care, Rouen University Hospital, Rouen, France
| | - Philippe Leroux
- Haute-Normandie-INSERM ERI-28, Institute for Research and Innovation in Biomedicine of Rouen University, Rouen, France
- * E-mail:
| |
Collapse
|
44
|
Cayabyab FS, Gowribai K, Walz W. Involvement of matrix metalloproteinases-2 and -9 in the formation of a lacuna-like cerebral cavity. J Neurosci Res 2013; 91:920-33. [PMID: 23606560 DOI: 10.1002/jnr.23223] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 02/22/2013] [Accepted: 03/01/2013] [Indexed: 12/20/2022]
Abstract
We used a modified pial vessel disruption (PVD) protocol with adult male Wistar rats to mimic small-vessel stroke in the cerebral cortex. Within 3 weeks, this lesion develops into a single lacuna-like cavity, which is fluid-filled and encapsulated by reactive astrocytes. Minocycline treatment that commences 1 hr after lesion and continues for 6 days prevents the cavitation and causes a filling of the lesion with reactive astrocytes and no barrier. Here, we determined whether inhibition of matrix metalloproteinases-2 and -9 (MMPs) mediates this minocycline action. Confocal microscopy revealed increased punctate staining of MMPs inside the lesion sites after 2 days of PVD. Astrocytes lined the lesion border but showed sparse localization inside the lesion. In contrast, increased MMP levels inside the lesion coincided with increased ED1 or OX-42 immunostaining, suggesting that MMP elevation reflected increased secretions from microglia/macrophages. Imaging analyses also revealed that minocycline administered for 2 days before animal euthanasia, significantly decreased MMP levels within the lesion. Moreover, Western blot analysis of cortical tissue extracts showed a significant 30-40% upregulation of MMPs 2 days after lesion. Minocycline administered 2 hr before the lesion significantly inhibited both MMP-9 and MMP-2 levels by ∼40%. In contrast, minocycline administered 1 hr after the lesion only decreased MMP-9 levels by ∼30%. Because MMP inhibition with batimastat injection also prevented cavity formation at 21 days, we conclude that minocycline prevented the creation of a lacuna-like cyst in the cerebral cortex by inhibiting the MMP secretion from microglia in the affected tissue.
Collapse
Affiliation(s)
- Francisco S Cayabyab
- Department of Physiology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.
| | | | | |
Collapse
|
45
|
Abstract
The brain is in many ways an immunologically and pharmacologically privileged site. The blood-brain barrier (BBB) of the cerebrovascular endothelium and its participation in the complex structure of the neurovascular unit (NVU) restrict access of immune cells and immune mediators to the central nervous system (CNS). In pathologic conditions, very well-organized immunologic responses can develop within the CNS, raising important questions about the real nature and the intrinsic and extrinsic regulation of this immune privilege. We assess the interactions of immune cells and immune mediators with the BBB and NVU in neurologic disease, cerebrovascular disease, and intracerebral tumors. The goals of this review are to outline key scientific advances and the status of the science central to both the neuroinflammation and CNS barriers fields, and highlight the opportunities and priorities in advancing brain barriers research in the context of the larger immunology and neuroscience disciplines. This review article was developed from reports presented at the 2011 Annual Blood-Brain Barrier Consortium Meeting.
Collapse
|
46
|
Chen XM, Chen HS, Xu MJ, Shen JG. Targeting reactive nitrogen species: a promising therapeutic strategy for cerebral ischemia-reperfusion injury. Acta Pharmacol Sin 2013; 34:67-77. [PMID: 22842734 DOI: 10.1038/aps.2012.82] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Ischemic stroke accounts for nearly 80% of stroke cases. Recanalization with thrombolysis is a currently crucial therapeutic strategy for re-building blood supply, but the thrombolytic therapy often companies with cerebral ischemia-reperfusion injury, which are mediated by free radicals. As an important component of free radicals, reactive nitrogen species (RNS), including nitric oxide (NO) and peroxynitrite (ONOO(-)), play important roles in the process of cerebral ischemia-reperfusion injury. Ischemia-reperfusion results in the production of nitric oxide (NO) and peroxynitrite (ONOO(-)) in ischemic brain, which trigger numerous molecular cascades and lead to disruption of the blood brain barrier and exacerbate brain damage. There are few therapeutic strategies available for saving ischemic brains and preventing the subsequent brain damage. Recent evidence suggests that RNS could be a therapeutic target for the treatment of cerebral ischemia-reperfusion injury. Herein, we reviewed the recent progress regarding the roles of RNS in the process of cerebral ischemic-reperfusion injury and discussed the potentials of drug development that target NO and ONOO(-) to treat ischemic stroke. We conclude that modulation for RNS level could be an important therapeutic strategy for preventing cerebral ischemia-reperfusion injury.
Collapse
|
47
|
Abstract
The Nobel laureate Max Delbrück often said that it is the crossover between disciplines where advances are possible in science. This certainly has been true for our understanding of the vascular biology of the central nervous system in the setting of ischemic stroke. The ability to cross the boundaries of hemostasis, neurology, hematology, and neuroscience has facilitated our research direction to define the relation of the microvasculature to neuron function. Work begun with the clinical scientific exploration of the contributions of arterial thrombosis to the acute injury processes initiated by focal cerebral ischemia has led to an increased understanding of the effects of ischemia on microvessel integrity.
Collapse
Affiliation(s)
- Gregory J. del Zoppo
- From the Department of Medicine (Division of Hematology) and the Department of Neurology, University of Washington, Seattle, WA
| |
Collapse
|
48
|
Abstract
With the demonstration that acute recanalization of obstructed symptomatic cerebral arteries during ischemic stroke can result in substantial improvement in clinical outcome, the variability in clinical responses, and in hemorrhagic transformation, requires attention. This short review addresses the effect of aging and amyloid deposition disease on microvessel integrity, interactions within the neurovascular unit, cerebral tissue susceptibility to ischemic injury, and postischemic inflammation, and ultimately on the outcomes and safety of acute recanalization during ischemic stroke. Microvessels and neighboring neurons respond simultaneously to focal ischemia. The cellular components and matrix barriers of the neurovascular unit all respond to ischemia; however, their coordinate interactions are not understood. Furthermore, there is little known about the cell-cell and cell-matrix interactions within the unit, or about the effect of β-amyloid on microvessel responses during ischemia. These considerations indicate the need for a coordinated research effort to understand the origins of the variability in recanalization outcome.
Collapse
Affiliation(s)
- Gregory J del Zoppo
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA.
| |
Collapse
|
49
|
Baeten KM, Akassoglou K. Extracellular matrix and matrix receptors in blood-brain barrier formation and stroke. Dev Neurobiol 2012; 71:1018-39. [PMID: 21780303 DOI: 10.1002/dneu.20954] [Citation(s) in RCA: 292] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The blood-brain barrier (BBB) is formed primarily to protect the brain microenvironment from the influx of plasma components, which may disturb neuronal functions. The BBB is a functional unit that consists mainly of specialized endothelial cells (ECs) lining the cerebral blood vessels, astrocytes, and pericytes. The BBB is a dynamic structure that is altered in neurologic diseases, such as stroke. ECs and astrocytes secrete extracellular matrix (ECM) proteins to generate and maintain the basement membranes (BMs). ECM receptors, such as integrins and dystroglycan, are also expressed at the brain microvasculature and mediate the connections between cellular and matrix components in physiology and disease. ECM proteins and receptors elicit diverse molecular signals that allow cell adaptation to environmental changes and regulate growth and cell motility. The composition of the ECM is altered upon BBB disruption and directly affects the progression of neurologic disease. The purpose of this review is to discuss the dynamic changes of ECM composition and integrin receptor expression that control BBB functions in physiology and pathology.
Collapse
Affiliation(s)
- Kim M Baeten
- Gladstone Institute of Neurological Disease, University of California, San Francisco, California 94158, USA
| | | |
Collapse
|
50
|
Lee JY, Kim HS, Choi HY, Oh TH, Yune TY. Fluoxetine inhibits matrix metalloprotease activation and prevents disruption of blood-spinal cord barrier after spinal cord injury. ACTA ACUST UNITED AC 2012; 135:2375-89. [PMID: 22798270 DOI: 10.1093/brain/aws171] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
After spinal cord injury, the disruption of blood-spinal cord barrier by activation of matrix metalloprotease is a critical event leading to infiltration of blood cells, inflammatory responses and neuronal cell death, contributing to permanent neurological disability. Recent evidence indicates that fluoxetine, an anti-depressant drug, is shown to have neuroprotective effects in ischaemic brain injury, but the precise mechanism underlying its protective effects is largely unknown. Here, we show that fluoxetine prevented blood-spinal cord barrier disruption via inhibition of matrix metalloprotease activation after spinal cord injury. After a moderate contusion injury at the T9 level of spinal cord with an infinite horizon impactor in the mouse, fluoxetine (10 mg/kg) was injected intraperitoneally and further administered once a day for indicated time points. Fluoxetine treatment significantly inhibited messenger RNA expression of matrix metalloprotease 2, 9 and 12 after spinal cord injury. By zymography and fluorimetric enzyme activity assay, fluoxetine also significantly reduced matrix metalloprotease 2 and matrix metalloprotease 9 activities after injury. In addition, fluoxetine inhibited nuclear factor kappa B-dependent matrix metalloprotease 9 expression in bEnd.3, a brain endothelial cell line, after oxygen-glucose deprivation/reoxygenation. Fluoxetine also attenuated the loss of tight junction molecules such as zona occludens 1 and occludin after injury in vivo as well as in bEnd.3 cultures. By immunofluorescence staining, fluoxetine prevented the breakdown of the tight junction integrity in endothelial cells of blood vessel after injury. Furthermore, fluoxetine inhibited the messenger RNA expression of chemokines such as Groα, MIP1α and 1β, and prevented the infiltration of neutrophils and macrophages, and reduced the expression of inflammatory mediators after injury. Finally, fluoxetine attenuated apoptotic cell death and improved locomotor function after injury. Thus, our results indicate that fluoxetine improved functional recovery in part by inhibiting matrix metalloprotease activation and preventing blood-spinal cord barrier disruption after spinal cord injury. Furthermore, our study suggests that fluoxetine may represent a potential therapeutic agent for preserving blood-brain barrier integrity following ischaemic brain injury and spinal cord injury in humans.
Collapse
Affiliation(s)
- Jee Y Lee
- Age-Related and Brain Diseases Research Centre, School of Medicine, Kyung Hee University, Seoul 130-701, Korea
| | | | | | | | | |
Collapse
|