1
|
Kawa H, Ahmed Z, Majid A, Chen R. Inhibition of matrix metalloproteinases to reduce blood brain barrier disruption and haemorrhagic transformation in ischaemic stroke: Go broad or go narrow? Neuropharmacology 2025; 262:110192. [PMID: 39419277 DOI: 10.1016/j.neuropharm.2024.110192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/19/2024] [Accepted: 10/14/2024] [Indexed: 10/19/2024]
Abstract
Ischaemic stroke characterises impulsive cerebral-region hypoxia due to deep intracerebral arteriole blockage, often accompanied by permanent cerebral infarction and cognitive impairment. Thrombolysis with recombinant tissue plasminogen activator (rtPA) and thrombectomy remain the only guidance-approved therapies. However, emerging data draws clear links between such therapies and haemorrhage transformation, which occur when cerebral vasculature is damaged during ischaemia/reperfusion. Studies have shown that matrix metalloproteinases (MMPs) play a significant role in haemorrhage transformation, by depleting the extracellular matrix (ECM) and disrupting the blood brain barrier (BBB). Inhibitors of MMPs may be used to prevent ischaemic stroke patients from BBB disruption and haemorrhage transformation, particularly for those receiving rtPA treatment. Preclinical studies found that inhibition of MMPs with agents or in knock out mice, effectively reduced BBB disruption and infarct volume, leading to improved ischaemic stroke outcomes. At present, MMP inhibition is not an approved therapy for stroke patients. There remain concerns about timing, dosing, duration of MMP inhibition and selection of either broad spectrum or specific MMP inhibitors for stroke patients. This review aims to summarize current knowledge on MMP inhibition in ischaemic stroke and explore whether a broad spectrum or a specific MMP inhibitor should be used for ischaemic stroke patient treatment. It is crucial to inhibit MMP activities early and sufficiently to ensure BBB intact during ischaemia and reperfusion, but also to reduce side effects of MMP inhibitors to minimum. Recent advance in stroke therapy by thrombectomy could aid in such treatment with intra-arterially delivery of MMP inhibitors (and/or antioxidants).
Collapse
Affiliation(s)
- Hala Kawa
- School of Pharmacy and Bioengineering, Keele University, Staffordshire, ST5 5BG, UK
| | - Zubair Ahmed
- Institute of Inflammation and Ageing, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Arshad Majid
- Division of Neurosciences, School of Medicine and Population Health, University of Sheffield, Sheffield, S10 2HQ, UK
| | - Ruoli Chen
- School of Pharmacy and Bioengineering, Keele University, Staffordshire, ST5 5BG, UK.
| |
Collapse
|
2
|
Fang YT, Liao SF, Chen PL, Yeh TS, Chen CI, Piravej K, Wu CC, Chiu WT, Lam C. Risk of Traumatic Intracranial Hemorrhage After Stroke: A Nationwide Population-Based Cohort Study in Taiwan. J Am Heart Assoc 2024; 13:e035725. [PMID: 39291491 DOI: 10.1161/jaha.124.035725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 08/22/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND Stroke and traumatic intracranial hemorrhage (tICH) are major causes of disability worldwide, with stroke exerting significant negative effects on the brain, potentially elevating tICH risk. In this study, we investigated tICH risk in stroke survivors. METHODS AND RESULTS Using relevant data (2017-2019) from Taiwan's National Health Insurance Research Database, we conducted a population-based retrospective cohort study. Patients were categorized into stroke and nonstroke groups, and tICH risk was compared using a Cox proportional-hazards model. Among 164 628 patients with stroke, 1004 experienced tICH. Patients with stroke had a higher tICH risk than nonstroke counterparts (adjusted hazard ratio [HR], 3.49 [95% CI, 3.17-3.84]). Subgroup analysis by stroke type revealed higher tICH risk in hemorrhagic stroke survivors compared with ischemic stroke survivors (HR, 5.64 [95% CI, 4.97-6.39] versus 2.87 [95% CI, 2.58-3.18], respectively). Older patients (≥45 years) with stroke had a higher tICH risk compared with their younger counterparts (<45 years), in contrast to younger patients without stroke (HR, 7.89 [95% CI, 6.41-9.70] versus 4.44 [95% CI, 2.99-6.59], respectively). Dementia and Parkinson disease emerged as significant tICH risk factors (HR, 1.69 [95% CI, 1.44-2.00] versus 2.17 [95% CI, 1.71-2.75], respectively). In the stroke group, the highest tICH incidence density occurred 3 months after stroke, particularly in patients aged >65 years. CONCLUSIONS Stroke survivors, particularly those with hemorrhagic stroke and those aged ≥45 years, face elevated tICH risk. Interventions targeting the high-risk period are vital, with fall injuries potentially contributing to tICH incidence.
Collapse
Affiliation(s)
- Yun-Ting Fang
- School of Medicine, College of Medicine Taipei Medical University Taipei Taiwan
- Taipei Veterans General Hospital Taipei Taiwan
| | - Shu-Fen Liao
- Department of Medical Research, Wan Fang Hospital Taipei Medical University Taipei Taiwan
- School of Public Health, College of Public Health Taipei Medical University Taipei Taiwan
| | - Ping-Ling Chen
- Graduate Institute of Injury Prevention and Control, College of Public Health Taipei Medical University Taipei Taiwan
| | - Tian-Shin Yeh
- Department of Physical Medicine and Rehabilitation, School of Medicine, College of Medicine Taipei Medical University Taipei Taiwan
- Department of Physical Medicine and Rehabilitation, Wan Fang Hospital Taipei Medical University Taipei Taiwan
- Department of Epidemiology and Nutrition, Harvard T. H. Chan School of Public Health Harvard University Boston MA
- Department of Physical Medicine and Rehabilitation National Taiwan University Hospital Taipei Taiwan
- Department of Physical Medicine and Rehabilitation, College of Medicine National Taiwan University Taipei Taiwan
| | - Chin-I Chen
- Department of Neurology, Wan Fang Hospital Taipei Medical University Taipei Taiwan
- Department of Neurology, School of Medicine, College of Medicine Taipei Medical University Taipei Taiwan
| | - Krisna Piravej
- Department of Rehabilitation Medicine, Faculty of Medicine Chulalongkorn University Bangkok Thailand
- Department of Chula Neuroscience Center King Chulalongkorn Memorial Hospital Bangkok Thailand
| | - Chia-Chieh Wu
- Emergency Department, Wan Fang Hospital Taipei Medical University Taipei Taiwan
| | - Wen-Ta Chiu
- Graduate Institute of Injury Prevention and Control, College of Public Health Taipei Medical University Taipei Taiwan
- Department of Neurosurgery, Shuang Ho Hospital Taipei Medical University New Taipei Taiwan
- AHMC Health System Alhambra CA
| | - Carlos Lam
- Emergency Department, Wan Fang Hospital Taipei Medical University Taipei Taiwan
- Department of Emergency, School of Medicine, College of Medicine Taipei Medical University Taipei Taiwan
| |
Collapse
|
3
|
Huang X, Lan Z, Hu Z. Role and mechanisms of mast cells in brain disorders. Front Immunol 2024; 15:1445867. [PMID: 39253085 PMCID: PMC11381262 DOI: 10.3389/fimmu.2024.1445867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 08/12/2024] [Indexed: 09/11/2024] Open
Abstract
Mast cells serve as crucial effector cells within the innate immune system and are predominantly localized in the skin, airways, gastrointestinal tract, urinary and reproductive tracts, as well as in the brain. Under physiological conditions, brain-resident mast cells secrete a diverse array of neuro-regulatory mediators to actively participate in neuroprotection. Meanwhile, as the primary source of molecules causing brain inflammation, mast cells also function as the "first responders" in brain injury. They interact with neuroglial cells and neurons to facilitate the release of numerous inflammatory mediators, proteases, and reactive oxygen species. This process initiates and amplifies immune-inflammatory responses in the brain, thereby contributing to the regulation of neuroinflammation and blood-brain barrier permeability. This article provides a comprehensive overview of the potential mechanisms through which mast cells in the brain may modulate neuroprotection and their pathological implications in various neurological disorders. It is our contention that the inhibition of mast cell activation in brain disorders could represent a novel avenue for therapeutic breakthroughs.
Collapse
Affiliation(s)
- Xuanyu Huang
- Department of Neurology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Ziwei Lan
- Department of Neurology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Zhiping Hu
- Department of Neurology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
4
|
Veeravalli KK. Implications of MMP-12 in the pathophysiology of ischaemic stroke. Stroke Vasc Neurol 2024; 9:97-107. [PMID: 37336584 PMCID: PMC11103161 DOI: 10.1136/svn-2023-002363] [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: 02/03/2023] [Accepted: 06/05/2023] [Indexed: 06/21/2023] Open
Abstract
This article focuses on the emerging role of matrix metalloproteinase-12 (MMP-12) in ischaemic stroke (IS). MMP-12 expression in the brain increases dramatically in animal models of IS, and its suppression reduces brain damage and promotes neurological, sensorimotor and cognitive functional outcomes. Thus, MMP-12 could represent a potential target for the management of IS. This article provides an overview of MMP-12 upregulation in the brain following IS, its deleterious role in the post-stroke pathogenesis (blood-brain barrier disruption, inflammation, apoptosis and demyelination), possible molecular interactions and mechanistic insights, its involvement in post-ischaemic functional deficits and recovery as well as the limitations, perspectives, challenges and future directions for further research. Prior to testing any MMP-12-targeted therapy in patients with acute IS, additional research is needed to establish the effectiveness of MMP-12 suppression against IS in older animals and in animals with comorbidities. This article also examines the clinical implications of suppressing MMP-12 alone or in combination with MMP-9 for extending the currently limited tissue plasminogen activator therapy time window. Targeting of MMP-12 is expected to have a profound influence on the therapeutic management of IS in the future.
Collapse
Affiliation(s)
- Krishna Kumar Veeravalli
- Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois, USA
| |
Collapse
|
5
|
Li Y, Chen J, Quan X, Chen Y, Han Y, Chen J, Yang L, Xu Y, Shen X, Wang R, Zhao Y. Extracellular Vesicles Maintain Blood-Brain Barrier Integrity by the Suppression of Caveolin-1/CD147/VEGFR2/MMP Pathway After Ischemic Stroke. Int J Nanomedicine 2024; 19:1451-1467. [PMID: 38371456 PMCID: PMC10874237 DOI: 10.2147/ijn.s444009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 02/08/2024] [Indexed: 02/20/2024] Open
Abstract
Background Ischemic stroke (IS) causes tragic death and disability worldwide. However, effective therapeutic interventions are finite. After IS, blood-brain barrier (BBB) integrity is disrupted, resulting in deteriorating neurological function. As a novel therapeutic, extracellular vesicles (EVs) have shown ideal restorative effects on BBB integrity post-stroke; however, the definite mechanisms remain ambiguous. In the present study, we investigated the curative effects and the mechanisms of EVs derived from bone marrow mesenchymal stem cells and brain endothelial cells (BMSC-EVs and BEC-EVs) on BBB integrity after acute IS. Methods EVs were isolated from BMSCs and BECs, and we investigated the therapeutic effect in vitro oxygen-glucose deprivation (OGD) insulted BECs model and in vivo rat middle cerebral artery occlusion (MCAo) model. The cell monolayer leakage, tight junction expression, and metalloproteinase (MMP) activity were evaluated, and rat brain infarct volume and neurological function were also analyzed. Results The administration of two kinds of EVs not only enhanced ZO-1 and Occludin expressions but also reduced the permeability and the activity of MMP-2/9 in OGD-insulted BECs. The amelioration of the cerebral infarction, BBB leakage, neurological function deficits, and the increasing ZO-1 and Occludin levels, as well as MMP activity inhibition was observed in MCAo rats. Additionally, the increased levels of Caveolin-1, CD147, vascular endothelial growth factor receptor 2 (VEGFR2), and vascular endothelial growth factor A (VEGFA) in isolated brain microvessels were downregulated after EVs treatment. In vitro, the employment of Caveolin-1 and CD147 siRNA partly suppressed the expressions of VEGFR2, VEGFA and MMP-2/9 activity and reduced the leakage of OGD insulted BECs and enhanced ZO-1 and Occludin expressions. Conclusion Our study firstly demonstrates that BEC and BMSC-EVs administrations maintain BBB integrity via the suppression of Caveolin-1/CD147/VEGFR2/MMP pathway after IS, and the efficacy of BMSC-EVs is superior to that of BEC-EVs.
Collapse
Affiliation(s)
- Yiyang Li
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR, People’s Republic of China
| | - Jiali Chen
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR, People’s Republic of China
| | - Xingping Quan
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR, People’s Republic of China
| | - Ying Chen
- School of Health Economics and Management, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
| | - Yan Han
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR, People’s Republic of China
| | - Jinfen Chen
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR, People’s Republic of China
| | - Li Yang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR, People’s Republic of China
| | - Youhua Xu
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR, People’s Republic of China
| | - Xu Shen
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Ruibing Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR, People’s Republic of China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, People’s Republic of China
| | - Yonghua Zhao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR, People’s Republic of China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, People’s Republic of China
| |
Collapse
|
6
|
Chai YL, Rajeev V, Poh L, Selvaraji S, Hilal S, Chen CP, Jo DG, Koo EH, Arumugam TV, Lai MKP. Chronic cerebral hypoperfusion alters the CypA-EMMPRIN-gelatinase pathway: Implications for vascular dementia. J Cereb Blood Flow Metab 2023; 43:722-735. [PMID: 36537035 PMCID: PMC10108186 DOI: 10.1177/0271678x221146401] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 11/17/2022] [Accepted: 11/17/2022] [Indexed: 03/21/2023]
Abstract
Chronic cerebral hypoperfusion (CCH) is postulated to underlie multiple pathophysiological processes in vascular dementia (VaD), including extracellular matrix dysfunction. While several extracellular matrix proteins, namely cyclophilin A (CypA), extracellular matrix metalloproteinase inducer (EMMPRIN) and gelatinases (matrix metalloproteinases, MMP-2 and -9) have been investigated in acute stroke, their involvement in CCH and VaD remains unclear. In this study, CypA-EMMPRIN-gelatinase proteins were analysed in a clinical cohort of 36 aged, cognitively unimpaired subjects and 48 VaD patients, as well as in a bilateral carotid artery stenosis mouse model of CCH. Lower CypA and higher EMMPRIN levels were found in both VaD serum and CCH mouse brain. Furthermore, gelatinases were differentially altered in CCH mice and VaD patients, with significant MMP-2 increase in CCH brain and serum, whilst serum MMP-9 was elevated in VaD but reduced in CCH, suggesting complex CypA-EMMPRIN-gelatinase regulatory mechanisms. Interestingly, subjects with cortical infarcts had higher serum MMP-2, while white matter hyperintensities, cortical infarcts and lacunes were associated with higher serum MMP-9. Taken together, our data indicate that perturbations of CypA-EMMPRIN signalling may be associated with gelatinase-mediated vascular sequelae, highlighting the potential utility of the CypA-EMMPRIN-gelatinase pathway as clinical biomarkers and therapeutic targets in VaD.
Collapse
Affiliation(s)
- Yuek Ling Chai
- Department of Pharmacology, Yong
Loo Lin School of Medicine, National University of Singapore, Kent Ridge,
Singapore
- Memory, Aging and Cognition Centre,
National University Health System, Kent Ridge, Singapore
| | - Vismitha Rajeev
- Department of Pharmacology, Yong
Loo Lin School of Medicine, National University of Singapore, Kent Ridge,
Singapore
| | - Luting Poh
- Department of Pharmacology, Yong
Loo Lin School of Medicine, National University of Singapore, Kent Ridge,
Singapore
| | - Sharmelee Selvaraji
- Department of Pharmacology, Yong
Loo Lin School of Medicine, National University of Singapore, Kent Ridge,
Singapore
| | - Saima Hilal
- Department of Pharmacology, Yong
Loo Lin School of Medicine, National University of Singapore, Kent Ridge,
Singapore
- Saw Swee Hock School of Public
Health, National University of Singapore, Kent Ridge, Singapore
| | - Christopher P Chen
- Department of Pharmacology, Yong
Loo Lin School of Medicine, National University of Singapore, Kent Ridge,
Singapore
- Memory, Aging and Cognition Centre,
National University Health System, Kent Ridge, Singapore
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan
University, Suwon, Republic of Korea
| | - Edward H Koo
- Department of Medicine, National
University of Singapore, Kent Ridge, Singapore
- Graduate School for Integrative
Sciences and Engineering, National University of Singapore, Kent Ridge,
Singapore
- Department of Neurosciences,
University of California San Diego, San Diego, CA, USA
| | - Thiruma V Arumugam
- School of Pharmacy, Sungkyunkwan
University, Suwon, Republic of Korea
- Centre for Cardiovascular Biology
and Disease Research, Department of Microbiology, Anatomy, Physiology and
Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe
University, Bundoora, VIC, Australia
| | - Mitchell KP Lai
- Department of Pharmacology, Yong
Loo Lin School of Medicine, National University of Singapore, Kent Ridge,
Singapore
- Memory, Aging and Cognition Centre,
National University Health System, Kent Ridge, Singapore
| |
Collapse
|
7
|
Pluta R, Kiś J, Januszewski S, Jabłoński M, Czuczwar SJ. Cross-Talk between Amyloid, Tau Protein and Free Radicals in Post-Ischemic Brain Neurodegeneration in the Form of Alzheimer’s Disease Proteinopathy. Antioxidants (Basel) 2022; 11:antiox11010146. [PMID: 35052650 PMCID: PMC8772936 DOI: 10.3390/antiox11010146] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 01/05/2022] [Accepted: 01/09/2022] [Indexed: 02/04/2023] Open
Abstract
Recent years have seen remarkable progress in research into free radicals oxidative stress, particularly in the context of post-ischemic recirculation brain injury. Oxidative stress in post-ischemic tissues violates the integrity of the genome, causing DNA damage, death of neuronal, glial and vascular cells, and impaired neurological outcome after brain ischemia. Indeed, it is now known that DNA damage and repair play a key role in post-stroke white and gray matter remodeling, and restoring the integrity of the blood-brain barrier. This review will present one of the newly characterized mechanisms that emerged with genomic and proteomic development that led to brain ischemia to a new level of post-ischemic neuropathological mechanisms, such as the presence of amyloid plaques and the development of neurofibrillary tangles, which further exacerbate oxidative stress. Finally, we hypothesize that modified amyloid and the tau protein, along with the oxidative stress generated, are new key elements in the vicious circle important in the development of post-ischemic neurodegeneration in a type of Alzheimer’s disease proteinopathy.
Collapse
Affiliation(s)
- Ryszard Pluta
- Laboratory of Ischemic and Neurodegenerative Brain Research, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5 Str., 02-106 Warsaw, Poland;
- Correspondence: ; Tel.: +48-22-608-6540
| | - Jacek Kiś
- Department of Urology, 1st Military Clinical Hospital with the Outpatient Clinic, Al. Racławickie 23, 20-049 Lublin, Poland;
| | - Sławomir Januszewski
- Laboratory of Ischemic and Neurodegenerative Brain Research, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5 Str., 02-106 Warsaw, Poland;
| | - Mirosław Jabłoński
- Department of Rehabilitation and Orthopedics, Medical University of Lublin, Jaczewskiego 8 Str., 20-090 Lublin, Poland;
| | - Stanisław J. Czuczwar
- Department of Pathophysiology, Medical University of Lublin, Jaczewskiego 8b Str., 20-090 Lublin, Poland;
| |
Collapse
|
8
|
Liew SL, Lin DJ, Cramer SC. Interventions to Improve Recovery After Stroke. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00061-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
9
|
Xia M, Su Y, Fu J, Xu J, Wang Q, Gao F, Shen Y, Dong Q, Cheng X. The Use of Serum Matrix Metalloproteinases in Cerebral Amyloid Angiopathy-Related Intracerebral Hemorrhage and Cognitive Impairment. J Alzheimers Dis 2021; 82:1159-1170. [PMID: 34151802 DOI: 10.3233/jad-210288] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Neuroimaging has played a primary role in predicting intracerebral hemorrhage (ICH) recurrence of cerebral amyloid angiopathy (CAA); however, the utilities of biomarkers in CAA-related ICH and cognitive impairment remain unexplored. OBJECTIVE To investigate the correlations of serum levels of matrix metalloproteinase-2 (MMP-2), MMP-3, and MMP-9 with CAA-related MRI markers, ICH recurrence, and cognitive status. METHODS 68 cases with first probable CAA-ICH and 69 controls were recruited. Clinical and imaging data were obtained at baseline and serum MMPs in the acute phase were measured by Luminex multiplex assays. Cognitive status was assessed with the Chinese version of Mini-Mental State Examination within 10-14 days after ICH onset. RESULTS Serum MMP-2 level was significantly lower in CAA-ICH patients than controls while MMP-9 was significantly higher. In CAA-ICH patients, MMP-3 level was significantly associated with lobar cerebral microbleeds count after adjusting age, sex, and hypertension (adjusted coefficient 0.368, 95%CI 0.099-0.637, p = 0.008). During a median follow-up of 2.4 years, higher level of MMP-2 predicted lower CAA-ICH recurrence after adjusting age (adjusted HR 0.326, 95%CI 0.122-0.871, p = 0.025), ICH volume (adjusted HR 0.259, 95%CI 0.094-0.715, p = 0.009), total MRI burden of SVD score (adjusted HR 0.350, 95%CI 0.131-0.936, p = 0.037) respectively. Besides, higher level of MMP-2 was significantly associated with decreased risk of cognitive impairment independent of age and ICH volume (adjusted OR 0.054, 95%CI 0.005-0.570, p = 0.015). CONCLUSION Serum MMP-2 in acute phase might be a promising biomarker to predict CAA-ICH recurrence and to evaluate the risk of cognitive impairment.
Collapse
Affiliation(s)
- Mingxu Xia
- Department of Neurology, National Centre for Neurological Disorders, National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Ya Su
- Department of Neurology, National Centre for Neurological Disorders, National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiayu Fu
- Department of Neurology, National Centre for Neurological Disorders, National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiajie Xu
- Department of Neurology, National Centre for Neurological Disorders, National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Qiong Wang
- Department of Neurology, First Affiliated Hospital of University of Science and Technology of China, Hefei, China.,Neurodegenerative Disorder Research Centre and Institute on Aging and Brain Disorders, University of Science and Technology of China, Hefei, China
| | - Feng Gao
- Neurodegenerative Disorder Research Centre and Institute on Aging and Brain Disorders, University of Science and Technology of China, Hefei, China
| | - Yong Shen
- Neurodegenerative Disorder Research Centre and Institute on Aging and Brain Disorders, University of Science and Technology of China, Hefei, China.,Centre for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Qiang Dong
- Department of Neurology, National Centre for Neurological Disorders, National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Xin Cheng
- Department of Neurology, National Centre for Neurological Disorders, National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
10
|
Makkiyah F, Sadewo W, Nurrizka R. Comparative Dose of Intracarotid Autologous Bone Marrow Mononuclear Therapy in Chronic Ischemic Stroke in Rats. Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.5675] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Research on chronic ischemic stroke is limited. One of the more promising approaches showing positive effects in the acute stage is mononuclear bone marrow cell therapy. This research may be the first which presents data about the optimum dose of bone marrow mononuclear cells (BM-MNCs) for chronic ischemic stroke in rats and discusses factors influencing recovery in the chronic stage.
We performed temporary middle cerebral artery occlusion (MCAO) procedures on the rats which were then randomly assigned to one of two experimental groups in which they were given either low or high doses of autologous BM-MNCs (5 million or 10 million cells per kg body weight).
Rat brains were fixed for HE, CD31, and doublecortin staining for analysis of the effects. Rat behavior was assessed weekly using the cylinder test and a modified neurological severity score (NSS) test.
In the four weeks prior to administration of BM-MNC, cylinder test scores improved to near normal, and NSS test scores improved moderately. The infarct zone decreased significantly (p <0,01), there was an improvement in angiogenesis (p = 0.1590) and a significant improvement in neurogenesis (p <0,01). Reduction of the infarct zone was associated with a higher dose whereas both higher and lower doses were found to have a similar effect on improving angiogenesis, and neurogenesis. Recovery was superior after twelve weeks compared with the recovery assessment at eight weeks.
In conclusion, a dose of 10 million cells was more effective than a dose of 5 million cells per kg body weight for reducing the infarct zone and ameliorating neurogenesis. There was an improvement of histopathological parameters associated with the longer infarct period.
Collapse
|
11
|
Lin DJ, Cramer SC. Principles of Neural Repair and Their Application to Stroke Recovery Trials. Semin Neurol 2021; 41:157-166. [PMID: 33663003 DOI: 10.1055/s-0041-1725140] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Neural repair is the underlying therapeutic strategy for many treatments currently under investigation to improve recovery after stroke. Repair-based therapies are distinct from acute stroke strategies: instead of salvaging threatened brain tissue, the goal is to improve behavioral outcomes on the basis of experience-dependent brain plasticity. Furthermore, timing, concomitant behavioral experiences, modality specific outcome measures, and careful patient selection are fundamental concepts for stroke recovery trials that can be deduced from principles of neural repair. Here we discuss core principles of neural repair and their implications for stroke recovery trials, highlighting related issues from key studies in humans. Research suggests a future in which neural repair therapies are personalized based on measures of brain structure and function, genetics, and lifestyle factors.
Collapse
Affiliation(s)
- David J Lin
- Center for Neurotechnology and Neurorecovery, Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts.,VA RR&D Center for Neurorestoration and Neurotechnology, Rehabilitation R&D Service, Department of VA Medical Center, Providence, Rhode Island
| | - Steven C Cramer
- Department of Neurology, University of California, Los Angeles, California.,California Rehabilitation Institute, Los Angeles, California
| |
Collapse
|
12
|
Igarashi T, Sastre C, Wolcott Z, Kimberly WT. Continuous Glibenclamide Prevents Hemorrhagic Transformation in a Rodent Model of Severe Ischemia-Reperfusion. J Stroke Cerebrovasc Dis 2021; 30:105595. [PMID: 33450605 PMCID: PMC7894607 DOI: 10.1016/j.jstrokecerebrovasdis.2020.105595] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 12/20/2020] [Accepted: 12/29/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Endovascular thrombectomy (EVT) is highly effective but may also lead to hemorrhagic transformation (HT) and edema, which may be more pronounced in severe ischemia. We sought to determine whether glibenclamide can attenuate HT and edema in a severe ischemia-reperfusion model that reflects EVT. METHODS Using a transient middle cerebral artery occlusion (tMCAo) rodent model of stroke, we studied two rat cohorts, one without rt-PA and a second cohort treated with rt-PA. Glibenclamide or vehicle control was administered as an intravenous bolus at reperfusion, followed by continuous subcutaneous administration with an osmotic pump. RESULTS Compared to vehicle control, glibenclamide improved neurological outcome (median 7, interquartile range [IQR 6-8] vs. control median 6 [IQR 0-6], p = 0.025), reduced stroke volume (323 ± 42 vs. 484 ± 60 mm3, p < 0.01), swelling volume (10 ± 4 vs. 28 ± 7%, p < 0.01) and water content (84 ± 1 vs. 85 ± 1%, p < 0.05). Glibenclamide administration also reduced HT based on ECASS criteria, densitometry (0.94 ± 0.1 vs. 1.15 ± 0.2, p < 0.01), and quantitative hemoglobin concentration (2.7 ± 1.5 vs. 6.2 ± 4.6 uL, p = 0.011). In the second cohort with rt-PA coadministration, concordant effects on HT were observed with glibenclamide. CONCLUSIONS Taken together, these studies demonstrated that glibenclamide reduced the amount of edema and HT after severe ischemia. This study suggests that co-administration of glibenclamide may be worth further study in severe stroke patients treated with EVT with or without IV rt-PA.
Collapse
Affiliation(s)
- Takahiro Igarashi
- Division of Neurocritical Care, Center for Genomic Medicine and Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Cristina Sastre
- Division of Neurocritical Care, Center for Genomic Medicine and Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Zoe Wolcott
- Division of Neurocritical Care, Center for Genomic Medicine and Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - W Taylor Kimberly
- Division of Neurocritical Care, Center for Genomic Medicine and Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
13
|
Treatment with Atorvastatin During Vascular Remodeling Promotes Pericyte-Mediated Blood-Brain Barrier Maturation Following Ischemic Stroke. Transl Stroke Res 2021; 12:905-922. [PMID: 33423214 DOI: 10.1007/s12975-020-00883-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 10/22/2022]
Abstract
We previously showed that newly formed vessels in ischemic rat brain have high blood-brain barrier (BBB) permeability at 3 weeks after stroke due to a lack of major endothelial tight junction proteins (TJPs), which may exacerbate edema in stroke patients. Atorvastatin was suggested a dose-dependent pro-angiogenic effect and ameliorating BBB permeability beyond its cholesterol-lowering effects. This study examined our hypothesis that, during vascular remodeling after stroke, treatment with atorvastatin could facilitate BBB maturation in remodeling vasculature in ischemic brain. Adult spontaneously hypertensive rats underwent middle cerebral artery occlusion with reperfusion (MCAO/RP). Atorvastatin, at dose of 3 mg/kg, was delivered daily starting at 14 days after MCAO/RP onset for 7 days. The rats were studied at multiple time points up to 8 weeks with multimodal-MRI, behavior tests, immunohistochemistry, and biochemistry. The delayed treatment of atorvastatin significantly reduced infarct size and BBB permeability, restored cerebral blood flow, and improved the neurological outcome at 8 weeks after MCAO/RP. Postmortem studies showed that atorvastatin promoted angiogenesis and stabilized the newly formed vessels in peri-infarct areas. Importantly, atorvastatin facilitated maturation of BBB properties in the new vessels by promoting endothelial tight junction (TJ) formation. Further in vivo and in vitro studies demonstrated that proliferating peri-vascular pericytes expressing neural-glial antigen 2 (NG2) mediated the role of atorvastatin on BBB maturation through regulating endothelial TJ strand formations. Our results suggested a therapeutic potential of atorvastatin in facilitating a full BBB integrity and functional stroke recovery, and an essential role for pericyte-mediated endothelial TJ formation in remodeling vasculature.
Collapse
|
14
|
Hansen C, Sastre C, Wolcott Z, Bevers MB, Kimberly WT. Time-dependent, dynamic prediction of fatty acid-binding protein 4, Galectin-3, and soluble ST2 measurement with poor outcome after acute stroke. Int J Stroke 2020; 16:660-668. [PMID: 33167787 DOI: 10.1177/1747493020971166] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Time-dependent change in the level of biomarkers after stroke is not well understood. We sought to compare fatty acid-binding protein 4 (FABP4), Galectin-3, and soluble ST2 to ascertain for a change in prediction of outcome at admission and 48 h later. METHODS Plasma FABP4, Galectin-3, and soluble ST2 were measured in biospecimens from acute stroke patients at the time of admission (n = 383) and 48 h later (n = 244). Functional outcome was assessed at 90 days using the modified Rankin Scale and dichotomized into good (modified Rankin Scale 0-2) and poor outcome (modified Rankin Scale 3-6). RESULTS On admission, elevated levels of each biomarker predicted poor outcome (FABP4: OR 1.92, 95% CI 1.42-2.59, P < 0.0001; Galectin-3: OR 1.85, 95% CI 1.42-2.40, P < 0.0001; soluble ST2: OR 1.55, 95% CI 1.22-1.97, P < 0.0001) and death (FABP4: OR 2.45; 95% CI 1.51-3.98; P < 0.0001; Galectin-3: OR 2.12; 95% CI 1.50-3.30; P < 0.0001; soluble ST2: OR 2.17; 95% CI 1.58-2.99; P < 0.0001). At 48 h, soluble ST2 predicted poor outcome (OR 2.62, 95% CI 1.77-3.88, P < 0.0001) and mortality (OR 3.36, 95% CI 2.06-5.48, P < 0.0001), and Galectin-3 predicted mortality only (OR 1.81, 95% CI 1.05-3.10, P = 0.033). FABP4 measured at 48 h was not predictive of outcome or death. Associations of Galectin-3 and soluble ST2 with outcome or mortality were independent of age, sex, and NIHSS, whereas those with FABP4 were not. CONCLUSIONS Galectin-3 performed better when measured on admission, whereas soluble ST2 was predictive at admission and better at 48 h after stroke. The time-dependent differences may reflect the evolving role of these pathways after acute stroke.
Collapse
Affiliation(s)
- Christina Hansen
- Department of Neurology, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Cristina Sastre
- Department of Neurology, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Zoe Wolcott
- Department of Neurology, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Matthew B Bevers
- Divisions of Stroke, Cerebrovascular and Critical Care Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - W Taylor Kimberly
- Department of Neurology, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
15
|
Yazdanian M, Moazzami M, Shabani M, Cheragh Birjandi S. The Effect of Eight-Week Aerobic Exercise before Cerebral Ischemia on the Expression of NT-3 and TrkC Genes in Male Rats. MEDICAL LABORATORY JOURNAL 2020. [DOI: 10.29252/mlj.14.5.35] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
|
16
|
Rouleau N, Murugan NJ, Rusk W, Koester C, Kaplan DL. Matrix Deformation with Ectopic Cells Induced by Rotational Motion in Bioengineered Neural Tissues. Ann Biomed Eng 2020; 48:2192-2203. [PMID: 32671625 PMCID: PMC7405955 DOI: 10.1007/s10439-020-02561-6] [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: 03/21/2020] [Accepted: 06/29/2020] [Indexed: 11/30/2022]
Abstract
The brain's extracellular matrix (ECM) is a dynamic protein-based scaffold within which neural networks can form, self-maintain, and re-model. When the brain incurs injuries, microscopic tissue tears and active ECM re-modelling give way to abnormal brain structure and function including the presence of ectopic cells. Post-mortem and neuroimaging data suggest that the brains of jet pilots and astronauts, who are exposed to rotational forces, accelerations, and microgravity, display brain anomalies which could be indicative of a mechanodisruptive pathology. Here we present a model of non-impact-based brain injury induced by matrix deformation following mechanical shaking. Using a bioengineered 3D neural tissue platform, we designed a repetitive shaking paradigm to simulate subtle rotational acceleration. Our results indicate shaking induced ectopic cell clustering that could be inhibited by physically restraining tissue movement. Imaging revealed that the collagen substrate surrounding cells was deformed following shaking. Applied to neonatal rat brains, shaking induced deformation of extracellular spaces within the cerebral cortices and reduced the number of cell bodies at higher accelerations. We hypothesize that ECM deformation may represent a more significant role in brain injury progression than previously assumed and that the present model system contributes to its understanding as a phenomenon.
Collapse
Affiliation(s)
- Nicolas Rouleau
- Department of Biomedical Engineering, Science & Technology Center, Tufts University, Medford, MA, USA
- Initiative for Neural Science, Disease, and Engineering (INSciDE), Tufts University, Medford, USA
- The Allen Discovery Center, Tufts University, Medford, USA
| | - Nirosha J Murugan
- Department of Biomedical Engineering, Science & Technology Center, Tufts University, Medford, MA, USA
- The Allen Discovery Center, Tufts University, Medford, USA
- Department of Biology, Tufts University, Medford, USA
| | - William Rusk
- Department of Biomedical Engineering, Science & Technology Center, Tufts University, Medford, MA, USA
| | - Cole Koester
- Department of Biomedical Engineering, Science & Technology Center, Tufts University, Medford, MA, USA
| | - David L Kaplan
- Department of Biomedical Engineering, Science & Technology Center, Tufts University, Medford, MA, USA.
- Initiative for Neural Science, Disease, and Engineering (INSciDE), Tufts University, Medford, USA.
- The Allen Discovery Center, Tufts University, Medford, USA.
| |
Collapse
|
17
|
Lattanzi S, Di Napoli M, Ricci S, Divani AA. Matrix Metalloproteinases in Acute Intracerebral Hemorrhage. Neurotherapeutics 2020; 17:484-496. [PMID: 31975152 PMCID: PMC7283398 DOI: 10.1007/s13311-020-00839-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Spontaneous intracerebral hemorrhage (ICH) accounts for 10-30% of all strokes and affects more than one million people every year worldwide, and it is the stroke subtype associated with the highest rates of mortality and residual disability. So far, clinical trials have mainly targeted primary cerebral injury and have substantially failed to improve clinical outcomes. The understanding of the pathophysiology of early and delayed injury after ICH is, hence, of paramount importance to identify potential targets of intervention and develop effective therapeutic strategies. Matrix metalloproteinases (MMPs) represent a ubiquitous superfamily of structurally related zinc-dependent endopeptidases able to degrade any component of the extracellular matrix. They are upregulated after ICH, in which different cell types, including leukocytes, activated microglia, neurons, and endothelial cells, are involved in their synthesis and secretion. The aim of this review is to summarize the available experimental and clinical evidence about the role of MMPs in brain injury following spontaneous ICH and provide critical insights into the underlying mechanisms.
Collapse
Affiliation(s)
- Simona Lattanzi
- Neurological Clinic, Department of Experimental and Clinical Medicine, Marche Polytechnic University, Ancona, Italy
| | - Mario Di Napoli
- Department of Neurology and Stroke Unit, San Camillo de' Lellis District General Hospital, Rieti, Italy
| | - Silvia Ricci
- Department of Neurology and Stroke Unit, San Camillo de' Lellis District General Hospital, Rieti, Italy
| | - Afshin A Divani
- Department of Neurology, University of New Mexico, Albuquerque, New Mexico, USA.
| |
Collapse
|
18
|
Dharmalingam P, Talakatta G, Mitra J, Wang H, Derry PJ, Nilewski LG, McHugh EA, Fabian RH, Mendoza K, Vasquez V, Hegde PM, Kakadiaris E, Roy T, Boldogh I, Hegde VL, Mitra S, Tour JM, Kent TA, Hegde ML. Pervasive Genomic Damage in Experimental Intracerebral Hemorrhage: Therapeutic Potential of a Mechanistic-Based Carbon Nanoparticle. ACS NANO 2020; 14:2827-2846. [PMID: 32049495 PMCID: PMC7850811 DOI: 10.1021/acsnano.9b05821] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Therapy for intracerebral hemorrhage (ICH) remains elusive, in part dependent on the severity of the hemorrhage itself as well as multiple deleterious effects of blood and its breakdown products such as hemin and free iron. While oxidative injury and genomic damage have been seen following ICH, the details of this injury and implications remain unclear. Here, we discovered that, while free iron produced mostly reactive oxygen species (ROS)-related single-strand DNA breaks, hemin unexpectedly induced rapid and persistent nuclear and mitochondrial double-strand breaks (DSBs) in neuronal and endothelial cell genomes and in mouse brains following experimental ICH comparable to that seen with γ radiation and DNA-complexing chemotherapies. Potentially as a result of persistent DSBs and the DNA damage response, hemin also resulted in senescence phenotype in cultured neurons and endothelial cells. Subsequent resistance to ferroptosis reported in other senescent cell types was also observed here in neurons. While antioxidant therapy prevented senescence, cells became sensitized to ferroptosis. To address both senescence and resistance to ferroptosis, we synthesized a modified, catalytic, and rapidly internalized carbon nanomaterial, poly(ethylene glycol)-conjugated hydrophilic carbon clusters (PEG-HCC) by covalently bonding the iron chelator, deferoxamine (DEF). This multifunctional nanoparticle, DEF-HCC-PEG, protected cells from both senescence and ferroptosis and restored nuclear and mitochondrial genome integrity in vitro and in vivo. We thus describe a potential molecular mechanism of hemin/iron-induced toxicity in ICH that involves a rapid induction of DSBs, senescence, and the consequent resistance to ferroptosis and provide a mechanistic-based combinatorial therapeutic strategy.
Collapse
Affiliation(s)
- Prakash Dharmalingam
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, Texas 77030, United States
| | - Girish Talakatta
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, Texas 77030, United States
| | - Joy Mitra
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, Texas 77030, United States
| | - Haibo Wang
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, Texas 77030, United States
| | - Paul J Derry
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas 77030, United States
| | | | - Emily A McHugh
- Department of Chemistry, Rice University, Houston, Texas 77005, United States
| | - Roderic H Fabian
- Department of Neurology, Baylor College of Medicine, and Michael E. DeBakey VA Medical Center, Houston, Texas 77030, United States
| | - Kimberly Mendoza
- Department of Chemistry, Rice University, Houston, Texas 77005, United States
| | - Velmarini Vasquez
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, Texas 77030, United States
| | - Pavana M Hegde
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, Texas 77030, United States
| | - Eugenia Kakadiaris
- Department of Chemistry, Rice University, Houston, Texas 77005, United States
| | - Trenton Roy
- Department of Chemistry, Rice University, Houston, Texas 77005, United States
| | - Istvan Boldogh
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Venkatesh L Hegde
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, Texas 77030, United States
| | - Sankar Mitra
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, Texas 77030, United States
- Weill Medical College of Cornell University, New York, New York 10065, United States
| | - James M Tour
- Departments of Chemistry, Computer Science, Materials Science and NanoEngineering, Smalley-Curl Institute and the NanoCarbon Center, Rice University, Houston, Texas 77005, United States
| | - Thomas A Kent
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas 77030, United States
- Department of Chemistry, Rice University, Houston, Texas 77005, United States
- Stanley H. Appel Department of Neurology, Houston Methodist Hospital and Research Institute, Houston, Texas 77030, United States
| | - Muralidhar L Hegde
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, Texas 77030, United States
- Weill Medical College of Cornell University, New York, New York 10065, United States
- Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Neurological Institute, Houston Methodist, Houston, Texas 77030, United States
| |
Collapse
|
19
|
Zhu X, Hatfield J, Sullivan JK, Xu F, Van Nostrand WE. Robust neuroinflammation and perivascular pathology in rTg-DI rats, a novel model of microvascular cerebral amyloid angiopathy. J Neuroinflammation 2020; 17:78. [PMID: 32127016 PMCID: PMC7055091 DOI: 10.1186/s12974-020-01755-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 02/21/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Cerebral amyloid angiopathy (CAA) is a common cerebral small vessel disease of the aged and a prominent comorbidity of Alzheimer's disease (AD). CAA can promote a variety of vascular-related pathologies including neuroinflammation, cerebral infarction, and hemorrhages, which can all contribute to vascular cognitive impairment and dementia (VCID). Our understanding of the pathogenesis of CAA remains limited and further investigation of this condition requires better preclinical animal models that more accurately reflect the human disease. Recently, we generated a novel transgenic rat model for CAA (rTg-DI) that develops robust and progressive microvascular CAA, consistent microhemorrhages and behavioral deficits. METHODS In the current study, we investigated perivascular pathological processes that accompany the onset and progressive accumulation of microvascular CAA in this model. Cohorts of rTg-DI rats were aged to 3 months with the onset of CAA and to 12 months with advanced stage disease and then quantitatively analyzed for progression of CAA, perivascular glial activation, inflammatory markers, and perivascular stress. RESULTS The rTg-DI rats developed early-onset and robust accumulation of microvascular amyloid. As the disease progressed, rTg-DI rats exhibited increased numbers of astrocytes and activated microglia which were accompanied by expression of a distinct subset of inflammatory markers, perivascular pericyte degeneration, astrocytic caspase 3 activation, and disruption of neuronal axonal integrity. CONCLUSIONS Taken together, these results demonstrate that rTg-DI rats faithfully mimic numerous aspects of human microvascular CAA and provide new experimental insight into the pathogenesis of neuroinflammation and perivascular stress associated with the onset and progression of this condition, suggesting new potential therapeutic targets for this condition. The rTg-DI rats provide an improved preclinical platform for developing new biomarkers and testing therapeutic strategies for microvascular CAA.
Collapse
Affiliation(s)
- Xiaoyue Zhu
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, George & Anne Ryan Institute for Neuroscience, University of Rhode Island, 130 Flagg Road, Kingston, RI, 02881, USA
| | - Joshua Hatfield
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, George & Anne Ryan Institute for Neuroscience, University of Rhode Island, 130 Flagg Road, Kingston, RI, 02881, USA
- Present Address: Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY, 11724, USA
| | - Joseph K Sullivan
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, George & Anne Ryan Institute for Neuroscience, University of Rhode Island, 130 Flagg Road, Kingston, RI, 02881, USA
- Present Address: New York Medical College, 40 Sunshine Cottage Road, Valhalla, NY, 10595, USA
| | - Feng Xu
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, George & Anne Ryan Institute for Neuroscience, University of Rhode Island, 130 Flagg Road, Kingston, RI, 02881, USA
| | - William E Van Nostrand
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, George & Anne Ryan Institute for Neuroscience, University of Rhode Island, 130 Flagg Road, Kingston, RI, 02881, USA.
| |
Collapse
|
20
|
Zhang W, Zhu L, An C, Wang R, Yang L, Yu W, Li P, Gao Y. The blood brain barrier in cerebral ischemic injury – Disruption and repair. BRAIN HEMORRHAGES 2020. [DOI: 10.1016/j.hest.2019.12.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
21
|
Hamanaka G, Kubo T, Ohtomo R, Takase H, Reyes-Bricio E, Oribe S, Osumi N, Lok J, Lo EH, Arai K. Microglial responses after phagocytosis: Escherichia coli bioparticles, but not cell debris or amyloid beta, induce matrix metalloproteinase-9 secretion in cultured rat primary microglial cells. Glia 2020; 68:1435-1444. [PMID: 32057146 DOI: 10.1002/glia.23791] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 01/24/2020] [Accepted: 01/27/2020] [Indexed: 12/13/2022]
Abstract
Upon infection or brain damage, microglia are activated to play roles in immune responses, including phagocytosis and soluble factor release. However, little is known whether the event of phagocytosis could be a trigger for releasing soluble factors from microglia. In this study, we tested if microglia secrete a neurovascular mediator matrix metalloproteinase-9 (MMP-9) after phagocytosis in vitro. Primary microglial cultures were prepared from neonatal rat brains. Cultured microglia phagocytosed Escherichia coli bioparticles within 2 hr after incubation and started to secrete MMP-9 at around 12 hr after the phagocytosis. A TLR4 inhibitor TAK242 suppressed the E. coli-bioparticle-induced MMP-9 secretion. However, TAK242 did not change the engulfment of E. coli bioparticles in microglial cultures. Because lipopolysaccharides (LPS), the major component of the outer membrane of E. coli, also induced MMP-9 secretion in a dose-response manner and because the response was inhibited by TAK242 treatment, we assumed that the LPS-TLR4 pathway, which was activated by adhering to the substance, but not through the engulfing process of phagocytosis, would play a role in releasing MMP-9 from microglia after E. coli bioparticle treatment. To support the finding that the engulfing step would not be a critical trigger for MMP-9 secretion after the event of phagocytosis in microglia, we confirmed that cell debris and amyloid beta were both captured into microglia via phagocytosis, but neither of them induced MMP-9 secretion from microglia. Taken together, these data demonstrate that microglial response in MMP-9 secretion after phagocytosis differs depending on the types of particles/substances that microglia encountered.
Collapse
Affiliation(s)
- Gen Hamanaka
- Neuroprotection Research Laboratory, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.,Neuroprotection Research Laboratory, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Tomoya Kubo
- Neuroprotection Research Laboratory, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.,Neuroprotection Research Laboratory, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Ryo Ohtomo
- Neuroprotection Research Laboratory, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.,Neuroprotection Research Laboratory, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Hajime Takase
- Neuroprotection Research Laboratory, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.,Neuroprotection Research Laboratory, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Estefania Reyes-Bricio
- Neuroprotection Research Laboratory, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.,Neuroprotection Research Laboratory, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Shuntaro Oribe
- Department of Developmental Neuroscience, United Centers for Advanced Research and Translational Medicine, Tohoku University School of Medicine, Sendai, Japan
| | - Noriko Osumi
- Department of Developmental Neuroscience, United Centers for Advanced Research and Translational Medicine, Tohoku University School of Medicine, Sendai, Japan
| | - Josephine Lok
- Neuroprotection Research Laboratory, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.,Neuroprotection Research Laboratory, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.,Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts
| | - Eng H Lo
- Neuroprotection Research Laboratory, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.,Neuroprotection Research Laboratory, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Ken Arai
- Neuroprotection Research Laboratory, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.,Neuroprotection Research Laboratory, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
22
|
Bell AH, Miller SL, Castillo-Melendez M, Malhotra A. The Neurovascular Unit: Effects of Brain Insults During the Perinatal Period. Front Neurosci 2020; 13:1452. [PMID: 32038147 PMCID: PMC6987380 DOI: 10.3389/fnins.2019.01452] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 12/30/2019] [Indexed: 12/31/2022] Open
Abstract
The neurovascular unit (NVU) is a relatively recent concept in neuroscience that broadly describes the relationship between brain cells and their blood vessels. The NVU incorporates cellular and extracellular components involved in regulating cerebral blood flow and blood-brain barrier function. The NVU within the adult brain has attracted strong research interest and its structure and function is well described, however, the NVU in the developing brain over the fetal and neonatal period remains much less well known. One area of particular interest in perinatal brain development is the impact of known neuropathological insults on the NVU. The aim of this review is to synthesize existing literature to describe structure and function of the NVU in the developing brain, with a particular emphasis on exploring the effects of perinatal insults. Accordingly, a brief overview of NVU components and function is provided, before discussion of NVU development and how this may be affected by perinatal pathologies. We have focused this discussion around three common perinatal insults: prematurity, acute hypoxia, and chronic hypoxia. A greater understanding of processes affecting the NVU in the perinatal period may enable application of targeted therapies, as well as providing a useful basis for research as it expands further into this area.
Collapse
Affiliation(s)
- Alexander H. Bell
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Suzanne L. Miller
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Margie Castillo-Melendez
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Atul Malhotra
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Monash Newborn, Monash Children’s Hospital, Melbourne, VIC, Australia
| |
Collapse
|
23
|
Minarelli J, Davis EL, Dickerson A, Moore WC, Mejia JA, Gugala Z, Olmsted-Davis EA, Davis AR. Characterization of neuromas in peripheral nerves and their effects on heterotopic bone formation. Mol Pain 2019; 15:1744806919838191. [PMID: 30813850 PMCID: PMC6440042 DOI: 10.1177/1744806919838191] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The formation of neuromas involves expansion of the cellular components of peripheral nerves. The onset of these disorganized tumors involves activation of sensory nerves and neuroinflammation. Particularly problematic in neuroma is arborization of axons leading to extreme, neuropathic pain. The most common sites for neuroma are the ends of transected nerves following injury; however, this rodent model does not reliably result in neuroma formation. In this study, we established a rodent model of neuroma in which the sciatic nerve was loosely ligated with two chromic gut sutures. This model formed neuromas reliably (∼95%), presumably through activation of the neural inflammatory cascade. Resulting neuromas had a disorganized structure and a significant number of replicating cells. Quantification of changes in perineurial and Schwann cells showed a significant increase in these populations. Immunohistochemical analysis showed the presence of β-tubulin 3 in the rapidly expanding nerve and a decrease in neurofilament heavy chain compared to the normal nerve, suggesting the axons forming a disorganized structure. Measurement of the permeability of the blood-nerve barrier shows that it opened almost immediately and remained open as long as 10 days. Studies using an antagonist of the β3-adrenergic receptor (L-748,337) or cromolyn showed a significant reduction in tumor size and cell expansion as determined by flow cytometry, with an improvement in the animal's gait detected using a Catwalk system. Previous studies in our laboratory have shown that heterotopic ossification is also a result of the activation of neuroinflammation. Since heterotopic ossification and neuroma often occur together in amputees, they were induced in the same limbs of the study animals. More heterotopic bone was formed in animals with neuromas as compared to those without. These data collectively suggest that perturbation of early neuroinflammation with compounds such as L-748,337 and cromolyn may reduce formation of neuromas.
Collapse
Affiliation(s)
- Jordan Minarelli
- 1 Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - Eleanor L Davis
- 1 Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - Austin Dickerson
- 1 Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - William C Moore
- 1 Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - Julio A Mejia
- 1 Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - Zbigniew Gugala
- 2 Department of Orthopedic Surgery and Rehabilitation, University of Texas Medical Branch, Galveston, TX, USA
| | - Elizabeth A Olmsted-Davis
- 1 Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA.,3 Department of Pediatrics - Section Hematology/Oncology, Baylor College of Medicine, Houston, TX, USA.,4 Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Alan R Davis
- 1 Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA.,3 Department of Pediatrics - Section Hematology/Oncology, Baylor College of Medicine, Houston, TX, USA.,4 Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
24
|
Rubenstein R, Sharma DR, Chang B, Oumata N, Cam M, Vaucelle L, Lindberg MF, Chiu A, Wisniewski T, Wang KKW, Meijer L. Novel Mouse Tauopathy Model for Repetitive Mild Traumatic Brain Injury: Evaluation of Long-Term Effects on Cognition and Biomarker Levels After Therapeutic Inhibition of Tau Phosphorylation. Front Neurol 2019; 10:124. [PMID: 30915013 PMCID: PMC6421297 DOI: 10.3389/fneur.2019.00124] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 01/30/2019] [Indexed: 12/20/2022] Open
Abstract
Traumatic brain injury (TBI) is a risk factor for a group of neurodegenerative diseases termed tauopathies, which includes Alzheimer's disease and chronic traumatic encephalopathy (CTE). Although TBI is stratified by impact severity as either mild (m), moderate or severe, mTBI is the most common and the most difficult to diagnose. Tauopathies are pathologically related by the accumulation of hyperphosphorylated tau (P-tau) and increased total tau (T-tau). Here we describe: (i) a novel human tau-expressing transgenic mouse model, TghTau/PS1, to study repetitive mild closed head injury (rmCHI), (ii) quantitative comparison of T-tau and P-tau from brain and plasma in TghTau/PS1 mice over a 12 month period following rmCHI (and sham), (iii) the usefulness of P-tau as an early- and late-stage blood-based biochemical biomarker for rmCHI, (iii) the influence of kinase-targeted therapeutic intervention on rmCHI-associated cognitive deficits using a combination of lithium chloride (LiCl) and R-roscovitine (ros), and (iv) correlation of behavioral and cognitive changes with concentrations of the brain and blood-based T-tau and P-tau. Compared to sham-treated mice, behavior changes and cognitive deficits of rmCHI-treated TghTau/PS1 mice correlated with increases in both cortex and plasma T-tau and P-tau levels over 12 months. In addition, T-tau, but more predominantly P-tau, levels were significantly reduced in the cortex and plasma by LiCl + ros approaching the biomarker levels in sham and drug-treated sham mice (the drugs had only modest effects on the T-tau and P-tau levels in sham mice) throughout the 12 month study period. Furthermore, although we also observed a reversal of the abnormal behavior and cognitive deficits in the drug-treated rmCHI mice (compared to the untreated rmCHI mice) throughout the time course, these drug-treated effects were most pronounced up until 10 and 12 months where the abnormal behavior and cognition deficits began to gradually increase. These studies describe: (a) a translational relevant animal model for TBI-linked tauopathies, and (b) utilization of T-tau and P-tau as rmCHI biomarkers in plasma to monitor novel therapeutic strategies and treatment regimens for these neurodegenerative diseases.
Collapse
Affiliation(s)
- Richard Rubenstein
- Laboratory of Neurodegenerative Diseases and CNS Biomarker Discovery, Departments of Neurology and Physiology/Pharmacology, SUNY Downstate Medical Center, Brooklyn, NY, United States
| | - Deep R Sharma
- Laboratory of Neurodegenerative Diseases and CNS Biomarker Discovery, Departments of Neurology and Physiology/Pharmacology, SUNY Downstate Medical Center, Brooklyn, NY, United States
| | - Binggong Chang
- Laboratory of Neurodegenerative Diseases and CNS Biomarker Discovery, Departments of Neurology and Physiology/Pharmacology, SUNY Downstate Medical Center, Brooklyn, NY, United States
| | - Nassima Oumata
- ManRos Therapeutics, Centre de Perharidy, Roscoff, France
| | - Morgane Cam
- ManRos Therapeutics, Centre de Perharidy, Roscoff, France
| | - Lise Vaucelle
- ManRos Therapeutics, Centre de Perharidy, Roscoff, France
| | | | - Allen Chiu
- Laboratory of Neurodegenerative Diseases and CNS Biomarker Discovery, Departments of Neurology and Physiology/Pharmacology, SUNY Downstate Medical Center, Brooklyn, NY, United States
| | - Thomas Wisniewski
- Center for Cognitive Neurology and Departments of Neurology, Pathology and Psychiatry, New York University School of Medicine, New York, NY, United States
| | - Kevin K W Wang
- Program for Neurotrauma, Neuroproteomics and Biomarker Research, Departments of Emergency Medicine, Psychiatry and Neuroscience, University of Florida, Gainesville, FL, United States
| | - Laurent Meijer
- ManRos Therapeutics, Centre de Perharidy, Roscoff, France
| |
Collapse
|
25
|
Chen H, Chen X, Luo Y, Shen J. Potential molecular targets of peroxynitrite in mediating blood–brain barrier damage and haemorrhagic transformation in acute ischaemic stroke with delayed tissue plasminogen activator treatment. Free Radic Res 2018; 52:1220-1239. [PMID: 30468092 DOI: 10.1080/10715762.2018.1521519] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Hansen Chen
- School of Chinese Medicine, the University of Hong Kong, PR China
- Shenzhen Institute of Research and Innovation (HKU-SIRI), University of Hong Kong, Hong Kong, PR China
| | - Xi Chen
- Department of Core Facility, the People’s Hospital of Bao-an Shenzhen, Shenzhen, PR China
- The 8th People’s Hospital of Shenzhen, the Affiliated Bao-an Hospital of Southern Medical University, Shenzhen, PR China
| | - Yunhao Luo
- School of Chinese Medicine, the University of Hong Kong, PR China
| | - Jiangang Shen
- School of Chinese Medicine, the University of Hong Kong, PR China
- Shenzhen Institute of Research and Innovation (HKU-SIRI), University of Hong Kong, Hong Kong, PR China
| |
Collapse
|
26
|
Setyopranoto I, Malueka RG, Panggabean AS, Widyadharma IPE, Sadewa AH, Lamsudin R, Wibowo S. Association between Increased Matrix Metalloproteinase-9 (MMP-9) Levels with Hyperglycaemia Incidence in Acute Ischemic Stroke Patients. Open Access Maced J Med Sci 2018; 6:2067-2072. [PMID: 30559862 PMCID: PMC6290450 DOI: 10.3889/oamjms.2018.459] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 10/29/2018] [Accepted: 10/30/2018] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND: Hyperglycemia is common in acute stroke patients. Hyperglycemia can induce the production of reactive oxygen species, causing increased activity of matrix metalloproteinase-9 (MMP-9). AIM: This study aimed to determine an association between the increased levels of MMP-9 and the incidence of hyperglycemia in acute ischemic stroke patients. METHODS: This is a case-control study. Acute ischemic stroke patients admitted to the Stroke Unit of a reference hospital in Yogyakarta, Indonesia was divided into the hyperglycemic and non-hyperglycemic group. Demographic and clinical characteristics of each subject were recorded, and blood levels of MMP-9 were measured. Seventy-one patients were recruited, 40 subjects in the hyperglycemic group and 31 subjects in the non-hyperglycemic group. RESULTS: The median levels of blood MMP-9 level in the hyperglycemic and non-hyperglycemic group were 974.37 and 748.48 ng/mL, respectively, and the difference was statistically not significant (95% CI, 191.24-2849.53; p = 0.07). When the calculated cut-off point of 600.99 ng/mL was used, the proportion of patients with higher MMP-9 levels was significantly more in the hyperglycemic group compared with the ones in the non-hyperglycemic group (82.5% and 54.8%, respectively; OR = 3.88; p = 0.011). CONCLUSION: We concluded that the proportion of patients with MMP-9 level >600.99 ng/mL was significantly higher in acute ischemic stroke patients with hyperglycemia.
Collapse
Affiliation(s)
- Ismail Setyopranoto
- Department of Neurology, Faculty of Medicine, Universitas Gadjah Mada and Dr Sardjito General Hospital, Yogyakarta, Indonesia
| | - Rusdy Ghazali Malueka
- Department of Neurology, Faculty of Medicine, Universitas Gadjah Mada and Dr Sardjito General Hospital, Yogyakarta, Indonesia
| | - Andre Stefanus Panggabean
- Department of Neurology, Faculty of Medicine, Universitas Gadjah Mada and Dr Sardjito General Hospital, Yogyakarta, Indonesia
| | - I Putu Eka Widyadharma
- Department of Neurology, Faculty of Medicine, Udayana University and Sanglah General Hospital, Bali, Indonesia
| | - Ahmad Hamim Sadewa
- Department of Biochemistry, Faculty of Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Rusdi Lamsudin
- Department of Neurology, Faculty of Medicine, Universitas Gadjah Mada and Dr Sardjito General Hospital, Yogyakarta, Indonesia
| | - Samekto Wibowo
- Department of Neurology, Faculty of Medicine, Universitas Gadjah Mada and Dr Sardjito General Hospital, Yogyakarta, Indonesia
| |
Collapse
|
27
|
Abstract
PRIMARY OBJECTIVE The purpose of this paper is to review the clinical and research utility and applications of blood, cerebrospinal fluid (CSF), and cerebral microdialysis biomarkers in traumatic brain injury (TBI). RESEARCH DESIGN Not applicable. METHODS AND PROCEDURES A selective review was performed on these biofluid biomarkers in TBI. MAIN OUTCOME AND RESULTS Neurofilament heavy chain protein (NF-H), glial fibrillary acidic protein (GFAP), ubiquitin C-terminal hydrolase-L1 (UCHL1), neuron-specific enolase (NSE), myelin basic protein (MBP), tau, and s100β blood biomarkers are elevated during the acute phase of severe head trauma but have key limitations in their research and clinical applications to mild TBI (mTBI). CSF biomarkers currently provide the best reflection of the central nervous system (CNS) pathobiological processes in TBI. Both animal and human studies of TBI have demonstrated the importance of serial sampling of biofluids and suggest that CSF biomarkers may be better equipped to characterize both TBI severity and temporal profiles. CONCLUSIONS The identification of biofluid biomarkers could play a vital role in identifying, diagnosing, and treating the underlying individual pathobiological changes of TBI. CNS-derived exosomes analyzed by ultra-high sensitivity detection methods have the potential to identify blood biomarkers for the range of TBI severity and time course.
Collapse
Affiliation(s)
- Denes V Agoston
- a Department of Anatomy, Physiology and Genetics , Uniformed Services University , Bethesda , MD , USA.,b Department of Neuroscience , Karolinska Institutet , Stockholm , Sweden
| | - Andrew Shutes-David
- c VA Northwest Network Mental Illness Research, Education, and Clinical Center , Veterans Affairs Puget Sound Health Care System , Seattle , WA , USA.,d Geriatric Research, Education, and Clinical Center , Veterans Affairs Puget Sound Health Care System , Seattle , WA , USA
| | - Elaine R Peskind
- c VA Northwest Network Mental Illness Research, Education, and Clinical Center , Veterans Affairs Puget Sound Health Care System , Seattle , WA , USA.,e Department of Psychiatry and Behavioral Sciences , University of Washington , Seattle , WA , USA
| |
Collapse
|
28
|
Chen HS, Chen X, Li WT, Shen JG. Targeting RNS/caveolin-1/MMP signaling cascades to protect against cerebral ischemia-reperfusion injuries: potential application for drug discovery. Acta Pharmacol Sin 2018; 39:669-682. [PMID: 29595191 PMCID: PMC5943912 DOI: 10.1038/aps.2018.27] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 01/26/2018] [Indexed: 02/07/2023] Open
Abstract
Reactive nitrogen species (RNS) play important roles in mediating cerebral ischemia-reperfusion injury. RNS activate multiple signaling pathways and participate in different cellular events in cerebral ischemia-reperfusion injury. Recent studies have indicated that caveolin-1 and matrix metalloproteinase (MMP) are important signaling molecules in the pathological process of ischemic brain injury. During cerebral ischemia-reperfusion, the production of nitric oxide (NO) and peroxynitrite (ONOO−), two representative RNS, down-regulates the expression of caveolin-1 (Cav-1) and, in turn, further activates nitric oxide synthase (NOS) to promote RNS generation. The increased RNS further induce MMP activation and mediate disruption of the blood-brain barrier (BBB), aggravating the brain damage in cerebral ischemia-reperfusion injury. Therefore, the feedback interaction among RNS/Cav-1/MMPs provides an amplified mechanism for aggravating ischemic brain damage during cerebral ischemia-reperfusion injury. Targeting the RNS/Cav-1/MMP pathway could be a promising therapeutic strategy for protecting against cerebral ischemia-reperfusion injury. In this mini-review article, we highlight the important role of the RNS/Cav-1/MMP signaling cascades in ischemic stroke injury and review the current progress of studies seeking therapeutic compounds targeting the RNS/Cav-1/MMP signaling cascades to attenuate cerebral ischemia-reperfusion injury. Several representative natural compounds, including calycosin-7-O-β-D-glucoside, baicalin, Momordica charantia polysaccharide (MCP), chlorogenic acid, lutein and lycopene, have shown potential for targeting the RNS/Cav-1/MMP signaling pathway to protect the brain in ischemic stroke. Therefore, the RNS/Cav-1/MMP pathway is an important therapeutic target in ischemic stroke treatment.
Collapse
|
29
|
Free Radical Damage in Ischemia-Reperfusion Injury: An Obstacle in Acute Ischemic Stroke after Revascularization Therapy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:3804979. [PMID: 29770166 PMCID: PMC5892600 DOI: 10.1155/2018/3804979] [Citation(s) in RCA: 298] [Impact Index Per Article: 42.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 12/07/2017] [Indexed: 12/16/2022]
Abstract
Acute ischemic stroke is a common cause of morbidity and mortality worldwide. Thrombolysis with recombinant tissue plasminogen activator and endovascular thrombectomy are the main revascularization therapies for acute ischemic stroke. However, ischemia-reperfusion injury after revascularization therapy can result in worsening outcomes. Among all possible pathological mechanisms of ischemia-reperfusion injury, free radical damage (mainly oxidative/nitrosative stress injury) has been found to play a key role in the process. Free radicals lead to protein dysfunction, DNA damage, and lipid peroxidation, resulting in cell death. Additionally, free radical damage has a strong connection with inducing hemorrhagic transformation and cerebral edema, which are the major complications of revascularization therapy, and mainly influencing neurological outcomes due to the disruption of the blood-brain barrier. In order to get a better clinical prognosis, more and more studies focus on the pharmaceutical and nonpharmaceutical neuroprotective therapies against free radical damage. This review discusses the pathological mechanisms of free radicals in ischemia-reperfusion injury and adjunctive neuroprotective therapies combined with revascularization therapy against free radical damage.
Collapse
|
30
|
Redondo-Castro E, Cunningham CJ, Miller J, Brown H, Allan SM, Pinteaux E. Changes in the secretome of tri-dimensional spheroid-cultured human mesenchymal stem cells in vitro by interleukin-1 priming. Stem Cell Res Ther 2018; 9:11. [PMID: 29343288 PMCID: PMC5773162 DOI: 10.1186/s13287-017-0753-5] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 12/18/2017] [Accepted: 12/19/2017] [Indexed: 12/30/2022] Open
Abstract
Background Mesenchymal stem cells (MSCs) are one of the most promising candidates for the treatment of major neurological disorders. Desirable therapeutic properties of MSCs include reparative and regenerative potential but, despite their proven safety, the efficacy of MSCs remains controversial. Therefore, it is essential to optimise culture protocols to enhance the therapeutic potential of the MSC secretome. Here we aimed to: assess the increase in secretion of cytokines that may induce repair, regeneration, or immunomodulation when cultured in three dimensions; study the effect of interleukin (IL)-1 priming on two- (2D) and three-dimensional (3D) cultures of MSC; and evaluate the potential use of the modified secretome using microglial-MSC co-cultures. Methods We established a 3D spheroid culture of human MSCs, and compared the secretome in 2D and 3D cultures under primed (IL-1) and unprimed conditions. BV2 microglial cells were stimulated with lipopolysaccharide (LPS) and treated with spheroid conditioned media (CM) or were co-cultured with whole spheroids. Concentrations of secreted cytokines were determined by enzyme-linked immunosorbent assay (ELISA). Protein arrays were used to further evaluate the effect of IL-1 priming in 2D and 3D cultures. Results 3D culture of MSCs significantly increased secretion of the IL-1 receptor antagonist (IL-1Ra), vascular endothelial growth factor (VEGF), and granulocyte-colony stimulating factor (G-CSF) compared with 2D culture, despite priming treatments with IL-1 being more effective in 2D than in 3D. The addition of CM of 3D-MSCs reduced LPS-induced tumour necrosis factor (TNF)-α secretion from BV2 cells, while the 3D spheroid co-cultured with the BV2 cells induced an increase in IL-6, but had no effect on TNF-α release. Protein arrays indicated that priming treatments trigger a more potent immune profile which is necessary to orchestrate an effective tissue repair. This effect was lost in 3D, partly because of the overexpression of IL-6. Conclusions Increased secretion of anti-inflammatory markers occurs when MSCs are cultured in 3D, but this specific secretome did not translate into anti-inflammatory effects on LPS-treated BV2 cells in co-culture. These data highlight the importance of optimising priming treatments and culture conditions to maximise the therapeutic potential of MSC spheroids.
Collapse
Affiliation(s)
- Elena Redondo-Castro
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Catriona J Cunningham
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Jonjo Miller
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Helena Brown
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Stuart M Allan
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Emmanuel Pinteaux
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| |
Collapse
|
31
|
Wang TH, Xiong LL, Yang SF, You C, Xia QJ, Xu Y, Zhang P, Wang SF, Liu J. LPS Pretreatment Provides Neuroprotective Roles in Rats with Subarachnoid Hemorrhage by Downregulating MMP9 and Caspase3 Associated with TLR4 Signaling Activation. Mol Neurobiol 2017; 54:7746-7760. [PMID: 27844284 DOI: 10.1007/s12035-016-0259-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 10/24/2016] [Indexed: 02/05/2023]
Abstract
Subarachnoid hemorrhage (SAH), as a severe brain disease, has high morbidity and mortality. SAH usually induced neurological dysfunction or death and the treatment is far from satisfaction. Here, we investigated the effect of low dose of LPS pretreatment and underlying molecular mechanism in rat SAH model. Firstly, SAH model was induced by prechiasmal cistern injection method (SAH1) and common carotid artery-prechiasmal cistern shunt method (SAH2), respectively, to select the more suitable SAH model. At 6, 12, 24, 48, and 72 h after SAH, brain injury including neurological dysfunction, blood-brain barrier disruption, brain edema, and cell apoptosis were detected. And the expression of MMP9, HMGB1/TLR4, and caspase3 in cortex were also explored. Then, SB-3CT, an inhibitor of MMP9, was administrated to investigate the exact function of MMP9 in the brain injury at 24 h after SAH. Moreover, low dose of LPS was used to verify whether it had nerve protection after SAH and the mechanism involving in MMP9 and caspase 3 was investigated. Our results showed SAH1 seems to be the most suitable SAH model. In addition, MMP9 activated by HMGB1/TLR4 may promote or aggravate brain injury, while inhibiting MMP9 via SB-3CT exerted a neuroprotective effect. Moreover, LPS improved the neurological dysfunction, reduced Evans blue extravasation and brain edema, and inhibited cell apoptosis of cortex in rats with brain injury induced by SAH. Importantly, LPS pretreatment increased the expression level of TLR4, and decreased the level of MMP9 and caspase3. Therefore, the present study revealed that low dose of LPS pretreatment could provide neuroprotective effects on brain injury caused by SAH via downregulating MMP9 and caspase3 and activating TLR4 signal pathway.
Collapse
Affiliation(s)
- Ting-Hua Wang
- Institute of Neurological Disease, and Department of Neurosurgery, Translational Neuroscience Center, the state key laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Liu-Lin Xiong
- Institute of Neurological Disease, and Department of Neurosurgery, Translational Neuroscience Center, the state key laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Shuai-Fen Yang
- Institute of Neurological Disease, and Department of Neurosurgery, Translational Neuroscience Center, the state key laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Chao You
- Institute of Neurological Disease, and Department of Neurosurgery, Translational Neuroscience Center, the state key laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Qing-Jie Xia
- Institute of Neurological Disease, and Department of Neurosurgery, Translational Neuroscience Center, the state key laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Yang Xu
- Institute of Neurological Disease, and Department of Neurosurgery, Translational Neuroscience Center, the state key laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Piao Zhang
- Animal Zoology Department, Institute of Neuroscience, Kunming medical University, Kunming, 650000, China
| | - Shu-Fen Wang
- Yunnan Key Laboratory of Stem Cells and Regenerative Medicine, Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming, 650000, China.
| | - Jia Liu
- Institute of Neurological Disease, and Department of Neurosurgery, Translational Neuroscience Center, the state key laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China.
- Animal Zoology Department, Institute of Neuroscience, Kunming medical University, Kunming, 650000, China.
| |
Collapse
|
32
|
Li Y, Xu L, Zeng K, Xu Z, Suo D, Peng L, Ren T, Sun Z, Yang W, Jin X, Yang L. Propane-2-sulfonic acid octadec-9-enyl-amide, a novel PPARα/γ dual agonist, protects against ischemia-induced brain damage in mice by inhibiting inflammatory responses. Brain Behav Immun 2017; 66:289-301. [PMID: 28736035 DOI: 10.1016/j.bbi.2017.07.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 07/14/2017] [Accepted: 07/19/2017] [Indexed: 01/24/2023] Open
Abstract
Propane-2-sulfonic acid octadec-9-enyl-amide (N15), an analogue of oleoylethanolamide (OEA), is a novel PPARα/γ dual agonist. Our previous studies verified the positive effects of OEA on the acute and delayed stages of cerebral ischemia. However, it is not clear whether N15 is effective against ischemic cerebral injury. In the present study, male Kunming mice were subjected to middle cerebral artery occlusion (MCAO). To evaluate its preventive effects, N15 (50, 100 or 200mg/kg, ip) was administered for 3days before ischemia. To evaluate its therapeutic effects, N15 (200mg/kg, ip) was administered 1h before reperfusion or 0, 1, 2 or 4h after reperfusion. Neurological deficit scores, infarct volume and the degree of brain oedema were determined at 24h after reperfusion. Blood brain barrier (BBB) disruption was evaluated by Evans blue (EB) and FITC-dextran leakages at 6h after reperfusion. The activation/inflammatory responses of microglia/macrophages were detected using immunohistochemistry and western blot. N15 pretreatment improved neurological dysfunction, reduced infarct volume and alleviated brain oedema in a dose-dependent manner; the most effective dose was 200mg/kg. The therapeutic time window was within 2h after reperfusion. N15 treatment preserved the BBB integrity and suppressed the activation of microglia/macrophages. N15 inhibited inflammatory cytokine expression not only in MCAO mice but also in lipopolysaccharide (LPS)-stimulated BV-2 microglial cells. Additionally, N15 markedly decreased the phosphorylation levels of NF-κBp65, STAT3, and ERK1/2 both in vivo and in vitro. Furthermore, the PPARα antagonist MK886 or PPARγ antagonist T0070907 respectively partly abolished the anti-inflammatory effects of N15 in vitro. Our findings demonstrated that N15 can exert neuroprotective effects against cerebral ischemic insult. Moreover, the neuroprotective effects of N15 on cerebral ischemia may be attributed to its anti-inflammatory properties, at least in part, by enhancing PPARα/γ dual signaling and inhibiting the activation of the NF-κB, STAT3, and ERK1/2 signaling pathways. These findings suggest that N15 may be a potential therapeutic choice for the prevention and treatment of ischemic stroke.
Collapse
Affiliation(s)
- Ying Li
- Xiamen Key Laboratory of Chiral Drugs, Medical College, Xiamen University, Xiamen, China; Department of Pharmacy, Xiamen Medical College, Xiamen, China
| | - Lanxi Xu
- Xiamen Key Laboratory of Chiral Drugs, Medical College, Xiamen University, Xiamen, China
| | - Kaiyue Zeng
- Xiamen Key Laboratory of Chiral Drugs, Medical College, Xiamen University, Xiamen, China
| | - Zhentian Xu
- Xiamen Key Laboratory of Chiral Drugs, Medical College, Xiamen University, Xiamen, China
| | - Daqin Suo
- Xiamen Key Laboratory of Chiral Drugs, Medical College, Xiamen University, Xiamen, China
| | - Lu Peng
- Xiamen Key Laboratory of Chiral Drugs, Medical College, Xiamen University, Xiamen, China
| | - Tong Ren
- Xiamen Key Laboratory of Chiral Drugs, Medical College, Xiamen University, Xiamen, China
| | - Zhiheng Sun
- Xiamen Key Laboratory of Chiral Drugs, Medical College, Xiamen University, Xiamen, China
| | - Wushuang Yang
- Department of Neurosurgery, Xiamen Hospital of Traditional Chinese Medicine, Xiamen, China
| | - Xin Jin
- Xiamen Key Laboratory of Chiral Drugs, Medical College, Xiamen University, Xiamen, China.
| | - Lichao Yang
- Xiamen Key Laboratory of Chiral Drugs, Medical College, Xiamen University, Xiamen, China.
| |
Collapse
|
33
|
Immunoregulatory effect of mast cells influenced by microbes in neurodegenerative diseases. Brain Behav Immun 2017; 65:68-89. [PMID: 28676349 DOI: 10.1016/j.bbi.2017.06.017] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 05/17/2017] [Accepted: 06/30/2017] [Indexed: 02/06/2023] Open
Abstract
When related to central nervous system (CNS) health and disease, brain mast cells (MCs) can be a source of either beneficial or deleterious signals acting on neural cells. We review the current state of knowledge about molecular interactions between MCs and glia in neurodegenerative diseases such as Multiple Sclerosis, Alzheimer's disease, Amyotrophic Lateral Sclerosis, Parkinson's disease, Epilepsy. We also discuss the influence on MC actions evoked by the host microbiota, which has a profound effect on the host immune system, inducing important consequences in neurodegenerative disorders. Gut dysbiosis, reduced intestinal motility and increased intestinal permeability, that allow bacterial products to circulate and pass through the blood-brain barrier, are associated with neurodegenerative disease. There are differences between the microbiota of neurologic patients and healthy controls. Distinguishing between cause and effect is a challenging task, and the molecular mechanisms whereby remote gut microbiota can alter the brain have not been fully elucidated. Nevertheless, modulation of the microbiota and MC activation have been shown to promote neuroprotection. We review this new information contributing to a greater understanding of MC-microbiota-neural cells interactions modulating the brain, behavior and neurodegenerative processes.
Collapse
|
34
|
Lee JY, Xu K, Nguyen H, Guedes VA, Borlongan CV, Acosta SA. Stem Cell-Induced Biobridges as Possible Tools to Aid Neuroreconstruction after CNS Injury. Front Cell Dev Biol 2017; 5:51. [PMID: 28540289 PMCID: PMC5424542 DOI: 10.3389/fcell.2017.00051] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 04/21/2017] [Indexed: 12/12/2022] Open
Abstract
Notch-induced mesenchymal stromal cells (MSCs) mediate a distinct mechanism of repair after brain injury by forming a biobridge that facilitates biodistribution of host cells from a neurogenic niche to the area of injury. We have observed the biobridge in an area between the subventricular zone and the injured cortex using immunohistochemistry and laser capture. Cells in the biobridge express high levels of extracellular matrix metalloproteinases (MMPs), specifically MMP-9, which co-localized with a trail of MSCs graft. The transplanted stem cells then become almost undetectable, being replaced by newly recruited host cells. This stem cell-paved biobridge provides support for distal migration of host cells from the subventricular zone to the site of injury. Biobridge formation by transplanted stem cells seems to have a fundamental role in initiating endogenous repair processes. Two major stem cell-mediated repair mechanisms have been proposed thus far: direct cell replacement by transplanted grafts and bystander effects through the secretion of trophic factors including fibroblast growth factor 2 (FGF-2), epidermal growth factor (EGF), stem cell factor (SCF), erythropoietin, and brain-derived neurotrophic factor (BDNF) among others. This groundbreaking observation of biobridge formation by transplanted stem cells represents a novel mechanism for stem cell mediated brain repair. Future studies on graft-host interaction will likely establish biobridge formation as a fundamental mechanism underlying therapeutic effects of stem cells and contribute to the scientific pursuit of developing safe and efficient therapies not only for traumatic brain injury but also for other neurological disorders. The aim of this review is to hypothetically extend concepts related to the formation of biobridges in other central nervous system disorders.
Collapse
Affiliation(s)
- Jea Y Lee
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida College of MedicineTampa, FL, USA
| | - Kaya Xu
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida College of MedicineTampa, FL, USA
| | - Hung Nguyen
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida College of MedicineTampa, FL, USA
| | - Vivian A Guedes
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida College of MedicineTampa, FL, USA
| | - Cesar V Borlongan
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida College of MedicineTampa, FL, USA
| | - Sandra A Acosta
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida College of MedicineTampa, FL, USA
| |
Collapse
|
35
|
Millar LJ, Shi L, Hoerder-Suabedissen A, Molnár Z. Neonatal Hypoxia Ischaemia: Mechanisms, Models, and Therapeutic Challenges. Front Cell Neurosci 2017; 11:78. [PMID: 28533743 PMCID: PMC5420571 DOI: 10.3389/fncel.2017.00078] [Citation(s) in RCA: 213] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 03/07/2017] [Indexed: 12/11/2022] Open
Abstract
Neonatal hypoxia-ischaemia (HI) is the most common cause of death and disability in human neonates, and is often associated with persistent motor, sensory, and cognitive impairment. Improved intensive care technology has increased survival without preventing neurological disorder, increasing morbidity throughout the adult population. Early preventative or neuroprotective interventions have the potential to rescue brain development in neonates, yet only one therapeutic intervention is currently licensed for use in developed countries. Recent investigations of the transient cortical layer known as subplate, especially regarding subplate's secretory role, opens up a novel set of potential molecular modulators of neonatal HI injury. This review examines the biological mechanisms of human neonatal HI, discusses evidence for the relevance of subplate-secreted molecules to this condition, and evaluates available animal models. Neuroserpin, a neuronally released neuroprotective factor, is discussed as a case study for developing new potential pharmacological interventions for use post-ischaemic injury.
Collapse
Affiliation(s)
- Lancelot J. Millar
- Molnár Group, Department of Physiology, Anatomy and Genetics, University of OxfordOxford, UK
| | - Lei Shi
- Molnár Group, Department of Physiology, Anatomy and Genetics, University of OxfordOxford, UK
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of Pharmacy, Jinan UniversityGuangzhou, China
| | | | - Zoltán Molnár
- Molnár Group, Department of Physiology, Anatomy and Genetics, University of OxfordOxford, UK
| |
Collapse
|
36
|
Kanazawa M, Takahashi T, Nishizawa M, Shimohata T. Therapeutic Strategies to Attenuate Hemorrhagic Transformation After Tissue Plasminogen Activator Treatment for Acute Ischemic Stroke. J Atheroscler Thromb 2017; 24:240-253. [PMID: 27980241 PMCID: PMC5383539 DOI: 10.5551/jat.rv16006] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 11/09/2016] [Indexed: 01/11/2023] Open
Abstract
This review focuses on the mechanisms and emerging concepts of stroke and therapeutic strategies for attenuating hemorrhagic transformation (HT) after tissue plasminogen activator (tPA) treatment for acute ischemic stroke (AIS). The therapeutic time window for tPA treatment has been extended. However, the patients who are eligible for tPA treatment are still <5% of all patients with AIS. The risk of serious or fatal symptomatic hemorrhage increases with delayed initiation of treatment. HT is thought to be caused by 1) ischemia/reperfusion injury; 2) the toxicity of tPA itself; 3) inflammation; and/or 4) remodeling factor-mediated effects. Modulation of these pathophysiologies is the basis of direct therapeutic strategies to attenuate HT after tPA treatment. Several studies have revealed that matrix metalloproteinases and free radicals are potential therapeutic targets. In addition, we have demonstrated that the inhibition of the vascular endothelial growth factor-signaling pathway and supplemental treatment with a recombinant angiopoietin-1 protein might be a promising therapeutic strategy for attenuating HT after tPA treatment through vascular protection. Moreover, single-target therapies could be insufficient for attenuating HT after tPA treatment and improving the therapeutic outcome of patients with AIS. We recently identified progranulin, which is a growth factor and a novel target molecule with multiple therapeutic effects. Progranulin might be a therapeutic target that protects the brain through suppression of vascular remodeling (vascular protection), neuroinflammation, and/or neuronal death (neuroprotection). Clinical trials which evaluate the effects of anti-VEGF drugs or PGRN-based treatment with tPA will be might worthwhile.
Collapse
Affiliation(s)
- Masato Kanazawa
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Tetsuya Takahashi
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Masatoyo Nishizawa
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Takayoshi Shimohata
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| |
Collapse
|
37
|
Hu X, De Silva TM, Chen J, Faraci FM. Cerebral Vascular Disease and Neurovascular Injury in Ischemic Stroke. Circ Res 2017; 120:449-471. [PMID: 28154097 PMCID: PMC5313039 DOI: 10.1161/circresaha.116.308427] [Citation(s) in RCA: 280] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 10/13/2016] [Accepted: 10/26/2016] [Indexed: 12/13/2022]
Abstract
The consequences of cerebrovascular disease are among the leading health issues worldwide. Large and small cerebral vessel disease can trigger stroke and contribute to the vascular component of other forms of neurological dysfunction and degeneration. Both forms of vascular disease are driven by diverse risk factors, with hypertension as the leading contributor. Despite the importance of neurovascular disease and subsequent injury after ischemic events, fundamental knowledge in these areas lag behind our current understanding of neuroprotection and vascular biology in general. The goal of this review is to address select key structural and functional changes in the vasculature that promote hypoperfusion and ischemia, while also affecting the extent of injury and effectiveness of therapy. In addition, as damage to the blood-brain barrier is one of the major consequences of ischemia, we discuss cellular and molecular mechanisms underlying ischemia-induced changes in blood-brain barrier integrity and function, including alterations in endothelial cells and the contribution of pericytes, immune cells, and matrix metalloproteinases. Identification of cell types, pathways, and molecules that control vascular changes before and after ischemia may result in novel approaches to slow the progression of cerebrovascular disease and lessen both the frequency and impact of ischemic events.
Collapse
Affiliation(s)
- Xiaoming Hu
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| | - T. Michael De Silva
- Biomedicine Discovery Institute, Department of Pharmacology, 9 Ancora Imparo Way, Monash University, Clayton, Vic, Australia
| | - Jun Chen
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| | - Frank M. Faraci
- Departments of Internal Medicine and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City Veterans Affairs Healthcare System, Iowa City, IA, USA
| |
Collapse
|
38
|
Matrix Metalloproteinase-9 and Recovery of Acute Ischemic Stroke. J Stroke Cerebrovasc Dis 2017; 26:733-740. [PMID: 28063771 DOI: 10.1016/j.jstrokecerebrovasdis.2016.09.043] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 08/17/2016] [Accepted: 09/24/2016] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Stroke outcome can be predicted by clinical features, biochemical parameters, and some risk factors. Matrix metalloproteinase-9 (MMP-9) is involved in various stages of stroke pathology. MMP-9 inhibitors are potential stroke therapeutic agents. Little is known about the relation between MMP-9-after the acute stage-and clinical recovery. OBJECTIVE The study aimed to investigate the serum level of MMP-9 at stroke onset as predictor of stroke outcome and the relation between the level of MMP-9 after 30 days and stroke recovery. METHODS The National Institutes of Health Stroke Scale, modified Rankin Scale, and serum level of MMP-9 were assessed in 30 patients with acute ischemic stroke during the first 24 hours of onset and then a month later. None of the patients received thrombolytic therapy. Thirty normal volunteers of matched age and sex were included in the control group. RESULTS The serum level of MMP-9 at stroke onset was independently positively correlated with stroke outcome. The serum level of MMP-9 30 days after stroke onset was positively correlated with initial stroke severity and outcome, as well as with clinical recovery. CONCLUSION Higher serum level of MMP-9 at stroke onset can be a predictor of poor stroke outcome. However, beyond the acute stage, MMP-9 may play beneficial role in stroke recovery.
Collapse
|
39
|
Gautam J, Zhang X, Yao Y. The role of pericytic laminin in blood brain barrier integrity maintenance. Sci Rep 2016; 6:36450. [PMID: 27808256 PMCID: PMC5093438 DOI: 10.1038/srep36450] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 10/14/2016] [Indexed: 11/09/2022] Open
Abstract
Laminin, a major component of the basement membrane, plays an important role in blood brain barrier regulation. At the neurovascular unit, brain endothelial cells, astrocytes, and pericytes synthesize and deposit different laminin isoforms into the basement membrane. It has been shown that laminin α4 (endothelial laminin) regulates vascular integrity at embryonic/neonatal stage, while astrocytic laminin maintains vascular integrity in adulthood. Here, we investigate the function of pericyte-derived laminin in vascular integrity. Using a conditional knockout mouse line, we report that loss of pericytic laminin leads to hydrocephalus and BBB breakdown in a small percentage (10.7%) of the mutants. Interestingly, BBB disruption always goes hand-in-hand with hydrocephalus in these mutants, and neither symptom is observed in the rest 89.3% of the mutants. Further mechanistic studies show that reduced tight junction proteins, diminished AQP4 expression, and decreased pericyte coverage are responsible for the BBB disruption. Together, these data suggest that pericyte-derived laminin is involved in the maintenance of BBB integrity and regulation of ventricular size/development.
Collapse
Affiliation(s)
- Jyoti Gautam
- College of Pharmacy, University of Minnesota, 1110 Kirby Drive, Duluth, MN, 55812, USA
| | - Xuanming Zhang
- College of Pharmacy, University of Minnesota, 1110 Kirby Drive, Duluth, MN, 55812, USA
| | - Yao Yao
- College of Pharmacy, University of Minnesota, 1110 Kirby Drive, Duluth, MN, 55812, USA
| |
Collapse
|
40
|
Schuldt G, Galanis C, Strehl A, Hick M, Schiener S, Lenz M, Deller T, Maggio N, Vlachos A. Inhibition of Protease-Activated Receptor 1 Does not Affect Dendritic Homeostasis of Cultured Mouse Dentate Granule Cells. Front Neuroanat 2016; 10:64. [PMID: 27378862 PMCID: PMC4904007 DOI: 10.3389/fnana.2016.00064] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 05/27/2016] [Indexed: 12/25/2022] Open
Abstract
Protease-activated receptors (PARs) are widely expressed in the central nervous system (CNS). While a firm link between PAR1-activation and functional synaptic and intrinsic neuronal properties exists, studies on the role of PAR1 in neural structural plasticity are scarce. The physiological function of PAR1 in the brain remains not well understood. We here sought to determine whether prolonged pharmacologic PAR1-inhibition affects dendritic morphologies of hippocampal neurons. To address this question we employed live-cell microscopy of mouse dentate granule cell dendrites in 3-week old entorhino-hippocampal slice cultures prepared from Thy1-GFP mice. A subset of cultures were treated with the PAR1-inhibitor SCH79797 (1 μM; up to 3 weeks). No major effects of PAR1-inhibition on static and dynamic parameters of dentate granule cell dendrites were detected under control conditions. Granule cells of PAR1-deficient slice cultures showed unaltered dendritic morphologies, dendritic spine densities and excitatory synaptic strength. Furthermore, we report that PAR1-inhibition does not prevent dendritic retraction following partial deafferentation in vitro. Consistent with this finding, no major changes in PAR1-mRNA levels were detected in the denervated dentate gyrus (DG). We conclude that neural PAR1 is not involved in regulating the steady-state dynamics or deafferentation-induced adaptive changes of cultured dentate granule cell dendrites. These results indicate that drugs targeting neural PAR1-signals may not affect the stability and structural integrity of neuronal networks in healthy brain regions.
Collapse
Affiliation(s)
- Gerlind Schuldt
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University Frankfurt Frankfurt, Germany
| | - Christos Galanis
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University Frankfurt Frankfurt, Germany
| | - Andreas Strehl
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University Frankfurt Frankfurt, Germany
| | - Meike Hick
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University Frankfurt Frankfurt, Germany
| | - Sabine Schiener
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University Frankfurt Frankfurt, Germany
| | - Maximilian Lenz
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University FrankfurtFrankfurt, Germany; Institute of Anatomy II, Faculty of Medicine, Heinrich-Heine-University DüsseldorfDüsseldorf, Germany
| | - Thomas Deller
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University Frankfurt Frankfurt, Germany
| | - Nicola Maggio
- Department of Neurology, The Sagol Center for Neurosciences, Sheba Medical Center, Affiliated to the Sackler Faculty of Medicine, Tel Aviv UniversityTel Aviv, Israel; Talpiot Medical Leadership Program, Department of Neurology and J. Sagol Neuroscience Center, The Chaim Sheba Medical CenterTel HaShomer, Israel; Sagol School of Neuroscience, Tel Aviv UniversityTel Aviv, Israel
| | - Andreas Vlachos
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University FrankfurtFrankfurt, Germany; Institute of Anatomy II, Faculty of Medicine, Heinrich-Heine-University DüsseldorfDüsseldorf, Germany
| |
Collapse
|
41
|
Abstract
Stroke not only causes initial cell death, but also a limited process of repair and recovery. As an overall biological process, stroke has been most often considered from the perspective of early phases of ischemia, how these inter-relate and lead to expansion of the infarct. However, just as the biology of later stages of stroke becomes better understood, the clinical realities of stroke indicate that it is now more a chronic disease than an acute killer. As an overall biological process, it is now more important to understand how early cell death leads to the later, limited recovery so as develop an integrative view of acute to chronic stroke. This progression from death to repair involves sequential stages of primary cell death, secondary injury events, reactive tissue progenitor responses, and formation of new neuronal circuits. This progression is radial: from the tissue that suffers the infarct secondary injury signals, including free radicals and inflammatory cytokines, radiate out from the stroke core to trigger later regenerative events. Injury and repair processes occur not just in the local stroke site, but are also triggered in the connected networks of neurons that had existed in the stroke center: damage signals are relayed throughout a brain network. From these relayed, distributed damage signals, reactive astrocytosis, inflammatory processes, and the formation of new connections occur in distant brain areas. In short, emerging data in stroke cell death studies and the development of the field of stroke neural repair now indicate a continuum in time and in space of progressive events that can be considered as the 3 Rs of stroke biology: radial, relayed, and regenerative.
Collapse
Affiliation(s)
- S Thomas Carmichael
- Departments of Neurology and Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
| |
Collapse
|
42
|
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: 313] [Impact Index Per Article: 34.8] [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
|
43
|
Oxidative DNA Damage Mediated by Intranuclear MMP Activity Is Associated with Neuronal Apoptosis in Ischemic Stroke. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:6927328. [PMID: 26925194 PMCID: PMC4748094 DOI: 10.1155/2016/6927328] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 12/26/2015] [Accepted: 12/31/2015] [Indexed: 11/18/2022]
Abstract
Evidence of the pathological roles of matrix metalloproteinases (MMPs) in various neurological disorders has made them attractive therapeutic targets. MMPs disrupt the blood-brain barrier and cause neuronal death and neuroinflammation in acute cerebral ischemia and are critical for angiogenesis during recovery. However, some challenges have to be overcome before MMPs can be further validated as drug targets in stroke injury. Identifying in vivo substrates of MMPs should greatly improve our understanding of the mechanisms of ischemic injury and is critical for providing more precise drug targets. Recent works have uncovered nontraditional roles for MMPs in the cytosol and nucleus. These have shed light on intracellular targets and biological actions of MMPs, adding additional layers of complexity for therapeutic MMP inhibition. In this review, we discussed the recent advances made in understanding nuclear location of MMPs, their regulation of intranuclear sorting, and their intranuclear proteolytic activity and substrates. In particular, we highlighted the roles of intranuclear MMPs in oxidative DNA damage, neuronal apoptosis, and neuroinflammation at an early stage of stroke insult. These novel data point to new putative MMP-mediated intranuclear actions in stroke-induced pathological processes and may lead to novel approaches to treatment of stroke and other neurological diseases.
Collapse
|
44
|
Marrow-Derived Mesenchymal Stromal Cells in the Treatment of Stroke. Transl Neurosci 2016. [DOI: 10.1007/978-1-4899-7654-3_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
45
|
|
46
|
Chelluboina B, Klopfenstein JD, Pinson DM, Wang DZ, Vemuganti R, Veeravalli KK. Matrix Metalloproteinase-12 Induces Blood–Brain Barrier Damage After Focal Cerebral Ischemia. Stroke 2015; 46:3523-31. [DOI: 10.1161/strokeaha.115.011031] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 10/07/2015] [Indexed: 12/12/2022]
Abstract
Background and Purpose—
Matrix metalloproteinases (MMPs) have a central role in compromising the integrity of the blood–brain barrier (BBB). The role of MMP-12 in brain damage after ischemic stroke remains unknown. The main objective of the current study is to investigate the effect of MMP-12 suppression at an early time point before reperfusion on the BBB damage in rats.
Methods—
Sprague–Dawley rats were subjected to middle cerebral artery occlusion and reperfusion. MMP-12 shRNA–expressing plasmids formulated as nanoparticles were administered at a dose of 1 mg/kg body weight. The involvement of MMP-12 on BBB damage was assessed by performing various techniques, including Evans blue dye extravasation, 2,3,5-triphenyltetrazolium chloride staining, immunoblot, gelatin zymography, and immunofluorescence analysis.
Results—
MMP-12 is upregulated ≈31-, 47-, and 66-fold in rats subjected 1–, 2-, or 4-hour ischemia, respectively, followed by 1-day reperfusion. MMP-12 suppression protected the BBB integrity by inhibiting the degradation of tight-junction proteins. Either intravenous or intra-arterial delivery of MMP-12 shRNA-expressing plasmid significantly reduced the percent Evans blue dye extravasation and infarct size. Furthermore, MMP-12 suppression reduced the endogenous levels of other proteases, such as tissue-type plasminogen activator and MMP-9, which are also known to be the key players involved in BBB damage.
Conclusions—
These results demonstrate the adverse role of MMP-12 in acute brain damage that occurs after ischemic stroke and, thereby, suggesting that MMP-12 suppression could be a promising therapeutic target for cerebral ischemia.
Collapse
Affiliation(s)
- Bharath Chelluboina
- From the Departments of Cancer Biology and Pharmacology (B.C., K.K.V.), Neurosurgery (J.D.K.), Pathology (D.M.P.), and Neurology (D.Z.W.), University of Illinois College of Medicine at Peoria; Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin, Madison (R.V.); and Comprehensive Stroke Center, Illinois Neurological Institute, Peoria (J.D.K., D.Z.W.)
| | - Jeffrey D. Klopfenstein
- From the Departments of Cancer Biology and Pharmacology (B.C., K.K.V.), Neurosurgery (J.D.K.), Pathology (D.M.P.), and Neurology (D.Z.W.), University of Illinois College of Medicine at Peoria; Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin, Madison (R.V.); and Comprehensive Stroke Center, Illinois Neurological Institute, Peoria (J.D.K., D.Z.W.)
| | - David M. Pinson
- From the Departments of Cancer Biology and Pharmacology (B.C., K.K.V.), Neurosurgery (J.D.K.), Pathology (D.M.P.), and Neurology (D.Z.W.), University of Illinois College of Medicine at Peoria; Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin, Madison (R.V.); and Comprehensive Stroke Center, Illinois Neurological Institute, Peoria (J.D.K., D.Z.W.)
| | - David Z. Wang
- From the Departments of Cancer Biology and Pharmacology (B.C., K.K.V.), Neurosurgery (J.D.K.), Pathology (D.M.P.), and Neurology (D.Z.W.), University of Illinois College of Medicine at Peoria; Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin, Madison (R.V.); and Comprehensive Stroke Center, Illinois Neurological Institute, Peoria (J.D.K., D.Z.W.)
| | - Raghu Vemuganti
- From the Departments of Cancer Biology and Pharmacology (B.C., K.K.V.), Neurosurgery (J.D.K.), Pathology (D.M.P.), and Neurology (D.Z.W.), University of Illinois College of Medicine at Peoria; Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin, Madison (R.V.); and Comprehensive Stroke Center, Illinois Neurological Institute, Peoria (J.D.K., D.Z.W.)
| | - Krishna Kumar Veeravalli
- From the Departments of Cancer Biology and Pharmacology (B.C., K.K.V.), Neurosurgery (J.D.K.), Pathology (D.M.P.), and Neurology (D.Z.W.), University of Illinois College of Medicine at Peoria; Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin, Madison (R.V.); and Comprehensive Stroke Center, Illinois Neurological Institute, Peoria (J.D.K., D.Z.W.)
| |
Collapse
|
47
|
Teng Z, Jiang L, Hu Q, He Y, Guo Z, Wu Y, Huang Z, Cao F, Cheng C, Sun X, Guo Z. Peroxisome Proliferator-Activated Receptor β/δ Alleviates Early Brain Injury After Subarachnoid Hemorrhage in Rats. Stroke 2015; 47:196-205. [PMID: 26628385 DOI: 10.1161/strokeaha.115.011701] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 10/19/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND PURPOSE Early brain injury is proposed to be the primary cause of the poor outcome after subarachnoid hemorrhage (SAH), which is closely related to the neural apoptosis. To date, the relationship between peroxisome proliferator-activated receptor β/δ (PPARβ/δ) and nuclear factor-κB/matrix metalloproteinase-9 (NF-κB/MMP-9) pathway, both of which are closely related to apoptotic effects, has been poorly studied in SAH. The present study was undertaken to evaluate the effects of PPARβ/δ on early brain injury and NF-κB/MMP-9 pathway after SAH in rats. METHODS SAH model was established by injecting nonheparinized autologous arterial blood into the prechiasmatic cistern in male Sprague-Dawley rats. Adenoviruses or small interfering RNAs were injected into the right lateral cerebral ventricle to, respectively, up- or downregulate PPARβ/δ expression before SAH. All animals were assessed with a neurological score and then killed at 24 hours after SAH surgery. The indexes of brain water content, blood-brain barrier permeability, and apoptosis were used to detect brain injury. The expression of PPARβ/δ, NF-κB, and MMP-9 were measured by immunohistochemistry, gelatin zymography, and Western Blot methods, respectively. In addition, GW0742, a specific agonist of PPARβ/δ, was used to treat SAH in rats, the effects of which were evaluated by neurological scoring and Evans blue extravasation. RESULTS Overexpression of PPARβ/δ by adenoviruses treatment significantly ameliorated brain injury with improvement in neurological deficits, brain edema, blood-brain barrier impairment, and neural cell apoptosis at 24 hours after SAH in rats, whereas downregulation of PPARβ/δ by small interfering RNAs administration resulted in the reverse effects of the above. The expression levels of NF-κB and MMP-9 were markedly downregulated when PPARβ/δ increased after PPARβ/δ adenovirus transfection and upregulated when PPARβ/δ decreased by PPARβ/δ small interfering RNAs treatment. Moreover, GW0742 improved neurological deficits and reduced Evans blue extravasation at 24 hours after SAH. CONCLUSIONS PPARβ/δ's overexpression may attenuate early brain injury after rats' SAH administration, which reduces neural apoptosis possibly through blocking NF-κB/MMP-9 pathway.
Collapse
Affiliation(s)
- Zhipeng Teng
- From the Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Z.T., L.J., Y.W., Z.H., F.C., C.C., X.S., Zongduo Guo); Discipline of Neuroscience and Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Q.H.); Department of Neurosurgery, Tong-ji Hospital, Wuhan, China (Y.H.); and Department of Neurology, the First Hospital of Jilin University, Changchun, China (Zhenni Guo)
| | - Li Jiang
- From the Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Z.T., L.J., Y.W., Z.H., F.C., C.C., X.S., Zongduo Guo); Discipline of Neuroscience and Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Q.H.); Department of Neurosurgery, Tong-ji Hospital, Wuhan, China (Y.H.); and Department of Neurology, the First Hospital of Jilin University, Changchun, China (Zhenni Guo)
| | - Qin Hu
- From the Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Z.T., L.J., Y.W., Z.H., F.C., C.C., X.S., Zongduo Guo); Discipline of Neuroscience and Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Q.H.); Department of Neurosurgery, Tong-ji Hospital, Wuhan, China (Y.H.); and Department of Neurology, the First Hospital of Jilin University, Changchun, China (Zhenni Guo)
| | - Yue He
- From the Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Z.T., L.J., Y.W., Z.H., F.C., C.C., X.S., Zongduo Guo); Discipline of Neuroscience and Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Q.H.); Department of Neurosurgery, Tong-ji Hospital, Wuhan, China (Y.H.); and Department of Neurology, the First Hospital of Jilin University, Changchun, China (Zhenni Guo)
| | - Zhenni Guo
- From the Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Z.T., L.J., Y.W., Z.H., F.C., C.C., X.S., Zongduo Guo); Discipline of Neuroscience and Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Q.H.); Department of Neurosurgery, Tong-ji Hospital, Wuhan, China (Y.H.); and Department of Neurology, the First Hospital of Jilin University, Changchun, China (Zhenni Guo)
| | - Yue Wu
- From the Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Z.T., L.J., Y.W., Z.H., F.C., C.C., X.S., Zongduo Guo); Discipline of Neuroscience and Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Q.H.); Department of Neurosurgery, Tong-ji Hospital, Wuhan, China (Y.H.); and Department of Neurology, the First Hospital of Jilin University, Changchun, China (Zhenni Guo)
| | - Zhijian Huang
- From the Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Z.T., L.J., Y.W., Z.H., F.C., C.C., X.S., Zongduo Guo); Discipline of Neuroscience and Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Q.H.); Department of Neurosurgery, Tong-ji Hospital, Wuhan, China (Y.H.); and Department of Neurology, the First Hospital of Jilin University, Changchun, China (Zhenni Guo)
| | - Fang Cao
- From the Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Z.T., L.J., Y.W., Z.H., F.C., C.C., X.S., Zongduo Guo); Discipline of Neuroscience and Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Q.H.); Department of Neurosurgery, Tong-ji Hospital, Wuhan, China (Y.H.); and Department of Neurology, the First Hospital of Jilin University, Changchun, China (Zhenni Guo)
| | - Chongjie Cheng
- From the Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Z.T., L.J., Y.W., Z.H., F.C., C.C., X.S., Zongduo Guo); Discipline of Neuroscience and Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Q.H.); Department of Neurosurgery, Tong-ji Hospital, Wuhan, China (Y.H.); and Department of Neurology, the First Hospital of Jilin University, Changchun, China (Zhenni Guo)
| | - Xiaochuan Sun
- From the Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Z.T., L.J., Y.W., Z.H., F.C., C.C., X.S., Zongduo Guo); Discipline of Neuroscience and Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Q.H.); Department of Neurosurgery, Tong-ji Hospital, Wuhan, China (Y.H.); and Department of Neurology, the First Hospital of Jilin University, Changchun, China (Zhenni Guo).
| | - Zongduo Guo
- From the Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China (Z.T., L.J., Y.W., Z.H., F.C., C.C., X.S., Zongduo Guo); Discipline of Neuroscience and Department of Anatomy, Histology and Embryology, Collaborative Innovation Center for Brain Science, and Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Q.H.); Department of Neurosurgery, Tong-ji Hospital, Wuhan, China (Y.H.); and Department of Neurology, the First Hospital of Jilin University, Changchun, China (Zhenni Guo).
| |
Collapse
|
48
|
Upregulation of EMMPRIN (OX47) in Rat Dorsal Root Ganglion Contributes to the Development of Mechanical Allodynia after Nerve Injury. Neural Plast 2015; 2015:249756. [PMID: 26697232 PMCID: PMC4677233 DOI: 10.1155/2015/249756] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 06/24/2015] [Accepted: 06/28/2015] [Indexed: 12/14/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are widely implicated in inflammation and tissue remodeling associated with various neurodegenerative diseases and play an important role in nociception and allodynia. Extracellular Matrix Metalloproteinase Inducer (EMMPRIN) plays a key regulatory role for MMP activities. However, the role of EMMPRIN in the development of neuropathic pain is not clear. Western blotting, real-time quantitative RT-PCR (qRT-PCR), and immunofluorescence were performed to determine the changes of messenger RNA and protein of EMMPRIN/OX47 and their cellular localization in the rat dorsal root ganglion (DRG) after nerve injury. Paw withdrawal threshold test was examined to evaluate the pain behavior in spinal nerve ligation (SNL) model. The lentivirus containing OX47 shRNA was injected into the DRG one day before SNL. The expression level of both mRNA and protein of OX47 was markedly upregulated in ipsilateral DRG after SNL. OX47 was mainly expressed in the extracellular matrix of DRG. Administration of shRNA targeted against OX47 in vivo remarkably attenuated mechanical allodynia induced by SNL. In conclusion, peripheral nerve injury induced upregulation of OX47 in the extracellular matrix of DRG. RNA interference against OX47 significantly suppressed the expression of OX47 mRNA and the development of mechanical allodynia. The altered expression of OX47 may contribute to the development of neuropathic pain after nerve injury.
Collapse
|
49
|
Zhang S, Dong H, Zhang X, Li N, Sun J, Qian Y. Cerebral mast cells contribute to postoperative cognitive dysfunction by promoting blood brain barrier disruption. Behav Brain Res 2015; 298:158-66. [PMID: 26554724 DOI: 10.1016/j.bbr.2015.11.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Revised: 11/02/2015] [Accepted: 11/04/2015] [Indexed: 12/18/2022]
Abstract
Trauma induced neuroinflammation plays a key role in the development of postoperative cognitive dysfunction (POCD). The blood-brain barrier (BBB), a highly specialized endothelial layer, is exquisitely sensitive to inflammatory insults, which can result in numerous neurocognitive syndromes. While brain mast cells are the "first responder" in the injury, the functional interactions between mast cells and the BBB remain poorly understood. Our results demonstrate that tibial fracture surgery can induce cognitive impairment relating to an inflammatory response and destabilization of the BBB. Disodium cromoglycate (cromolyn)--which acts as a mast cell stabilizer--inhibited this effect. Specifically, cromolyn resulted in ameliorated cognitive ability, decrease of inflammatory cytokines and increase of BBB stability. Taken together, these results suggest that activated mast cells contributed to central nervous system inflammation and cognitive dysfunction by promoting BBB disruption, and interactions between mast cells and the BBB could constitute a new and unique therapeutic target for POCD.
Collapse
Affiliation(s)
- Susu Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| | - Hongquan Dong
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| | - Xiang Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| | - Nana Li
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| | - Jie Sun
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China.
| | - Yanning Qian
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China.
| |
Collapse
|
50
|
Zhang XS, Zhang X, Zhang QR, Wu Q, Li W, Jiang TW, Hang CH. Astaxanthin reduces matrix metalloproteinase-9 expression and activity in the brain after experimental subarachnoid hemorrhage in rats. Brain Res 2015; 1624:113-124. [PMID: 26210617 DOI: 10.1016/j.brainres.2015.07.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Revised: 06/25/2015] [Accepted: 07/03/2015] [Indexed: 11/30/2022]
Abstract
We have previously shown that astaxanthin (ATX) reduces the blood-brain barrier (BBB) disruption and neurovascular dysfunction following subarachnoid hemorrhage (SAH) insults. However, the underlying mechanisms remain unclear. It is known that the matrix metalloproteinases (MMPs), especially matrix metalloproteinase-9 (MMP-9) plays a crucial role in the pathogenesis of secondary brain injury after SAH. And ATX has the ability to regulate MMP-9 in other models. Herein, we investigated whether ATX could ameliorate MMP-9 activation and expression in a rat model of SAH. A total of 144 rats were randomly divided into the following groups: control group (n=36), SAH group (n=36), SAH+vehicle group (n=36), and SAH+ATX group (n=36). The SAH model was induced by injection of 0.3 ml autologous blood into the prechiasmatic cistern. ATX (20 μl of 0.1 mmol) or vehicle was administered intracerebroventricularly 30 min after SAH induction. Mortality, neurological function, brain edema and blood-brain barrier (BBB) permeability were measured at 24 and 72 h after SAH. Biochemical and zymographic methods were used to analyze MMP-9 expression and activity in brain samples. Immunohistochemistry and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) staining were also evaluated at 24h. Our data indicated that ATX could significantly reduce the expression and activity of MMP-9, leading to the amelioration of brain edema, BBB impairment, neurological deficits and TUNEL-positive cells at 24h but not 72 h after SAH. The ATX-mediated down-regulation of MMP-9 was correlated with the decreased levels of IL-1β, TNF-α, oxidative stress, activated microglia and infiltrating neutrophils. These results suggest that the neurovascular protection of ATX in SAH is partly associated with the inhibition of MMP-9 expression and activity.
Collapse
Affiliation(s)
- Xiang-Sheng Zhang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Xin Zhang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China.
| | - Qing-Rong Zhang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Qi Wu
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Wei Li
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Tian-Wei Jiang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Chun-Hua Hang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China.
| |
Collapse
|