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
|
Ganguly K, Adhikary K, Acharjee A, Acharjee P, Trigun SK, Mutlaq AS, Ashique S, Yasmin S, Alshahrani AM, Ansari MY. Biological significance and pathophysiological role of Matrix Metalloproteinases in the Central Nervous System. Int J Biol Macromol 2024; 280:135967. [PMID: 39322129 DOI: 10.1016/j.ijbiomac.2024.135967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 09/21/2024] [Accepted: 09/21/2024] [Indexed: 09/27/2024]
Abstract
Matrix Metalloproteinases (MMPs), which are endopeptidase reliant on zinc, are low in embryonic tissues but increases in response to a variety of physiological stimulus and pathological stresses. Neuro-glial cells, endothelial cells, fibroblasts, and leucocytes secrete MMPs, which cleave extracellular matrix proteins in a time-dependent manner. MMPs affect synaptic plasticity and the development of short-term memory by controlling the size, shape, and excitatory synapses' function through the lateral diffusion of receptors. In addition, MMPs influence the Extracellular Matrix proteins in the Peri-Neuronal Net at the Neuro-glial interface, which aids in the establishment of long-term memory. Through modulating neuronal, and glial cells migration, differentiation, Neurogenesis, and survival, MMPs impact brain development in mammals. In adult brains, MMPs play a beneficial role in physiological plasticity, which includes learning, memory consolidation, social interaction, and complex behaviors, by proteolytically altering a wide variety of factors, including growth factors, cytokines, receptors, DNA repair enzymes, and matrix proteins. Additionally, stress, depression, addiction, hepatic encephalopathy, and stroke may all have negative effects on MMPs. In addition to their role in glioblastoma development, MMPs influence neurological diseases such as epilepsy, schizophrenia, autism spectrum disorder, brain damage, pain, neurodegeneration, and Alzheimer's and Parkinson's. To help shed light on the potential of MMPs as a therapeutic target for neurodegenerative diseases, this review summarizes their regulation, mode of action, and participation in brain physiological plasticity and pathological damage. Finally, by employing different MMP-based nanotools and inhibitors, MMPs may also be utilized to map the anatomical and functional connectome of the brain, analyze its secretome, and treat neurodegenerative illnesses.
Collapse
Affiliation(s)
- Krishnendu Ganguly
- Department of Medical Lab Technology, Paramedical College Durgapur, Helen Keller Sarani, Durgapur 713212, West Bengal, India.
| | - Krishnendu Adhikary
- Department of Medical Lab Technology, Paramedical College Durgapur, Helen Keller Sarani, Durgapur 713212, West Bengal, India.
| | - Arup Acharjee
- Molecular Omics Laboratory, Department of Zoology, University of Allahabad, Allahabad, Uttar Pradesh, India.
| | - Papia Acharjee
- Biochemistry Section, Department of Zoology, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India.
| | - Surendra Kumar Trigun
- Biochemistry Section, Department of Zoology, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India.
| | | | - Sumel Ashique
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Department of Pharmaceutics, Bengal College of Pharmaceutical Sciences & Research, Durgapur 713212, West Bengal, India.
| | - Sabina Yasmin
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia.
| | - Asma M Alshahrani
- Department of Clinical Pharmacy, Faculty of Pharmacy, King Khalid University, Abha, Saudi Arabia; Department of Clinical Pharmacy, Shaqra University, Saudi Arabia.
| | - Mohammad Yousuf Ansari
- MM college of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana 133207, India.
| |
Collapse
|
3
|
Li X, Wang X, Yang Y, Zhou J, Wu X, Zhao J, Zhang J, Guo X, Shao M, Song M, Su X, Han Y, Liu Q, Chen T, Zhang L, Liu B, Yue W, Lv L, Li W. Elevated plasma matrix metalloproteinase 9 in schizophrenia patients associated with poor antipsychotic treatment response and white matter density deficits. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2024; 10:71. [PMID: 39191778 DOI: 10.1038/s41537-024-00494-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 08/09/2024] [Indexed: 08/29/2024]
Abstract
Oxidative stress and neuroinflammation contribute to schizophrenia (SCZ) pathology and may influence treatment efficacy. Matrix metalloproteinase 9 (MMP9) is a critical molecular node mediating the interaction between oxidative stress and inflammation, and so may influence treatment efficacy. Here we examined the associations of plasma MMP9 concentration with antipsychotic drug responses, clinical symptoms, and brain structure. A total of 129 healthy controls and 124 patients with SCZ were included in this study. Patients were monitored clinically during 8 weeks of antipsychotic treatment and classified as poor responders (n = 49) or good responders (n = 75). We then compared plasma MMP9 concentrations in healthy controls at baseline and both SCZ responder groups at baseline and after the 8-week antipsychotic treatment regimen. Cognitive function was also examined using the MATRICS Consensus Cognitive Battery. In addition, we extracted regional white matter density from magnetic resonance images of patients. Compared to healthy controls, plasma MMP9 levels were significantly elevated in poor responders at baseline and negatively correlated with both white matter density in the right superior temporal gyrus and the change in cognitive symptoms after treatment. Conversely, there was no significant difference in plasma MMP9 between good responders and healthy controls, and no associations of plasma MMP9 with cognitive symptoms or regional white matter density among good responders. Elevated plasma MMP9 is associated with poor antipsychotic drug efficacy and white matter deficits in SCZ patients, and so may be a useful biomarker to guide personalized treatment.
Collapse
Affiliation(s)
- Xiaojing Li
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang, 453002, China
| | - Xiujuan Wang
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
| | - Yongfeng Yang
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
| | - Jiahui Zhou
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
| | - Xufei Wu
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, 453002, China
| | - Jingyuan Zhao
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
| | - Jianhong Zhang
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
| | - Xiaoge Guo
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
| | - Minglong Shao
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
| | - Meng Song
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
| | - Xi Su
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
| | - Yong Han
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
| | - Qing Liu
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
| | - Tengfei Chen
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
| | - Luwen Zhang
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
| | - Bing Liu
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, 100875, Beijing, China
- Chinese Institute for Brain Research, 102206, Beijing, China
| | - Weihua Yue
- Institute of Mental Health, Peking University, 100191, Beijing, China
- Key Laboratory for Mental Health, Ministry of Health, 100191, Beijing, China
| | - Luxian Lv
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China.
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, 453002, China.
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China.
| | - Wenqiang Li
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China.
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, 453002, China.
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China.
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang, 453002, China.
| |
Collapse
|
4
|
Owjfard M, Rahimian Z, Ghaderpanah R, Rafiei E, Sadrian S, Sabaghan M, Karimi F. Therapeutic Effects of Intranasal Administration of Resveratrol on the Rat Model of Brain Ischemia. Heliyon 2024; 10:e32592. [PMID: 38952360 PMCID: PMC11215267 DOI: 10.1016/j.heliyon.2024.e32592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/16/2024] [Accepted: 06/05/2024] [Indexed: 07/03/2024] Open
Abstract
Background Resveratrol is a natural phenolic compound widely found in plants. Previous studies have suggested its neuroprotective role in cerebral ischemia due to its anti-oxidative, anti-inflammatory, and anti-apoptotic effects. Intranasal administration of resveratrol enhances its capacity to penetrate the blood-brain barrier, increasing therapeutic efficacy and safety. Objective We aimed to examine the therapeutic potential of intranasal administration of resveratrol treatment in rats exposed to cerebral ischemia. Methods Sixty-four male rats were divided into three groups: the sham group, which was exposed to only surgical stress; the vehicle and resveratrol groups, which received intranasal vehicle or 50 mg/kg resveratrol for 7 days following middle cerebral artery occlusion, respectively. We assessed the modified neurologic severity scores, wire hanging tests, blood-brain barrier disruption, brain water content, and infarct volume. Levels of matrix metalloproteinase-9, nuclear factor-kappa B, B-cell lymphoma protein 2, and B-cell lymphoma protein 2-associated X messenger RNA expression were examined. Results At 3- and 7-days post-ischemia, rats receiving intranasal resveratrol had lower modified neurological severity scores and a smaller brain infarct volume than the rats receiving vehicle. Additionally, the intranasal resveratrol-treated rats showed significantly prolonged wire-hanging performance at the 7-day mark post-ischemia compared to the vehicle group. The blood-brain barrier disruption and brain water content were significantly lower in the resveratrol group than in the vehicle group. Furthermore, the resveratrol-treated group displayed lower expression of Matrix Metalloproteinase-9 and Nuclear Factor-Kappa B in contrast to the vehicle group, while the difference in expression levels of B-cell lymphoma protein 2-associated X and B-cell lymphoma protein 2 were not significant. Conclusion Intranasal administration of resveratrol showed neuroprotective effects on ischemic stroke by improving neurobehavioral function, reducing blood-brain barrier disruption, cerebral edema, and infarct volume. This treatment also downregulated Matrix Metalloproteinase-9 and Nuclear Factor-Kappa B expression, indicating its potential as a therapeutic option for ischemic stroke.
Collapse
Affiliation(s)
- Maryam Owjfard
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Shiraz University of Applied Science and Technology (UAST), Shiraz, Iran
| | - Zahra Rahimian
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Rezvan Ghaderpanah
- Students Research Committee, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Elahe Rafiei
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyedhassan Sadrian
- Students Research Committee, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | |
Collapse
|
5
|
Dhapola R, Medhi B, HariKrishnaReddy D. Insight into the pathophysiological advances and molecular mechanisms underlying cerebral stroke: current status. Mol Biol Rep 2024; 51:649. [PMID: 38733445 DOI: 10.1007/s11033-024-09597-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 04/30/2024] [Indexed: 05/13/2024]
Abstract
Molecular pathways involved in cerebral stroke are diverse. The major pathophysiological events that are observed in stroke comprises of excitotoxicity, oxidative stress, mitochondrial damage, endoplasmic reticulum stress, cellular acidosis, blood-brain barrier disruption, neuronal swelling and neuronal network mutilation. Various biomolecules are involved in these pathways and several major proteins are upregulated and/or suppressed following stroke. Different types of receptors, ion channels and transporters are activated. Fluctuations in levels of various ions and neurotransmitters have been observed. Cells involved in immune responses and various mediators involved in neuro-inflammation get upregulated progressing the pathogenesis of the disease. Despite of enormity of the problem, there is not a single therapy that can limit infarction and neurological disability due to stroke. This is because of poor understanding of the complex interplay between these pathophysiological processes. This review focuses upon the past to present research on pathophysiological events that are involved in stroke and various factors that are leading to neuronal death following cerebral stroke. This will pave a way to researchers for developing new potent therapeutics that can aid in the treatment of cerebral stroke.
Collapse
Affiliation(s)
- Rishika Dhapola
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India
| | - Bikash Medhi
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, Punjab, 160012, India
| | - Dibbanti HariKrishnaReddy
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India.
| |
Collapse
|
6
|
López-Sánchez C, Lagoa R, Poejo J, García-López V, García-Martínez V, Gutierrez-Merino C. An Update of Kaempferol Protection against Brain Damage Induced by Ischemia-Reperfusion and by 3-Nitropropionic Acid. Molecules 2024; 29:776. [PMID: 38398528 PMCID: PMC10893315 DOI: 10.3390/molecules29040776] [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: 12/19/2023] [Revised: 02/01/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
Kaempferol, a flavonoid present in many food products, has chemical and cellular antioxidant properties that are beneficial for protection against the oxidative stress caused by reactive oxygen and nitrogen species. Kaempferol administration to model experimental animals can provide extensive protection against brain damage of the striatum and proximal cortical areas induced by transient brain cerebral ischemic stroke and by 3-nitropropionic acid. This article is an updated review of the molecular and cellular mechanisms of protection by kaempferol administration against brain damage induced by these insults, integrated with an overview of the contributions of the work performed in our laboratories during the past years. Kaempferol administration at doses that prevent neurological dysfunctions inhibit the critical molecular events that underlie the initial and delayed brain damage induced by ischemic stroke and by 3-nitropropionic acid. It is highlighted that the protection afforded by kaempferol against the initial mitochondrial dysfunction can largely account for its protection against the reported delayed spreading of brain damage, which can develop from many hours to several days. This allows us to conclude that kaempferol administration can be beneficial not only in preventive treatments, but also in post-insult therapeutic treatments.
Collapse
Affiliation(s)
- Carmen López-Sánchez
- Institute of Molecular Pathology Biomarkers, University of Extremadura, 06006 Badajoz, Spain; (J.P.); (V.G.-L.); (V.G.-M.)
- Department of Human Anatomy and Embryology, Faculty of Medicine and Health Sciences, University of Extremadura, 06006 Badajoz, Spain
| | - Ricardo Lagoa
- School of Technology and Management, Polytechnic Institute of Leiria, Morro do Lena-Alto do Vieiro, 2411-901 Leiria, Portugal;
- Laboratory of Separation and Reaction Engineering-Laboratory of Catalysis and Materials (LSRE-LCM), Polytechnic Institute of Leiria, 2411-901 Leiria, Portugal
| | - Joana Poejo
- Institute of Molecular Pathology Biomarkers, University of Extremadura, 06006 Badajoz, Spain; (J.P.); (V.G.-L.); (V.G.-M.)
| | - Virginio García-López
- Institute of Molecular Pathology Biomarkers, University of Extremadura, 06006 Badajoz, Spain; (J.P.); (V.G.-L.); (V.G.-M.)
- Department of Medical and Surgical Therapeutics, Pharmacology Area, Faculty of Medicine and Health Sciences, University of Extremadura, 06006 Badajoz, Spain
| | - Virginio García-Martínez
- Institute of Molecular Pathology Biomarkers, University of Extremadura, 06006 Badajoz, Spain; (J.P.); (V.G.-L.); (V.G.-M.)
- Department of Human Anatomy and Embryology, Faculty of Medicine and Health Sciences, University of Extremadura, 06006 Badajoz, Spain
| | - Carlos Gutierrez-Merino
- Institute of Molecular Pathology Biomarkers, University of Extremadura, 06006 Badajoz, Spain; (J.P.); (V.G.-L.); (V.G.-M.)
| |
Collapse
|
7
|
Di Martino E, Rayasam A, Vexler ZS. Brain Maturation as a Fundamental Factor in Immune-Neurovascular Interactions in Stroke. Transl Stroke Res 2024; 15:69-86. [PMID: 36705821 PMCID: PMC10796425 DOI: 10.1007/s12975-022-01111-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 01/28/2023]
Abstract
Injuries in the developing brain cause significant long-term neurological deficits. Emerging clinical and preclinical data have demonstrated that the pathophysiology of neonatal and childhood stroke share similar mechanisms that regulate brain damage, but also have distinct molecular signatures and cellular pathways. The focus of this review is on two different diseases-neonatal and childhood stroke-with emphasis on similarities and distinctions identified thus far in rodent models of these diseases. This includes the susceptibility of distinct cell types to brain injury with particular emphasis on the role of resident and peripheral immune populations in modulating stroke outcome. Furthermore, we discuss some of the most recent and relevant findings in relation to the immune-neurovascular crosstalk and how the influence of inflammatory mediators is dependent on specific brain maturation stages. Finally, we comment on the current state of treatments geared toward inducing neuroprotection and promoting brain repair after injury and highlight that future prophylactic and therapeutic strategies for stroke should be age-specific and consider gender differences in order to achieve optimal translational success.
Collapse
Affiliation(s)
- Elena Di Martino
- Department of Neurology, University California San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158-0663, USA
| | - Aditya Rayasam
- Department of Neurology, University California San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158-0663, USA
| | - Zinaida S Vexler
- Department of Neurology, University California San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158-0663, USA.
| |
Collapse
|
8
|
He Q, Wang Y, Fang C, Feng Z, Yin M, Huang J, Ma Y, Mo Z. Advancing stroke therapy: A deep dive into early phase of ischemic stroke and recanalization. CNS Neurosci Ther 2024; 30:e14634. [PMID: 38379112 PMCID: PMC10879038 DOI: 10.1111/cns.14634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 01/16/2024] [Accepted: 01/23/2024] [Indexed: 02/22/2024] Open
Abstract
Ischemic stroke, accounting for the majority of stroke events, significantly contributes to global morbidity and mortality. Vascular recanalization therapies, namely intravenous thrombolysis and mechanical thrombectomy, have emerged as critical interventions, yet their success hinges on timely application and patient-specific factors. This review focuses on the early phase pathophysiological mechanisms of ischemic stroke and the nuances of recanalization. It highlights the dual role of neutrophils in tissue damage and repair, and the critical involvement of the blood-brain barrier (BBB) in stroke outcomes. Special emphasis is placed on ischemia-reperfusion injury, characterized by oxidative stress, inflammation, and endothelial dysfunction, which paradoxically exacerbates cerebral damage post-revascularization. The review also explores the potential of targeting molecular pathways involved in BBB integrity and inflammation to enhance the efficacy of recanalization therapies. By synthesizing current research, this paper aims to provide insights into optimizing treatment protocols and developing adjuvant neuroprotective strategies, thereby advancing stroke therapy and improving patient outcomes.
Collapse
Affiliation(s)
- Qianyan He
- Department of Neurology, Stroke CenterThe First Hospital of Jilin UniversityJilinChina
- Institute of Biomedicine and BiotechnologyShenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenGuangdongChina
| | - Yueqing Wang
- Institute of Biomedicine and BiotechnologyShenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenGuangdongChina
| | - Cheng Fang
- Institute of Biomedicine and BiotechnologyShenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenGuangdongChina
| | - Ziying Feng
- Institute of Biomedicine and BiotechnologyShenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenGuangdongChina
| | - Meifang Yin
- Institute of Biomedicine and BiotechnologyShenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenGuangdongChina
| | - Juyang Huang
- School of Pharmaceutical Sciences (Shenzhen)Sun Yat‐sen UniversityShenzhenGuangdongChina
| | - Yinzhong Ma
- Institute of Biomedicine and BiotechnologyShenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenGuangdongChina
| | - Zhizhun Mo
- Emergency Department, Shenzhen Traditional Chinese Medicine HospitalThe Fourth Clinical Medical College of Guangzhou University of Chinese MedicineShenzhenGuangdongChina
| |
Collapse
|
9
|
Sunny A, James RR, Menon SR, Rayaroth S, Daniel A, Thompson NA, Tharakan B. Matrix Metalloproteinase-9 inhibitors as therapeutic drugs for traumatic brain injury. Neurochem Int 2024; 172:105642. [PMID: 38008261 DOI: 10.1016/j.neuint.2023.105642] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 11/04/2023] [Accepted: 11/09/2023] [Indexed: 11/28/2023]
Abstract
Traumatic brain injury (TBI) is one of the leading causes of morbidity and mortality among young adults and the elderly. In the United States, TBI is responsible for around 30 percent of all injuries brought on by injuries in general. Vasogenic cerebral edema due to blood-brain barrier (BBB) dysfunction and the associated elevation of intracranial pressure (ICP) are some of the major causes of secondary injuries following traumatic brain injury. Matrix metalloproteinase-9 (MMP-9) is a therapeutic target for being an enzyme that degrades the proteins that make up a part of the microvascular basal lamina as well as inter-endothelial tight junctions of the blood-brain barrier. MMP-9-mediated BBB dysfunctions and the compromise of the BBB is a major pathway that leads the development of vasogenic cerebral edema, elevation of ICP, poor cerebral perfusion and brain herniation following traumatic brain injury. That makes MMP-9 an effective therapeutic target and endogenous or exogenous MMP-9 inhibitors as therapeutic drugs for preventing secondary brain damage after traumatic brain injury. Although our understanding of the mechanisms that underlie the primary and secondary stages of damage following a TBI has significantly improved in recent years, such information has not yet resulted in the successful development of novel pharmacological treatment options for traumatic brain injury. Recent pre-clinical and/or clinical studies have demonstrated that there are several compounds with specific or non-specific MMP-9 inhibitory properties either directly binding and inhibiting MMP-9 or by indirectly inhibiting MMP-9, with potential as therapeutic agents for traumatic brain injury. This article reviews the efficacy of several such medications and potential agents that include endogenous and exogeneous compounds that are at various levels of research and development. MMP-9-based therapeutic drug development has enormous potential in the pharmacological treatment of cerebral edema and/or neuronal injury resulting from traumatic brain injury.
Collapse
Affiliation(s)
- Angel Sunny
- Icahn School of Medicine at Mount Sinai, Elmhurst, NY, USA
| | | | | | | | - Abhijith Daniel
- Pushpagiri Institute of Medical Sciences and Research Centre, Thiruvalla, India
| | - Namita Ann Thompson
- Pushpagiri Institute of Medical Sciences and Research Centre, Thiruvalla, India
| | - Binu Tharakan
- Department of Surgery, Morehouse School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
10
|
Cipriani R, Domerq M, Martín A, Matute C. Role of Microglia in Stroke. ADVANCES IN NEUROBIOLOGY 2024; 37:405-422. [PMID: 39207705 DOI: 10.1007/978-3-031-55529-9_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Ischemic stroke is a complex brain pathology caused by an interruption of blood supply to the brain. It results in neurological deficits which that reflect the localization and the size of the compromised brain area and are the manifestation of complex pathogenic events triggered by energy depletion. Inflammation plays a prominent role, worsening the injury in the early phase and influencing poststroke recovery in the late phase. Activated microglia are one of the most important cellular components of poststroke inflammation, appearing from the first few hours and persisting for days and weeks after stroke injury. In this chapter, we will discuss the nature of the inflammatory response in brain ischemia, the contribution of microglia to injury and regeneration after stroke, and finally, how ischemic stroke directly affects microglia functions and survival.
Collapse
Affiliation(s)
| | - Maria Domerq
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU) and CIBERNED, Leioa, Spain
| | - Abraham Martín
- Achucarro Basque Center for Neuroscience, Leioa, Spain.
- Ikerbasque Basque Foundation for Science, Bilbao, Spain.
| | - Carlos Matute
- Achucarro Basque Center for Neuroscience, Leioa, Spain.
- Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU) and CIBERNED, Leioa, Spain.
| |
Collapse
|
11
|
Chiang HL, Wu KC, Chen YY, Ho CJ, Wang HL, Fu YH, Chen WY, Lin CJ. The Critical Role of Equilibrative Nucleoside Transporter-2 in Modulating Cerebral Damage and Vascular Dysfunction in Mice with Brain Ischemia-Reperfusion. Pharm Res 2023; 40:2541-2554. [PMID: 37498500 DOI: 10.1007/s11095-023-03565-2] [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: 05/18/2023] [Accepted: 07/11/2023] [Indexed: 07/28/2023]
Abstract
BACKGROUND Cerebral vascular protection is critical for stroke treatment. Adenosine modulates vascular flow and exhibits neuroprotective effects, in which brain extracellular concentration of adenosine is dramatically increased during ischemic events and ischemia-reperfusion. Since the equilibrative nucleoside transporter-2 (Ent2) is important in regulating brain adenosine homeostasis, the present study aimed to investigate the role of Ent2 in mice with cerebral ischemia-reperfusion. METHODS Cerebral ischemia-reperfusion injury was examined in mice with transient middle cerebral artery occlusion (tMCAO) for 90 minutes, followed by 24-hour reperfusion. Infarct volume, brain edema, neuroinflammation, microvascular structure, regional cerebral blood flow (rCBF), cerebral metabolic rate of oxygen (CMRO2), and the production of reactive oxygen species (ROS) were examined following the reperfusion. RESULTS Ent2 deletion reduced the infarct volume, brain edema, and neuroinflammation in mice with cerebral ischemia-reperfusion. tMCAO-induced disruption of brain microvessels was ameliorated in Ent2-/- mice, with a reduced expression of matrix metalloproteinases-9 and aquaporin-4 proteins. Following the reperfusion, the rCBF of the wild-type (WT) mice was quickly restored to the baseline, whereas, in Ent2-/- mice, rCBF was slowly recovered initially, but was then higher than that in the WT mice at the later phase of reperfusion. The improved CMRO2 and reduced ROS level support the beneficial effects caused by the changes in the rCBF of Ent2-/- mice. Further studies showed that the protective effects of Ent2 deletion in mice with tMCAO involve adenosine receptor A2AR. CONCLUSIONS Ent2 plays a critical role in modulating cerebral collateral circulation and ameliorating pathological events of brain ischemia and reperfusion injury.
Collapse
Affiliation(s)
- Hui-Ling Chiang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Kuo-Chen Wu
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
- Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan
| | - You-Yin Chen
- Department of Biomedical Engineering, National Yang-Ming-Chiao-Tung University, Taipei, Taiwan
| | - Chin-Jui Ho
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Han-Lin Wang
- Department of Biomedical Engineering, National Yang-Ming-Chiao-Tung University, Taipei, Taiwan
| | - Yu-Hua Fu
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wen-Yu Chen
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chun-Jung Lin
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
12
|
Williams EI, Betterton RD, Stanton JA, Moreno-Rodriguez VM, Lochhead JJ, Davis TP, Ronaldson PT. Oatp (Organic Anion Transporting Polypeptide)-Mediated Transport: A Mechanism for Atorvastatin Neuroprotection in Stroke. Stroke 2023; 54:2875-2885. [PMID: 37750296 PMCID: PMC10615849 DOI: 10.1161/strokeaha.123.043649] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 08/13/2023] [Accepted: 08/31/2023] [Indexed: 09/27/2023]
Abstract
BACKGROUND Drug discovery for stroke is challenging as indicated by poor clinical translatability. In contrast, HMG-CoA (3-hydroxy-3-methylglutaryl coenzyme A) reductase inhibitors (ie, statins) improve poststroke neurological outcomes. This property requires transport across the blood-brain barrier via an endogenous uptake transporter (ie, Oatp1a4 [organic anion transporting polypeptide 1a4]). Our goal was to study Oatp1a4 as a drug delivery mechanism because the blood-brain barrier cannot be assumed to be completely open for all drugs in ischemic stroke. METHODS Male Sprague-Dawley rats (200-250 g) were subjected to middle cerebral artery occlusion (90 minutes) followed by reperfusion for up to 7 days. Atorvastatin (20 mg/kg, IV) was administered 2 hours following intraluminal suture removal. Involvement of Oatp-mediated transport was determined using fexofenadine (3.2 mg/kg, IV), a competitive Oatp inhibitor. Oatp1a4 transport activity was measured by in situ brain perfusion. Infarction volumes/brain edema ratios and neuronal nuclei expression were determined using 2,3,5-triphenyltetrazolium chloride-stained brain tissue slices and confocal microscopy, respectively. Poststroke functional outcomes were assessed via neurological deficit scores and rotarod analysis. RESULTS At 2-hour post-middle cerebral artery occlusion, [3H]atorvastatin uptake was increased in ischemic brain tissue. A single dose of atorvastatin significantly reduced post-middle cerebral artery occlusion infarction volume, decreased brain edema ratio, increased caudoputamen neuronal nuclei expression, and improved functional neurological outcomes. All middle cerebral artery occlusion positive effects of atorvastatin were attenuated by fexofenadine coadministration (ie, an Oatp transport inhibitor). CONCLUSIONS Our data demonstrate that neuroprotective effects of atorvastatin may require central nervous system delivery by Oatp-mediated transport at the blood-brain barrier, a mechanism that persists despite increased cerebrovascular permeability in ischemic stroke. These novel and translational findings support utility of blood-brain barrier transporters in drug delivery for neuroprotective agents.
Collapse
Affiliation(s)
- Erica I. Williams
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Robert D. Betterton
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Joshua A. Stanton
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | | | - Jeffrey J. Lochhead
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Thomas P. Davis
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Patrick T. Ronaldson
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
13
|
Rahman Z, Ghuge S, Dandekar MP. Partial blood replacement ameliorates middle cerebral artery occlusion generated neurological aberrations by intervening TLR4 and NLRP3 cascades in rats. Metab Brain Dis 2023; 38:2339-2354. [PMID: 37402080 DOI: 10.1007/s11011-023-01259-7] [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: 12/03/2022] [Accepted: 06/26/2023] [Indexed: 07/05/2023]
Abstract
Acute ischemic stroke is a catastrophic medical condition that causes severe disability and mortality if the sufferer escapes treatment within a stipulated timeframe. While timely intervention with clot-bursting agents like tissue-plasminogen activators abrogates some post-stroke neurologic deficits, no neuroprotective therapy is yet promisingly addresses the post-recanalization neuroinflammation in post-stroke survivors. Herein, we investigated the effect of partial blood replacement therapy (BRT), obtained from healthy and treadmill-trained donor rats, on neurological deficits, and peripheral and central inflammatory cascades using the ischemia-reperfusion animal paradigm. The cerebral ischemia-reperfusion was induced in rats by occlusion of the middle cerebral artery (MCAO) for 90 min, followed by reperfusion. Rats underwent MCAO surgery displayed remarkable sensorimotor and motor deficits in rotarod, foot fault, adhesive removal, and paw whisker tests till 5 days post-surgery. These behavior abnormalities were ameliorated in the BRT-recipient MCAO rats. BRT also reduced the infarct volume and neuronal death in the ipsilateral hemisphere revealed by TTC and cresyl violet staining compared to the MCAO group. Rats received BRT infusion exhibited the reduced expression of glial fibrillary acidic protein, ionized calcium-binding adaptor molecule-1 (Iba-1), and MyD88 on day 5 post-MCAO in immunohistochemistry and immunofluorescent assays. Moreover, elevated levels of toll-like receptor 4 (TLR4) and mRNA expression of IL-1β, TNF-α, matrix metalloproteinase-9 and NLRP3, and decreased levels of zonula occludens-1 in MCAO rats, were reversed following BRT. These findings suggest that the partial BRT may rescind MCAO-induced neurological dysfunctions and cerebral injury by intervening in the TLR4 and NLRP3 pathways in rats.
Collapse
Affiliation(s)
- Ziaur Rahman
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India
| | - Shubham Ghuge
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India
| | - Manoj P Dandekar
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, 500037, Telangana, India.
| |
Collapse
|
14
|
Wendt TS, Gonzales RJ. Ozanimod differentially preserves human cerebrovascular endothelial barrier proteins and attenuates matrix metalloproteinase-9 activity following in vitro acute ischemic injury. Am J Physiol Cell Physiol 2023; 325:C951-C971. [PMID: 37642239 DOI: 10.1152/ajpcell.00342.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 08/31/2023]
Abstract
Endothelial integrity is critical in mitigating a vicious cascade of secondary injuries following acute ischemic stroke (AIS). Matrix metalloproteinase-9 (MMP-9), a contributor to endothelial integrity loss, is elevated during stroke and is associated with worsened stroke outcome. We investigated the FDA-approved selective sphingosine-1-phosphate receptor 1 (S1PR1) ligand, ozanimod, on the regulation/activity of MMP-9 as well as endothelial barrier components [platelet endothelial cell adhesion molecule 1 (PECAM-1), claudin-5, and zonula occludens 1 (ZO-1)] in human brain microvascular endothelial cells (HBMECs) following hypoxia plus glucose deprivation (HGD). We previously reported that S1PR1 activation improves HBMEC integrity; however, mechanisms underlying S1PR1 involvement in endothelial cell barrier integrity have not been clearly elucidated. We hypothesized that ozanimod would attenuate an HGD-induced increase in MMP-9 activity that would concomitantly attenuate the loss of integral barrier components. Male HBMECs were treated with ozanimod or vehicle and exposed to 3 h of normoxia (21% O2) or HGD (1% O2). Immunoblotting, zymography, qRT-PCR, and immunocytochemical labeling techniques assessed processes related to MMP-9 and barrier markers. We observed that HGD acutely increased MMP-9 activity and reduced claudin-5 and PECAM-1 levels, and ozanimod attenuated these responses. In situ analysis, via PROSPER, suggested that attenuation of MMP-9 activity may be a primary factor in maintaining these integral barrier proteins. We also observed that HGD increased intracellular mechanisms associated with augmented MMP-9 activation; however, ozanimod had no effect on these select factors. Thus, we conclude that ozanimod has the potential to attenuate HGD-mediated decreases in HBMEC integrity in part by decreasing MMP-9 activity as well as preserving barrier properties.NEW & NOTEWORTHY We have identified a potential novel mechanism by which ozanimod, a selective sphingosine-1-phosphate receptor 1 (S1PR1) agonist, attenuates hypoxia plus glucose deprivation (HGD)-induced matrix metalloproteinase-9 (MMP-9) activity and disruptions in integral human brain endothelial cell barrier proteins. Our results suggest that ischemic-like injury elicits increased MMP-9 activity and alterations of barrier integrity proteins in human brain microvascular endothelial cells (HBMECs) and that ozanimod via S1PR1 attenuates these HGD-induced responses, adding to its therapeutic potential in cerebrovascular protection during the acute phase of ischemic stroke.
Collapse
Affiliation(s)
- Trevor S Wendt
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, Arizona, United States
| | - Rayna J Gonzales
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, Arizona, United States
| |
Collapse
|
15
|
Balog BM, Sonti A, Zigmond RE. Neutrophil biology in injuries and diseases of the central and peripheral nervous systems. Prog Neurobiol 2023; 228:102488. [PMID: 37355220 PMCID: PMC10528432 DOI: 10.1016/j.pneurobio.2023.102488] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 05/24/2023] [Accepted: 06/16/2023] [Indexed: 06/26/2023]
Abstract
The role of inflammation in nervous system injury and disease is attracting increased attention. Much of that research has focused on microglia in the central nervous system (CNS) and macrophages in the peripheral nervous system (PNS). Much less attention has been paid to the roles played by neutrophils. Neutrophils are part of the granulocyte subtype of myeloid cells. These cells, like macrophages, originate and differentiate in the bone marrow from which they enter the circulation. After tissue damage or infection, neutrophils are the first immune cells to infiltrate into tissues and are directed there by specific chemokines, which act on chemokine receptors on neutrophils. We have reviewed here the basic biology of these cells, including their differentiation, the types of granules they contain, the chemokines that act on them, the subpopulations of neutrophils that exist, and their functions. We also discuss tools available for identification and further study of neutrophils. We then turn to a review of what is known about the role of neutrophils in CNS and PNS diseases and injury, including stroke, Alzheimer's disease, multiple sclerosis, amyotrophic lateral sclerosis, spinal cord and traumatic brain injuries, CNS and PNS axon regeneration, and neuropathic pain. While in the past studies have focused on neutrophils deleterious effects, we will highlight new findings about their benefits. Studies on their actions should lead to identification of ways to modify neutrophil effects to improve health.
Collapse
Affiliation(s)
- Brian M Balog
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106-4975, USA
| | - Anisha Sonti
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106-4975, USA
| | - Richard E Zigmond
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106-4975, USA.
| |
Collapse
|
16
|
Peng T, Booher K, Moody MR, Yin X, Aronowski J, McPherson DD, Savitz SI, Kim H, Huang SL. Enhanced Cerebroprotection of Xenon-Loaded Liposomes in Combination with rtPA Thrombolysis for Embolic Ischemic Stroke. Biomolecules 2023; 13:1256. [PMID: 37627321 PMCID: PMC10452377 DOI: 10.3390/biom13081256] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/12/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Xenon (Xe) has shown great potential as a stroke treatment due to its exceptional ability to protect brain tissue without inducing side effects. We have previously developed Xe-loaded liposomes for the ultrasound-activated delivery of Xe into the cerebral region and demonstrated their therapeutic efficacy. At present, the sole FDA-approved thrombolytic agent for stroke treatment is recombinant tissue plasminogen activator (rtPA). In this study, we aimed to investigate the potential of combining Xe-liposomes with an intravenous rtPA treatment in a clinically relevant embolic rat stroke model. We evaluated the combinational effect using an in vitro clot lysis model and an in vivo embolic middle cerebral artery occlusion (eMCAO) rat model. The treatment groups received intravenous administration of Xe-liposomes (20 mg/kg) at 2 h post-stroke onset, followed by the administration of rtPA (10 mg/kg) at either 2 or 4 h after the onset. Three days after the stroke, behavioral tests were conducted, and brain sections were collected for triphenyltetrazolium chloride (TTC) and TUNEL staining. Infarct size was determined as normalized infarct volume (%). Both in vitro and in vivo clot lysis experiments demonstrated that Xe-liposomes in combination with rtPA resulted in effective clot lysis comparable to the treatment with free rtPA alone. Animals treated with Xe-liposomes in combination with rtPA showed reduced TUNEL-positive cells and demonstrated improved neurological recovery. Importantly, Xe-liposomes in combination with late rtPA treatment reduced rtPA-induced hemorrhage, attributing to the reduction of MMP9 immunoreactivity. This study demonstrates that the combined therapy of Xe-liposomes and rtPA provides enhanced therapeutic efficacy, leading to decreased neuronal cell death and a potential to mitigate hemorrhagic side effects associated with late rtPA treatment.
Collapse
Affiliation(s)
- Tao Peng
- Division of Cardiovascular Medicine, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (T.P.); (M.R.M.); (X.Y.); (D.D.M.)
| | - Keith Booher
- Zymo Research Corporation, Irvine, CA 92614, USA;
| | - Melanie R. Moody
- Division of Cardiovascular Medicine, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (T.P.); (M.R.M.); (X.Y.); (D.D.M.)
| | - Xing Yin
- Division of Cardiovascular Medicine, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (T.P.); (M.R.M.); (X.Y.); (D.D.M.)
| | - Jaroslaw Aronowski
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (J.A.); (S.I.S.)
- Institute for Stroke and Cerebrovascular Disease, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - David D. McPherson
- Division of Cardiovascular Medicine, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (T.P.); (M.R.M.); (X.Y.); (D.D.M.)
| | - Sean I. Savitz
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (J.A.); (S.I.S.)
- Institute for Stroke and Cerebrovascular Disease, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Hyunggun Kim
- Division of Cardiovascular Medicine, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (T.P.); (M.R.M.); (X.Y.); (D.D.M.)
- Department of Biomechatronic Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Shao-Ling Huang
- Division of Cardiovascular Medicine, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (T.P.); (M.R.M.); (X.Y.); (D.D.M.)
- Institute for Stroke and Cerebrovascular Disease, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
17
|
Sheu ML, Pan LY, Yang CN, Sheehan J, Pan LY, You WC, Wang CC, Pan HC. Thrombin-Induced Microglia Activation Modulated through Aryl Hydrocarbon Receptors. Int J Mol Sci 2023; 24:11416. [PMID: 37511175 PMCID: PMC10380349 DOI: 10.3390/ijms241411416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 07/01/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023] Open
Abstract
Thrombin is a multifunctional serine protein which is closely related to neurodegenerative disorders. The Aryl hydrocarbon receptor (AhR) is well expressed in microglia cells involving inflammatory disorders of the brain. However, it remains unclear as to how modulation of AhR expression by thrombin is related to the development of neurodegeneration disorders. In this study, we investigated the role of AhR in the development of thrombin-induced neurodegenerative processes, especially those concerning microglia. The primary culture of either wild type or AhR deleted microglia, as well as BV-2 cell lines, was used for an in vitro study. Hippocampal slice culture and animals with either wild type or with AhR deleted were used for the ex vivo and in vivo studies. Simulations of ligand protein docking showed a strong integration between the thrombin and AhR. In thrombin-triggered microglia cells, deleting AhR escalated both the NO release and iNOS expression. Such effects were abolished by the administration of the AhR agonist. In thrombin-activated microglia cells, downregulating AhR increased the following: vascular permeability, pro-inflammatory genetic expression, MMP-9 activity, and the ratio of M1/M2 phenotype. In the in vivo study, thrombin induced the activation of microglia and their volume, thereby contributing to the deterioration of neurobehavior. Deleting AhR furthermore aggravated the response in terms of impaired neurobehavior, increasing brain edema, aggregating microglia, and increasing neuronal death. In conclusion, thrombin caused the activation of microglia through increased vessel permeability, expression of inflammatory response, and phenotype of M1 microglia, as well the MMP activity. Deleting AhR augmented the above detrimental effects. These findings indicate that the modulation of AhR is essential for the regulation of thrombin-induced brain damages and that the AhR agonist may harbor the potentially therapeutic effect in thrombin-induced neurodegenerative disorder.
Collapse
Affiliation(s)
- Meei-Ling Sheu
- Institute of Biomedical Sciences, National Chung-Hsing University, Taichung 40227, Taiwan;
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 40210, Taiwan
- Ph.D. Program in Translational Medicine, Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 40227, Taiwan
| | - Liang-Yi Pan
- Faculty of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Cheng-Ning Yang
- Department of Dentistry, School of Dentistry, College of Medicine, National Taiwan University, Taipei 106319, Taiwan;
| | - Jason Sheehan
- Department of Neurosurgery, University of Virginia, Charlottesville, VA 22904, USA;
| | - Liang-Yu Pan
- Faculty of Medicine, Poznan University of Medical Sciences, 61-701 Poznań, Poland;
| | - Weir-Chiang You
- Department of Radiation Oncology, Taichung Veterans General Hospital, Taichung 40210, Taiwan;
| | - Chien-Chia Wang
- Department of Life Sciences, National Central University, Taoyuan 32001, Taiwan;
| | - Hung-Chuan Pan
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 40210, Taiwan
- Ph.D. Program in Translational Medicine, Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 40227, Taiwan
- Department of Neurosurgery, Taichung Veterans General Hospital, Taichung 40210, Taiwan
| |
Collapse
|
18
|
Ji Y, Gao Q, Ma Y, Wang F, Tan X, Song D, Hoo RLC, Wang Z, Ge X, Han H, Guo F, Chang J. An MMP-9 exclusive neutralizing antibody attenuates blood-brain barrier breakdown in mice with stroke and reduces stroke patient-derived MMP-9 activity. Pharmacol Res 2023; 190:106720. [PMID: 36893823 DOI: 10.1016/j.phrs.2023.106720] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 02/27/2023] [Accepted: 03/06/2023] [Indexed: 03/09/2023]
Abstract
Rapid upregulation of matrix metalloproteinase 9 (MMP-9) leads to blood-brain barrier (BBB) breakdown following stroke, but no MMP-9 inhibitors have been approved in clinic largely due to their low specificities and side effects. Here, we explored the therapeutic potential of a human IgG monoclonal antibody (mAb), L13, which was recently developed with exclusive neutralizing specificity to MMP-9, nanomolar potency, and biological function, using mouse stroke models and stroke patient samples. We found that L13 treatment at the onset of reperfusion following cerebral ischemia or after intracranial hemorrhage (ICH) significantly reduced brain tissue injury and improved the neurological outcomes of mice. Compared to control IgG, L13 substantially attenuated BBB breakdown in both types of stroke model by inhibiting MMP-9 activity-mediated degradations of basement membrane and endothelial tight junction proteins. Importantly, these BBB-protective and neuroprotective effects of L13 in wild-type mice were comparable to Mmp9 genetic deletion and fully abolished in Mmp9 knockout mice, highlighting the in vivo target specificity of L13. Meanwhile, ex vivo co-incubation with L13 significantly neutralized the enzymatic activities of human MMP-9 in the sera of ischemic and hemorrhagic stroke patients, or in the peri-hematoma brain tissues from hemorrhagic stroke patients. Overall, we demonstrated that MMP-9 exclusive neutralizing mAbs constitute a potential feasible therapeutic approach for both ischemic and hemorrhagic stroke.
Collapse
Affiliation(s)
- Yabin Ji
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Qiang Gao
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450001, China; Department of Neurosurgery, Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100084, China
| | - Yinzhong Ma
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Fang Wang
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450001, China
| | - Xixi Tan
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Neurology, Yangjiang People's Hospital, Yangjiang 529500, China
| | - Dengpan Song
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450001, China
| | - Ruby L C Hoo
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong 999077, China
| | - Zening Wang
- Department of Chemical and Environmental Engineering, University of California, Riverside, CA 92521, USA
| | - Xin Ge
- Department of Chemical and Environmental Engineering, University of California, Riverside, CA 92521, USA
| | - Hongjie Han
- Department of Neurosurgery, Pingdingshan Second People's Hospital, Pingdingshan 467000, China
| | - Fuyou Guo
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450001, China.
| | - Junlei Chang
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| |
Collapse
|
19
|
Wang Z, Song Y, Bai S, Xiang W, Zhou X, Han L, Zhu D, Guan Y. Imaging of microglia in post-stroke inflammation. Nucl Med Biol 2023; 118-119:108336. [PMID: 37028196 DOI: 10.1016/j.nucmedbio.2023.108336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 03/21/2023] [Accepted: 03/21/2023] [Indexed: 04/03/2023]
Abstract
Microglia constantly survey the central nervous system microenvironment and maintain brain homeostasis. Microglia activation, polarization and inflammatory response are of great importance in the pathophysiology of ischemic stroke. For exploring biochemical processes in vivo, positron emission tomography (PET) is a superior imaging tool. Translocator protein 18 kDa (TSPO), is a validated neuroinflammatory biomarker which is widely used to evaluate various central nervous system (CNS) pathologies in both preclinical and clinical studies. TSPO level can be elevated due to peripheral inflammatory cells infiltration and glial cells activation. Therefore, a clear understanding of the dynamic changes between microglia and TSPO is critical for interpreting PET studies and understanding the pathophysiology after ischemic stroke. Our review discusses alternative biological targets that have attracted considerable interest for the imaging of microglia activation in recent years, and the potential value of imaging of microglia in the assessment of stroke therapies.
Collapse
|
20
|
Matrix Metalloproteinases in Cardioembolic Stroke: From Background to Complications. Int J Mol Sci 2023; 24:ijms24043628. [PMID: 36835040 PMCID: PMC9959608 DOI: 10.3390/ijms24043628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/20/2023] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
Abstract
Matrix metalloproteinases (MMPs) are endopeptidases participating in physiological processes of the brain, maintaining the blood-brain barrier integrity and playing a critical role in cerebral ischemia. In the acute phase of stroke activity, the expression of MMPs increase and is associated with adverse effects, but in the post-stroke phase, MMPs contribute to the process of healing by remodeling tissue lesions. The imbalance between MMPs and their inhibitors results in excessive fibrosis associated with the enhanced risk of atrial fibrillation (AF), which is the main cause of cardioembolic strokes. MMPs activity disturbances were observed in the development of hypertension, diabetes, heart failure and vascular disease enclosed in CHA2DS2VASc score, the scale commonly used to evaluate the risk of thromboembolic complications risk in AF patients. MMPs involved in hemorrhagic complications of stroke and activated by reperfusion therapy may also worsen the stroke outcome. In the present review, we briefly summarize the role of MMPs in the ischemic stroke with particular consideration of the cardioembolic stroke and its complications. Moreover, we discuss the genetic background, regulation pathways, clinical risk factors and impact of MMPs on the clinical outcome.
Collapse
|
21
|
Chandrabalan A, Firth A, Litchfield RB, Appleton CT, Getgood A, Ramachandran R. Human osteoarthritis knee joint synovial fluids cleave and activate Proteinase-Activated Receptor (PAR) mediated signaling. Sci Rep 2023; 13:1124. [PMID: 36670151 PMCID: PMC9859807 DOI: 10.1038/s41598-023-28068-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 01/12/2023] [Indexed: 01/22/2023] Open
Abstract
Osteoarthritis (OA) is the most prevalent joint disorder with increasing worldwide incidence. Mechanistic insights into OA pathophysiology are evolving and there are currently no disease-modifying OA drugs. An increase in protease activity is linked to progressive degradation of the cartilage in OA. Proteases also trigger inflammation through a family of G protein-coupled receptors (GPCRs) called the Proteinase-Activated Receptors (PARs). PAR signaling can trigger pro-inflammatory responses and targeting PARs is proposed as a therapeutic approach in OA. Several enzymes can cleave the PAR N-terminus, but the endogenous protease activators of PARs in OA remain unclear. Here we characterized PAR activating enzymes in knee joint synovial fluids from OA patients and healthy donors using genetically encoded PAR biosensor expressing cells. Calcium signaling assays were performed to examine receptor activation. The class and type of enzymes cleaving the PARs was further characterized using protease inhibitors and fluorogenic substrates. We find that PAR1, PAR2 and PAR4 activating enzymes are present in knee joint synovial fluids from healthy controls and OA patients. Compared to healthy controls, PAR1 activating enzymes are elevated in OA synovial fluids while PAR4 activating enzyme levels are decreased. Using enzyme class and type selective inhibitors and fluorogenic substrates we find that multiple PAR activating enzymes are present in OA joint fluids and identify serine proteinases (thrombin and trypsin-like) and matrix metalloproteinases as the major classes of PAR activating enzymes in the OA synovial fluids. Synovial fluid driven increase in calcium signaling was significantly reduced in cells treated with PAR1 and PAR2 antagonists, but not in PAR4 antagonist treated cells. OA associated elevation of PAR1 cleavage suggests that targeting this receptor may be beneficial in the treatment of OA.
Collapse
Affiliation(s)
- Arundhasa Chandrabalan
- Department of Physiology and Pharmacology, Bone and Joint Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
| | - Andrew Firth
- Division of Orthopedic Surgery, Bone and Joint Institute, Fowler Kennedy Sport Medicine Clinic, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Robert B Litchfield
- Division of Orthopedic Surgery, Bone and Joint Institute, Fowler Kennedy Sport Medicine Clinic, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - C Thomas Appleton
- Department of Physiology and Pharmacology, Bone and Joint Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada.,Department of Medicine, Bone and Joint Institute, Schulich School of Medicine and Dentistry, The Dr. Sandy Kirkley Centre for Musculoskeletal Research, London, ON, Canada
| | - Alan Getgood
- Division of Orthopedic Surgery, Bone and Joint Institute, Fowler Kennedy Sport Medicine Clinic, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Rithwik Ramachandran
- Department of Physiology and Pharmacology, Bone and Joint Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada.
| |
Collapse
|
22
|
Constantakis JW, Reed-McBain CA, Famakin B. Astrocyte innate immune activation and injury amplification following experimental focal cerebral ischemia. Neurochem Int 2023; 162:105456. [PMID: 36509233 DOI: 10.1016/j.neuint.2022.105456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/17/2022] [Accepted: 11/19/2022] [Indexed: 12/13/2022]
Abstract
Astrocytes are a distinct population of glial cells responsible for many homeostatic functions in normal neural architecture. In the healthy brain, astrocyte functions range from maintenance of the blood brain barrier to modulation of synaptic transmission and neuronal plasticity to glial scar formation post-ischemic injury. In humans, this group of cells exhibits far greater heterogeneity than previously thought-with distinct subpopulations that likely carry out specialized functions. Following ischemic injury, astrocytes take on a distinct phenotype-known as the reactive astrocyte. This phenotype is responsible for both the propagation and amelioration of neuronal injury during ischemia. Following ischemia, astrocytes undergo temporal and spatial-dependent changes in morphology, gene expression, hypertrophy and hyperplasia as a result of signaling within the local microenvironment of the penumbra compared to the core infarct. This elicits a cascade of downstream effects, including inflammation and activation of the innate immune system, which both propagates and ameliorates local injury within the brain parenchyma. This review will focus upon the double-edged sword-that are astrocytes and the innate immune system. We will discuss the role that astrocytes and the innate immune system play in amplifying secondary brain injury, as well as attenuating ischemic damage. Specifically, we will focus on molecular signaling and processes that could be targeted as potential therapeutic interventions.
Collapse
Affiliation(s)
- John W Constantakis
- Department of Neurology, University of Wisconsin, School of Medicine and Public Health, Madison, WI, 53705, USA
| | - Catherine A Reed-McBain
- Department of Dermatology, University of Wisconsin, School of Medicine and Public Health, Madison, WI, 53705, USA
| | - Bolanle Famakin
- Department of Neurology, University of Wisconsin, School of Medicine and Public Health, Madison, WI, 53705, USA.
| |
Collapse
|
23
|
Xu J, Li P, Lu F, Chen Y, Guo Q, Yang Y. Domino reaction of neurovascular unit in neuropathic pain after spinal cord injury. Exp Neurol 2023; 359:114273. [PMID: 36375510 DOI: 10.1016/j.expneurol.2022.114273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/25/2022] [Accepted: 11/08/2022] [Indexed: 11/12/2022]
Abstract
The mechanism of neuropathic pain after spinal cord injury is complex, and the communication between neurons, glia, and blood vessels in neurovascular units significantly affects the occurrence and development of neuropathic pain. After spinal cord injury, a domino chain reaction occurs in the neuron-glia-vessel, which affects the permeability of the blood-spinal cord barrier and jointly promotes the development of neuroinflammation. This article discusses the signal transduction between neuro-glial-endothelial networks from a multidimensional point of view and reviews its role in neuropathic pain after spinal cord injury.
Collapse
Affiliation(s)
- Jingmei Xu
- Department of Anesthesiology, Xiangya Hospital, Central South University, 87th Xiangya Road, Kaifu District, Changsha, Hunan, China
| | - Ping Li
- National Clinical Research Center for Geriatric Disorders,Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Department of Obstetrics, Xiangya Hospital, Central South University, 87th Xiangya Road, Kaifu District, Changsha, Hunan, China
| | - Feng Lu
- Department of Anesthesiology, First Affiliated Hospital of Gannan medical university, Ganzhou 341000, China
| | - Yulu Chen
- Department of Anesthesiology, Xiangya Hospital, Central South University, 87th Xiangya Road, Kaifu District, Changsha, Hunan, China
| | - Qulian Guo
- Department of Anesthesiology, Xiangya Hospital, Central South University, 87th Xiangya Road, Kaifu District, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders,Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Yong Yang
- Department of Anesthesiology, Xiangya Hospital, Central South University, 87th Xiangya Road, Kaifu District, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders,Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China.
| |
Collapse
|
24
|
Lu H, Ashiqueali R, Lin CI, Walchale A, Clendaniel V, Matheson R, Fisher M, Lo EH, Selim M, Shehadah A. Histone Deacetylase 3 Inhibition Decreases Cerebral Edema and Protects the Blood–Brain Barrier After Stroke. Mol Neurobiol 2022; 60:235-246. [DOI: 10.1007/s12035-022-03083-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 10/03/2022] [Indexed: 11/30/2022]
Abstract
AbstractWe have previously shown that selective inhibition of histone deacetylase 3 (HDAC3) decreases infarct volume and improves long-term functional outcomes after stroke. In this study, we examined the effects of HDAC3 inhibition on cerebral edema and blood–brain barrier (BBB) leakage and explored its underlying mechanisms. Adult male Wistar rats were subjected to 2-h middle cerebral artery occlusion (MCAO) and randomly treated i.p. with either vehicle or a selective HDAC3 inhibitor (RGFP966) at 2 and 24 h after stroke. Modified neurological severity scores (mNSS) were calculated at 2 h, 1 day, and 3 days. H&E, Evans blue dye (EBD) assay, and fluorescein isothiocyanate (FITC)-dextran were employed to assess cerebral edema and BBB leakage. Western blot for matrix metalloproteinase-9 (MMP9), MMP-9 zymography, and immunostaining for HDAC3, GFAP, Iba-1, albumin, aquaporin-4, claudin-5, ZO-1, and NF-kB were performed. Early RGFP966 administration decreased cerebral edema (p = 0.002) and BBB leakage, as measured by EBD assay, FITC-dextran, and albumin extravasation (p < 0.01). RGFP966 significantly increased tight junction proteins (claudin-5 and ZO-1) in the peri-infarct area. RGFP966 also significantly decreased HDAC3 in GFAP + astrocytes, which correlated with better mNSS (r = 0.67, p = 0.03) and decreased cerebral edema (r = 0.64, p = 0.04). RGFP966 decreased aquaporin-4 in GFAP + astrocytes (p = 0.002), as well as, the inflammatory markers Iba-1, NF-kB, and MMP9 in the ischemic brain (p < 0.05). Early HDAC3 inhibition decreases cerebral edema and BBB leakage. BBB protection by RGFP966 is mediated in part by the upregulation of tight junction proteins, downregulation of aquaporin-4 and HDAC3 in astrocytes, and decreased neuroinflammation.
Collapse
|
25
|
Need for a Paradigm Shift in the Treatment of Ischemic Stroke: The Blood-Brain Barrier. Int J Mol Sci 2022; 23:ijms23169486. [PMID: 36012745 PMCID: PMC9409167 DOI: 10.3390/ijms23169486] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/04/2022] [Accepted: 08/18/2022] [Indexed: 11/17/2022] Open
Abstract
Blood-brain barrier (BBB) integrity is essential to maintaining brain health. Aging-related alterations could lead to chronic progressive leakiness of the BBB, which is directly correlated with cerebrovascular diseases. Indeed, the BBB breakdown during acute ischemic stroke is critical. It remains unclear, however, whether BBB dysfunction is one of the first events that leads to brain disease or a down-stream consequence. This review will focus on the BBB dysfunction associated with cerebrovascular disease. An added difficulty is its association with the deleterious or reparative effect, which depends on the stroke phase. We will first outline the BBB structure and function. Then, we will focus on the spatiotemporal chronic, slow, and progressive BBB alteration related to ischemic stroke. Finally, we will propose a new perspective on preventive therapeutic strategies associated with brain aging based on targeting specific components of the BBB. Understanding BBB age-evolutions will be beneficial for new drug development and the identification of the best performance window times. This could have a direct impact on clinical translation and personalised medicine.
Collapse
|
26
|
Baidya SK, Banerjee S, Adhikari N, Jha T. Selective Inhibitors of Medium-Size S1' Pocket Matrix Metalloproteinases: A Stepping Stone of Future Drug Discovery. J Med Chem 2022; 65:10709-10754. [PMID: 35969157 DOI: 10.1021/acs.jmedchem.1c01855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Among various matrix metalloproteinases (MMPs), MMPs having medium-size S1' pockets are established as promising biomolecular targets for executing crucial roles in cancer, cardiovascular diseases, and neurodegenerative diseases. However, no such MMP inhibitors (MMPIs) are available to date as drug candidates despite a lot of continuous research work for more than three decades. Due to a high degree of structural resemblance among these MMPs, designing selective MMPIs is quite challenging. However, the variability and uniqueness of the S1' pockets of these MMPs make them promising targets for designing selective MMPIs. In this perspective, the overall structural aspects of medium-size S1' pocket MMPs including the unique binding patterns of enzyme-inhibitor interactions have been discussed in detail to acquire knowledge regarding selective inhibitor designing. This overall knowledge will surely be a curtain raiser for the designing of selective MMPIs as drug candidates in the future.
Collapse
Affiliation(s)
- Sandip Kumar Baidya
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Suvankar Banerjee
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Nilanjan Adhikari
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Tarun Jha
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| |
Collapse
|
27
|
Green Tea ( Camellia sinensis): A Review of Its Phytochemistry, Pharmacology, and Toxicology. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27123909. [PMID: 35745040 PMCID: PMC9231383 DOI: 10.3390/molecules27123909] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/11/2022] [Accepted: 06/13/2022] [Indexed: 12/21/2022]
Abstract
Objectives Green tea (Camellia sinensis) is a kind of unfermented tea that retains the natural substance in fresh leaves to a great extent. It is regarded as the second most popular drink in the world besides water. In this paper, the phytochemistry, pharmacology, and toxicology of green tea are reviewed systematically and comprehensively. Key findings Green tea has been demonstrated to be good for human health. Nowadays, multiple pharmacologically active components have been isolated and identified from green tea, including tea polyphenols, alkaloids, amino acids, polysaccharides, and volatile components. Recent studies have demonstrated that green tea shows versatile pharmacological activities, such as antioxidant, anticancer, hypoglycemic, antibacterial, antiviral, and neuroprotective. Studies on the toxic effects of green tea extract and its main ingredients have also raised concerns including hepatotoxicity and DNA damage. Summary Green tea can be used to assist the treatment of diabetes, Alzheimer’s disease, oral cancer, and dermatitis. Consequently, green tea has shown promising practical prospects in health care and disease prevention.
Collapse
|
28
|
Fisher RA, Miners JS, Love S. Pathological changes within the cerebral vasculature in Alzheimer's disease: New perspectives. Brain Pathol 2022; 32:e13061. [PMID: 35289012 PMCID: PMC9616094 DOI: 10.1111/bpa.13061] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/11/2022] [Accepted: 02/21/2022] [Indexed: 12/14/2022] Open
Abstract
Cerebrovascular disease underpins vascular dementia (VaD), but structural and functional changes to the cerebral vasculature contribute to disease pathology and cognitive decline in Alzheimer's disease (AD). In this review, we discuss the contribution of cerebral amyloid angiopathy and non‐amyloid small vessel disease in AD, and the accompanying changes to the density, maintenance and remodelling of vessels (including alterations to the composition and function of the cerebrovascular basement membrane). We consider how abnormalities of the constituent cells of the neurovascular unit – particularly of endothelial cells and pericytes – and impairment of the blood‐brain barrier (BBB) impact on the pathogenesis of AD. We also discuss how changes to the cerebral vasculature are likely to impair Aβ clearance – both intra‐periarteriolar drainage (IPAD) and transport of Aβ peptides across the BBB, and how impaired neurovascular coupling and reduced blood flow in relation to metabolic demand increase amyloidogenic processing of APP and the production of Aβ. We review the vasoactive properties of Aβ peptides themselves, and the probable bi‐directional relationship between vascular dysfunction and Aβ accumulation in AD. Lastly, we discuss recent methodological advances in transcriptomics and imaging that have provided novel insights into vascular changes in AD, and recent advances in assessment of the retina that allow in vivo detection of vascular changes in the early stages of AD.
Collapse
Affiliation(s)
- Robert A Fisher
- Dementia Research Group, University of Bristol Medical School, Bristol, UK
| | - J Scott Miners
- Dementia Research Group, University of Bristol Medical School, Bristol, UK
| | - Seth Love
- Dementia Research Group, University of Bristol Medical School, Bristol, UK
| |
Collapse
|
29
|
Acute Hyperglycemia Exacerbates Hemorrhagic Transformation after Embolic Stroke and Reperfusion with tPA: A Possible Role of TXNIP-NLRP3 Inflammasome. J Stroke Cerebrovasc Dis 2022; 31:106226. [PMID: 34847489 PMCID: PMC8792268 DOI: 10.1016/j.jstrokecerebrovasdis.2021.106226] [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] [Received: 07/06/2021] [Revised: 11/08/2021] [Accepted: 11/11/2021] [Indexed: 02/03/2023] Open
Abstract
OBJECTIVES Acute hyperglycemia (HG) exacerbates reperfusion injury after stroke. Our recent studies showed that acute HG upregulates thioredoxin-interacting protein (TXNIP) expression, which in turn induces inflammation and neurovascular damage in a suture model of ischemic stroke. The aim of the present study was to investigate the effect of acute HG on TXNIP-associated neurovascular damage, in a more clinically relevant murine model of embolic stroke and intravenous tissue plasminogen activator (IV-tPA) reperfusion. MATERIALS AND METHODS HG was induced in adult male mice, by intraperitoneal injection of 20% glucose. This was followed by embolic middle cerebral artery occlusion (eMCAO), with or without IV-tPA (10 mg/kg) given 3 h post embolization. Brain infarction, edema, hemoglobin content, expression of matrix metalloproteinase (MMP-9), vascular endothelial growth factor A (VEGFA), tight junction proteins (claudin-5, occluding, and zonula occludens-1), TXNIP, and NOD-like receptor protein3 (NLRP3)-inflammasome activation were evaluated at 24 h after eMCAO. RESULTS HG alone significantly increased TXNIP in the brain after eMCAO, and this was associated with exacerbated hemorrhagic transformation (HT; as measured by hemoglobin content). IV-tPA in HG conditions showed a trend to decrease infarct volume, but worsened HT after eMCAO, suggesting that HG reduces the therapeutic efficacy of IV-tPA. Further, HG and tPA-reperfusion did not show significant differences in expression of MMP-9, VEGFA, junction proteins, and NLRP3 inflammasome activation between the groups. CONCLUSION The current findings suggest a potential role for TXNIP in the occurrence of HT in hyperglycemic conditions following eMCAO. Further studies are needed to understand the precise role of vascular TXNIP on HG/tPA-induced neurovascular damage after stroke.
Collapse
|
30
|
Li W, Cao F, Takase H, Arai K, Lo EH, Lok J. Blood-Brain Barrier Mechanisms in Stroke and Trauma. Handb Exp Pharmacol 2022; 273:267-293. [PMID: 33580391 DOI: 10.1007/164_2020_426] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The brain microenvironment is tightly regulated. The blood-brain barrier (BBB), which is composed of cerebral endothelial cells, astrocytes, and pericytes, plays an important role in maintaining the brain homeostasis by regulating the transport of both beneficial and detrimental substances between circulating blood and brain parenchyma. After brain injury and disease, BBB tightness becomes dysregulated, thus leading to inflammation and secondary brain damage. In this chapter, we overview the fundamental mechanisms of BBB damage and repair after stroke and traumatic brain injury (TBI). Understanding these mechanisms may lead to therapeutic opportunities for brain injury.
Collapse
Affiliation(s)
- Wenlu Li
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Fang Cao
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hajime Takase
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ken Arai
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Eng H Lo
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Josephine Lok
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
31
|
Mechanisms of Thrombosis and Thrombolysis. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00002-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
32
|
Finger CE, Moreno-Gonzalez I, Gutierrez A, Moruno-Manchon JF, McCullough LD. Age-related immune alterations and cerebrovascular inflammation. Mol Psychiatry 2022; 27:803-818. [PMID: 34711943 PMCID: PMC9046462 DOI: 10.1038/s41380-021-01361-1] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 09/20/2021] [Accepted: 10/12/2021] [Indexed: 12/11/2022]
Abstract
Aging is associated with chronic systemic inflammation, which contributes to the development of many age-related diseases, including vascular disease. The world's population is aging, leading to an increasing prevalence of both stroke and vascular dementia. The inflammatory response to ischemic stroke is critical to both stroke pathophysiology and recovery. Age is a predictor of poor outcomes after stroke. The immune response to stroke is altered in aged individuals, which contributes to the disparate outcomes between young and aged patients. In this review, we describe the current knowledge of the effects of aging on the immune system and the cerebral vasculature and how these changes alter the immune response to stroke and vascular dementia in animal and human studies. Potential implications of these age-related immune alterations on chronic inflammation in vascular disease outcome are highlighted.
Collapse
Affiliation(s)
- Carson E. Finger
- Department of Neurology, McGovern Medical School, UTHealth Science Center at Houston, Houston, TX USA
| | - Ines Moreno-Gonzalez
- Department of Neurology, McGovern Medical School, UTHealth Science Center at Houston, Houston, TX USA ,grid.10215.370000 0001 2298 7828Department of Cell Biology, Genetics and Physiology, Instituto de Investigacion Biomedica de Malaga-IBIMA, Faculty of Sciences, Malaga University, Malaga, Spain ,grid.418264.d0000 0004 1762 4012Biomedical Research Networking Center on Neurodegenerative Diseases (CIBERNED), Malaga, Spain
| | - Antonia Gutierrez
- grid.10215.370000 0001 2298 7828Department of Cell Biology, Genetics and Physiology, Instituto de Investigacion Biomedica de Malaga-IBIMA, Faculty of Sciences, Malaga University, Malaga, Spain ,grid.418264.d0000 0004 1762 4012Biomedical Research Networking Center on Neurodegenerative Diseases (CIBERNED), Malaga, Spain
| | - Jose Felix Moruno-Manchon
- Department of Neurology, McGovern Medical School, UTHealth Science Center at Houston, Houston, TX USA
| | - Louise D. McCullough
- Department of Neurology, McGovern Medical School, UTHealth Science Center at Houston, Houston, TX USA
| |
Collapse
|
33
|
Stevenson TK, Moore SJ, Murphy GG, Lawrence DA. Tissue Plasminogen Activator in Central Nervous System Physiology and Pathology: From Synaptic Plasticity to Alzheimer's Disease. Semin Thromb Hemost 2021; 48:288-300. [DOI: 10.1055/s-0041-1740265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
AbstractTissue plasminogen activator's (tPA) fibrinolytic function in the vasculature is well-established. This specific role for tPA in the vasculature, however, contrasts with its pleiotropic activities in the central nervous system. Numerous physiological and pathological functions have been attributed to tPA in the central nervous system, including neurite outgrowth and regeneration; synaptic and spine plasticity; neurovascular coupling; neurodegeneration; microglial activation; and blood–brain barrier permeability. In addition, multiple substrates, both plasminogen-dependent and -independent, have been proposed to be responsible for tPA's action(s) in the central nervous system. This review aims to dissect a subset of these different functions and the different molecular mechanisms attributed to tPA in the context of learning and memory. We start from the original research that identified tPA as an immediate-early gene with a putative role in synaptic plasticity to what is currently known about tPA's role in a learning and memory disorder, Alzheimer's disease. We specifically focus on studies demonstrating tPA's involvement in the clearance of amyloid-β and neurovascular coupling. In addition, given that tPA has been shown to regulate blood–brain barrier permeability, which is perturbed in Alzheimer's disease, this review also discusses tPA-mediated vascular dysfunction and possible alternative mechanisms of action for tPA in Alzheimer's disease pathology.
Collapse
Affiliation(s)
- Tamara K. Stevenson
- Michigan Neuroscience Institute, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Molecular and Integrative Physiology, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - Shannon J. Moore
- Michigan Neuroscience Institute, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Molecular and Integrative Physiology, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - Geoffrey G. Murphy
- Michigan Neuroscience Institute, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Molecular and Integrative Physiology, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - Daniel A. Lawrence
- Department of Molecular and Integrative Physiology, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
34
|
Evans LE, Taylor JL, Smith CJ, Pritchard HAT, Greenstein AS, Allan SM. Cardiovascular co-morbidities, inflammation and cerebral small vessel disease. Cardiovasc Res 2021; 117:2575-2588. [PMID: 34499123 DOI: 10.1093/cvr/cvab284] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Indexed: 12/15/2022] Open
Abstract
Cerebral small vessel disease (cSVD) is the most common cause of vascular cognitive impairment and affects all levels of the brain's vasculature. Features include diverse structural and functional changes affecting small arteries and capillaries that lead to a decline in cerebral perfusion. Due to an aging population, incidence of cerebral small vessel disease (cSVD) is continually rising. Despite its prevalence and its ability to cause multiple debilitating illnesses, such as stroke and dementia, there are currently no therapeutic strategies for the treatment of cSVD. In the healthy brain, interactions between neuronal, vascular and inflammatory cells are required for normal functioning. When these interactions are disturbed, chronic pathological inflammation can ensue. The interplay between cSVD and inflammation has attracted much recent interest and this review discusses chronic cardiovascular diseases, particularly hypertension, and explores how the associated inflammation may impact on the structure and function of the small arteries of the brain in cSVD. Molecular approaches in animal studies are linked to clinical outcomes in patients and novel hypotheses regarding inflammation and cSVD are proposed that will hopefully stimulate further discussion and study in this important area.
Collapse
Affiliation(s)
- Lowri E Evans
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom.,Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
| | - Jade L Taylor
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom.,Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
| | - Craig J Smith
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom.,Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK.,Manchester Centre for Clinical Neurosciences, Manchester Academic Health Science Centre, Salford Royal Hospital, Manchester Academic Health Sciences Centre (MAHSC)
| | - Harry A T Pritchard
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom.,Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
| | - Adam S Greenstein
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom.,Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
| | - Stuart M Allan
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK.,Division of Neuroscience and Experimental Psychology, The University of Manchester, Manchester, UK
| |
Collapse
|
35
|
Chen C, Huang T, Zhai X, Ma Y, Xie L, Lu B, Zhang Y, Li Y, Chen Z, Yin J, Li P. Targeting neutrophils as a novel therapeutic strategy after stroke. J Cereb Blood Flow Metab 2021; 41:2150-2161. [PMID: 33691513 PMCID: PMC8393299 DOI: 10.1177/0271678x211000137] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Stroke is followed by an intricate immune interaction involving the engagement of multiple immune cells, including neutrophils. As one of the first responders recruited to the brain, the crucial roles of neutrophils in the ischemic brain damage are receiving increasing attention in recent years. Notably, neutrophils are not homogenous, and yet there is still a lack of full knowledge about the extent and impact of neutrophil heterogeneity. The biological understanding of the neutrophil response to both innate and pathological conditions is rapidly evolving as single-cell-RNA sequencing uncovers overall neutrophil profiling across maturation and differentiation contexts. In this review, we scrutinize the latest research that points to the multifaceted role of neutrophils in different conditions and summarize the regulatory signals that may determine neutrophil diversity. In addition, we list several potential targets or therapeutic strategies targeting neutrophils to limit brain damage following ischemic stroke.
Collapse
Affiliation(s)
- Chen Chen
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tingting Huang
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaozhu Zhai
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yezhi Ma
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lv Xie
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bingwei Lu
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yueman Zhang
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Li
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zengai Chen
- Department of Radiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiemin Yin
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peiying Li
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
36
|
Ringland C, Schweig JE, Eisenbaum M, Paris D, Ait-Ghezala G, Mullan M, Crawford F, Abdullah L, Bachmeier C. MMP9 modulation improves specific neurobehavioral deficits in a mouse model of Alzheimer's disease. BMC Neurosci 2021; 22:39. [PMID: 34034683 PMCID: PMC8152085 DOI: 10.1186/s12868-021-00643-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 05/10/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Matrix metallopeptidase 9 (MMP9) has been implicated in a variety of neurological disorders, including Alzheimer's disease (AD), where MMP9 levels are elevated in the brain and cerebrovasculature. Previously our group demonstrated apolipoprotein E4 (apoE4) was less efficient in regulating MMP9 activity in the brain than other apoE isoforms, and that MMP9 inhibition facilitated beta-amyloid (Aβ) elimination across the blood-brain barrier (BBB) METHODS: In the current studies, we evaluated the impact of MMP9 modulation on Aβ disposition and neurobehavior in AD using two approaches, (1) pharmacological inhibition of MMP9 with SB-3CT in apoE4 x AD (E4FAD) mice, and (2) gene deletion of MMP9 in AD mice (MMP9KO/5xFAD) RESULTS: Treatment with the MMP9 inhibitor SB-3CT in E4FAD mice led to reduced anxiety compared to placebo using the elevated plus maze. Deletion of the MMP9 gene in 5xFAD mice also reduced anxiety using the open field test, in addition to improving sociability and social recognition memory, particularly in male mice, as assessed through the three-chamber task, indicating certain behavioral alterations in AD may be mediated by MMP9. However, neither pharmacological inhibition of MMP9 or gene deletion of MMP9 affected spatial learning or memory in the AD animals, as determined through the radial arm water maze. Moreover, the effect of MMP9 modulation on AD neurobehavior was not due to changes in Aβ disposition, as both brain and plasma Aβ levels were unchanged in the SB-3CT-treated E4FAD animals and MMP9KO/AD mice compared to their respective controls. CONCLUSIONS In total, while MMP9 inhibition did improve specific neurobehavioral deficits associated with AD, such as anxiety and social recognition memory, modulation of MMP9 did not alter spatial learning and memory or Aβ tissue levels in AD animals. While targeting MMP9 may represent a therapeutic strategy to mitigate aspects of neurobehavioral decline in AD, further work is necessary to understand the nature of the relationship between MMP9 activity and neurological dysfunction.
Collapse
Affiliation(s)
- Charis Ringland
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
- The Open University, Milton Keynes, UK
| | | | - Maxwell Eisenbaum
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
- The Open University, Milton Keynes, UK
| | - Daniel Paris
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
| | - Ghania Ait-Ghezala
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
- The Open University, Milton Keynes, UK
| | - Michael Mullan
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
- The Open University, Milton Keynes, UK
| | - Fiona Crawford
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
- The Open University, Milton Keynes, UK
- James A. Haley Veterans' Hospital, Tampa, FL, USA
| | - Laila Abdullah
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
- The Open University, Milton Keynes, UK
- James A. Haley Veterans' Hospital, Tampa, FL, USA
| | - Corbin Bachmeier
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA.
- The Open University, Milton Keynes, UK.
- Bay Pines VA Healthcare System, Bay Pines, FL, USA.
| |
Collapse
|
37
|
Cao Y, Wang F, Wang Y, Long J. Agomelatine prevents macrophage infiltration and brain endothelial cell damage in a stroke mouse model. Aging (Albany NY) 2021; 13:13548-13559. [PMID: 33839700 PMCID: PMC8202857 DOI: 10.18632/aging.202836] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 11/15/2020] [Indexed: 01/01/2023]
Abstract
Background and purpose: Ischemic/reperfusions are regarded as the clinical consensus for stroke treatment, which results in secondary injury of brain tissues. Increased blood-brain barrier (BBB) permeability and infiltration of inflammatory cells are responsible for the ischemic/reperfusion injury. In the present study, we aimed to investigate the effects of Agomelatine on brain ischemic/reperfusions injury and the underlying mechanism. Methods: MCAO model was established in mice. The expressions of CD68 and claudin-5 in the cerebral cortex were determined using an immunofluorescence assay. Brain permeability was evaluated using Evans blue staining assay. A two-chamber and two-cell trans-well assay was used to detect the migration ability of macrophages through endothelial cells. The expression levels of claudin-5 and MCP-1 in the endothelial cells were determined using qRT-PCR and ELISA. Results: CD68 was found to be up-regulated in the cerebral cortex of MCAO mice but was down-regulated by treatment with Agomelatine. The expression level of down-regulated claudin-5 in the cerebral cortex of MCAO mice was significantly suppressed by Agomelatine. Deeper staining of Evans blue was found in the MCAO group, which was however faded significantly in the Agomelatine treated MCAO mice. The migrated macrophages were significantly increased by hypoxia incubation but were greatly suppressed by the introduction of Agomelatine. The down-regulated claudin-5 by hypoxic incubation in endothelial cells was up-regulated by treatment with Agomelatine. Furthermore, the increased expression of MCP-1 in endothelial cells under hypoxic conditions was significantly inhibited by Agomelatine. Conclusion: Agomelatine prevents macrophage infiltration and brain endothelial cell damage in a stroke mouse model.
Collapse
Affiliation(s)
- Yiqiang Cao
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Fei Wang
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Yonggang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Jiang Long
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| |
Collapse
|
38
|
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
|
39
|
Takase H, Regenhardt RW. Motor tract reorganization after acute central nervous system injury: a translational perspective. Neural Regen Res 2021; 16:1144-1149. [PMID: 33269763 PMCID: PMC8224132 DOI: 10.4103/1673-5374.300330] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Acute central nervous system injuries are among the most common causes of disability worldwide, with widespread social and economic implications. Motor tract injury accounts for the majority of this disability; therefore, there is impetus to understand mechanisms underlying the pathophysiology of injury and subsequent reorganization of the motor tract that may lead to recovery. After acute central nervous system injury, there are changes in the microenvironment and structure of the motor tract. For example, ischemic stroke involves decreased local blood flow and tissue death from lack of oxygen and nutrients. Traumatic injury, in contrast, causes stretching and shearing injury to microstructures, including myelinated axons and their surrounding vessels. Both involve blood-brain barrier dysfunction, which is an important initial event. After acute central nervous system injury, motor tract reorganization occurs in the form of cortical remapping in the gray matter and axonal regeneration and rewiring in the white matter. Cortical remapping involves one cortical region taking on the role of another. cAMP-response-element binding protein is a key transcription factor that can enhance plasticity in the peri-infarct cortex. Axonal regeneration and rewiring depend on complex cell-cell interactions between axons, oligodendrocytes, and other cells. The RhoA/Rho-associated coiled-coil containing kinase signaling pathway plays a central role in axon growth/regeneration through interactions with myelin-derived axonal growth inhibitors and regulation of actin cytoskeletal dynamics. Oligodendrocytes and their precursors play a role in myelination, and neurons are involved through their voltage-gated calcium channels. Understanding the pathophysiology of injury and the biology of motor tract reorganization may allow the development of therapies to enhance recovery after acute central nervous system injury. These include targeted rehabilitation, novel pharmacotherapies, such as growth factors and axonal growth inhibitor blockade, and the implementation of neurotechnologies, such as central nervous system stimulators and robotics. The translation of these advances depends on careful alignment of preclinical studies and human clinical trials. As experimental data mount, the future is one of optimism.
Collapse
Affiliation(s)
- Hajime Takase
- Department of Neurosurgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan; Department of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Robert W Regenhardt
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
40
|
Sharma R, Kumar V, Logani A, Chawla A, Mir RA, Sharma S, Kalaivani M. Association between concentration of active MMP-9 in pulpal blood and pulpotomy outcome in permanent mature teeth with irreversible pulpitis - a preliminary study. Int Endod J 2020; 54:479-489. [PMID: 33128238 DOI: 10.1111/iej.13437] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/26/2020] [Accepted: 10/27/2020] [Indexed: 12/11/2022]
Abstract
AIM To investigate the correlation between the concentration of active-matrix metalloproteinases-9 (aMMP-9) in pulpal blood and the outcome of pulpotomy in mature permanent teeth with symptomatic irreversible pulpitis (SIP). METHODOLOGY Forty permanent molar teeth with a clinical diagnosis of SIP and normal apical tissues with periapical index (PAI) score ≤ 2 and ten permanent teeth (8 molars and two premolars) with a diagnosis of normal pulp that required root canal treatment for prosthetic reasons from patients between the ages of 15-35 years were recruited. All clinical procedures were performed under local anaesthesia and rubber dam isolation. After access opening, the coronal pulp tissue was amputated up to the canal orifice. A 100 μL volume of the pulpal blood was collected using a micropipette and transported to the laboratory. Sodium hypochlorite (2.5 %) was used as a haemostatic agent, and mineral trioxide aggregate (MTA) was used as the pulp capping material. The tooth was restored with composite at the same visit. Teeth with normal pulps were treated with single-visit root canal treatment. Patients with pulpotomy were recalled at 6 and 12 months. Outcome assessment of teeth with pulpotomy was carried out at 12 months and was categorized as success (asymptomatic patients with PAI score ≤ 2) or failure (symptomatic patients or PAI score ≥ 3). Quantification of aMMP-9 in pulpal blood was achieved using a fluorometric assay. The following statistical analyses were performed to assess the data: t-test, Fisher's exact test, kappa coefficient, non-parametric test, Wilcoxon rank-sum test, Spearman rank correlation test and receiver operating characteristic curve (ROC). RESULT The success rate of pulpotomy was 88 % at 12-months. There was a significant difference between the median concentrations of aMMP-9 in pulpal blood of teeth with normal pulps (52 (12-96) ng mL-1 :) and SIP (193.3 (25.8-607.7) ng mL-1 :) (P = 0.0003) and successful (132.3 (25.8-548.3) ng mL-1 :) and failed cases (512.4 (334.8-607.7 ng mL-1 :) (P = 0.0015) of MTA pulpotomy. A significant association was established between aMMP-9 concentration and outcome of pulpotomy. The area under the receiver operating characteristics curve (0.9484, 95%CI) suggested excellent discriminatory power of aMMP-9 concentration in pulpal blood to predict the pulpotomy outcome. CONCLUSION The pulpal blood concentration of aMMP-9 was significantly associated with the outcome of pulpotomy in teeth with symptomatic irreversible pulpitis, where it may be used as a potential prognostic biomarker.
Collapse
Affiliation(s)
- R Sharma
- Division of Conservative Dentistry and Endodontics, Centre for Dental Education and Research, All India Institute of Medical Sciences, New Delhi, India
| | - V Kumar
- Division of Conservative Dentistry and Endodontics, Centre for Dental Education and Research, All India Institute of Medical Sciences, New Delhi, India
| | - A Logani
- Division of Conservative Dentistry and Endodontics, Centre for Dental Education and Research, All India Institute of Medical Sciences, New Delhi, India
| | - A Chawla
- Division of Conservative Dentistry and Endodontics, Centre for Dental Education and Research, All India Institute of Medical Sciences, New Delhi, India
| | - R A Mir
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - S Sharma
- Division of Conservative Dentistry and Endodontics, Centre for Dental Education and Research, All India Institute of Medical Sciences, New Delhi, India
| | - M Kalaivani
- Department of Biostatistics, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
41
|
Tuttolomondo A, Puleo MG, Velardo MC, Corpora F, Daidone M, Pinto A. Molecular Biology of Atherosclerotic Ischemic Strokes. Int J Mol Sci 2020; 21:ijms21249372. [PMID: 33317034 PMCID: PMC7763838 DOI: 10.3390/ijms21249372] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 12/03/2020] [Indexed: 02/07/2023] Open
Abstract
Among the causes of global death and disability, ischemic stroke (also known as cerebral ischemia) plays a pivotal role, by determining the highest number of worldwide mortality, behind cardiomyopathies, affecting 30 million people. The etiopathogenetic burden of a cerebrovascular accident could be brain ischemia (~80%) or intracranial hemorrhage (~20%). The most common site when ischemia occurs is the one is perfused by middle cerebral arteries. Worse prognosis and disablement consequent to brain damage occur in elderly patients or affected by neurological impairment, hypertension, dyslipidemia, and diabetes. Since, in the coming years, estimates predict an exponential increase of people who have diabetes, the disease mentioned above constitutes together with stroke a severe social and economic burden. In diabetic patients after an ischemic stroke, an exorbitant activation of inflammatory molecular pathways and ongoing inflammation is responsible for more severe brain injury and impairment, promoting the advancement of ischemic stroke and diabetes. Considering that the ominous prognosis of ischemic brain damage could by partially clarified by way of already known risk factors the auspice would be modifying poor outcome in the post-stroke phase detecting novel biomolecules associated with poor prognosis and targeting them for revolutionary therapeutic strategies.
Collapse
|
42
|
Kumar L, Planas-Iglesias J, Harms C, Kamboj S, Wright D, Klein-Seetharaman J, Sarkar SK. Activity-dependent interdomain dynamics of matrix metalloprotease-1 on fibrin. Sci Rep 2020; 10:20615. [PMID: 33244162 PMCID: PMC7692495 DOI: 10.1038/s41598-020-77699-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 11/13/2020] [Indexed: 01/18/2023] Open
Abstract
The roles of protein conformational dynamics and allostery in function are well-known. However, the roles that interdomain dynamics have in function are not entirely understood. We used matrix metalloprotease-1 (MMP1) as a model system to study the relationship between interdomain dynamics and activity because MMP1 has diverse substrates. Here we focus on fibrin, the primary component of a blood clot. Water-soluble fibrinogen, following cleavage by thrombin, self-polymerize to form water-insoluble fibrin. We studied the interdomain dynamics of MMP1 on fibrin without crosslinks using single-molecule Forster Resonance Energy Transfer (smFRET). We observed that the distance between the catalytic and hemopexin domains of MMP1 increases or decreases as the MMP1 activity increases or decreases, respectively. We modulated the activity using (1) an active site mutant (E219Q) of MMP1, (2) MMP9, another member of the MMP family that increases the activity of MMP1, and (3) tetracycline, an inhibitor of MMP1. We fitted the histograms of smFRET values to a sum of two Gaussians and the autocorrelations to an exponential and power law. We modeled the dynamics as a two-state Poisson process and calculated the kinetic rates from the histograms and autocorrelations. Activity-dependent interdomain dynamics may enable allosteric control of the MMP1 function.
Collapse
Affiliation(s)
- Lokender Kumar
- Department of Physics, Colorado School of Mines, 1500 Illinois Street, Golden, CO, 80401, USA
| | - Joan Planas-Iglesias
- Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK
- Loschmidt Laboratories, Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5/A13, 625 00, Brno, Czech Republic
| | - Chase Harms
- Department of Physics, Colorado School of Mines, 1500 Illinois Street, Golden, CO, 80401, USA
| | - Sumaer Kamboj
- Department of Physics, Colorado School of Mines, 1500 Illinois Street, Golden, CO, 80401, USA
| | - Derek Wright
- Department of Physics, Colorado School of Mines, 1500 Illinois Street, Golden, CO, 80401, USA
| | - Judith Klein-Seetharaman
- Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK
- Department of Chemistry, Colorado School of Mines, 1500 Illinois Street, Golden, CO, 80401, USA
| | - Susanta K Sarkar
- Department of Physics, Colorado School of Mines, 1500 Illinois Street, Golden, CO, 80401, USA.
| |
Collapse
|
43
|
Douglas AS, Shearer JA, Okolo A, Pandit A, Gilvarry M, Doyle KM. The Relationship Between Cerebral Reperfusion And Regional Expression Of Matrix Metalloproteinase-9 In Rat Brain Following Focal Cerebral Ischemia. Neuroscience 2020; 453:256-265. [PMID: 33220187 DOI: 10.1016/j.neuroscience.2020.10.034] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 10/23/2020] [Accepted: 10/27/2020] [Indexed: 12/29/2022]
Abstract
We investigated the effect of full and partial mechanical reperfusion on MMP-9 expression in rat brain following middle cerebral artery occlusion, mimicking mechanical thrombectomy. Using percentage hemispheric lesion volume and oedema as measures, partial reperfusion reduced extent of brain damage caused by MCA occlusion, but the protective effect was less pronounced than with complete reperfusion. Using ELISA quantification in fresh frozen tissue, confirmed by immunofluorescence in perfusion fixed tissue, increased MMP-9 expression was observed in infarcted tissue. MMP-9 was increased in lesioned tissue of the anterior and posterior temporal cortex and underlying striatal tissue, but also the normal appearing frontal cortex. No significant increase in MMP-9 in the hippocampus was observed, nor in the unlesioned contralateral hemisphere. Both partial reperfusion and full reperfusion reduced the regional MMP expression significantly. The highest levels of MMP-9 were observed in lesioned brain regions in the non-reperfused group. MMP-9 expression was evident in microvessels and in neuronal cell bodies of affected tissue. This study shows that MMP-9 brain levels are reduced relative to the extent of reperfusion. These observations suggest targeting early increases in MMP-9 expression as a possible neuroprotective therapeutic strategy and highlight the rat MCA occlusion model as an ideal model in which to study candidate therapeutics.
Collapse
Affiliation(s)
- A S Douglas
- Department of Physiology and Galway Neuroscience Centre, School of Medicine, National University of Ireland, Galway, Ireland; CÚRAM-Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland.
| | - J A Shearer
- Department of Physiology and Galway Neuroscience Centre, School of Medicine, National University of Ireland, Galway, Ireland; CÚRAM-Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - A Okolo
- Department of Physiology and Galway Neuroscience Centre, School of Medicine, National University of Ireland, Galway, Ireland; CÚRAM-Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - A Pandit
- CÚRAM-Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | | | - K M Doyle
- Department of Physiology and Galway Neuroscience Centre, School of Medicine, National University of Ireland, Galway, Ireland; CÚRAM-Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
44
|
Takarada-Iemata M. Roles of N-myc downstream-regulated gene 2 in the central nervous system: molecular basis and relevance to pathophysiology. Anat Sci Int 2020; 96:1-12. [PMID: 33174183 DOI: 10.1007/s12565-020-00587-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 10/30/2020] [Indexed: 12/12/2022]
Abstract
N-myc downstream-regulated gene 2 (NDRG2) is a member of the NDRG family, whose members have multiple functions in cell proliferation, differentiation, and stress responses. NDRG2 is widely distributed in the central nervous system and is uniquely expressed by astrocytes; however, its role in brain function remains elusive. The clinical relevance of NDRG2 and the molecular mechanisms in which it participates have been reported by studies using cultured cells and specimens of patients with neurological disorders. In recent years, genetic tools, including several lines of Ndrg2-knockout mice and virus-mediated gene transfer, have improved understanding of the roles of NDRG2 in vivo. This review aims to provide an update of recent growing in vivo evidence that NDRG2 is involved in brain function, focusing on research of Ndrg2-knockout mice with neurological disorders such as brain tumors, chronic neurodegenerative diseases, and acute brain insults including brain injury and cerebral stroke. These studies demonstrate that NDRG2 plays diverse roles in the regulation of astrocyte reactivity, blood-brain barrier integrity, and glutamate excitotoxicity. Further elucidation of the roles of NDRG2 and their molecular basis may provide novel therapeutic approaches for various neurological disorders.
Collapse
Affiliation(s)
- Mika Takarada-Iemata
- Department of Neuroanatomy, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan.
| |
Collapse
|
45
|
Chao CM, Chen CL, Niu KC, Lin CH, Tang LY, Lin LS, Chang CP. Hypobaric hypoxia preconditioning protects against hypothalamic neuron apoptosis in heat-exposed rats by reversing hypothalamic overexpression of matrix metalloproteinase-9 and ischemia. Int J Med Sci 2020; 17:2622-2634. [PMID: 33162790 PMCID: PMC7645337 DOI: 10.7150/ijms.47560] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 08/24/2020] [Indexed: 12/03/2022] Open
Abstract
Background: Hypoxia-inducible factor-1α (HIF-1α), heat shock protein-72 (HSP-72), hemeoxygenase-1 (HO-1), and matrix metalloproteinase-9 (MMP-9) have been identified as potential therapeutic targets in the brain for cerebral ischemia. To elucidate their underlying mechanisms, we first aimed to ascertain whether these proteins participate in the pathogenesis of heat-induced ischemic damage to the hypothalamus of rats. Second, we investigated whether hypobaric hypoxia preconditioning (HHP) attenuates heat-induced hypothalamic ischemic/hypoxic injury by modulating these proteins in situ. Methods: Anesthetized rats treated with or without HHP were subjected to heat stress. Hypothalamic ischemic/hypoxic damage was evaluated by measuring hypothalamic levels of cerebral blood flow (CBF), partial oxygen pressure (PO2), and hypothalamic temperature via an implanted probe. Hypothalamic apoptotic neurons were counted by measuring the number of NeuN/caspase-3/DAPI triple-stained cells. Hypothalamic protein expression of HIF-1α, HSP-72, HO-1, and MMP-9 was determined biochemically. Results: Before the start of the thermal experiments, rats were subjected to 5 hours of HHP (0.66 ATA or 18.3% O2) daily for 5 consecutive days per week for 2 weeks, which led to significant loss of body weight, reduced brown adipose tissue (BAT) wet weight and decreased body temperature. The animals were then subjected to thermal studies. Twenty minutes after heat stress, heat-exposed rats not treated with HHP displayed significantly higher core and hypothalamic temperatures, hypothalamic MMP-9 levels, and numbers of hypothalamic apoptotic neurons but significantly lower mean blood pressure, hypothalamic blood flow, and PO2 values than control rats not exposed to heat. In heat-exposed rats, HHP significantly increased the hypothalamic levels of HIF-1α, HSP-72, and HO-1 but significantly alleviated body and hypothalamic hyperthermia, hypotension, hypothalamic ischemia, hypoxia, neuronal apoptosis and degeneration. Conclusions: HHP may protect against hypothalamic ischemic/hypoxic injury and overexpression of MMP-9 by upregulating the hypothalamic expression of HIF-1α, HSP-72, and HO-1 in rats subjected to heatstroke.
Collapse
Affiliation(s)
- Chien-Ming Chao
- Department of Intensive Care Medicine, Chi Mei Medical Center, Liouying, Tainan, Taiwan
- Department of Nursing, Min-Hwei College of Health Care Management, Tainan, Taiwan
| | - Chun-Liang Chen
- Department of Gastroenterology and General Surgery, Chi Mei Medical Hospital, Chiali, Tainan, Taiwan
| | - Ko-Chi Niu
- Department of Hyperbaric Oxygen, Chi Mei Medical Center, Tainan, Taiwan
| | - Cheng-Hsien Lin
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| | - Ling-Yu Tang
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
| | - Lieh-Sheng Lin
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei, Taiwan
| | - Ching-Ping Chang
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
| |
Collapse
|
46
|
Blood substitution therapy rescues the brain of mice from ischemic damage. Nat Commun 2020; 11:4078. [PMID: 32843630 PMCID: PMC7447645 DOI: 10.1038/s41467-020-17930-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 07/22/2020] [Indexed: 12/15/2022] Open
Abstract
Acute stroke causes complex, pathological, and systemic responses that have not been treatable by any single medication. In this study, using a murine transient middle cerebral artery occlusion stroke model, a novel therapeutic strategy is proposed, where blood replacement (BR) robustly reduces infarctions and improves neurological deficits in mice. Our analyses of immune cell subsets suggest that BR therapy substantially decreases neutrophils in blood following a stroke. Electrochemiluminescence detection demonstrates that BR therapy reduces cytokine storm in plasma and ELISA demonstrates reduced levels of matrix metalloproteinase-9 (MMP-9) in the plasma and brains at different time points post-stroke. Further, we have demonstrated that the addition of MMP-9 to the blood diminishes the protective effect of the BR therapy. Our study is the first to show that BR therapy leads to profoundly improved stroke outcomes in mice and that the improved outcomes are mediated via MMP-9. These results offer new insights into the mechanisms of stroke damage. Acute stroke causes complex, pathological, and systemic responses which remain challenging to treat. Here, the authors show that substituting the blood of stroke model mice with whole-blood from naive healthy donor mice reduces infarct volume and improves neurological deficits.
Collapse
|
47
|
Kumar L, Nash A, Harms C, Planas-Iglesias J, Wright D, Klein-Seetharaman J, Sarkar SK. Allosteric Communications between Domains Modulate the Activity of Matrix Metalloprotease-1. Biophys J 2020; 119:360-374. [PMID: 32585130 PMCID: PMC7376139 DOI: 10.1016/j.bpj.2020.06.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 05/30/2020] [Accepted: 06/09/2020] [Indexed: 11/23/2022] Open
Abstract
An understanding of the structure-dynamics relationship is essential for understanding how a protein works. Prior research has shown that the activity of a protein correlates with intradomain dynamics occurring at picosecond to millisecond timescales. However, the correlation between interdomain dynamics and the function of a protein is poorly understood. Here, we show that communications between the catalytic and hemopexin domains of matrix metalloprotease-1 (MMP1) on type 1 collagen fibrils correlate with its activity. Using single-molecule Förster resonance energy transfer, we identified functionally relevant open conformations in which the two MMP1 domains are well separated, which were significantly absent for catalytically inactive point mutant (E219Q) of MMP1 and could be modulated by an inhibitor or an enhancer of activity. The observed relevance of open conformations resolves the debate about the roles of open and closed MMP1 structures in function. We fitted the histograms of single-molecule Förster resonance energy transfer values to a sum of two Gaussians and the autocorrelations to an exponential and power law. We used a two-state Poisson process to describe the dynamics and calculate the kinetic rates from the fit parameters. All-atom and coarse-grained simulations reproduced some of the experimental features and revealed substrate-dependent MMP1 dynamics. Our results suggest that an interdomain separation facilitates opening up the catalytic pocket so that the collagen chains come closer to the MMP1 active site. Coordination of functional conformations at different parts of MMP1 occurs via allosteric communications that can take place via interactions mediated by collagen even if the linker between the domains is absent. Modeling dynamics as a Poisson process enables connecting the picosecond timescales of molecular dynamics simulations with the millisecond timescales of single-molecule measurements. Water-soluble MMP1 interacting with water-insoluble collagen fibrils poses challenges for biochemical studies that the single-molecule tracking can overcome for other insoluble substrates. Interdomain communications are likely important for multidomain proteins.
Collapse
Affiliation(s)
- Lokender Kumar
- Department of Physics, Colorado School of Mines, Golden, Colorado
| | - Anthony Nash
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Chase Harms
- Department of Physics, Colorado School of Mines, Golden, Colorado
| | - Joan Planas-Iglesias
- Warwick Medical School, University of Warwick, Coventry, United Kingdom; Loschmidt Laboratories, Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Derek Wright
- Department of Physics, Colorado School of Mines, Golden, Colorado
| | - Judith Klein-Seetharaman
- Warwick Medical School, University of Warwick, Coventry, United Kingdom; Department of Chemistry, Colorado School of Mines, Golden, Colorado
| | - Susanta K Sarkar
- Department of Physics, Colorado School of Mines, Golden, Colorado.
| |
Collapse
|
48
|
González-Nieto D, Fernández-Serra R, Pérez-Rigueiro J, Panetsos F, Martinez-Murillo R, Guinea GV. Biomaterials to Neuroprotect the Stroke Brain: A Large Opportunity for Narrow Time Windows. Cells 2020; 9:E1074. [PMID: 32357544 PMCID: PMC7291200 DOI: 10.3390/cells9051074] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/20/2020] [Accepted: 04/23/2020] [Indexed: 12/14/2022] Open
Abstract
Ischemic stroke represents one of the most prevalent pathologies in humans and is a leading cause of death and disability. Anti-thrombolytic therapy with tissue plasminogen activator (t-PA) and surgical thrombectomy are the primary treatments to recanalize occluded vessels and normalize the blood flow in ischemic and peri-ischemic regions. A large majority of stroke patients are refractory to treatment or are not eligible due to the narrow time window of therapeutic efficacy. In recent decades, we have significantly increased our knowledge of the molecular and cellular mechanisms that inexorably lead to progressive damage in infarcted and peri-lesional brain areas. As a result, promising neuroprotective targets have been identified and exploited in several stroke models. However, these considerable advances have been unsuccessful in clinical contexts. This lack of clinical translatability and the emerging use of biomaterials in different biomedical disciplines have contributed to developing a new class of biomaterial-based systems for the better control of drug delivery in cerebral disorders. These systems are based on specific polymer formulations structured in nanoparticles and hydrogels that can be administered through different routes and, in general, bring the concentrations of drugs to therapeutic levels for prolonged times. In this review, we first provide the general context of the molecular and cellular mechanisms impaired by cerebral ischemia, highlighting the role of excitotoxicity, inflammation, oxidative stress, and depolarization waves as the main pathways and targets to promote neuroprotection avoiding neuronal dysfunction. In the second part, we discuss the versatile role played by distinct biomaterials and formats to support the sustained administration of particular compounds to neuroprotect the cerebral tissue at risk of damage.
Collapse
Affiliation(s)
- Daniel González-Nieto
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28040 Madrid, Spain; (R.F.-S.); (J.P.-R.); (G.V.G.)
- Departamento de Tecnología Fotónica y Bioingeniería, ETSI Telecomunicaciones, Universidad Politécnica de Madrid, 28040 Madrid, Spain
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Rocío Fernández-Serra
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28040 Madrid, Spain; (R.F.-S.); (J.P.-R.); (G.V.G.)
- Departamento de Tecnología Fotónica y Bioingeniería, ETSI Telecomunicaciones, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - José Pérez-Rigueiro
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28040 Madrid, Spain; (R.F.-S.); (J.P.-R.); (G.V.G.)
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - Fivos Panetsos
- Neurocomputing and Neurorobotics Research Group: Faculty of Biology and Faculty of Optics, Universidad Complutense de Madrid, 28040 Madrid, Spain;
- Brain Plasticity Group, Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | | | - Gustavo V. Guinea
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28040 Madrid, Spain; (R.F.-S.); (J.P.-R.); (G.V.G.)
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| |
Collapse
|
49
|
Kestner RI, Mayser F, Vutukuri R, Hansen L, Günther S, Brunkhorst R, Devraj K, Pfeilschifter W. Gene Expression Dynamics at the Neurovascular Unit During Early Regeneration After Cerebral Ischemia/Reperfusion Injury in Mice. Front Neurosci 2020; 14:280. [PMID: 32300291 PMCID: PMC7142359 DOI: 10.3389/fnins.2020.00280] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 03/11/2020] [Indexed: 12/21/2022] Open
Abstract
With increasing distribution of endovascular stroke therapies, transient middle cerebral artery occlusion (tMCAO) in mice now more than ever depicts a relevant patient population with recanalized M1 occlusion. In this case, the desired therapeutic effect of blood flow restauration is accompanied by breakdown of the blood-brain barrier (BBB) and secondary reperfusion injury. The aim of this study was to elucidate short and intermediate-term transcriptional patterns and the involved pathways covering the different cellular players at the neurovascular unit after transient large vessel occlusion. To achieve this, male C57Bl/6J mice were treated according to an intensive post-stroke care protocol after 60 min occlusion of the middle cerebral artery or sham surgery to allow a high survival rate. After 24 h or 7 days, RNA from microvessel fragments from the ipsilateral and the contralateral hemispheres was isolated and used for mRNA sequencing. Bioinformatic analyses allowed us to depict gene expression changes at two timepoints of neurovascular post-stroke injury and regeneration. We validated our dataset by quantitative real time PCR of BBB-associated targets with well-characterized post-stroke dynamics. Hence, this study provides a well-controlled transcriptome dataset of a translationally relevant mouse model 24 h and 7 days after stroke which might help to discover future therapeutic targets in cerebral ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Roxane-Isabelle Kestner
- Department of Neurology, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany.,Department of General Pharmacology and Toxicology, Pharmazentrum Frankfurt, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Franziska Mayser
- Department of Neurology, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany.,Department of General Pharmacology and Toxicology, Pharmazentrum Frankfurt, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Rajkumar Vutukuri
- Department of General Pharmacology and Toxicology, Pharmazentrum Frankfurt, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Lena Hansen
- Department of General Pharmacology and Toxicology, Pharmazentrum Frankfurt, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Stefan Günther
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Robert Brunkhorst
- Department of Neurology, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Kavi Devraj
- Department of General Pharmacology and Toxicology, Pharmazentrum Frankfurt, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany.,Institute of Neurology (Edinger Institute), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Waltraud Pfeilschifter
- Department of Neurology, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
50
|
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
|