1
|
Quan X, Liu C, Chen J, Li Y, Yuan Z, Zheng Y, Mok GSP, Wang R, Zhao Y. Neutrophil-Mimetic Upconversion Photosynthetic Nanosystem Derived from Microalgae for Targeted Treatment of Thromboembolic Stroke. ACS NANO 2024; 18:30307-30320. [PMID: 39465976 DOI: 10.1021/acsnano.4c06247] [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: 10/29/2024]
Abstract
Thromboembolic stroke constitutes the majority of brain strokes, resulting in elevated mortality and morbidity rates, as well as significant societal and economic burdens. Although intravenous thrombolysis serves as the standard clinical treatment, its narrow therapeutic window and the inflammatory response induced by tissue plasminogen activator (tPA) administration limit its efficacy. In the initial stages of stroke, the abrupt cessation of blood flow leads to an energy metabolism disorder, marked by a substantial decrease in adenosine triphosphate (ATP) and nicotinamide adenine dinucleotide phosphate (NADPH) levels, causing irreversible damage to neural cells. In this study, we introduce a neutrophil-mimetic, microalgae-derived upconversion photosynthetic nanosystem designed for targeted treatment of thromboembolic stroke. This system features upconversion nanoparticles coated with a thylakoid membrane and wrapped in an activated neutrophil membrane, further decorated with ROS-responsive thrombolytic tPA on its surface. The neutrophil-mimetic design facilitates high targeting specificity and accumulation at the thrombus site after intravenous administration. Upon exposure to elevated levels of reactive oxygen species (ROS) at the thrombus location, the nanosystem promptly demonstrated potent thrombolytic efficacy through the surface-modified tPA. Furthermore, near-infrared II (NIR-II) laser irradiation activated the generation of ATP and NADPH, which inhibited inflammatory cell infiltration, platelet activation, oxidative stress, and neuronal injury. This constructed nanoplatform not only showcases exceptional targeting efficiency at the stroke site and controllable release of the thrombolytic agent but also facilitates ATP/NADPH-mediated thrombolytic, anti-inflammatory, antioxidative stress, and neuroprotective effects. Additionally, it offers valuable insights into the potential therapeutic applications of microalgae-based derivatives in managing thromboembolic stroke.
Collapse
Affiliation(s)
- Xingping Quan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, SAR 999078, China
| | - Chang Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, SAR 999078, China
| | - Jinfen Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, SAR 999078, China
| | - Yiyang Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, SAR 999078, China
| | - Zhen Yuan
- Centre for Cognitive and Brain Sciences, University of Macau, Macau, SAR 999078, China
| | - Ying Zheng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, SAR 999078, China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macau, SAR 999078, China
| | - Greta S P Mok
- Department of Electrical and Computer Engineering, University of Macau, Macau, SAR 999078, China
| | - Ruibing Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, SAR 999078, China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macau, SAR 999078, China
| | - Yonghua Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, SAR 999078, China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macau, SAR 999078, China
| |
Collapse
|
2
|
Won SJ, Zhang Y, Butler NJ, Kim K, Mocanu E, Nzoutchoum OT, Lakkaraju R, Davis J, Ghosh S, Swanson RA. Stress hyperglycemia exacerbates inflammatory brain injury after stroke. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.14.594195. [PMID: 38798486 PMCID: PMC11118312 DOI: 10.1101/2024.05.14.594195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Post-stroke hyperglycemia occurs in 30% - 60% of ischemic stroke patients as part of the systemic stress response, but neither clinical evidence nor pre-clinical studies indicate whether post-stroke hyperglycemia affects stroke outcome. Here we investigated this issue using a mouse model of permanent ischemia. Mice were maintained either normoglycemic or hyperglycemic during the interval of 17 - 48 hours after ischemia onset. Post-stroke hyperglycemia was found to increase infarct volume, blood-brain barrier disruption, and hemorrhage formation, and to impair motor recovery. Post-stroke hyperglycemia also increased superoxide formation by peri-infarct microglia/macrophages. In contrast, post-stroke hyperglycemia did not increase superoxide formation or exacerbate motor impairment in p47 phox-/- mice, which cannot form an active superoxide-producing NADPH oxidase-2 complex. These results suggest that hyperglycemia occurring hours-to-days after ischemia can increase oxidative stress in peri-infarct tissues by fueling NADPH oxidase activity in reactive microglia/macrophages, and by this mechanism contribute to worsened functional outcome.
Collapse
|
3
|
Liu J, Zhang X, Xu Y, Zhang S. Regulation of Microglial Activation by Wnt/β-Catenin Signaling After Global Cerebral Ischemia in Mice. Mol Neurobiol 2024; 61:308-325. [PMID: 37607993 DOI: 10.1007/s12035-023-03557-8] [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: 08/05/2023] [Indexed: 08/24/2023]
Abstract
Microglia are immunocompetent cells in the central nervous system. Following cerebral ischemia, microglia will be rapidly activated and undergo proliferation, morphological transformation, and changes in gene expression and function. At present, the regulatory mechanisms of microglial activation following ischemia remain largely unclear. In this study, we took advantage of CX3CR1GFP/+ fluorescent mice and a global cerebral ischemia-reperfusion model to investigate the mechanisms of microglial activation following different degrees of global ischemia. Our results showed that the proliferation of microglia was gated by the degree of ischemia. Marked microglial de-ramification and proliferation were observed after 60 min of ischemia but not in transient ischemia (20 min). Immunohistology, qRT-PCR, and Western blotting analysis showed that microglial activation was accompanied with a reduction in Wnt/β-catenin signaling after cerebral ischemia. Downregulation of Wnt/β-catenin signaling using Wnt antagonist XAV939 during 20 min ischemia promoted microglial de-ramification and proliferation. In contrast, enhancing Wnt/β-catenin signaling using Wnt agonist LiCl during 60 min ischemia-reduced microglial de-ramification and proliferation. Importantly, we found that Wnt agonist inhibited inflammation in the ischemic brain and was conducive to animal behavioral recovery. Collectively, these data demonstrated that Wnt/β-catenin signaling played a key role in microglial activation following cerebral ischemia, and regulating microglial activation may be a potential therapeutic strategy for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Junru Liu
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, 730000, China
| | - Xinying Zhang
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, 730000, China
| | - Yanyi Xu
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, 730000, China
| | - Shengxiang Zhang
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, 730000, China.
| |
Collapse
|
4
|
Wu X, Li JR, Fu Y, Chen DY, Nie H, Tang ZP. From static to dynamic: live observation of the support system after ischemic stroke by two photon-excited fluorescence laser-scanning microscopy. Neural Regen Res 2023; 18:2093-2107. [PMID: 37056116 PMCID: PMC10328295 DOI: 10.4103/1673-5374.369099] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/21/2022] [Accepted: 01/13/2023] [Indexed: 02/17/2023] Open
Abstract
Ischemic stroke is one of the most common causes of mortality and disability worldwide. However, treatment efficacy and the progress of research remain unsatisfactory. As the critical support system and essential components in neurovascular units, glial cells and blood vessels (including the blood-brain barrier) together maintain an optimal microenvironment for neuronal function. They provide nutrients, regulate neuronal excitability, and prevent harmful substances from entering brain tissue. The highly dynamic networks of this support system play an essential role in ischemic stroke through processes including brain homeostasis, supporting neuronal function, and reacting to injuries. However, most studies have focused on postmortem animals, which inevitably lack critical information about the dynamic changes that occur after ischemic stroke. Therefore, a high-precision technique for research in living animals is urgently needed. Two-photon fluorescence laser-scanning microscopy is a powerful imaging technique that can facilitate live imaging at high spatiotemporal resolutions. Two-photon fluorescence laser-scanning microscopy can provide images of the whole-cortex vascular 3D structure, information on multicellular component interactions, and provide images of structure and function in the cranial window. This technique shifts the existing research paradigm from static to dynamic, from flat to stereoscopic, and from single-cell function to multicellular intercommunication, thus providing direct and reliable evidence to identify the pathophysiological mechanisms following ischemic stroke in an intact brain. In this review, we discuss exciting findings from research on the support system after ischemic stroke using two-photon fluorescence laser-scanning microscopy, highlighting the importance of dynamic observations of cellular behavior and interactions in the networks of the brain's support systems. We show the excellent application prospects and advantages of two-photon fluorescence laser-scanning microscopy and predict future research developments and directions in the study of ischemic stroke.
Collapse
Affiliation(s)
- Xuan Wu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Jia-Rui Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yu Fu
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Dan-Yang Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Hao Nie
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Zhou-Ping Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
5
|
Roseborough AD, Myers SJ, Khazaee R, Zhu Y, Zhao L, Iorio E, Elahi FM, Pasternak SH, Whitehead SN. Plasma derived extracellular vesicle biomarkers of microglia activation in an experimental stroke model. J Neuroinflammation 2023; 20:20. [PMID: 36721258 PMCID: PMC9890769 DOI: 10.1186/s12974-023-02708-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 01/27/2023] [Indexed: 02/02/2023] Open
Abstract
Chronic microglia activation post-stroke is associated with worse neurological and cognitive outcomes. However, measurement of microglia activation in vivo is currently limited. Plasma derived extracellular vesicles (EVs) are cell-specific indicators that may allow for non-invasive measurement of microglia phenotype. The aim of this study was to identify activation-state specific microglia EVs (MEVs) in vitro followed by validation in an experimental stroke model. Following pro-inflammatory activation, MEVs contain the microglia protein TMEM119 alongside increased expression of the Toll-like receptor 4 co-receptor CD14. Immunoprecipitation followed by fluorescent nanoparticle tracking analysis (ONI Nanoimager) was used to confirm the isolation of TMEM119+/CD14+ EVs from rat plasma. Electron microscopy confirmed that TMEM119 and CD14 localize to the MEV membrane. To model ischemia, plasma was collected from 3-month wildtype Fischer344 rats prior to, 7 and 28 days after endothelin-1 or saline injection into the dorsal right striatum. Fluorescently labelled MEVs were directly measured in the plasma using nanoflow cytometry (Apogee A60 Microplus). We report a significant increase in circulating TMEM119+/CD14+ EVs 28-days post-stroke in comparison to baseline levels and saline-injected rats, which correlated weakly with stroke volume. TMEM119+/MHC-II+ EVs were also increased post-stroke in comparison to baseline and saline-injected animals. This study is the first to describe an EV biomarker of activated microglia detected directly in plasma following stroke and represents a future tool for the measurement of microglia activity in vivo.
Collapse
Affiliation(s)
- A. D. Roseborough
- grid.39381.300000 0004 1936 8884Vulnerable Brain Laboratory, Department of Anatomy and Cell Biology, The Schulich School of Medicine and Dentistry, The University of Western Ontario, 458 Medical Sciences Building, ON N6A 3K London, Canada
| | - S. J. Myers
- grid.39381.300000 0004 1936 8884Vulnerable Brain Laboratory, Department of Anatomy and Cell Biology, The Schulich School of Medicine and Dentistry, The University of Western Ontario, 458 Medical Sciences Building, ON N6A 3K London, Canada
| | - R. Khazaee
- grid.39381.300000 0004 1936 8884Biotron Integrated Microscopy Facility, The University of Western Ontario, London, ON Canada ,grid.39381.300000 0004 1936 8884Deparment of Biology, The University of Western Ontario, London, ON Canada
| | - Y. Zhu
- grid.39381.300000 0004 1936 8884Vulnerable Brain Laboratory, Department of Anatomy and Cell Biology, The Schulich School of Medicine and Dentistry, The University of Western Ontario, 458 Medical Sciences Building, ON N6A 3K London, Canada
| | - L. Zhao
- grid.39381.300000 0004 1936 8884Vulnerable Brain Laboratory, Department of Anatomy and Cell Biology, The Schulich School of Medicine and Dentistry, The University of Western Ontario, 458 Medical Sciences Building, ON N6A 3K London, Canada
| | - E. Iorio
- grid.266102.10000 0001 2297 6811Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA USA
| | - F. M. Elahi
- grid.266102.10000 0001 2297 6811Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA USA ,grid.59734.3c0000 0001 0670 2351Icahn School of Medicine at Mount Sinai, New York, USA ,grid.39381.300000 0004 1936 8884Department of Clinical Neurological Sciences, The Schulich School of Medicine and Dentistry, The University of Western Ontario, ON London, Canada
| | - S. H. Pasternak
- grid.59734.3c0000 0001 0670 2351Icahn School of Medicine at Mount Sinai, New York, USA ,grid.39381.300000 0004 1936 8884Department of Clinical Neurological Sciences, The Schulich School of Medicine and Dentistry, The University of Western Ontario, ON London, Canada ,grid.39381.300000 0004 1936 8884Robarts Research Institute, The Schulich School of Medicine and Dentistry, The University of Western Ontario, ON London, Canada
| | - S. N. Whitehead
- grid.39381.300000 0004 1936 8884Vulnerable Brain Laboratory, Department of Anatomy and Cell Biology, The Schulich School of Medicine and Dentistry, The University of Western Ontario, 458 Medical Sciences Building, ON N6A 3K London, Canada
| |
Collapse
|
6
|
Truong SHT, Bonnici B, Rupasinghe S, Kemp-Harper BK, Samuel CS, Broughton BRS. Post-stroke administration of H2 relaxin reduces functional deficits, neuronal apoptosis and immune cell infiltration into the mouse brain. Pharmacol Res 2023; 187:106611. [PMID: 36526079 DOI: 10.1016/j.phrs.2022.106611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 12/11/2022] [Accepted: 12/11/2022] [Indexed: 12/15/2022]
Abstract
Brain inflammation and apoptosis contribute to neuronal damage and loss following ischaemic stroke, leading to cognitive and functional disability. It is well-documented that the human gene-2 (H2)-relaxin hormone exhibits pleiotropic properties via its cognate receptor, Relaxin Family Peptide Receptor 1 (RXFP1), including anti-inflammatory and anti-apoptotic effects, thus making it a potential therapeutic for stroke. Hence, the current study investigated whether post-stroke H2-relaxin administration could improve functional and histological outcomes. 8-12-week-old male C57BL/6 mice were subjected to sham operation or photothrombotic stroke and intravenously-administered with either saline (vehicle) or 0.02, 0.2 or 2 mg/kg doses of recombinant H2-relaxin at 6, 24 and 48 h post-stroke. Motor function was assessed using the hanging wire and cylinder test pre-surgery, and at 24 and 72 h post-stroke. Brains were removed after 72 h and infarct volume was assessed via thionin staining, and RXFP1 expression, leukocyte infiltration and apoptosis were determined by immunofluorescence. RXFP1 was identified on neurons, astrocytes and macrophages, and increased post-stroke. Whilst H2-relaxin did not alter infarct volume, it did cause a dose-dependent improvement in motor function at 24 and 72 h post-stroke. Moreover, 2 mg/kg H2-relaxin significantly decreased the number of apoptotic cells as well as macrophages and neutrophils within the ischaemic hemisphere, but did not alter T or B cells numbers. The anti-inflammatory and anti-apoptotic effects of H2-relaxin when administered at 6 h post-cerebral ischaemia may provide a novel therapeutic option for patients following ischaemic stroke.
Collapse
Affiliation(s)
- Shirley H T Truong
- Cardiovascular & Pulmonary Pharmacology Group, Department of Pharmacology and Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Benjamin Bonnici
- Cardiovascular & Pulmonary Pharmacology Group, Department of Pharmacology and Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Samoda Rupasinghe
- Cardiovascular & Pulmonary Pharmacology Group, Department of Pharmacology and Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Barbara K Kemp-Harper
- Cardiovascular & Pulmonary Pharmacology Group, Department of Pharmacology and Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Chrishan S Samuel
- Fibrosis Group, Department of Pharmacology and Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia; Department of Biochemistry and Molecular Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Brad R S Broughton
- Cardiovascular & Pulmonary Pharmacology Group, Department of Pharmacology and Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia.
| |
Collapse
|
7
|
Brain Microvascular Endothelial Cell-Derived Exosomes Protect Neurons from Ischemia–Reperfusion Injury in Mice. Pharmaceuticals (Basel) 2022; 15:ph15101287. [PMID: 36297399 PMCID: PMC9608440 DOI: 10.3390/ph15101287] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/14/2022] [Accepted: 10/16/2022] [Indexed: 11/17/2022] Open
Abstract
Stroke often results in neurological and neuropsychiatric sequela. Exosomes derived from brain endothelial cells (EC-Exo) protect neurons from hypoxic injury. However, the biological role of exosomes in apoptosis and synaptic plasticity remains unclear. This research aimed to assess whether cerebral microvascular endothelial cells inhibit apoptosis and promote synaptic remodeling through exosome-mediated cell–cell interaction after the ischemic attack. The effects of EC-Exo on primary neuronal apoptosis and synapses in oxyglucose deprivation reoxygenation (OGD/R) injury were first assessed in vitro. Animal experiments were performed using C57BL/6J mice, divided into three groups: a sham group, a model (middle cerebral artery occlusion/reperfusion, MCAO/R) group, and an EC-Exo group (tail vein injection of EC-Exo, once/2 days for 14 days) to evaluate the neuromotor and exploratory abilities of mice after MCAO/R. Apoptosis and synaptic protein expression levels were detected. The results demonstrated that EC-Exo inhibited neuronal apoptosis and increased synaptic length after OGD/R. In vivo, EC-Exo not only improved neural motor behavior and increased regional cerebral blood flow (rCBF) in MCAO/R-injured mice but also promoted the expression of synaptic regulatory proteins and inhibited apoptosis in the brain. These results suggest that EC-Exo may provide neuroprotection against stroke by promoting synaptic remodeling and inhibiting apoptosis from protecting neurons.
Collapse
|
8
|
Seong D, Yi S, Han S, Lee J, Park S, Hwang YH, Kim J, Kim HK, Jeon M. Target ischemic stroke model creation method using photoacoustic microscopy with simultaneous vessel monitoring and dynamic photothrombosis induction. PHOTOACOUSTICS 2022; 27:100376. [PMID: 35734368 PMCID: PMC9207728 DOI: 10.1016/j.pacs.2022.100376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 06/02/2022] [Indexed: 06/02/2023]
Abstract
The ischemic stroke animal model evaluates the efficacy of reperfusion and neuroprotective strategies for ischemic injuries. Various conventional methods have been reported to induce the ischemic models; however, controlling specific neurological deficits, mortality rates, and the extent of the infarction is difficult as the size of the affected region is not precisely controlled. In this paper, we report a single laser-based localized target ischemic stroke model development method by simultaneous vessel monitoring and photothrombosis induction using photoacoustic microscopy (PAM), which has minimized the infarct size at precise location with high reproducibility. The proposed method has significantly reduced the infarcted region by illuminating the precise localization. The reproducibility and validity of suggested method have been demonstrated through repeated experiments and histological analyses. These results demonstrate that our method can provide the ischemic stroke model closest to the clinical pathology for brain ischemia research from inducement, occurrence mechanisms to the recovery process.
Collapse
Affiliation(s)
- Daewoon Seong
- School of Electronic and Electrical Engineering, College of IT Engineering, Kyungpook National University, Daegu 41566, the Republic of Korea
| | - Soojin Yi
- Bio-Medical Institute, Kyungpook National University Hospital, Daegu 41404, the Republic of Korea
- Department of Ophthalmology, School of Medicine, Kyungpook National University, Daegu 41944, the Republic of Korea
- Department of Biomedical Science, The Graduate School, Kyungpook National University, Daegu 41944, the Republic of Korea
| | - Sangyeob Han
- School of Electronic and Electrical Engineering, College of IT Engineering, Kyungpook National University, Daegu 41566, the Republic of Korea
- Institute of Biomedical Engineering, School of Medicine, Kyungpook National University, Daegu 41566, the Republic of Korea
| | - Jaeyul Lee
- School of Electronic and Electrical Engineering, College of IT Engineering, Kyungpook National University, Daegu 41566, the Republic of Korea
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Sungjo Park
- Pohang Innotown Center, Pohang University of Science and Technology, Pohang 37673, the Republic of Korea
| | - Yang-Ha Hwang
- Department of Neurology, School of Medicine, Kyungpook National University, Daegu 41944, the Republic of Korea
| | - Jeehyun Kim
- School of Electronic and Electrical Engineering, College of IT Engineering, Kyungpook National University, Daegu 41566, the Republic of Korea
| | - Hong Kyun Kim
- Bio-Medical Institute, Kyungpook National University Hospital, Daegu 41404, the Republic of Korea
- Department of Ophthalmology, School of Medicine, Kyungpook National University, Daegu 41944, the Republic of Korea
- Department of Biomedical Science, The Graduate School, Kyungpook National University, Daegu 41944, the Republic of Korea
| | - Mansik Jeon
- School of Electronic and Electrical Engineering, College of IT Engineering, Kyungpook National University, Daegu 41566, the Republic of Korea
| |
Collapse
|
9
|
Singh AA, Kharwar A, Dandekar MP. A Review on Preclinical Models of Ischemic Stroke: Insights Into the Pathomechanisms and New Treatment Strategies. Curr Neuropharmacol 2022; 20:1667-1686. [PMID: 34493185 PMCID: PMC9881062 DOI: 10.2174/1570159x19666210907092928] [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: 06/20/2021] [Revised: 07/21/2021] [Accepted: 08/26/2021] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Stroke is a serious neurovascular problem and the leading cause of disability and death worldwide. The disrupted demand to supply ratio of blood and glucose during cerebral ischemia develops hypoxic shock, and subsequently necrotic neuronal death in the affected regions. Multiple causal factors like age, sex, race, genetics, diet, and lifestyle play an important role in the occurrence as well as progression of post-stroke deleterious events. These biological and environmental factors may be contributed to vasculature variable architecture and abnormal neuronal activity. Since recombinant tissue plasminogen activator is the only clinically effective clot bursting drug, there is a huge unmet medical need for newer therapies for the treatment of stroke. Innumerous therapeutic interventions have shown promise in the experimental models of stroke but failed to translate it into clinical counterparts. METHODS Original publications regarding pathophysiology, preclinical experimental models, new targets and therapies targeting ischemic stroke have been reviewed since the 1970s. RESULTS We highlighted the critical underlying pathophysiological mechanisms of cerebral stroke and preclinical stroke models. We discuss the strengths and caveats of widely used ischemic stroke models, and commented on the potential translational problems. We also describe the new emerging treatment strategies, including stem cell therapy, neurotrophic factors and gut microbiome-based therapy for the management of post-stroke consequences. CONCLUSION There are still many inter-linked pathophysiological alterations with regards to stroke, animal models need not necessarily mimic the same conditions of stroke pathology and newer targets and therapies are the need of the hour in stroke research.
Collapse
Affiliation(s)
- Aditya A. Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Balanagar, TS 500037, India
| | - Akash Kharwar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Balanagar, TS 500037, India
| | - Manoj P. Dandekar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Balanagar, TS 500037, India,Address correspondence to this author at the Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Balanagar, TS 500037, India; Tel: +91-40-23074750; E-mail:
| |
Collapse
|
10
|
Pereira-Figueiredo D, Nascimento AA, Cunha-Rodrigues MC, Brito R, Calaza KC. Caffeine and Its Neuroprotective Role in Ischemic Events: A Mechanism Dependent on Adenosine Receptors. Cell Mol Neurobiol 2022; 42:1693-1725. [PMID: 33730305 PMCID: PMC11421760 DOI: 10.1007/s10571-021-01077-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 03/05/2021] [Indexed: 02/07/2023]
Abstract
Ischemia is characterized by a transient, insufficient, or permanent interruption of blood flow to a tissue, which leads to an inadequate glucose and oxygen supply. The nervous tissue is highly active, and it closely depends on glucose and oxygen to satisfy its metabolic demand. Therefore, ischemic conditions promote cell death and lead to a secondary wave of cell damage that progressively spreads to the neighborhood areas, called penumbra. Brain ischemia is one of the main causes of deaths and summed with retinal ischemia comprises one of the principal reasons of disability. Although several studies have been performed to investigate the mechanisms of damage to find protective/preventive interventions, an effective treatment does not exist yet. Adenosine is a well-described neuromodulator in the central nervous system (CNS), and acts through four subtypes of G-protein-coupled receptors. Adenosine receptors, especially A1 and A2A receptors, are the main targets of caffeine in daily consumption doses. Accordingly, caffeine has been greatly studied in the context of CNS pathologies. In fact, adenosine system, as well as caffeine, is involved in neuroprotection effects in different pathological situations. Therefore, the present review focuses on the role of adenosine/caffeine in CNS, brain and retina, ischemic events.
Collapse
Affiliation(s)
- D Pereira-Figueiredo
- Neurobiology of the Retina Laboratory, Biomedical Sciences Program, Biomedical Institute, Fluminense Federal University, Niterói, RJ, Brazil
| | - A A Nascimento
- Neurobiology of the Retina Laboratory, Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, RJ, Brazil
| | - M C Cunha-Rodrigues
- Neurobiology of the Retina Laboratory, Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, RJ, Brazil
| | - R Brito
- Laboratory of Neuronal Physiology and Pathology, Cellular and Molecular Biology Department, Institute of Biology, Fluminense Federal University, Niterói, RJ, Brazil
| | - K C Calaza
- Neurobiology of the Retina Laboratory, Biomedical Sciences Program, Biomedical Institute, Fluminense Federal University, Niterói, RJ, Brazil.
- Neurobiology of the Retina Laboratory, Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, RJ, Brazil.
- Neurobiology Department, Biology Institute of Fluminense Federal University, Niteroi, RJ, Brazil.
| |
Collapse
|
11
|
Vicente-Acosta A, Ceprian M, Sobrino P, Pazos MR, Loría F. Cannabinoids as Glial Cell Modulators in Ischemic Stroke: Implications for Neuroprotection. Front Pharmacol 2022; 13:888222. [PMID: 35721207 PMCID: PMC9199389 DOI: 10.3389/fphar.2022.888222] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 05/10/2022] [Indexed: 11/13/2022] Open
Abstract
Stroke is the second leading cause of death worldwide following coronary heart disease. Despite significant efforts to find effective treatments to reduce neurological damage, many patients suffer from sequelae that impair their quality of life. For this reason, the search for new therapeutic options for the treatment of these patients is a priority. Glial cells, including microglia, astrocytes and oligodendrocytes, participate in crucial processes that allow the correct functioning of the neural tissue, being actively involved in the pathophysiological mechanisms of ischemic stroke. Although the exact mechanisms by which glial cells contribute in the pathophysiological context of stroke are not yet completely understood, they have emerged as potentially therapeutic targets to improve brain recovery. The endocannabinoid system has interesting immunomodulatory and protective effects in glial cells, and the pharmacological modulation of this signaling pathway has revealed potential neuroprotective effects in different neurological diseases. Therefore, here we recapitulate current findings on the potential promising contribution of the endocannabinoid system pharmacological manipulation in glial cells for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Andrés Vicente-Acosta
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain.,Departamento de Biología Molecular, Universidad Autónoma de Madrid, Madrid, Spain
| | - Maria Ceprian
- ERC Team, PGNM, INSERM U1315, CNRS UMR5261, University of Lyon 1, University of Lyon, Lyon, France
| | - Pilar Sobrino
- Departamento de Neurología, Hospital Universitario Fundación Alcorcón, Alcorcón, Spain
| | - Maria Ruth Pazos
- Laboratorio de Apoyo a la Investigación, Hospital Universitario Fundación Alcorcón, Alcorcón, Spain
| | - Frida Loría
- Laboratorio de Apoyo a la Investigación, Hospital Universitario Fundación Alcorcón, Alcorcón, Spain
| |
Collapse
|
12
|
Early Application of Ipsilateral Cathodal-tDCS in a Mouse Model of Brain Ischemia Results in Functional Improvement and Perilesional Microglia Modulation. Biomolecules 2022; 12:biom12040588. [PMID: 35454177 PMCID: PMC9027610 DOI: 10.3390/biom12040588] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 04/12/2022] [Accepted: 04/15/2022] [Indexed: 02/01/2023] Open
Abstract
Early stroke therapeutic approaches rely on limited options, further characterized by a narrow therapeutic time window. In this context, the application of transcranial direct current stimulation (tDCS) in the acute phases after brain ischemia is emerging as a promising non-invasive tool. Despite the wide clinical application of tDCS, the cellular mechanisms underlying its positive effects are still poorly understood. Here, we explored the effects of cathodal tDCS (C-tDCS) 6 h after focal forelimb M1 ischemia in Cx3CR1GFP/+ mice. C-tDCS improved motor functionality of the affected forelimb, as assessed by the cylinder and foot-fault tests at 48 h, though not changing the ischemic volume. In parallel, histological analysis showed that motor recovery is associated with decreased microglial cell density in the area surrounding the ischemic core, while astrocytes were not affected. Deeper analysis of microglia morphology within the perilesional area revealed a shift toward a more ramified healthier state, with increased processes’ complexity and a less phagocytic anti-inflammatory activity. Taken together, our findings suggest a positive role for early C-tDCS after ischemia, which is able to modulate microglia phenotype and morphology in parallel to motor recovery.
Collapse
|
13
|
Liu C, Xie J, Sun S, Li H, Li T, Jiang C, Chen X, Wang J, Le A, Wang J, Li Z, Wang J, Wang W. Hemorrhagic Transformation After Tissue Plasminogen Activator Treatment in Acute Ischemic Stroke. Cell Mol Neurobiol 2022; 42:621-646. [PMID: 33125600 PMCID: PMC11441267 DOI: 10.1007/s10571-020-00985-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 10/22/2020] [Indexed: 12/17/2022]
Abstract
Hemorrhagic transformation (HT) is a common complication after thrombolysis with recombinant tissue-type plasminogen activator (rt-PA) in ischemic stroke. In this article, recent research progress of HT in vivo and in vitro studies was reviewed. We have discussed new potential mechanisms and possible experimental models of HT development, as well as possible biomarkers and treatment methods. Meanwhile, we compared and analyzed rodent models, large animal models and in vitro BBB models of HT, and the limitations of these models were discussed. The molecular mechanism of HT was investigated in terms of BBB disruption, rt-PA neurotoxicity and the effect of neuroinflammation, matrix metalloproteinases, reactive oxygen species. The clinical features to predict HT were represented including blood biomarkers and clinical factors. Recent progress in neuroprotective strategies to improve HT after stroke treated with rt-PA is outlined. Further efforts need to be made to reduce the risk of HT after rt-PA therapy and improve the clinical prognosis of patients with ischemic stroke.
Collapse
Affiliation(s)
- Chengli Liu
- Department of Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Jie Xie
- Department of Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Shanshan Sun
- Department of Ultrasound Imaging, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Hui Li
- Department of Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Tianyu Li
- Department of Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Chao Jiang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
| | - Xuemei Chen
- Department of Anatomy, College of Basic Medical Sciences, Zhengzhou University, Henan, 450000, People's Republic of China
| | - Junmin Wang
- Department of Anatomy, College of Basic Medical Sciences, Zhengzhou University, Henan, 450000, People's Republic of China
| | - Anh Le
- Washington University in St. Louis, Saint Louis, MO, 63130, USA
| | - Jiarui Wang
- The Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Zhanfei Li
- Department of Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Jian Wang
- Department of Anatomy, College of Basic Medical Sciences, Zhengzhou University, Henan, 450000, People's Republic of China.
| | - Wei Wang
- Department of Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
| |
Collapse
|
14
|
Chen X, Wang J, Ge L, Lu G, Wan H, Jiang Y, Yao Z, Deng G, Zhang X. A fibrin targeted molecular imaging evaluation of microvascular no-reflow in acute ischemic stroke. Brain Behav 2022; 12:e2474. [PMID: 35025138 PMCID: PMC8865146 DOI: 10.1002/brb3.2474] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 11/21/2021] [Accepted: 12/13/2021] [Indexed: 11/09/2022] Open
Abstract
OBJECTIVE To investigate the relationship between fibrin deposition and "no-reflow" within microcirculation after thrombolysis in acute ischemic stroke (AIS). MATERIALS AND METHODS Experiments were approved by the institutional animal care and use committee. An experimental AIS model was induced in C57BL/6 mice by middle cerebral artery occlusion (MCAO) via the photothrombotic method. Mice were randomly assigned to non-thrombolytic or thrombolytic treated groups (n = 12 per group). The modified Neurological Severity Score and Fast Beam Balance Test were performed by a researcher blinded to the treatment method. MRI was utilized to evaluate all of the mice. An FXIIIa-targeted probe was applied to detect fibrin deposition in acute ischemic brain regions by fluorescence imaging. Necrosis and pathological changes of brain tissue were estimated via Hematoxylin and eosin staining while fibrin deposition was observed by immunohistochemistry. RESULTS Thrombolytic therapy improved AIS clinical symptoms. The infarct area of non-thrombolytic treated mice was significantly greater than that of the thrombolytic treated mice (p < .0001). Fluorescent imaging indicated fibrin deposition in ischemic brain tissue in both groups, with less fibrin in non-thrombolytic treated mice than thrombolytic treated mice, though the difference was not significant. Brain cells with abnormal morphology, necrosis, and liquefication were observed in the infarcted area for both groups. Clotted red blood cells (RBCs) and fibrin build-up in capillaries were found near the ischemic area in both non-thrombolytic and thrombolytic treated groups of mice. CONCLUSION Fibrin deposition and stacked RBCs contribute to microcirculation no-reflow in AIS after thrombolytic therapy.
Collapse
Affiliation(s)
- Xi Chen
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jing Wang
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Liang Ge
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Gang Lu
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Hailin Wan
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yeqing Jiang
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhenwei Yao
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Gang Deng
- Department of Intervention and Vascular Surgery, Zhongda Hospital, Southeast University, Nanjing, China
| | - Xiaolong Zhang
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
15
|
Change in the central control of the bladder function of rats with focal cerebral infarction induced by photochemically-induced thrombosis. PLoS One 2021; 16:e0255200. [PMID: 34752461 PMCID: PMC8577768 DOI: 10.1371/journal.pone.0255200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 10/15/2021] [Indexed: 11/19/2022] Open
Abstract
The photochemically-induced thrombosis (photothrombosis) method can create focal cerebral infarcts anywhere in the relatively superficial layers of the cerebrum; it is easy to implement and minimally invasive. Taking advantage of this versatility, we aimed to establish a new rat model of urinary frequency with focal cerebral infarction, which was characterized by its simplicity, nonlethal nature, and high reproducibility. The prefrontal cortex and the anterior cingulate cortex, which are involved in lower urinary tract control, were targeted for focal cerebral infarction, and urinary parameters were measured by cystometrogram. Cystometric analysis indicated that micturition intervals significantly shortened in photothrombosis-treated rats compared with those in the sham operative group on Days 1 and 7 (P < 0.01), but prolonged after 14 days, with no difference between the two groups. Immunopathological evaluation showed an accumulation of activated microglia, followed by an increase in reactive astrocytes at the peri-infarct zone after photothrombotic stroke. Throughout this study, all postphotothrombosis rats showed cerebral infarction in the prefrontal cortex and anterior cingulate cortex; there were no cases of rats with fatal cerebral infarction. This model corresponded to the clinical presentation, in that the micturition status changed after stroke. In conclusion, this novel model combining nonlethality and high reproducibility may be a suitable model of urinary frequency after focal cerebral infarction.
Collapse
|
16
|
Ortiz-Villatoro NN, Reyes-Garcia SZ, Freitas L, Rodrigues LD, Santos LEC, Faber J, Cavalheiro EA, Finsterer J, Scorza FA, de Almeida ACG, Scorza CA. Amazon rainforest rodents (Proechimys) are resistant to post-stroke epilepsy. Sci Rep 2021; 11:16780. [PMID: 34408211 PMCID: PMC8373885 DOI: 10.1038/s41598-021-96235-5] [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: 09/01/2020] [Accepted: 04/12/2021] [Indexed: 02/07/2023] Open
Abstract
There are no clinical interventions to prevent post-injury epilepsy, a common and devastating outcome after brain insults. Epileptogenic events that run from brain injury to epilepsy are poorly understood. Previous studies in our laboratory suggested Proechimys, an exotic Amazonian rodent, as resistant to acquired epilepsy development in post-status epilepticus models. The present comparative study was conducted to assess (1) stroke-related brain responses 24-h and 30 days after cortical photothrombosis and (2) post-stroke epilepsy between Proechimys rodents and Wistar rats, a traditional animal used for laboratory research. Proechimys group showed smaller volume of ischemic infarction and lesser glial activation than Wistar group. In contrast to Wistar rats, post-stroke decreased levels of pro-inflammatory cytokines and increased levels of anti-inflammatory mediators and growth factors were found in Proechimys. Electrophysiological signaling changes assessed by cortical spreading depression, in vitro and in vivo, showed that Wistar's brain is most severely affected by stroke. Chronic electrocorticographic recordings showed that injury did not lead to epilepsy in Proechimys whereas 88% of the Wistar rats developed post-stroke epilepsy. Science gains insights from comparative studies on diverse species. Proechimys rodents proved to be a useful animal model to study antiepileptogenic mechanisms after brain insults and complement conventional animal models.
Collapse
Affiliation(s)
- Nancy N. Ortiz-Villatoro
- grid.411249.b0000 0001 0514 7202Disciplina de Neurociência, Departamento de Neurologia/Neurocirurgia, Escola Paulista de Medicina/Universidade Federal de São Paulo (EPM/UNIFESP), São Paulo, 04039-032 Brazil
| | - Selvin Z. Reyes-Garcia
- grid.10601.360000 0001 2297 2829Posgrado de Neurología, Facultad de Ciencias Médicas, Universidad Nacional Autónoma de Honduras, Tegucigalpa, Honduras
| | - Leandro Freitas
- grid.411249.b0000 0001 0514 7202Disciplina de Neurociência, Departamento de Neurologia/Neurocirurgia, Escola Paulista de Medicina/Universidade Federal de São Paulo (EPM/UNIFESP), São Paulo, 04039-032 Brazil
| | - Laís D. Rodrigues
- grid.411249.b0000 0001 0514 7202Disciplina de Neurociência, Departamento de Neurologia/Neurocirurgia, Escola Paulista de Medicina/Universidade Federal de São Paulo (EPM/UNIFESP), São Paulo, 04039-032 Brazil
| | - Luiz E. C. Santos
- grid.428481.30000 0001 1516 3599Neurociência Experimental e Computacional, Universidade Federal São João Del-Rey, São João del-Rei, Brazil
| | - Jean Faber
- grid.411249.b0000 0001 0514 7202Disciplina de Neurociência, Departamento de Neurologia/Neurocirurgia, Escola Paulista de Medicina/Universidade Federal de São Paulo (EPM/UNIFESP), São Paulo, 04039-032 Brazil
| | - Esper A. Cavalheiro
- grid.411249.b0000 0001 0514 7202Disciplina de Neurociência, Departamento de Neurologia/Neurocirurgia, Escola Paulista de Medicina/Universidade Federal de São Paulo (EPM/UNIFESP), São Paulo, 04039-032 Brazil
| | - Josef Finsterer
- grid.413303.60000 0004 0437 0893Krankenanstalt Rudolfstiftung, Mersserli Institute, Vienna, Austria
| | - Fulvio A. Scorza
- grid.411249.b0000 0001 0514 7202Disciplina de Neurociência, Departamento de Neurologia/Neurocirurgia, Escola Paulista de Medicina/Universidade Federal de São Paulo (EPM/UNIFESP), São Paulo, 04039-032 Brazil
| | - Antônio C. G. de Almeida
- grid.428481.30000 0001 1516 3599Neurociência Experimental e Computacional, Universidade Federal São João Del-Rey, São João del-Rei, Brazil
| | - Carla A. Scorza
- grid.411249.b0000 0001 0514 7202Disciplina de Neurociência, Departamento de Neurologia/Neurocirurgia, Escola Paulista de Medicina/Universidade Federal de São Paulo (EPM/UNIFESP), São Paulo, 04039-032 Brazil
| |
Collapse
|
17
|
Li F, Xu D, Hou K, Gou X, Li Y. The role of P2Y12 receptor inhibition in ischemic stroke on microglia, platelets and vascular smooth muscle cells. J Thromb Thrombolysis 2021; 50:874-885. [PMID: 32248335 DOI: 10.1007/s11239-020-02098-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
P2Y12 receptors on platelets have long been the main target of antiplatelet drugs. However, a growing number of studies have revealed that P2Y12 receptor activation on microglia and vascular smooth muscle cells (VSMCs) also aggravates ischemic stroke injury. The proliferation and migration of VSMCs in the vascular wall have important influence on the early lesion of atherosclerosis, which may lead to the origin of cerebral ischemic attack of atherosclerosis. Blockage of cellular P2Y12 receptors could inhibit microglial activation, block formation of platelet-leukocyte aggregates, reduce proinflammatory cytokine levels and suppress migration and proliferation of VSMCs, implying that apart from anti-thrombotic effect, P2Y12 inhibitors have additional neuroprotective, anti-inflammatory and anti-atherosclerotic therapeutic benefits against ischemic stroke. In this review, we will summarize recent advances in studies on P2Y12 receptors and emphatically introduce their significance in microglia, platelets and VSMCs after ischemic stroke, discussing how to exert the beneficial effects of P2Y12 inhibition.
Collapse
Affiliation(s)
- Fengyang Li
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Dan Xu
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Kai Hou
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Xue Gou
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Yunman Li
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
18
|
Heindl S, Ricci A, Carofiglio O, Zhou Q, Arzberger T, Lenart N, Franzmeier N, Hortobagyi T, Nelson PT, Stowe AM, Denes A, Edbauer D, Liesz A. Chronic T cell proliferation in brains after stroke could interfere with the efficacy of immunotherapies. J Exp Med 2021; 218:e20202411. [PMID: 34037669 PMCID: PMC8160576 DOI: 10.1084/jem.20202411] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 03/31/2021] [Accepted: 04/28/2021] [Indexed: 01/07/2023] Open
Abstract
Neuroinflammation is an emerging focus of translational stroke research. Preclinical studies have demonstrated a critical role for brain-invading lymphocytes in post-stroke pathophysiology. Reducing cerebral lymphocyte invasion by anti-CD49d antibodies consistently improves outcome in the acute phase after experimental stroke models. However, clinical trials testing this approach failed to show efficacy in stroke patients for the chronic outcome 3 mo after stroke. Here, we identify a potential mechanistic reason for this phenomenon by detecting chronic T cell accumulation-evading the systemic therapy-in the post-ischemic brain. We observed a persistent accumulation of T cells in mice and human autopsy samples for more than 1 mo after stroke. Cerebral T cell accumulation in the post-ischemic brain was driven by increased local T cell proliferation rather than by T cell invasion. This observation urges re-evaluation of current immunotherapeutic approaches, which target circulating lymphocytes for promoting recovery after stroke.
Collapse
Affiliation(s)
- Steffanie Heindl
- Institute for Stroke and Dementia Research, University Hospital, Ludwig Maximilians University Munich, Munich, Germany
| | - Alessio Ricci
- Institute for Stroke and Dementia Research, University Hospital, Ludwig Maximilians University Munich, Munich, Germany
| | - Olga Carofiglio
- Institute for Stroke and Dementia Research, University Hospital, Ludwig Maximilians University Munich, Munich, Germany
| | - Qihui Zhou
- German Center for Neurodegenerative Diseases, Munich, Germany
| | - Thomas Arzberger
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig Maximilians University Munich, Munich, Germany
- Center for Neuropathology and Prion Research, Ludwig Maximilians University Munich, Munich, Germany
| | - Nikolett Lenart
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research, University Hospital, Ludwig Maximilians University Munich, Munich, Germany
| | - Tibor Hortobagyi
- ELKH-DE Cerebrovascular and Neurodegenerative Research Group, Department of Neurology, University of Debrecen, Debrecen, Hungary
| | | | | | - Adam Denes
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Dieter Edbauer
- German Center for Neurodegenerative Diseases, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Arthur Liesz
- Institute for Stroke and Dementia Research, University Hospital, Ludwig Maximilians University Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
19
|
Vogel DDS, Ortiz-Villatoro NN, Araújo NS, Marques MJG, Aimbire F, Scorza FA, Scorza CA, Albertini R. Transcranial low-level laser therapy in an in vivo model of stroke: Relevance to the brain infarct, microglia activation and neuroinflammation. JOURNAL OF BIOPHOTONICS 2021; 14:e202000500. [PMID: 33580734 DOI: 10.1002/jbio.202000500] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/04/2021] [Accepted: 02/06/2021] [Indexed: 06/12/2023]
Abstract
Stroke is the main cause of death and functional disability. The available therapy affects only 5% of patients, and new therapeutic approaches have been constantly tested. Transcranial photobiomodulation (PBM) is promising for its neuroprotective effect on brain injuries. Thus, the present study investigated the PBM effects in an in vivo model of ischemic stroke induced by photothrombosis (PT). Five different groups of Wistar rats were submitted or not to a daily dose of fish oil or/and laser sessions for 2 months. The ischemia volume was evaluated by stereology; GFAP, Iba and NeuN by immunohistochemistry; TNF-α, IL-1β, IL-6, IL-10 and TGF-β by ELISA assay. PBM influenced both the lesion volume and the GFAP. Furthermore, PBM and Ω-3 or both reduced Iba RNAm. PBM reduced TNF-α, IL-1β, IL-6, brain damage, neuroinflammation and microglial activation, and it increased astroglial activity in peri-lesioned region after stroke.
Collapse
Affiliation(s)
- Débora D S Vogel
- Disciplina de Neurociência, Departamento de Neurologia e Neurocirurgia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Nancy N Ortiz-Villatoro
- Disciplina de Neurociência, Departamento de Neurologia e Neurocirurgia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Noemi S Araújo
- Disciplina de Neurociência, Departamento de Neurologia e Neurocirurgia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Márcia Jonathas Guimarães Marques
- Disciplina de Neurociência, Departamento de Neurologia e Neurocirurgia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Flavio Aimbire
- Programa de Pós-graduação em Medicina Translacional, Departamento de Ciência e Tecnologia, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Fúlvio A Scorza
- Disciplina de Neurociência, Departamento de Neurologia e Neurocirurgia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Carla A Scorza
- Disciplina de Neurociência, Departamento de Neurologia e Neurocirurgia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Regiane Albertini
- Programa de Pós-graduação em Ciência do Movimento Humano e Reabilitação, Departamento de Ciência e Tecnologia, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| |
Collapse
|
20
|
Brait VH, Wright DK, Nategh M, Oman A, Syeda WT, Ermine CM, O'Brien KR, Werden E, Churilov L, Johnston LA, Thompson LH, Nithianantharajah J, Jackman KA, Brodtmann A. Longitudinal hippocampal volumetric changes in mice following brain infarction. Sci Rep 2021; 11:10269. [PMID: 33986303 PMCID: PMC8119705 DOI: 10.1038/s41598-021-88284-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 04/05/2021] [Indexed: 01/14/2023] Open
Abstract
Hippocampal atrophy is increasingly described in many neurodegenerative syndromes in humans, including stroke and vascular cognitive impairment. However, the progression of brain volume changes after stroke in rodent models is poorly characterized. We aimed to monitor hippocampal atrophy occurring in mice up to 48-weeks post-stroke. Male C57BL/6J mice were subjected to an intraluminal filament-induced middle cerebral artery occlusion (MCAO). At baseline, 3-days, and 1-, 4-, 12-, 24-, 36- and 48-weeks post-surgery, we measured sensorimotor behavior and hippocampal volumes from T2-weighted MRI scans. Hippocampal volume-both ipsilateral and contralateral-increased over the life-span of sham-operated mice. In MCAO-subjected mice, different trajectories of ipsilateral hippocampal volume change were observed dependent on whether the hippocampus contained direct infarction, with a decrease in directly infarcted tissue and an increase in non-infarcted tissue. To further investigate these volume changes, neuronal and glial cell densities were assessed in histological brain sections from the subset of MCAO mice lacking hippocampal infarction. Our findings demonstrate previously uncharacterized changes in hippocampal volume and potentially brain parenchymal cell density up to 48-weeks in both sham- and MCAO-operated mice.
Collapse
Affiliation(s)
- Vanessa H Brait
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia.
| | - David K Wright
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia.,The Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Mohsen Nategh
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Alexander Oman
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Warda T Syeda
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Charlotte M Ermine
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Katrina R O'Brien
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Emilio Werden
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Leonid Churilov
- Melbourne Medical School, University of Melbourne, Parkville, VIC, Australia
| | - Leigh A Johnston
- Department of Biomedical Engineering, University of Melbourne, Parkville, VIC, Australia.,Melbourne Brain Centre Imaging Unit, University of Melbourne, Parkville, VIC, Australia
| | - Lachlan H Thompson
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Jess Nithianantharajah
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Katherine A Jackman
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Amy Brodtmann
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
21
|
Zhao Y, Ye S, Lin J, Liang F, Chen J, Hu J, Chen K, Fang Y, Chen X, Xiong Y, Lin L, Tan X. NmFGF1-Regulated Glucolipid Metabolism and Angiogenesis Improves Functional Recovery in a Mouse Model of Diabetic Stroke and Acts via the AMPK Signaling Pathway. Front Pharmacol 2021; 12:680351. [PMID: 34025437 PMCID: PMC8139577 DOI: 10.3389/fphar.2021.680351] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 04/23/2021] [Indexed: 12/16/2022] Open
Abstract
Diabetes increases the risk of stroke, exacerbates neurological deficits, and increases mortality. Non-mitogenic fibroblast growth factor 1 (nmFGF1) is a powerful neuroprotective factor that is also regarded as a metabolic regulator. The present study aimed to investigate the effect of nmFGF1 on the improvement of functional recovery in a mouse model of type 2 diabetic (T2D) stroke. We established a mouse model of T2D stroke by photothrombosis in mice that were fed a high-fat diet and injected with streptozotocin (STZ). We found that nmFGF1 reduced the size of the infarct and attenuated neurobehavioral deficits in our mouse model of T2D stroke. Angiogenesis plays an important role in neuronal survival and functional recovery post-stroke. NmFGF1 promoted angiogenesis in the mouse model of T2D stroke. Furthermore, nmFGF1 reversed the reduction of tube formation and migration in human brain microvascular endothelial cells (HBMECs) cultured in high glucose conditions and treated with oxygen glucose deprivation/re-oxygenation (OGD). Amp-activated protein kinase (AMPK) plays a critical role in the regulation of angiogenesis. Interestingly, we found that nmFGF1 increased the protein expression of phosphorylated AMPK (p-AMPK) both in vivo and in vitro. We found that nmFGF1 promoted tube formation and migration and that this effect was further enhanced by an AMPK agonist (A-769662). In contrast, these processes were inhibited by the application of an AMPK inhibitor (compound C) or siRNA targeting AMPK. Furthermore, nmFGF1 ameliorated neuronal loss in diabetic stroke mice via AMPK-mediated angiogenesis. In addition, nmFGF1 ameliorated glucose and lipid metabolic disorders in our mouse model of T2D stroke without causing significant changes in body weight. These results revealed that nmFGF1-regulated glucolipid metabolism and angiogenesis play a key role in the improvement of functional recovery in a mouse model of T2D stroke and that these effects are mediated by the AMPK signaling pathway.
Collapse
Affiliation(s)
- Yeli Zhao
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Shasha Ye
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jingjing Lin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Fei Liang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jun Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jian Hu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Kun Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yani Fang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xiongjian Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Ye Xiong
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Li Lin
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
- Research Units of Clinical Translation of Cell Growth Factors and Diseases Research, Chinese Academy of Medical Science, Wenzhou, China
| | - Xianxi Tan
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
22
|
Kim Y, Lee YB, Bae SK, Oh SS, Choi JR. Development of a photochemical thrombosis investigation system to obtain a rabbit ischemic stroke model. Sci Rep 2021; 11:5787. [PMID: 33707580 PMCID: PMC7970995 DOI: 10.1038/s41598-021-85348-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 02/25/2021] [Indexed: 11/15/2022] Open
Abstract
Photochemical thrombosis is a method for the induction of ischemic stroke in the cerebral cortex. It can generate localized ischemic infarcts in the desired region; therefore, it has been actively employed in establishing an ischemic stroke animal model and in vivo assays of diagnostic and therapeutic techniques for stroke. To establish a rabbit ischemic stroke model and overcome the shortcoming of previous studies that were difficult to build a standardized photothrombotic rabbit model, we developed a photochemical thrombosis induction system that can produce consistent brain damage on a specific area. To verify the generation of photothrombotic brain damage using the system, longitudinal magnetic resonance imaging, 2,3,5-triphenyltetrazolium chloride staining, and histological staining were applied. These analytical methods have a high correlation for ischemic infarction and are appropriate for analyzing photothrombotic brain damage in the rabbit brain. The results indicated that the photothrombosis induction system has a main advantage of being accurately controlled a targeted region of photothrombosis and can produce cerebral hemisphere lesions on the target region of the rabbit brain. In conjugation with brain atlas, it can induce photochemical ischemic stroke locally in the part of the brain that is responsible for a particular brain function and the system can be used to develop animal models with degraded specific functions. Also, the photochemical thrombosis induction system and a standardized rabbit ischemic stroke model that uses this system have the potential to be used for verifications of biomedical techniques for ischemic stroke at a preclinical stage in parallel with further performance improvements.
Collapse
Affiliation(s)
- Yoonhee Kim
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu, 41061, Republic of Korea
| | - Yoon Bum Lee
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu, 41061, Republic of Korea
| | - Seung Kuk Bae
- Department of Biofibers and Biomaterials Science, Kyungpook National University, Daegu, 41566, Korea
| | - Sung Suk Oh
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu, 41061, Republic of Korea.
| | - Jong-Ryul Choi
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu, 41061, Republic of Korea.
| |
Collapse
|
23
|
Karpati S, Hubert V, Hristovska I, Lerouge F, Chaput F, Bretonnière Y, Andraud C, Banyasz A, Micouin G, Monteil M, Lecouvey M, Mercey-Ressejac M, Dey AK, Marche PN, Lindgren M, Pascual O, Wiart M, Parola S. Hybrid multimodal contrast agent for multiscale in vivo investigation of neuroinflammation. NANOSCALE 2021; 13:3767-3781. [PMID: 33555278 DOI: 10.1039/d0nr07026b] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Neuroinflammation is a process common to several brain pathologies. Despites its medical relevance, it still remains poorly understood; there is therefore a need to develop new in vivo preclinical imaging strategies to monitor inflammatory processes longitudinally. We here present the development of a hybrid imaging nanoprobe named NP3, that was specifically designed to get internalized by phagocytic cells and imaged in vivo with MRI and bi-photon microscopy. NP3 is composed of a 16 nm core of gadolinium fluoride (GdF3), coated with bisphosphonate polyethylene glycol (PEG) and functionalized with a Lemke-type fluorophore. It has a hydrodynamic diameter of 28 ± 8 nm and a zeta potential of -42 ± 6 mV. The MR relaxivity ratio at 7 T is r1/r2 = 20; therefore, NP3 is well suited as a T2/T2* contrast agent. In vitro cytotoxicity assessments performed on four human cell lines revealed no toxic effects of NP3. In addition, NP3 is internalized by macrophages in vitro without inducing inflammation or cytotoxicity. In vivo, uptake of NP3 has been observed in the spleen and the liver. NP3 has a prolonged vascular remanence, which is an advantage for macrophage uptake in vivo. The proof-of-concept that NP3 may be used as a contrast agent targeting phagocytic cells is provided in an animal model of ischemic stroke in transgenic CX3CR1-GFP/+ mice using three complementary imaging modalities: MRI, intravital two-photon microscopy and phase contrast imaging with synchrotron X-rays. In summary, NP3 is a promising preclinical tool for the multiscale and multimodal investigation of neuroinflammation.
Collapse
Affiliation(s)
- Szilvia Karpati
- Université de Lyon, Ecole Normale Supérieure de Lyon, CNRS UMR 5182, Université Lyon 1, Laboratoire de Chimie, 46 allée d'Italie, F69364 Lyon, France.
| | - Violaine Hubert
- Univ-Lyon, CarMeN laboratory, Inserm U1060, INRA U1397, INSA Lyon, Université Claude Bernard Lyon 1, F-69600, Oullins, France
| | - Inès Hristovska
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217, Université Lyon, Villeurbanne 69100, France
| | - Frédéric Lerouge
- Université de Lyon, Ecole Normale Supérieure de Lyon, CNRS UMR 5182, Université Lyon 1, Laboratoire de Chimie, 46 allée d'Italie, F69364 Lyon, France.
| | - Frédéric Chaput
- Université de Lyon, Ecole Normale Supérieure de Lyon, CNRS UMR 5182, Université Lyon 1, Laboratoire de Chimie, 46 allée d'Italie, F69364 Lyon, France.
| | - Yann Bretonnière
- Université de Lyon, Ecole Normale Supérieure de Lyon, CNRS UMR 5182, Université Lyon 1, Laboratoire de Chimie, 46 allée d'Italie, F69364 Lyon, France.
| | - Chantal Andraud
- Université de Lyon, Ecole Normale Supérieure de Lyon, CNRS UMR 5182, Université Lyon 1, Laboratoire de Chimie, 46 allée d'Italie, F69364 Lyon, France.
| | - Akos Banyasz
- Université de Lyon, Ecole Normale Supérieure de Lyon, CNRS UMR 5182, Université Lyon 1, Laboratoire de Chimie, 46 allée d'Italie, F69364 Lyon, France.
| | - Guillaume Micouin
- Université de Lyon, Ecole Normale Supérieure de Lyon, CNRS UMR 5182, Université Lyon 1, Laboratoire de Chimie, 46 allée d'Italie, F69364 Lyon, France.
| | - Maëlle Monteil
- Université Sorbonne Paris Nord, Laboratoire CSPBAT, CNRS UMR 7244, F-93017 Bobigny Cedex, France
| | - Marc Lecouvey
- Université Sorbonne Paris Nord, Laboratoire CSPBAT, CNRS UMR 7244, F-93017 Bobigny Cedex, France
| | - Marion Mercey-Ressejac
- Institute for Advanced Biosciences, Université Grenoble-Alpes, INSERM U1209, CNRS UMR5309, La Tronche, France
| | - Arindam K Dey
- Institute for Advanced Biosciences, Université Grenoble-Alpes, INSERM U1209, CNRS UMR5309, La Tronche, France
| | - Patrice N Marche
- Institute for Advanced Biosciences, Université Grenoble-Alpes, INSERM U1209, CNRS UMR5309, La Tronche, France
| | - Mikael Lindgren
- Norwegian University of Science and Technology - Department of Physics, Høgskoleringen 5, Realfagbygget, 7491 Trondheim, Norway
| | - Olivier Pascual
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217, Université Lyon, Villeurbanne 69100, France
| | - Marlène Wiart
- Univ-Lyon, CarMeN laboratory, Inserm U1060, INRA U1397, INSA Lyon, Université Claude Bernard Lyon 1, F-69600, Oullins, France
| | - Stephane Parola
- Université de Lyon, Ecole Normale Supérieure de Lyon, CNRS UMR 5182, Université Lyon 1, Laboratoire de Chimie, 46 allée d'Italie, F69364 Lyon, France.
| |
Collapse
|
24
|
Sillerud LO, Yang Y, Yang LY, Duval KB, Thompson J, Yang Y. Longitudinal monitoring of microglial/macrophage activation in ischemic rat brain using Iba-1-specific nanoparticle-enhanced magnetic resonance imaging. J Cereb Blood Flow Metab 2020; 40:S117-S133. [PMID: 32960690 PMCID: PMC7687035 DOI: 10.1177/0271678x20953913] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Microglial/macrophage activation plays a dual role in response to brain injury after a stroke, promoting early neuroinflammation and benefit for neurovascular recovery. Therefore, the dynamics of stroke-induced cerebral microglial/macrophage activation are of substantial interest. This study used novel anti-Iba-1-targeted superparamagnetic iron-platinum (FePt) nanoparticles in conjunction with magnetic resonance imaging (MRI) to measure the spatiotemporal changes of the microglial/macrophage activation in living rat brain for four weeks post-stroke. Ischemic lesion areas were identified and measured using T2-weighted MR images. After injection of the FePt-nanoparticles, T2*-weighted MR images showed that the nanoparticles were seen solely in brain regions that coincided with areas of active microglia/macrophages detected by post-mortem immunohistochemistry. Good agreement in morphological and distributive dynamic changes was also observed between the Fe+-cells and the Iba-1+-microglia/macrophages. The spatiotemporal changes of nanoparticle detected by T2*-weighted images paralleled the changes of microglial/macrophage activation and phenotypes measured by post-mortem immunohistochemistry over the four weeks post-stroke. Maximum microglial/macrophage activation occurred seven days post-stroke for both measures, and the diminished activation found after two weeks continued to four weeks. Our results suggest that nanoparticle-enhanced MRI may constitute a novel approach for monitoring the dynamic development of neuroinflammation in living animals during the progression and treatment of stroke.
Collapse
Affiliation(s)
- Laurel O Sillerud
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
- BRaIN Imaging Center, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Yirong Yang
- BRaIN Imaging Center, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
- College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Lisa Y Yang
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Kelsey B Duval
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Jeffrey Thompson
- Center for Memory and Aging, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Yi Yang
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
- Yi Yang, Department of Neurology, University of New Mexico, MSC11 6035, Albuquerque, NM 87131-0001, USA.
| |
Collapse
|
25
|
Lubart A, Benbenishty A, Har-Gil H, Laufer H, Gdalyahu A, Assaf Y, Blinder P. Single Cortical Microinfarcts Lead to Widespread Microglia/Macrophage Migration Along the White Matter. Cereb Cortex 2020; 31:248-266. [PMID: 32954425 DOI: 10.1093/cercor/bhaa223] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 05/13/2020] [Accepted: 06/08/2020] [Indexed: 12/31/2022] Open
Abstract
Loss of cognitive function with aging is a complex and poorly understood process. Recently, clinical research has linked the occurrence of cortical microinfarcts to cognitive decline. Cortical microinfarcts form following the occlusion of penetrating vessels and are considered to be restricted to the proximity of the occluded vessel. Whether and how such local events propagate and affect remote brain regions remain unknown. To this end, we combined histological analysis and longitudinal diffusion tensor imaging (DTI), following the targeted-photothrombotic occlusion of single cortical penetrating vessels. Occlusions resulted in distant tissue reorganization across the mouse brain. This remodeling co-occurred with the formation of a microglia/macrophage migratory path along subcortical white matter tracts, reaching the contralateral hemisphere through the corpus callosum and leaving a microstructural signature detected by DTI-tractography. CX3CR1-deficient mice exhibited shorter trail lengths, differential remodeling, and only ipsilateral white matter tract changes. We concluded that microinfarcts lead to brain-wide remodeling in a microglial CX3CR1-dependent manner.
Collapse
Affiliation(s)
- Alisa Lubart
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Amit Benbenishty
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv-Yafo, Israel.,Biological Regulation Department, The Weizmann Institute of Science, Rehovot, Israel
| | - Hagai Har-Gil
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Hadas Laufer
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Amos Gdalyahu
- Neurobiology, Biochemistry and Biophysics School, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Yaniv Assaf
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv-Yafo, Israel.,Neurobiology, Biochemistry and Biophysics School, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Pablo Blinder
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv-Yafo, Israel.,Neurobiology, Biochemistry and Biophysics School, Tel Aviv University, Tel Aviv-Yafo, Israel
| |
Collapse
|
26
|
Freitas-Andrade M, Raman-Nair J, Lacoste B. Structural and Functional Remodeling of the Brain Vasculature Following Stroke. Front Physiol 2020; 11:948. [PMID: 32848875 PMCID: PMC7433746 DOI: 10.3389/fphys.2020.00948] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 07/14/2020] [Indexed: 12/12/2022] Open
Abstract
Maintenance of cerebral blood vessel integrity and regulation of cerebral blood flow ensure proper brain function. The adult human brain represents only a small portion of the body mass, yet about a quarter of the cardiac output is dedicated to energy consumption by brain cells at rest. Due to a low capacity to store energy, brain health is heavily reliant on a steady supply of oxygen and nutrients from the bloodstream, and is thus particularly vulnerable to stroke. Stroke is a leading cause of disability and mortality worldwide. By transiently or permanently limiting tissue perfusion, stroke alters vascular integrity and function, compromising brain homeostasis and leading to widespread consequences from early-onset motor deficits to long-term cognitive decline. While numerous lines of investigation have been undertaken to develop new pharmacological therapies for stroke, only few advances have been made and most clinical trials have failed. Overall, our understanding of the acute and chronic vascular responses to stroke is insufficient, yet a better comprehension of cerebrovascular remodeling following stroke is an essential prerequisite for developing novel therapeutic options. In this review, we present a comprehensive update on post-stroke cerebrovascular remodeling, an important and growing field in neuroscience, by discussing cellular and molecular mechanisms involved, sex differences, limitations of preclinical research design and future directions.
Collapse
Affiliation(s)
| | - Joanna Raman-Nair
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Baptiste Lacoste
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
27
|
Galkov M, Kiseleva E, Gulyaev M, Sidorova M, Gorbacheva L. New PAR1 Agonist Peptide Demonstrates Protective Action in a Mouse Model of Photothrombosis-Induced Brain Ischemia. Front Neurosci 2020; 14:335. [PMID: 32547356 PMCID: PMC7273131 DOI: 10.3389/fnins.2020.00335] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 03/20/2020] [Indexed: 11/23/2022] Open
Abstract
Protease-activated receptors (PARs) are involved not only in hemostasis but also in the development of ischemic brain injury. In the present work, we examined in vivo effects of a new peptide (AP9) composing Asn47-Phen55 of PAR1 “tethered ligand” generated by activated protein C. We chose a mouse model of photothrombosis (PT)-induced ischemia to assess AP9 effects in vivo. To reveal the molecular mechanism of AP9 action, mice lacking β-arrestin-2 were used. AP9 was injected intravenously once 10 min before PT at doses of 0.2, 2, or 20 mg/kg, or twice, that is, 10 min before and 1 h after PT at a dose of 20 mg/kg. Lesion volume was measured by magnetic resonance imaging and staining of brain sections with tetrazolium salt. Neurologic deficit was estimated using the cylinder and the grid-walk tests. Blood–brain barrier (BBB) disruption was assessed by Evans blue dye extraction. Eosin-hematoxylin staining and immunohistochemical staining were applied to evaluate the number of undamaged neurons and activated glial cells in the penumbra. A single administration of AP9 (20 mg/kg), as well as its two injections (20 mg/kg), decreased brain lesion volume. A double administration of AP9 also reduced BBB disruption and neurological deficit in mice. We did not observe the protective effect of AP9 in mice lacking β-arrestin-2 after PT. Thus, we demonstrated for the first time protective properties of a PAR1 agonist peptide, AP9, in vivo. β-Arrestin-2 was required for the protective action of AP9 in PT-induced brain ischemia.
Collapse
Affiliation(s)
- Maksim Galkov
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia.,Electrophysiology Laboratory, Translational Medicine Institute, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Ekaterina Kiseleva
- Electrophysiology Laboratory, Translational Medicine Institute, Pirogov Russian National Research Medical University, Moscow, Russia.,Department of Cell Biology, Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Moscow, Russia
| | - Mikhail Gulyaev
- Faculty of Fundamental Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Maria Sidorova
- Laboratory of Peptide Synthesis, Institute of Experimental Cardiology, National Medical Research Center for Cardiology of Russian Ministry of Health, Moscow, Russia
| | - Liubov Gorbacheva
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia.,Electrophysiology Laboratory, Translational Medicine Institute, Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
28
|
Zou Y, Hu J, Huang W, Ye S, Han F, Du J, Shao M, Guo R, Lin J, Zhao Y, Xiong Y, Wang X. Non-Mitogenic Fibroblast Growth Factor 1 Enhanced Angiogenesis Following Ischemic Stroke by Regulating the Sphingosine-1-Phosphate 1 Pathway. Front Pharmacol 2020; 11:59. [PMID: 32194396 PMCID: PMC7063943 DOI: 10.3389/fphar.2020.00059] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 01/22/2020] [Indexed: 12/11/2022] Open
Abstract
Ischemic strokes account for about 80% of all strokes and are associated with a high risk of mortality. Angiogenesis of brain microvascular endothelial cells may contribute to functional restoration following ischemia. Fibroblast growth factor 1 (FGF1), a member of FGF superfamily, involved in embryonic development, angiogenesis, wound healing, and neuron survival. However, the mitogenic activity of FGF1 is known to contribute to several human pathologies, thereby questioning the safety of its clinical applications. Here, we explored the effects and mechanism of action of non-mitogenic FGF1 (nmFGF1) on angiogenesis in mice after ischemia stroke and an oxygen-glucose deprivation (OGD)-induced human brain microvascular endothelial cells (HBMECs) injury model. We found that intranasal administration nmFGF1 significantly promoted angiogenesis in mice after stroke, and significantly increased the formation of matrigel tube and promoted scratch migration in a dose-dependent manner in OGD-induced HBMECs in vitro. However, the co-administration of an FGF receptor 1 (FGFR1)-specific inhibitor PD173074 significantly reversed the effects of nmFGF1 in vitro, suggesting that nmFGF1 functions via FGFR1 activation. Moreover, nmFGF1 activated sphingosine-1-phosphate receptor 1 (S1PR1, S1P1) in mice after stroke in vivo. S1P1 protein antagonist VPC23019 and agonist FTY720 were used to confirm that nmFGF1 promotes angiogenesis in vitro partially through the S1P1 pathway. OGD induced downregulation of S1P1 expression. The S1P1 antagonist VPC23019 blocked the stimulatory effects of nmFGF1, whereas the S1P1 agonist FTY720 exerted effects comparable with those of nmFGF1. Furthermore, PD173074 reversed the effect of nmFGF1 on upregulating S1P1 signaling. In conclusion, nmFGF1 enhanced angiogenesis in mice following stroke and OGD-induced HBMECs through S1P1 pathway regulation mediated via FGFR1 activation. This new discovery suggests the potential therapeutic role of nmFGF1 for the treatment of ischemic strokes.
Collapse
Affiliation(s)
- Yuchi Zou
- The Frist Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jian Hu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wenting Huang
- School of the First Clinical Medical Science, Wenzhou Medical University, Wenzhou, China
| | - Shasha Ye
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Fanyi Han
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jingting Du
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Mingjie Shao
- School of the First Clinical Medical Science, Wenzhou Medical University, Wenzhou, China
| | - Ruili Guo
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jingjing Lin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yeli Zhao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.,Engineering Laboratory of Zhejiang Province for Pharmaceutical Development of Growth Factors, Biomedical Collaborative Innovation Center of Wenzhou, Wenzhou, China
| | - Ye Xiong
- The Frist Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xue Wang
- The Frist Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
29
|
Lv J, Li S, Zhang J, Duan F, Wu Z, Chen R, Chen M, Huang S, Ma H, Nie L. In vivo photoacoustic imaging dynamically monitors the structural and functional changes of ischemic stroke at a very early stage. Am J Cancer Res 2020; 10:816-828. [PMID: 31903152 PMCID: PMC6929999 DOI: 10.7150/thno.38554] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 09/18/2019] [Indexed: 11/24/2022] Open
Abstract
Ischemic stroke (IS) is one of the leading causes of death and accounts for 85% of stroke cases. Since the symptoms are not obvious, diagnosis of IS, particularly at an early stage, is a great challenge. Photoacoustic imaging combines high sensitivity of optical imaging and fine resolution of ultrasonography to non-invasively provide structural and functional information of IS. Methods: We adopted three rapid photoacoustic imaging systems with varying characteristics, including a portable handheld photoacoustic system, high-sensitivity bowl-shaped array photoacoustic computed tomography (PACT), and high-resolution photoacoustic microscopy (PAM) to assess the stereoscopic and comprehensive pathophysiological status of IS at an early stage. Two representative models of IS, referring to photothrombosis and middle cerebral artery occlusion (MCAO) models, were established to verify the feasibility of photoacoustic imaging detection. Results: Non-invasive, rapid PACT of the IS model in mouse provided structural information of the brain lesion, achieving early disease identification (5 min after the onset of disease). Moreover, it was able to dynamically reflect disease progression. Quantitative high-resolution PAM allowed observation of pathological changes in the microvascular system of mouse brain. In terms of functional imaging, significant differences in oxygen saturation (sO2) levels between infarcted and normal areas could be observed by PACT, permitting effective functional parameters for the diagnosis of IS. Conclusions: We used PACT to perform full-view structural imaging and functional imaging of sO2 in IS at the macroscopic level, and then observed the microvascular changes in the infarcted area at the microscopic level by using PAM. This work may provide new tools for the early diagnosis of IS and its subsequent complications as well as assessment of disease progression.
Collapse
|
30
|
Galkov M, Gulyaev M, Kiseleva E, Andreev-Andrievskiy A, Gorbacheva L. Methods for detection of brain injury after photothrombosis-induced ischemia in mice: Characteristics and new aspects of their application. J Neurosci Methods 2020; 329:108457. [PMID: 31614160 DOI: 10.1016/j.jneumeth.2019.108457] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 09/12/2019] [Accepted: 10/06/2019] [Indexed: 10/25/2022]
Abstract
BACKGROUND Photothrombosis is a minimally invasive method for induction of cortical ischemia. However, different ways of applying some methods to assess photothrombosis-induced damage need to be developed. NEW METHODS We applied the tongue protrusion test and H&E staining of brain sections to detect ischemic damage after photothrombosis. Evaluation of the local status of the BBB using Evans blue dye was proposed. We also assessed the sensitivity of the grid-walk test. Moreover, we examined the interchangeability of MRI and TTC staining to measure lesion volume. RESULTS We evaluated ischemic outcomes at 24 h after photothrombosis in mice. The tongue protrusion test did not reveal impairments of the neurological status whereas the grid-walk test showed the high sensitivity. Using histological techniques, we determined the reduction in the number of neurons with normal morphology in the penumbra. 3D reconstruction of the brain, which reflected Evans blue dye distribution in the nervous tissue, revealed BBB disruption in areas remote from the ischemic core. We also showed the strong correlation between damage volumes assessed by MRI and TTC staining. COMPARISON WITH EXISTING METHODS The present work demonstrates the efficacy of the classical histological approach and TTC staining that are more affordable than MRI and immunohistochemical methods. Detection of 3D distribution of Evans blue dye in the brain in contrast to its total extraction reveals BBB damage in details. CONCLUSIONS We proposed the simple methods for describing the severity of brain ischemia at the cellular and whole organism levels without significant labor and financial expenditures.
Collapse
Affiliation(s)
- Maksim Galkov
- Lomonosov Moscow State University, 119991 Moscow, Russia; Pirogov Russian National Research Medical University, 117997 Moscow, Russia.
| | | | - Ekaterina Kiseleva
- Pirogov Russian National Research Medical University, 117997 Moscow, Russia; Koltzov Institute of Developmental Biology of Russian Academy of Sciences, 119334 Moscow, Russia
| | - Alexander Andreev-Andrievskiy
- Lomonosov Moscow State University, 119991 Moscow, Russia; Institute of Biomedical Problems of Russian Academy of Sciences, 123007 Moscow, Russia
| | - Liubov Gorbacheva
- Lomonosov Moscow State University, 119991 Moscow, Russia; Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| |
Collapse
|
31
|
Cirillo C, Brihmat N, Castel-Lacanal E, Le Friec A, Barbieux-Guillot M, Raposo N, Pariente J, Viguier A, Simonetta-Moreau M, Albucher JF, Olivot JM, Desmoulin F, Marque P, Chollet F, Loubinoux I. Post-stroke remodeling processes in animal models and humans. J Cereb Blood Flow Metab 2020; 40:3-22. [PMID: 31645178 PMCID: PMC6928555 DOI: 10.1177/0271678x19882788] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 08/28/2019] [Accepted: 09/05/2019] [Indexed: 01/05/2023]
Abstract
After cerebral ischemia, events like neural plasticity and tissue reorganization intervene in lesioned and non-lesioned areas of the brain. These processes are tightly related to functional improvement and successful rehabilitation in patients. Plastic remodeling in the brain is associated with limited spontaneous functional recovery in patients. Improvement depends on the initial deficit, size, nature and localization of the infarction, together with the sex and age of the patient, all of them affecting the favorable outcome of reorganization and repair of damaged areas. A better understanding of cerebral plasticity is pivotal to design effective therapeutic strategies. Experimental models and clinical studies have fueled the current understanding of the cellular and molecular processes responsible for plastic remodeling. In this review, we describe the known mechanisms, in patients and animal models, underlying cerebral reorganization and contributing to functional recovery after ischemic stroke. We also discuss the manipulations and therapies that can stimulate neural plasticity. We finally explore a new topic in the field of ischemic stroke pathophysiology, namely the brain-gut axis.
Collapse
Affiliation(s)
- Carla Cirillo
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Nabila Brihmat
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Evelyne Castel-Lacanal
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Alice Le Friec
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | | | - Nicolas Raposo
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Jérémie Pariente
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Alain Viguier
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Marion Simonetta-Moreau
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Jean-François Albucher
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Jean-Marc Olivot
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Franck Desmoulin
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Philippe Marque
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - François Chollet
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| | - Isabelle Loubinoux
- Toulouse NeuroImaging Center (ToNIC), INSERM, University Paul Sabatier, UPS, Toulouse, France
| |
Collapse
|
32
|
Microcirculatory Changes in Experimental Models of Stroke and CNS-Injury Induced Immunodepression. Int J Mol Sci 2019; 20:ijms20205184. [PMID: 31635068 PMCID: PMC6834192 DOI: 10.3390/ijms20205184] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 10/14/2019] [Accepted: 10/18/2019] [Indexed: 12/17/2022] Open
Abstract
Stroke is the second-leading cause of death globally and the leading cause of disability in adults. Medical complications after stroke, especially infections such as pneumonia, are the leading cause of death in stroke survivors. Systemic immunodepression is considered to contribute to increased susceptibility to infections after stroke. Different experimental models have contributed significantly to the current knowledge of stroke pathophysiology and its consequences. Each model causes different changes in the cerebral microcirculation and local inflammatory responses after ischemia. The vast majority of studies which focused on the peripheral immune response to stroke employed the middle cerebral artery occlusion method. We review various experimental stroke models with regard to microcirculatory changes and discuss the impact on local and peripheral immune response for studies of CNS-injury (central nervous system injury) induced immunodepression.
Collapse
|
33
|
Li XM, Wang XY, Feng XW, Shao MM, Liu WF, Ma QQ, Wang EP, Chen J, Shao B. Serum interleukin-33 as a novel marker for long-term prognosis and recurrence in acute ischemic stroke patients. Brain Behav 2019; 9:e01369. [PMID: 31397082 PMCID: PMC6749472 DOI: 10.1002/brb3.1369] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 06/03/2019] [Accepted: 06/08/2019] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVES Interleukin-33, a newly identified member of interleukin-1 family, had been confirmed to play a crucial role in regulating inflammatory responses in various disease. However, the exact role of interleukin-33 in the disease process of acute ischemic stroke still remains unclear. This study aims to demonstrate the relationship between interleukin-33 levels and long-term functional outcome as well as ischemic stroke recurrence. METHODS Three hundred and four first-ever acute ischemic stroke patients were recruited and basic information and history of all subjects taken within 72 hr on admission. The functional outcome was estimated by Barthel index. The multivariate logistic regression was used to analyze the prognosis, while the Cox proportional hazard model was applied to assess the recurrence risk. RESULTS Out of 304 subjects, 259 patients successfully completed scheduled two-year follow-up. We found that higher interleukin-33 levels correlated positively with better prognosis as compared with those with lower interleukin-33 levels who presented with poorer outcome (62.45 ± 20.50 ng/ml vs. 51.58 ± 19.16 ng/ml, p < .001). After adjustment of all confounders, interleukin-33 was associated with the one-year prognosis with an adjusted odds ratio of 0.956 (95% confidence interval, 0.937-0.976, p < .001). Furthermore, interleukin-33 levels were also closely related to recurrent ischemic stroke with an adjusted hazard ratio of 0.979 (95% confidence interval, 0.961-0.997, p = .025). CONCLUSIONS IL-33 can be used to predict the long-term outcomes and ischemic stroke recurrence in first-ever acute ischemic stroke patients.
Collapse
Affiliation(s)
- Xian-Mei Li
- Department of Rehabilitation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiao-Yang Wang
- Department of Rehabilitation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiao-Wen Feng
- Department of Rehabilitation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Meng-Meng Shao
- Department of Rehabilitation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wen-Fang Liu
- Department of Rehabilitation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qin-Qin Ma
- Department of Rehabilitation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - En-Pei Wang
- Department of Rehabilitation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jie Chen
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Bei Shao
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
34
|
Adrenergic receptor antagonism induces neuroprotection and facilitates recovery from acute ischemic stroke. Proc Natl Acad Sci U S A 2019; 116:11010-11019. [PMID: 31097598 DOI: 10.1073/pnas.1817347116] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Spontaneous waves of cortical spreading depolarization (CSD) are induced in the setting of acute focal ischemia. CSD is linked to a sharp increase of extracellular K+ that induces a long-lasting suppression of neural activity. Furthermore, CSD induces secondary irreversible damage in the ischemic brain, suggesting that K+ homeostasis might constitute a therapeutic strategy in ischemic stroke. Here we report that adrenergic receptor (AdR) antagonism accelerates normalization of extracellular K+, resulting in faster recovery of neural activity after photothrombotic stroke. Remarkably, systemic adrenergic blockade before or after stroke facilitated functional motor recovery and reduced infarct volume, paralleling the preservation of the water channel aquaporin-4 in astrocytes. Our observations suggest that AdR blockers promote cerebrospinal fluid exchange and rapid extracellular K+ clearance, representing a potent potential intervention for acute stroke.
Collapse
|
35
|
Morrison HW, Filosa JA. Stroke and the neurovascular unit: glial cells, sex differences, and hypertension. Am J Physiol Cell Physiol 2019; 316:C325-C339. [PMID: 30601672 DOI: 10.1152/ajpcell.00333.2018] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A functional neurovascular unit (NVU) is central to meeting the brain's dynamic metabolic needs. Poststroke damage to the NVU within the ipsilateral hemisphere ranges from cell dysfunction to complete cell loss. Thus, understanding poststroke cell-cell communication within the NVU is of critical importance. Loss of coordinated NVU function exacerbates ischemic injury. However, particular cells of the NVU (e.g., astrocytes) and those with ancillary roles (e.g., microglia) also contribute to repair mechanisms. Epidemiological studies support the notion that infarct size and recovery outcomes are heterogeneous and greatly influenced by modifiable and nonmodifiable factors such as sex and the co-morbid condition common to stroke: hypertension. The mechanisms whereby sex and hypertension modulate NVU function are explored, to some extent, in preclinical laboratory studies. We present a review of the NVU in the context of ischemic stroke with a focus on glial contributions to NVU function and dysfunction. We explore the impact of sex and hypertension as modifiable and nonmodifiable risk factors and the underlying cellular mechanisms that may underlie heterogeneous stroke outcomes. Most of the preclinical investigative studies of poststroke NVU dysfunction are carried out primarily in male stroke models lacking underlying co-morbid conditions, which is very different from the human condition. As such, the evolution of translational medicine to target the NVU for improved stroke outcomes remains elusive; however, it is attainable with further research.
Collapse
|
36
|
Fang M, Zhong L, Jin X, Cui R, Yang W, Gao S, Lv J, Li B, Liu T. Effect of Inflammation on the Process of Stroke Rehabilitation and Poststroke Depression. Front Psychiatry 2019; 10:184. [PMID: 31031649 PMCID: PMC6470379 DOI: 10.3389/fpsyt.2019.00184] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 03/13/2019] [Indexed: 11/13/2022] Open
Abstract
A considerable body of evidence has shown that inflammation plays an important role in the process of stroke rehabilitation and development of poststroke depression (PSD). However, the specific molecular and cellular mechanisms involved remain unclear. In this review, we summarize how neuroinflammation affects stroke rehabilitation and PSD. We mainly focus on the immune/inflammatory response, involving astrocytes, microglia, monocyte-derived macrophages, cytokines (tumor necrosis factor alpha, interleukin 1), and microRNAs (microRNA-124, microRNA 133b). This review provides new insights into the effect of inflammation on the process of stroke rehabilitation and PSD and potentially offer new therapeutic targets of stroke and PSD.
Collapse
Affiliation(s)
- Meidan Fang
- Department of General Surgery, Second Hospital of Jilin University, Changchun, China
| | - Lili Zhong
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Xin Jin
- Department of Oncology and Hematology, Second Hospital of Jilin University, Changchun, China
| | - Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Wei Yang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Shuohui Gao
- Department of Gastrointestinal Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jing Lv
- Chang Chun University of Chinese Medicine, Changchun, China
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Tongjun Liu
- Department of General Surgery, Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
37
|
Orczykowski ME, Arndt KR, Palitz LE, Kramer BC, Pessina MA, Oblak AL, Finklestein SP, Mortazavi F, Rosene DL, Moore TL. Cell based therapy enhances activation of ventral premotor cortex to improve recovery following primary motor cortex injury. Exp Neurol 2018. [PMID: 29540323 DOI: 10.1016/j.expneurol.2018.03.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Stroke results in enduring damage to the brain which is accompanied by innate neurorestorative processes, such as reorganization of surviving circuits. Nevertheless, patients are often left with permanent residual impairments. Cell based therapy is an emerging therapeutic that may function to enhance the innate neurorestorative capacity of the brain. We previously evaluated human umbilical tissue-derived cells (hUTC) in our non-human primate model of cortical injury limited to the hand area of primary motor cortex. Injection of hUTC 24 h after injury resulted in significantly enhanced recovery of fine motor function compared to vehicle treated controls (Moore et al., 2013). These monkeys also received an injection of Bromodeoxyuridine (BrdU) 8 days after cortical injury to label cells undergoing replication. This was followed by 12 weeks of behavioral testing, which culminated 3 h prior to perfusion in a final behavioral testing session using only the impaired hand. In this session, the neuronal activity initiating hand movements leads to the upregulation of the immediate early gene c-Fos in activated cells. Following perfusion-fixation of the brain, sections were processed using immunohistochemistry to label c-Fos activated cells, pre-synaptic vesicle protein synaptophysin, and BrdU labeled neuroprogenitor cells to investigate the hypothesis that hUTC treatment enhanced behavioral recovery by facilitating reorganization of surviving cortical tissues. Quantitative analysis revealed that c-Fos activated cells were significantly increased in the ipsi- and contra-lesional ventral premotor but not the dorsal premotor cortices in the hUTC treated monkeys compared to placebo controls. Furthermore, the increase in c-Fos activated cells in the ipsi- and contra-lesional ventral premotor cortex correlated with a decrease in recovery time and improved grasp topography. Interestingly, there was no difference between treatment groups in the number of synaptophysin positive puncta in either ipsi- or contra-lesional ventral or dorsal premotor cortices. Nor was there a significant difference in the density of BrdU labeled cells in the subgranular zone of the hippocampus or the subventricular zone of the lateral ventricle. These findings support the hypothesis that hUTC treatment enhances the capacity of the brain to reorganize after cortical injury and that bilateral plasticity in ventral premotor cortex is a critical locus for this recovery of function. This reorganization may be accomplished through enhanced activation of pre-existing circuits within ventral premotor, but it could also reflect ventral premotor projections to the brainstem or spinal cord.
Collapse
Affiliation(s)
- Mary E Orczykowski
- Department of Anatomy & Neurobiology, 72 E. Concord Street, L-1004, Boston University School of Medicine, Boston, MA 02118, USA.
| | - Kevin R Arndt
- Department of Anatomy & Neurobiology, 72 E. Concord Street, L-1004, Boston University School of Medicine, Boston, MA 02118, USA
| | - Lauren E Palitz
- Department of Anatomy & Neurobiology, 72 E. Concord Street, L-1004, Boston University School of Medicine, Boston, MA 02118, USA
| | - Brian C Kramer
- Janssen Scientific Affairs, LLC 800 Ridgeview Drive, Horsham 19044, PA, USA
| | - Monica A Pessina
- Department of Anatomy & Neurobiology, 72 E. Concord Street, L-1004, Boston University School of Medicine, Boston, MA 02118, USA
| | - Adrian L Oblak
- Department of Anatomy & Neurobiology, 72 E. Concord Street, L-1004, Boston University School of Medicine, Boston, MA 02118, USA
| | | | - Farzad Mortazavi
- Department of Anatomy & Neurobiology, 72 E. Concord Street, L-1004, Boston University School of Medicine, Boston, MA 02118, USA
| | - Douglas L Rosene
- Department of Anatomy & Neurobiology, 72 E. Concord Street, L-1004, Boston University School of Medicine, Boston, MA 02118, USA; Yerkes National Primate Research Center, 201 Dowman Drive, Emory University, Atlanta 30322, GA, USA
| | - Tara L Moore
- Department of Anatomy & Neurobiology, 72 E. Concord Street, L-1004, Boston University School of Medicine, Boston, MA 02118, USA; Department of Neurology, 72 E. Concord Street C3, Boston University School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
38
|
Amiri A, Goudarzi R, Amiresmaili M, Iranmanesh F. Cost-effectiveness analysis of tissue plasminogen activator in acute ischemic stroke in Iran. J Med Econ 2018; 21:282-287. [PMID: 29105528 DOI: 10.1080/13696998.2017.1401545] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
AIMS Tissue plasminogen activator (tPA) is used to treat acute ischemic stroke up to 4.5 h after symptom onset. Its cost-effectiveness in developing countries is not specified yet. This study aimed to study cost-effectiveness of tPA in Iran. METHODS This is a cost-effectiveness analysis from the perspective of the third party payer to compare IV tPA with no tPA of ischemic stroke. A Markov model with a lifetime horizon was used to analyze the costs and outcomes. Cost data were extracted from the 94 patients admitted in two hospitals in Iran. All costs were calculated based on US dollars in 2016. Quality-adjusted life years (QALY) were extracted from previously published literature. Cost-effectiveness was determined by calculating ICER by TreeAge Pro 2011 software. RESULTS Lifetime costs of no tPA strategy were higher than tPA ($10,718 in the no tPA group compared with $8,796 in the tPA group). The tPA arm gained 0.20 QALY compared with no tPA. ICER was $8,471 per QALY. ICER value suggests that tPA is cost-effective compared with no tPA. LIMITATIONS The limitations of the present study are the reliance on calculated QALY value of other countries and difficulty in accessing patients treated with tPA. CONCLUSIONS The balance of hospitalization and rehabilitation costs and QALYs support the conclusion that treatment with intravenous tPA in the 4.5-h time window is cost-effective from the perspectives of the third party payer and inclusion of tPA in the insurance benefit package being reasonable.
Collapse
Affiliation(s)
- Asrin Amiri
- a Faculty of Management and Medical Informatics , Kerman University of Medical Sciences , Kerman , Iran
| | - Reza Goudarzi
- b Modeling in Health Research Center, Institute for Futures Studies in Health, Kerman University of Medical Sciences , Kerman , Iran
| | - Mohammadreza Amiresmaili
- a Faculty of Management and Medical Informatics , Kerman University of Medical Sciences , Kerman , Iran
| | - Farhad Iranmanesh
- c Neurology Research Center, Kerman University of Medical Sciences , Kerman , Iran
| |
Collapse
|
39
|
Jones EV, Bernardinelli Y, Zarruk JG, Chierzi S, Murai KK. SPARC and GluA1-Containing AMPA Receptors Promote Neuronal Health Following CNS Injury. Front Cell Neurosci 2018; 12:22. [PMID: 29449802 PMCID: PMC5799273 DOI: 10.3389/fncel.2018.00022] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 01/15/2018] [Indexed: 12/22/2022] Open
Abstract
The proper formation and maintenance of functional synapses in the central nervous system (CNS) requires communication between neurons and astrocytes and the ability of astrocytes to release neuromodulatory molecules. Previously, we described a novel role for the astrocyte-secreted matricellular protein SPARC (Secreted Protein, Acidic and Rich in Cysteine) in regulating α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) and plasticity at developing synapses. SPARC is highly expressed by astrocytes and microglia during CNS development but its level is reduced in adulthood. Interestingly, SPARC has been shown to be upregulated in CNS injury and disease. However, the role of SPARC upregulation in these contexts is not fully understood. In this study, we investigated the effect of chronic SPARC administration on glutamate receptors on mature hippocampal neuron cultures and following CNS injury. We found that SPARC treatment increased the number of GluA1-containing AMPARs at synapses and enhanced synaptic function. Furthermore, we determined that the increase in synaptic strength induced by SPARC could be inhibited by Philanthotoxin-433, a blocker of homomeric GluA1-containing AMPARs. We then investigated the effect of SPARC treatment on neuronal health in an injury context where SPARC expression is upregulated. We found that SPARC levels are increased in astrocytes and microglia following middle cerebral artery occlusion (MCAO) in vivo and oxygen-glucose deprivation (OGD) in vitro. Remarkably, chronic pre-treatment with SPARC prevented OGD-induced loss of synaptic GluA1. Furthermore, SPARC treatment reduced neuronal death through Philanthotoxin-433 sensitive GluA1 receptors. Taken together, this study suggests a novel role for SPARC and GluA1 in promoting neuronal health and recovery following CNS damage.
Collapse
Affiliation(s)
- Emma V Jones
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, QC, Canada
| | | | - Juan G Zarruk
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, QC, Canada
| | - Sabrina Chierzi
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, QC, Canada
| | - Keith K Murai
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, QC, Canada
| |
Collapse
|
40
|
Feng Y, Liao S, Wei C, Jia D, Wood K, Liu Q, Wang X, Shi FD, Jin WN. Infiltration and persistence of lymphocytes during late-stage cerebral ischemia in middle cerebral artery occlusion and photothrombotic stroke models. J Neuroinflammation 2017; 14:248. [PMID: 29246244 PMCID: PMC5732427 DOI: 10.1186/s12974-017-1017-0] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 11/29/2017] [Indexed: 01/05/2023] Open
Abstract
Background Evidence suggests that brain infiltration of lymphocytes contributes to acute neural injury after cerebral ischemia. However, the spatio-temporal dynamics of brain-infiltrating lymphocytes during the late stage after cerebral ischemia remains unclear. Methods C57BL/6 (B6) mice were subjected to sham, photothrombosis, or 60-min transient middle cerebral artery occlusion (MCAO) procedures. Infarct volume, neurodeficits, production of reactive oxygen species (ROS) and inflammatory factors, brain-infiltrating lymphocytes, and their activation as well as pro-inflammatory cytokine IFN-γ production were assessed. Brain-infiltrating lymphocytes were also measured in tissue sections from post-mortem patients after ischemic stroke by immunostaining. Results In mice subjected to transient MCAO or photothrombotic stroke, we found that lymphocyte infiltration persists in the ischemic brain until at least day 14 after surgery, during which brain infarct volume significantly diminished. These brain-infiltrating lymphocytes express activation marker CD69 and produce proinflammatory cytokines such as IFN-γ, accompanied with a sustained increase of reactive oxygen species (ROS) and inflammatory cytokines release in the brain. In addition, brain-infiltrating lymphocytes were observed in post-mortem brain sections from patients during the late stage of ischemic stroke. Conclusion Our results demonstrate that brain-infiltration of lymphocytes persists after the acute stage of cerebral ischemia, facilitating future advanced studies to reveal the precise role of lymphocytes during late stage of stroke. Electronic supplementary material The online version of this article (10.1186/s12974-017-1017-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yan Feng
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Center for Neuroinflammation, Beijing TianTan Hospital, Beijing, 100070, China
| | - Shiwei Liao
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Changjuan Wei
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Dongmei Jia
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Kristofer Wood
- Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, 85013, AZ, USA
| | - Qiang Liu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, 85013, AZ, USA
| | - Xiaoying Wang
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, 02129, MA, USA
| | - Fu-Dong Shi
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Center for Neuroinflammation, Beijing TianTan Hospital, Beijing, 100070, China.,Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, 85013, AZ, USA
| | - Wei-Na Jin
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China. .,Center for Neuroinflammation, Beijing TianTan Hospital, Beijing, 100070, China. .,Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, 85013, AZ, USA.
| |
Collapse
|