1
|
Gao L, Wang B, Cui X, Xia L, Li X, Figueredo YN, Li D, Liu K, Wang H, Jin M. Neochlorogenic acid ameliorates Alzheimer's disease-like pathology via scavenging oxidative stress and restoring blood-brain barrier function in zebrafish. Prog Neuropsychopharmacol Biol Psychiatry 2025; 138:111334. [PMID: 40122506 DOI: 10.1016/j.pnpbp.2025.111334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 03/03/2025] [Accepted: 03/12/2025] [Indexed: 03/25/2025]
Abstract
Alzheimer's disease is the most widespread neurodegenerative disease characterized by insidious onset and slow progression. At present, most available medications serve to attenuate the progression of Alzheimer's disease with side effects and drug resistance. Neochlorogenic acid is a natural polyphenolic compound with excellent antioxidant properties. Based on zebrafish Alzheimer's disease model induced by AlCl3, we found that neochlorogenic acid significantly improved motor dysfunction, reduced brain cell apoptosis, and Aβ plaque. Because of antioxidant stress and improvement of blood-brain barrier dysfunction are important in treating Alzheimer's disease, we explored the interaction between these two mechanisms in alleviating the pathological course of Alzheimer's disease. Neochlorogenic acid inhibited the overproduction of reactive oxygen species, suppressed the gene expression encoding antioxidant-related proteins, and protected brain cell integrity while enhancing Nrf2, improving blood-brain barrier nerve resilience. Meanwhile, neochlorogenic acid attenuated blood-brain barrier dysfunction in Alzheimer's disease zebrafish by reducing blood hemoglobin leakage and upregulating the gene expression encoding blood-brain barrier endothelial cell-related proteins, resulting in reactive oxygen species in a controllable state. In conclusion, our research suggests that neochlorogenic acid ameliorates Alzheimer's disease-like pathology by inhibiting oxidative stress and restoring blood-brain barrier function, indicating that neochlorogenic acid may be a potential drug for treating Alzheimer's disease.
Collapse
Affiliation(s)
- Li Gao
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; School of Psychology and Mental Health, North China University of Science and Technology, 21 Bohai Road, Tang'shan 063210, Hebei Province, People's Republic of China
| | - Baokun Wang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; School of Pharmacy, Qingdao University, Qing'dao 266003, Shandong Province, People's Republic of China
| | - Xiaotong Cui
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China
| | - Lijie Xia
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; School of Psychology and Mental Health, North China University of Science and Technology, 21 Bohai Road, Tang'shan 063210, Hebei Province, People's Republic of China
| | - Xinjia Li
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China
| | - Yanier Nuñez Figueredo
- Centro de Investigación y Desarrollo de Medicamentos (CIDEM), Ave 26, No.1605, e/Boyeros y Puentes Grandes, La Habana CP10600, Cuba
| | - Dong Li
- R&D Department, Jinan Perfect Biological Technology Co., Ltd., Jinan 250101, Shandong Province, People's Republic of China
| | - Kechun Liu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China
| | - Haitao Wang
- School of Psychology and Mental Health, North China University of Science and Technology, 21 Bohai Road, Tang'shan 063210, Hebei Province, People's Republic of China.
| | - Meng Jin
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; School of Psychology and Mental Health, North China University of Science and Technology, 21 Bohai Road, Tang'shan 063210, Hebei Province, People's Republic of China.
| |
Collapse
|
2
|
Beard RS, Hoettels BA, McAllister JM, Meegan JE, Wertz TS, Self DA, Hrkach DE, Greiner D, Chapman K, Villalba N, Yang X, Cha BJ, Jorcyk CL, Oxford JT, Wu MH, Yuan SY. Progression of experimental autoimmune encephalomyelitis in mice and neutrophil-mediated blood-brain barrier dysfunction requires non-muscle myosin light chain kinase. J Cereb Blood Flow Metab 2025:271678X251318620. [PMID: 39917847 PMCID: PMC11806455 DOI: 10.1177/0271678x251318620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 12/22/2024] [Accepted: 01/10/2025] [Indexed: 02/11/2025]
Abstract
Blood-brain barrier (BBB) dysfunction occurs in numerous central nervous system disorders. Unfortunately, a limited understanding of the mechanisms governing barrier function hinders the identification and assessment of BBB-targeted therapies. Previously, we found that non-muscle myosin light chain kinase (nmMLCK) negatively regulates the tight junction protein claudin-5 in brain microvascular endothelial cells (BMVECs) under inflammatory conditions. Here, we used complementary animal and primary cell co-culture models to further investigate nmMLCK and claudin-5 during neuroinflammation. We found that nmMLCK-knockout mice resisted experimental autoimmune encephalomyelitis (EAE), including paralysis, demyelination, neutrophil infiltration, and BBB dysfunction. However, transiently silencing claudin-5 culminated in a fulminant disease course. In parallel, we found that neutrophil-secreted factors triggered a biphasic loss in the barrier quality of wild-type BMVEC monolayers, plus pronounced neutrophil migration during the second phase. Conversely, nmMLCK-knockout monolayers resisted barrier dysfunction and neutrophil migration. Lastly, we found an inverse relationship between claudin-5 expression in BMVECs and neutrophil migration. Overall, our findings support a pathogenic role for nmMLCK in BMVECs during EAE that includes BBB dysfunction and neutrophil infiltration, reveal that claudin-5 contributes to the immune barrier properties of BMVECs, and underscore the harmful effects of claudin-5 loss during neuroinflammation.
Collapse
Affiliation(s)
- Richard S Beard
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
- Department of Biological Sciences, Boise State University, Boise, ID, USA
- Biomedical Research Institute, Boise State University, Boise, ID, USA
| | - Brian A Hoettels
- Department of Biological Sciences, Boise State University, Boise, ID, USA
- Biomedical Research Institute, Boise State University, Boise, ID, USA
| | - Jessica M McAllister
- Department of Biological Sciences, Boise State University, Boise, ID, USA
- Biomedical Research Institute, Boise State University, Boise, ID, USA
| | - Jamie E Meegan
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Travis S Wertz
- Department of Biological Sciences, Boise State University, Boise, ID, USA
- Biomedical Research Institute, Boise State University, Boise, ID, USA
| | - Desiree A Self
- Department of Biological Sciences, Boise State University, Boise, ID, USA
- Biomedical Research Institute, Boise State University, Boise, ID, USA
| | - Dylan E Hrkach
- Department of Biological Sciences, Boise State University, Boise, ID, USA
- Biomedical Research Institute, Boise State University, Boise, ID, USA
| | - Daniel Greiner
- Department of Biological Sciences, Boise State University, Boise, ID, USA
| | - Kristina Chapman
- Department of Biological Sciences, Boise State University, Boise, ID, USA
- Biomedical Research Institute, Boise State University, Boise, ID, USA
| | - Nuria Villalba
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Xiaoyuan Yang
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Byeong J Cha
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Cheryl L Jorcyk
- Department of Biological Sciences, Boise State University, Boise, ID, USA
| | - Julia T Oxford
- Department of Biological Sciences, Boise State University, Boise, ID, USA
- Biomedical Research Institute, Boise State University, Boise, ID, USA
| | - Mack H Wu
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Sarah Y Yuan
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| |
Collapse
|
3
|
Deng HJ, Xu YH, Wu K, Li YC, Zhang YJ, Yu HF, Li C, Xu D, Wang F. The sentinel against brain injury post-subarachnoid hemorrhage: efferocytosis of erythrocytes by leptomeningeal lymphatic endothelial cells. Theranostics 2025; 15:2487-2509. [PMID: 39990222 PMCID: PMC11840724 DOI: 10.7150/thno.103701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 01/06/2025] [Indexed: 02/25/2025] Open
Abstract
Rationale: The clearance of extravasated erythrocytes represents the most reasonable strategy against brain injury post-subarachnoid hemorrhage (SAH). There is little knowledge about the autologous clearance of extravasated erythrocytes post-SAH. The leptomeningeal lymphatic endothelial cells (LLECs) have been less studied functionally, which were firstly harvested and cultured in vitro by our group previously and are probably related to the clearance of extravasated erythrocytes post-SAH for they closely surround subarachnoid space. Methods: We established a SAH animal model, employed primary LLECs in vitro, mimicked the conditions of the SAH in vitro, performed RNA sequencing, and transfected LLECs with adenovirus and adeno-associated virus both in vivo and in vitro to reveal the molecular mechanisms of efferocytosis of erythrocytes by LLECs and its neuroprotection post-SAH. Results: Firstly, we demonstrated the eryptosis-initiated degradation of extravasated erythrocytes in vitro. Furthermore, we found LLECs preferentially adhered and engulfed apoptotic erythrocytes in vivo and in vitro while sparing from intact erythrocytes, suggesting their novel capacity in the efferocytosis of erythrocytes. Additionally, the efferocytosis of erythrocytes by LLECs plays a role on neuroprotection via improving neurological functions, maintaining neurostructural integrity, and alleviating neuropathological consequences post-SAH. During efferocytosis, phosphatidylserine (PS) and phosphatidylserine receptor (PSR) mediated the recognition of apoptotic erythrocytes by LLECs. We also confirmed that NHL repeat-containing 2 (NHLRC2) positively regulated the efferocytosis of erythrocytes by LLECs to serve as a central regulator in it mediated neuroprotection post-SAH. Conclusions: This study elucidated the efferocytosis of erythrocytes by LLECs and subsequently neuroprotection post-SAH. These findings highlight a prompt, efficient, and regulable pathway for the autologous clearance of extravasated erythrocytes that performs as a sentinel against brain injury post-SAH.
Collapse
Affiliation(s)
- Hong-Ji Deng
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yun-Huo Xu
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Kun Wu
- Department of Clinical Laboratory, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yun-Cong Li
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yong-Jin Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China
- Clinical Medical Research Center, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Han-Fu Yu
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Chong Li
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Dan Xu
- Department of Dermatology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Fei Wang
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
4
|
Wroe W, Dienel A, Hong S, Matsumura K, Guzman J, Torres K, Bernal A, Zeineddine HA, Pandit PT, Blackburn SL, McBride DW. Incidence and Factors in Delayed Neurological Deficits after Subarachnoid Hemorrhage in Mice. BRAIN HEMORRHAGES 2024; 5:99-106. [PMID: 39830728 PMCID: PMC11741540 DOI: 10.1016/j.hest.2023.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025] Open
Abstract
Background Delayed cerebral ischemia (DCI) is one of the most feared complications in aneurysmal subarachnoid hemorrhage (SAH). Animal models are crucial to studying the disease mechanisms and potential treatments. DCI in rodents was thought to not exist; herein we examine literature and our experience with DCI in rodents. Methods Daily behavioral performance was assessed every day from day 1 to up to 7 days post-SAH on mice from 5 different studies that used the endovascular perforation model. Performance was graded using an 8-test sensorimotor neuroscore previously described. The daily neuroscore was then used to identify the incidence and timing of delayed neurological deficits, a clinical surrogate for DCI. A total number of 298 mice (134 males, 164 females) were subjected to SAH. Fifty-one mice had histological staining done to identify infarct volume. Results The overall incidence of DND was 33.9%; 27.6% in males and 39.0% in females, but this difference was not statistically significant. The overall incidence of delayed death was 21.1%, and there was no significant difference for delayed mortality in females versus male mice. There is a non-statistically significant trend towards increased infarct volume in mice suffering DND. Conclusions Mice with endovascular puncture induced SAH develop DND at rates comparable to human patients. Future work needs to correlate the DND seen with decreased regional cerebral blood flow, another hallmark of DCI, but in spite of this need, researchers may use the murine models to test therapies for DCI after SAH.
Collapse
Affiliation(s)
- William Wroe
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Ari Dienel
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Sungha Hong
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Kanako Matsumura
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Jose Guzman
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Kiara Torres
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | | | - Hussein A. Zeineddine
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Peeyush Thankamani Pandit
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Spiros L. Blackburn
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Devin W. McBride
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
5
|
Mu C, Gao M, Xu W, Sun X, Chen T, Xu H, Qiu H. Mechanisms of microRNA-132 in central neurodegenerative diseases: A comprehensive review. Biomed Pharmacother 2024; 170:116029. [PMID: 38128185 DOI: 10.1016/j.biopha.2023.116029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/12/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023] Open
Abstract
MicroRNA-132 (miR-132) is a highly conserved molecule that plays a crucial regulatory role in central nervous system (CNS) disorders. The expression levels of miR-132 exhibit variability in various neurological disorders and have been closely linked to disease onset and progression. The expression level of miR-132 in the CNS is regulated by a diverse range of stimuli and signaling pathways, including neuronal migration and integration, dendritic outgrowth, and complexity, synaptogenesis, synaptic plasticity, as well as inflammation and apoptosis activation. The aberrant expression of miR-132 in various central neurodegenerative diseases has garnered widespread attention. Clinical studies have revealed altered miR-132 expression levels in both chronic and acute CNS diseases, positioning miR-132 as a potential biomarker or therapeutic target. An in-depth exploration of miR-132 holds the promise of enhancing our understanding of the mechanisms underlying CNS diseases, thereby offering novel insights and strategies for disease diagnosis and treatment. It is anticipated that this review will assist researchers in recognizing the potential value of miR-132 and in generating innovative ideas for clinical trials related to CNS degenerative diseases.
Collapse
Affiliation(s)
- Chenxi Mu
- Basic Medical College, Jiamusi University, Jiamusi 154007, Heilongjiang, China; Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, Jiamusi 154007, Heilongjiang, China
| | - Meng Gao
- Basic Medical College, Jiamusi University, Jiamusi 154007, Heilongjiang, China; Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, Jiamusi 154007, Heilongjiang, China
| | - Weijing Xu
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, Jiamusi 154007, Heilongjiang, China; School of Public Health, Jiamusi University, Jiamusi 154007, Heilongjiang, China
| | - Xun Sun
- Basic Medical College, Jiamusi University, Jiamusi 154007, Heilongjiang, China; Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, Jiamusi 154007, Heilongjiang, China
| | - Tianhao Chen
- Basic Medical College, Jiamusi University, Jiamusi 154007, Heilongjiang, China; Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, Jiamusi 154007, Heilongjiang, China
| | - Hui Xu
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, Jiamusi 154007, Heilongjiang, China.
| | - Hongbin Qiu
- School of Public Health, Jiamusi University, Jiamusi 154007, Heilongjiang, China.
| |
Collapse
|
6
|
Xie Y, He W, Ma L, Ren R, Yang S, Lu Q. Endothelial TREM-1 receptor regulates the blood-brain barrier integrity after intracerebral hemorrhage in mice via SYK/β-catenin signaling. CNS Neurosci Ther 2023; 29:3228-3238. [PMID: 37170484 PMCID: PMC10580358 DOI: 10.1111/cns.14255] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 04/11/2023] [Accepted: 04/24/2023] [Indexed: 05/13/2023] Open
Abstract
BACKGROUND Intracerebral hemorrhage (ICH) is a high mortality and disability stroke subtype. Destruction of the blood-brain barrier (BBB) is a crucial contributor to brain edema and neurological deficit after ICH. Triggering receptor expressed on myeloid cells 1 (TREM-1) has been reported to be expressed in endothelial cells, but its role in ICH remains unclear. This study aims to evaluate the role of TREM-1 on BBB permeability after ICH in mice. METHODS Two hundred and forty-two CD1 mice were used in this study. The ICH model was established by collagenase injection. LP17 was administered intranasally at 2 or 8 h after ICH to inhibit TREM-1. To explore the underlying mechanism, SYK activation CRISPR was administered intracerebroventricularly with LP17, and Anti-mouse TREM-1 rat IgG2a (a specific TREM-1 agonist) was injected intracerebroventricularly with R406 (a specific SYK inhibitor) intraperitoneally. Neurobehavioral outcome, brain water content, BBB permeability, and protein expression were evaluated. RESULTS The expression level of the TREM-1 receptor increased rapidly as early as 6 h after ICH, and it was mainly expressed on the endotheliocytes in the neurovascular unit. Early and delayed administration of LP17 significantly decreased brain edema and improved neurobehavioral outcomes at 24 h after ICH. LP17 reduced the BBB permeability by increasing β-catenin, claudin-5 and ZO-1 expression. Furthermore, SYK activation CRISPR abolished the beneficial effect of LP17 on the expression of the above junction molecules. Meanwhile, R406 reversed the impact of the TREM-1 activator on the downregulation of β-catenin, claudin-5 and ZO-1 expression. CONCLUSIONS This study demonstrated that TREM-1 deteriorated BBB permeability via modulating the expression of interendothelial junction molecules after ICH, and this regulation is partly mediated by the SYK/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Yonglin Xie
- Department of Emergency, Sir Run Run Shaw HospitalZhejiang University, School of MedicineHangzhouChina
| | - Wei He
- Department of Pharmacy, Second Affiliated HospitalZhejiang University, School of MedicineHangzhouChina
| | - Li Ma
- Department of Neurosurgery, Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
| | - Reng Ren
- Department of Neurointensive Care Unit, The Second Affiliated HospitalZhejiang University, School of MedicineHangzhouChina
| | - Shuxu Yang
- Department of Neurosurgery, Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
| | - Qin Lu
- Department of Neurosurgery, Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
| |
Collapse
|
7
|
Zheng W, Li W, Zeng Y, Yuan H, Yang H, Chen R, Zhu A, Wu J, Song Z, Yan W. Endogenous FGF21 attenuates blood-brain barrier disruption in penumbra after delayed recanalization in MCAO rats through FGFR1/PI3K/Akt pathway. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2023; 48:648-662. [PMID: 37539567 PMCID: PMC10930414 DOI: 10.11817/j.issn.1672-7347.2023.220380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Indexed: 08/05/2023]
Abstract
OBJECTIVES Restoration of blood circulation within "time window" is the principal treating goal for treating acute ischemic stroke. Previous studies revealed that delayed recanalization might cause serious ischemia/reperfusion injury. However, plenty of evidences showed delayed recanalization improved neurological outcomes in acute ischemic stroke. This study aims to explore the role of delayed recanalization on blood-brain barrier (BBB) in the penumbra (surrounding ischemic core) and neurological outcomes after middle cerebral artery occlusion (MCAO). METHODS Recanalization was performed on the 3rd day after MCAO. BBB disruption was tested by Western blotting, Evans blue dye, and immunofluorescence staining. Infarct volume and neurological outcomes were evaluated on the 7th day after MCAO. The expression of fibroblast growth factor 21 (FGF21), fibroblast growth factor receptor 1 (FGFR1), phosphatidylinositol-3-kinase (PI3K), and serine/threonine kinase (Akt) in the penumbra were observed by immunofluorescence staining and/or Western blotting. RESULTS The extraversion of Evans blue, IgG, and albumin increased surrounding ischemic core after MCAO, but significantly decreased after recanalization. The expression of Claudin-5, Occludin, and zona occludens 1 (ZO-1) decreased surrounding ischemic core after MCAO, but significantly increased after recanalization. Infarct volume reduced and neurological outcomes improved following recanalization (on the 7th day after MCAO). The expressions of Claudin-5, Occludin, and ZO-1 decreased surrounding ischemic core following MCAO, which were up-regulated corresponding to the increases of FGF21, p-FGFR1, PI3K, and p-Akt after recanalization. Intra-cerebroventricular injection of FGFR1 inhibitor SU5402 down-regulated the expression of PI3K, p-Akt, Occludin, Claudin-5, and ZO-1 in the penumbra, which weakened the beneficial effects of recanalization on neurological outcomes after MCAO. CONCLUSIONS Delayed recanalization on the 3rd day after MCAO increases endogenous FGF21 in the penumbra and activates FGFR1/PI3K/Akt pathway, which attenuates BBB disruption in the penumbra and improves neurobehavior in MCAO rats.
Collapse
Affiliation(s)
- Wen Zheng
- Department of Neurology, Third Xiangya Hospital, Central South University, Changsha 410013.
| | - Wenjun Li
- Department of Neurology, Third Xiangya Hospital, Central South University, Changsha 410013
| | - Yini Zeng
- Department of Neurology, Third Xiangya Hospital, Central South University, Changsha 410013
| | - Hui Yuan
- Department of Neurology, Third Xiangya Hospital, Central South University, Changsha 410013
| | - Heng Yang
- Department of Neurology, Third Xiangya Hospital, Central South University, Changsha 410013
| | - Ru Chen
- Department of Neurology, Third Xiangya Hospital, Central South University, Changsha 410013
| | - Anding Zhu
- Department of Neurology, Third Xiangya Hospital, Central South University, Changsha 410013
| | - Jinze Wu
- Department of Neurology, Third Xiangya Hospital, Central South University, Changsha 410013
| | - Zhi Song
- Department of Neurology, Third Xiangya Hospital, Central South University, Changsha 410013
| | - Wenguang Yan
- Department of Rihabilitation Medicine, Third Xiangya Hospital, Central South University, Changsha 410013, China.
| |
Collapse
|
8
|
Lin J, Xu Y, Guo P, Chen YJ, Zhou J, Xia M, Tan B, Liu X, Feng H, Chen Y. CCL5/CCR5-mediated peripheral inflammation exacerbates blood‒brain barrier disruption after intracerebral hemorrhage in mice. J Transl Med 2023; 21:196. [PMID: 36918921 PMCID: PMC10015963 DOI: 10.1186/s12967-023-04044-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 03/08/2023] [Indexed: 03/16/2023] Open
Abstract
BACKGROUND Owing to metabolic disequilibrium and immune suppression, intracerebral hemorrhage (ICH) patients are prone to infections; according to a recent global analysis of stroke cases, approximately 10 million new-onset ICH patients had experienced concurrent infection. However, the intrinsic mechanisms underlying the effects of infection related peripheral inflammation after ICH remain unclear. METHODS Lipopolysaccharide (LPS) was intraperitoneally injected into ICH model mice to induce peripheral inflammation. Neurobehavioral deficits, blood‒brain barrier (BBB) disruption, and the expression of CCR5, JAK2, STAT3, and MMP9 were evaluated after treatment with recombinant CCL5 (rCCL5) (a CCR5 ligand), maraviroc (MVC) (an FDA-approved selective CCR5 antagonist), or JAK2 CRISPR plasmids. RESULTS Our study revealed that severe peripheral inflammation increased CCL5/CCR5 axis activation in multiple inflammatory cell types, including microglia, astrocytes, and monocytes, and aggravated BBB disruption and neurobehavioral dysfunction after ICH, possibly in part through the JAK2/STAT3 signaling pathway. CONCLUSIONS CCR5 might be a potential target for the clinical treatment of infection-induced exacerbation of BBB disruption following ICH.
Collapse
Affiliation(s)
- Jie Lin
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.,Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Ya Xu
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.,Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Peiwen Guo
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.,Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yù-Jié Chen
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.,Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jiru Zhou
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.,Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Min Xia
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.,Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Binbin Tan
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.,Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xin Liu
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Hua Feng
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China. .,Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China. .,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Yujie Chen
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China. .,Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China. .,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| |
Collapse
|
9
|
Cheng M, Yang Z, Qiao L, Yang Y, Deng Y, Zhang C, Mi T. AGEs induce endothelial cells senescence and endothelial barrier dysfunction via miR-1-3p/MLCK signaling pathways. Gene 2022; 851:147030. [DOI: 10.1016/j.gene.2022.147030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 10/10/2022] [Accepted: 10/31/2022] [Indexed: 11/08/2022]
|
10
|
Hemorrhagic Cerebral Insults and Secondary Takotsubo Syndrome: Findings in a Novel In Vitro Model Using Human Blood Samples. Int J Mol Sci 2022; 23:ijms231911557. [PMID: 36232860 PMCID: PMC9569517 DOI: 10.3390/ijms231911557] [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: 07/18/2022] [Revised: 09/23/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
Intracranial hemorrhage results in devastating forms of cerebral damage. Frequently, these results also present with cardiac dysfunction ranging from ECG changes to Takotsubo syndrome (TTS). This suggests that intracranial bleeding due to subarachnoid hemorrhage (SAH) disrupts the neuro-cardiac axis leading to neurogenic stress cardiomyopathy (NSC) of different degrees. Following this notion, SAH and secondary TTS could be directly linked, thus contributing to poor outcomes. We set out to test if blood circulation is the driver of the brain-heart axis by investigating serum samples of TTS patients. We present a novel in vitro model combining SAH and secondary TTS to mimic the effects of blood or serum, respectively, on blood-brain barrier (BBB) integrity using in vitro monolayers of an established murine model. We consistently demonstrated decreased monolayer integrity and confirmed reduced Claudin-5 and Occludin levels by RT-qPCR and Western blot and morphological reorganization of actin filaments in endothelial cells. Both tight junction proteins show a time-dependent reduction. Our findings highlight a faster and more prominent disintegration of BBB in the presence of TTS and support the importance of the bloodstream as a causal link between intracerebral bleeding and cardiac dysfunction. This may represent potential targets for future therapeutic inventions in SAH and TTS.
Collapse
|
11
|
Dienel A, Hong SH, Guzman J, Peeyush KT, Blackburn SL, McBride DW. Confirming Subarachnoid Hemorrhage Induction in the Endovascular Puncture Mouse Model. BRAIN HEMORRHAGES 2022; 3:111-116. [PMID: 39831000 PMCID: PMC11741539 DOI: 10.1016/j.hest.2022.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Background and Purpose Experimental studies of subarachnoid hemorrhage (SAH) using the endovascular perforation model routinely use either cerebral blood flow (CBF) or intracranial pressure (ICP) monitoring to confirm SAH induction. We designed this study to test the hypothesis that contralateral CBF is a better marker of SAH induction than ipsilateral CBF. Methods Sixty-eight adult C57BL/6J mice were subjected to endovascular perforation. Mice were monitored using laser Doppler (for relative CBF) or ICP probes. Results The ipsilateral CBF significantly decreased prior to ICP rising. However, reduction of the CBF in the contralateral hemisphere occurred at the same time as the rise in ICP. When CBF is monitored on both hemispheres simultaneously, the drop in ipsilateral CBF precedes the drop in the contralateral CBF. Conclusions The most suitable method for confirming puncture and induction of SAH in the endovascular mouse model is by ICP. Monitoring the CBF of the contralateral hemisphere is also able to detect the moment of SAH induction. However, monitoring the ipsilateral CBF is not satisfactory for determining puncture and induction of SAH due to the changes in CBF caused by blood flow occlusion from the endovascular filament as it enters into the circle of Willis.
Collapse
Affiliation(s)
- Ari Dienel
- The Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Sung-Ha Hong
- The Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Jose Guzman
- The Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Kumar T. Peeyush
- The Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Spiros L. Blackburn
- The Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Devin W. McBride
- The Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
12
|
Solár P, Zamani A, Lakatosová K, Joukal M. The blood-brain barrier and the neurovascular unit in subarachnoid hemorrhage: molecular events and potential treatments. Fluids Barriers CNS 2022; 19:29. [PMID: 35410231 PMCID: PMC8996682 DOI: 10.1186/s12987-022-00312-4] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/24/2022] [Indexed: 12/12/2022] Open
Abstract
The response of the blood-brain barrier (BBB) following a stroke, including subarachnoid hemorrhage (SAH), has been studied extensively. The main components of this reaction are endothelial cells, pericytes, and astrocytes that affect microglia, neurons, and vascular smooth muscle cells. SAH induces alterations in individual BBB cells, leading to brain homeostasis disruption. Recent experiments have uncovered many pathophysiological cascades affecting the BBB following SAH. Targeting some of these pathways is important for restoring brain function following SAH. BBB injury occurs immediately after SAH and has long-lasting consequences, but most changes in the pathophysiological cascades occur in the first few days following SAH. These changes determine the development of early brain injury as well as delayed cerebral ischemia. SAH-induced neuroprotection also plays an important role and weakens the negative impact of SAH. Supporting some of these beneficial cascades while attenuating the major pathophysiological pathways might be decisive in inhibiting the negative impact of bleeding in the subarachnoid space. In this review, we attempt a comprehensive overview of the current knowledge on the molecular and cellular changes in the BBB following SAH and their possible modulation by various drugs and substances.
Collapse
Affiliation(s)
- Peter Solár
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, 625 00, Brno, Czech Republic
- Department of Neurosurgery, Faculty of Medicine, Masaryk University and St. Anne's University Hospital Brno, Pekařská 53, 656 91, Brno, Czech Republic
| | - Alemeh Zamani
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, 625 00, Brno, Czech Republic
| | - Klaudia Lakatosová
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, 625 00, Brno, Czech Republic
| | - Marek Joukal
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, 625 00, Brno, Czech Republic.
| |
Collapse
|
13
|
Yan J, Xu W, Lenahan C, Huang L, Ocak U, Wen J, Li G, He W, Le C, Zhang JH, Mo L, Tang J. Met-RANTES preserves the blood–brain barrier through inhibiting CCR1/SRC/Rac1 pathway after intracerebral hemorrhage in mice. Fluids Barriers CNS 2022; 19:7. [PMID: 35062973 PMCID: PMC8781527 DOI: 10.1186/s12987-022-00305-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 01/06/2022] [Indexed: 11/27/2022] Open
Abstract
Background C–C chemokine receptor type 1 (CCR1) and its endogenous ligand, CCL5, participate in the pathogenesis of neuroinflammatory diseases. However, much remains unknown regarding CCL5/CCR1 signaling in blood–brain barrier (BBB) permeability after intracerebral hemorrhage (ICH). Methods A total of 250 CD1 male mice were used and ICH was induced via autologous whole blood injection. Either Met-RANTES, a selective CCR1 antagonist, or Met-RANTES combined with a Rac1 CRISPR activator was administered to the mice 1 h after ICH. Post-ICH assessments included neurobehavioral tests, brain water content, BBB integrity, hematoma volume, Western blot, and immunofluorescence staining. The CCR1 ligand, rCCL5, and SRC CRISPR knockout in naïve mice were used to further elucidate detrimental CCL5/CCR1/SRC signaling. Results Brain endogenous CCR1 and CCL5 were upregulated after ICH in mice with a peak at 24 h, and CCR1 was expressed in endothelial cells, astrocytes, and neurons. Met-R treatment reduced brain edema and neurobehavioral impairment, as well as preserved BBB integrity and tight junction protein expression in ICH mice. Met-R treatment decreased expression of p-SRC, Rac1, albumin, and MMP9, but increased claudin-5, occludin, and ZO-1 tight junction proteins after ICH. These effects were regressed using the Rac1 CRISPR activator. Administration of rCCL5 in naïve mice increased expression of p-SRC, Rac1, albumin, and MMP9, but decreased levels of claudin-5, occludin, and ZO-1 tight junction proteins. These effects in naïve mice were reversed with SRC CRISPR (KO). Conclusions Our findings demonstrate that CCR5 inhibition by Met-R improves neurological deficits after ICH by preserving BBB integrity through inhibiting CCR1/SRC/Rac1 signaling pathway in mice. Thus, Met-R has therapeutic potential in the management of ICH patients. Supplementary Information The online version contains supplementary material available at 10.1186/s12987-022-00305-3.
Collapse
|
14
|
Cai L, Ge B, Xu S, Chen X, Yang H. Up-regulation of circARF3 reduces blood-brain barrier damage in rat subarachnoid hemorrhage model via miR-31-5p/MyD88/NF-κB axis. Aging (Albany NY) 2021; 13:21345-21363. [PMID: 34511434 PMCID: PMC8457610 DOI: 10.18632/aging.203468] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 07/08/2021] [Indexed: 12/12/2022]
Abstract
Inflammation events have been found to aggravate brain injury and blood-brain barrier (BBB) damage following subarachnoid hemorrhage (SAH). This study probed the role and mechanism of a novel circRNA, circARF3, in regulating the BBB injury in SAH rats and hypoxia-induced vascular endothelial cell (VEC) injury in vitro. Levels of circARF3 and miR-31-5p were monitored by RT-PCR. The expression of inflammatory factors IL-1β and TNF-α was verified by ELISA. In vivo SAH model was constructed in Sprague Dawley (SD) rats. The BBB integrity and cerebral edema, as well as the neurological functions of the rats were evaluated. The apoptotic neurons and microglia in brain lesions were examined by immunohistochemistry (IHC). The MyD88/NF-κB pathway was tested by Western blot. Furthermore, gain-of functional assay were constructed to explore the effects of circARF3 and miR-31-5p in primary cultured brain microvascular endothelial cell (BMEC) injury and microglial inflammation induced by oxygen and glucose deprivation (OGD). circARF3 was significantly down-regulated in plasma and CSF in SAH patients with higher Fisher stages. In the SAH rat model, overexpressing circARF3 improved BBB integrity and neurological score, decreased neuronal apoptosis and microglial activation in ipsilateral basal cortex, with declined miR-31-5p expression and MyD88-NF-κB activation. In vitro, overexpressing circARF3 attenuated OGD-mediated integrity destruction of BMECs and microglial induced neuroinflammation, while overexpressing miR-31-5p had opposite effects. Mechanistically, circARF3 sponged miR-31-5p as an endogenous competitive RNA and dampens its expression, thus inactivating MyD88-NF-κB pathway. CircARF3 attenuates BBB destruction in SAH rats by regulating the miR-31-5p-activated MyD88-NF-κB pathway.
Collapse
Affiliation(s)
- Li Cai
- Department of Neurosurgery, Guangxi International Zhuang Medicine Hospital, Nanning 530221, Guangxi, China
| | - Beihai Ge
- Department of Neurology, Guangxi Zhuang Autonomous Region Brain Hospital, Liuzhou 545005, Guangxi, China
| | - Shengbo Xu
- Department of Neurosurgery, Guangxi International Zhuang Medicine Hospital, Nanning 530221, Guangxi, China
| | - Xiangwen Chen
- Department of Neurosurgery, Guangxi International Zhuang Medicine Hospital, Nanning 530221, Guangxi, China
| | - Hong Yang
- Department of Neurosurgery, Guangxi International Zhuang Medicine Hospital, Nanning 530221, Guangxi, China
| |
Collapse
|
15
|
Yang Q, Yu J, Qin H, Liu L, Di C, Zhuang Q, Yin H. Irbesartan suppresses lipopolysaccharide (LPS)-induced blood-brain barrier (BBB) dysfunction by inhibiting the activation of MLCK/MLC. Int Immunopharmacol 2021; 98:107834. [PMID: 34174702 DOI: 10.1016/j.intimp.2021.107834] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 05/26/2021] [Accepted: 05/27/2021] [Indexed: 12/26/2022]
Abstract
The basic function of the blood-brain barrier (BBB) is to selectively regulate the infiltration of solutes from the circulating blood into the central nervous system (CNS). Impaired BBB activity is related to brain damage caused by stroke, traumatic injury, neurodegenerative diseases, etc. Comprised of a monolayer of endothelial cells, the integrity of the BBB is determined by the expression of tight junction proteins and the contractile activity of the perijunctional apical actomyosin ring. Irbesartan, an AT1R antagonist, has been widely used for the treatment of hypertension. However, the pharmacological function of Irbesartan in the balance of the BBB is still unknown. In the present study, we performed both in-vivo and in-vitro experiments using lipopolysaccharide (LPS) to explore the mechanism behind the protective effects of Irbesartan against the BBB impairment. The results of our mouse model study revealed that Irbesartan could reduce BBB permeability, restore the expression of Occludin, and suppress the expression of inflammatory mediators, including interleukin-6, monocyte chemoattractant protein-1, and intercellular adhesion molecule-1. Additionally, Irbesartan improved LPS-induced depressive-like behavior. In our in vitro experiments, human brain microvascular endothelial cells (HBMVECs) stimulated with LPS demonstrated decreased endothelial permeability and increased occludin expression in response to Irbesartan treatment. Importantly, we found that the protective effects of Irbesartan were mediated through the NF-κB/MLC/MLCK signaling pathway, as blockage of NF-κB abolished the effects of Irbesartan. Our findings provide a basis for further research into the neuroprotective mechanism of Irbesartan.
Collapse
Affiliation(s)
- Qixia Yang
- Department of Pharmacy, Zaozhuang Municipal Hospital, Zaozhuang, Shandong 277100, China
| | - Juanjuan Yu
- Department of Pharmacy, Zaozhuang Municipal Hospital, Zaozhuang, Shandong 277100, China
| | - Hao Qin
- Department of Neurosurgery, Zaozhuang Municipal Hospital, Zaozhuang, Shandong 277100, China
| | - Long Liu
- Department of Neurosurgery, Zaozhuang Municipal Hospital, Zaozhuang, Shandong 277100, China
| | - Chao Di
- Department of Neurosurgery, Zaozhuang Municipal Hospital, Zaozhuang, Shandong 277100, China
| | - Qiang Zhuang
- Department of Neurosurgery, Zaozhuang Municipal Hospital, Zaozhuang, Shandong 277100, China
| | - Hang Yin
- Department of Neurosurgery, Zaozhuang Municipal Hospital, Zaozhuang, Shandong 277100, China.
| |
Collapse
|
16
|
Wang J, Wang Y, Zuo Y, Duan J, Pan A, Li JM, Yan XX, Liu F. MFGE8 mitigates brain injury in a rat model of SAH by maintaining vascular endothelial integrity via TIGβ5/PI3K/CXCL12 signaling. Exp Brain Res 2021; 239:2193-2205. [PMID: 33991211 DOI: 10.1007/s00221-021-06111-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 04/08/2021] [Indexed: 12/25/2022]
Abstract
Leaked blood components, injured endothelial cells, local inflammatory response and vasospasm may converge to promote microthrombosis following subarachnoid hemorrhage (SAH). Previously, we showed that the milk fat globule-epidermal growth factor 8 (MFGE8) can mitigate SAH-induced microthrombosis. This present study was aimed to explore the molecular pathway participated in MFGE8-dependent protection on vascular endothelium. Immunofluorescence, immunoblot and behavioral tests were used to determine the molecular partner and signaling pathway mediating the effect of MFGE8 in vascular endothelium in rats with experimental SAH and controls, together with the applications of RNA silencing and pharmacological intervention methods. Relative to control, recombinant human MFGE8 (rhMFGE8) treatment increased 5-bromo-2'-deoxyuridine (BrdU) labeled new endothelial cells, reduced TUNUL-positive endothelial cells and elevated the expression of phosphatidylinositol 3-kinase (PI3K) and chemokine (C-X-C motif) ligand 12 (CXCL12), in the brains of SAH rats. These effects were reversed by MFGE8 RNA silencing, as well as following cilengitide and wortmannin intervention. These results suggest that MFGE8 promotes endothelial regeneration and mitigates endothelial DNA damage through the activation of the TIGβ5/PI3K/CXCL12 signaling pathway.
Collapse
Affiliation(s)
- Jikai Wang
- Department of Neurosurgery, The Fifth Affiliated Hospital of Sun Yat-sen University, No. 52 Meihuadong Road, Zhuhai, 519000, Guangdong, China
| | - Yiping Wang
- Department of Neurosurgery, The Fifth Affiliated Hospital of Sun Yat-sen University, No. 52 Meihuadong Road, Zhuhai, 519000, Guangdong, China
| | - Yuchun Zuo
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Jiajia Duan
- Department of Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Aihua Pan
- Department of Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Jian-Ming Li
- Department of Anatomy, School of Basic Sciences, Changsha Medical University, Changsha, 410219, Hunan, China
| | - Xiao-Xin Yan
- Department of Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Fei Liu
- Department of Neurosurgery, The Fifth Affiliated Hospital of Sun Yat-sen University, No. 52 Meihuadong Road, Zhuhai, 519000, Guangdong, China.
| |
Collapse
|
17
|
Lu Q, Liu R, Sherchan P, Ren R, He W, Fang Y, Huang Y, Shi H, Tang L, Yang S, Zhang JH, Tang J. TREM (Triggering Receptor Expressed on Myeloid Cells)-1 Inhibition Attenuates Neuroinflammation via PKC (Protein Kinase C) δ/CARD9 (Caspase Recruitment Domain Family Member 9) Signaling Pathway After Intracerebral Hemorrhage in Mice. Stroke 2021; 52:2162-2173. [PMID: 33947214 DOI: 10.1161/strokeaha.120.032736] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Qin Lu
- Department of Neurosurgery, Sir Run Run Shaw Hospital (Q.L., S.Y.), School of Medicine, Zhejiang University, Hangzhou, China.,Department of Physiology and Pharmacology (Q.L., R.L., P.S., R.R., W.H., Y.F., Y.H., H.S., L.T., J.H.Z., J.T.), Loma Linda University, CA
| | - Rui Liu
- Department of Neurosurgery, The Second Affiliated Hospital (R.R., Y.F., Y.H., L.T.), School of Medicine, Zhejiang University, Hangzhou, China.,Department of Neurology, Guizhou Provincial People's Hospital, Guiyang, China (R.L.).,Department of Physiology and Pharmacology (Q.L., R.L., P.S., R.R., W.H., Y.F., Y.H., H.S., L.T., J.H.Z., J.T.), Loma Linda University, CA
| | - Prativa Sherchan
- Department of Physiology and Pharmacology (Q.L., R.L., P.S., R.R., W.H., Y.F., Y.H., H.S., L.T., J.H.Z., J.T.), Loma Linda University, CA
| | - Reng Ren
- Department of Physiology and Pharmacology (Q.L., R.L., P.S., R.R., W.H., Y.F., Y.H., H.S., L.T., J.H.Z., J.T.), Loma Linda University, CA
| | - Wei He
- Department of Physiology and Pharmacology (Q.L., R.L., P.S., R.R., W.H., Y.F., Y.H., H.S., L.T., J.H.Z., J.T.), Loma Linda University, CA.,Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China (W.H.)
| | - Yuanjian Fang
- Department of Neurosurgery, The Second Affiliated Hospital (R.R., Y.F., Y.H., L.T.), School of Medicine, Zhejiang University, Hangzhou, China.,Department of Physiology and Pharmacology (Q.L., R.L., P.S., R.R., W.H., Y.F., Y.H., H.S., L.T., J.H.Z., J.T.), Loma Linda University, CA
| | - Yi Huang
- Department of Neurosurgery, The Second Affiliated Hospital (R.R., Y.F., Y.H., L.T.), School of Medicine, Zhejiang University, Hangzhou, China.,Department of Physiology and Pharmacology (Q.L., R.L., P.S., R.R., W.H., Y.F., Y.H., H.S., L.T., J.H.Z., J.T.), Loma Linda University, CA
| | - Hui Shi
- Department of Physiology and Pharmacology (Q.L., R.L., P.S., R.R., W.H., Y.F., Y.H., H.S., L.T., J.H.Z., J.T.), Loma Linda University, CA.,Department of Neurosurgery, Yongchuan Hospital, Chongqing Medical University, China (H.S.)
| | - Lihui Tang
- Department of Neurosurgery, The Second Affiliated Hospital (R.R., Y.F., Y.H., L.T.), School of Medicine, Zhejiang University, Hangzhou, China.,Department of Physiology and Pharmacology (Q.L., R.L., P.S., R.R., W.H., Y.F., Y.H., H.S., L.T., J.H.Z., J.T.), Loma Linda University, CA
| | - Shuxu Yang
- Department of Neurosurgery, Sir Run Run Shaw Hospital (Q.L., S.Y.), School of Medicine, Zhejiang University, Hangzhou, China
| | - John H Zhang
- Department of Physiology and Pharmacology (Q.L., R.L., P.S., R.R., W.H., Y.F., Y.H., H.S., L.T., J.H.Z., J.T.), Loma Linda University, CA.,Department of Neurosurgery (J.H.Z.), Loma Linda University, CA.,Department of Anesthesiology (J.H.Z.), Loma Linda University, CA
| | - Jiping Tang
- Department of Physiology and Pharmacology (Q.L., R.L., P.S., R.R., W.H., Y.F., Y.H., H.S., L.T., J.H.Z., J.T.), Loma Linda University, CA
| |
Collapse
|
18
|
Amoo M, Henry J, Pender N, Brennan P, Campbell M, Javadpour M. Blood-brain barrier permeability imaging as a predictor for delayed cerebral ischaemia following subarachnoid haemorrhage. A narrative review. Acta Neurochir (Wien) 2021; 163:1457-1467. [PMID: 33404877 DOI: 10.1007/s00701-020-04670-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 12/01/2020] [Indexed: 01/17/2023]
Abstract
BACKGROUND Aneurysmal subarachnoid haemorrhage is associated with significant morbidity and mortality due to the myriad of complications contributing to early brain injury and delayed cerebral ischaemia. There is increasing interest in the exploration of the association between blood-brain barrier integrity and risks of delayed cerebral ischaemia and poor outcomes. Despite recent advances in cerebral imaging, radiographic imaging of blood-brain barrier disruption, as a biomarker for outcome prediction, has not been adopted in clinical practice. METHODS We performed a narrative review by searching for articles describing molecular changes or radiological identification of changes in BBB permeability following subarachnoid haemorrhage (SAH) on MEDLINE. Preclinical studies were analysed if reported structural changes and clinical studies were included if they investigated for radiological markers of BBB disruption and its correlation with delayed cerebral ischaemia. RESULTS There is ample preclinical evidence to suggest that there are structural changes in BBB permeability following SAH. The available clinical literature has demonstrated correlations between permeability imaging and outcomes following aneurysmal subarachnoid haemorrhage (aSAH). CONCLUSION Radiological biomarkers offer a potential non-invasive prognostication tool and may also allow early identifications of patients who may be at risk of DCI.
Collapse
|
19
|
Liu H, He J, Wu Y, Du Y, Jiang Y, Chen C, Yu Z, Zhong J, Wang Z, Cheng C, Sun X, Huang Z. Endothelial Regulation by Exogenous Annexin A1 in Inflammatory Response and BBB Integrity Following Traumatic Brain Injury. Front Neurosci 2021; 15:627110. [PMID: 33679307 PMCID: PMC7930239 DOI: 10.3389/fnins.2021.627110] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 01/15/2021] [Indexed: 12/19/2022] Open
Abstract
Background and Target Following brain trauma, blood–brain barrier (BBB) disruption and inflammatory response are critical pathological steps contributing to secondary injury, leading to high mortality and morbidity. Both pathologies are closely associated with endothelial remodeling. In the present study, we concentrated on annexin A1 (ANXA1) as a novel regulator of endothelial function after traumatic brain injury. Methods After establishing controlled cortical impact (CCI) model in male mice, human recombinant ANXA1 (rANXA1) was administered intravenously, followed by assessments of BBB integrity, brain edema, inflammatory response, and neurological deficits. Result Animals treated with rANXA1 (1 μg/kg) at 1 h after CCI exhibited optimal BBB protection including alleviated BBB disruption and brain edema, as well as endothelial junction proteins loss. The infiltrated neutrophils and inflammatory cytokines were suppressed by rANXA1, consistent with decreased adhesive and transmigrating molecules from isolated microvessels. Moreover, rANXA1 attenuated the neurological deficits induced by CCI. We further found that the Ras homolog gene family member A (RhoA) inhibition has similar effect as rANXA1 in ameliorating brain injuries after CCI, whereas rANXA1 suppressed CCI-induced RhoA activation. Conclusion Our findings suggest that the endothelial remodeling by exogenous rANXA1 corrects BBB disruption and inflammatory response through RhoA inhibition, hence improving functional outcomes in CCI mice.
Collapse
Affiliation(s)
- Han Liu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Department of Neurosurgery, Qilu Hospital of Shandong University (Qingdao Campus), Qingdao, China
| | - Junchi He
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yue Wu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yang Du
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Yinghua Jiang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Chengzhi Chen
- Department of Occupational and Environmental Health, School of Public Health and Management, Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University, Chongqing, China
| | - Zhanyang Yu
- Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Jianjun Zhong
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhigang Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University (Qingdao Campus), Qingdao, China
| | - Chongjie Cheng
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaochuan Sun
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhijian Huang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
20
|
Neulen A, Molitor M, Kosterhon M, Pantel T, Holzbach E, Rudi WS, Karbach SH, Wenzel P, Ringel F, Thal SC. Correlation of cardiac function and cerebral perfusion in a murine model of subarachnoid hemorrhage. Sci Rep 2021; 11:3317. [PMID: 33558609 PMCID: PMC7870815 DOI: 10.1038/s41598-021-82583-9] [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: 05/13/2020] [Accepted: 01/18/2021] [Indexed: 12/28/2022] Open
Abstract
Cerebral hypoperfusion is a key factor for determining the outcome after subarachnoid hemorrhage (SAH). A subset of SAH patients develop neurogenic stress cardiomyopathy (NSC), but it is unclear to what extent cerebral hypoperfusion is influenced by cardiac dysfunction after SAH. The aims of this study were to examine the association between cardiac function and cerebral perfusion in a murine model of SAH and to identify electrocardiographic and echocardiographic signs indicative of NSC. We quantified cortical perfusion by laser SPECKLE contrast imaging, and myocardial function by serial high-frequency ultrasound imaging, for up to 7 days after experimental SAH induction in mice by endovascular filament perforation. Cortical perfusion decreased significantly whereas cardiac output and left ventricular ejection fraction increased significantly shortly post-SAH. Transient pathological ECG and echocardiographic abnormalities, indicating NSC (right bundle branch block, reduced left ventricular contractility), were observed up to 3 h post-SAH in a subset of model animals. Cerebral perfusion improved over time after SAH and correlated significantly with left ventricular end-diastolic volume at 3, 24, and 72 h. The murine SAH model is appropriate to experimentally investigate NSC. We conclude that in addition to cerebrovascular dysfunction, cardiac dysfunction may significantly influence cerebral perfusion, with LVEDV presenting a potential parameter for risk stratification.
Collapse
Affiliation(s)
- Axel Neulen
- Department of Neurosurgery, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany.
| | - Michael Molitor
- Center for Cardiology-Cardiology I, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.,Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.,German Center for Cardiovascular Research (DZHK)-Partner Site Rhine-Main, Mainz, Germany
| | - Michael Kosterhon
- Department of Neurosurgery, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Tobias Pantel
- Department of Neurosurgery, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Elisa Holzbach
- Department of Neurosurgery, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Wolf-Stephan Rudi
- Center for Cardiology-Cardiology I, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.,Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.,German Center for Cardiovascular Research (DZHK)-Partner Site Rhine-Main, Mainz, Germany
| | - Susanne H Karbach
- Center for Cardiology-Cardiology I, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.,Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.,German Center for Cardiovascular Research (DZHK)-Partner Site Rhine-Main, Mainz, Germany
| | - Philip Wenzel
- Center for Cardiology-Cardiology I, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.,Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.,German Center for Cardiovascular Research (DZHK)-Partner Site Rhine-Main, Mainz, Germany
| | - Florian Ringel
- Department of Neurosurgery, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Serge C Thal
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany. .,Center for Molecular Surgical Research (MFO), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.
| |
Collapse
|
21
|
AT1R/GSK-3 β/mTOR Signaling Pathway Involved in Angiotensin II-Induced Neuronal Apoptosis after HIE Both In Vitro and In Vivo. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8864323. [PMID: 33425219 PMCID: PMC7773460 DOI: 10.1155/2020/8864323] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/17/2020] [Accepted: 12/02/2020] [Indexed: 12/11/2022]
Abstract
Objective The focus of the present study is to evaluate the effects of Angiotensin II (Ang II) on neuronal apoptosis after HIE and the potential underlying mechanisms. Methods Primary neonatal rat cortical neurons were used to study the oxygen-glucose deprivation (OGD) cell model. The expressions of Ang II, AT1R, GSK-3β, p-GSK-3β, mTOR, p-mTOR, Bax, Bcl-2, and cleaved caspase-3 were detected via western blot. IF and flow cytometry were used to evaluate neuronal apoptosis. Hypoxic-ischemic encephalopathy (HIE) was established to evaluate the therapeutic effects of Ang II in vivo. Cerebral infarction areas were detected by 2,3,5-Triphenyltetrazolium chloride staining. The righting and geotaxis reflexes were also recorded. In addition, Fluoro-Jade C staining and TUNEL staining were performed to evaluate neuronal degeneration and apoptosis. Results Ang II significantly increased the rate of neuronal apoptosis, upregulated the expression of cleaved caspase-3, and downregulated Bcl-2/Bax ratio after OGD insult. For vivo assay, the expressions of endogenous Ang II and AT1R gradually increased and peaked at 24 h after HIE. Ang II increased NeuN-positive AT1R cell expression. In addition, Ang II increased the area of cerebral infarction, promoted neuronal degeneration and apoptosis, aggravated neurological deficits on righting and geotaxis reflexes, and was accompanied by increased expressions of phosphorylated GSK-3β and mTOR. The application of valsartan (Ang II inhibitor) or SB216763 (GSK-3β inhibitor) reversed these phenomena triggered by Ang II following HIE. Conclusion Ang II increased neuronal apoptosis through the AT1R/GSK-3β/mTOR signaling pathway after experimental HIE both in vitro and in vivo, and Ang II may serve as a novel therapeutic target to ameliorate brain injury after HIE.
Collapse
|
22
|
Yang Y, Torbey MT. Angiogenesis and Blood-Brain Barrier Permeability in Vascular Remodeling after Stroke. Curr Neuropharmacol 2020; 18:1250-1265. [PMID: 32691713 PMCID: PMC7770645 DOI: 10.2174/1570159x18666200720173316] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/27/2020] [Accepted: 07/11/2020] [Indexed: 11/22/2022] Open
Abstract
Angiogenesis, the growth of new blood vessels, is a natural defense mechanism helping to restore oxygen and nutrient supply to the affected brain tissue following an ischemic stroke. By stimulating vessel growth, angiogenesis may stabilize brain perfusion, thereby promoting neuronal survival, brain plasticity, and neurologic recovery. However, therapeutic angiogenesis after stroke faces challenges: new angiogenesis-induced vessels have a higher than normal permeability, and treatment to promote angiogenesis may exacerbate outcomes in stroke patients. The development of therapies requires elucidation of the precise cellular and molecular basis of the disease. Microenvironment homeostasis of the central nervous system is essential for its normal function and is maintained by the blood-brain barrier (BBB). Tight junction proteins (TJP) form the tight junction (TJ) between vascular endothelial cells (ECs) and play a key role in regulating the BBB permeability. We demonstrated that after stroke, new angiogenesis-induced vessels in peri-infarct areas have abnormally high BBB permeability due to a lack of major TJPs in ECs. Therefore, promoting TJ formation and BBB integrity in the new vessels coupled with speedy angiogenesis will provide a promising and safer treatment strategy for improving recovery from stroke. Pericyte is a central neurovascular unite component in vascular barriergenesis and are vital to BBB integrity. We found that pericytes also play a key role in stroke-induced angiogenesis and TJ formation in the newly formed vessels. Based on these findings, in this article, we focus on regulation aspects of the BBB functions and describe cellular and molecular special features of TJ formation with an emphasis on role of pericytes in BBB integrity during angiogenesis after stroke.
Collapse
Affiliation(s)
- Yi Yang
- Department of Neurology, University of New Mexico Health Sciences Center; Albuquerque, New Mexico, 87131, United States
| | - Michel T Torbey
- Department of Neurology, University of New Mexico Health Sciences Center; Albuquerque, New Mexico, 87131, United States
| |
Collapse
|
23
|
Valproate Sodium Protects Blood Brain Barrier Integrity in Intracerebral Hemorrhage Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8884320. [PMID: 33224434 PMCID: PMC7676278 DOI: 10.1155/2020/8884320] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 09/28/2020] [Accepted: 10/15/2020] [Indexed: 01/04/2023]
Abstract
Valproate sodium (VPA) is a traditional antiepileptic drug with a neuroprotective role in cerebrovascular disease. After intracerebral hemorrhage (ICH), mechanical compression by hematoma, neuroinflammation, oxidative stress, and cytotoxicity of hematoma lysates caused the destruction of the blood brain barrier (BBB). Targeting BBB is a major therapeutic method for patients with ICH. The purpose of the present study was to explore the role of VPA in preserving BBB integrity in the ICH model and investigate the underlying molecular mechanisms. One hundred and thirty-six adult male CD1 mice were randomly divided into five groups in the study. Mice subjected to ICH were administered intraperitoneally with VPA at 3, 24, and 48 h post-ICH, respectively. Neurobehavioral assessments, BBB permeability, Evans blue fluorescence, hematoma volume, and protein expression were evaluated. The administration of VPA reduced BBB permeability and improved the neurobehavior significantly post-ICH. VPA administration significantly decreased the expression of phosphorylated nuclear factor-kappa B (p-NFκB), matrix metalloproteinases 9 (MMP9), tumor necrosis factorα (TNFα), and interleukin-6 (IL-6), while it enhanced the expression of claudin 5 and occludin in the brain. In conclusion, VPA administration maintained the integrity of BBB after experimental ICH, thus reducing brain edema and improving the neurological outcomes. Therefore, VPA administration might be a new therapeutic method to protect BBB integrity for patients with ICH.
Collapse
|
24
|
Fang Y, Shi H, Ren R, Huang L, Okada T, Lenahan C, Gamdzyk M, Travis ZD, Lu Q, Tang L, Huang Y, Zhou K, Tang J, Zhang J, Zhang JH. Pituitary Adenylate Cyclase-Activating Polypeptide Attenuates Brain Edema by Protecting Blood-Brain Barrier and Glymphatic System After Subarachnoid Hemorrhage in Rats. Neurotherapeutics 2020; 17:1954-1972. [PMID: 32918234 PMCID: PMC7851266 DOI: 10.1007/s13311-020-00925-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2020] [Indexed: 12/14/2022] Open
Abstract
Brain edema is a vital contributor to early brain injury after subarachnoid hemorrhage (SAH), which is responsible for prolonged hospitalization and poor outcomes. Pharmacological therapeutic targets on edema formation have been the focus of research for decades. Pituitary adenylate cyclase-activating polypeptide (PACAP) has been shown to participate in neural development and brain injury. Here, we used PACAP knockout CRISPR to demonstrate that endogenous PACAP plays an endogenous neuroprotective role against brain edema formation after SAH in rats. The exogenous PACAP treatment provided both short- and long-term neurological benefits by preserving the function of the blood-brain barrier and glymphatic system after SAH. Pretreatment of inhibitors of PACAP receptors showed that the PACAP-involved anti-edema effect and neuroprotection after SAH was facilitated by the selective PACAP receptor (PAC1). Further administration of adenylyl cyclase (AC) inhibitor and sulfonylurea receptor 1 (SUR1) CRISPR activator suggested that the AC-cyclic adenosine monophosphate (cAMP)-protein kinase A (PKA) axis participated in PACAP signaling after SAH, which inhibited the expression of edema-related proteins, SUR1 and aquaporin-4 (AQP4), through SUR1 phosphorylation. Thus, PACAP may serve as a potential clinical treatment to alleviate brain edema in patients with SAH.
Collapse
Affiliation(s)
- Yuanjian Fang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Zhejiang, 310009, Hangzhou, China
| | - Hui Shi
- Department of Neurosurgery, Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Reng Ren
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Zhejiang, 310009, Hangzhou, China
| | - Lei Huang
- Department of Neurosurgery, Loma Linda University, Loma Linda, California, USA
- Department of Physiology and Pharmacology, Loma Linda University, 11041 Campus St, Risley Hall, Room 219, Loma Linda, California, 92354, USA
| | - Takeshi Okada
- Department of Neurosurgery, Loma Linda University, Loma Linda, California, USA
- Department of Physiology and Pharmacology, Loma Linda University, 11041 Campus St, Risley Hall, Room 219, Loma Linda, California, 92354, USA
| | - Cameron Lenahan
- Department of Neurosurgery, Loma Linda University, Loma Linda, California, USA
- Burrell College of Osteopathic Medicine, Las Cruces, New Mexico, USA
| | - Marcin Gamdzyk
- Department of Neurosurgery, Loma Linda University, Loma Linda, California, USA
| | - Zachary D Travis
- Department of Neurosurgery, Loma Linda University, Loma Linda, California, USA
| | - Qin Lu
- Department of Neurosurgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Lihui Tang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Zhejiang, 310009, Hangzhou, China
| | - Yi Huang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Zhejiang, 310009, Hangzhou, China
| | - Keren Zhou
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Zhejiang, 310009, Hangzhou, China
| | - Jiping Tang
- Department of Neurosurgery, Loma Linda University, Loma Linda, California, USA
- Department of Physiology and Pharmacology, Loma Linda University, 11041 Campus St, Risley Hall, Room 219, Loma Linda, California, 92354, USA
- Department of Anesthesiology, Loma Linda University, Loma Linda, California, USA
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Zhejiang, 310009, Hangzhou, China.
| | - John H Zhang
- Department of Neurosurgery, Loma Linda University, Loma Linda, California, USA.
- Department of Physiology and Pharmacology, Loma Linda University, 11041 Campus St, Risley Hall, Room 219, Loma Linda, California, 92354, USA.
- Department of Anesthesiology, Loma Linda University, Loma Linda, California, USA.
| |
Collapse
|
25
|
Suzuki H, Kanamaru H, Kawakita F, Asada R, Fujimoto M, Shiba M. Cerebrovascular pathophysiology of delayed cerebral ischemia after aneurysmal subarachnoid hemorrhage. Histol Histopathol 2020; 36:143-158. [PMID: 32996580 DOI: 10.14670/hh-18-253] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Aneurysmal subarachnoid hemorrhage (SAH) remains a serious cerebrovascular disease. Even if SAH patients survive the initial insults, delayed cerebral ischemia (DCI) may occur at 4 days or later post-SAH. DCI is characteristics of SAH, and is considered to develop by blood breakdown products and inflammatory reactions, or secondary to early brain injury, acute pathophysiological events that occur in the brain within the first 72 hours of aneurysmal SAH. The pathology underlying DCI may involve large artery vasospasm and/or microcirculatory disturbances by microvasospasm, microthrombosis, dysfunction of venous outflow and compression of microvasculature by vasogenic or cytotoxic tissue edema. Recent clinical evidence has shown that large artery vasospasm is not the only cause of DCI, and that both large artery vasospasm-dependent and -independent cerebral infarction causes poor outcome. Animal studies suggest that mechanisms of vasospasm may differ between large artery and arterioles or capillaries, and that many kinds of cells in the vascular wall and brain parenchyma may be involved in the pathogenesis of microcirculatory disturbances. The impairment of the paravascular and glymphatic systems also may play important roles in the development of DCI. As pathological mediators for DCI, glutamate and several matricellular proteins have been investigated in addition to inflammatory molecules. Glutamate is involved in excitotoxicity contributing to cortical spreading ischemia and epileptic activity-related events. Microvascular dysfunction is an attractive mechanism to explain the cause of poor outcomes independently of large cerebral artery vasospasm, but needs more studies to clarify the pathophysiologies or mechanisms and to develop a novel therapeutic strategy.
Collapse
Affiliation(s)
- Hidenori Suzuki
- Department of Neurosurgery, Mie University Graduate School of Medicine, Tsu, Japan.
| | - Hideki Kanamaru
- Department of Neurosurgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Fumihiro Kawakita
- Department of Neurosurgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Reona Asada
- Department of Neurosurgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Masashi Fujimoto
- Department of Neurosurgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Masato Shiba
- Department of Neurosurgery, Mie University Graduate School of Medicine, Tsu, Japan
| |
Collapse
|
26
|
Qu C, Song H, Shen J, Xu L, Li Y, Qu C, Li T, Zhang J. Mfsd2a Reverses Spatial Learning and Memory Impairment Caused by Chronic Cerebral Hypoperfusion via Protection of the Blood-Brain Barrier. Front Neurosci 2020; 14:461. [PMID: 32612494 PMCID: PMC7308492 DOI: 10.3389/fnins.2020.00461] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 04/15/2020] [Indexed: 12/12/2022] Open
Abstract
Disruption of the blood–brain barrier (BBB) can lead to cognitive impairment. Major facilitator superfamily domain-containing protein 2a (Mfsd2a) is a newly discovered protein that is essential for maintaining BBB integrity. However, the role of Mfsd2a in vascular cognitive impairment has not been explored yet. In this study, a rat model of chronic cerebral hypoperfusion (CCH) was established by producing permanent bilateral common carotid artery occlusion (2VO) in rats. We found that after the 2VO procedure, the rats exhibited cognitive impairment, showed increased BBB leakage within the hippocampus, and had reduced expression of the Mfsd2a protein. The overexpression of Mfsd2a in the rat hippocampus reversed these changes. Further investigations using transmission electron microscopy revealed a significantly increased rate of vesicular transcytosis in the BBB of the hippocampus of the CCH rats; the rate reduced after overexpression of Mfsd2a. Moreover, Mfsd2a overexpression did not cause changes in the expression of tight junction-associated proteins and in the ultrastructures of the tight junctions. In conclusion, Mfsd2a attenuated BBB damage and ameliorated cognitive impairment in CCH rats, and its protective effect on the BBB was achieved via inhibition of vesicular transcytosis.
Collapse
Affiliation(s)
- Changhua Qu
- Department of Neurology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Hao Song
- Department of Neurology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Jun Shen
- Department of Neurology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Linling Xu
- Department of Neurology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Yaqing Li
- Department of Neurology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Chujie Qu
- Department of Neurology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Tian Li
- Department of Neurology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Junjian Zhang
- Department of Neurology, Zhongnan Hospital, Wuhan University, Wuhan, China
| |
Collapse
|
27
|
Zhang H, Pan Q, Xie Z, Chen Y, Wang J, Bihl J, Zhong W, Chen Y, Zhao B, Ma X. Implication of MicroRNA503 in Brain Endothelial Cell Function and Ischemic Stroke. Transl Stroke Res 2020; 11:1148-1164. [PMID: 32285355 DOI: 10.1007/s12975-020-00794-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 02/18/2020] [Accepted: 02/20/2020] [Indexed: 12/12/2022]
Abstract
The role of miR-503 in brain endothelium and ischemic stroke (IS) remains unclear. We aimed to study the relationship between plasma miR-503 and the onset time, severity, subtypes, and von Willebrand Factor (vWF) level in IS patients and to investigate the roles and underlying mechanisms of miR-503 in middle cerebral artery occlusion (MCAO) mice and cultured cerebral vascular endothelial cells (ECs). In MCAO mice, the effects of plasma from acute severe IS patients (ASS) with or without miR-503 antagomir on brain and ECs damage were determined. In cultured human ECs, the effects of miR-503 overexpression or knockdown on the monolayer permeability, apoptosis, ROS, and NO generation were investigated. For mechanism study, the PI3K/Akt/eNOS pathway, cleaved caspase-3, and bcl-2 were analyzed. Results showed that plasma miR-503 was significantly increased in IS patients, especially in acute period and severe cases and subtypes of LAA and TACI, and was positively correlated with vWF. Logistic analysis indicated that miR-503 was an independent risk factor for IS, with the area under curve of 0.796 in ROC analysis. In MCAO mice, ASS pretreatment aggravated neurological injury, BBB damage, brain edema, CBF reduction, and decreased NO production while increased apoptosis and ROS generation in brain ECs, which were partly abolished by miR-503 antagomir. In cultured ECs, miR-503 overexpression and knockdown confirmed its effects on regulating monolayer permeability, cell apoptosis, NO, and ROS generation via PI3K/Akt/eNOS pathway or bcl-2 and cleaved caspase-3 proteins. These together indicate that miR-503 is a promising biomarker and novel therapeutic target for IS.
Collapse
Affiliation(s)
- Huiting Zhang
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, 57 South Renmin Road, Zhanjiang, 524001, China
| | - Qunwen Pan
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, 57 South Renmin Road, Zhanjiang, 524001, China
| | - Zi Xie
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, 57 South Renmin Road, Zhanjiang, 524001, China
| | - Yanyu Chen
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, 57 South Renmin Road, Zhanjiang, 524001, China
| | - Jinju Wang
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, OH, 45430, USA
| | - Ji Bihl
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, OH, 45430, USA
| | - Wangtao Zhong
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, 57 South Renmin Road, Zhanjiang, 524001, China
| | - Yanfang Chen
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, OH, 45430, USA
| | - Bin Zhao
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, 57 South Renmin Road, Zhanjiang, 524001, China.
| | - Xiaotang Ma
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, 57 South Renmin Road, Zhanjiang, 524001, China.
| |
Collapse
|
28
|
Nonionotropic Action of Endothelial NMDA Receptors on Blood-Brain Barrier Permeability via Rho/ROCK-Mediated Phosphorylation of Myosin. J Neurosci 2020; 40:1778-1787. [PMID: 31953371 DOI: 10.1523/jneurosci.0969-19.2019] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 11/14/2019] [Accepted: 12/20/2019] [Indexed: 12/19/2022] Open
Abstract
Increase in blood-brain barrier (BBB) permeability is a crucial step in neuroinflammatory processes. We previously showed that N Methyl D Aspartate Receptor (NMDARs), expressed on cerebral endothelial cells forming the BBB, regulate immune cell infiltration across this barrier in the mouse. Here, we describe the mechanism responsible for the action of NMDARs on BBB permeabilization. We report that mouse CNS endothelial NMDARs display the regulatory GluN3A subunit. This composition confers to NMDARs' unconventional properties: these receptors do not induce Ca2+ influx but rather show nonionotropic properties. In inflammatory conditions, costimulation of human brain endothelial cells by NMDA agonists (NMDA or glycine) and the serine protease tissue plasminogen activator, previously shown to potentiate NMDAR activity, induces metabotropic signaling via the Rho/ROCK pathway. This pathway leads to an increase in permeability via phosphorylation of myosin light chain and subsequent shrinkage of human brain endothelial cells. Together, these data draw a link between NMDARs and the cytoskeleton in brain endothelial cells that regulates BBB permeability in inflammatory conditions.SIGNIFICANCE STATEMENT The authors describe how NMDARs expressed on endothelial cells regulate blood-brain barrier function via myosin light chain phosphorylation and increase in permeability. They report that these non-neuronal NMDARs display distinct structural, functional, and pharmacological features than their neuronal counterparts.
Collapse
|
29
|
Zhao C, Ma J, Wang Z, Li H, Shen H, Li X, Chen G. Mfsd2a Attenuates Blood-Brain Barrier Disruption After Sub-arachnoid Hemorrhage by Inhibiting Caveolae-Mediated Transcellular Transport in Rats. Transl Stroke Res 2020; 11:1012-1027. [PMID: 31907728 DOI: 10.1007/s12975-019-00775-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 12/09/2019] [Accepted: 12/20/2019] [Indexed: 02/06/2023]
Abstract
Blood-brain barrier (BBB) disruption is one of the critical mechanisms of brain injury induced by subarachnoid hemorrhage (SAH). Past studies have often focused on the tight junctions of endothelial cells. However, low transcellular transport levels also play an important role in the normal functioning of the BBB. Major facilitator superfamily domain-containing 2a (Mfsd2a) has been demonstrated to be essential for the maintenance of the normal BBB. Our present study aimed to explore the roles and mechanisms of Mfsd2a in BBB disruption after SAH. In this study, a prechiasmatic cistern single-injection model was used to produce experimental SAH in Sprague-Dawley rats. Specific small-interfering RNA and plasmids were used to downregulate and upregulate the expression of Mfsd2a prior to assessments in our SAH model. Omega-3 fatty acid deficiency diet was used to reduce DHA in rat brain. The expression level of Mfsd2a decreased significantly after SAH and reached its lowest level at 72 h post-SAH, which then gradually recovered. At 72 h after SAH, BBB function was disrupted; upregulation of Mfsd2a reversed this damage, whereas downregulation of Mfsd2a exacerbated this damage. These effects were primarily mediated through transcellular transport, especially for changes in caveolae compared to those of tight junctions. After stopping the supply of omega-3 fatty acids, the effect of Mfsd2a on inhibition of caveolae and protection of the blood-brain barrier was eliminated. Taken together, Mfsd2a inhibits caveolae-based transcellular transport by transporting omega-3 fatty acids to protect the BBB after SAH.
Collapse
Affiliation(s)
- Chongshun Zhao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street,, Suzhou, 215006, Jiangsu Province, China
| | - Junwei Ma
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, Shandong Province, China
| | - Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street,, Suzhou, 215006, Jiangsu Province, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street,, Suzhou, 215006, Jiangsu Province, China
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street,, Suzhou, 215006, Jiangsu Province, China
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street,, Suzhou, 215006, Jiangsu Province, China.
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street,, Suzhou, 215006, Jiangsu Province, China.
| |
Collapse
|
30
|
Griemert EV, Hedrich J, Hirnet T, Thal SC. Deficiency of Plasminogen Activator Inhibitor Type 2 Limits Brain Edema Formation after Traumatic Brain Injury. J Neurotrauma 2019; 36:2272-2278. [DOI: 10.1089/neu.2018.6126] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Eva-Verena Griemert
- Department of Anesthesiology, University Medical Center of Johannes Gutenberg-University, Mainz, Germany
| | - Jana Hedrich
- Institute of Physiology, University Medical Center of Johannes Gutenberg-University, Mainz, Germany
- Max Planck Institute for Polymer Research, Mainz, Germany
| | - Tobias Hirnet
- Department of Anesthesiology, University Medical Center of Johannes Gutenberg-University, Mainz, Germany
| | - Serge C. Thal
- Department of Anesthesiology, University Medical Center of Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
31
|
Neutrophils mediate early cerebral cortical hypoperfusion in a murine model of subarachnoid haemorrhage. Sci Rep 2019; 9:8460. [PMID: 31186479 PMCID: PMC6560094 DOI: 10.1038/s41598-019-44906-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 05/28/2019] [Indexed: 02/07/2023] Open
Abstract
Cerebral hypoperfusion in the first hours after subarachnoid haemorrhage (SAH) is a major determinant of poor neurological outcome. However, the underlying pathophysiology is only partly understood. Here we induced neutropenia in C57BL/6N mice by anti-Ly6G antibody injection, induced SAH by endovascular filament perforation, and analysed cerebral cortical perfusion with laser SPECKLE contrast imaging to investigate the role of neutrophils in mediating cerebral hypoperfusion during the first 24 h post-SAH. SAH induction significantly increased the intracranial pressure (ICP), and significantly reduced the cerebral perfusion pressure (CPP). At 3 h after SAH, ICP had returned to baseline and CPP was similar between SAH and sham mice. However, in SAH mice with normal neutrophil counts cortical hypoperfusion persisted. Conversely, despite similar CPP, cortical perfusion was significantly higher at 3 h after SAH in mice with neutropenia. The levels of 8-iso-prostaglandin-F2α in the subarachnoid haematoma increased significantly at 3 h after SAH in animals with normal neutrophil counts indicating oxidative stress, which was not the case in neutropenic SAH animals. These results suggest that neutrophils are important mediators of cortical hypoperfusion and oxidative stress early after SAH. Targeting neutrophil function and neutrophil-induced oxidative stress could be a promising new approach to mitigate cerebral hypoperfusion early after SAH.
Collapse
|
32
|
Braun DJ, Bachstetter AD, Sudduth TL, Wilcock DM, Watterson DM, Van Eldik LJ. Genetic knockout of myosin light chain kinase (MLCK210) prevents cerebral microhemorrhages and attenuates neuroinflammation in a mouse model of vascular cognitive impairment and dementia. GeroScience 2019; 41:671-679. [PMID: 31104189 PMCID: PMC6885026 DOI: 10.1007/s11357-019-00072-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 04/25/2019] [Indexed: 12/15/2022] Open
Abstract
The blood-brain barrier (BBB) is critical in maintenance of brain homeostasis, and loss of its functional integrity is a key feature across a broad range of neurological insults. This includes both acute injuries such as traumatic brain injury and stroke, as well as more chronic pathologies associated with aging, such as vascular cognitive impairment and dementia (VCID). A specific form of myosin light chain kinase (MLCK210) is a major regulator of barrier integrity in general, including the BBB. Studies have demonstrated the potential of MLCK210 as a therapeutic target for peripheral disorders involving tissue barrier dysfunction, but less is known about its potential as a target for chronic neurologic disorders. We report here that genetic knockout (KO) of MLCK210 protects against cerebral microhemorrhages and neuroinflammation induced by chronic dietary hyperhomocysteinemia. Overall, the results are consistent with an accumulating body of evidence supporting MLCK210 as a potential therapeutic target for tissue barrier dysfunction and specifically implicate it in BBB dysfunction and neuroinflammation in a model of VCID.
Collapse
Affiliation(s)
- David J Braun
- Sanders-Brown Center on Aging, University of Kentucky, 101 Sanders-Brown Bldg., 800 S. Limestone Street, Lexington, KY, 40536, USA
| | - Adam D Bachstetter
- Sanders-Brown Center on Aging, University of Kentucky, 101 Sanders-Brown Bldg., 800 S. Limestone Street, Lexington, KY, 40536, USA
- Department of Neuroscience, University of Kentucky, Lexington, KY, 40536, USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, 40536, USA
| | - Tiffany L Sudduth
- Sanders-Brown Center on Aging, University of Kentucky, 101 Sanders-Brown Bldg., 800 S. Limestone Street, Lexington, KY, 40536, USA
- Department of Physiology, University of Kentucky, Lexington, KY, 40536, USA
| | - Donna M Wilcock
- Sanders-Brown Center on Aging, University of Kentucky, 101 Sanders-Brown Bldg., 800 S. Limestone Street, Lexington, KY, 40536, USA
- Department of Physiology, University of Kentucky, Lexington, KY, 40536, USA
| | - D Martin Watterson
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Linda J Van Eldik
- Sanders-Brown Center on Aging, University of Kentucky, 101 Sanders-Brown Bldg., 800 S. Limestone Street, Lexington, KY, 40536, USA.
- Department of Neuroscience, University of Kentucky, Lexington, KY, 40536, USA.
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, 40536, USA.
| |
Collapse
|