1
|
Xia X, Ren Q, Zhang J, Guan S, Jiang Q, Wei Y, Hua R, Zhao S, Hu X, Shi F, Meng X. Altered brain glymphatic function on diffusion-tensor MRI in patients with spontaneous intracerebral hemorrhage: an exploratory study. Front Aging Neurosci 2024; 16:1506980. [PMID: 39737332 PMCID: PMC11683096 DOI: 10.3389/fnagi.2024.1506980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 12/02/2024] [Indexed: 01/01/2025] Open
Abstract
Objectives To investigate the function of the glymphatic system (GS) and its association with neuropsychological tests in spontaneous intracerebral hemorrhage (sICH) by diffusion tensor imaging analysis along the perivascular space (DTI-ALPS). Methods This retrospective study included 58 patients with sICH and 63 age- and sex-matched healthy controls (HCs). Partial correlation analyses were performed to examine the relationships between the DTI-ALPS index and radiological as well as clinical data. Mediation analyses were performed to explore the mediating role of the grey matter proportion (GM%) in the relationship between DTI-ALPS index and Montreal Cognitive Assessment (MoCA) score. Results Significantly lower DTI-ALPS index values were observed in sICH compared with HCs (FDR-p < 0.001). In the acute-subacute sICH group, the ALPS index was significantly correlated with hematoma volume (r = -0.572, FDR-p = 0.031). In the chronic sICH group, the ALPS index was significantly correlated with MoCA scores (r = 0.425, FDR-p = 0.014). In chronic sICH groups, GM% served as a significant mediator in the relationship between the DTI-ALPS index and MoCA scores (indirect effects β = 4.925, 95%CI: 0.028, 11.841). The ALPS index was identified as an independent prognostic indicator for unfavorable outcomes in sICH (β = -9.851, p = 0.018). Conclusion Our study demonstrated that the DTI-ALPS index decreased in sICH patients, suggesting potential functional impairment of the lymphoid system. Additionally, the DTI-ALPS index served as an independent predictor of poor 90-day prognosis. In the acute-subacute stage of sICH, the DTI-ALPS index had negative correlation with hematoma volume. In the chronic sICH group, the GM% partially mediated the relationship between the GS and cognitive function.
Collapse
Affiliation(s)
- Xiaona Xia
- Department of Radiology, Qilu Hospital (Qingdao) of Shandong University, Qingdao, China
| | - Qingguo Ren
- Department of Radiology, Qilu Hospital (Qingdao) of Shandong University, Qingdao, China
| | - Juntao Zhang
- GE Healthcare PDX GMS Medical Affairs, Shanghai, China
| | - Shuai Guan
- Department of Radiology, Qilu Hospital (Qingdao) of Shandong University, Qingdao, China
| | - Qingjun Jiang
- Department of Radiology, Qilu Hospital (Qingdao) of Shandong University, Qingdao, China
| | - Ying Wei
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China
| | - Rui Hua
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China
| | - Shen Zhao
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China
| | - Xiangjun Hu
- Department of Rehabilitation Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Baoshan District Wusong Central Hospital, Shanghai, China
| | - Feng Shi
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China
| | - Xiangshui Meng
- Department of Radiology, Qilu Hospital (Qingdao) of Shandong University, Qingdao, China
| |
Collapse
|
2
|
Liao J, Duan Y, Liu Y, Chen H, An Z, Chen Y, Su Z, Usman AM, Xiao G. Simvastatin alleviates glymphatic system damage via the VEGF-C/VEGFR3/PI3K-Akt pathway after experimental intracerebral hemorrhage. Brain Res Bull 2024; 216:111045. [PMID: 39097032 DOI: 10.1016/j.brainresbull.2024.111045] [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: 06/21/2024] [Revised: 07/28/2024] [Accepted: 07/31/2024] [Indexed: 08/05/2024]
Abstract
Current clinical practice primarily relies on surgical intervention to remove hematomas in patients with intracerebral hemorrhage (ICH), given the lack of effective drug therapies. Previous research indicates that simvastatin (SIM) may enhance hematoma absorption and resolution in the acute phase of ICH, though the precise mechanisms remain unclear. Recent findings have highlighted the glymphatic system (GS) as a crucial component in intracranial cerebrospinal fluid circulation, playing a significant role in hematoma clearance post-ICH. This study investigates the link between SIM efficacy in hematoma resolution and the GS. Our experimental results show that SIM alleviates GS damage in ICH-induced rats, resulting in improved outcomes such as reduced brain edema, neuronal apoptosis, and degeneration. Further analysis reveals that SIM's effects are mediated through the VEGF-C/VEGFR3/PI3K-Akt pathway. This study advances our understanding of SIM's mechanism in promoting intracranial hematoma clearance and underscores the potential of targeting the GS for ICH treatment.
Collapse
Affiliation(s)
- Junbo Liao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China; Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yingxing Duan
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
| | - Yaxue Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China; Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Haolong Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China; Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhihan An
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China; Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yibing Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China; Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhangjie Su
- Department of Neurosurgery, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge CB2 0QQ, UK
| | - Ahsan Muhammad Usman
- Department of Neurosurgery, Allied Hospital Faisalabad, Sargodha Road, Faisalabad 38000, Pakistan
| | - Gelei Xiao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China; Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
3
|
Rümenapp JE, Sendel M, Kersebaum D, Larsen N, Jansen O, Baron R. Impaired glymphatic flow as a potential driver of pain chronification. Pain 2023; 164:2191-2195. [PMID: 37433183 DOI: 10.1097/j.pain.0000000000002979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/27/2023] [Indexed: 07/13/2023]
Affiliation(s)
- Johanna Elisabeth Rümenapp
- Division of Neurological Pain Research and Therapy, Clinic for Neurology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Manon Sendel
- Division of Neurological Pain Research and Therapy, Clinic for Neurology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Dilara Kersebaum
- Division of Neurological Pain Research and Therapy, Clinic for Neurology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Naomi Larsen
- Clinic for Radiology and Neuroradiology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Olav Jansen
- Clinic for Radiology and Neuroradiology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Ralf Baron
- Division of Neurological Pain Research and Therapy, Clinic for Neurology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| |
Collapse
|
4
|
Li X, Tan X, Zhou Q, Xie Z, Meng W, Pang Y, Huang L, Ding Z, Hu Y, Li R, Huang G, Li H. Limb Remote Ischemic Postconditioning Improves Glymphatic Dysfunction After Cerebral Ischemia-Reperfusion Injury. Neuroscience 2023; 521:20-30. [PMID: 37121383 DOI: 10.1016/j.neuroscience.2023.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 03/29/2023] [Accepted: 04/22/2023] [Indexed: 05/02/2023]
Abstract
BACKGROUND Delayed neuronal damage can be caused or aggravated after cerebral ischemia-reperfusion (I/R) injury. Recent studies have shown that glymphatic system dysfunction after cerebral ischemia-reperfusion injury is involved in ischemic brain edema and neuroinflammation, thereby regulating cerebral ischemia-reperfusion injury. The aim of this study is to investigate the changes of glymphatic system after cerebral ischemia-reperfusion injury and whether limb remote ischemic postconditioning (LRIP) can improve the function of glymphatic system to protect the brain. METHODS To establish a focal brain I/R injury mouse model, this study utilized the middle cerebral artery occlusion/reperfusion (MCAO/R) method. The present study classified eight-week-old C57BL/6 male mice into three groups. The changes in glymphatic function in different periods of ischemia and reperfusion were analyzed through immunofluorescence, transmission electron microscopy (TEM), and Western-Blot (WB) assays. The contents of the evaluation included cerebrospinal fluid flow, swelling degree of brain tissue, aquaporin-4 (AQP4) expression and polarization, and amyloid-β (Aβ) excretion. RESULTS In the early stages of cerebral ischemia, cerebrospinal fluid (CSF) flow is disturbed, accompanied by a decrease in AQP4 polarization. The polarity of AQP4 decreased from 12 h to 72 h of reperfusion, the Aβ deposition. LRIP can increase the expression of β-DG and AQP4 polarization, reduce the deposition of Aβ, improve the function of the glymphatic system, and reduce the expression of AQP4 to play A protective role in brain. CONCLUSION Glymphatic system impaired after cerebral ischemia-reperfusion injury in mice. LRIP may play a neuroprotective role by improving glymphatic function after I/R.
Collapse
Affiliation(s)
- Xiaohong Li
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Xiaoli Tan
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Qian Zhou
- Department of Neurology, the Second Affiliated Hospital of Guilin Medical University, Guilin 541199, China
| | - Zhuoxi Xie
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Weiting Meng
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Yeyu Pang
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Lizhen Huang
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Zhihao Ding
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Yuanhong Hu
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Ruhua Li
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Guilan Huang
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Hao Li
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin 541001, China.
| |
Collapse
|
5
|
Chojnowski K, Opiełka M, Gozdalski J, Radziwon J, Dańczyszyn A, Aitken AV, Biancardi VC, Winklewski PJ. The Role of Arginine-Vasopressin in Stroke and the Potential Use of Arginine-Vasopressin Type 1 Receptor Antagonists in Stroke Therapy: A Narrative Review. Int J Mol Sci 2023; 24:ijms24032119. [PMID: 36768443 PMCID: PMC9916514 DOI: 10.3390/ijms24032119] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 01/15/2023] [Accepted: 01/16/2023] [Indexed: 01/25/2023] Open
Abstract
Stroke is a life-threatening condition in which accurate diagnoses and timely treatment are critical for successful neurological recovery. The current acute treatment strategies, particularly non-invasive interventions, are limited, thus urging the need for novel therapeutical targets. Arginine vasopressin (AVP) receptor antagonists are emerging as potential targets to treat edema formation and subsequent elevation in intracranial pressure, both significant causes of mortality in acute stroke. Here, we summarize the current knowledge on the mechanisms leading to AVP hyperexcretion in acute stroke and the subsequent secondary neuropathological responses. Furthermore, we discuss the work supporting the predictive value of measuring copeptin, a surrogate marker of AVP in stroke patients, followed by a review of the experimental evidence suggesting AVP receptor antagonists in stroke therapy. As we highlight throughout the narrative, critical gaps in the literature exist and indicate the need for further research to understand better AVP mechanisms in stroke. Likewise, there are advantages and limitations in using copeptin as a prognostic tool, and the translation of findings from experimental animal models to clinical settings has its challenges. Still, monitoring AVP levels and using AVP receptor antagonists as an add-on therapeutic intervention are potential promises in clinical applications to alleviate stroke neurological consequences.
Collapse
Affiliation(s)
- Karol Chojnowski
- Student Scientific Circle of the Department of Adult Neurology, Medical University of Gdansk, 17 Smoluchowskiego Street, 80-214 Gdansk, Poland
| | - Mikołaj Opiełka
- Student Scientific Circle of the Department of Adult Neurology, Medical University of Gdansk, 17 Smoluchowskiego Street, 80-214 Gdansk, Poland
| | - Jacek Gozdalski
- Department of Adult Neurology, Medical University of Gdansk, 17 Smoluchowskiego Street, 80-214 Gdansk, Poland
- Correspondence: (J.G.); (P.J.W.)
| | - Jakub Radziwon
- Student Scientific Circle of the Department of Adult Neurology, Medical University of Gdansk, 17 Smoluchowskiego Street, 80-214 Gdansk, Poland
| | - Aleksandra Dańczyszyn
- Student Scientific Circle of the Department of Adult Neurology, Medical University of Gdansk, 17 Smoluchowskiego Street, 80-214 Gdansk, Poland
| | - Andrew Vieira Aitken
- Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
- Center for Neurosciences Initiative, Auburn University, Auburn, AL 36849, USA
| | - Vinicia Campana Biancardi
- Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
- Center for Neurosciences Initiative, Auburn University, Auburn, AL 36849, USA
| | - Paweł Jan Winklewski
- Department of Human Physiology, Medical University of Gdansk, 15 Tuwima Street, 80-210 Gdansk, Poland
- 2nd Department of Radiology, Medical University of Gdansk, 17 Smoluchowskiego Street, 80-214 Gdansk, Poland
- Correspondence: (J.G.); (P.J.W.)
| |
Collapse
|
6
|
Fan X, Chen H, Xu C, Wang Y, Yin P, Li M, Tang Z, Jiang F, Wei W, Song J, Li G, Zhong D. S1PR3, as a Core Protein Related to Ischemic Stroke, is Involved in the Regulation of Blood–Brain Barrier Damage. Front Pharmacol 2022; 13:834948. [PMID: 35685645 PMCID: PMC9173650 DOI: 10.3389/fphar.2022.834948] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 04/15/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Ischemic stroke is the most common stroke incident. Sphingosine-1-phosphate (S1P) receptor 3 (S1PR3) is a member of the downstream G protein-coupled receptor family of S1P. The effect of S1PR3 on ischemic stroke remains elusive. Methods: We downloaded two middle cerebral artery occlusion (MCAO) microarray datasets from the Gene Expression Omnibus (GEO) database and screened differentially expressed genes (DEGs). Then, we performed a weighted gene coexpression network analysis (WGCNA) and identified the core module genes related to ischemic stroke. We constructed a protein–protein interaction (PPI) network for the core genes in which DEGs and WGCNA intersected. Finally, we discovered that S1PR3 was involved as the main member of the red proteome. Then, we explored the mechanism of S1PR3 in the mouse tMCAO model. The S1PR3-specific inhibitor CAY10444 was injected into the abdominal cavity of mice after cerebral ischemia/reperfusion (I/R) injury, and changes in the expression of blood–brain barrier-related molecules were measured using PCR, western blotting, and immunofluorescence staining. Results: Both GEO datasets showed that S1PR3 was upregulated during cerebral I/R in mice. WGCNA revealed that the light yellow module had the strongest correlation with the occurrence of IS. We determined the overlap with DEGs, identified 146 core genes that are potentially related to IS, and constructed a PPI network. Finally, S1PR3 was found to be the main member of the red proteome. In the mouse cerebral I/R model, S1PR3 expression increased 24 h after ischemia. After the administration of CAY10444, brain edema and neurological deficits in mice were ameliorated. CAY10444 rescued the decreased expression of the tight junction (TJ) proteins zonula occludens 1 (ZO1) and occludin after ischemia induced by transient MCAO (tMCAO) and reduced the increase in aquaporin 4 (AQP4) levels after tMCAO, preserving the integrity of the BBB. Finally, we found that S1PR3 is involved in regulating the mitogen-activated protein kinase (MAPK) and (phosphatidylinositol-3 kinase/serine-threonine kinase) PI3K-Akt signaling pathways. Conclusion: S1PR3 participates in the regulation of blood–brain barrier damage after cerebral I/R. S1PR3 is expected to be an indicator and predictor of cerebral ischemia, and drugs targeting S1PR3 may also provide new ideas for clinical medications.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Di Zhong
- *Correspondence: Guozhong Li, ; Di Zhong,
| |
Collapse
|
7
|
Glutamate Efflux across the Blood–Brain Barrier: New Perspectives on the Relationship between Depression and the Glutamatergic System. Metabolites 2022; 12:metabo12050459. [PMID: 35629963 PMCID: PMC9143347 DOI: 10.3390/metabo12050459] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/11/2022] [Accepted: 05/17/2022] [Indexed: 02/04/2023] Open
Abstract
Depression is a significant cause of disability and affects millions worldwide; however, antidepressant therapies often fail or are inadequate. Current medications for treating major depressive disorder can take weeks or months to reach efficacy, have troubling side effects, and are limited in their long-term capabilities. Recent studies have identified a new set of glutamate-based approaches, such as blood glutamate scavengers, which have the potential to provide alternatives to traditional antidepressants. In this review, we hypothesize as to the involvement of the glutamate system in the development of depression. We identify the mechanisms underlying glutamate dysregulation, offering new perspectives on the therapeutic modalities of depression with a focus on its relationship to blood–brain barrier (BBB) permeability. Ultimately, we conclude that in diseases with impaired BBB permeability, such as depression following stroke or traumatic brain injury, or in neurogenerative diseases, the glutamate system should be considered as a pathway to treatment. We propose that drugs such as blood glutamate scavengers should be further studied for treatment of these conditions.
Collapse
|
8
|
Zhou XB, Zhang YX, Zhou CX, Ma JJ. Chinese Herbal Medicine Adjusting Brain Microenvironment via Mediating Central Nervous System Lymphatic Drainage in Alzheimer's Disease. Chin J Integr Med 2022; 28:176-184. [PMID: 34731433 DOI: 10.1007/s11655-021-3342-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/02/2021] [Indexed: 02/05/2023]
Abstract
Due to its complex pathogenesis and lack of effective therapeutic methods, Alzheimer's disease (AD) has become a severe public health problem worldwide. Recent studies have discovered the function of central nervous system lymphatic drainage, which provides a new strategy for the treatment of AD. Chinese herbal medicine (CHM) has been considered as a cure for AD for hundreds of years in China, and its effect on scavenging β-amyloid protein in the brain of AD patients has been confirmed. In this review, the mechanism of central nervous system lymphatic drainage and the regulatory functions of CHM on correlation factors were briefly summarized. The advances in our understanding regarding the treatment of AD via regulating the central lymphatic system with CHM will promote the clinical application of CHM in AD patients and the discovery of new therapeutic drugs.
Collapse
Affiliation(s)
- Xi-Bin Zhou
- Department of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210046, China
| | - Yu-Xing Zhang
- Department of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210046, China
| | - Chun-Xiang Zhou
- Department of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210046, China
- Department of Traditional Chinese Medicine, Nanjing BenQ Hospital, Nanjing, 210036, China
| | - Jun-Jie Ma
- Department of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210046, China.
| |
Collapse
|
9
|
Szczygielski J, Kopańska M, Wysocka A, Oertel J. Cerebral Microcirculation, Perivascular Unit, and Glymphatic System: Role of Aquaporin-4 as the Gatekeeper for Water Homeostasis. Front Neurol 2021; 12:767470. [PMID: 34966347 PMCID: PMC8710539 DOI: 10.3389/fneur.2021.767470] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/12/2021] [Indexed: 12/13/2022] Open
Abstract
In the past, water homeostasis of the brain was understood as a certain quantitative equilibrium of water content between intravascular, interstitial, and intracellular spaces governed mostly by hydrostatic effects i.e., strictly by physical laws. The recent achievements in molecular bioscience have led to substantial changes in this regard. Some new concepts elaborate the idea that all compartments involved in cerebral fluid homeostasis create a functional continuum with an active and precise regulation of fluid exchange between them rather than only serving as separate fluid receptacles with mere passive diffusion mechanisms, based on hydrostatic pressure. According to these concepts, aquaporin-4 (AQP4) plays the central role in cerebral fluid homeostasis, acting as a water channel protein. The AQP4 not only enables water permeability through the blood-brain barrier but also regulates water exchange between perivascular spaces and the rest of the glymphatic system, described as pan-cerebral fluid pathway interlacing macroscopic cerebrospinal fluid (CSF) spaces with the interstitial fluid of brain tissue. With regards to this, AQP4 makes water shift strongly dependent on active processes including changes in cerebral microcirculation and autoregulation of brain vessels capacity. In this paper, the role of the AQP4 as the gatekeeper, regulating the water exchange between intracellular space, glymphatic system (including the so-called neurovascular units), and intravascular compartment is reviewed. In addition, the new concepts of brain edema as a misbalance in water homeostasis are critically appraised based on the newly described role of AQP4 for fluid permeation. Finally, the relevance of these hypotheses for clinical conditions (including brain trauma and stroke) and for both new and old therapy concepts are analyzed.
Collapse
Affiliation(s)
- Jacek Szczygielski
- Department of Neurosurgery, Institute of Medical Sciences, University of Rzeszów, Rzeszów, Poland.,Department of Neurosurgery, Faculty of Medicine and Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Marta Kopańska
- Department of Pathophysiology, Institute of Medical Sciences, University of Rzeszów, Rzeszów, Poland
| | - Anna Wysocka
- Chair of Internal Medicine and Department of Internal Medicine in Nursing, Faculty of Health Sciences, Medical University of Lublin, Lublin, Poland
| | - Joachim Oertel
- Department of Neurosurgery, Faculty of Medicine and Saarland University Medical Center, Saarland University, Homburg, Germany
| |
Collapse
|
10
|
Lv T, Zhao B, Hu Q, Zhang X. The Glymphatic System: A Novel Therapeutic Target for Stroke Treatment. Front Aging Neurosci 2021; 13:689098. [PMID: 34305569 PMCID: PMC8297504 DOI: 10.3389/fnagi.2021.689098] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/07/2021] [Indexed: 12/25/2022] Open
Abstract
The glymphatic system (GS) is a novel defined brain-wide perivascular transit network between cerebrospinal fluid (CSF) and interstitial solutes that facilitates the clearance of brain metabolic wastes. The complicated network of the GS consists of the periarterial CSF influx pathway, astrocytes-mediated convective transport of fluid and solutes supported by AQP4 water channels, and perivenous efflux pathway. Recent researches indicate that the GS dysfunction is associated with various neurological disorders, including traumatic brain injury, hydrocephalus, epilepsy, migraine, and Alzheimer’s disease (AD). Meanwhile, the GS also plays a pivotal role in the pathophysiological process of stroke, including brain edema, blood–brain barrier (BBB) disruption, immune cell infiltration, neuroinflammation, and neuronal apoptosis. In this review, we illustrated the key anatomical structures of the GS, the relationship between the GS and the meningeal lymphatic system, the interaction between the GS and the BBB, and the crosstalk between astrocytes and other GS cellular components. In addition, we contributed to the current knowledge about the role of the GS in the pathology of stroke and the role of AQP4 in stroke. We further discussed the potential use of the GS in early risk assessment, diagnostics, prognostics, and therapeutics of stroke.
Collapse
Affiliation(s)
- Tao Lv
- Department of Neurosurgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bing Zhao
- Department of Neurosurgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qin Hu
- Central Laboratory, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaohua Zhang
- Department of Neurosurgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
11
|
Enterovirus 71 infection induced Aquaporin-4 depolarization by increasing matrix metalloproteinase-9 activity. Neurosci Lett 2021; 759:136049. [PMID: 34126180 DOI: 10.1016/j.neulet.2021.136049] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 06/08/2021] [Accepted: 06/09/2021] [Indexed: 01/23/2023]
Abstract
Aquaporin-4 (AQP4) is the key water channel protein that regulates brain water homeostasis. Polarized expression of AQP4 on the astroglial endfeet facilitates its role in bi-directional brain water flux control. In the current study, we found that enterovirus 71 (EV71) infection induced depolarization of AQP4 in mouse brain, and demonstrated that β-dystroglycan (β-DG), the key component of dystrophin glycoprotein complex (DGC) that anchors AQP4 to the astroglial endfeet, was degraded upon infection. Elevated activity or expression of matrix metalloproteinase 9 (MMP9) upon infection was found in both mouse brains and patient cerebrospinal fluid (CSF) samples. Inhibiting MMP9 activity by SB-3CT rescued the decay of β-DG and reduced the depolarization of AQP4. Brain edema induced by viral infection was also ameliorated by SB-3CT treatment in mice.
Collapse
|
12
|
Shi Y, Fan X, Li G, Zhong D, Zhang X. Association of Serum Dystroglycan, MMP-2/9 and AQP-4 with Haematoma Expansion in Patients with Intracerebral Haemorrhage. Neuropsychiatr Dis Treat 2021; 17:11-18. [PMID: 33442252 PMCID: PMC7797333 DOI: 10.2147/ndt.s283016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/23/2020] [Indexed: 12/05/2022] Open
Abstract
OBJECTIVE The purpose of this study was to explore association of serum dystroglycan (DG), matrix metalloproteinase-2/matrix metalloproteinase-9 (MMP-2/9), and aquaporin-4 (AQP-4) expression and haematoma expansion in patients with intracerebral haemorrhage (ICH), which are proteins involved in maintaining the integrity of the blood-brain barrier. METHODS We included patients older than 18 years old with ICH who had undergone baseline CT within 6 hours after intracerebral haemorrhage symptom onset in our hospital between April 2018 and December 2018. Two readers independently assessed haematoma volume and other imaging information upon admission and again within 24 hours. All patients underwent 5 mL of venous blood collection 6 and 24 hours after admission. Serum expression levels of dystroglycan, matrix metalloproteinase-2/matrix metalloproteinase-9 and aquaporin-4 were determined by quantitative enzyme-linked immunosorbent assay (ELISA). Repeated analysis of variance was used to determine whether expression of the four proteins in patients with cerebral haemorrhage changed within 24 hours and whether there were differences between the haematoma enlargement and non-haematoma enlargement groups over time. Univariate and multivariate logistic regression analyses were used to compare the correlation among expression of the four proteins, clinical characteristics of patients and haematoma enlargement. RESULTS Expression levels of serum matrix metalloproteinase-2/matrix metalloproteinases-9 and aquaporin-4 gradually increased within 24 hours in patients with cerebral haemorrhage (P<0.001), while expression levels of dystroglycan gradually decreased (P<0.01). Expression of serum matrix metalloproteinases-9 6 hours after onset was independently correlated with the expansion of cerebral haemorrhage. The ROC curve (AUC=0.778, 95% Cl: 0.661-0.894, P<0.001) exhibited high sensitivity (0.900) and low specificity (0.642). CONCLUSION These data support that expression of MMP-9 in peripheral blood is independently correlated with the enlargement of haematoma in patients with intracerebral haemorrhage 6 hours after onset and can be used as an independent predictor of haematoma enlargement in patients with intracerebral haemorrhage. However, although the expression of MMP-2, AQP-4 and DG exhibited some changes within 6 and 24 hours after onset, they were not independently correlated with early haematoma enlargement in patients with intracerebral haemorrhage. Further multi-time point exploration and expansion of the sample size is necessary in future studies.
Collapse
Affiliation(s)
- Yue Shi
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, People's Republic of China
| | - Xuehui Fan
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, People's Republic of China
| | - Guozhong Li
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, People's Republic of China
| | - Di Zhong
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, People's Republic of China
| | - Xin Zhang
- Department of Neurology, Liuzhou People's Hospital, Liuzhou, Guangxi, 545006, People's Republic of China
| |
Collapse
|
13
|
Zhang H, Wang Y, Lian L, Zhang C, He Z. Glycine-Histidine-Lysine (GHK) Alleviates Astrocytes Injury of Intracerebral Hemorrhage via the Akt/miR-146a-3p/AQP4 Pathway. Front Neurosci 2020; 14:576389. [PMID: 33192260 PMCID: PMC7658812 DOI: 10.3389/fnins.2020.576389] [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: 06/26/2020] [Accepted: 09/03/2020] [Indexed: 12/22/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a major type of cerebrovascular disease with poor prognosis. Recent studies have shown that Glycyl-l-histidyl-l-lysine (GHK) is a kind of natural human tripeptide which could inhibit inflammation and against neurodegenerative diseases, but neither its role nor the mechanisms in ICH have yet been explicit. Currently, we investigated the possible strategies of GHK on ICH injury. Neurological deficit scores, brain water content, Nissl staining, and aquaporin 4 (AQP4) immunohistochemistry were detected in different groups of rats. The expression of microRNAs (miRNAs) was examined by real-time PCR. Inflammatory factors were detected using enzyme-linked immunosorbent assay (ELISA). Cell viability and cell proliferation were detected by Cell Counting Kit-8 (CCK-8). Matrix metalloproteinase 2 (MMP2), MMP9, tissue inhibitors of metalloproteinase-1 (TIMP1), AQP4 expression were detected/assessed using western blot. We observed that 5 and 10 μg/g of GHK improved neurological recovery by significantly reducing brain water content, improving neurological deficits, and promoting neuron survival. Besides, GHK alleviated inflammatory reaction and downregulated AQP4 expression. Furthermore, the effects of GHK on astrocyte were associated with the upregulation of miRNA-146a-3p, which partially regulated the expression of AQP4. Our results demonstrated that the phosphatidylinositol 3-kinase (PI3K)/AKT pathway participated in the GHK-induced upregulation of miR-146a-3p and miR-146a-3p/AQP4 interaction plays a role in the injury following ICH. These findings suggested that GHK could provide a novel therapeutic strategy for ICH.
Collapse
Affiliation(s)
- Heyu Zhang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, China.,Department of Neurology, First Hospital of China Medical University, Shenyang, China
| | - Yanzhe Wang
- Department of Neurology, First Hospital of China Medical University, Shenyang, China
| | - Ling Lian
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, China
| | - Cheng Zhang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, China
| | - Zhiyi He
- Department of Neurology, First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
14
|
Gao M, Lu W, Shu Y, Yang Z, Sun S, Xu J, Gan S, Zhu S, Qiu G, Zhuo F, Xu S, Wang Y, Chen J, Wu X, Huang J. Poldip2 mediates blood-brain barrier disruption and cerebral edema by inducing AQP4 polarity loss in mouse bacterial meningitis model. CNS Neurosci Ther 2020; 26:1288-1302. [PMID: 32790044 PMCID: PMC7702237 DOI: 10.1111/cns.13446] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 06/27/2020] [Accepted: 07/05/2020] [Indexed: 12/20/2022] Open
Abstract
Background Specific highly polarized aquaporin‐4 (AQP4) expression is reported to play a crucial role in blood‐brain barrier (BBB) integrity and brain water transport balance. The upregulation of polymerase δ‐interacting protein 2 (Poldip2) was involved in aggravating BBB disruption following ischemic stroke. This study aimed to investigate whether Poldip2‐mediated BBB disruption and cerebral edema formation in mouse bacterial meningitis (BM) model occur via induction of AQP4 polarity loss. Methods and Results Mouse BM model was induced by injecting mice with group B hemolytic streptococci via posterior cistern. Recombinant human Poldip2 (rh‐Poldip2) was administered intranasally at 1 hour after BM induction. Small interfering ribonucleic acid (siRNA) targeting Poldip2 was administered by intracerebroventricular (i.c.v) injection at 48 hours before BM induction. A specific inhibitor of matrix metalloproteinases (MMPs), UK383367, was administered intravenously at 0.5 hour before BM induction. Western blotting, immunofluorescence staining, quantitative real‐time PCR, neurobehavioral test, brain water content test, Evans blue (EB) permeability assay, transmission electron microscopy (TEM), and gelatin zymography were carried out. The results showed that Poldip2 was upregulated and AQP4 polarity was lost in mouse BM model. Both Poldip2 siRNA and UK383367 improved neurobehavioral outcomes, alleviated brain edema, preserved the integrity of BBB, and relieved the loss of AQP4 polarity in BM model. Rh‐Poldip2 upregulated the expression of MMPs and glial fibrillary acidic protein (GFAP) and downregulated the expression of β‐dystroglycan (β‐DG), zonula occludens‐1 (ZO‐1), occludin, and claudin‐5; whereas Poldip2 siRNA downregulated the expression of MMPs and GFAP, and upregulated β‐DG, ZO‐1, occludin, and claudin‐5. Similarly, UK383367 downregulated the expression of GFAP and upregulated the expression of β‐DG, ZO‐1, occludin, and claudin‐5. Conclusion Poldip2 inhibition alleviated brain edema and preserved the integrity of BBB partially by relieving the loss of AQP4 polarity via MMPs/β‐DG pathway.
Collapse
Affiliation(s)
- Meng Gao
- Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Weitian Lu
- Department of Anatomy, Chongqing Medical University, Chongqing, China.,Institute of Neuroscience, Chongqing Medical University, Chongqing, China
| | - Yue Shu
- Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Zhengyu Yang
- Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Shanquan Sun
- Department of Anatomy, Chongqing Medical University, Chongqing, China.,Institute of Neuroscience, Chongqing Medical University, Chongqing, China
| | - Jin Xu
- Department of Anatomy, Chongqing Medical University, Chongqing, China.,Institute of Neuroscience, Chongqing Medical University, Chongqing, China
| | - Shengwei Gan
- Department of Anatomy, Chongqing Medical University, Chongqing, China.,Institute of Neuroscience, Chongqing Medical University, Chongqing, China
| | - Shujuan Zhu
- Department of Anatomy, Chongqing Medical University, Chongqing, China.,Institute of Neuroscience, Chongqing Medical University, Chongqing, China
| | - Guoping Qiu
- Department of Anatomy, Chongqing Medical University, Chongqing, China.,Institute of Neuroscience, Chongqing Medical University, Chongqing, China
| | - Fei Zhuo
- Department of Anatomy, Chongqing Medical University, Chongqing, China.,Institute of Neuroscience, Chongqing Medical University, Chongqing, China
| | - Shiye Xu
- Department of Anatomy, Chongqing Medical University, Chongqing, China.,Institute of Neuroscience, Chongqing Medical University, Chongqing, China
| | - Yiying Wang
- Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Junhong Chen
- Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Xuan Wu
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Juan Huang
- Department of Anatomy, Chongqing Medical University, Chongqing, China.,Institute of Neuroscience, Chongqing Medical University, Chongqing, China
| |
Collapse
|
15
|
Patwa J, Flora SJS. Heavy Metal-Induced Cerebral Small Vessel Disease: Insights into Molecular Mechanisms and Possible Reversal Strategies. Int J Mol Sci 2020; 21:ijms21113862. [PMID: 32485831 PMCID: PMC7313017 DOI: 10.3390/ijms21113862] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/24/2020] [Accepted: 05/25/2020] [Indexed: 12/11/2022] Open
Abstract
Heavy metals are considered a continuous threat to humanity, as they cannot be eradicated. Prolonged exposure to heavy metals/metalloids in humans has been associated with several health risks, including neurodegeneration, vascular dysfunction, metabolic disorders, cancer, etc. Small blood vessels are highly vulnerable to heavy metals as they are directly exposed to the blood circulatory system, which has comparatively higher concentration of heavy metals than other organs. Cerebral small vessel disease (CSVD) is an umbrella term used to describe various pathological processes that affect the cerebral small blood vessels and is accepted as a primary contributor in associated disorders, such as dementia, cognitive disabilities, mood disorder, and ischemic, as well as a hemorrhagic stroke. In this review, we discuss the possible implication of heavy metals/metalloid exposure in CSVD and its associated disorders based on in-vitro, preclinical, and clinical evidences. We briefly discuss the CSVD, prevalence, epidemiology, and risk factors for development such as genetic, traditional, and environmental factors. Toxic effects of specific heavy metal/metalloid intoxication (As, Cd, Pb, Hg, and Cu) in the small vessel associated endothelium and vascular dysfunction too have been reviewed. An attempt has been made to highlight the possible molecular mechanism involved in the pathophysiology, such as oxidative stress, inflammatory pathway, matrix metalloproteinases (MMPs) expression, and amyloid angiopathy in the CSVD and related disorders. Finally, we discussed the role of cellular antioxidant defense enzymes to neutralize the toxic effect, and also highlighted the potential reversal strategies to combat heavy metal-induced vascular changes. In conclusion, heavy metals in small vessels are strongly associated with the development as well as the progression of CSVD. Chelation therapy may be an effective strategy to reduce the toxic metal load and the associated complications.
Collapse
|
16
|
Zhang G, Ma P, Wan S, Xu J, Yang M, Qiu G, Zhuo F, Xu S, Huo J, Ju Y, Liu H. Dystroglycan is involved in the activation of ERK pathway inducing the change of AQP4 expression in scratch-injured astrocytes. Brain Res 2019; 1721:146347. [DOI: 10.1016/j.brainres.2019.146347] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 06/28/2019] [Accepted: 07/21/2019] [Indexed: 01/28/2023]
|
17
|
Huang Y, Li SN, Zhou XY, Zhang LX, Chen GX, Wang TH, Xia QJ, Liang N, Zhang X. The Dual Role of AQP4 in Cytotoxic and Vasogenic Edema Following Spinal Cord Contusion and Its Possible Association With Energy Metabolism via COX5A. Front Neurosci 2019; 13:584. [PMID: 31258460 PMCID: PMC6587679 DOI: 10.3389/fnins.2019.00584] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 05/23/2019] [Indexed: 02/05/2023] Open
Abstract
Spinal cord edema, mainly including vasogenic and cytotoxic edema, influences neurological outcome after spinal cord contusion (SCC). Aquaporin 4 (AQP4) is the most ubiquitous water channel in the central nervous system (CNS), which is a rate-limiting factor in vasogenic edema expressing in brain injury, and it contributes to the formation of cytotoxic edema locating in astrocytes. However, little is known about the regulatory mechanism of AQP4 within vasogenic and cytotoxic edema in SCC, and whether the regulation mechanism of AQP4 is related to Cytochrome coxidase (COX5A) affecting energy metabolism. Therefore, the SCC model is established by Allen’s method, and the degree of edema and neuronal area is measured. The motor function of rats is evaluated by the Basso, Beattie, and Bresnahan (BBB) scoring system. Meanwhile, AQP4 and COX5A are detected by real-time quantitative PCR (qRT-PCR) and western blot (WB). The localization of targeted protein is exhibited by immunohistochemical staining (IHC) and immunofluorescence (IF). Additionally, the methodology of AQP4 lentivirus-mediated RNA interference (AQP4-RNAi) is used to reveal the effect on edema of SCC and the regulating molecular mechanism. Firstly, we observe that the tissue water content increases after SCC and decreases after the peak value of tissue water content at 3 days (P < 0.05) with abundant expression of AQP4 protein locating around vascular endothelial cells (VECs), which suggests that the increasing AQP4 promotes water reabsorption and improves vasogenic edema in the early stage of SCC. However, the neuronal area is larger than in the sham group in the 7 days (P < 0.05) with the total water content of spinal cord decrease. Meanwhile, AQP4 migrates from VECs to neuronal cytomembrane, which indicates that AQP4 plays a crucial role in aggravating the formation and development of cytotoxic edema in the middle stages of SCC. Secondly, AQP4-RNAi is used to elucidate the mechanism of AQP4 to edema of SCC. The neuronal area shrinks and the area of cytotoxic edema reduces after AQP4 downregulation. The BBB scores are significantly higher than in the vector group after AQP4-RNAi at 5, 7, and 14 (P < 0.05). There is a relationship between AQP4 and COX5A shown by bioinformatics analysis. After AQP4 inhibition, the expression of COX5A is significantly upregulated in the swelling astrocytes. Therefore, the inhibition of AQP4 expression reduces cytotoxic edema in SCC and improves motor function, which may be associated with upregulation of COX5A via affecting energy metabolism. Moreover, it is not clear how the inhibition of AQP4 directly causes the upregulation of COX5A.
Collapse
Affiliation(s)
- Yuan Huang
- Clinical Laboratory Medicine, Chengdu Medical College, Chengdu, China
| | - Sheng-Nan Li
- Center for Experimental Technology of Preclinical Medicine, Chengdu Medical College, Chengdu, China
| | - Xiu-Ya Zhou
- Center for Experimental Technology of Preclinical Medicine, Chengdu Medical College, Chengdu, China
| | | | - Gang-Xian Chen
- Center for Experimental Technology of Preclinical Medicine, Chengdu Medical College, Chengdu, China
| | - Ting-Hua Wang
- Institute of Neuroscience, Kunming Medical University, Kunming, China.,Institute of Neurological Diseases, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qing-Jie Xia
- Institute of Neurological Diseases, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Nan Liang
- Center for Experimental Technology of Preclinical Medicine, Chengdu Medical College, Chengdu, China
| | - Xiao Zhang
- Center for Experimental Technology of Preclinical Medicine, Chengdu Medical College, Chengdu, China
| |
Collapse
|
18
|
Zhang X, Gu Y, Li P, Jiang A, Sheng X, Jin X, Shi Y, Li G. Matrix Metalloproteases-Mediated Cleavage on β-Dystroglycan May Play a Key Role in the Blood-Brain Barrier After Intracerebral Hemorrhage in Rats. Med Sci Monit 2019; 25:794-800. [PMID: 30686819 PMCID: PMC6362757 DOI: 10.12659/msm.908500] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND It is well documented that the Blood-Brain barrier (BBB) can be damaged by matrix metalloproteases (MMPs) after intracerebral hemorrhage (ICH), but little is known about the mechanism of this effect. MATERIAL AND METHODS We established an ICH model in rats by injecting collagenase VII into the striatum. Afterwards, intraperitoneal injection of these rats with 40 mg/kg GM6001 (a MMPs inhibitor). The effects of GM6001 on ICH were investigated by neurological severity score, brain water content, Evans blue staining, hematoxylin-eosin staining, immunohistochemical staining, and Western blot assays. RESULTS We demonstrated that the neurological damage caused by ICH was relieved at 5 and 7 days following administration of GM6001. The impaired BBB induced by ICH was improved in response to GM6001 treatment at around 3 days, as evidenced by alleviated cerebral edema, decreased Evans blue extravasation, and a reduction in inflammatory cellular infiltration. Mechanism analysis revealed that ICH induced the generation of β-dystroglycan cleavage, which could be suppressed by GM6001 treatment. Furthermore, we found that recombinant MMP2 and MMP9 triggered the cleavage of β-dystroglycan in vitro, and this action could be inhibited by GM6001 administration. CONCLUSIONS Taken together, our results suggest that MMPs-mediated cleavage on β-dystroglycan may play an important role in BBB after ICH.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Yunhe Gu
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Peitong Li
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Anqi Jiang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Xiaomeng Sheng
- Department of Neurology, Harbin Fourth Hospital, Harbin, Heilongjiang, China (mainland)
| | - Xin Jin
- Department of Neurology, Jixi People's Hospital, Jixi, Heilongjiang, China (mainland)
| | - Yue Shi
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Guozhong Li
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| |
Collapse
|
19
|
Szczygielski J, Glameanu C, Müller A, Klotz M, Sippl C, Hubertus V, Schäfer KH, Mautes AE, Schwerdtfeger K, Oertel J. Changes in Posttraumatic Brain Edema in Craniectomy-Selective Brain Hypothermia Model Are Associated With Modulation of Aquaporin-4 Level. Front Neurol 2018; 9:799. [PMID: 30333785 PMCID: PMC6176780 DOI: 10.3389/fneur.2018.00799] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Accepted: 09/04/2018] [Indexed: 12/19/2022] Open
Abstract
Both hypothermia and decompressive craniectomy have been considered as a treatment for traumatic brain injury. In previous experiments we established a murine model of decompressive craniectomy and we presented attenuated edema formation due to focal brain cooling. Since edema development is regulated via function of water channel proteins, our hypothesis was that the effects of decompressive craniectomy and of hypothermia are associated with a change in aquaporin-4 (AQP4) concentration. Male CD-1 mice were assigned into following groups (n = 5): sham, decompressive craniectomy, trauma, trauma followed by decompressive craniectomy and trauma + decompressive craniectomy followed by focal hypothermia. After 24 h, magnetic resonance imaging with volumetric evaluation of edema and contusion were performed, followed by ELISA analysis of AQP4 concentration in brain homogenates. Additional histopathological analysis of AQP4 immunoreactivity has been performed at more remote time point of 28d. Correlation analysis revealed a relationship between AQP4 level and both volume of edema (r2 = 0.45, p < 0.01, **) and contusion (r2 = 0.41, p < 0.01, **) 24 h after injury. Aggregated analysis of AQP4 level (mean ± SEM) presented increased AQP4 concentration in animals subjected to trauma and decompressive craniectomy (52.1 ± 5.2 pg/mL, p = 0.01; *), but not to trauma, decompressive craniectomy and hypothermia (45.3 ± 3.6 pg/mL, p > 0.05; ns) as compared with animals subjected to decompressive craniectomy only (32.8 ± 2.4 pg/mL). However, semiquantitative histopathological analysis at remote time point revealed no significant difference in AQP4 immunoreactivity across the experimental groups. This suggests that AQP4 is involved in early stages of brain edema formation after surgical decompression. The protective effect of selective brain cooling may be related to change in AQP4 response after decompressive craniectomy. The therapeutic potential of this interaction should be further explored.
Collapse
Affiliation(s)
- Jacek Szczygielski
- Department of Neurosurgery, Faculty of Medicine, Saarland University Medical Center, Saarland University, Homburg, Germany.,Institute of Neuropathology, Faculty of Medicine, Saarland University Medical Center, Saarland University, Homburg, Germany.,Faculty of Medicine, University of Rzeszów, Rzeszów, Poland
| | - Cosmin Glameanu
- Department of Neurosurgery, Faculty of Medicine, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Andreas Müller
- Department of Radiology, Faculty of Medicine, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Markus Klotz
- Working Group Enteric Nervous System (AGENS), University of Applied Sciences Kaiserslautern, Kaiserslautern, Germany
| | - Christoph Sippl
- Department of Neurosurgery, Faculty of Medicine, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Vanessa Hubertus
- Department of Neurosurgery, Faculty of Medicine, Saarland University Medical Center, Saarland University, Homburg, Germany.,Department of Neurosurgery, Charité University Medicine, Berlin, Germany
| | - Karl-Herbert Schäfer
- Working Group Enteric Nervous System (AGENS), University of Applied Sciences Kaiserslautern, Kaiserslautern, Germany
| | - Angelika E Mautes
- Department of Neurosurgery, Faculty of Medicine, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Karsten Schwerdtfeger
- Department of Neurosurgery, Faculty of Medicine, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Joachim Oertel
- Department of Neurosurgery, Faculty of Medicine, Saarland University Medical Center, Saarland University, Homburg, Germany
| |
Collapse
|
20
|
Bai W, Zhou YG. Homeostasis of the Intraparenchymal-Blood Glutamate Concentration Gradient: Maintenance, Imbalance, and Regulation. Front Mol Neurosci 2017; 10:400. [PMID: 29259540 PMCID: PMC5723322 DOI: 10.3389/fnmol.2017.00400] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 11/20/2017] [Indexed: 12/25/2022] Open
Abstract
It is widely accepted that glutamate is the most important excitatory neurotransmitter in the central nervous system (CNS). However, there is also a large amount of glutamate in the blood. Generally, the concentration gradient of glutamate between intraparenchymal and blood environments is stable. However, this gradient is dramatically disrupted under a variety of pathological conditions, resulting in an amplifying cascade that causes a series of pathological reactions in the CNS and peripheral organs. This eventually seriously worsens a patient’s prognosis. These two “isolated” systems are rarely considered as a whole even though they mutually influence each other. In this review, we summarize what is currently known regarding the maintenance, imbalance and regulatory mechanisms that control the intraparenchymal-blood glutamate concentration gradient, discuss the interrelationships between these systems and further explore their significance in clinical practice.
Collapse
Affiliation(s)
- Wei Bai
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yuan-Guo Zhou
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
21
|
Kálmán M, Tóth L, Szöllosi D, Oszwald E, Mahalek J, Sadeghian S. Correlation Between Extravasation and Alterations of Cerebrovascular Laminin and β-Dystroglycan Immunoreactivity Following Cryogenic Lesions in Rats. J Neuropathol Exp Neurol 2017; 76:929-941. [PMID: 29044412 DOI: 10.1093/jnen/nlx081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The blood-brain barrier becomes "leaky" following lesions. Former studies revealed that following lesions the immunoreactivity of cerebrovascular laminin becomes detectable whereas that of β-dystroglycan disappears. These alterations may be indicators of glio-vascular decoupling that may result in the impairment of the blood-brain-barrier. This study investigates correlation between the post-lesion extravasation and the above-mentioned immunohistochemical alterations. Following cryogenic lesions, the survival periods lasted 5, 10, 30 minutes, 1 or 12 hours, or 1 day. Some brains were fixed immediately post-lesion. Immunofluorescent reactions were performed in floating sections. The extravasation was detected with immunostaining for plasma fibronectin and rat immunoglobulins. When the survival period was 30 minutes or longer, the area of extravasation corresponded to the area of altered laminin and β-dystroglycan immunoreactivities. Following immediate fixation some laminin immunoreactivity was already detected. The extravasation seemed to precede this early appearance of laminin immunoreactivity. The β-dystroglycan immunoreactivity disappeared later. When the extravasation spread into the corpus callosum, vascular laminin immunoreactivity appeared but the β-dystroglycan immunoreactivity persisted. It seems that extravasation separates the glial and vascular basal laminae, which results in the appearance of laminin immunoreactivity. The disappearance of β-dystroglycan immunoreactivity is neither a condition nor an inevitable consequence of the 2 other phenomena.
Collapse
Affiliation(s)
- Mihály Kálmán
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - László Tóth
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Dávid Szöllosi
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Erzsébet Oszwald
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Judit Mahalek
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Sam Sadeghian
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
22
|
Aquaporin-4 and Cerebrovascular Diseases. Int J Mol Sci 2016; 17:ijms17081249. [PMID: 27529222 PMCID: PMC5000647 DOI: 10.3390/ijms17081249] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 07/20/2016] [Accepted: 07/26/2016] [Indexed: 12/16/2022] Open
Abstract
Cerebrovascular diseases are conditions caused by problems with brain vasculature, which have a high morbidity and mortality. Aquaporin-4 (AQP4) is the most abundant water channel in the brain and crucial for the formation and resolution of brain edema. Considering brain edema is an important pathophysiological change after stoke, AQP4 is destined to have close relation with cerebrovascular diseases. However, this relation is not limited to brain edema due to other biological effects elicited by AQP4. Till now, multiple studies have investigated roles of AQP4 in cerebrovascular diseases. This review focuses on expression of AQP4 and the effects of AQP4 on brain edema and neural cells injuries in cerebrovascular diseases including cerebral ischemia, intracerebral hemorrhage and subarachnoid hemorrhage. In the current review, we pay more attention to the studies of recent years directly from cerebrovascular diseases animal models or patients, especially those using AQP4 gene knockout mice. This review also elucidates the potential of AQP4as an excellent therapeutic target.
Collapse
|
23
|
β-Dystroglycan cleavage by matrix metalloproteinase-2/-9 disturbs aquaporin-4 polarization and influences brain edema in acute cerebral ischemia. Neuroscience 2016; 326:141-157. [PMID: 27038751 DOI: 10.1016/j.neuroscience.2016.03.055] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2015] [Revised: 03/19/2016] [Accepted: 03/23/2016] [Indexed: 01/31/2023]
Abstract
Dystroglycan (DG) is widely expressed in various tissues, and throughout the cerebral microvasculature. It consists of two subunits, α-DG and β-DG, and the cleavage of the latter by matrix metalloproteinase (MMP)-2 and -9 underlies a number of physiological and pathological processes. However, the involvement of MMP-2/-9-mediated β-DG cleavage in cerebral ischemia remains uncertain. In astrocytes, DG is crucial for maintaining the polarization of aquaporin-4 (AQP4), which plays a role in the regulation of cytotoxic and vasogenic edema. The present study aimed to explore the effects of MMP-2/-9-mediated β-DG cleavage on AQP4 polarization and brain edema in acute cerebral ischemia. A model of cerebral ischemia was established via permanent middle cerebral artery occlusion (pMCAO) in male C57BL/6 mice. Western blotting, real-time polymerase chain reaction (PCR), immunohistochemical staining, immunofluorescent staining, electron microscopy, and light microscopy were used. Captopril was applied as a selective MMP-2/-9 inhibitor. Recombinant mouse MMP (rmMMP)-2 and -9 were used in an in vitro cleavage experiment. The present study demonstrated evidence of β-DG cleavage by MMP-2/-9 in pMCAO mouse brains; this cleavage was implicated in AQP4 redistribution and brain edema in cerebral ischemia. In addition, captopril exacerbated cytotoxic edema and ameliorated vasogenic edema at 24h after pMCAO, and alleviated brain edema and neurological deficit at 48h and 72h. In conclusion, this study provides novel insight into the effects of MMP-2/-9-mediated β-DG cleavage in acute cerebral ischemia. Such findings might facilitate the development of a therapeutic strategy for the optimization of MMP-2/-9 targeted treatment in cerebral ischemia.
Collapse
|