1
|
Che J, Sun Y, Deng Y, Zhang J. Blood-brain barrier disruption: a culprit of cognitive decline? Fluids Barriers CNS 2024; 21:63. [PMID: 39113115 PMCID: PMC11305076 DOI: 10.1186/s12987-024-00563-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 07/31/2024] [Indexed: 08/10/2024] Open
Abstract
Cognitive decline covers a broad spectrum of disorders, not only resulting from brain diseases but also from systemic diseases, which seriously influence the quality of life and life expectancy of patients. As a highly selective anatomical and functional interface between the brain and systemic circulation, the blood-brain barrier (BBB) plays a pivotal role in maintaining brain homeostasis and normal function. The pathogenesis underlying cognitive decline may vary, nevertheless, accumulating evidences support the role of BBB disruption as the most prevalent contributing factor. This may mainly be attributed to inflammation, metabolic dysfunction, cell senescence, oxidative/nitrosative stress and excitotoxicity. However, direct evidence showing that BBB disruption causes cognitive decline is scarce, and interestingly, manipulation of the BBB opening alone may exert beneficial or detrimental neurological effects. A broad overview of the present literature shows a close relationship between BBB disruption and cognitive decline, the risk factors of BBB disruption, as well as the cellular and molecular mechanisms underlying BBB disruption. Additionally, we discussed the possible causes leading to cognitive decline by BBB disruption and potential therapeutic strategies to prevent BBB disruption or enhance BBB repair. This review aims to foster more investigations on early diagnosis, effective therapeutics, and rapid restoration against BBB disruption, which would yield better cognitive outcomes in patients with dysregulated BBB function, although their causative relationship has not yet been completely established.
Collapse
Affiliation(s)
- Ji Che
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No.270 Dong'An Road, Xuhui District, Shanghai, 200032, P. R. China
| | - Yinying Sun
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No.270 Dong'An Road, Xuhui District, Shanghai, 200032, P. R. China
| | - Yixu Deng
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No.270 Dong'An Road, Xuhui District, Shanghai, 200032, P. R. China
| | - Jun Zhang
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No.270 Dong'An Road, Xuhui District, Shanghai, 200032, P. R. China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, P. R. China.
| |
Collapse
|
2
|
Lorin C, Guiet R, Chiaruttini N, Ambrosini G, Boci E, Abdellah M, Markram H, Keller D. Structural and molecular characterization of astrocyte and vasculature connectivity in the mouse hippocampus and cortex. Glia 2024. [PMID: 39007459 DOI: 10.1002/glia.24594] [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] [Received: 12/19/2023] [Revised: 07/02/2024] [Accepted: 07/04/2024] [Indexed: 07/16/2024]
Abstract
The relation of astrocytic endfeet to the vasculature plays a key functional role in the neuro-glia-vasculature unit. We characterize the spatial organization of astrocytes and the structural aspects that facilitate their involvement in molecular exchanges. Using double transgenic mice, we performed co-immunostaining, confocal microscopy, and three-dimensional digital segmentation to investigate the biophysical and molecular organization of astrocytes and their intricate endfoot network at the micrometer level in the isocortex and hippocampus. The results showed that hippocampal astrocytes had smaller territories, reduced endfoot dimensions, and fewer contacts with blood vessels compared with those in the isocortex. Additionally, we found that both connexins 43 and 30 have a higher density in the endfoot and the former is overexpressed relative to the latter. However, due to the limitations of the method, further studies are needed to determine the exact localization on the endfoot. The quantitative information obtained in this study will be useful for modeling the interactions of astrocytes with the vasculature.
Collapse
Affiliation(s)
- Charlotte Lorin
- Blue Brain Project, Swiss Federal Institute of Technology Lausanne (EPFL), Campus Biotech, Geneva, Switzerland
| | - Romain Guiet
- Bioimaging and Optics Platform, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Nicolas Chiaruttini
- Bioimaging and Optics Platform, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Giovanna Ambrosini
- Bioinformatics Competence Center, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
- Bioinformatics Competence Center, University of Lausanne, Lausanne, Switzerland
| | - Elvis Boci
- Blue Brain Project, Swiss Federal Institute of Technology Lausanne (EPFL), Campus Biotech, Geneva, Switzerland
| | - Marwan Abdellah
- Blue Brain Project, Swiss Federal Institute of Technology Lausanne (EPFL), Campus Biotech, Geneva, Switzerland
| | - Henry Markram
- Blue Brain Project, Swiss Federal Institute of Technology Lausanne (EPFL), Campus Biotech, Geneva, Switzerland
| | - Daniel Keller
- Blue Brain Project, Swiss Federal Institute of Technology Lausanne (EPFL), Campus Biotech, Geneva, Switzerland
| |
Collapse
|
3
|
Thormann M, Traube N, Yehia N, Koestler R, Galabova G, MacAulay N, Toft-Bertelsen TL. Toward New AQP4 Inhibitors: ORI-TRN-002. Int J Mol Sci 2024; 25:924. [PMID: 38255997 PMCID: PMC10815436 DOI: 10.3390/ijms25020924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/04/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Cerebral edema is a life-threatening condition that can cause permanent brain damage or death if left untreated. Existing therapies aim at mitigating the associated elevated intracranial pressure, yet they primarily alleviate pressure rather than prevent edema formation. Prophylactic anti-edema therapy necessitates novel drugs targeting edema formation. Aquaporin 4 (AQP4), an abundantly expressed water pore in mammalian glia and ependymal cells, has been proposed to be involved in cerebral edema formation. A series of novel compounds have been tested for their potential inhibitory effects on AQP4. However, selectivity, toxicity, functional inhibition, sustained therapeutic concentration, and delivery into the central nervous system are major challenges. Employing extensive density-functional theory (DFT) calculations, we identified a previously unreported thermodynamically stable tautomer of the recently identified AQP4-specific inhibitor TGN-020 (2-(nicotinamide)-1,3,4-thiadiazol). This novel form, featuring a distinct hydrogen-bonding pattern, served as a template for a COSMOsim-3D-based virtual screen of proprietary compounds from Origenis™. The screening identified ORI-TRN-002, an electronic homologue of TGN-020, demonstrating high solubility and low protein binding. Evaluating ORI-TRN-002 on AQP4-expressing Xenopus laevis oocytes using a high-resolution volume recording system revealed an IC50 of 2.9 ± 0.6 µM, establishing it as a novel AQP4 inhibitor. ORI-TRN-002 exhibits superior solubility and overcomes free fraction limitations compared to other reported AQP4 inhibitors, suggesting its potential as a promising anti-edema therapy for treating cerebral edema in the future.
Collapse
Affiliation(s)
| | - Nadine Traube
- Origenis GmbH, Am Klopferspitz 19A, 82152 Martinsried, Germany
| | - Nasser Yehia
- Origenis GmbH, Am Klopferspitz 19A, 82152 Martinsried, Germany
| | - Roland Koestler
- Origenis GmbH, Am Klopferspitz 19A, 82152 Martinsried, Germany
| | | | - Nanna MacAulay
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen, Denmark
| | - Trine L. Toft-Bertelsen
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen, Denmark
| |
Collapse
|
4
|
Zhao Z, He J, Chen Y, Wang Y, Wang C, Tan C, Liao J, Xiao G. The pathogenesis of idiopathic normal pressure hydrocephalus based on the understanding of AQP1 and AQP4. Front Mol Neurosci 2022; 15:952036. [PMID: 36204139 PMCID: PMC9530743 DOI: 10.3389/fnmol.2022.952036] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 08/12/2022] [Indexed: 11/21/2022] Open
Abstract
Idiopathic normal pressure hydrocephalus (iNPH) is a neurological disorder without a recognized cause. Aquaporins (AQPs) are transmembrane channels that carry water through cell membranes and are critical for cerebrospinal fluid circulation and cerebral water balance. The function of AQPs in developing and maintaining hydrocephalus should be studied in greater detail as a possible diagnostic and therapeutic tool. Recent research indicates that patients with iNPH exhibited high levels of aquaporin 1 and low levels of aquaporin 4 expression, suggesting that these AQPs are essential in iNPH pathogenesis. To determine the source of iNPH and diagnose and treat it, it is necessary to examine and appreciate their function in the genesis and maintenance of hydrocephalus. The expression, function, and regulation of AQPs in iNPH are reviewed in this article, in order to provide fresh targets and suggestions for future research.
Collapse
Affiliation(s)
- Zitong Zhao
- 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
| | - Jian He
- Department of Pediatrics, 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
| | - Yuchang Wang
- 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
| | - Chuansen Wang
- 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
| | - Changwu Tan
- 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
| | - 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
| | - 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
- *Correspondence: Gelei Xiao
| |
Collapse
|
5
|
Abstract
The central nervous system (CNS) has been viewed as an immunologically privileged site, but emerging works are uncovering a large array of neuroimmune interactions primarily occurring at its borders. CNS barriers sites host diverse population of both innate and adaptive immune cells capable of, directly and indirectly, influence the function of the residing cells of the brain parenchyma. These structures are only starting to reveal their role in controlling brain function under normal and pathological conditions and represent an underexplored therapeutic target for the treatment of brain disorders. This review will highlight the development of the CNS barriers to host neuro-immune interactions and emphasize their newly described roles in neurodevelopmental, neurological, and neurodegenerative disorders, particularly for the meninges.
Collapse
Affiliation(s)
- Natalie M Frederick
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Gabriel A Tavares
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Antoine Louveau
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Molecular Medicine, Cleveland Clinic College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA.,Kent University, Neurosciences, School of Biomedical Sciences, Cleveland, Ohio, USA
| |
Collapse
|
6
|
MacAulay N. Reply to 'Aquaporin 4 and glymphatic flow have central roles in brain fluid homeostasis'. Nat Rev Neurosci 2021; 22:651-652. [PMID: 34408335 DOI: 10.1038/s41583-021-00515-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Nanna MacAulay
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
7
|
Abstract
Our brains consist of 80% water, which is continuously shifted between different compartments and cell types during physiological and pathophysiological processes. Disturbances in brain water homeostasis occur with pathologies such as brain oedema and hydrocephalus, in which fluid accumulation leads to elevated intracranial pressure. Targeted pharmacological treatments do not exist for these conditions owing to our incomplete understanding of the molecular mechanisms governing brain water transport. Historically, the transmembrane movement of brain water was assumed to occur as passive movement of water along the osmotic gradient, greatly accelerated by water channels termed aquaporins. Although aquaporins govern the majority of fluid handling in the kidney, they do not suffice to explain the overall brain water movement: either they are not present in the membranes across which water flows or they appear not to be required for the observed flow of water. Notably, brain fluid can be secreted against an osmotic gradient, suggesting that conventional osmotic water flow may not describe all transmembrane fluid transport in the brain. The cotransport of water is an unconventional molecular mechanism that is introduced in this Review as a missing link to bridge the gap in our understanding of cellular and barrier brain water transport.
Collapse
Affiliation(s)
- Nanna MacAulay
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
8
|
Pro-Inflammatory Role of AQP4 in Mice Subjected to Intrastriatal Injections of the Parkinsonogenic Toxin MPP. Cells 2020; 9:cells9112418. [PMID: 33167342 PMCID: PMC7694382 DOI: 10.3390/cells9112418] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/01/2020] [Accepted: 11/02/2020] [Indexed: 12/28/2022] Open
Abstract
Aquaporin-4 (AQP4) is critically involved in brain water and volume homeostasis and has been implicated in a wide range of pathological conditions. Notably, evidence has been accrued to suggest that AQP4 plays a proinflammatory role by promoting release of astrocytic cytokines that activate microglia and other astrocytes. Neuroinflammation is a hallmark of Parkinson’s disease (PD), and we have previously shown that astrocytes in substantia nigra (SN) are enriched in AQP4 relative to cortical astrocytes, and that their complement of AQP4 is further increased following treatment with the parkinsonogenic toxin MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine). Here, we investigated the effect of Aqp4 deletion on microglial activation in mice subjected to unilateral intrastriatal injection of 1-methyl-4-phenylpyridinium (MPP+, the toxic metabolite of MPTP). Our results show that MPP+ injections lead to a pronounced increase in the expression level of microglial activating genes in the ventral mesencephalon of wild type (WT) mice, but not Aqp4−/− mice. We also show, in WT mice, that MPP+ injections cause an upregulation of nigral AQP4 and swelling of astrocytic endfeet. These findings are consistent with the idea that AQP4 plays a pro-inflammatory role in Parkinson’s disease, secondary to the dysregulation of astrocytic volume homeostasis.
Collapse
|
9
|
Cash A, Theus MH. Mechanisms of Blood-Brain Barrier Dysfunction in Traumatic Brain Injury. Int J Mol Sci 2020; 21:ijms21093344. [PMID: 32397302 PMCID: PMC7246537 DOI: 10.3390/ijms21093344] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/02/2020] [Accepted: 05/04/2020] [Indexed: 12/16/2022] Open
Abstract
Traumatic brain injuries (TBIs) account for the majority of injury-related deaths in the United States with roughly two million TBIs occurring annually. Due to the spectrum of severity and heterogeneity in TBIs, investigation into the secondary injury is necessary in order to formulate an effective treatment. A mechanical consequence of trauma involves dysregulation of the blood–brain barrier (BBB) which contributes to secondary injury and exposure of peripheral components to the brain parenchyma. Recent studies have shed light on the mechanisms of BBB breakdown in TBI including novel intracellular signaling and cell–cell interactions within the BBB niche. The current review provides an overview of the BBB, novel detection methods for disruption, and the cellular and molecular mechanisms implicated in regulating its stability following TBI.
Collapse
Affiliation(s)
- Alison Cash
- The Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA;
| | - Michelle H. Theus
- The Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA;
- The Center for Regenerative Medicine, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA
- Correspondence: ; Tel.: 1-540-231-0909; Fax: 1-540-231-7425
| |
Collapse
|
10
|
MacAulay N. Molecular mechanisms of K + clearance and extracellular space shrinkage-Glia cells as the stars. Glia 2020; 68:2192-2211. [PMID: 32181522 DOI: 10.1002/glia.23824] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 02/28/2020] [Accepted: 03/04/2020] [Indexed: 12/17/2022]
Abstract
Neuronal signaling in the central nervous system (CNS) associates with release of K+ into the extracellular space resulting in transient increases in [K+ ]o . This elevated K+ is swiftly removed, in part, via uptake by neighboring glia cells. This process occurs in parallel to the [K+ ]o elevation and glia cells thus act as K+ sinks during the neuronal activity, while releasing it at the termination of the pulse. The molecular transport mechanisms governing this glial K+ absorption remain a point of debate. Passive distribution of K+ via Kir4.1-mediated spatial buffering of K+ has become a favorite within the glial field, although evidence for a quantitatively significant contribution from this ion channel to K+ clearance from the extracellular space is sparse. The Na+ /K+ -ATPase, but not the Na+ /K+ /Cl- cotransporter, NKCC1, shapes the activity-evoked K+ transient. The different isoform combinations of the Na+ /K+ -ATPase expressed in glia cells and neurons display different kinetic characteristics and are thereby distinctly geared toward their temporal and quantitative contribution to K+ clearance. The glia cell swelling occurring with the K+ transient was long assumed to be directly associated with K+ uptake and/or AQP4, although accumulating evidence suggests that they are not. Rather, activation of bicarbonate- and lactate transporters appear to lead to glial cell swelling via the activity-evoked alkaline transient, K+ -mediated glial depolarization, and metabolic demand. This review covers evidence, or lack thereof, accumulated over the last half century on the molecular mechanisms supporting activity-evoked K+ and extracellular space dynamics.
Collapse
Affiliation(s)
- Nanna MacAulay
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
11
|
Hoddevik EH, Rao SB, Zahl S, Boldt HB, Ottersen OP, Amiry-Moghaddam M. Organisation of extracellular matrix proteins laminin and agrin in pericapillary basal laminae in mouse brain. Brain Struct Funct 2020; 225:805-816. [PMID: 32072250 PMCID: PMC7046580 DOI: 10.1007/s00429-020-02036-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 01/29/2020] [Indexed: 01/08/2023]
Abstract
Evidence suggests that extracellular matrix molecules of perivascular basal laminae help orchestrate the molecular assemblies at the gliovascular interface. Specifically, laminin and agrin are thought to tether the dystrophin-associated protein (DAP) complex to the astrocytic basal lamina. This complex includes α-syntrophin (α-Syn), which is believed to anchor aquaporin-4 (AQP4) to astrocytic endfoot membrane domains. We have previously shown that the size of the perivascular AQP4 pool differs considerably between brain regions in an α-Syn-dependent manner. Also, both AQP4 and α-Syn occur at higher densities in endfoot membrane domains facing pericytes than in endfoot membrane domains facing endothelial cells. The heterogeneous distribution of AQP4 at the regional and capillary level has been attributed to a direct interaction between AQP4 and α-Syn. This would be challenged (1) if the microdistributions of laminin and agrin fail to align with those of DAP and AQP4 and (2) if targeted deletion of α-Syn leads to a loss of laminin and/or agrin. Here, we provide the first detailed and quantitative analysis of laminin and agrin in brain basal laminae of mice. We show that the microdistributions of these molecules vary in a fashion that is well aligned with the previously reported microdistribution of AQP4. We also demonstrate that the expression patterns of laminin and agrin are insensitive to targeted deletion of α-Syn, suggesting that α-Syn deletion affects AQP4 directly and not indirectly via laminin or agrin. These data fill remaining voids in the current model of how key molecules are assembled and tethered at the gliovascular interface.
Collapse
Affiliation(s)
- Eystein Hellstrøm Hoddevik
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Blindern, Post box 1105, 0317, Oslo, Norway
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Shreyas Balachandra Rao
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Blindern, Post box 1105, 0317, Oslo, Norway
| | - Soulmaz Zahl
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Blindern, Post box 1105, 0317, Oslo, Norway
| | - Henning Bünsow Boldt
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Blindern, Post box 1105, 0317, Oslo, Norway
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Ole Petter Ottersen
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Blindern, Post box 1105, 0317, Oslo, Norway
- President's Office, Karolinska Institutet, Nobels väg 6, 171 77, Stockholm, Sweden
| | - Mahmood Amiry-Moghaddam
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Blindern, Post box 1105, 0317, Oslo, Norway.
| |
Collapse
|
12
|
Zhang Y, Xu K, Liu Y, Erokwu BO, Zhao P, Flask CA, Ramos-Estebanez C, Farr GW, LaManna JC, Boron WF, Yu X. Increased cerebral vascularization and decreased water exchange across the blood-brain barrier in aquaporin-4 knockout mice. PLoS One 2019; 14:e0218415. [PMID: 31220136 PMCID: PMC6586297 DOI: 10.1371/journal.pone.0218415] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 05/31/2019] [Indexed: 12/26/2022] Open
Abstract
Aquaporin-4 (AQP4) plays an important role in regulating water exchange across the blood-brain barrier (BBB) and brain-cerebrospinal fluid interface. Studies on AQP-4 knockout mice (AQP4-KO) have reported considerable protection from brain edema induced by acute water intoxication and ischemic stroke, identifying AQP4 as a potential target for therapeutic interventions. However, the long-term effects of chronic AQP4 suppression are yet to be elucidated. In the current study, we evaluated the physiological and structural changes in adult AQP4-KO mice using magnetic resonance imaging (MRI) and immunohistochemical analysis. Water exchange across BBB was assessed by tracking an intravenous bolus injection of oxygen-17 (17O) water (H217O) using 17O-MRI. Cerebral blood flow (CBF) was quantified using arterial spin-labeling (ASL) MRI. Capillary density was determined by immunohistochemical staining for glucose transporter-1 (GLUT1). Compared to wildtype control mice, AQP4-KO mice showed a significant reduction in peak and steady-state H217O uptake despite unaltered CBF. Interestingly, a 22% increase in cortical capillary density was observed in AQP4-KO mice. These results suggest that increased cerebral vascularization may be an adaptive response to chronic reduction in water exchange across BBB in AQP4-KO mice.
Collapse
Affiliation(s)
- Yifan Zhang
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States of America
- Department of Radiology, Case Western Reserve University, Cleveland, OH, United States of America
- * E-mail: (YZ); (XY)
| | - Kui Xu
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH, United States of America
| | - Yuchi Liu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States of America
| | - Bernadette O. Erokwu
- Department of Radiology, Case Western Reserve University, Cleveland, OH, United States of America
| | - Pan Zhao
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH, United States of America
| | - Chris A. Flask
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States of America
- Department of Radiology, Case Western Reserve University, Cleveland, OH, United States of America
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, United States of America
| | - Ciro Ramos-Estebanez
- Department of Neurology, Case Western Reserve University, Cleveland, OH, United States of America
| | - George W. Farr
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH, United States of America
- Aeromics, LLC, Cleveland, OH, United States of America
| | - Joseph C. LaManna
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH, United States of America
| | - Walter F. Boron
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH, United States of America
| | - Xin Yu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States of America
- Department of Radiology, Case Western Reserve University, Cleveland, OH, United States of America
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH, United States of America
- * E-mail: (YZ); (XY)
| |
Collapse
|
13
|
The potential roles of aquaporin 4 in amyotrophic lateral sclerosis. Neurol Sci 2019; 40:1541-1549. [PMID: 30980198 DOI: 10.1007/s10072-019-03877-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 03/28/2019] [Indexed: 12/13/2022]
Abstract
Aquaporin 4 (AQP4) is a primary water channel found on astrocytes in the central nervous system (CNS). Besides its function in water and ion homeostasis, AQP4 has also been documented to be involved in a myriad of acute and chronic cerebral pathologies, including autoimmune neurodegenerative diseases. AQP4 has been postulated to be associated with the incidence of a progressive neurodegenerative disorder known as amyotrophic lateral sclerosis (ALS), a disease that targets the motor neurons, causing muscle weakness and eventually paralysis. Raised AQP4 levels were noted in association with vessels surrounded with swollen astrocytic processes as well as in the brainstem, cortex, and gray matter in patients with terminal ALS. AQP4 depolarization may lead to motor neuron degeneration in ALS via GLT-1. Besides, alterations in AQP4 expression in ALS may result in the loss of blood-brain barrier (BBB) integrity. Changes in AQP4 function may also disrupt K+ homeostasis and cause connexin dysregulation, the latter of which is associated to ALS disease progression. Furthermore, AQP4 suppression augments recovery in motor function in ALS, a phenomenon thought to be associated to NGF. No therapeutic drug targeting AQP4 has been developed to date. Nevertheless, the plethora of suggestive experimental results underscores the significance of further exploration into this area.
Collapse
|
14
|
Chronic stress impairs the aquaporin-4-mediated glymphatic transport through glucocorticoid signaling. Psychopharmacology (Berl) 2019; 236:1367-1384. [PMID: 30607477 DOI: 10.1007/s00213-018-5147-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 12/03/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND The glymphatic system has recently been proposed to function as a brain-wide macroscopic system for the clearance of potentially harmful molecules, such as amyloid beta (e.g., Aβ), from the brain parenchyma. Previous literatures have established that the glymphatic function is dramatically suppressed by aging, traumatic brain injury, and some diseases. However, the effect of chronic stress on the glymphatic function and its underlying mechanism remains largely unknown. METHODS Adult mice were randomly divided into four groups: chronic unpredictable mild stress (CUMS)-treated group, CUMS simultaneously treated with mifepristone (MFP) group, dexamethasone (DEX)-treated group, and control group. Stress response was observed by assessing the change of body weight, plasma corticosterone level, and behavior tests. The level of Aβ42 in cerebral tissue was assessed by ELISA. The glymphatic function was determined by using fluorescence tracer injection. The expression and localization of aquaporin-4 (AQP4) were evaluated by immunohistochemistry and western blot. The transcription level of AQP4 and anchoring molecules was evaluated by real-time PCR. FINDINGS Compared with control group, CUMS-treated mice exhibited the impairment of global glymphatic function especially in the anterior brain. This change was accompanied by the decreased expression and polarization of AQP4, reduced transcription of AQP4, agrin, laminin, and dystroglycan in the anterior cortex. Similarly, the glucocorticoid receptor (GR) agonist DEX exposure could reduce the glymphatic function and AQP4 expression. Moreover, the GR antagonist MFP treatment could significantly rescue the glymphatic function and reverse the expression and polarization of AQP4 impaired by CUMS. CONCLUSION Chronic stress could impair the AQP4-mediated glymphatic transport in the brain through glucocorticoid signaling. Our results also suggest that GR antagonist could be beneficial to rescue the glymphatic function suppressed by chronic stress.
Collapse
|
15
|
Hasan-Olive MM, Enger R, Hansson HA, Nagelhus EA, Eide PK. Loss of perivascular aquaporin-4 in idiopathic normal pressure hydrocephalus. Glia 2018; 67:91-100. [DOI: 10.1002/glia.23528] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 08/10/2018] [Accepted: 08/15/2018] [Indexed: 12/31/2022]
Affiliation(s)
- Md Mahdi Hasan-Olive
- Department of Neurosurgery; Oslo University Hospital - Rikshospitalet; Oslo Norway
- Institute of Clinical Medicine, Faculty of Medicine; University of Oslo; Oslo Norway
| | - Rune Enger
- GliaLab and Letten Centre, Division of Physiology, Department of Molecular Medicine; Institute of Basic Medical Sciences, University of Oslo; Oslo Norway
- Department of Neurology; Oslo University Hospital - Rikshospitalet; Oslo Norway
| | | | - Erlend A. Nagelhus
- GliaLab and Letten Centre, Division of Physiology, Department of Molecular Medicine; Institute of Basic Medical Sciences, University of Oslo; Oslo Norway
- Department of Neurology; Oslo University Hospital - Rikshospitalet; Oslo Norway
| | - Per Kristian Eide
- Department of Neurosurgery; Oslo University Hospital - Rikshospitalet; Oslo Norway
- Institute of Clinical Medicine, Faculty of Medicine; University of Oslo; Oslo Norway
| |
Collapse
|
16
|
Lan YL, Wang X, Lou JC, Ma XC, Zhang B. The potential roles of aquaporin 4 in malignant gliomas. Oncotarget 2018; 8:32345-32355. [PMID: 28423683 PMCID: PMC5458289 DOI: 10.18632/oncotarget.16017] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 02/22/2017] [Indexed: 11/25/2022] Open
Abstract
Aquaporin 4 (AQP4) is the major water channel expressed in the central nervous system and is primarily expressed in astrocytes. Recently, accumulated evidence has pointed to AQP4 as a key molecule that could play a critical role in glioma development. Discoveries of the role of AQP4 in cell migration suggest that AQP4 could be a significant factor regarding glioma malignancies. However, the AQP4 expression levels in glioma have not been fully elucidated; furthermore, the correlation of AQP4 expression with glioma malignancy remains controversial. Here, we review the expression pattern and predictive significance of AQP4 in malignant glioma. The molecular mechanism of AQP4 as it pertains to the migration and invasion of human glioma cells has been summarized. In addition, the important roles of AQP4 in combating drug resistance as well as potential pharmacological blockers of AQP4 have been systematically discussed. More research should be conducted to elucidate the potential roles of AQP4 in malignant glioma for identifying the tumor type, progression stages and optimal treatment strategies. The observed experimental results strongly emphasize the importance of this topic for future investigations.
Collapse
Affiliation(s)
- Yu-Long Lan
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, China.,Department of Pharmacy, Dalian Medical University, Dalian, China.,Department of Physiology, Dalian Medical University, Dalian, China.,Department of Neurosurgery, The Third People's Hospital of Dalian, Non-Directly Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xun Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, China.,Department of Neurosurgery, The Third People's Hospital of Dalian, Non-Directly Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jia-Cheng Lou
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiao-Chi Ma
- Department of Pharmacy, Dalian Medical University, Dalian, China
| | - Bo Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
17
|
Prydz A, Stahl K, Puchades M, Davarpaneh N, Nadeem M, Ottersen OP, Gundersen V, Amiry-Moghaddam M. Subcellular expression of aquaporin-4 in substantia nigra of normal and MPTP-treated mice. Neuroscience 2017; 359:258-266. [DOI: 10.1016/j.neuroscience.2017.07.029] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 06/17/2017] [Accepted: 07/12/2017] [Indexed: 02/06/2023]
|
18
|
Temporal Profiling of Astrocyte Precursors Reveals Parallel Roles for Asef during Development and after Injury. J Neurosci 2017; 36:11904-11917. [PMID: 27881777 DOI: 10.1523/jneurosci.1658-16.2016] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 09/12/2016] [Accepted: 09/18/2016] [Indexed: 12/21/2022] Open
Abstract
Lineage development is a stepwise process, governed by stage-specific regulatory factors and associated markers. Astrocytes are one of the principle cell types in the CNS and the stages associated with their development remain very poorly defined. To identify these stages, we performed gene-expression profiling on astrocyte precursor populations in the spinal cord, identifying distinct patterns of gene induction during their development that are strongly correlated with human astrocytes. Validation studies identified a new cohort of astrocyte-associated genes during development and demonstrated their expression in reactive astrocytes in human white matter injury (WMI). Functional studies on one of these genes revealed that mice lacking Asef exhibited impaired astrocyte differentiation during development and repair after WMI, coupled with compromised blood-brain barrier integrity in the adult CNS. These studies have identified distinct stages of astrocyte lineage development associated with human WMI and, together with our functional analysis of Asef, highlight the parallels between astrocyte development and their reactive counterparts associated with injury. SIGNIFICANCE STATEMENT Astrocytes play a central role in CNS function and associated diseases. Yet the mechanisms that control their development remain poorly defined. Using the developing mouse spinal cord as a model system, we identify molecular changes that occur in developing astrocytes. These molecular signatures are strongly correlated with human astrocyte expression profiles and validation in mouse spinal cord identifies a host of new genes associated with the astrocyte lineage. These genes are present in reactive astrocytes in human white matter injury, and functional studies reveal that one of these genes, Asef, contributes to reactive astrocyte responses after injury. These studies identify distinct stages of astrocyte lineage development and highlight the parallels between astrocyte development and their reactive counterparts associated with injury.
Collapse
|
19
|
Abstract
Neuronal survival, electrical signaling and synaptic activity require a well-balanced micro-environment in the central nervous system. This is achieved by the blood-brain barrier (BBB), an endothelial barrier situated in the brain capillaries, that controls near-to-all passage in and out of the brain. The endothelial barrier function is highly dependent on signaling interactions with surrounding glial, neuronal and vascular cells, together forming the neuro-glio-vascular unit. Within this functional unit, connexin (Cx) channels are of utmost importance for intercellular communication between the different cellular compartments. Connexins are best known as the building blocks of gap junction (GJ) channels that enable direct cell-cell transfer of metabolic, biochemical and electric signals. In addition, beyond their role in direct intercellular communication, Cxs also form unapposed, non-junctional hemichannels in the plasma membrane that allow the passage of several paracrine messengers, complementing direct GJ communication. Within the NGVU, Cxs are expressed in vascular endothelial cells, including those that form the BBB, and are eminent in astrocytes, especially at their endfoot processes that wrap around cerebral vessels. However, despite the density of Cx channels at this so-called gliovascular interface, it remains unclear as to how Cx-based signaling between astrocytes and BBB endothelial cells may converge control over BBB permeability in health and disease. In this review we describe available evidence that supports a role for astroglial as well as endothelial Cxs in the regulation of BBB permeability during development as well as in disease states.
Collapse
|
20
|
Enger R, Dukefoss DB, Tang W, Pettersen KH, Bjørnstad DM, Helm PJ, Jensen V, Sprengel R, Vervaeke K, Ottersen OP, Nagelhus EA. Deletion of Aquaporin-4 Curtails Extracellular Glutamate Elevation in Cortical Spreading Depression in Awake Mice. Cereb Cortex 2017; 27:24-33. [PMID: 28365776 PMCID: PMC5939213 DOI: 10.1093/cercor/bhw359] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 10/25/2016] [Accepted: 10/29/2016] [Indexed: 12/20/2022] Open
Abstract
Cortical spreading depression (CSD) is a phenomenon that challenges the homeostatic mechanisms on which normal brain function so critically depends. Analyzing the sequence of events in CSD holds the potential of providing new insight in the physiological processes underlying normal brain function as well as the pathophysiology of neurological conditions characterized by ionic dyshomeostasis. Here, we have studied the sequential progression of CSD in awake wild-type mice and in mice lacking aquaporin-4 (AQP4) or inositol 1,4,5-triphosphate type 2 receptor (IP3R2). By the use of a novel combination of genetically encoded sensors that a novel combination - an unprecedented temporal and spatial resolution, we show that CSD leads to brisk Ca2+ signals in astrocytes and that the duration of these Ca2+ signals is shortened in the absence of AQP4 but not in the absence of IP3R2. The decrease of the astrocytic, AQP4-dependent Ca2+ signals, coincides in time and space with a decrease in the duration of extracellular glutamate overflow but not with the initial peak of the glutamate release suggesting that in CSD, extracellular glutamate accumulation is extended through AQP4-dependent glutamate release from astrocytes. The present data point to a salient glial contribution to CSD and identify AQP4 as a new target for therapy.
Collapse
Affiliation(s)
- Rune Enger
- Department of Neurology, Oslo University Hospital, N-0027 Oslo, Norway
- GliaLab and Letten Centre, Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, N-0317 Oslo, Norway
| | - Didrik B. Dukefoss
- GliaLab and Letten Centre, Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, N-0317 Oslo, Norway
| | - Wannan Tang
- GliaLab and Letten Centre, Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, N-0317 Oslo, Norway
| | - Klas H. Pettersen
- GliaLab and Letten Centre, Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, N-0317 Oslo, Norway
| | - Daniel M. Bjørnstad
- GliaLab and Letten Centre, Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, N-0317 Oslo, Norway
| | - P. Johannes Helm
- GliaLab and Letten Centre, Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, N-0317 Oslo, Norway
| | - Vidar Jensen
- GliaLab and Letten Centre, Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, N-0317 Oslo, Norway
| | - Rolf Sprengel
- Department of Molecular Neurobiology, Max Planck Institute for Medical Research, D-69120 Heidelberg, Germany
- Max Planck Research Group at the Institute for Anatomy and Cell Biology, Heidelberg University, D-69120 Heidelberg, Germany
| | - Koen Vervaeke
- Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, N-0317 Oslo, Norway
| | - Ole P. Ottersen
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, N-0317 Oslo, Norway
| | - Erlend A. Nagelhus
- Department of Neurology, Oslo University Hospital, N-0027 Oslo, Norway
- GliaLab and Letten Centre, Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, N-0317 Oslo, Norway
| |
Collapse
|
21
|
Factors determining the density of AQP4 water channel molecules at the brain-blood interface. Brain Struct Funct 2016; 222:1753-1766. [PMID: 27629271 PMCID: PMC5406442 DOI: 10.1007/s00429-016-1305-y] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 09/04/2016] [Indexed: 10/27/2022]
Abstract
Perivascular endfeet of astrocytes are enriched with aquaporin-4 (AQP4)-a water channel that is critically involved in water transport at the brain-blood interface and that recently was identified as a key molecule in a system for waste clearance. The factors that determine the size of the perivascular AQP4 pool remain to be identified. Here we show that the size of this pool differs considerably between brain regions, roughly mirroring regional differences in Aqp4 mRNA copy numbers. We demonstrate that a targeted deletion of α-syntrophin-a member of the dystrophin complex responsible for AQP4 anchoring-removes a substantial and fairly constant proportion (79-94 %) of the perivascular AQP4 pool across the central nervous system (CNS). Quantitative immunogold analyses of AQP4 and α-syntrophin in perivascular membranes indicate that there is a fixed stoichiometry between these two molecules. Both molecules occur at higher densities in endfoot membrane domains facing pericytes than in endfoot membrane domains facing endothelial cells. Our data suggest that irrespective of region, endfoot targeting of α-syntrophin is the single most important factor determining the size of the perivascular AQP4 pool and hence the capacity for water transport at the brain-blood interface.
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: 31] [Impact Index Per Article: 3.9] [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
|
Liang F, Luo C, Xu G, Su F, He X, Long S, Ren H, Liu Y, Feng Y, Pei Z. Deletion of aquaporin-4 is neuroprotective during the acute stage of micro traumatic brain injury in mice. Neurosci Lett 2015; 598:29-35. [PMID: 25957560 DOI: 10.1016/j.neulet.2015.05.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 04/24/2015] [Accepted: 05/02/2015] [Indexed: 10/23/2022]
Abstract
Micro traumatic brain injury (TBI) is the most common type of brain injury, but the mechanisms underlying it are poorly understood. Aquaporin-4 (AQP4) is a water channel expressed in astrocyte end-feet, which plays an important role in brain edema. However, little is known about the role of AQP4 in micro TBI. Here, we examined the role of AQP4 in the pathogenesis of micro TBI in a closed-skull brain injury model, using two-photon microscopy. Our results indicate that AQP4 deletion reduced cell death, water content, astrocyte swelling and lesion volume during the acute stage of micro TBI. Our data revealed that astrocyte swelling is a decisive pathophysiological factor in the acute phase of this form of micro brain injury. Thus, treatments that inhibit AQP4 could be used as a neuroprotective strategy for micro TBI.
Collapse
Affiliation(s)
- Fengyin Liang
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Chuanming Luo
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Guangqing Xu
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Fengjuan Su
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiaofei He
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Simei Long
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Huixia Ren
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Yaning Liu
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Yanqing Feng
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China.
| | - Zhong Pei
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
24
|
Assentoft M, Larsen BR, MacAulay N. Regulation and Function of AQP4 in the Central Nervous System. Neurochem Res 2015; 40:2615-27. [PMID: 25630715 DOI: 10.1007/s11064-015-1519-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 01/08/2015] [Accepted: 01/12/2015] [Indexed: 01/09/2023]
Abstract
Aquaporin 4 (AQP4) is the predominant water channel in the mammalian brain and is mainly expressed in the perivascular glial endfeet at the brain-blood interface. Based on studies on AQP4(-/-) mice, AQP4 has been assigned physiological roles in stimulus-induced K(+) clearance, paravascular fluid flow, and brain edema formation. Conflicting data have been presented on the role of AQP4 in K(+) clearance and associated extracellular space shrinkage and on the stroke-induced alterations of AQP4 expression levels during edema formation, raising questions about the functional importance of AQP4 in these (patho)physiological aspects. Phosphorylation-dependent gating of AQP4 has been proposed as a regulatory mechanism for AQP4-mediated osmotic water transport. This paradigm was, however, recently challenged by experimental evidence and molecular dynamics simulations. Regulatory patterns and physiological roles for AQP4 thus remain to be fully explored.
Collapse
Affiliation(s)
- Mette Assentoft
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, Bldg. 12.6, 2200, Copenhagen, Denmark
| | - Brian Roland Larsen
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, Bldg. 12.6, 2200, Copenhagen, Denmark
| | - Nanna MacAulay
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, Bldg. 12.6, 2200, Copenhagen, Denmark.
| |
Collapse
|
25
|
Bond WS, Rex TS. Evidence That Erythropoietin Modulates Neuroinflammation through Differential Action on Neurons, Astrocytes, and Microglia. Front Immunol 2014; 5:523. [PMID: 25374571 PMCID: PMC4205853 DOI: 10.3389/fimmu.2014.00523] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 10/06/2014] [Indexed: 12/15/2022] Open
Abstract
Neuroinflammation is a normal and healthy response to neuronal damage. However, excessive or chronic neuroinflammation exacerbates neurodegeneration after trauma and in progressive diseases such as Alzheimer’s, Parkinson’s, age-related macular degeneration, and glaucoma. Therefore, molecules that modulate neuroinflammation are candidates as neuroprotective agents. Erythropoietin (EPO) is a known neuroprotective agent that indirectly attenuates neuroinflammation, in part, by inhibiting neuronal apoptosis. In this review, we provide evidence that EPO also modulates neuroinflammation upstream of apoptosis by acting directly on glia. Further, the signaling induced by EPO may differ depending on cell type and context possibly as a result of activation of different receptors. While significant progress has been made in our understanding of EPO signaling, this review also identifies areas for future study in terms of the role of EPO in modulating neuroinflammation.
Collapse
Affiliation(s)
- Wesley S Bond
- Vanderbilt Eye Institute, Vanderbilt University Medical Center , Nashville, TN , USA ; Vanderbilt Brain Institute, Vanderbilt University Medical Center , Nashville, TN , USA
| | - Tonia S Rex
- Vanderbilt Eye Institute, Vanderbilt University Medical Center , Nashville, TN , USA ; Vanderbilt Brain Institute, Vanderbilt University Medical Center , Nashville, TN , USA
| |
Collapse
|
26
|
Chu H, Ding H, Tang Y, Dong Q. Erythropoietin protects against hemorrhagic blood-brain barrier disruption through the effects of aquaporin-4. J Transl Med 2014; 94:1042-53. [PMID: 24978642 DOI: 10.1038/labinvest.2014.84] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Revised: 04/08/2014] [Accepted: 04/10/2014] [Indexed: 11/09/2022] Open
Abstract
Erythropoietin (EPO) has protective effects against many neurological diseases, including intracerebral hemorrhage (ICH). Here, we aimed to test EPO's effects on blood-brain barrier (BBB) disruption morphologically and functionally following ICH, which has not been well investigated. We also examined whether the effects were dependent on aquaporin-4 (AQP4). We detected the expression of perihematomal AQP4 and EPO receptor (EPOR) induced by EPO injection at 1, 3 and 7 days after ICH. We also examined the effects of EPO on BBB disruption by ICH in wild-type mice, and tested whether such effects were AQP4 dependent by using AQP4 knock-out mice. Furthermore, we assessed the related signal transduction pathways via astrocyte cultures. We found that EPO highly increased perihematomal AQP4 and EPOR expression. Specifically, EPO led to BBB protection in both types of mice by functionally reducing brain edema and BBB permeability, as well as morphologically suppressing tight junction (TJ) opening and endothelial cell swelling, and increasing expression of the TJ proteins occludin and zonula occluden-1 (ZO-1). Statistical analysis indicated that AQP4 was required for these effects. In addition, EPO upregulated phosphorylation of C-Jun amino-terminal kinase (JNK) and p38-mitogen-activated protein kinase (MAPK) as well as EPOR and AQP4 proteins in cultured astrocytes. The latter was inhibited by JNK and p38-MAPK inhibitors. Our data suggest that EPO protects BBB from disruption after ICH and that the main targets are the TJ proteins occludin and ZO-1. The effects of EPO are associated with increased levels of AQP4, and may occur through activation of JNK and p38-MAPK pathways after binding to EPOR.
Collapse
Affiliation(s)
- Heling Chu
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Hongyan Ding
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Yuping Tang
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| |
Collapse
|
27
|
Stokum JA, Kurland DB, Gerzanich V, Simard JM. Mechanisms of astrocyte-mediated cerebral edema. Neurochem Res 2014; 40:317-28. [PMID: 24996934 DOI: 10.1007/s11064-014-1374-3] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Revised: 06/18/2014] [Accepted: 06/26/2014] [Indexed: 11/26/2022]
Abstract
Cerebral edema formation stems from disruption of blood brain barrier (BBB) integrity and occurs after injury to the CNS. Due to the restrictive skull, relatively small increases in brain volume can translate into impaired tissue perfusion and brain herniation. In excess, cerebral edema can be gravely harmful. Astrocytes are key participants in cerebral edema by virtue of their relationship with the cerebral vasculature, their unique compliment of solute and water transport proteins, and their general role in brain volume homeostasis. Following the discovery of aquaporins, passive conduits of water flow, aquaporin 4 (AQP4) was identified as the predominant astrocyte water channel. Normally, AQP4 is highly enriched at perivascular endfeet, the outermost layer of the BBB, whereas after injury, AQP4 expression disseminates to the entire astrocytic plasmalemma, a phenomenon termed dysregulation. Arguably, the most important role of AQP4 is to rapidly neutralize osmotic gradients generated by ionic transporters. In pathological conditions, AQP4 is believed to be intimately involved in the formation and clearance of cerebral edema. In this review, we discuss aquaporin function and localization in the BBB during health and injury, and we examine post-injury ionic events that modulate AQP4-dependent edema formation.
Collapse
Affiliation(s)
- Jesse A Stokum
- Department of Neurosurgery, University of Maryland School of Medicine, 22 S. Greene St., Suite S12D, Baltimore, MD, 21201-1595, USA
| | | | | | | |
Collapse
|
28
|
Engelhardt B, Liebner S. Novel insights into the development and maintenance of the blood-brain barrier. Cell Tissue Res 2014; 355:687-99. [PMID: 24590145 PMCID: PMC3972432 DOI: 10.1007/s00441-014-1811-2] [Citation(s) in RCA: 211] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 01/13/2014] [Indexed: 01/20/2023]
Abstract
The blood-brain barrier (BBB) is essential for maintaining homeostasis within the central nervous system (CNS) and is a prerequisite for proper neuronal function. The BBB is localized to microvascular endothelial cells that strictly control the passage of metabolites into and out of the CNS. Complex and continuous tight junctions and lack of fenestrae combined with low pinocytotic activity make the BBB endothelium a tight barrier for water soluble moleucles. In combination with its expression of specific enzymes and transport molecules, the BBB endothelium is unique and distinguishable from all other endothelial cells in the body. During embryonic development, the CNS is vascularized by angiogenic sprouting from vascular networks originating outside of the CNS in a precise spatio-temporal manner. The particular barrier characteristics of BBB endothelial cells are induced during CNS angiogenesis by cross-talk with cellular and acellular elements within the developing CNS. In this review, we summarize the currently known cellular and molecular mechanisms mediating brain angiogenesis and introduce more recently discovered CNS-specific pathways (Wnt/β-catenin, Norrin/Frizzled4 and hedgehog) and molecules (GPR124) that are crucial in BBB differentiation and maturation. Finally, based on observations that BBB dysfunction is associated with many human diseases such as multiple sclerosis, stroke and brain tumors, we discuss recent insights into the molecular mechanisms involved in maintaining barrier characteristics in the mature BBB endothelium.
Collapse
Affiliation(s)
- Britta Engelhardt
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, 3012 Bern, Switzerland
| | - Stefan Liebner
- Institute of Neurology (Edinger Institute), University Hospital Frankfurt, Heinrich-Hoffmann-Strasse 7, 60528 Frankfurt/Main, Germany
| |
Collapse
|
29
|
Abstract
Aquaporin-4 (AQP4) is one of the most abundant molecules in the brain and is particularly prevalent in astrocytic membranes at the blood-brain and brain-liquor interfaces. While AQP4 has been implicated in a number of pathophysiological processes, its role in brain physiology has remained elusive. Only recently has evidence accumulated to suggest that AQP4 is involved in such diverse functions as regulation of extracellular space volume, potassium buffering, cerebrospinal fluid circulation, interstitial fluid resorption, waste clearance, neuroinflammation, osmosensation, cell migration, and Ca(2+) signaling. AQP4 is also required for normal function of the retina, inner ear, and olfactory system. A review will be provided of the physiological roles of AQP4 in brain and of the growing list of data that emphasize the polarized nature of astrocytes.
Collapse
|
30
|
Alvarez JI, Katayama T, Prat A. Glial influence on the blood brain barrier. Glia 2013; 61:1939-58. [PMID: 24123158 PMCID: PMC4068281 DOI: 10.1002/glia.22575] [Citation(s) in RCA: 385] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 08/13/2013] [Accepted: 08/19/2013] [Indexed: 12/14/2022]
Abstract
The Blood Brain Barrier (BBB) is a specialized vascular structure tightly regulating central nervous system (CNS) homeostasis. Endothelial cells are the central component of the BBB and control of their barrier phenotype resides on astrocytes and pericytes. Interactions between these cells and the endothelium promote and maintain many of the physiological and metabolic characteristics that are unique to the BBB. In this review we describe recent findings related to the involvement of astroglial cells, including radial glial cells, in the induction of barrier properties during embryogenesis and adulthood. In addition, we describe changes that occur in astrocytes and endothelial cells during injury and inflammation with a particular emphasis on alterations of the BBB phenotype. GLIA 2013;61:1939–1958
Collapse
Affiliation(s)
- Jorge Ivan Alvarez
- Neuroimmunology unit, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | | | | |
Collapse
|
31
|
Gundersen GA, Vindedal GF, Skare O, Nagelhus EA. Evidence that pericytes regulate aquaporin-4 polarization in mouse cortical astrocytes. Brain Struct Funct 2013; 219:2181-6. [PMID: 23982198 PMCID: PMC4223569 DOI: 10.1007/s00429-013-0629-0] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2013] [Accepted: 08/16/2013] [Indexed: 11/29/2022]
Abstract
Aquaporin-4 (AQP4) water channels are concentrated in astrocytic endfoot membranes at the brain–blood and brain–cerebrospinal fluid interfaces. The mechanisms underpinning the polarized distribution of AQP4 are poorly understood. Here we tested the hypothesis that pericytes regulate AQP4 anchoring to perivascular astrocytic endfoot membranes. AQP4 immunofluorescence of brain sections obtained from novel transgenic double reporter mice expressing enhanced green fluorescent protein (eGFP) in astrocytes and Discoma Red (DsRed) in pericytes revealed strong AQP4 signal in astrocytic processes adjacent to pericytes. Quantitative immunogold analysis of C57BL/6 mice showed that the AQP4 expression was higher in endfoot membranes abutting pericytes than in those facing endothelial cells. Similar findings were made for α-syntrophin, a member of the dystrophin-associated protein complex (DAPC). The enrichment of α-syntrophin in membranes ensheathing pericytes persisted after Aqp4 gene deletion. Our data support the concept that pericytes regulate AQP4 polarization.
Collapse
Affiliation(s)
- Georg Andreas Gundersen
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo, P.O. Box 1137, Blindern, 0318, Oslo, Norway
| | | | | | | |
Collapse
|
32
|
Jin BJ, Zhang H, Binder DK, Verkman AS. Aquaporin-4-dependent K(+) and water transport modeled in brain extracellular space following neuroexcitation. ACTA ACUST UNITED AC 2013; 141:119-32. [PMID: 23277478 PMCID: PMC3536523 DOI: 10.1085/jgp.201210883] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Potassium (K(+)) ions released into brain extracellular space (ECS) during neuroexcitation are efficiently taken up by astrocytes. Deletion of astrocyte water channel aquaporin-4 (AQP4) in mice alters neuroexcitation by reducing ECS [K(+)] accumulation and slowing K(+) reuptake. These effects could involve AQP4-dependent: (a) K(+) permeability, (b) resting ECS volume, (c) ECS contraction during K(+) reuptake, and (d) diffusion-limited water/K(+) transport coupling. To investigate the role of these mechanisms, we compared experimental data to predictions of a model of K(+) and water uptake into astrocytes after neuronal release of K(+) into the ECS. The model computed the kinetics of ECS [K(+)] and volume, with input parameters including initial ECS volume, astrocyte K(+) conductance and water permeability, and diffusion in astrocyte cytoplasm. Numerical methods were developed to compute transport and diffusion for a nonstationary astrocyte-ECS interface. The modeling showed that mechanisms b-d, together, can predict experimentally observed impairment in K(+) reuptake from the ECS in AQP4 deficiency, as well as altered K(+) accumulation in the ECS after neuroexcitation, provided that astrocyte water permeability is sufficiently reduced in AQP4 deficiency and that solute diffusion in astrocyte cytoplasm is sufficiently low. The modeling thus provides a potential explanation for AQP4-dependent K(+)/water coupling in the ECS without requiring AQP4-dependent astrocyte K(+) permeability. Our model links the physical and ion/water transport properties of brain cells with the dynamics of neuroexcitation, and supports the conclusion that reduced AQP4-dependent water transport is responsible for defective neuroexcitation in AQP4 deficiency.
Collapse
Affiliation(s)
- Byung-Ju Jin
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | | | | |
Collapse
|
33
|
Alvestad S, Hammer J, Hoddevik EH, Skare Ø, Sonnewald U, Amiry-Moghaddam M, Ottersen OP. Mislocalization of AQP4 precedes chronic seizures in the kainate model of temporal lobe epilepsy. Epilepsy Res 2013; 105:30-41. [DOI: 10.1016/j.eplepsyres.2013.01.006] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Revised: 01/04/2013] [Accepted: 01/06/2013] [Indexed: 10/27/2022]
|
34
|
Iliff JJ, Wang M, Liao Y, Plogg BA, Peng W, Gundersen GA, Benveniste H, Vates GE, Deane R, Goldman SA, Nagelhus EA, Nedergaard M. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid β. Sci Transl Med 2013; 4:147ra111. [PMID: 22896675 DOI: 10.1126/scitranslmed.3003748] [Citation(s) in RCA: 3231] [Impact Index Per Article: 293.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Because it lacks a lymphatic circulation, the brain must clear extracellular proteins by an alternative mechanism. The cerebrospinal fluid (CSF) functions as a sink for brain extracellular solutes, but it is not clear how solutes from the brain interstitium move from the parenchyma to the CSF. We demonstrate that a substantial portion of subarachnoid CSF cycles through the brain interstitial space. On the basis of in vivo two-photon imaging of small fluorescent tracers, we showed that CSF enters the parenchyma along paravascular spaces that surround penetrating arteries and that brain interstitial fluid is cleared along paravenous drainage pathways. Animals lacking the water channel aquaporin-4 (AQP4) in astrocytes exhibit slowed CSF influx through this system and a ~70% reduction in interstitial solute clearance, suggesting that the bulk fluid flow between these anatomical influx and efflux routes is supported by astrocytic water transport. Fluorescent-tagged amyloid β, a peptide thought to be pathogenic in Alzheimer's disease, was transported along this route, and deletion of the Aqp4 gene suppressed the clearance of soluble amyloid β, suggesting that this pathway may remove amyloid β from the central nervous system. Clearance through paravenous flow may also regulate extracellular levels of proteins involved with neurodegenerative conditions, its impairment perhaps contributing to the mis-accumulation of soluble proteins.
Collapse
Affiliation(s)
- Jeffrey J Iliff
- Center for Translational Neuromedicine, Department of Neurosurgery, University of Rochester Medical Center, Rochester, NY 14642, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
The aquaporins (AQPs) are plasma membrane water-transporting proteins. AQP4 is the principal member of this protein family in the CNS, where it is expressed in astrocytes and is involved in water movement, cell migration and neuroexcitation. AQP1 is expressed in the choroid plexus, where it facilitates cerebrospinal fluid secretion, and in dorsal root ganglion neurons, where it tunes pain perception. The AQPs are potential drug targets for several neurological conditions. Astrocytoma cells strongly express AQP4, which may facilitate their infiltration into the brain, and the neuroinflammatory disease neuromyelitis optica is caused by AQP4-specific autoantibodies that produce complement-mediated astrocytic damage.
Collapse
Affiliation(s)
- Marios C Papadopoulos
- Academic Neurosurgery Unit, St. George's, University of London, Tooting, London, SW17 0RE, UK. mpapadop@sgul. ac.uk
| | | |
Collapse
|
36
|
Cruz NF, Ball KK, Froehner SC, Adams ME, Dienel GA. Regional registration of [6-(14)C]glucose metabolism during brain activation of α-syntrophin knockout mice. J Neurochem 2013; 125:247-59. [PMID: 23346911 DOI: 10.1111/jnc.12166] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 01/02/2013] [Accepted: 01/20/2013] [Indexed: 12/16/2022]
Abstract
α-Syntrophin is a component of the dystrophin scaffold-protein complex that serves as an adaptor for recruitment of key proteins to the cytoplasmic side of plasma membranes. α-Syntrophin knockout (KO) causes loss of the polarized localization of aquaporin4 (AQP4) at astrocytic endfeet and interferes with water and K(+) homeostasis. During brain activation, release of ions and metabolites from endfeet is anticipated to increase perivascular fluid osmolarity, AQP4-mediated osmotic water flow from endfeet, and metabolite washout from brain. This study tests the hypothesis that reduced levels of endfoot AQP4 increase retention of [(14)C]metabolites during sensory stimulation. Conscious KO and wild-type mice were pulse-labeled with [6-(14)C] glucose during unilateral acoustic stimulation or bilateral acoustic plus whisker stimulation, and label retention was assayed by computer-assisted brain imaging or analysis of [(14)C]metabolites in extracts, respectively. High-resolution autoradiographic assays detected a 17% side-to-side difference (p < 0.05) in inferior colliculus of KO mice, not wild-type mice. However, there were no labeling differences between KO and wild-type mice for five major HPLC fractions from four dissected regions, presumably because of insufficient anatomical resolution. The results suggest a role for AQP4-mediated water flow in support of washout of metabolites, and underscore the need for greater understanding of astrocytic water and metabolite fluxes.
Collapse
Affiliation(s)
- Nancy F Cruz
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Kelly K Ball
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Stanley C Froehner
- Department of Physiology and Biophysics, University of Washington, Seattle, Washington, USA
| | - Marvin E Adams
- Department of Physiology and Biophysics, University of Washington, Seattle, Washington, USA
| | - Gerald A Dienel
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
37
|
Bataveljić D, Nikolić L, Milosević M, Todorović N, Andjus PR. Changes in the astrocytic aquaporin-4 and inwardly rectifying potassium channel expression in the brain of the amyotrophic lateral sclerosis SOD1(G93A) rat model. Glia 2012; 60:1991-2003. [PMID: 22987392 DOI: 10.1002/glia.22414] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2012] [Accepted: 08/10/2012] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease affecting upper and lower motor neurons. Dysfunction and death of motor neurons are closely related to the modified astrocytic environment. Astrocytic endfeet, lining the blood-brain barrier (BBB), are enriched in two proteins, aquaporin-4 (AQP4) and inwardly rectifying potassium channel (Kir) 4.1. Both channels are important for the maintainance of a functional BBB astrocytic lining. In this study, expression levels of AQP4 and Kir4.1 were for the first time examined in the brainstem and cortex, along with the functional properties of Kir channels in cultured cortical astrocytes of the SOD1(G93A) rat model of ALS. Western blot analysis showed increased expression of AQP4 and decreased expression of Kir4.1 in the brainstem and cortex of the ALS rat. In addition, higher immunoreactivity of AQP4 and reduced immunolabeling of Kir4.1 in facial and trigeminal nuclei as well as in the motor cortex were also observed. Particularly, the observed changes in the expression of both channels were retained in cultured astrocytes. Furthermore, whole-cell patch-clamp recordings from cultured ALS cortical astrocytes showed a significantly lower Kir current density. Importantly, the potassium uptake current in ALS astrocytes was significantly reduced at all extracellular potassium concentrations. Consequently, the Kir-specific Cs(+)- and Ba(2+)-sensitive currents were also decreased. The changes in the studied channels, notably at the upper CNS level, could underline the hampered ability of astrocytes to maintain water and potassium homeostasis, thus affecting the BBB, disturbing the neuronal microenvironment, and causing motoneuronal dysfunction and death.
Collapse
Affiliation(s)
- Danijela Bataveljić
- Center for Laser Microscopy, Faculty of Biology, University of Belgrade, Studentski trg 3, POB 52, Belgrade, Serbia
| | | | | | | | | |
Collapse
|
38
|
Benabdesselam R, Dorbani-Mamine L, Benmessaoud-Mesbah O, Rendon A, Mhaouty-Kodja S, Hardin-Pouzet H. Dp71 gene disruption alters the composition of the dystrophin-associated protein complex and neuronal nitric oxide synthase expression in the hypothalamic supraoptic and paraventricular nuclei. J Endocrinol 2012; 213:239-49. [PMID: 22493004 DOI: 10.1530/joe-12-0066] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
DP71 is the major cerebral dystrophin isoform and exerts its multiple functions via the dystrophin-associated protein complex (DAPC), also comprised of β-dystroglycan (β-DG) and α1-syntrophin (α1-Syn). Since DP71 disruption leads to impairment in the central control of the osmoregulatory axis, we investigated: 1) the DAPC composition in the hypothalamic supraoptic nucleus (SON) and paraventricular nucleus (PVN) of Dp71-null mice; and 2) the expression and activity of neuronal nitric oxide synthase (nNOS), because it is a potential partner of the DAPC and a functional index of osmoregulatory axis activity. In wild-type mice, dystrophins and their autosomal homologs the utrophins, β-DG, and α1-Syn were localized in astrocyte end feet. In Dp71-null mice, the levels of β-DG and α1-Syn were lower and utrophin expression did not change. The location of the DAPC in astrocytic end feet suggests that it could be involved in hypothalamic osmosensitivity, which adapts the osmotic response. The altered composition of the DAPC in Dp71-null mice could thus explain why these mice manifest an hypo-osmolar status. In the SON and PVN neurons of Dp71-null mice, nNOS expression and activity were increased. Although we previously established that DP140 is expressed de novo in these neurons, the DAPC remained incomplete due to the low levels of β-DG and α1-Syn produced in these cells. Our data reveal the importance of DP71 for the constitution of a functional DAPC in the hypothalamus. Such DAPC disorganization may lead to modification of the microenvironment of the SON and PVN neurons and thus may result in a perturbed osmoregulation.
Collapse
Affiliation(s)
- Roza Benabdesselam
- Unité de Recherches, Faculté des Sciences Biologiques/UMMTO, BP 17, Tizi-Ouzou, Algeria
| | | | | | | | | | | |
Collapse
|
39
|
Binder DK, Nagelhus EA, Ottersen OP. Aquaporin-4 and epilepsy. Glia 2012; 60:1203-14. [DOI: 10.1002/glia.22317] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Accepted: 02/09/2012] [Indexed: 12/17/2022]
|