1
|
Wu X, Li JR, Fu Y, Chen DY, Nie H, Tang ZP. From static to dynamic: live observation of the support system after ischemic stroke by two photon-excited fluorescence laser-scanning microscopy. Neural Regen Res 2023; 18:2093-2107. [PMID: 37056116 PMCID: PMC10328295 DOI: 10.4103/1673-5374.369099] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/21/2022] [Accepted: 01/13/2023] [Indexed: 02/17/2023] Open
Abstract
Ischemic stroke is one of the most common causes of mortality and disability worldwide. However, treatment efficacy and the progress of research remain unsatisfactory. As the critical support system and essential components in neurovascular units, glial cells and blood vessels (including the blood-brain barrier) together maintain an optimal microenvironment for neuronal function. They provide nutrients, regulate neuronal excitability, and prevent harmful substances from entering brain tissue. The highly dynamic networks of this support system play an essential role in ischemic stroke through processes including brain homeostasis, supporting neuronal function, and reacting to injuries. However, most studies have focused on postmortem animals, which inevitably lack critical information about the dynamic changes that occur after ischemic stroke. Therefore, a high-precision technique for research in living animals is urgently needed. Two-photon fluorescence laser-scanning microscopy is a powerful imaging technique that can facilitate live imaging at high spatiotemporal resolutions. Two-photon fluorescence laser-scanning microscopy can provide images of the whole-cortex vascular 3D structure, information on multicellular component interactions, and provide images of structure and function in the cranial window. This technique shifts the existing research paradigm from static to dynamic, from flat to stereoscopic, and from single-cell function to multicellular intercommunication, thus providing direct and reliable evidence to identify the pathophysiological mechanisms following ischemic stroke in an intact brain. In this review, we discuss exciting findings from research on the support system after ischemic stroke using two-photon fluorescence laser-scanning microscopy, highlighting the importance of dynamic observations of cellular behavior and interactions in the networks of the brain's support systems. We show the excellent application prospects and advantages of two-photon fluorescence laser-scanning microscopy and predict future research developments and directions in the study of ischemic stroke.
Collapse
Affiliation(s)
- Xuan Wu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Jia-Rui Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yu Fu
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Dan-Yang Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Hao Nie
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Zhou-Ping Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
2
|
Yi T, Gao P, Hou M, Lv H, Huang M, Gao S, He J, Yang D, Chen W, Zhu T, Yu C, Liu F, Yin H, Jin S. The mechanisms underlying the actions of Xuefu Zhuyu decoction pretreatment against neurological deficits after ischemic stroke in mice: The mediation of glymphatic function by aquaporin-4 and its anchoring proteins. Front Pharmacol 2022; 13:1053253. [PMID: 36582539 PMCID: PMC9792381 DOI: 10.3389/fphar.2022.1053253] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 11/30/2022] [Indexed: 12/15/2022] Open
Abstract
Ischemic stroke (IS) has been associated with an impairment in glymphatic function. Xuefu Zhuyu Decoction (XFZYD) is widely used in the prevention and treatment of ischemic stroke. We hypothesized that Xuefu Zhuyu decoction pretreatment could attenuate early neurological deficits after ischemic stroke by enhancing the function of the glymphatic system. To prove our hypothesis, we carried out temporary middle cerebral artery occlusion and reperfusion surgery on C57BL/6 mice and then measured neurological score, infarct size and performed hematoxylin-eosin staining to assess stroke outcomes after 24 h of reperfusion. Subsequently, we injected fluorescent tracers in to the cisterna magna and evaluated tracer distribution in coronal brain sections. The polarization of aquaporin-4 (AQP4), colocalization of aquaporin-4, α-dystroglycan, β-dystroglycan and agrin were determined by immunofluorescence. Our research showed that pretreatment with Xuefu Zhuyu decoction significantly alleviated neurological scores, neurological deficits and pathological abnormalities in a mouse model of ischemic stroke. Importantly, Xuefu Zhuyu decoction pretreatment enhanced cerebrospinal fluid influx, protected aquaporin-4 depolarization and promoted the colocalization of aquaporin-4 with its anchoring proteins in the brain. Our findings highlight novel mechanisms underlying the neuroprotective effect of Xuefu Zhuyu decoction pretreatment on ischemic stroke-induced brain damage through the glymphatic system. Xuefu Zhuyu decoction pretreatment may offer a promising approach to slow the onset and progression of ischemic stroke.
Collapse
Affiliation(s)
- Ting Yi
- School of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ping Gao
- School of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Meng Hou
- Department of Neurology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Huan Lv
- Department of Neurology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mengyuan Huang
- Department of Neurology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shanshan Gao
- Department of Neurology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinrong He
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dongdong Yang
- Department of Neurology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Weiyin Chen
- Department of Neurology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tianmin Zhu
- School of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chang Yu
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fuyou Liu
- Department of Neurology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Haiyan Yin
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China,*Correspondence: Haiyan Yin, ; Shuoguo Jin,
| | - Shuoguo Jin
- Department of Neurology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China,*Correspondence: Haiyan Yin, ; Shuoguo Jin,
| |
Collapse
|
3
|
The Water Transport System in Astrocytes–Aquaporins. Cells 2022; 11:cells11162564. [PMID: 36010640 PMCID: PMC9406552 DOI: 10.3390/cells11162564] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/26/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022] Open
Abstract
Highlights (AQPs) are transmembrane proteins responsible for fast water movement across cell membranes, including those of astrocytes. The expression and subcellular localization of AQPs in astrocytes are highly dynamic under physiological and pathological conditions. Besides their primary function in water homeostasis, AQPs participate in many ancillary functions including glutamate clearance in tripartite synapses and cell migration.
Abstract Astrocytes have distinctive morphological and functional characteristics, and are found throughout the central nervous system. Astrocytes are now known to be far more than just housekeeping cells in the brain. Their functions include contributing to the formation of the blood–brain barrier, physically and metabolically supporting and communicating with neurons, regulating the formation and functions of synapses, and maintaining water homeostasis and the microenvironment in the brain. Aquaporins (AQPs) are transmembrane proteins responsible for fast water movement across cell membranes. Various subtypes of AQPs (AQP1, AQP3, AQP4, AQP5, AQP8 and AQP9) have been reported to be expressed in astrocytes, and the expressions and subcellular localizations of AQPs in astrocytes are highly correlated with both their physiological and pathophysiological functions. This review describes and summarizes the recent advances in our understanding of astrocytes and AQPs in regard to controlling water homeostasis in the brain. Findings regarding the features of different AQP subtypes, such as their expression, subcellular localization, physiological functions, and the pathophysiological roles of astrocytes are presented, with brain edema and glioma serving as two representative AQP-associated pathological conditions. The aim is to provide a better insight into the elaborate “water distribution” system in cells, exemplified by astrocytes, under normal and pathological conditions.
Collapse
|
4
|
Yu YH, Kim SW, Kang J, Song Y, Im H, Kim SJ, Yoo DY, Lee MR, Park DK, Oh JS, Kim DS. Phosphodiesterase-5 Inhibitor Attenuates Anxious Phenotypes and Movement Disorder Induced by Mild Ischemic Stroke in Rats. J Korean Neurosurg Soc 2022; 65:665-679. [PMID: 35430790 PMCID: PMC9452378 DOI: 10.3340/jkns.2021.0101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 02/03/2022] [Indexed: 11/27/2022] Open
Abstract
Objective Patients with mild ischemic stroke experience various sequela and residual symptoms, such as anxious behavior and deficits in movement. Few approaches have been proved to be effective and safe therapeutic approaches for patients with mild ischemic stroke by acute stroke. Sildenafil (SIL), a phosphodiesterase-5 inhibitor (PDE5i), is a known remedy for neurodegenerative disorders and vascular dementia through its angiogenesis and neurogenesis effects. In this study, we investigated the efficacy of PDE5i in the emotional and behavioral abnormalities in rats with mild ischemic stroke.
Methods We divided the rats into four groups as follows (n=20, respectively) : group 1, naïve; group 2, middle cerebral artery occlusion (MCAo30); group 3, MCAo30+SIL-pre; and group 4, MCAo30+SIL-post. In the case of drug administration groups, single dose of PDE5i (sildenafil citrate, 20 mg/kg) was given at 30-minute before and after reperfusion of MCAo in rats. After surgery, we investigated and confirmed the therapeutic effect of sildenafil on histology, immunofluorescence, behavioral assays and neural oscillations.
Results Sildenafil alleviated a neuronal loss and reduced the infarction volume. And results of behavior task and immunofluorescence shown possibility that anti-inflammation process and improve motor deficits sildenafil treatment after mild ischemic stroke. Furthermore, sildenafil treatment attenuated the alteration of theta-frequency rhythm in the CA1 region of the hippocampus, a known neural oscillatory marker for anxiety disorder in rodents, induced by mild ischemic stroke.
Conclusion PDE5i as effective therapeutic agents for anxiety and movement disorders and provide robust preclinical evidence to support the development and use of PDE5i for the treatment of mild ischemic stroke residual disorders.
Collapse
|
5
|
Sylvain NJ, Salman MM, Pushie MJ, Hou H, Meher V, Herlo R, Peeling L, Kelly ME. The effects of trifluoperazine on brain edema, aquaporin-4 expression and metabolic markers during the acute phase of stroke using photothrombotic mouse model. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2021; 1863:183573. [PMID: 33561476 DOI: 10.1016/j.bbamem.2021.183573] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 01/23/2021] [Accepted: 01/29/2021] [Indexed: 12/22/2022]
Abstract
Stroke is the second leading cause of death and the third leading cause of disability globally. Edema is a hallmark of stroke resulting from dysregulation of water homeostasis in the central nervous system (CNS) and plays the major role in stroke-associated morbidity and mortality. The overlap between cellular and vasogenic edema makes treating this condition complicated, and to date, there is no pathogenically oriented drug treatment for edema. Water balance in the brain is tightly regulated, primarily by aquaporin 4 (AQP4) channels, which are mainly expressed in perivascular astrocytic end-feet. Targeting AQP4 could be a useful therapeutic approach for treating brain edema; however, there is no approved drug for stroke treatment that can directly block AQP4. In this study, we demonstrate that the FDA-approved drug trifluoperazine (TFP) effectively reduces cerebral edema during the early acute phase in post-stroke mice using a photothrombotic stroke model. This effect was combined with an inhibition of AQP4 expression at gene and protein levels. Importantly, TFP does not appear to induce any deleterious changes on brain electrolytes or metabolic markers, including total protein or lipid levels. Our results support a possible role for TFP in providing a beneficial extra-osmotic effect on brain energy metabolism, as indicated by the increase of glycogen levels. We propose that targeting AQP4-mediated brain edema using TFP is a viable therapeutic strategy during the early and acute phase of stroke that can be further investigated during later stages to help in developing novel CNS edema therapies.
Collapse
Affiliation(s)
- Nicole J Sylvain
- Division of Neurosurgery, Department of Surgery, College of Medicine, University of Saskatchewan, Canada
| | - Mootaz M Salman
- Medical Sciences Division, Department of Physiology, Anatomy and Genetics, Oxford University, South Parks Road, Oxford OX1 3QX, UK.
| | - M Jake Pushie
- Division of Neurosurgery, Department of Surgery, College of Medicine, University of Saskatchewan, Canada
| | - Huishu Hou
- Division of Neurosurgery, Department of Surgery, College of Medicine, University of Saskatchewan, Canada
| | - Vedashree Meher
- Department of Anatomy and Cell Biology, College of Medicine University of Saskatchewan, Canada
| | - Rasmus Herlo
- Neurotechnology Center, Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Lissa Peeling
- Division of Neurosurgery, Department of Surgery, College of Medicine, University of Saskatchewan, Canada
| | - Michael E Kelly
- Division of Neurosurgery, Department of Surgery, College of Medicine, University of Saskatchewan, Canada
| |
Collapse
|
6
|
Steliga A, Kowiański P, Czuba E, Waśkow M, Moryś J, Lietzau G. Neurovascular Unit as a Source of Ischemic Stroke Biomarkers-Limitations of Experimental Studies and Perspectives for Clinical Application. Transl Stroke Res 2020; 11:553-579. [PMID: 31701356 PMCID: PMC7340668 DOI: 10.1007/s12975-019-00744-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 09/23/2019] [Accepted: 09/24/2019] [Indexed: 01/13/2023]
Abstract
Cerebral stroke, which is one of the most frequent causes of mortality and leading cause of disability in developed countries, often leads to devastating and irreversible brain damage. Neurological and neuroradiological diagnosis of stroke, especially in its acute phase, is frequently uncertain or inconclusive. This results in difficulties in identification of patients with poor prognosis or being at high risk for complications. It also makes difficult identification of these stroke patients who could benefit from more aggressive therapies. In contrary to the cardiovascular disease, no single biomarker is available for the ischemic stroke, addressing the abovementioned issues. This justifies the need for identifying of effective diagnostic measures characterized by high specificity and sensitivity. One of the promising avenues in this area is studies on the panels of biomarkers characteristic for processes which occur in different types and phases of ischemic stroke and represent all morphological constituents of the brains' neurovascular unit (NVU). In this review, we present the current state of knowledge concerning already-used or potentially applicable biomarkers of the ischemic stroke. We also discuss the perspectives for identification of biomarkers representative for different types and phases of the ischemic stroke, as well as for different constituents of NVU, which concentration levels correlate with extent of brain damage and patients' neurological status. Finally, a critical analysis of perspectives on further improvement of the ischemic stroke diagnosis is presented.
Collapse
Affiliation(s)
- Aleksandra Steliga
- Faculty of Health Sciences, Pomeranian University of Slupsk, 64 Bohaterów Westerplatte St., 76-200, Slupsk, Poland
| | - Przemysław Kowiański
- Faculty of Health Sciences, Pomeranian University of Slupsk, 64 Bohaterów Westerplatte St., 76-200, Slupsk, Poland.
- Department of Anatomy and Neurobiology, Medical University of Gdansk, 1 Debinki St., 80-211, Gdansk, Poland.
| | - Ewelina Czuba
- Department of Anatomy and Neurobiology, Medical University of Gdansk, 1 Debinki St., 80-211, Gdansk, Poland
| | - Monika Waśkow
- Faculty of Health Sciences, Pomeranian University of Slupsk, 64 Bohaterów Westerplatte St., 76-200, Slupsk, Poland
| | - Janusz Moryś
- Department of Anatomy and Neurobiology, Medical University of Gdansk, 1 Debinki St., 80-211, Gdansk, Poland
| | - Grażyna Lietzau
- Department of Anatomy and Neurobiology, Medical University of Gdansk, 1 Debinki St., 80-211, Gdansk, Poland
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
7
|
Andjelkovic AV, Stamatovic SM, Phillips CM, Martinez-Revollar G, Keep RF. Modeling blood-brain barrier pathology in cerebrovascular disease in vitro: current and future paradigms. Fluids Barriers CNS 2020; 17:44. [PMID: 32677965 PMCID: PMC7367394 DOI: 10.1186/s12987-020-00202-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/15/2020] [Indexed: 12/16/2022] Open
Abstract
The complexity of the blood-brain barrier (BBB) and neurovascular unit (NVU) was and still is a challenge to bridge. A highly selective, restrictive and dynamic barrier, formed at the interface of blood and brain, the BBB is a "gatekeeper" and guardian of brain homeostasis and it also acts as a "sensor" of pathological events in blood and brain. The majority of brain and cerebrovascular pathologies are associated with BBB dysfunction, where changes at the BBB can lead to or support disease development. Thus, an ultimate goal of BBB research is to develop competent and highly translational models to understand mechanisms of BBB/NVU pathology and enable discovery and development of therapeutic strategies to improve vascular health and for the efficient delivery of drugs. This review article focuses on the progress being made to model BBB injury in cerebrovascular diseases in vitro.
Collapse
Affiliation(s)
- Anuska V Andjelkovic
- Department of Pathology, University of Michigan Medical School, 7520 MSRB I, 1150 West Medical Center Dr, Ann Arbor, MI, 48109-5602, USA.
| | - Svetlana M Stamatovic
- Department of Pathology, University of Michigan Medical School, 7520 MSRB I, 1150 West Medical Center Dr, Ann Arbor, MI, 48109-5602, USA
| | - Chelsea M Phillips
- Graduate Program in Neuroscience, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Gabriela Martinez-Revollar
- Department of Pathology, University of Michigan Medical School, 7520 MSRB I, 1150 West Medical Center Dr, Ann Arbor, MI, 48109-5602, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Molecular Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
8
|
Maroli N, Kalagatur NK, Bhasuran B, Jayakrishnan A, Manoharan RR, Kolandaivel P, Natarajan J, Kadirvelu K. Molecular Mechanism of T-2 Toxin-Induced Cerebral Edema by Aquaporin-4 Blocking and Permeation. J Chem Inf Model 2019; 59:4942-4958. [DOI: 10.1021/acs.jcim.9b00711] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
| | | | | | | | | | | | - Jeyakumar Natarajan
- Data Mining and Text Mining Laboratory, Department of Bioinformatics, Bharathiar University, Coimbatore 641046, Tamil Nadu, India
| | | |
Collapse
|
9
|
Stamatovic SM, Phillips CM, Martinez-Revollar G, Keep RF, Andjelkovic AV. Involvement of Epigenetic Mechanisms and Non-coding RNAs in Blood-Brain Barrier and Neurovascular Unit Injury and Recovery After Stroke. Front Neurosci 2019; 13:864. [PMID: 31543756 PMCID: PMC6732937 DOI: 10.3389/fnins.2019.00864] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 07/31/2019] [Indexed: 12/13/2022] Open
Abstract
Cessation of blood flow leads to a complex cascade of pathophysiological events at the blood-vascular-parenchymal interface which evolves over time and space, and results in damage to neural cells and edema formation. Cerebral ischemic injury evokes a profound and deleterious upregulation in inflammation and triggers multiple cell death pathways, but it also induces a series of the events associated with regenerative responses, including vascular remodeling, angiogenesis, and neurogenesis. Emerging evidence suggests that epigenetic reprograming could play a pivotal role in ongoing post-stroke neurovascular unit (NVU) changes and recovery. This review summarizes current knowledge about post-stroke recovery processes at the NVU, as well as epigenetic mechanisms and modifiers (e.g., DNA methylation, histone modifying enzymes and microRNAs) associated with stroke injury, and NVU repair. It also discusses novel drug targets and therapeutic strategies for enhancing post-stroke recovery.
Collapse
Affiliation(s)
- Svetlana M. Stamatovic
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Chelsea M. Phillips
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI, United States
| | | | - Richard F. Keep
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Molecular Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Anuska V. Andjelkovic
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
10
|
Morris DC, Zhang ZG, Chopp M. Thymosin β4 for the treatment of acute stroke: neurorestorative or neuroprotective? Expert Opin Biol Ther 2019; 18:149-158. [PMID: 30063858 DOI: 10.1080/14712598.2018.1484100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Thymosin β4 (Tβ4) is a 5K peptide which influences cellular migration by inhibiting organization of the actin-cytoskeleton. Treatment of acute stroke presently involves use of rt-PA and/or endovascular treatment with thrombectomy, both of which have time limitations. Therefore, development of a treatment beyond these times is necessary as most stroke patients present beyond these time limits. A drug which could be administered within 24 h from symptom onset would provide substantial benefit. AREAS COVERED This review summarizes the data and results of two in-vivo studies testing Tβ4 in an embolic stroke model of young and aged rats. In addition, we describe in-vitro investigations of the neurorestorative and neuroprotective properties of Tβ4 in a variety of neuroprogenitor and oligoprogenitor cell models. EXPERT OPINION Tβ4 acts as a neurorestorative agent when employed in a young male rat model of embolic stroke while in an aged model it acts a neuroprotectant. However evaluation of Tβ4 as a treatment of stroke requires further preclinical evaluation in females and in males and females with comorbidities such as, hypertension and diabetes in models of embolic stroke to further define the mechanism of action and potential as a treatment of stroke in humans.
Collapse
Affiliation(s)
- Daniel C Morris
- a Department of Emergency Medicine , Henry Ford Health Systems , Detroit , MI , USA
| | - Zheng G Zhang
- b Department of Neurology , Henry Ford Health Systems , Detroit , MI , USA
| | - Michael Chopp
- b Department of Neurology , Henry Ford Health Systems , Detroit , MI , USA.,c Department of Physics , Oakland University , Rochester , MI , USA
| |
Collapse
|
11
|
Clément T, Rodriguez-Grande B, Badaut J. Aquaporins in brain edema. J Neurosci Res 2018; 98:9-18. [DOI: 10.1002/jnr.24354] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Accepted: 10/15/2018] [Indexed: 01/08/2023]
Affiliation(s)
- Tifenn Clément
- CNRS UMR 5287, INCIA, University of Bordeaux; Bordeaux France
| | | | - Jérôme Badaut
- CNRS UMR 5287, INCIA, University of Bordeaux; Bordeaux France
- Department of Basic Science; Loma Linda University School of Medicine; Loma Linda California
| |
Collapse
|
12
|
Woo J, Kim JE, Im JJ, Lee J, Jeong HS, Park S, Jung SY, An H, Yoon S, Lim SM, Lee S, Ma J, Shin EY, Han YE, Kim B, Lee EH, Feng L, Chun H, Yoon BE, Kang I, Dager SR, Lyoo IK, Lee CJ. Astrocytic water channel aquaporin-4 modulates brain plasticity in both mice and humans: a potential gliogenetic mechanism underlying language-associated learning. Mol Psychiatry 2018; 23:1021-1030. [PMID: 29565042 DOI: 10.1038/mp.2017.113] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 03/21/2017] [Accepted: 04/17/2017] [Indexed: 01/04/2023]
Abstract
The role of astrocytes in brain plasticity has not been extensively studied compared with that of neurons. Here we adopted integrative translational and reverse-translational approaches to explore the role of an astrocyte-specific major water channel in the brain, aquaporin-4 (AQP4), in brain plasticity and learning. We initially identified the most prevalent genetic variant of AQP4 (single nucleotide polymorphism of rs162008 with C or T variation, which has a minor allele frequency of 0.21) from a human database (n=60 706) and examined its functionality in modulating the expression level of AQP4 in an in vitro luciferase reporter assay. In the following experiments, AQP4 knock-down in mice not only impaired hippocampal volumetric plasticity after exposure to enriched environment but also caused loss of long-term potentiation after theta-burst stimulation. In humans, there was a cross-sectional association of rs162008 with gray matter (GM) volume variation in cortices, including the vicinity of the Perisylvian heteromodal language area (Sample 1, n=650). GM volume variation in these brain regions was positively associated with the semantic verbal fluency. In a prospective follow-up study (Sample 2, n=45), the effects of an intensive 5-week foreign language (English) learning experience on regional GM volume increase were modulated by this AQP4 variant, which was also associated with verbal learning capacity change. We then delineated in mice mechanisms that included AQP4-dependent transient astrocytic volume changes and astrocytic structural elaboration. We believe our study provides the first integrative evidence for a gliogenetic basis that involves AQP4, underlying language-associated brain plasticity.
Collapse
Affiliation(s)
- J Woo
- Center for Neural Science and Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea.,Neuroscience Program, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - J E Kim
- Department of Brain and Cognitive Sciences, Scranton College, Ewha Womans University, Seoul, Republic of Korea.,Ewha Brain Institute, Ewha Womans University, Seoul, Republic of Korea
| | - J J Im
- Ewha Brain Institute, Ewha Womans University, Seoul, Republic of Korea.,Interdisciplinary Program in Neuroscience, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea
| | - J Lee
- Center for Neural Science and Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - H S Jeong
- Department of Radiology, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - S Park
- Center for Neural Science and Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - S-Y Jung
- Center for Neural Science and Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea.,Neuroscience Program, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - H An
- Center for Neural Science and Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea.,Department of Radiology, University of Washington, Seattle, WA, USA.,Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - S Yoon
- Ewha Brain Institute, Ewha Womans University, Seoul, Republic of Korea
| | - S M Lim
- Department of Radiology, College of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - S Lee
- Ewha Brain Institute, Ewha Womans University, Seoul, Republic of Korea.,Interdisciplinary Program in Neuroscience, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea
| | - J Ma
- Ewha Brain Institute, Ewha Womans University, Seoul, Republic of Korea.,Interdisciplinary Program in Neuroscience, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea
| | - E Y Shin
- Department of Brain and Cognitive Sciences, Scranton College, Ewha Womans University, Seoul, Republic of Korea.,Ewha Brain Institute, Ewha Womans University, Seoul, Republic of Korea
| | - Y-E Han
- Center for Neural Science and Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea.,Neuroscience Program, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - B Kim
- Ewha Brain Institute, Ewha Womans University, Seoul, Republic of Korea.,Interdisciplinary Program in Neuroscience, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea
| | - E H Lee
- Green Cross Laboratories, Yongin, Republic of Korea
| | - L Feng
- Center for Neural Science and Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - H Chun
- Center for Neural Science and Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - B-E Yoon
- Center for Neural Science and Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea.,Department of Nanobiomedical Science, Dankook University, Cheonan, Republic of Korea
| | - I Kang
- Department of Brain and Cognitive Sciences, Scranton College, Ewha Womans University, Seoul, Republic of Korea.,Ewha Brain Institute, Ewha Womans University, Seoul, Republic of Korea
| | - S R Dager
- Department of Radiology, University of Washington, Seattle, WA, USA.,Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - I K Lyoo
- Department of Brain and Cognitive Sciences, Scranton College, Ewha Womans University, Seoul, Republic of Korea.,Ewha Brain Institute, Ewha Womans University, Seoul, Republic of Korea.,Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - C J Lee
- Center for Neural Science and Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea.,Neuroscience Program, University of Science and Technology (UST), Daejeon, Republic of Korea.,KU-KIST, Graduate School of Convergence Technology, Korea University, Seoul, Republic of Korea
| |
Collapse
|
13
|
Szöllősi D, Tóth L, Kálmán M. Postmortem immunohistochemical alterations following cerebral lesions: A possible pathohistological importance of the β-dystroglycan immunoreactivity. Neuropathology 2017; 38:207-217. [PMID: 29266551 DOI: 10.1111/neup.12447] [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] [Received: 08/01/2017] [Revised: 11/06/2017] [Accepted: 11/07/2017] [Indexed: 12/29/2022]
Abstract
The frequency of cerebrovascular injuries raises the importance of their immunohistological investigation in postmortem materials. Most injuries involve the impairment of the blood-brain barrier. The barrier is maintained by the glio-vascular connections which break up following injuries. Some immunohistochemical alterations may refer to the impairment of the gliovascular connections. Laminin and the components of the dystroglycan complex show characteristic immunohistochemical alterations following various experimental injuries (stab wound, cryogenic lesion, arterial occlusions): immunoreactivity of β-dystroglycan, α-dystrobrevin and aquaporin 4 disappeared while that of utrophin and laminin appeared along the vessels, whereas α-syntrophin visualized the reactive astrocytes but not the resting ones. The aims of the present study were to investigate whether these post-lesion alterations: (i) are reproducible with immersive fixation, which is used in postmortem histology; (ii) are resistant to a postmortem delay before fixation; and (iii) are to be attributed to a direct effect of the lesion, or are mediated by processes occurring only in the living brain. Three models were investigated: (i) following lesions, some brains were fixed by transcardial perfusion, others by immersion; (ii) following lesions, the animals were decapitated and stored at room temperature for 8 or 16 h before fixation; and (iii) the lesions were performed after decapitation. Cryogenic lesions were performed by applying a dry ice cooled copper rod to the brain surface of ketamine-xylazine anesthetized rats. The immunohistochemical reactions were performed on free-floating sections cut with vibratome. Both immunoperoxidase and immunofluorescence methods were used. The fixation method - perfusive or immersive - did not change the post-lesion phenomena investigated. The postmortem delay did not influence the β-dystroglycan immunoreactivity, that is its lack delineated the area of the lesion. However, in the case of the other substances, various lengths of postmortem delay rendered the immunohistochemistry uninterpretable. The results suggest β-dystroglycan immunostaining could be applied in the neuropathology to detect cerebrovascular impairments.
Collapse
Affiliation(s)
- Dávid Szöllősi
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - László Tóth
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Mihály Kálmán
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
14
|
Bi C, Tham DKL, Perronnet C, Joshi B, Nabi IR, Moukhles H. The Oxidative Stress-Induced Increase in the Membrane Expression of the Water-Permeable Channel Aquaporin-4 in Astrocytes Is Regulated by Caveolin-1 Phosphorylation. Front Cell Neurosci 2017; 11:412. [PMID: 29326556 PMCID: PMC5742350 DOI: 10.3389/fncel.2017.00412] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 12/08/2017] [Indexed: 01/14/2023] Open
Abstract
The reperfusion of ischemic brain tissue following a cerebral stroke causes oxidative stress, and leads to the generation of reactive oxygen species (ROS). Apart from inflicting oxidative damage, the latter may also trigger the upregulation of aquaporin 4 (AQP4), a water-permeable channel expressed by astroglial cells of the blood-brain barrier (BBB), and contribute to edema formation, the severity of which is known to be the primary determinant of mortality and morbidity. The mechanism through which this occurs remains unknown. In the present study, we have attempted to address this question using primary astrocyte cultures treated with hydrogen peroxide (H2O2) as a model system. First, we showed that H2O2 induces a significant increase in AQP4 protein levels and that this is inhibited by the antioxidant N-acetylcysteine (NAC). Second, we demonstrated using cell surface biotinylation that H2O2 increases AQP4 cell-surface expression independently of it's increased synthesis. In parallel, we found that caveolin-1 (Cav1) is phosphorylated in response to H2O2 and that this is reversed by the Src kinase inhibitor 4-Amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4-d]pyrimidine (PP2). PP2 also abrogated the H2O2-induced increase in AQP4 surface levels, suggesting that the phosphorylation of tyrosine-14 of Cav1 regulates this process. We further showed that dominant-negative Y14F and phosphomimetic Y14D mutants caused a decrease and increase in AQP4 membrane expression respectively, and that the knockdown of Cav1 inhibits the increase in AQP4 cell-surface, expression following H2O2 treatment. Together, these findings suggest that oxidative stress-induced Cav1 phosphorylation modulates AQP4 subcellular distribution and therefore may indirectly regulate AQP4-mediated water transport.
Collapse
Affiliation(s)
- Chongshan Bi
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Daniel K L Tham
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Caroline Perronnet
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Bharat Joshi
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Ivan R Nabi
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Hakima Moukhles
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
15
|
Morris DC, Cheung WL, Loi R, Zhang T, Lu M, Zhang ZG, Chopp M. Thymosin β4 for the treatment of acute stroke in aged rats. Neurosci Lett 2017; 659:7-13. [PMID: 28864242 DOI: 10.1016/j.neulet.2017.08.064] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 08/10/2017] [Accepted: 08/28/2017] [Indexed: 12/21/2022]
Abstract
Thymosin β4 (Tβ4) is a 5K peptide which influences cellular migration by inhibiting organization of the actin-cytoskeleton. Tβ4 has neurorestorative properties and is a potential candidate for the treatment of sub-acute stroke. Previous research demonstrated that Tβ4 improved neurological outcome in a young (3 months) rat model of embolic stroke. We hypothesized that Tβ4 would improve neurological outcome in an aged rat model of embolic stroke when administered 24h after embolic stroke. Aged Male Wistar rats (Charles River, France 18-21 months) were subjected to embolic middle cerebral artery occlusion (MCAo). Rats were randomized to receive Tβ4 (12mg/kg, RegeneRx Biopharmaceuticals, Inc.) or control 24h after MCAo and then every 3days for 4 additional doses. The dose of 12mg/kg was the maximal dose of Tβ4 that showed functional improvement in a young rat model of embolic stroke. Functional tests (adhesive-removal test (ART), foot fault test (FFT) and the modified Neurological Severity Score (mNSS)) were performed weekly. The rats were sacrificed 56days after MCAo and lesion volumes were measured. Immunohistochemical analysis for oligodendrogenesis, myelination and gliosis was also performed. Twenty-three rats were included in the study: control group (n=12) and Tβ4 group (n=11). After randomization, there were three deaths in both the control and Tβ4 groups. The Tβ4 treatment reduced infarct volume by more than 50% (12.8%±9.3%, mean±SE, p<0.05) compared to the control group (26.0%±4.3%). However, Tβ4 did not show improvement in functional outcome compared to control. There was no significant increase in oligodendrogenesis, myelination and gliosis between control and treatment with Tβ4, however, we unexpectedly observed that overall (control and Tβ4 groups) astrocytic gliosis as measured by GFAP immunoreactivity was significantly inversely correlated with neurological outcome measured using the modified Neurological Severity Score (mNSS) (p<0.01), suggesting that greater gliosis may be related to improvement of neurological outcome in aged rats. In summary, Tβ4 treatment of stroke aged rats significantly reduces infarct volume compared to vehicle treated stroke, however, Tβ4 treatment did not show improvement in functional outcome, myelination or gliosis when compared to control. GFAP staining was significantly inversely correlated to improvement in the mNSS, suggesting that gliosis in the aged rat may be of benefit in improvement of functional outcome.
Collapse
Affiliation(s)
- Daniel C Morris
- Department of Emergency Medicine, Henry Ford Health System, Detroit, MI 48202, USA.
| | - Wing Lee Cheung
- Department of Emergency Medicine, Henry Ford Health System, Detroit, MI 48202, USA.
| | - Richard Loi
- Department of Emergency Medicine, Henry Ford Health System, Detroit, MI 48202, USA
| | - Talan Zhang
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI 48202, USA.
| | - Mei Lu
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI 48202, USA.
| | - Zheng G Zhang
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA.
| | - Michael Chopp
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; Department of Physics, Oakland University, Rochester, MI 48309, USA.
| |
Collapse
|
16
|
Osmotherapy With Hypertonic Saline Attenuates Global Cerebral Edema Following Experimental Cardiac Arrest via Perivascular Pool of Aquaporin-4. Crit Care Med 2017; 44:e702-10. [PMID: 27035238 DOI: 10.1097/ccm.0000000000001671] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVES We tested the hypothesis that osmotherapy with hypertonic saline attenuates cerebral edema following experimental cardiac arrest and cardiopulmonary resuscitation by exerting its effect via the perivascular pool of aquaporin-4. We used mice with targeted disruption of the gene encoding α-syntrophin (α-Syn) that demonstrate diminished perivascular aquaporin-4 pool but retain the non-endfoot and ependymal pools. DESIGN Laboratory animal study. SETTING University animal research laboratory. INTERVENTIONS Isoflurane-anesthetized adult male wild-type C57B/6 or α-Syn mice were subjected to cardiac arrest/cardiopulmonary resuscitation and treated with either a continuous IV infusion of 0.9% saline or various concentrations of hypertonic saline. Serum osmolality, regional brain water content, blood-brain barrier disruption, and aquaporin-4 protein expression were determined at 24 hours after cardiac arrest/cardiopulmonary resuscitation. MEASUREMENTS AND MAIN RESULTS Hypertonic saline (7.5%) treatment significantly attenuated water content in the caudoputamen complex and cortex compared with 0.9% saline treatment in wild-type mice subjected to cardiac arrest/cardiopulmonary resuscitation. In contrast, in α-Syn mice subjected to cardiac arrest/cardiopulmonary resuscitation, 7.5% hypertonic saline treatment did not attenuate water content. Treatment with 7.5% hypertonic saline attenuated blood-brain barrier disruption at 24 hours following cardiac arrest/cardiopulmonary resuscitation in wild-type mice but not in α-Syn mice. Total aquaporin-4 protein expression was not different between 0.9% saline and hypertonic saline-treated wild-type mice. CONCLUSIONS Following experimental cardiac arrest/cardiopulmonary resuscitation: 1) continuous hypertonic saline therapy maintained to achieve serum osmolality of ≈ 350 mOsm/L is beneficial for the treatment of cerebral edema; 2) perivascular pool of aquaporin-4 plays a critical role in water egress from brain; and 3) hypertonic saline attenuates blood-brain barrier disruption via perivascular aquaporin-4 pool.
Collapse
|
17
|
Hubbard JA, Szu JI, Binder DK. The role of aquaporin-4 in synaptic plasticity, memory and disease. Brain Res Bull 2017; 136:118-129. [PMID: 28274814 DOI: 10.1016/j.brainresbull.2017.02.011] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 02/27/2017] [Accepted: 02/28/2017] [Indexed: 12/25/2022]
Abstract
Since the discovery of aquaporins, it has become clear that the various mammalian aquaporins play critical physiological roles in water and ion balance in multiple tissues. Aquaporin-4 (AQP4), the principal aquaporin expressed in the central nervous system (CNS, brain and spinal cord), has been shown to mediate CNS water homeostasis. In this review, we summarize new and exciting studies indicating that AQP4 also plays critical and unanticipated roles in synaptic plasticity and memory formation. Next, we consider the role of AQP4 in Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), Parkinson's disease (PD), multiple sclerosis (MS), neuromyelitis optica (NMO), epilepsy, traumatic brain injury (TBI), and stroke. Each of these conditions involves changes in AQP4 expression and/or distribution that may be functionally relevant to disease physiology. Insofar as AQP4 is exclusively expressed on astrocytes, these data provide new evidence of "astrocytopathy" in the etiology of diverse neurological diseases.
Collapse
Affiliation(s)
- Jacqueline A Hubbard
- Center for Glial-Neuronal Interactions, Division of Biomedical Sciences, School of Medicine, University of California, Riverside, United States
| | - Jenny I Szu
- Center for Glial-Neuronal Interactions, Division of Biomedical Sciences, School of Medicine, University of California, Riverside, United States
| | - Devin K Binder
- Center for Glial-Neuronal Interactions, Division of Biomedical Sciences, School of Medicine, University of California, Riverside, United States.
| |
Collapse
|
18
|
Venkat P, Chopp M, Chen J. New insights into coupling and uncoupling of cerebral blood flow and metabolism in the brain. Croat Med J 2017; 57:223-8. [PMID: 27374823 PMCID: PMC4937223 DOI: 10.3325/cmj.2016.57.223] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The brain has high metabolic and energy needs and requires continuous cerebral blood flow (CBF), which is facilitated by a tight coupling between neuronal activity, CBF, and metabolism. Upon neuronal activation, there is an increase in energy demand, which is then met by a hemodynamic response that increases CBF. Such regional CBF increase in response to neuronal activation is observed using neuroimaging techniques such as functional magnetic resonance imaging and positron emission tomography. The mechanisms and mediators (eg, nitric oxide, astrocytes, and ion channels) that regulate CBF-metabolism coupling have been extensively studied. The neurovascular unit is a conceptual model encompassing the anatomical and metabolic interactions between the neurons, vascular components, and glial cells in the brain. It is compromised under disease states such as stroke, diabetes, hypertension, dementias, and with aging, all of which trigger a cascade of inflammatory responses that exacerbate brain damage. Hence, tight regulation and maintenance of neurovascular coupling is central for brain homeostasis. This review article also discusses the waste clearance pathways in the brain such as the glymphatic system. The glymphatic system is a functional waste clearance pathway that removes metabolic wastes and neurotoxins from the brain along paravascular channels. Disruption of the glymphatic system burdens the brain with accumulating waste and has been reported in aging as well as several neurological diseases.
Collapse
Affiliation(s)
| | | | - Jieli Chen
- Jieli Chen, Senior Staff Investigator, Henry Ford Hospital, Neurology Research, E&R Building, 3091, Detroit, MI, 48202, USA,
| |
Collapse
|
19
|
Hirt L, Fukuda AM, Ambadipudi K, Rashid F, Binder D, Verkman A, Ashwal S, Obenaus A, Badaut J. Improved long-term outcome after transient cerebral ischemia in aquaporin-4 knockout mice. J Cereb Blood Flow Metab 2017; 37:277-290. [PMID: 26767580 PMCID: PMC5363745 DOI: 10.1177/0271678x15623290] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Revised: 10/23/2015] [Accepted: 10/26/2015] [Indexed: 01/07/2023]
Abstract
A hallmark of stroke is water accumulation (edema) resulting from dysregulation of osmotic homeostasis. Brain edema contributes to tissue demise and may lead to increased intracranial pressure and lethal herniation. Currently, there are only limited treatments to prevent edema formation following stroke. Aquaporin 4 (AQP4), a brain water channel, has become a focus of interest for therapeutic approaches targeting edema. At present, there are no pharmacological tools to block AQP4. The role of AQP4 in edema after brain injury remains unclear with conflicting results from studies using AQP4-/- mice and of AQP4 expression following stroke. Here, we studied AQP4 and its role in edema formation by testing AQP4-/- mice in a model of middle cerebral artery occlusion using novel quantitative MRI water content measurements, histology and behavioral changes as outcome measures. Absence of AQP4 was associated with decreased mortality and increased motor recovery 3 to 14 days after stroke. Behavioral improvement was associated with decreased lesion volume, neuronal cell death and neuroinflammation in AQP4-/- compared to wild type mice. Our data suggest that the lack of AQP4 confers an overall beneficial role at long term with improved neuronal survival and reduced neuroinflammation, but without a direct effect on edema formation.
Collapse
Affiliation(s)
- Lorenz Hirt
- Department of Clinical Neurosciences, Neurology Service, Centre Hospitalier Universitaire Vaudois and Lausanne University, Switzerland
| | - Andrew M Fukuda
- Department of Physiology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Kamalakar Ambadipudi
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Faisil Rashid
- Department of Physiology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Devin Binder
- Center for Glial-Neuronal Interactions, Division of Biomedical Sciences, University of California, Riverside, Riverside, CA, USA
| | - Alan Verkman
- Medicine and Physiology, Cardiovascular Research Institute, University of California San Francisco, CA, USA
| | - Stephen Ashwal
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Andre Obenaus
- Department of Physiology, Loma Linda University School of Medicine, Loma Linda, CA, USA.,Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, USA.,Center for Glial-Neuronal Interactions, Division of Biomedical Sciences, University of California, Riverside, Riverside, CA, USA
| | - Jerome Badaut
- Department of Physiology, Loma Linda University School of Medicine, Loma Linda, CA, USA .,Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, USA.,CNRS UMR5287, University of Bordeaux, Bordeaux, France
| |
Collapse
|
20
|
White matter damage and glymphatic dysfunction in a model of vascular dementia in rats with no prior vascular pathologies. Neurobiol Aging 2016; 50:96-106. [PMID: 27940353 DOI: 10.1016/j.neurobiolaging.2016.11.002] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 11/10/2016] [Accepted: 11/10/2016] [Indexed: 11/24/2022]
Abstract
We investigated cognitive function, axonal/white matter (WM) changes and glymphatic function of vascular dementia using a multiple microinfarction (MMI) model in retired breeder (RB) rats. The MMI model induces significant (p < 0.05) cognitive decline that worsens with age starting at 2 weeks, which persists until at least 6 weeks after MMI. RB rats subjected to MMI exhibit significant axonal/WM damage identified by decreased myelin thickness, oligodendrocyte progenitor cell numbers, axon density, synaptic protein expression in the cortex and striatum, cortical neuronal branching, and dendritic spine density in the cortex and hippocampus compared with age-matched controls. MMI evokes significant dilation of perivascular spaces as well as water channel dysfunction indicated by decreased Aquaporin-4 expression around blood vessels. MMI-induced glymphatic dysfunction with delayed cerebrospinal fluid penetration into the brain parenchyma via paravascular pathways as well as delayed waste clearance from the brain. The MMI model in RB rats decreases Aquaporin-4 and induces glymphatic dysfunction which may play an important role in MMI-induced axonal/WM damage and cognitive deficits.
Collapse
|
21
|
Kurisu K, Abumiya T, Nakamura H, Shimbo D, Shichinohe H, Nakayama N, Kazumata K, Shimizu H, Houkin K. Transarterial Regional Brain Hypothermia Inhibits Acute Aquaporin-4 Surge and Sequential Microvascular Events in Ischemia/Reperfusion Injury. Neurosurgery 2016; 79:125-34. [DOI: 10.1227/neu.0000000000001088] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
|
22
|
Wu X, Zhang JT, Li D, Zhou J, Yang J, Zheng HL, Chen JG, Wang F. Aquaporin-4 deficiency facilitates fear memory extinction in the hippocampus through excessive activation of extrasynaptic GluN2B-containing NMDA receptors. Neuropharmacology 2016; 112:124-134. [PMID: 27373674 DOI: 10.1016/j.neuropharm.2016.06.031] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Revised: 06/11/2016] [Accepted: 06/28/2016] [Indexed: 12/22/2022]
Abstract
Aquaporin-4 (AQP-4) is the predominant water channel in the brain and primarily expressed in astrocytes. Astrocytes have been generally believed to play important roles in regulating synaptic plasticity and information processing. A growing number of evidence shows that AQP-4 plays a potential role in the regulation of astrocyte function. However, little is known about the function of AQP-4 for synaptic plasticity in the hippocampus. Therefore, we evaluated long-term depression (LTD) in the hippocampus and the extinction of fear memory of AQP-4 knockout (KO) and wild-type (WT) mice. We found that AQP-4 deficiency facilitated fear memory extinction and NMDA receptors (NMDARs)-dependent LTD in the CA3-CA1 pathway. Furthermore, AQP-4 deficiency selectively increased GluN2B-NMDAR-mediated excitatory postsynaptic currents (EPSCs). The excessive activation of extrasynaptic GluN2B-NMDAR contributed to the facilitation of NMDAR-dependent LTD and enhancement of fear memory extinction in AQP-4 KO mice. Thus, it appears that AQP-4 may be a potential target for intervention in fear memory extinction. This article is part of the Special Issue entitled 'Ionotropic glutamate receptors'.
Collapse
Affiliation(s)
- Xin Wu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jie-Ting Zhang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Di Li
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jun Zhou
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jun Yang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hui-Ling Zheng
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jian-Guo Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; The Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, 430030, China; The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, 430030, China; Laboratory of Neuropsychiatric Diseases, The Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, 430030, China; The Collaborative Innovation Center for Brain Science, Wuhan, 430030, China
| | - Fang Wang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; The Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, 430030, China; The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, 430030, China; Laboratory of Neuropsychiatric Diseases, The Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, 430030, China; The Collaborative Innovation Center for Brain Science, Wuhan, 430030, China.
| |
Collapse
|
23
|
Hubbard JA, Hsu MS, Seldin MM, Binder DK. Expression of the Astrocyte Water Channel Aquaporin-4 in the Mouse Brain. ASN Neuro 2015; 7:7/5/1759091415605486. [PMID: 26489685 PMCID: PMC4623559 DOI: 10.1177/1759091415605486] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Aquaporin-4 (AQP4) is a bidirectional water channel that is found on astrocytes throughout the central nervous system. Expression is particularly high around areas in contact with cerebrospinal fluid, suggesting that AQP4 plays a role in fluid exchange between the cerebrospinal fluid compartments and the brain. Despite its significant role in the brain, the overall spatial and region-specific distribution of AQP4 has yet to be fully characterized. In this study, we used Western blotting and immunohistochemical techniques to characterize AQP4 expression and localization throughout the mouse brain. We observed AQP4 expression throughout the forebrain, subcortical areas, and brainstem. AQP4 protein levels were highest in the cerebellum with lower expression in the cortex and hippocampus. We found that AQP4 immunoreactivity was profuse on glial cells bordering ventricles, blood vessels, and subarachnoid space. Throughout the brain, AQP4 was expressed on astrocytic end-feet surrounding blood vessels but was also heterogeneously expressed in brain tissue parenchyma and neuropil, often with striking laminar specificity. In the cerebellum, we showed that AQP4 colocalized with the proteoglycan brevican, which is synthesized by and expressed on cerebellar astrocytes. Despite the high abundance of AQP4 in the cerebellum, its functional significance has yet to be investigated. Given the known role of AQP4 in synaptic plasticity in the hippocampus, the widespread and region-specific expression pattern of AQP4 suggests involvement not only in fluid balance and ion homeostasis but also local synaptic plasticity and function in distinct brain circuits.
Collapse
Affiliation(s)
- Jacqueline A Hubbard
- Center for Glial-Neuronal Interactions, Division of Biomedical Sciences, University of California, Riverside, CA, USA
| | - Mike S Hsu
- Center for Glial-Neuronal Interactions, Division of Biomedical Sciences, University of California, Riverside, CA, USA
| | - Marcus M Seldin
- Division of Cardiology, University of California, Los Angeles, CA, USA
| | - Devin K Binder
- Center for Glial-Neuronal Interactions, Division of Biomedical Sciences, University of California, Riverside, CA, USA
| |
Collapse
|
24
|
He ZP, Lu H. Aquaporin-4 gene silencing protects injured neurons after early cerebral infarction. Neural Regen Res 2015; 10:1082-7. [PMID: 26330830 PMCID: PMC4541238 DOI: 10.4103/1673-5374.160099] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2015] [Indexed: 02/04/2023] Open
Abstract
Aquaporin-4 regulates water molecule channels and is important in tissue regulation and water transportation in the brain. Upregulation of aquaporin-4 expression is closely related to cellular edema after early cerebral infarction. Cellular edema and aquaporin-4 expression can be determined by measuring cerebral infarct area and apparent diffusion coefficient using diffusion-weighted imaging (DWI). We examined the effects of silencing aquaporin-4 on cerebral infarction. Rat models of cerebral infarction were established by occlusion of the right middle cerebral artery and siRNA-aquaporin-4 was immediately injected via the right basal ganglia. In control animals, the area of high signal intensity and relative apparent diffusion coefficient value on T2-weighted imaging (T2WI) and DWI gradually increased within 0.5-6 hours after cerebral infarction. After aquaporin-4 gene silencing, the area of high signal intensity on T2WI and DWI reduced, relative apparent diffusion coefficient value was increased, and cellular edema was obviously alleviated. At 6 hours after cerebral infarction, the apparent diffusion coefficient value was similar between treatment and model groups, but angioedema was still obvious in the treatment group. These results indicate that aquaporin-4 gene silencing can effectively relieve cellular edema after early cerebral infarction; and when conducted accurately and on time, the diffusion coefficient value and the area of high signal intensity on T2WI and DWI can reflect therapeutic effects of aquaporin-4 gene silencing on cellular edema.
Collapse
Affiliation(s)
- Zhan-ping He
- Department of Radiology, Affiliated Haikou Hospital of Xiangya School of Medicine, Central South University (Department of Radiology, Haikou Municipal People's Hospital), Haikou, Hainan Province, China
| | - Hong Lu
- Department of Radiology, Affiliated Haikou Hospital of Xiangya School of Medicine, Central South University (Department of Radiology, Haikou Municipal People's Hospital), Haikou, Hainan Province, China
| |
Collapse
|
25
|
Noh JS, Pak HJ, Shin YJ, Riew TR, Park JH, Moon YW, Lee MY. Differential expression of the calcium-sensing receptor in the ischemic and border zones after transient focal cerebral ischemia in rats. J Chem Neuroanat 2015; 66-67:40-51. [DOI: 10.1016/j.jchemneu.2015.05.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 04/20/2015] [Accepted: 05/15/2015] [Indexed: 10/23/2022]
|
26
|
Kolpakova ME, Veselkina OS, Vlasov TD. Creatine in Cell Metabolism and Its Protective Action in Cerebral Ischemia. ACTA ACUST UNITED AC 2015. [DOI: 10.1007/s11055-015-0098-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
27
|
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: 3.6] [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
|
28
|
Signaling molecules regulating phenotypic conversions of astrocytes and glial scar formation in damaged nerve tissues. Neurochem Int 2014; 78:35-42. [DOI: 10.1016/j.neuint.2014.08.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 07/17/2014] [Accepted: 08/22/2014] [Indexed: 12/21/2022]
|
29
|
Wang WW, Xie CL, Zhou LL, Wang GS. The function of aquaporin4 in ischemic brain edema. Clin Neurol Neurosurg 2014; 127:5-9. [PMID: 25306413 DOI: 10.1016/j.clineuro.2014.09.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 06/17/2014] [Accepted: 09/19/2014] [Indexed: 02/01/2023]
Abstract
Cerebral ischemia injury is a primary cause of human death and long-term disability. We know that the cerebral edema induced by ischemia injury has a fatal effect on humans, which is the main cause of death for cerebral ischemia because it produces elevated intracranial pressure that leads to secondary brain damage, such as further impaired vascular perfusion and herniation of brain. Therefore, reducing the severity of brain edema has become the main therapeutic strategy for the treatment of CI. However, current treatment options for brain edema are limited and problematic. Therefore, finding novel strategies for overcoming this problem is crucial. Numerous studies demonstrated that cerebral edema may be attenuated via the regulation of AQP4 expression, thus initiating a novel therapeutic strategy against this possibly fatal condition. This review focuses on the role of AQP4 in ischemic brain edema, and its prospect as a therapeutic target.
Collapse
Affiliation(s)
- Wen-Wen Wang
- The Center of Traditional Chinese Medicine, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Cheng-long Xie
- Department of Neurology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Li-Li Zhou
- Department of Neurology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Guo-Sheng Wang
- Department of Oncology, The First Affiliated Hospital Beilun Branch of Zhejiang University, College of Medicine, Ningbo 315806, China.
| |
Collapse
|
30
|
Badaut J, Fukuda AM, Jullienne A, Petry KG. Aquaporin and brain diseases. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1840:1554-65. [PMID: 24513456 PMCID: PMC3960327 DOI: 10.1016/j.bbagen.2013.10.032] [Citation(s) in RCA: 146] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2013] [Revised: 10/09/2013] [Accepted: 10/17/2013] [Indexed: 11/28/2022]
Abstract
BACKGROUND The presence of water channel proteins, aquaporins (AQPs), in the brain led to intense research in understanding the underlying roles of each of them under normal conditions and pathological conditions. SCOPE OF REVIEW In this review, we summarize some of the recent knowledge on the 3 main AQPs (AQP1, AQP4 and AQP9), with a special focus on AQP4, the most abundant AQP in the central nervous system. MAJOR CONCLUSIONS AQP4 was most studied in several brain pathological conditions ranging from acute brain injuries (stroke, traumatic brain injury) to the chronic brain disease with autoimmune neurodegenerative diseases. To date, no specific therapeutic agents have been developed to either inhibit or enhance water flux through these channels. However, experimental results strongly underline the importance of this topic for future investigation. Early inhibition of water channels may have positive effects in prevention of edema formation in brain injuries but at later time points during the course of a disease, AQP is critical for clearance of water from the brain into blood vessels. GENERAL SIGNIFICANCE Thus, AQPs, and in particular AQP4, have important roles both in the formation and resolution of edema after brain injury. The dual, complex function of these water channel proteins makes them an excellent therapeutic target. This article is part of a Special Issue entitled Aquaporins.
Collapse
Affiliation(s)
- Jérôme Badaut
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA; Department of Physiology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA; Univ Bordeaux, CNRS UMR 5287, 146 rue Leo Saignat33076 Bordeaux cedex.
| | - Andrew M Fukuda
- Department of Physiology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| | - Amandine Jullienne
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| | - Klaus G Petry
- INSERM U1049 Neuroinflammation, Imagerie et Thérapie de la Sclérose en Plaques, F-33076 Bordeaux, France
| |
Collapse
|
31
|
Bhattacharya P, Pandey AK, Paul S, Patnaik R. Melatonin renders neuroprotection by protein kinase C mediated aquaporin-4 inhibition in animal model of focal cerebral ischemia. Life Sci 2014; 100:97-109. [PMID: 24530291 DOI: 10.1016/j.lfs.2014.01.085] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 01/02/2014] [Accepted: 01/31/2014] [Indexed: 11/30/2022]
Abstract
AIM Aquaporin-4(AQP4) expression in the brain with relation to edema formation following focal cerebral ischemia was investigated. Studies have shown that brain edema is one of the significant factors in worsening stroke outcomes. While many mechanisms may aggravate brain injury, one such potential system may involve AQP4 up regulation in stroke patients that could result in increased edema formation. Post administration of melatonin following ischemic stroke reduces AQP4 mediated brain edema and confers neuroprotection. MATERIALS AND METHODS An in-silico approach was undertaken to confirm effective melatonin-AQP4 binding. Rats were treated with 5mg/kg, i.p. melatonin or placebo at 30 min prior, 60 min post and 120 min post 60 min of middle cerebral artery occlusion (MCAO) followed by 24h reperfusion. Rats were evaluated for battery of neurological and motor function tests just before sacrifice. Brains were harvested for infarct size estimation, water content measurement, biochemical analysis, apoptosis study and western blot experiments. KEY FINDINGS Melatonin at 60 min post ischemia rendered neuroprotection as evident by reduction in cerebral infarct volume, improvement in motor and neurological deficit and reduction in brain edema. Furthermore, ischemia induced surge in levels of nitrite and malondialdehyde (MDA) were also found to be significantly reduced in ischemic brain regions in treated animals. Melatonin potentiated intrinsic antioxidant status, inhibited acid mediated rise in intracellular calcium levels, decreased apoptotic cell death and also markedly inhibited protein kinase C (PKC) influenced AQP4 expression in the cerebral cortex and dorsal striatum. SIGNIFICANCE Melatonin confers neuroprotection by protein kinase C mediated AQP4 inhibition in ischemic stroke.
Collapse
Affiliation(s)
- Pallab Bhattacharya
- Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi 221005, UP, India.
| | - Anand Kumar Pandey
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi 221005, UP, India
| | - Sudip Paul
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi 221005, UP, India; Department of Biomedical Engineering, North Eastern Hill University (NEHU), Shillong, 793022, Meghalaya, India
| | - Ranjana Patnaik
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi 221005, UP, India
| |
Collapse
|
32
|
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
|
33
|
Bhattacharya P, Pandey AK, Paul S, Patnaik R, Yavagal DR. Aquaporin-4 inhibition mediates piroxicam-induced neuroprotection against focal cerebral ischemia/reperfusion injury in rodents. PLoS One 2013; 8:e73481. [PMID: 24023878 PMCID: PMC3762750 DOI: 10.1371/journal.pone.0073481] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 07/22/2013] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND AND PURPOSE Aquaporin-4(AQP4) is an abundant water channel protein in brain that regulates water transport to maintain homeostasis. Cerebral edema resulting from AQP4 over expression is considered to be one of the major determinants for progressive neuronal insult during cerebral ischemia. Although, both upregulation and downregulation of AQP4 expression is associated with brain pathology, over expression of AQP4 is one of the chief contributors of water imbalance in brain during ischemic pathology. We have found that Piroxicam binds to AQP4 with optimal binding energy value. Thus, we hypothesized that Piroxicam is neuroprotective in the rodent cerebral ischemic model by mitigating cerebral edema via AQP4 regulation. METHODS Rats were treated with Piroxicam OR placebo at 30 min prior, 2 h post and 4 h post 60 minutes of MCAO followed by 24 hour reperfusion. Rats were evaluated for neurological deficits and motor function just before sacrifice. Brains were harvested for infarct size estimation, water content measurement, biochemical analysis, RT-PCR and western blot experiments. RESULTS Piroxicam pretreatment thirty minutes prior to ischemia and four hour post reperfusion afforded neuroprotection as evident through significant reduction in cerebral infarct volume, improvement in motor behavior, neurological deficit and reduction in brain edema. Furthermore, ischemia induced surge in levels of nitrite and malondialdehyde were also found to be significantly reduced in ischemic brain regions in treated animals. This neuroprotection was found to be associated with inhibition of acid mediated rise in intracellular calcium levels and also downregulated AQP4 expression. CONCLUSIONS Findings of the present study provide significant evidence that Piroxicam acts as a potent AQP4 regulator and renders neuroprotection in focal cerebral ischemia. Piroxicam could be clinically exploited for the treatment of brain stroke along with other anti-stroke therapeutics in future.
Collapse
Affiliation(s)
- Pallab Bhattacharya
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, (U.P.), India
- Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Anand Kumar Pandey
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, (U.P.), India
| | - Sudip Paul
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, (U.P.), India
- Department of Biomedical Engineering, North Eastern Hill University (NEHU), Shillong, Meghalaya, India
| | - Ranjana Patnaik
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, (U.P.), India
| | - Dileep R. Yavagal
- Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| |
Collapse
|
34
|
Yang J, Li MX, Luo Y, Chen T, Liu J, Fang P, Jiang B, Hu ZL, Jin Y, Chen JG, Wang F. Chronic ceftriaxone treatment rescues hippocampal memory deficit in AQP4 knockout mice via activation of GLT-1. Neuropharmacology 2013; 75:213-22. [PMID: 23973312 DOI: 10.1016/j.neuropharm.2013.08.009] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 07/03/2013] [Accepted: 08/08/2013] [Indexed: 10/26/2022]
Abstract
Aquaporin-4 (AQP4) is the predominant water channel protein in the mammalian brain, and is mainly expressed in astrocytes. Besides its important role in water transport across the blood-brain barrier, our present study demonstrated that AQP4 deficiency impaired hippocampal long-term potentiation (LTP) and hippocampus-dependent memory formation, accompanied by the increase in extracellular glutamate concentration and N-methyl-d-aspartate (NMDA) receptor-mediated currents in hippocampal dentate gyrus (DG) region. The impairment of LTP and memory formation of AQP4 knockout (KO) mice was mediated by the downregulation of glutamate transporter-1 (GLT-1) expression/function, since it can be rescued by β-lactam antibiotic ceftriaxone (Cef), a potent GLT-1 stimulator. These results suggest that AQP4 functions as the modulator of synaptic plasticity and memory, and chronic Cef treatment rescues hippocampal memory deficit induced by AQP4 knockout.
Collapse
Affiliation(s)
- Jun Yang
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Ming-Xing Li
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Yi Luo
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Tao Chen
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Jing Liu
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Peng Fang
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Bo Jiang
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Zhuang-Li Hu
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan 430030, China; The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan 430030, China.
| | - You Jin
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan 430030, China; The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan 430030, China.
| | - Jian-Guo Chen
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan 430030, China; The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan 430030, China.
| | - Fang Wang
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan 430030, China; The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan 430030, China.
| |
Collapse
|
35
|
Assentoft M, Kaptan S, Fenton RA, Hua SZ, de Groot BL, MacAulay N. Phosphorylation of rat aquaporin-4 at Ser(111) is not required for channel gating. Glia 2013; 61:1101-12. [PMID: 23616425 DOI: 10.1002/glia.22498] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Accepted: 02/27/2013] [Indexed: 01/17/2023]
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. AQP4 has been described as an important entry and exit site for water during formation of brain edema and regulation of AQP4 is therefore of therapeutic interest. Phosphorylation of some aquaporins has been proposed to regulate their water permeability via gating of the channel itself. Protein kinase (PK)-dependent phosphorylation of Ser(111) has been reported to increase the water permeability of AQP4 expressed in an astrocytic cell line. This possibility was, however, questioned based on the crystal structure of the human AQP4. Our study aimed to resolve if Ser(111) was indeed a site involved in phosphorylation-mediated gating of AQP4. The water permeability of AQP4-expressing Xenopus oocytes was not altered by a range of activators and inhibitors of PKG and PKA. Mutation of Ser(111) to alanine or aspartate (to prevent or mimic phosphorylation) did not change the water permeability of AQP4. PKG activation had no effect on the water permeability of AQP4 in primary cultures of rat astrocytes. Molecular dynamics simulations of a phosphorylation of AQP4.Ser(111) recorded no phosphorylation-induced change in water permeability. A phospho-specific antibody, exclusively recognizing AQP4 when phosphorylated on Ser(111) , failed to detect phosphorylation in cell lysate of rat brain stimulated by conditions proposed to induce phosphorylation of this residue. Thus, our data indicate a lack of phosphorylation of Ser(111) and of phosphorylation-dependent gating of AQP4.
Collapse
Affiliation(s)
- Mette Assentoft
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | |
Collapse
|
36
|
Abstract
The eye contains numerous water channel proteins and the roles of AQPs (aquaporins) in the retina are blurred, especially under disease conditions. The purpose of this study was to investigate the expression of AQP9 gene and proteins affected by elevated IOP (intraocular pressure) in a rat model of glaucoma induced by intravitreous injection of hypertonic saline into the episcleral veins. The gene and protein expressions of AQP9 were investigated by real-time PCR and Western blotting. The immunoreactive expression of AQP9, AQP4 and GFAP (glial fibrillary acidic protein) in the optic nerve of rats exposed to experimentally elevated IOP was detected by immunofluorescence microscopy. The mRNA and protein expression levels of AQP9 were up-regulated in the retina of an animal model of glaucoma. The immunoreactivities of the AQP9, AQP4 and GFAP were also detected and increased in the optic nerve region. The expression of AQP9 was up-regulated in this glaucoma model and the immunoreactivities of the AQP4 and GFAP were also detected as co-localizing with AQP9 in the optic nerve region, indicating retina ganglion cells were surrounded by activated astrocytes. This may indicate that the injured neurons may rely on the astrocytes. The alterations of AQP expression may compensate the glaucomatous damage.
Collapse
|
37
|
Shih AY, Blinder P, Tsai PS, Friedman B, Stanley G, Lyden PD, Kleinfeld D. The smallest stroke: occlusion of one penetrating vessel leads to infarction and a cognitive deficit. Nat Neurosci 2013; 16:55-63. [PMID: 23242312 PMCID: PMC3952571 DOI: 10.1038/nn.3278] [Citation(s) in RCA: 242] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 11/15/2012] [Indexed: 11/09/2022]
Abstract
Microinfarctions are present in the aged and injured human brain. Their clinical relevance is controversial, with postulated sequelae ranging from cognitive sparing to vascular dementia. To address the consequences of microinfarcts, we used controlled optical methods to create occlusions of individual penetrating arterioles or venules in rat cortex. Single microinfarcts, targeted to encompass all or part of a cortical column, impaired performance in a macrovibrissa-based behavioral task. Furthermore, the targeting of multiple vessels resulted in tissue damage that coalesced across cortex, even though the intervening penetrating vessels were acutely patent. Post-occlusion administration of memantine, a glutamate receptor antagonist that reduces cognitive decline in Alzheimer's disease, ameliorated tissue damage and perceptual deficits. Collectively, these data imply that microinfarcts likely contribute to cognitive decline. Strategies that have received limited success in the treatment of ischemic injury, which include therapeutics against excitotoxicity, may be successful against the progressive nature of vascular dementia.
Collapse
Affiliation(s)
- Andy Y. Shih
- Department of Physics, University of California at San Diego, La Jolla, CA, USA
| | - Pablo Blinder
- Department of Physics, University of California at San Diego, La Jolla, CA, USA
| | - Philbert S. Tsai
- Department of Physics, University of California at San Diego, La Jolla, CA, USA
| | - Beth Friedman
- Department of Pharmacology, University of California at San Diego, La Jolla, CA, USA
| | - Geoffrey Stanley
- Department of Physics, University of California at San Diego, La Jolla, CA, USA
| | - Patrick D. Lyden
- Department of Neurology, Cedars-Sinai Hospital, Los Angeles, CA, USA
| | - David Kleinfeld
- Department of Physics, University of California at San Diego, La Jolla, CA, USA
- Section of Neurobiology, University of California at San Diego, La Jolla, CA, USA
| |
Collapse
|
38
|
Zanotto C, Abib RT, Batassini C, Tortorelli LS, Biasibetti R, Rodrigues L, Nardin P, Hansen F, Gottfried C, Leite MC, Gonçalves CA. Non-specific inhibitors of aquaporin-4 stimulate S100B secretion in acute hippocampal slices of rats. Brain Res 2013; 1491:14-22. [DOI: 10.1016/j.brainres.2012.10.065] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Revised: 10/29/2012] [Accepted: 10/31/2012] [Indexed: 10/27/2022]
|
39
|
Aggravated inflammation and increased expression of cysteinyl leukotriene receptors in the brain after focal cerebral ischemia in AQP4-deficient mice. Neurosci Bull 2012; 28:680-92. [PMID: 23132680 DOI: 10.1007/s12264-012-1281-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2012] [Accepted: 04/06/2012] [Indexed: 10/27/2022] Open
Abstract
OBJECTIVE Aquaporin-4 (AQP4), the main water channel protein in the brain, plays a critical role in water homeostasis and brain edema. Here, we investigated its role in the inflammatory responses after focal cerebral ischemia. METHODS In AQP4-knockout (KO) and wild-type mice, focal cerebral ischemia was induced by 30 min of middle cerebral arterial occlusion (MCAO). Ischemic neuronal injury and cellular inflammatory responses, as well as the expression and localization of cysteinyl leukotriene CysLT(2) and CysLT(1) receptors, were determined at 24 and 72 h after MCAO. RESULTS AQP4-KO mice showed more neuronal loss, more severe microglial activation and neutrophil infiltration, but less astrocyte proliferation in the brain after MCAO than wild-type mice. In addition, the protein levels of both CysLT(1) and CysLT(2) receptors were up-regulated in the ischemic brain, and the up-regulation was more pronounced in AQP4-KO mice. The CysLT(1) and CysLT(2) receptors were primarily localized in neurons, microglia and neutrophils; those localized in microglia and neutrophils were enhanced in AQP4-KO mice. CONCLUSION AQP4 may play an inhibitory role in postischemic inflammation.
Collapse
|
40
|
Brain water channel proteins in health and disease. Mol Aspects Med 2012; 33:562-78. [DOI: 10.1016/j.mam.2012.03.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Revised: 03/28/2012] [Accepted: 03/31/2012] [Indexed: 02/07/2023]
|
41
|
Abstract
Mechanisms of ischemic neuronal and vascular injury remain obscure. Here we test the hypothesis that thrombin, a blood-borne coagulation factor, contributes to neurovascular injury during acute focal ischemia. Stroke was induced in adult Sprague Dawley rats by occluding the middle cerebral artery. Intra-arterial thrombin infusion during ischemia significantly increased vascular disruption and cellular injury. Intravenous infusion of argatroban, a direct thrombin inhibitor, alleviated neurovascular injury. Immunostaining showed thrombin on neurons in the ischemic core. Using an activatable cell-penetrating peptide engineered to detect thrombin activity, we discovered that thrombin proteolytic activity was specifically associated with neuronal damage during ischemia. Protease activated receptor-1, the presumptive thrombin receptor, appeared to mediate ischemic neurovascular injury. Furthermore, rats receiving thrombin during ischemia showed cognitive deficit, whereas rats receiving argatroban retained intact learning and memory. These results suggest a potential role for thrombin contributing to neurovascular injury and several potential avenues for neuroprotection.
Collapse
|
42
|
Aquaporin-4 deficiency impairs synaptic plasticity and associative fear memory in the lateral amygdala: involvement of downregulation of glutamate transporter-1 expression. Neuropsychopharmacology 2012; 37:1867-78. [PMID: 22473056 PMCID: PMC3376319 DOI: 10.1038/npp.2012.34] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Astrocytes are implicated in information processing, signal transmission, and regulation of synaptic plasticity. Aquaporin-4 (AQP4) is the major water channel in adult brain and is primarily expressed in astrocytes. A growing body of evidence indicates that AQP4 is a potential molecular target for the regulation of astrocytic function. However, little is known about the role of AQP4 in synaptic plasticity in the amygdala. Therefore, we evaluated long-term potentiation (LTP) in the lateral amygdala (LA) and associative fear memory of AQP4 knockout (KO) and wild-type mice. We found that AQP4 deficiency impaired LTP in the thalamo-LA pathway and associative fear memory. Furthermore, AQP4 deficiency significantly downregulated glutamate transporter-1 (GLT-1) expression and selectively increased NMDA receptor (NMDAR)-mediated EPSCs in the LA. However, low concentration of NMDAR antagonist reversed the impairment of LTP in KO mice. Upregulating GLT-1 expression by chronic treatment with ceftriaxone also reversed the impairment of LTP and fear memory in KO mice. These findings imply a role for AQP4 in synaptic plasticity and associative fear memory in the amygdala by regulating GLT-1 expression.
Collapse
|
43
|
6-Hydroxydopamine leads to T2 hyperintensity, decreased claudin-3 immunoreactivity and altered aquaporin 4 expression in the striatum. Behav Brain Res 2012; 232:148-58. [DOI: 10.1016/j.bbr.2012.04.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Revised: 03/30/2012] [Accepted: 04/03/2012] [Indexed: 11/23/2022]
|
44
|
Pozarowski P, Holden E, Darzynkiewicz Z. Laser scanning cytometry: principles and applications-an update. Methods Mol Biol 2012; 931:187-212. [PMID: 23027005 DOI: 10.1007/978-1-62703-056-4_11] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Laser scanning cytometer (LSC) is the microscope-based cytofluorometer that offers a plethora of unique analytical capabilities, not provided by flow cytometry (FCM). This review describes attributes of LSC and covers its numerous applications derived from plentitude of the parameters that can be measured. Among many LSC applications the following are emphasized: (a) assessment of chromatin condensation to identify mitotic, apoptotic cells, or senescent cells; (b) detection of nuclear or mitochondrial translocation of critical factors such as NF-κB, p53, or Bax; (c) semi-automatic scoring of micronuclei in mutagenicity assays; (d) analysis of fluorescence in situ hybridization (FISH) and use of the FISH analysis attribute to measure other punctuate fluorescence patterns such as γH2AX foci or receptor clustering; (e) enumeration and morphometry of nucleoli and other cell organelles; (f) analysis of progeny of individual cells in clonogenicity assay; (g) cell immunophenotyping; (h) imaging, visual examination, or sequential analysis using different probes of the same cells upon their relocation; (i) in situ enzyme kinetics, drug uptake, and other time-resolved processes; (j) analysis of tissue section architecture using fluorescent and chromogenic probes; (k) application for hypocellular samples (needle aspirate, spinal fluid, etc.); and (l) other clinical applications. Advantages and limitations of LSC are discussed and compared with FCM.
Collapse
Affiliation(s)
- Piotr Pozarowski
- The Brander Cancer Research Institute, New York Medical College, Valhalla, NY, USA
| | | | | |
Collapse
|
45
|
Medana IM, Day NPJ, Sachanonta N, Mai NTH, Dondorp AM, Pongponratn E, Hien TT, White NJ, Turner GDH. Coma in fatal adult human malaria is not caused by cerebral oedema. Malar J 2011; 10:267. [PMID: 21923924 PMCID: PMC3182981 DOI: 10.1186/1475-2875-10-267] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Accepted: 09/17/2011] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND The role of brain oedema in the pathophysiology of cerebral malaria is controversial. Coma associated with severe Plasmodium falciparum malaria is multifactorial, but associated with histological evidence of parasitized erythrocyte sequestration and resultant microvascular congestion in cerebral vessels. To determine whether these changes cause breakdown of the blood-brain barrier and resultant perivascular or parenchymal cerebral oedema, histology, immunohistochemistry and image analysis were used to define the prevalence of histological patterns of oedema and the expression of specific molecular pathways involved in water balance in the brain in adults with fatal falciparum malaria. METHODS The brains of 20 adult Vietnamese patients who died of severe malaria were examined for evidence of disrupted vascular integrity. Immunohistochemistry and image analysis was performed on brainstem sections for activation of the vascular endothelial growth factor (VEGF) receptor 2 and expression of the aquaporin 4 (AQP4) water channel protein. Fibrinogen immunostaining was assessed as evidence of blood-brain barrier leakage and perivascular oedema formation. Correlations were performed with clinical, biochemical and neuropathological parameters of severe malaria infection. RESULTS The presence of oedema, plasma protein leakage and evidence of VEGF signalling were heterogeneous in fatal falciparum malaria and did not correlate with pre-mortem coma. Differences in vascular integrity were observed between brain regions with the greatest prevalence of disruption in the brainstem, compared to the cortex or midbrain. There was a statistically non-significant trend towards higher AQP4 staining in the brainstem of cases that presented with coma (P = .02). CONCLUSIONS Histological evidence of cerebral oedema or immunohistochemical evidence of localised loss of vascular integrity did not correlate with the occurrence of pre-mortem coma in adults with fatal falciparum malaria. Enhanced expression of AQP4 water channels in the brainstem may, therefore, reflect a mix of both neuropathological or attempted neuroprotective responses to oedema formation.
Collapse
Affiliation(s)
- Isabelle M Medana
- Nuffield Department of Clinical Laboratory Sciences, The John Radcliffe Hospital, University of Oxford, Oxford, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Gankam Kengne F, Nicaise C, Soupart A, Boom A, Schiettecatte J, Pochet R, Brion JP, Decaux G. Astrocytes are an early target in osmotic demyelination syndrome. J Am Soc Nephrol 2011; 22:1834-45. [PMID: 21885671 DOI: 10.1681/asn.2010111127] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Abrupt osmotic changes during rapid correction of chronic hyponatremia result in demyelinative brain lesions, but the sequence of events linking rapid osmotic changes to myelin loss is not yet understood. Here, in a rat model of osmotic demyelination syndrome, we found that massive astrocyte death occurred after rapid correction of hyponatremia, delineating the regions of future myelin loss. Astrocyte death caused a disruption of the astrocyte-oligodendrocyte network, rapidly upregulated inflammatory cytokines genes, and increased serum S100B, which predicted clinical manifestations and outcome of osmotic demyelination. These results support a model for the pathophysiology of osmotic brain injury in which rapid correction of hyponatremia triggers apoptosis in astrocytes followed by a loss of trophic communication between astrocytes and oligodendrocytes, secondary inflammation, microglial activation, and finally demyelination.
Collapse
Affiliation(s)
- Fabrice Gankam Kengne
- Erasme University Hospital, Department of General Internal Medicine, Research Unit on Hydromineral Metabolism, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Nico B, Ribatti D. Role of aquaporins in cell migration and edema formation in human brain tumors. Exp Cell Res 2011; 317:2391-6. [PMID: 21784068 DOI: 10.1016/j.yexcr.2011.07.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Revised: 07/04/2011] [Accepted: 07/09/2011] [Indexed: 01/08/2023]
Abstract
The aquaporins (AQPs) are a family of transmembrane water channel proteins widely distributed and play a major role in transcellular and transepithelial water movement. Moreover, recent evidence indicates that AQPs may be involved in cell migration, angiogenesis, and tumor growth. This review article summarizes literature data concerning the involvement of AQP-1 and -4 in human brain tumor growth and edema formation and suggests a potential therapeutic approach by antagonizing their biological activity.
Collapse
Affiliation(s)
- Beatrice Nico
- Department of Human Anatomy and Histology, University of Bari Medical School, Bari, Italy
| | | |
Collapse
|
48
|
Madathil SK, Nelson PT, Saatman KE, Wilfred BR. MicroRNAs in CNS injury: potential roles and therapeutic implications. Bioessays 2011; 33:21-6. [PMID: 21053309 DOI: 10.1002/bies.201000069] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Sindhu K Madathil
- Spinal Cord and Brain Injury Research Center, University of Kentucky Medical Center, University of Kentucky, Lexington, KY, USA
| | | | | | | |
Collapse
|
49
|
Loreto C, Reggio E. Aquaporin and vascular diseases. Curr Neuropharmacol 2011; 8:105-11. [PMID: 21119881 PMCID: PMC2923364 DOI: 10.2174/157015910791233196] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2009] [Revised: 03/01/2010] [Accepted: 04/07/2010] [Indexed: 11/24/2022] Open
Abstract
Aquaporins (AQP) are family of water channels found in several epithelial and endothelial cells, whose recent identification has provided insights into water transport in several tissues, including the central nervous system (CNS). Since brain edema continues to be the main cause of death from several CNS diseases, such as stroke, much of the interest in AQPs and their functional contribution to the water balance is due to their possible role in clearing edema water from the brain and in managing hydrocephalus and benign intracranial hypertension, suggesting that they could be targets for future treatments of various brain conditions, particularly vascular diseases. AQPs also seem to be involved in cell migration, and a mechanism of AQP-facilitated cell migration has been proposed where local osmotic gradients created at the tip of the lamellipodium drive water influx, facilitating lamellipodial extension and cell migration. AQP-facilitated cell migration was also detected in tumour cells, suggesting that it may have an important role in tumour angiogenesis and spread, and accounting for AQP expression in many tumour cell types and for correlations found between AQP expression and tumour stage in some tumours.
Collapse
Affiliation(s)
- Carla Loreto
- Department of Anatomy, Diagnostic Pathology, Forensic Medicine, Hygene and Public Health, University of Catania, Via S. Sofia 87, 95123 Catania, Italy.
| | | |
Collapse
|
50
|
Badaut J, Ashwal S, Obenaus A. Aquaporins in cerebrovascular disease: a target for treatment of brain edema? Cerebrovasc Dis 2011; 31:521-31. [PMID: 21487216 PMCID: PMC3085520 DOI: 10.1159/000324328] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Accepted: 01/11/2011] [Indexed: 12/11/2022] Open
Abstract
In cerebrovascular disease, edema formation is frequently observed within the first 7 days and is characterized by molecular and cellular changes in the neurovascular unit. The presence of water channels, aquaporins (AQPs), within the neurovascular unit has led to intensive research in understanding the underlying roles of each of the AQPs under normal conditions and in different diseases. In this review, we summarize some of the recent knowledge on AQPs, focusing on AQP4, the most abundant AQP in the central nervous system. Several experimental models illustrate that AQPs have dual, complex regulatory roles in edema formation and resolution. To date, no specific therapeutic agents have been developed to inhibit water flux through these channels. However, experimental results strongly suggest that this is an important area for future investigation. In fact, early inhibition of water channels may have positive effects in the prevention of edema formation. At later time points during the course of disease, AQP is important for the clearance of water from the brain into blood vessels. Thus, AQPs, and in particular AQP4, have important roles in the resolution of edema after brain injury. The function of these water channel proteins makes them an excellent therapeutic target.
Collapse
Affiliation(s)
- J Badaut
- Department of Pediatrics, Loma Linda University School of Medicine, Calif., USA.
| | | | | |
Collapse
|