1
|
Chen HJ, Zhao L, Wang L, Wang ZG, Pang DW, Liu SL. Simultaneous Mapping of the Nanoscale Organization and Redox State of Extracellular Space in Living Brain Tissue. ACS NANO 2024; 18:22245-22256. [PMID: 39116272 DOI: 10.1021/acsnano.4c06059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
The spatial organization characteristics and redox status of the extracellular space (ECS) are crucial in the development of brain diseases. However, it remains a challenge to simultaneously capture dynamic changes in microstructural features and redox states at the submicron level within the ECS. Here, we developed a reversible glutathione (GSH)-responsive nanoprobe (RGN) for mapping the spatial organization features and redox status of the ECS in brain tissues with nanoscale resolution. The RGN is composed of polymer nanoparticles modified with GSH-responsive molecules and amino-functionalized methoxypoly(ethylene glycol), which exhibit exceptional single-particle brightness and excellent free diffusion capability in the ECS of brain tissues. Tracking single RGNs in acute brain slices allowed us to dynamically map spatial organizational features and redox levels within the ECS of brain tissues in disease models. This provides a powerful super-resolution imaging method that offers a potential opportunity to study the dynamic changes in the ECS microenvironment and to understand the physiological and pathological roles of the ECS in vivo.
Collapse
Affiliation(s)
- Hua-Jie Chen
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Sciences, College of Chemistry, and School of Medicine, Nankai University, Tianjin 300071, P. R. China
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, P. R. China
| | - Liang Zhao
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Sciences, College of Chemistry, and School of Medicine, Nankai University, Tianjin 300071, P. R. China
| | - Lei Wang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Sciences, College of Chemistry, and School of Medicine, Nankai University, Tianjin 300071, P. R. China
| | - Zhi-Gang Wang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Sciences, College of Chemistry, and School of Medicine, Nankai University, Tianjin 300071, P. R. China
| | - Dai-Wen Pang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Sciences, College of Chemistry, and School of Medicine, Nankai University, Tianjin 300071, P. R. China
| | - Shu-Lin Liu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Center for Analytical Sciences, College of Chemistry, and School of Medicine, Nankai University, Tianjin 300071, P. R. China
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, P. R. China
| |
Collapse
|
2
|
Naseri Kouzehgarani G, Feldsien T, Engelhard HH, Mirakhur KK, Phipps C, Nimmrich V, Clausznitzer D, Lefebvre DR. Harnessing cerebrospinal fluid circulation for drug delivery to brain tissues. Adv Drug Deliv Rev 2021; 173:20-59. [PMID: 33705875 DOI: 10.1016/j.addr.2021.03.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/10/2021] [Accepted: 03/01/2021] [Indexed: 12/31/2022]
Abstract
Initially thought to be useful only to reach tissues in the immediate vicinity of the CSF circulatory system, CSF circulation is now increasingly viewed as a viable pathway to deliver certain therapeutics deeper into brain tissues. There is emerging evidence that this goal is achievable in the case of large therapeutic proteins, provided conditions are met that are described herein. We show how fluid dynamic modeling helps predict infusion rate and duration to overcome high CSF turnover. We posit that despite model limitations and controversies, fluid dynamic models, pharmacokinetic models, preclinical testing, and a qualitative understanding of the glymphatic system circulation can be used to estimate drug penetration in brain tissues. Lastly, in addition to highlighting landmark scientific and medical literature, we provide practical advice on formulation development, device selection, and pharmacokinetic modeling. Our review of clinical studies suggests a growing interest for intra-CSF delivery, particularly for targeted proteins.
Collapse
|
3
|
Soria FN, Miguelez C, Peñagarikano O, Tønnesen J. Current Techniques for Investigating the Brain Extracellular Space. Front Neurosci 2020; 14:570750. [PMID: 33177979 PMCID: PMC7591815 DOI: 10.3389/fnins.2020.570750] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 09/17/2020] [Indexed: 12/11/2022] Open
Abstract
The brain extracellular space (ECS) is a continuous reticular compartment that lies between the cells of the brain. It is vast in extent relative to its resident cells, yet, at the same time the nano- to micrometer dimensions of its channels and reservoirs are commonly finer than the smallest cellular structures. Our conventional view of this compartment as largely static and of secondary importance for brain function is rapidly changing, and its active dynamic roles in signaling and metabolite clearance have come to the fore. It is further emerging that ECS microarchitecture is highly heterogeneous and dynamic and that ECS geometry and diffusional properties directly modulate local diffusional transport, down to the nanoscale around individual synapses. The ECS can therefore be considered an extremely complex and diverse compartment, where numerous physiological events are unfolding in parallel on spatial and temporal scales that span orders of magnitude, from milliseconds to hours, and from nanometers to centimeters. To further understand the physiological roles of the ECS and identify new ones, researchers can choose from a wide array of experimental techniques, which differ greatly in their applicability to a given sample and the type of data they produce. Here, we aim to provide a basic introduction to the available experimental techniques that have been applied to address the brain ECS, highlighting their main characteristics. We include current gold-standard techniques, as well as emerging cutting-edge modalities based on recent super-resolution microscopy. It is clear that each technique comes with unique strengths and limitations and that no single experimental method can unravel the unknown physiological roles of the brain ECS on its own.
Collapse
Affiliation(s)
- Federico N. Soria
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- Department of Neuroscience, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Cristina Miguelez
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
- Autonomic and Movement Disorders Unit, Neurodegenerative Diseases, Biocruces Health Research Institute, Barakaldo, Spain
| | - Olga Peñagarikano
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Jan Tønnesen
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- Department of Neuroscience, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| |
Collapse
|
4
|
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: 7.2] [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
|
5
|
Smith AJ, Verkman AS. The "glymphatic" mechanism for solute clearance in Alzheimer's disease: game changer or unproven speculation? FASEB J 2018; 32:543-551. [PMID: 29101220 DOI: 10.1096/fj.201700999] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
How solutes and macromolecules are removed from brain tissue is of central importance in normal brain physiology and in how toxic protein aggregates are cleared in neurodegenerative conditions, including Alzheimer's disease (AD). Conventionally, solute transport in the narrow and tortuous extracellular space in brain parenchyma has been thought to be primarily diffusive and nondirectional. The recently proposed "glymphatic" (glial-lymphatic) hypothesis posits that solute clearance is convective and driven by active fluid transport from para-arterial to paravenous spaces though aquaporin-4 water channels in astrocyte endfeet. Glymphatic, convective solute clearance has received much attention because of its broad implications for AD and other brain pathologies and even the function of sleep. However, the theoretical plausibility of glymphatic transport has been questioned, and recent data have challenged its experimental underpinnings. A substantiated mechanism of solute clearance in the brain is of considerable importance because of its implications for pathogenic mechanisms of neurologic diseases and delivery of therapeutics.-Smith, A. J., Verkman, A. S. The "glymphatic" mechanism for solute clearance in Alzheimer's disease: game changer or unproven speculation?
Collapse
Affiliation(s)
- Alex J Smith
- Department of Medicine, University of California San Francisco, San Francisco, California, USA.,Department of Physiology, University of California San Francisco, San Francisco, California, USA
| | - Alan S Verkman
- Department of Medicine, University of California San Francisco, San Francisco, California, USA.,Department of Physiology, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
6
|
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: 10.4] [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
|
7
|
Jin BJ, Smith AJ, Verkman AS. Spatial model of convective solute transport in brain extracellular space does not support a "glymphatic" mechanism. J Gen Physiol 2016; 148:489-501. [PMID: 27836940 PMCID: PMC5129742 DOI: 10.1085/jgp.201611684] [Citation(s) in RCA: 138] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 10/21/2016] [Indexed: 01/12/2023] Open
Abstract
A “glymphatic mechanism” has been proposed to mediate convective fluid transport from para-arterial to paravenous extracellular space in the brain. Jin et al. model such a system and find that diffusion, rather than convection, can account for the transport of solutes. A “glymphatic system,” which involves convective fluid transport from para-arterial to paravenous cerebrospinal fluid through brain extracellular space (ECS), has been proposed to account for solute clearance in brain, and aquaporin-4 water channels in astrocyte endfeet may have a role in this process. Here, we investigate the major predictions of the glymphatic mechanism by modeling diffusive and convective transport in brain ECS and by solving the Navier–Stokes and convection–diffusion equations, using realistic ECS geometry for short-range transport between para-arterial and paravenous spaces. Major model parameters include para-arterial and paravenous pressures, ECS volume fraction, solute diffusion coefficient, and astrocyte foot-process water permeability. The model predicts solute accumulation and clearance from the ECS after a step change in solute concentration in para-arterial fluid. The principal and robust conclusions of the model are as follows: (a) significant convective transport requires a sustained pressure difference of several mmHg between the para-arterial and paravenous fluid and is not affected by pulsatile pressure fluctuations; (b) astrocyte endfoot water permeability does not substantially alter the rate of convective transport in ECS as the resistance to flow across endfeet is far greater than in the gaps surrounding them; and (c) diffusion (without convection) in the ECS is adequate to account for experimental transport studies in brain parenchyma. Therefore, our modeling results do not support a physiologically important role for local parenchymal convective flow in solute transport through brain ECS.
Collapse
Affiliation(s)
- Byung-Ju Jin
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143.,Department of Physiology, University of California, San Francisco, San Francisco, CA 94143
| | - Alex J Smith
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143.,Department of Physiology, University of California, San Francisco, San Francisco, CA 94143
| | - Alan S Verkman
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143 .,Department of Physiology, University of California, San Francisco, San Francisco, CA 94143
| |
Collapse
|
8
|
Affiliation(s)
- Chia-Chun Lin
- Department of Materials Science
and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6272, United States
| | - Emmabeth Parrish
- Department of Materials Science
and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6272, United States
| | - Russell J. Composto
- Department of Materials Science
and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6272, United States
| |
Collapse
|
9
|
Tang YT, Mendez JM, Theriot JJ, Sawant PM, López-Valdés HE, Ju YS, Brennan KC. Minimum conditions for the induction of cortical spreading depression in brain slices. J Neurophysiol 2014; 112:2572-9. [PMID: 25122714 DOI: 10.1152/jn.00205.2014] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Cortical spreading depression (CSD) occurs during various forms of brain injury such as stroke, subarachnoid hemorrhage, and brain trauma, but it is also thought to be the mechanism of the migraine aura. It is therefore expected to occur over a range of conditions including the awake behaving state. Yet it is unclear how such a massive depolarization could occur under relatively benign conditions. Using a microfluidic device with focal stimulation capability in a mouse brain slice model, we varied extracellular potassium concentration as well as the area exposed to increased extracellular potassium to determine the minimum conditions necessary to elicit CSD. Importantly, we focused on potassium levels that are physiologically plausible (≤145 mM; the intracellular potassium concentration). We found a strong correlation between the threshold concentration and the slice area exposed to increased extracellular potassium: minimum area of exposure was needed with the highest potassium concentration, while larger areas were needed at lower concentrations. We also found that moderate elevations of extracellular potassium were able to elicit CSD in relatively small estimated tissue volumes that might be activated under noninjury conditions. Our results thus show that CSD may be inducible under the conditions that expected in migraine aura as well as those related to brain trauma.
Collapse
Affiliation(s)
- Yujie T Tang
- Department of Mechanical and Aerospace Engineering and Department of Bioengineering, University of California, Los Angeles, California
| | - Jorge M Mendez
- Department of Neurology, University of Utah, Salt Lake City, Utah; and
| | - Jeremy J Theriot
- Department of Neurology, University of Utah, Salt Lake City, Utah; and
| | - Punam M Sawant
- Department of Neurology, University of Utah, Salt Lake City, Utah; and
| | | | - Y Sungtaek Ju
- Department of Mechanical and Aerospace Engineering and Department of Bioengineering, University of California, Los Angeles, California
| | - K C Brennan
- Department of Neurology, University of Utah, Salt Lake City, Utah; and
| |
Collapse
|
10
|
Becker S, Zorec B, Miklavčič D, Pavšelj N. Transdermal transport pathway creation: Electroporation pulse order. Math Biosci 2014; 257:60-8. [PMID: 25017876 DOI: 10.1016/j.mbs.2014.07.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 06/04/2014] [Accepted: 07/03/2014] [Indexed: 11/29/2022]
Abstract
In this study we consider the physics underlying electroporation which is administered to skin in order to radically increase transdermal drug delivery. The method involves the application of intense electric fields to alter the structure of the impermeable outer layer, the stratum corneum. A generally held view in the field of skin electroporation is that the skin's drop in resistance (to transport) is proportional to the total power of the pulses (which may be inferred by the number of pulses administered). Contrary to this belief, experiments conducted in this study show that the application of high voltage pulses prior to the application of low voltage pulses result in lower transport than when low voltage pulses alone are applied (when less total pulse power is administered). In order to reconcile these unexpected experimental results, a computational model is used to conduct an analysis which shows that the high density distribution of very small aqueous pathways through the stratum corneum associated with high voltage pulses is detrimental to the evolution of larger pathways that are associated with low voltage pulses.
Collapse
Affiliation(s)
- Sid Becker
- University of Canterbury, Christchurch, New Zealand.
| | | | | | | |
Collapse
|
11
|
Boisselier L, Ferry B, Gervais R. Involvement of the lateral entorhinal cortex for the formation of cross-modal olfactory-tactile associations in the rat. Hippocampus 2014; 24:877-91. [PMID: 24715601 DOI: 10.1002/hipo.22277] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/01/2014] [Indexed: 01/08/2023]
Abstract
While the olfactory and tactile vibrissal systems have been extensively studied in the rat, the neural basis of these cross-modal associations is still elusive. Here we tested the hypothesis that the lateral entorhinal cortex (LEC) could be particularly involved. In order to tackle this question, we have developed a new behavioral paradigm which consists in finding one baited cup (+) among three, each of the cups presenting a different and specific odor/texture (OT) combination. During the acquisition of a first task (Task OT1), the three cups were associated with the following OT combination: O1T1 for the baited cup; O2T1 and O1T2 for non-baited ones. Most rats learn this task within three training sessions (20 trials/session). In a second task (Task OT2) animals had to pair another OT combination with the reward using a new set of stimuli (O3T3+, O4T3, and O3T4). Results showed that rats manage to learn Task OT2 within one session only. In a third task (Task OT3) animals had to learn another OT combination based on previously learned items (e.g. O4T4+, O1T4 and O4T1). This task is called the "recombination task." Results showed that control rats solve the recombination task within one session. Animals bilaterally implanted with cannulae in the LEC were microinfused with d-APV (3 µg/0.6 µL) just before the acquisition or the test session of each task. The results showed that NMDA receptor blockade in LEC did not affect recall of Task OT1 but strongly impaired acquisition of both Task OT2 and OT3. Moreover, two control groups of animals infused with d-APV showed no deficit in the acquisition of unimodal olfactory and tactile tasks. Taken together, these data show that the NMDA system in the LEC is involved in the acquisition of association between an olfactory and a tactile stimulus during cross-modal learning task.
Collapse
Affiliation(s)
- Lise Boisselier
- Centre de Recherche en Neurosciences de Lyon, Team Olfaction: From Coding to Memory, UMR CNRS 5292INSERM U 1028, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | | | | |
Collapse
|
12
|
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: 14.6] [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
|
13
|
Skin electroporation for transdermal drug delivery: the influence of the order of different square wave electric pulses. Int J Pharm 2013; 457:214-23. [PMID: 24076397 DOI: 10.1016/j.ijpharm.2013.09.020] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Revised: 09/11/2013] [Accepted: 09/15/2013] [Indexed: 11/20/2022]
Abstract
Electroporation can be used as an active enhancement method for intra- and transdermal drug delivery. Differences in response of skin to electric pulses depend on their amplitude, duration and number and have been a point of interest in the past. While protocols consisting of the same repetitive, mostly exponentially decaying pulses have been used before, this study is focused on comparing different combinations of square wave short high voltage (HV) and longer low voltage (LV) electroporation pulses. Our in vitro experimental results show that longer LV pulses significantly increase subsequent passive transport of calcein through dermatomed pig skin, while short HV pulses alone result in negligible calcein passive transdermal transport. Surprisingly, when the long LV pulses are preceded by short duration HV pulses, the total calcein transported is reduced significantly. This result is explained using a theoretical physics based model of individual local transport region (LTR) evolution during the applied LV pulse. The theoretical model shows that HV pulses alter the structure of the stratum corneum in such a way that when the LV pulses are applied, insufficient thermal energy is generated to initiate LTR expansion. Together, the experimental results and theoretical predictions show that the total pulse energy alone cannot account for total solute transport: that the order of the types of pulses administered must also be considered. Our findings open a direction for further improvement of the method using new protocols.
Collapse
|
14
|
FRAP in Pharmaceutical Research: Practical Guidelines and Applications in Drug Delivery. Pharm Res 2013; 31:255-70. [DOI: 10.1007/s11095-013-1146-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 07/09/2013] [Indexed: 01/02/2023]
|
15
|
Abstract
Diffusion of solutes and macromolecules in the extracellular space (ECS) in brain is important for non-synaptic intercellular communication, extracellular ionic buffering, and delivery of drugs and metabolites. Diffusion in tumor ECS is important for delivery of anti-tumor drugs. The ECS in brain comprises ∼20% of brain parenchymal volume and contains cell-cell gaps down to ∼50 nm. We have developed fluorescence methods to quantify solute diffusion in the ECS, allowing measurements deep in solid tissues using microfiberoptics with micron tip size. Diffusion through the tortuous ECS in brain is generally slowed by ∼3-5-fold compared with that in water, with approximately half of the slowing due to tortuous ECS geometry and half due to the mildly viscous extracellular matrix (ECM). Mathematical modeling of slowed diffusion in an ECS with reasonable anatomical accuracy is in good agreement with experiment. In tumor tissue, diffusion of small macromolecules is only mildly slowed (<3-fold slower than in water) in superficial tumor, but is greatly slowed (>10-fold) at a depth of few millimeters as the tumor tissue becomes more compact. Slowing by ECM components such as collagen contribute to the slowed diffusion. Therefore, as found within cells, cellular crowding and highly tortuous transport can produce only minor slowing of diffusion in the ECS.
Collapse
Affiliation(s)
- A S Verkman
- Departments of Medicine and Physiology, University of California, San Francisco, CA 94143-0521, USA
| |
Collapse
|
16
|
Dmytrenko L, Cicanic M, Anderova M, Vorisek I, Ottersen OP, Sykova E, Vargova L. The impact of alpha-syntrophin deletion on the changes in tissue structure and extracellular diffusion associated with cell swelling under physiological and pathological conditions. PLoS One 2013; 8:e68044. [PMID: 23861848 PMCID: PMC3702576 DOI: 10.1371/journal.pone.0068044] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 05/23/2013] [Indexed: 01/08/2023] Open
Abstract
Aquaporin-4 (AQP4) is the primary cellular water channel in the brain and is abundantly expressed by astrocytes along the blood-brain barrier and brain-cerebrospinal fluid interfaces. Water transport via AQP4 contributes to the activity-dependent volume changes of the extracellular space (ECS), which affect extracellular solute concentrations and neuronal excitability. AQP4 is anchored by α-syntrophin (α-syn), the deletion of which leads to reduced AQP4 levels in perivascular and subpial membranes. We used the real-time iontophoretic method and/or diffusion-weighted magnetic resonance imaging to clarify the impact of α-syn deletion on astrocyte morphology and changes in extracellular diffusion associated with cell swelling in vitro and in vivo. In mice lacking α-syn, we found higher resting values of the apparent diffusion coefficient of water (ADCW) and the extracellular volume fraction (α). No significant differences in tortuosity (λ) or non-specific uptake (k'), were found between α-syn-negative (α-syn -/-) and α-syn-positive (α-syn +/+) mice. The deletion of α-syn resulted in a significantly smaller relative decrease in α observed during elevated K(+) (10 mM) and severe hypotonic stress (-100 mOsmol/l), but not during mild hypotonic stress (-50 mOsmol/l). After the induction of terminal ischemia/anoxia, the final values of ADCW as well as of the ECS volume fraction α indicate milder cell swelling in α-syn -/- in comparison with α-syn +/+ mice. Shortly after terminal ischemia/anoxia induction, the onset of a steep rise in the extracellular potassium concentration and an increase in λ was faster in α-syn -/- mice, but the final values did not differ between α-syn -/- and α-syn +/+ mice. This study reveals that water transport through AQP4 channels enhances and accelerates astrocyte swelling. The substantially altered ECS diffusion parameters will likely affect the movement of neuroactive substances and/or trophic factors, which in turn may modulate the extent of tissue damage and/or drug distribution.
Collapse
Affiliation(s)
- Lesia Dmytrenko
- Institute of Experimental Medicine AS CR, v.v.i., Prague, Czech Republic
| | - Michal Cicanic
- Institute of Experimental Medicine AS CR, v.v.i., Prague, Czech Republic
- Charles University, 2nd Faculty of Medicine, Prague, Czech Republic
| | - Miroslava Anderova
- Institute of Experimental Medicine AS CR, v.v.i., Prague, Czech Republic
- Charles University, 2nd Faculty of Medicine, Prague, Czech Republic
| | - Ivan Vorisek
- Institute of Experimental Medicine AS CR, v.v.i., Prague, Czech Republic
- Charles University, 2nd Faculty of Medicine, Prague, Czech Republic
| | - Ole Petter Ottersen
- Center for Molecular Biology and Neuroscience and Department of Anatomy, University of Oslo, Oslo, Norway
| | - Eva Sykova
- Institute of Experimental Medicine AS CR, v.v.i., Prague, Czech Republic
- Charles University, 2nd Faculty of Medicine, Prague, Czech Republic
| | - Lydia Vargova
- Institute of Experimental Medicine AS CR, v.v.i., Prague, Czech Republic
- Charles University, 2nd Faculty of Medicine, Prague, Czech Republic
| |
Collapse
|
17
|
Jin BJ, Zhang H, Binder DK, Verkman AS. Aquaporin-4-dependent K(+) and water transport modeled in brain extracellular space following neuroexcitation. ACTA ACUST UNITED AC 2013; 141:119-32. [PMID: 23277478 PMCID: PMC3536523 DOI: 10.1085/jgp.201210883] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Potassium (K(+)) ions released into brain extracellular space (ECS) during neuroexcitation are efficiently taken up by astrocytes. Deletion of astrocyte water channel aquaporin-4 (AQP4) in mice alters neuroexcitation by reducing ECS [K(+)] accumulation and slowing K(+) reuptake. These effects could involve AQP4-dependent: (a) K(+) permeability, (b) resting ECS volume, (c) ECS contraction during K(+) reuptake, and (d) diffusion-limited water/K(+) transport coupling. To investigate the role of these mechanisms, we compared experimental data to predictions of a model of K(+) and water uptake into astrocytes after neuronal release of K(+) into the ECS. The model computed the kinetics of ECS [K(+)] and volume, with input parameters including initial ECS volume, astrocyte K(+) conductance and water permeability, and diffusion in astrocyte cytoplasm. Numerical methods were developed to compute transport and diffusion for a nonstationary astrocyte-ECS interface. The modeling showed that mechanisms b-d, together, can predict experimentally observed impairment in K(+) reuptake from the ECS in AQP4 deficiency, as well as altered K(+) accumulation in the ECS after neuroexcitation, provided that astrocyte water permeability is sufficiently reduced in AQP4 deficiency and that solute diffusion in astrocyte cytoplasm is sufficiently low. The modeling thus provides a potential explanation for AQP4-dependent K(+)/water coupling in the ECS without requiring AQP4-dependent astrocyte K(+) permeability. Our model links the physical and ion/water transport properties of brain cells with the dynamics of neuroexcitation, and supports the conclusion that reduced AQP4-dependent water transport is responsible for defective neuroexcitation in AQP4 deficiency.
Collapse
Affiliation(s)
- Byung-Ju Jin
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | | | | |
Collapse
|
18
|
Kinney JP, Spacek J, Bartol TM, Bajaj CL, Harris KM, Sejnowski TJ. Extracellular sheets and tunnels modulate glutamate diffusion in hippocampal neuropil. J Comp Neurol 2013; 521:448-64. [PMID: 22740128 PMCID: PMC3540825 DOI: 10.1002/cne.23181] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2011] [Revised: 04/20/2012] [Accepted: 06/22/2012] [Indexed: 12/30/2022]
Abstract
Although the extracellular space in the neuropil of the brain is an important channel for volume communication between cells and has other important functions, its morphology on the micron scale has not been analyzed quantitatively owing to experimental limitations. We used manual and computational techniques to reconstruct the 3D geometry of 180 μm(3) of rat CA1 hippocampal neuropil from serial electron microscopy and corrected for tissue shrinkage to reflect the in vivo state. The reconstruction revealed an interconnected network of 40-80 nm diameter tunnels, formed at the junction of three or more cellular processes, spanned by sheets between pairs of cell surfaces with 10-40 nm width. The tunnels tended to occur around synapses and axons, and the sheets were enriched around astrocytes. Monte Carlo simulations of diffusion within the reconstructed neuropil demonstrate that the rate of diffusion of neurotransmitter and other small molecules was slower in sheets than in tunnels. Thus, the non-uniformity found in the extracellular space may have specialized functions for signaling (sheets) and volume transmission (tunnels).
Collapse
Affiliation(s)
- Justin P Kinney
- Howard Hughes Medical Institute, Salk Institute for Biological Studies, La Jolla, California 92037, USA
| | | | | | | | | | | |
Collapse
|
19
|
Wolak DJ, Thorne RG. Diffusion of macromolecules in the brain: implications for drug delivery. Mol Pharm 2013; 10:1492-504. [PMID: 23298378 DOI: 10.1021/mp300495e] [Citation(s) in RCA: 187] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Therapeutics must diffuse through the brain extracellular space (ECS) in order to distribute within the central nervous system (CNS) compartment; this requirement holds both for drugs that are directly placed within the CNS (i.e., central input) and for drugs that cross the barriers separating blood and brain following systemic administration. The diffusion of any substance within the CNS may be affected by a number of properties associated with the brain microenvironment, e.g., the volume fraction, geometry, width, and local viscosity of the ECS, as well as interactions with cell surfaces, the extracellular matrix, and components of the interstitial fluid. Here, we discuss ECS properties important in governing the distribution of macromolecules (e.g., antibodies and other protein therapeutics), nanoparticles and viral vectors within the CNS. We also provide an introduction to some of the methods commonly applied to measure diffusion of molecules in the brain ECS, with a particular emphasis on those used for determining the diffusion properties of macromolecules. Finally, we discuss how quantitative diffusion measurements can be used to better understand and potentially even improve upon CNS drug delivery by modeling delivery within and across species, screening drugs and drug conjugates, evaluating methods for altering drug distribution, and appreciating important changes in drug distribution that may occur with CNS disease or injury.
Collapse
Affiliation(s)
- Daniel J Wolak
- Pharmaceutical Sciences Division, University of Wisconsin-Madison School of Pharmacy, Madison, Wisconsin 53705, United States
| | | |
Collapse
|
20
|
McLean D, Cooke MJ, Wang Y, Fraser P, St George-Hyslop P, Shoichet MS. Targeting the amyloid-β antibody in the brain tissue of a mouse model of Alzheimer's disease. J Control Release 2011; 159:302-8. [PMID: 22245684 DOI: 10.1016/j.jconrel.2011.12.036] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Revised: 12/16/2011] [Accepted: 12/25/2011] [Indexed: 11/28/2022]
Abstract
Alzheimer's disease is a neurodegenerative disease characterized pathologically by amyloid-β (Aβ) aggregates in the brain. Notwithstanding many promising therapeutics that are under development, early diagnosis of Alzheimer's disease is limited. By targeting the Aβ aggregates, diagnosis can be improved and disease progression reduced. Molecular imaging using monoclonal antibodies to target specific isoforms of Aβ aggregates offer increased specificity in comparison to conventional imaging tracers; however, antibodies that are widely used in histology do not necessarily show similar binding in a dynamic in vivo environment. In this study, the diffusion and binding were studied of a classical monoclonal antibody, 6E10, in the brain of the TgCRND8 mouse model of AD. After intracranial injection of fluorescent 6E10, we observed broad and rapid labelling of Aβ deposits in the cortex and corpus callosum within 4h. Aβ plaques were detected up to 2.5mm away from the injection site in TgCRND8 mice and not in wild type mice at all, demonstrating specificity of binding. The apparent diffusivity and elimination constant of the anti-Aβ antibody were found to be independent of both the age of the animal and the accumulation of Aβ in the extracellular space, suggesting broad applicability of this targeting molecule. Mathematical modelling of the diffusion profiles of the anti-Aβ antibody in the brain parenchyma provides insights into the utility of antibodies as molecular imaging tools and targeted therapeutics.
Collapse
Affiliation(s)
- Daniel McLean
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
21
|
Ratelade J, Bennett JL, Verkman AS. Intravenous neuromyelitis optica autoantibody in mice targets aquaporin-4 in peripheral organs and area postrema. PLoS One 2011; 6:e27412. [PMID: 22076159 PMCID: PMC3208637 DOI: 10.1371/journal.pone.0027412] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Accepted: 10/17/2011] [Indexed: 01/09/2023] Open
Abstract
The pathogenesis of neuromyelitis optica (NMO) involves binding of IgG autoantibodies (NMO-IgG) to aquaporin-4 (AQP4) on astrocytes in the central nervous system (CNS). We studied the in vivo processing in mice of a recombinant monoclonal human NMO-IgG that binds strongly to mouse AQP4. Following intravenous administration, serum [NMO-IgG] decreased with t(½) ∼18 hours in wildtype mice and ∼41 hours in AQP4 knockout mice. NMO-IgG was localized to AQP4-expressing cell membranes in kidney (collecting duct), skeletal muscle, trachea (epithelial cells) and stomach (parietal cells). NMO-IgG was seen on astrocytes in the area postrema in brain, but not elsewhere in brain, spinal cord, optic nerve or retina. Intravenously administered NMO-IgG was also seen in brain following mechanical disruption of the blood-brain barrier. Selective cellular localization was not found for control (non-NMO) IgG, or for NMO-IgG in AQP4 knockout mice. NMO-IgG injected directly into brain parenchyma diffused over an area of ∼5 mm² over 24 hours and targeted astrocyte foot-processes. Our data establish NMO-IgG pharmacokinetics and tissue distribution in mice. The rapid access of serum NMO-IgG to AQP4 in peripheral organs but not the CNS indicates that restricted antibody access cannot account for the absence of NMO pathology in peripheral organs.
Collapse
Affiliation(s)
- Julien Ratelade
- Departments of Medicine and Physiology, University of California San Francisco, San Francisco, California, United States of America
| | - Jeffrey L. Bennett
- Departments of Neurology and Ophthalmology, University of Colorado Denver, Aurora, Colorado, United States of America
| | - A. S. Verkman
- Departments of Medicine and Physiology, University of California San Francisco, San Francisco, California, United States of America
| |
Collapse
|
22
|
Aquaporin-4: orthogonal array assembly, CNS functions, and role in neuromyelitis optica. Acta Pharmacol Sin 2011; 32:702-10. [PMID: 21552296 DOI: 10.1038/aps.2011.27] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Aquaporin-4 (AQP4) is a water-selective transporter expressed in astrocytes throughout the central nervous system, as well as in kidney, lung, stomach and skeletal muscle. The two AQP4 isoforms produced by alternative spicing, M1 and M23 AQP4, form heterotetramers that assemble in cell plasma membranes in supramolecular structures called orthogonal arrays of particles (OAPs). Phenotype analysis of AQP4-null mice indicates the involvement of AQP4 in brain and spinal cord water balance, astrocyte migration, neural signal transduction and neuroinflammation. AQP4-null mice manifest reduced brain swelling in cytotoxic cerebral edema, but increased brain swelling in vasogenic edema and hydrocephalus. AQP4 deficiency also increases seizure duration, impairs glial scarring, and reduces the severity of autoimmune neuroinflammation. Each of these phenotypes is likely explicable on the basis of reduced astrocyte water permeability in AQP4 deficiency. AQP4 is also involved in the neuroinflammatory demyelinating disease neuromyelitis optica (NMO), where autoantibodies (NMO-IgG) targeting AQP4 produce astrocyte damage and inflammation. Mice administered NMO-IgG and human complement by intracerebral injection develop characteristic NMO lesions with neuroinflammation, demyelination, perivascular complement deposition and loss of glial fibrillary acidic protein and AQP4 immunoreactivity. Our findings suggest the potential utility of AQP4-based therapeutics, including small-molecule modulators of AQP4 water transport function for therapy of brain swelling, injury and epilepsy, as well as small-molecule or monoclonal antibody blockers of NMO-IgG binding to AQP4 for therapy of NMO.
Collapse
|
23
|
Zhang H, Verkman AS. Microfiberoptic measurement of extracellular space volume in brain and tumor slices based on fluorescent dye partitioning. Biophys J 2010; 99:1284-91. [PMID: 20713014 DOI: 10.1016/j.bpj.2010.06.023] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2010] [Revised: 05/24/2010] [Accepted: 06/03/2010] [Indexed: 01/24/2023] Open
Abstract
The fractional volume occupied by extracellular space in tissues, termed alpha, is an important parameter of tissue architecture that affects cellular functions and drug delivery. We report a technically simple fluorescent dye partitioning method to measure alpha in tissue slices based on microfiberoptic detection of dye fluorescence in tissue versus overlying solution. Microfiberoptic tip geometry and dyes were selected for alpha determination from fluorescence intensity ratios, without the need to correct for illumination profile, light scattering/absorption, or dye binding. The method was validated experimentally using cell-embedded gels of specified alpha-values and optical properties. In mouse brain slices, alpha was strongly location-dependent, ranging from 0.16 in thalamus to 0.22 in brainstem, and was sensitive to cell volume changes. Aquaporin-4 water channel gene deletion caused significant extracellular space expansion, with alpha = 0.181 +/- 0.002 in cortex in wild-type mice and 0.211 +/- 0.003 in Aquaporin-4 knockout mice. In slices of LLC1 cell tumors grown in mice to approximately 5 mm diameter, alpha decreased remarkably from approximately 0.45 in superficial tumor to <0.25 in deeper (>100 mum) tumor. Fluorescent dye partitioning with microfiberoptic detection permits rapid, accurate, and anisotropy-insensitive determination of alpha-values in tissue slices.
Collapse
Affiliation(s)
- Hua Zhang
- Department of Medicine and Department of Physiology, University of California, San Francisco, California, USA
| | | |
Collapse
|
24
|
Proceedings of 156 thMeeting of the Society of British Neurological Surgeons. Br J Neurosurg 2010. [DOI: 10.3109/02688697.2010.508972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
25
|
Zhang H, Verkman AS. Aquaporin-1 tunes pain perception by interaction with Na(v)1.8 Na+ channels in dorsal root ganglion neurons. J Biol Chem 2009; 285:5896-906. [PMID: 20018876 DOI: 10.1074/jbc.m109.090233] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Aquaporin-1 (AQP1) water channels are expressed in the plasma membrane of dorsal root ganglion (DRG) neurons. We found reduced osmotic water permeability in freshly isolated DRG neurons from AQP1(-/-) versus AQP1(+/+) mice. Behavioral studies showed greatly reduced thermal inflammatory pain perception in AQP1(-/-) mice evoked by bradykinin, prostaglandin E(2), and capsaicin as well as reduced cold pain perception. Patch clamp of freshly isolated DRG neurons showed reduced action potential firing in response to current injections. Single action potentials after pulse current injections showed reduced maximum inward current, suggesting impaired Na(v)1.8 Na(+) function. Whole-cell Na(v)1.8 Na(+) currents in Na(v)1.8-expressing ND7-23 cells showed slowed frequency-dependent inactivation after AQP1 transfection. Immunoprecipitation studies showed AQP1- Na(v)1.8 Na(+) interaction, which was verified in live cells by single-particle tracking of quantum dot-labeled AQP1. Our results implicate the involvement of AQP1 in DRG neurons for the perception of inflammatory thermal pain and cold pain, whose molecular basis is accounted for, in part, by reduced Na(v)1.8-dependent membrane Na(+) current. AQP1 is, thus, a novel target for pain management.
Collapse
Affiliation(s)
- Hua Zhang
- Department of Medicine, University of California, San Francisco, California 94143-0521, USA
| | | |
Collapse
|
26
|
Verkman AS. Aquaporins: translating bench research to human disease. ACTA ACUST UNITED AC 2009; 212:1707-15. [PMID: 19448080 DOI: 10.1242/jeb.024125] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
There is considerable potential for translating knowledge of aquaporin structure, function and physiology to the clinic. One area is in aquaporin-based diagnostics. The discovery of AQP4 autoantibodies as a marker of the neuromyelitis optica form of multiple sclerosis has allowed precise diagnosis of this disease. Other aquaporin-based diagnostics are possible. Another area is in aquaporin-based genetics. Genetic diseases caused by loss-of-function mutations in aquaporins include nephrogenic diabetes insipidus and cataracts, and functionally significant aquaporin polymorphisms are beginning to be explored. Perhaps of greatest translational potential is aquaporin-based therapeutics. Information largely from aquaporin knockout mice has implicated key roles of aquaporin-facilitated water transport in transepithelial fluid transport (urinary concentrating, gland fluid secretion), water movement into and out of the brain, cell migration (angiogenesis, tumor metastasis, wound healing) and neural function (sensory signaling, seizures). A subset of aquaporins that transport both water and glycerol, the 'aquaglyceroporins', regulate glycerol content in epidermal, fat and other tissues, and are involved in skin hydration, cell proliferation, carcinogenesis and fat metabolism. Aquaporin-based modulator drugs are predicted to be of broad potential utility in the treatment of edematous states, cancer, obesity, wound healing, epilepsy and glaucoma. These exciting possibilities and their associated challenges are reviewed.
Collapse
Affiliation(s)
- A S Verkman
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, CA 94143, USA.
| |
Collapse
|
27
|
Crane JM, Tajima M, Verkman AS. Live-cell imaging of aquaporin-4 diffusion and interactions in orthogonal arrays of particles. Neuroscience 2009; 168:892-902. [PMID: 19699275 DOI: 10.1016/j.neuroscience.2009.08.034] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2009] [Revised: 08/03/2009] [Accepted: 08/12/2009] [Indexed: 11/26/2022]
Abstract
Orthogonal arrays of particles (OAPs) have been visualized for many years by freeze-fracture electron microscopy. Our laboratory discovered that aquaporin-4 (AQP4) is the protein responsible for OAP formation by demonstrating OAPs in AQP4-transfected cells and absence of OAPs in AQP4 knockout mice. We recently developed live-cell, single-molecule imaging methods to study AQP4 diffusion and interactions in OAPs. The methods include single particle tracking of quantum-dot labeled AQP4, and total internal reflection fluorescence microscopy of green fluorescent protein (GFP) and small fluorophore-labeled AQP4. The full-length (M1) form of AQP4 diffuses freely in membranes and does not form OAPs, whereas the shorter (M23) form of AQP4 forms OAPs and is nearly immobile. Analysis of a series of AQP4 truncations, point mutants and chimeras revealed that OAP formation by AQP4-M23 is stabilized by hydrophobic tetramer-tetramer interactions involving N-terminus residues, and that absence of OAPs in AQP4-M1 results from blocking of this interaction by residues just upstream from Met23. These biophysical methods are being extended to identify the cellular site of AQP4 assembly, AQP4 isoform interactions, OAP size and dynamics, and the determinants of regulated OAP assembly.
Collapse
Affiliation(s)
- J M Crane
- Departments of Medicine and Physiology, University of California, San Francisco, CA 94143-0521, USA
| | | | | |
Collapse
|
28
|
Magzoub M, Zhang H, Dix JA, Verkman AS. Extracellular space volume measured by two-color pulsed dye infusion with microfiberoptic fluorescence photodetection. Biophys J 2009; 96:2382-90. [PMID: 19289063 DOI: 10.1016/j.bpj.2008.12.3916] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2008] [Revised: 11/14/2008] [Accepted: 12/08/2008] [Indexed: 01/11/2023] Open
Abstract
The extracellular space (ECS) is the aqueous matrix surrounding cells in solid tissues. The only method to measure ECS volume fraction (alpha) in vivo has been tetramethylammonium iontophoresis, a technically challenging method developed more than 25 years ago. We report a simple, quantitative method to measure alpha by microfiberoptic fluorescence detection of a self-quenched green dye, calcein, and a reference red dye, sulforhodamine 101, after pulsed iontophoretic infusion. The idea is that the maximum increase in calcein fluorescence after iontophoresis is proportional to the aqueous volume into which the dye is deposited. We validated the method theoretically, and experimentally, using cell-embedded gels with specified alpha and ECS viscosity. Measurements in living mice gave alpha of 0.20 +/- 0.01 in brain, 0.13 +/- 0.02 in kidney and 0.074 +/- 0.01 in skeletal muscle. The technical simplicity of the "pulsed-infusion microfiberoptic photodetection" method developed here should allow elucidation of the relatively understudied biological roles of the ECS.
Collapse
Affiliation(s)
- Mazin Magzoub
- Department of Medicine, University of California, San Francisco, California, USA
| | | | | | | |
Collapse
|
29
|
AQP4 gene deletion in mice does not alter blood-brain barrier integrity or brain morphology. Neuroscience 2009; 161:764-72. [PMID: 19345723 DOI: 10.1016/j.neuroscience.2009.03.069] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2009] [Revised: 03/20/2009] [Accepted: 03/21/2009] [Indexed: 01/09/2023]
Abstract
The glial cell water channel aquaporin-4 (AQP4) plays an important role in brain edema, astrocyte migration, and neuronal excitability. Zhou et al. [Zhou J, Kong H, Hua X, Xiao M, Ding J, Hu G (2008) Altered blood-brain barrier integrity in adult aquaporin-4 knockout mice. Neuroreport 19:1-5] recently reported that AQP4 deletion significantly altered blood-brain barrier integrity and glial fibrillary acidic protein (GFAP) immunoreactivity in their AQP4 null mice. Here we describe a detailed characterization of baseline brain properties in our AQP4 null mice, including gross appearance, neuronal, astrocyte and oligodendrocyte characteristics, and blood-brain barrier integrity. Gross anatomical measurements included estimates of brain and ventricle size. Neurons, astrocytes and oligodendrocytes were assessed using the neuronal nuclear marker NeuN, the astrocyte marker GFAP, and the myelin stain Luxol Fast Blue. The blood-brain barrier was studied by electron microscopy and the horseradish peroxidase extravasation technique. There were no differences in brain and ventricle sizes between wild type and AQP4 null mice, nor were there differences in the cerebral cortical density of NeuN positive nuclei, perimicrovessel and glia limitans GFAP immunoreactivity, or the thickness and myelination of the corpus callosum. The ultrastructure of microvessels in the frontal cortex and caudate nucleus of wild type vs. AQP4 null mice was indistinguishable, with features including intact endothelial tight junctions, absence of perimicrovessel astrocyte foot process edema, and absence of horseradish peroxidase extravasation. In contrast to the report by Zhou et al. (2008), our data show that AQP4 deletion in mice does not produce major structural abnormalities in the brain.
Collapse
|
30
|
Xiao F, Hrabetová S. Enlarged extracellular space of aquaporin-4-deficient mice does not enhance diffusion of Alexa Fluor 488 or dextran polymers. Neuroscience 2009; 161:39-45. [PMID: 19303428 DOI: 10.1016/j.neuroscience.2009.03.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2009] [Revised: 03/02/2009] [Accepted: 03/05/2009] [Indexed: 10/21/2022]
Abstract
Aquaporin-4 (AQP4) water channels expressed on glia have been implicated in maintaining the volume of extracellular space (ECS). A previous diffusion study employing small cation tetramethylammonium and a real-time iontophoretic (RTI) method demonstrated an increase of about 25% in the ECS volume fraction (alpha) in the neocortex of AQP4(-/-) mice compared to AQP4(+/+) mice but no change in the hindrance imposed to diffusing molecules (tortuosity lambda). In contrast, other diffusion studies employing large molecules (dextran polymers) and a fluorescence recovery after photobleaching (FRAP) method measured a decrease of about 10%-20% in lambda in the neocortex of AQP4(-/-) mice. These conflicting findings on lambda would imply that large molecules diffuse more readily in the enlarged ECS of AQP4(-/-) mice than in wild type but small molecules do not. To test this hypothesis, we used integrative optical imaging (IOI) to measure tortuosity with a small Alexa Fluor 488 (molecular weight [MW] 547, lambda(AF)) and two large dextran polymers (MW 3000, lambda(dex3) and MW 75,000, lambda(dex75)) in the in vitro neocortex of AQP4(+/+) and AQP4(-/-) mice. We found that lambda(AF)=1.59, lambda(dex3)=1.76 and lambda(dex75)=2.30 obtained in AQP4(-/-) mice were not significantly different from lambda(AF)=1.61, lambda(dex3)=1.76, and lambda(dex75)=2.33 in AQP4(+/+) mice. These IOI results demonstrate that lambda measured with small and large molecules each remain unchanged in the enlarged ECS of AQP4(-/-) mice compared to values in AQP4(+/+) mice. Further analysis suggests that the FRAP method yields diffusion parameters not directly comparable with those obtained by IOI or RTI methods. Our findings have implications for the role of glial AQP4 in maintaining the ECS structure.
Collapse
Affiliation(s)
- F Xiao
- Department of Anatomy and Cell Biology, The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Medical Center, 450 Clarkson Avenue, Box 5, Brooklyn, NY 11203, USA
| | | |
Collapse
|
31
|
Abstract
Knockout mice have been informative in the discovery of unexpected biological functions of aquaporins. Knockout mice have confirmed the predicted roles of aquaporins in transepithelial fluid transport, as in the urinary concentrating mechanism and glandular fluid secretion. A less obvious, though predictable role of aquaporins is in tissue swelling under stress, as in the brain in stroke, tumor and infection. Phenotype analysis of aquaporin knockout mice has revealed several unexpected cellular roles of aquaporins whose mechanisms are being elucidated. Aquaporins facilitate cell migration, as seen in aquaporin-dependent tumor angiogenesis and tumor metastasis, by a mechanism that may involve facilitated water transport in lamellipodia of migrating cells. The ' aquaglyceroporins', aquaporins that transport both glycerol and water, regulate glycerol content in epidermis, fat and other tissues, and lead to a multiplicity of interesting consequences of gene disruption including dry skin, resistance to skin carcinogenesis, impaired cell proliferation and altered fat metabolism. An even more surprising role of a mammalian aquaporin is in neural signal transduction in the central nervous system. The many roles of aquaporins might be exploited for clinical benefit by modulation of aquaporin expression/function - as diuretics, and in the treatment of brain swelling, glaucoma, epilepsy, obesity and cancer.
Collapse
Affiliation(s)
- Alan S Verkman
- Departments of Medicine and Physiology, Cardiovascular Research Institute, University of California, San Francisco, CA, 94143-0521, USA.
| |
Collapse
|
32
|
Abstract
Diffusion in the extracellular space (ECS) of the brain is constrained by the volume fraction and the tortuosity and a modified diffusion equation represents the transport behavior of many molecules in the brain. Deviations from the equation reveal loss of molecules across the blood-brain barrier, through cellular uptake, binding, or other mechanisms. Early diffusion measurements used radiolabeled sucrose and other tracers. Presently, the real-time iontophoresis (RTI) method is employed for small ions and the integrative optical imaging (IOI) method for fluorescent macromolecules, including dextrans or proteins. Theoretical models and simulations of the ECS have explored the influence of ECS geometry, effects of dead-space microdomains, extracellular matrix, and interaction of macromolecules with ECS channels. Extensive experimental studies with the RTI method employing the cation tetramethylammonium (TMA) in normal brain tissue show that the volume fraction of the ECS typically is approximately 20% and the tortuosity is approximately 1.6 (i.e., free diffusion coefficient of TMA is reduced by 2.6), although there are regional variations. These parameters change during development and aging. Diffusion properties have been characterized in several interventions, including brain stimulation, osmotic challenge, and knockout of extracellular matrix components. Measurements have also been made during ischemia, in models of Alzheimer's and Parkinson's diseases, and in human gliomas. Overall, these studies improve our conception of ECS structure and the roles of glia and extracellular matrix in modulating the ECS microenvironment. Knowledge of ECS diffusion properties is valuable in contexts ranging from understanding extrasynaptic volume transmission to the development of paradigms for drug delivery to the brain.
Collapse
Affiliation(s)
- Eva Syková
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | | |
Collapse
|
33
|
Abstract
Aquaporin-4 (AQP4) is the major water channel expressed at fluid-tissue barriers throughout the brain and plays a crucial role in cerebral water balance. To assess whether these channels influence brain extracellular space (ECS) under resting physiological conditions, we used the established real-time iontophoresis method with tetramethylammonium (TMA(+)) to measure three diffusion parameters: ECS volume fraction (alpha), tortuosity (lambda), and TMA(+) loss (k'). In vivo measurements were performed in the somatosensory cortex of AQP4-deficient (AQP4(-/-)) mice and wild-type controls with matched age. Mice lacking AQP4 showed a 28% increase in alpha (0.23 +/- 0.007 vs 0.18 +/- 0.003) with no differences in lambda (1.62 +/- 0.04 vs 1.61 +/- 0.02) and k' (0.0045 +/- 0.0001 vs 0.0031 +/- 0.0009 s(-1)). Additional recordings in brain slices showed similarly elevated alpha in AQP4(-/-) mice, and no differences in lambda and k' between the two genotypes. This is the first direct comparison of ECS properties in adult mice lacking AQP4 water channels with wild-type animals and demonstrates a significant enlargement of the volume fraction but no difference in hindrance to TMA(+) diffusion, expressed as tortuosity. These findings provide direct evidence for involvement of AQP4 in modulation of the ECS volume fraction and provide a basis for future modeling of water and ion transport in the CNS.
Collapse
|
34
|
Lu DC, Zhang H, Zador Z, Verkman AS. Impaired olfaction in mice lacking aquaporin-4 water channels. FASEB J 2008; 22:3216-23. [PMID: 18511552 DOI: 10.1096/fj.07-104836] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Aquaporin-4 (AQP4) is a water-selective transport protein expressed in glial cells throughout the central nervous system. AQP4 deletion in mice produces alterations in several neuroexcitation phenomena, including hearing, vision, epilepsy, and cortical spreading depression. Here, we report defective olfaction and electroolfactogram responses in AQP4-null mice. Immunofluorescence indicated strong AQP4 expression in supportive cells of the nasal olfactory epithelium. The olfactory epithelium in AQP4-null mice had identical appearance, but did not express AQP4, and had approximately 12-fold reduced osmotic water permeability. Behavioral analysis showed greatly impaired olfaction in AQP4-null mice, with latency times of 17 +/- 0.7 vs. 55 +/- 5 s in wild-type vs. AQP4-null mice in a buried food pellet test, which was confirmed using an olfactory maze test. Electroolfactogram voltage responses to multiple odorants were reduced in AQP4-null mice, with maximal responses to triethylamine of 0.80 +/- 0.07 vs. 0.28 +/- 0.03 mV. Similar olfaction and electroolfactogram defects were found in outbred (CD1) and inbred (C57/bl6) mouse genetic backgrounds. Our results establish AQP4 as a novel determinant of olfaction, the deficiency of which probably impairs extracellular space K(+) buffering in the olfactory epithelium.
Collapse
Affiliation(s)
- Daniel C Lu
- Department of Medicine, University of California, San Francisco, CA 94143, USA
| | | | | | | |
Collapse
|
35
|
Abstract
The aquaporins (AQPs) are integral membrane proteins whose main function is to transport water across cell membranes in response to osmotic gradients. At the ocular surface, AQP1 is expressed in corneal endothelium, AQP3 and AQP5 in corneal epithelium, and AQP3 in conjunctival epithelium. AQPs are also expressed in lens fiber cells (AQP0), lens epithelium (AQP1), ciliary epithelium (AQP1, AQP4) and retinal Müller cells (AQP4). Mutations in AQP0 produce congenital cataracts in humans. Analysis of knockout mice lacking individual AQPs suggests their involvement in maintenance of corneal and lens transparency, corneal epithelial repair, intraocular pressure (IOP) regulation, retinal signal transduction and retinal swelling following injury. The mouse phenotype findings implicate AQPs as potential drug targets for therapy of elevated IOP and ocular disorders involving the cornea, lens and retina. However, much research remains in defining cell-level mechanisms for the ocular AQP functions, in establishing the relevance to human eye disease of conclusions from knockout mice, and in developing AQP-modulating drugs.
Collapse
Affiliation(s)
- A S Verkman
- Department of Medicine and Physiology, University of California, San Francisco, 1246 Health Sciences East Tower, San Francisco, CA 94143-0521, USA.
| | | | | |
Collapse
|
36
|
Mammalian aquaporins: diverse physiological roles and potential clinical significance. Expert Rev Mol Med 2008; 10:e13. [PMID: 18482462 DOI: 10.1017/s1462399408000690] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Aquaporins have multiple distinct roles in mammalian physiology. Phenotype analysis of aquaporin-knockout mice has confirmed the predicted role of aquaporins in osmotically driven transepithelial fluid transport, as occurs in the urinary concentrating mechanism and glandular fluid secretion. Aquaporins also facilitate water movement into and out of the brain in various pathologies such as stroke, tumour, infection and hydrocephalus. A major, unexpected cellular role of aquaporins was revealed by analysis of knockout mice: aquaporins facilitate cell migration, as occurs in angiogenesis, tumour metastasis, wound healing, and glial scar formation. Another unexpected role of aquaporins is in neural function - in sensory signalling and seizure activity. The water-transporting function of aquaporins is likely responsible for these roles. A subset of aquaporins that transport both water and glycerol, the 'aquaglyceroporins', regulate glycerol content in epidermal, fat and other tissues. Mice lacking various aquaglyceroporins have several interesting phenotypes, including dry skin, resistance to skin carcinogenesis, impaired cell proliferation, and altered fat metabolism. The various roles of aquaporins might be exploited clinically by development of drugs to alter aquaporin expression or function, which could serve as diuretics, and in the treatment of brain swelling, glaucoma, epilepsy, obesity and cancer.
Collapse
|
37
|
Random-walk model of diffusion in three dimensions in brain extracellular space: comparison with microfiberoptic photobleaching measurements. Biophys J 2008; 95:1785-94. [PMID: 18469079 DOI: 10.1529/biophysj.108.131466] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Diffusion through the extracellular space (ECS) in brain is important in drug delivery, intercellular communication, and extracellular ionic buffering. The ECS comprises approximately 20% of brain parenchymal volume and contains cell-cell gaps approximately 50 nm. We developed a random-walk model to simulate macromolecule diffusion in brain ECS in three dimensions using realistic ECS dimensions. Model inputs included ECS volume fraction (alpha), cell size, cell-cell gap geometry, intercellular lake (expanded regions of brain ECS) dimensions, and molecular size of the diffusing solute. Model output was relative solute diffusion in water versus brain ECS (D(o)/D). Experimental D(o)/D for comparison with model predictions was measured using a microfiberoptic fluorescence photobleaching method involving stereotaxic insertion of a micron-size optical fiber into mouse brain. D(o)/D for the small solute calcein in different regions of brain was in the range 3.0-4.1, and increased with brain cell swelling after water intoxication. D(o)/D also increased with increasing size of the diffusing solute, particularly in deep brain nuclei. Simulations of measured D(o)/D using realistic alpha, cell size and cell-cell gap required the presence of intercellular lakes at multicell contact points, and the contact length of cell-cell gaps to be least 50-fold smaller than cell size. The model accurately predicted D(o)/D for different solute sizes. Also, the modeling showed unanticipated effects on D(o)/D of changing ECS and cell dimensions that implicated solute trapping by lakes. Our model establishes the geometric constraints to account quantitatively for the relatively modest slowing of solute and macromolecule diffusion in brain ECS.
Collapse
|
38
|
Tait MJ, Saadoun S, Bell BA, Papadopoulos MC. Water movements in the brain: role of aquaporins. Trends Neurosci 2007; 31:37-43. [PMID: 18054802 DOI: 10.1016/j.tins.2007.11.003] [Citation(s) in RCA: 207] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2007] [Revised: 11/11/2007] [Accepted: 11/12/2007] [Indexed: 12/19/2022]
Abstract
About 80% of the brain is water. This review discusses the importance of the three brain water-channel proteins (AQP1, AQP4, AQP9) in brain physiology. AQP1 is expressed in the choroid plexus and participates in forming cerebrospinal fluid. AQP4, found in astrocyte foot processes, glia limitans and ependyma, facilitates water movement into and out of the brain, accelerates astrocyte migration and alters neuronal activity. Recently, AQP4 autoantibodies were discovered in patients with neuromyelitis optica, a demyelinating disease, and are now being used to diagnose this condition. AQP9 is present in some glia and neurons, but its function is unclear. Finally, we discuss how the discovery of AQP activators and inhibitors will be the next major step in this field.
Collapse
Affiliation(s)
- Matthew J Tait
- Academic Neurosurgery Unit, St. George's University of London, Cranmer Terrace, Tooting, London SW17 0RE, UK
| | | | | | | |
Collapse
|