1
|
Zhang X, Zhang Y, Su Q, Liu Y, Li Z, Yong VW, Xue M. Ion Channel Dysregulation Following Intracerebral Hemorrhage. Neurosci Bull 2024; 40:401-414. [PMID: 37755675 PMCID: PMC10912428 DOI: 10.1007/s12264-023-01118-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 06/14/2023] [Indexed: 09/28/2023] Open
Abstract
Injury to the brain after intracerebral hemorrhage (ICH) results from numerous complex cellular mechanisms. At present, effective therapy for ICH is limited and a better understanding of the mechanisms of brain injury is necessary to improve prognosis. There is increasing evidence that ion channel dysregulation occurs at multiple stages in primary and secondary brain injury following ICH. Ion channels such as TWIK-related K+ channel 1, sulfonylurea 1 transient receptor potential melastatin 4 and glutamate-gated channels affect ion homeostasis in ICH. They in turn participate in the formation of brain edema, disruption of the blood-brain barrier, and the generation of neurotoxicity. In this review, we summarize the interaction between ions and ion channels, the effects of ion channel dysregulation, and we discuss some therapeutics based on ion-channel modulation following ICH.
Collapse
Affiliation(s)
- Xiangyu Zhang
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China
| | - Yan Zhang
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China
| | - Qiuyang Su
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China
| | - Yang Liu
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China
| | - Zhe Li
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China
| | - V Wee Yong
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Calgary, AB, T2N 1N4, Canada.
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China.
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China.
| |
Collapse
|
2
|
Gao G, Sumrall ES, Pitchiaya S, Bitzer M, Alberti S, Walter NG. Biomolecular condensates in kidney physiology and disease. Nat Rev Nephrol 2023; 19:756-770. [PMID: 37752323 DOI: 10.1038/s41581-023-00767-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2023] [Indexed: 09/28/2023]
Abstract
The regulation and preservation of distinct intracellular and extracellular solute microenvironments is crucial for the maintenance of cellular homeostasis. In mammals, the kidneys control bodily salt and water homeostasis. Specifically, the urine-concentrating mechanism within the renal medulla causes fluctuations in extracellular osmolarity, which enables cells of the kidney to either conserve or eliminate water and electrolytes, depending on the balance between intake and loss. However, relatively little is known about the subcellular and molecular changes caused by such osmotic stresses. Advances have shown that many cells, including those of the kidney, rapidly (within seconds) and reversibly (within minutes) assemble membraneless, nano-to-microscale subcellular assemblies termed biomolecular condensates via the biophysical process of hyperosmotic phase separation (HOPS). Mechanistically, osmotic cell compression mediates changes in intracellular hydration, concentration and molecular crowding, rendering HOPS one of many related phase-separation phenomena. Osmotic stress causes numerous homo-multimeric proteins to condense, thereby affecting gene expression and cell survival. HOPS rapidly regulates specific cellular biochemical processes before appropriate protective or corrective action by broader stress response mechanisms can be initiated. Here, we broadly survey emerging evidence for, and the impact of, biomolecular condensates in nephrology, where initial concentration buffering by HOPS and its subsequent cellular escalation mechanisms are expected to have important implications for kidney physiology and disease.
Collapse
Affiliation(s)
- Guoming Gao
- Biophysics Graduate Program, University of Michigan, Ann Arbor, MI, USA
- Department of Chemistry and Center for RNA Biomedicine, University of Michigan, Ann Arbor, MI, USA
| | - Emily S Sumrall
- Biophysics Graduate Program, University of Michigan, Ann Arbor, MI, USA
- Department of Chemistry and Center for RNA Biomedicine, University of Michigan, Ann Arbor, MI, USA
| | | | - Markus Bitzer
- Department of Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Simon Alberti
- Technische Universität Dresden, Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Engineering (CMCB), Dresden, Germany
| | - Nils G Walter
- Department of Chemistry and Center for RNA Biomedicine, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
3
|
Randhawa K, Jahani-Asl A. CLIC1 regulation of cancer stem cells in glioblastoma. CURRENT TOPICS IN MEMBRANES 2023; 92:99-123. [PMID: 38007271 DOI: 10.1016/bs.ctm.2023.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2023]
Abstract
Chloride intracellular channel 1 (CLIC1) has emerged as a therapeutic target in various cancers. CLIC1 promotes cell cycle progression and cancer stem cell (CSC) self-renewal. Furthermore, CLIC1 is shown to play diverse roles in proliferation, cell volume regulation, tumour invasion, migration, and angiogenesis. In glioblastoma (GB), CLIC1 facilitates the G1/S phase transition and tightly regulates glioma stem-like cells (GSCs), a rare population of self-renewing CSCs with central roles in tumour resistance to therapy and tumour recurrence. CLIC1 is found as either a monomeric soluble protein or as a non-covalent dimeric protein that can form an ion channel. The ratio of dimeric to monomeric protein is altered in GSCs and depends on the cell redox state. Elucidating the mechanisms underlying the alterations in CLIC1 expression and structural transitions will further our understanding of its role in GSC biology. This review will highlight the role of CLIC1 in GSCs and its significance in facilitating different hallmarks of cancer.
Collapse
Affiliation(s)
- Kamaldeep Randhawa
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada; Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Arezu Jahani-Asl
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada; Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada; Regenerative Medicine Program and Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.
| |
Collapse
|
4
|
Lopina OD, Fedorov DA, Sidorenko SV, Bukach OV, Klimanova EA. Sodium Ions as Regulators of Transcription in Mammalian Cells. BIOCHEMISTRY. BIOKHIMIIA 2022; 87:789-799. [PMID: 36171659 DOI: 10.1134/s0006297922080107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/13/2022] [Accepted: 06/13/2022] [Indexed: 06/16/2023]
Abstract
The maintenance of an uneven distribution of Na+ and K+ ions between the cytoplasm and extracellular medium is the basis for the functioning of any animal cell. Changes in the intracellular ratio of these cations occur in response to numerous stimuli and are important for the cell activity regulation. Numerous experimental data have shown that gene transcription in mammalian cells can be regulated by changes in the intracellular [Na+]i/[K+]i ratio. Here, we discuss possible mechanisms of such regulation in various cell types, with special attention to the [Ca2+]-independent signaling pathways that suggest the presence of an intracellular sensor of monovalent cations. As such sensor, we propose the secondary structures of nucleic acids called G-quadruplexes. They are widely represented in mammalian genomes and are often found in the promoters of genes encoding transcription factors.
Collapse
Affiliation(s)
- Olga D Lopina
- Lomonosov Moscow State University, Faculty of Biology, Moscow, 119234, Russia.
| | - Dmitrii A Fedorov
- Lomonosov Moscow State University, Faculty of Biology, Moscow, 119234, Russia
| | | | - Olesya V Bukach
- Lomonosov Moscow State University, Faculty of Biology, Moscow, 119234, Russia
| | | |
Collapse
|
5
|
Population balance modelling captures host cell protein dynamics in CHO cell cultures. PLoS One 2022; 17:e0265886. [PMID: 35320326 PMCID: PMC8959726 DOI: 10.1371/journal.pone.0265886] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 03/09/2022] [Indexed: 11/19/2022] Open
Abstract
Monoclonal antibodies (mAbs) have been extensively studied for their wide therapeutic and research applications. Increases in mAb titre has been achieved mainly by cell culture media/feed improvement and cell line engineering to increase cell density and specific mAb productivity. However, this improvement has shifted the bottleneck to downstream purification steps. The higher accumulation of the main cell-derived impurities, host cell proteins (HCPs), in the supernatant can negatively affect product integrity and immunogenicity in addition to increasing the cost of capture and polishing steps. Mathematical modelling of bioprocess dynamics is a valuable tool to improve industrial production at fast rate and low cost. Herein, a single stage volume-based population balance model (PBM) has been built to capture Chinese hamster ovary (CHO) cell behaviour in fed-batch bioreactors. Using cell volume as the internal variable, the model captures the dynamics of mAb and HCP accumulation extracellularly under physiological and mild hypothermic culture conditions. Model-based analysis and orthogonal measurements of lactate dehydrogenase activity and double-stranded DNA concentration in the supernatant show that a significant proportion of HCPs found in the extracellular matrix is secreted by viable cells. The PBM then served as a platform for generating operating strategies that optimise antibody titre and increase cost-efficiency while minimising impurity levels.
Collapse
|
6
|
Engels M, Kalia M, Rahmati S, Petersilie L, Kovermann P, van Putten MJAM, Rose CR, Meijer HGE, Gensch T, Fahlke C. Glial Chloride Homeostasis Under Transient Ischemic Stress. Front Cell Neurosci 2021; 15:735300. [PMID: 34602981 PMCID: PMC8481871 DOI: 10.3389/fncel.2021.735300] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/23/2021] [Indexed: 12/17/2022] Open
Abstract
High water permeabilities permit rapid adjustments of glial volume upon changes in external and internal osmolarity, and pathologically altered intracellular chloride concentrations ([Cl–]int) and glial cell swelling are often assumed to represent early events in ischemia, infections, or traumatic brain injury. Experimental data for glial [Cl–]int are lacking for most brain regions, under normal as well as under pathological conditions. We measured [Cl–]int in hippocampal and neocortical astrocytes and in hippocampal radial glia-like (RGL) cells in acute murine brain slices using fluorescence lifetime imaging microscopy with the chloride-sensitive dye MQAE at room temperature. We observed substantial heterogeneity in baseline [Cl–]int, ranging from 14.0 ± 2.0 mM in neocortical astrocytes to 28.4 ± 3.0 mM in dentate gyrus astrocytes. Chloride accumulation by the Na+-K+-2Cl– cotransporter (NKCC1) and chloride outward transport (efflux) through K+-Cl– cotransporters (KCC1 and KCC3) or excitatory amino acid transporter (EAAT) anion channels control [Cl–]int to variable extent in distinct brain regions. In hippocampal astrocytes, blocking NKCC1 decreased [Cl–]int, whereas KCC or EAAT anion channel inhibition had little effect. In contrast, neocortical astrocytic or RGL [Cl–]int was very sensitive to block of chloride outward transport, but not to NKCC1 inhibition. Mathematical modeling demonstrated that higher numbers of NKCC1 and KCC transporters can account for lower [Cl–]int in neocortical than in hippocampal astrocytes. Energy depletion mimicking ischemia for up to 10 min did not result in pronounced changes in [Cl–]int in any of the tested glial cell types. However, [Cl–]int changes occurred under ischemic conditions after blocking selected anion transporters. We conclude that stimulated chloride accumulation and chloride efflux compensate for each other and prevent glial swelling under transient energy deprivation.
Collapse
Affiliation(s)
- Miriam Engels
- Institute of Biological Information Processing, Molekular-und Zellphysiologie (IBI-1), Forschungszentrum Jülich, Jülich, Germany
| | - Manu Kalia
- Applied Analysis, Department of Applied Mathematics, University of Twente, Enschede, Netherlands.,Institute of Neurobiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Sarah Rahmati
- Institute of Biological Information Processing, Molekular-und Zellphysiologie (IBI-1), Forschungszentrum Jülich, Jülich, Germany
| | - Laura Petersilie
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Peter Kovermann
- Institute of Biological Information Processing, Molekular-und Zellphysiologie (IBI-1), Forschungszentrum Jülich, Jülich, Germany
| | | | - Christine R Rose
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Hil G E Meijer
- Applied Analysis, Department of Applied Mathematics, University of Twente, Enschede, Netherlands
| | - Thomas Gensch
- Institute of Biological Information Processing, Molekular-und Zellphysiologie (IBI-1), Forschungszentrum Jülich, Jülich, Germany
| | - Christoph Fahlke
- Institute of Biological Information Processing, Molekular-und Zellphysiologie (IBI-1), Forschungszentrum Jülich, Jülich, Germany
| |
Collapse
|
7
|
Bustamante HA, Ehrich MF, Klein BG. Intracellular potassium depletion enhances apoptosis induced by staurosporine in cultured trigeminal satellite glial cells. Somatosens Mot Res 2021; 38:194-201. [PMID: 34187291 DOI: 10.1080/08990220.2021.1941843] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
PURPOSE Satellite glial cells (SGC) surrounding neurons in sensory ganglia can buffer extracellular potassium, regulating the excitability of injured neurons and possibly influencing a shift from acute to neuropathic pain. SGC apoptosis may be a key component in this process. This work evaluated induction or enhancement of apoptosis in cultured trigeminal SGC following changes in intracellular potassium [K]ic. MATERIALS AND METHODS We developed SGC primary cultures from rat trigeminal ganglia (TG). Purity of our cultures was confirmed using immunofluorescence and western blot analysis for the presence of the specific marker of SGC, glutamine synthetase (GS). SGC [K]ic was depleted using hypo-osmotic shock and 4 mM bumetanide plus 10 mM ouabain. [K]ic was measured using the K+ fluorescent indicator potassium benzofuran isophthalate (PBFI-AM). RESULTS SGC tested positive for GS and hypo-osmotic shock induced a significant decrease in [K]ic at every evaluated time. Cells were then incubated for 5 h with either 2 mM staurosporine (STS) or 20 ng/ml of TNF-α and evaluated for early apoptosis and late apoptosis/necrosis by flow cytometry using annexin V and propidium iodide. A significant increase in early apoptosis, from 16 to 38%, was detected in SGC with depleted [K]ic after incubation with STS. In contrast, TNF-α did not increase early apoptosis in normal or [K]ic depleted SGC. CONCLUSION Hypo-osmotic shock induced a decrease in intracellular potassium in cultured trigeminal SGC and this enhanced apoptosis induced by STS that is associated with the mitochondrial pathway. These results suggest that K+ dysregulation may underlie apoptosis in trigeminal SGC.
Collapse
Affiliation(s)
- Hedie A Bustamante
- Faculty of Veterinary Sciences, Veterinary Clinical Sciences Institute, Universidad Austral de Chile, Valdivia, Chile
| | - Marion F Ehrich
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Bradley G Klein
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| |
Collapse
|
8
|
Bazard P, Frisina RD, Acosta AA, Dasgupta S, Bauer MA, Zhu X, Ding B. Roles of Key Ion Channels and Transport Proteins in Age-Related Hearing Loss. Int J Mol Sci 2021; 22:6158. [PMID: 34200434 PMCID: PMC8201059 DOI: 10.3390/ijms22116158] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/30/2021] [Accepted: 05/31/2021] [Indexed: 12/25/2022] Open
Abstract
The auditory system is a fascinating sensory organ that overall, converts sound signals to electrical signals of the nervous system. Initially, sound energy is converted to mechanical energy via amplification processes in the middle ear, followed by transduction of mechanical movements of the oval window into electrochemical signals in the cochlear hair cells, and finally, neural signals travel to the central auditory system, via the auditory division of the 8th cranial nerve. The majority of people above 60 years have some form of age-related hearing loss, also known as presbycusis. However, the biological mechanisms of presbycusis are complex and not yet fully delineated. In the present article, we highlight ion channels and transport proteins, which are integral for the proper functioning of the auditory system, facilitating the diffusion of various ions across auditory structures for signal transduction and processing. Like most other physiological systems, hearing abilities decline with age, hence, it is imperative to fully understand inner ear aging changes, so ion channel functions should be further investigated in the aging cochlea. In this review article, we discuss key various ion channels in the auditory system and how their functions change with age. Understanding the roles of ion channels in auditory processing could enhance the development of potential biotherapies for age-related hearing loss.
Collapse
Affiliation(s)
- Parveen Bazard
- Department of Medical Engineering, College of Engineering, University of South Florida, Tampa, FL 33620, USA; (P.B.); (A.A.A.); (S.D.); (M.A.B.); (X.Z.); (B.D.)
- Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33612, USA
| | - Robert D. Frisina
- Department of Medical Engineering, College of Engineering, University of South Florida, Tampa, FL 33620, USA; (P.B.); (A.A.A.); (S.D.); (M.A.B.); (X.Z.); (B.D.)
- Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33612, USA
- Department Communication Sciences and Disorders, College of Behavioral & Communication Sciences, Tampa, FL 33620, USA
| | - Alejandro A. Acosta
- Department of Medical Engineering, College of Engineering, University of South Florida, Tampa, FL 33620, USA; (P.B.); (A.A.A.); (S.D.); (M.A.B.); (X.Z.); (B.D.)
- Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33612, USA
| | - Sneha Dasgupta
- Department of Medical Engineering, College of Engineering, University of South Florida, Tampa, FL 33620, USA; (P.B.); (A.A.A.); (S.D.); (M.A.B.); (X.Z.); (B.D.)
- Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33612, USA
| | - Mark A. Bauer
- Department of Medical Engineering, College of Engineering, University of South Florida, Tampa, FL 33620, USA; (P.B.); (A.A.A.); (S.D.); (M.A.B.); (X.Z.); (B.D.)
- Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33612, USA
| | - Xiaoxia Zhu
- Department of Medical Engineering, College of Engineering, University of South Florida, Tampa, FL 33620, USA; (P.B.); (A.A.A.); (S.D.); (M.A.B.); (X.Z.); (B.D.)
- Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33612, USA
| | - Bo Ding
- Department of Medical Engineering, College of Engineering, University of South Florida, Tampa, FL 33620, USA; (P.B.); (A.A.A.); (S.D.); (M.A.B.); (X.Z.); (B.D.)
- Global Center for Hearing and Speech Research, University of South Florida, Tampa, FL 33612, USA
| |
Collapse
|
9
|
Kung TFC, Wilkinson CM, Dirks CA, Jickling GC, Colbourne F. Glibenclamide does not improve outcome following severe collagenase-induced intracerebral hemorrhage in rats. PLoS One 2021; 16:e0252584. [PMID: 34081746 PMCID: PMC8174736 DOI: 10.1371/journal.pone.0252584] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 05/18/2021] [Indexed: 11/26/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a devastating insult with few effective treatments. Edema and raised intracranial pressure contribute to poor outcome after ICH. Glibenclamide blocks the sulfonylurea 1 transient receptor potential melastatin 4 (Sur1-Trpm4) channel implicated in edema formation. While glibenclamide has been found to improve outcome and reduce mortality in animal models of severe ischemic stroke, in ICH the effects are less clear. In our previous study, we found no benefit after a moderate-sized bleed, while others have reported benefit. Here we tested the hypothesis that glibenclamide may only be effective in severe ICH, where edema is an important contributor to outcome. Glibenclamide (10 μg/kg loading dose, 200 ng/h continuous infusion) was administered 2 hours post-ICH induced by collagenase injection into the striatum of adult rats. A survival period of 24 hours was maintained for experiments 1-3, and 72 hours for experiment 4. Glibenclamide did not affect hematoma volume (~81 μL) or other safety endpoints (e.g., glucose levels), suggesting the drug is safe. However, glibenclamide did not lessen striatal edema (~83% brain water content), ionic dyshomeostasis (Na+, K+), or functional impairment (e.g., neurological deficits (median = 10 out of 14), etc.) at 24 hours. It also did not affect edema at 72 h (~86% brain water content), or overall mortality rates (25% and 29.4% overall in vehicle vs. glibenclamide-treated severe strokes). Furthermore, glibenclamide appears to worsen cytotoxic edema in the peri-hematoma region (cell bodies were 46% larger at 24 h, p = 0.0017), but no effect on cell volume or density was noted elsewhere. Overall, these findings refute our hypothesis, as glibenclamide produced no favorable effects following severe ICH.
Collapse
Affiliation(s)
- Tiffany F. C. Kung
- Department of Psychology, University of Alberta, Edmonton, Alberta, Canada
| | | | - Christine A. Dirks
- Department of Psychology, University of Alberta, Edmonton, Alberta, Canada
| | - Glen C. Jickling
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
- Division of Neurology, Faculty of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Frederick Colbourne
- Department of Psychology, University of Alberta, Edmonton, Alberta, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
10
|
van Putten MJ, Fahlke C, Kafitz KW, Hofmeijer J, Rose CR. Dysregulation of Astrocyte Ion Homeostasis and Its Relevance for Stroke-Induced Brain Damage. Int J Mol Sci 2021; 22:5679. [PMID: 34073593 PMCID: PMC8198632 DOI: 10.3390/ijms22115679] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/21/2021] [Accepted: 05/22/2021] [Indexed: 12/14/2022] Open
Abstract
Ischemic stroke is a leading cause of mortality and chronic disability. Either recovery or progression towards irreversible failure of neurons and astrocytes occurs within minutes to days, depending on remaining perfusion levels. Initial damage arises from energy depletion resulting in a failure to maintain homeostasis and ion gradients between extra- and intracellular spaces. Astrocytes play a key role in these processes and are thus central players in the dynamics towards recovery or progression of stroke-induced brain damage. Here, we present a synopsis of the pivotal functions of astrocytes at the tripartite synapse, which form the basis of physiological brain functioning. We summarize the evidence of astrocytic failure and its consequences under ischemic conditions. Special emphasis is put on the homeostasis and stroke-induced dysregulation of the major monovalent ions, namely Na+, K+, H+, and Cl-, and their involvement in maintenance of cellular volume and generation of cerebral edema.
Collapse
Affiliation(s)
- Michel J.A.M. van Putten
- Department of Clinical Neurophysiology, University of Twente, 7522 NB Enschede, The Netherlands; (M.J.A.M.v.P.); (J.H.)
| | - Christoph Fahlke
- Institut für Biologische Informationsprozesse, Molekular-und Zellphysiologie (IBI-1), Forschungszentrum Jülich, 52425 Jülich, Germany;
| | - Karl W. Kafitz
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany;
| | - Jeannette Hofmeijer
- Department of Clinical Neurophysiology, University of Twente, 7522 NB Enschede, The Netherlands; (M.J.A.M.v.P.); (J.H.)
| | - Christine R. Rose
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany;
| |
Collapse
|
11
|
Sears SM, Hewett SJ. Influence of glutamate and GABA transport on brain excitatory/inhibitory balance. Exp Biol Med (Maywood) 2021; 246:1069-1083. [PMID: 33554649 DOI: 10.1177/1535370221989263] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
An optimally functional brain requires both excitatory and inhibitory inputs that are regulated and balanced. A perturbation in the excitatory/inhibitory balance-as is the case in some neurological disorders/diseases (e.g. traumatic brain injury Alzheimer's disease, stroke, epilepsy and substance abuse) and disorders of development (e.g. schizophrenia, Rhett syndrome and autism spectrum disorder)-leads to dysfunctional signaling, which can result in impaired cognitive and motor function, if not frank neuronal injury. At the cellular level, transmission of glutamate and GABA, the principle excitatory and inhibitory neurotransmitters in the central nervous system control excitatory/inhibitory balance. Herein, we review the synthesis, release, and signaling of GABA and glutamate followed by a focused discussion on the importance of their transport systems to the maintenance of excitatory/inhibitory balance.
Collapse
Affiliation(s)
- Sheila Ms Sears
- Department of Biology, Program in Neuroscience, 2029Syracuse University, Syracuse, NY 13244, USA
| | - Sandra J Hewett
- Department of Biology, Program in Neuroscience, 2029Syracuse University, Syracuse, NY 13244, USA
| |
Collapse
|
12
|
Shen W, Chen S, Xiang Y, Yao Z, Chen Z, Wu X, Li L, Zeng LH. Astroglial adrenoreceptors modulate synaptic transmission and contextual fear memory formation in dentate gyrus. Neurochem Int 2020; 143:104942. [PMID: 33340594 DOI: 10.1016/j.neuint.2020.104942] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/30/2020] [Accepted: 12/13/2020] [Indexed: 11/16/2022]
Abstract
Astrocytes perform various supporting functions, including ion buffering, metabolic supplying and neurotransmitter clearance. They can also sense neuronal activity owing to the presence of specific receptors for neurotransmitters. In turn, astrocytes can regulate synaptic activity through the release of gliotransmitters. Evidence has shown that astrocytes are very sensitive to the locus coeruleus (LC) afferents. However, little is known about how LC neuromodulatory norepinephrine (NE) modulates synaptic transmission through astrocytic activity. In mouse dentate gyrus (DG), we demonstrated an increase in the frequency of miniature excitatory postsynaptic currents (mEPSC) in response to NE, which required the release of glutamate from astrocytes. The rise in glutamate release probability is likely due to the activation of presynaptic GluN2B-containing NMDA receptors. Moreover, we showed that the activation of NE signaling in DG is necessary for the formation of contextual learning memory. Thus, NE signaling activation during fear conditioning training contributed to enduring changes in the frequency of mEPSC in DG. Our results strongly support the physiological neuromodulatory role of NE signaling, which is derived from activation of astrocytes.
Collapse
Affiliation(s)
- Weida Shen
- Department of Pharmacology, School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, 310015, China.
| | - Shishuo Chen
- Department of Pharmacology, School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, 310015, China; Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Yingchun Xiang
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Zheyu Yao
- Department of Pharmacology, School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, 310015, China
| | - Zhitao Chen
- Department of Pharmacology, School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, 310015, China
| | - Xitian Wu
- Department of Pharmacology, School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, 310015, China
| | - Ling Li
- Department of Pharmacology, School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, 310015, China
| | - Ling-Hui Zeng
- Department of Pharmacology, School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, 310015, China; Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
| |
Collapse
|
13
|
Kalisvaart ACJ, Wilkinson CM, Gu S, Kung TFC, Yager J, Winship IR, van Landeghem FKH, Colbourne F. An update to the Monro-Kellie doctrine to reflect tissue compliance after severe ischemic and hemorrhagic stroke. Sci Rep 2020; 10:22013. [PMID: 33328490 PMCID: PMC7745016 DOI: 10.1038/s41598-020-78880-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 12/01/2020] [Indexed: 02/06/2023] Open
Abstract
High intracranial pressure (ICP) can impede cerebral blood flow resulting in secondary injury or death following severe stroke. Compensatory mechanisms include reduced cerebral blood and cerebrospinal fluid volumes, but these often fail to prevent raised ICP. Serendipitous observations in intracerebral hemorrhage (ICH) suggest that neurons far removed from a hematoma may shrink as an ICP compliance mechanism. Here, we sought to critically test this observation. We tracked the timing of distal tissue shrinkage (e.g. CA1) after collagenase-induced striatal ICH in rat; cell volume and density alterations (42% volume reduction, 34% density increase; p < 0.0001) were highest day one post-stroke, and rebounded over a week across brain regions. Similar effects were seen in the filament model of middle cerebral artery occlusion (22% volume reduction, 22% density increase; p ≤ 0.007), but not with the Vannucci-Rice model of hypoxic-ischemic encephalopathy (2.5% volume increase, 14% density increase; p ≥ 0.05). Concerningly, this 'tissue compliance' appears to cause sub-lethal damage, as revealed by electron microscopy after ICH. Our data challenge the long-held assumption that 'healthy' brain tissue outside the injured area maintains its volume. Given the magnitude of these effects, we posit that 'tissue compliance' is an important mechanism invoked after severe strokes.
Collapse
Affiliation(s)
- Anna C J Kalisvaart
- Department of Psychology, Faculty of Science, University of Alberta, Edmonton, AB, Canada
| | - Cassandra M Wilkinson
- Department of Psychology, Faculty of Science, University of Alberta, Edmonton, AB, Canada
| | - Sherry Gu
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Tiffany F C Kung
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Jerome Yager
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Ian R Winship
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
- Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Frank K H van Landeghem
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
- Department of Laboratory Medicine and Pathology, University of Alberta Hospital, Edmonton, Canada
| | - Frederick Colbourne
- Department of Psychology, Faculty of Science, University of Alberta, Edmonton, AB, Canada.
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada.
| |
Collapse
|
14
|
Lopina OD, Tverskoi AM, Klimanova EA, Sidorenko SV, Orlov SN. Ouabain-Induced Cell Death and Survival. Role of α1-Na,K-ATPase-Mediated Signaling and [Na +] i/[K +] i-Dependent Gene Expression. Front Physiol 2020; 11:1060. [PMID: 33013454 PMCID: PMC7498651 DOI: 10.3389/fphys.2020.01060] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/31/2020] [Indexed: 12/12/2022] Open
Abstract
Ouabain is of cardiotonic steroids (CTS) family that is plant-derived compounds and is known for many years as therapeutic and cytotoxic agents. They are specific inhibitors of Na,K-ATPase, the enzyme, which pumps Na+ and K+ across plasma membrane of animal cells. Treatment of cells by CTS affects various cellular functions connected with the maintenance of the transmembrane gradient of Na+ and K+. Numerous studies demonstrated that binding of CTS to Na,K-ATPase not only suppresses its activity but also induces some signal pathways. This review is focused on different mechanisms of two ouabain effects: their ability (1) to protect rodent cells from apoptosis through the expression of [Na+]i-sensitive genes and (2) to trigger death of non-rodents cells (so-called «oncosis»), possessing combined markers of «classic» necrosis and «classic» apoptosis. Detailed study of oncosis demonstrated that the elevation of the [Na+]i/[K+]i ratio is not a sufficient for its triggering. Non-rodent cell death is determined by the characteristic property of "sensitive" to ouabain α1-subunit of Na,K-ATPase. In this case, ouabain binding leads to enzyme conformational changes triggering the activation of p38 mitogen-activated protein kinases (MAPK) signaling. The survival of rodent cells with ouabain-«resistant» α1-subunit is connected with another conformational transition induced by ouabain binding that results in the activation of ERK 1/2 signaling pathway.
Collapse
Affiliation(s)
- Olga Dmitrievna Lopina
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Artem Mikhaylovich Tverskoi
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences (RAS), Moscow, Russia
- Laboratory of Biological Membranes, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | | | | | - Sergei Nikolaevich Orlov
- Laboratory of Biological Membranes, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
15
|
Rana PS, Kurokawa M, Model MA. Evidence for macromolecular crowding as a direct apoptotic stimulus. J Cell Sci 2020; 133:jcs243931. [PMID: 32393677 PMCID: PMC7240305 DOI: 10.1242/jcs.243931] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/13/2020] [Indexed: 12/13/2022] Open
Abstract
Potassium loss and persistent shrinkage have both been implicated in apoptosis but their relationship and respective roles remain controversial. We approached this problem by clamping intracellular sodium and potassium in HeLa or MDCK cells using a combination of ionophores. Although ionophore treatment caused significant cell swelling, the initial volume could be restored and further reduced by application of sucrose. The swollen cells treated with ionophores remained viable for at least 8 h without any signs of apoptosis. Application of sucrose and the resulting shrinkage caused volume-dependent intrinsic apoptosis with all its classical features: inversion of phosphatidylserine, caspase activation and Bcl-2-dependent release of cytochrome c from mitochondria. In other experiments, apoptosis was induced by addition of the protein kinase inhibitor staurosporine at various degrees of swelling. Our results show that: (1) persistent shrinkage can cause apoptosis regardless of intracellular sodium or potassium composition or of the state of actin cytoskeleton; (2) strong potassium dependence of caspase activation is only observed in swollen cells with a reduced density of cytosolic proteins. We conclude that macromolecular crowding can be an important factor in determining the transition of cells to apoptosis.
Collapse
Affiliation(s)
- Priyanka S Rana
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
| | - Manabu Kurokawa
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
| | - Michael A Model
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
| |
Collapse
|
16
|
Chen L, König B, Liu T, Pervaiz S, Razzaque YS, Stauber T. More than just a pressure relief valve: physiological roles of volume-regulated LRRC8 anion channels. Biol Chem 2019; 400:1481-1496. [DOI: 10.1515/hsz-2019-0189] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 04/27/2019] [Indexed: 12/29/2022]
Abstract
Abstract
The volume-regulated anion channel (VRAC) is a key player in the volume regulation of vertebrate cells. This ubiquitously expressed channel opens upon osmotic cell swelling and potentially other cues and releases chloride and organic osmolytes, which contributes to regulatory volume decrease (RVD). A plethora of studies have proposed a wide range of physiological roles for VRAC beyond volume regulation including cell proliferation, differentiation and migration, apoptosis, intercellular communication by direct release of signaling molecules and by supporting the exocytosis of insulin. VRAC was additionally implicated in pathological states such as cancer therapy resistance and excitotoxicity under ischemic conditions. Following extensive investigations, 5 years ago leucine-rich repeat-containing family 8 (LRRC8) heteromers containing LRRC8A were identified as the pore-forming components of VRAC. Since then, molecular biological approaches have allowed further insight into the biophysical properties and structure of VRAC. Heterologous expression, siRNA-mediated downregulation and genome editing in cells, as well as the use of animal models have enabled the assessment of the proposed physiological roles, together with the identification of new functions including spermatogenesis and the uptake of antibiotics and platinum-based cancer drugs. This review discusses the recent molecular biological insights into the physiology of VRAC in relation to its previously proposed roles.
Collapse
Affiliation(s)
- Lingye Chen
- Institut für Chemie und Biochemie , Freie Universität Berlin , Thielallee 63 , D-14195 Berlin , Germany
| | - Benjamin König
- Institut für Chemie und Biochemie , Freie Universität Berlin , Thielallee 63 , D-14195 Berlin , Germany
| | - Tianbao Liu
- Institut für Chemie und Biochemie , Freie Universität Berlin , Thielallee 63 , D-14195 Berlin , Germany
| | - Sumaira Pervaiz
- Institut für Chemie und Biochemie , Freie Universität Berlin , Thielallee 63 , D-14195 Berlin , Germany
| | - Yasmin S. Razzaque
- Institut für Chemie und Biochemie , Freie Universität Berlin , Thielallee 63 , D-14195 Berlin , Germany
| | - Tobias Stauber
- Institut für Chemie und Biochemie , Freie Universität Berlin , Thielallee 63 , D-14195 Berlin , Germany
| |
Collapse
|
17
|
Toft-Bertelsen TL, Larsen BR, MacAulay N. Sensing and regulation of cell volume - we know so much and yet understand so little: TRPV4 as a sensor of volume changes but possibly without a volume-regulatory role? Channels (Austin) 2019; 12:100-108. [PMID: 29424275 PMCID: PMC5972811 DOI: 10.1080/19336950.2018.1438009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Cellular volume changes lead to initiation of cell volume regulatory events, the molecular identity of which remains unresolved. We here discuss experimental challenges associated with investigation of volume regulation during application of large, non-physiological osmotic gradients. The TRPV4 ion channel responds to volume increase irrespectively of the molecular mechanism underlying cell swelling, and is thus considered a sensor of volume changes. Evidence pointing towards the involvement of TRPV4 in subsequent volume regulatory mechanisms is intriguing, yet far from conclusive. We here present an experimental setting with astrocytic cell swelling in the absence of externally applied osmotic gradients, and the lack of evidence for involvement of TRPV4 in this regulatory volume response. Our aim with these new data and the preceding discussion is to stimulate further experimental effort in this area of research to clarify the role of TRPV4 and other channels and transporters in regulatory volume responses.
Collapse
Affiliation(s)
| | - Brian R Larsen
- a Department of Neuroscience , University of Copenhagen , Copenhagen , Denmark
| | - Nanna MacAulay
- a Department of Neuroscience , University of Copenhagen , Copenhagen , Denmark
| |
Collapse
|
18
|
Zhou DR, Eid R, Miller KA, Boucher E, Mandato CA, Greenwood MT. Intracellular second messengers mediate stress inducible hormesis and Programmed Cell Death: A review. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:773-792. [PMID: 30716408 DOI: 10.1016/j.bbamcr.2019.01.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 01/25/2019] [Accepted: 01/29/2019] [Indexed: 12/11/2022]
|
19
|
Bozza DC, Freire CA, Prodocimo V. Osmo-ionic regulation and carbonic anhydrase, Na+/K+-ATPase and V-H+-ATPase activities in gills of the ancient freshwater crustacean Aegla schmitti (Anomura) exposed to high salinities. Comp Biochem Physiol A Mol Integr Physiol 2019; 231:201-208. [DOI: 10.1016/j.cbpa.2019.02.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/18/2019] [Accepted: 02/21/2019] [Indexed: 12/13/2022]
|
20
|
Khedr S, Palygin O, Pavlov TS, Blass G, Levchenko V, Alsheikh A, Brands MW, El-Meanawy A, Staruschenko A. Increased ENaC activity during kidney preservation in Wisconsin solution. BMC Nephrol 2019; 20:145. [PMID: 31035971 PMCID: PMC6489205 DOI: 10.1186/s12882-019-1329-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 04/03/2019] [Indexed: 01/09/2023] Open
Abstract
Background The invention of an effective kidney preservation solution capable of prolonging harvested kidney viability is the core of kidney transplantation procedure. Researchers have been working on upgrading the preservation solution quality aiming at prolonging storage time while maintaining utmost organ viability and functionality. For many years, the University of Wisconsin (UW) solution has been considered the gold standard solution for kidney preservation. However, the lifespan of kidney preservation in the UW solution is still limited. Its impact on the epithelial Na+ channel (ENaC) activity and its mediated processes is unknown and the primary goal of this study. Methods Kidneys harvested from 8 weeks old Sprague Dawley rats were divided into 4 groups depending upon the period of preservation in UW solution. Additional analysis was performed using dogs’ kidneys. ENaC activity was measured using patch clamp technique; protein expression and mRNA transcription were tested through Western blot and RT-qPCR, respectively. A colorimetric LDH level estimation was performed at different time points during UW solution preservation. Results Kidney preservation in Wisconsin solution caused reduction of the kidney size and weight and elevation of LDH level. ENaC activity increased in both rat and dog kidneys preserved in the UW solution as assessed by patch clamp analysis. On the contrary, ENaC channel mRNA levels remained unchanged. Conclusions ENaC activity is significantly elevated in the kidneys during preservation in UW solution, which might affect the immediate post-implantation allograft function and trajectory post-transplant.
Collapse
Affiliation(s)
- Sherif Khedr
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Oleg Palygin
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Tengis S Pavlov
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.,Present address: Division of Hypertension and Vascular Research, Henry Ford Hospital, Detroit, MI, 48202, USA
| | - Gregory Blass
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.,Present address: Western Kentucky University, Bowling Green, KY, 42101, USA
| | - Vladislav Levchenko
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Ammar Alsheikh
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Michael W Brands
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, 30901, USA
| | - Ashraf El-Meanawy
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Alexander Staruschenko
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
| |
Collapse
|
21
|
Roles of volume-regulatory anion channels, VSOR and Maxi-Cl, in apoptosis, cisplatin resistance, necrosis, ischemic cell death, stroke and myocardial infarction. CURRENT TOPICS IN MEMBRANES 2019; 83:205-283. [PMID: 31196606 DOI: 10.1016/bs.ctm.2019.03.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
22
|
Koç Ş, Baysal S, Koç Z, Yener AÜ. Detection of Glycemia and Osmolarity Changes Using Eye Examinations. Metab Syndr Relat Disord 2018; 16:543-550. [PMID: 30183500 DOI: 10.1089/met.2018.0076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background: Glycated hemoglobin (HbA1c) is an index of the average blood glucose level over the preceding 2-3 months. In experimental studies, the lens responded to changes in osmolarity by forming vacuoles. By observing the vacuoles of the lens during eye examination, can we detect changes in osmolarity and glycemia over the last 6 months through HbA1c levels? Methods: In total, 400 patients (mean age, 67.7 ± 9.8 years), including those with diabetes mellitus, hypertension, and heart failure, were included in the study. The control group contained 70 patients matched in terms of age and sex and who had no prior disease (mean age, 67.8 ± 9.4 years). Monthly Na, glucose, and blood urea nitrogen values were used to calculate changes in osmolarity over 6 months. HbA1c values were also recorded. Biomicroscopy was used to evaluate lens vacuolation; all vacuoles were digitally photographed and converted to ImageJ format. Results: The sensitivity and specificity of using large vacuoles to detect HbA1c ≥10% were 88.0% (95% confidence interval [CI]: 68.8-97.4) and 82.6% (95% CI: 74.1-89.2), respectively. The sensitivity and specificity of detecting a 10 mOsm/kg change in osmolarity were 61% (95% CI: 48.9-72.4) and 94.5% (95% CI: 91.5-96.7), respectively. Conclusions: Lens vacuoles, which can be observed with a simple and quick examination, can be used to detect HbA1c levels and osmolarity changes over the last 6 months. Because of their relationship to the severity of retinopathy, vacuoles can also be used as a weak control indicator.
Collapse
Affiliation(s)
- Şahbender Koç
- Department of Cardiology and Keçiören Education and Training Hospital, University of Health Sciences, Ankara, Turkey
| | - Selçuk Baysal
- Department of Cardiology, Urfa Education and Training Hospital, University of Health Sciences, Urfa, Turkey
| | - Zuhal Koç
- Department of Internal Medicine, Onkoloji Education and Training Hospital, University of Health Sciences, Ankara, Turkey
| | - Arif Ülkü Yener
- Department of Ophthalmology, Keçiören Education and Training Hospital, University of Health Sciences, Ankara, Turkey
| |
Collapse
|
23
|
Wilson CS, Mongin AA. Cell Volume Control in Healthy Brain and Neuropathologies. CURRENT TOPICS IN MEMBRANES 2018; 81:385-455. [PMID: 30243438 DOI: 10.1016/bs.ctm.2018.07.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Regulation of cellular volume is a critical homeostatic process that is intimately linked to ionic and osmotic balance in the brain tissue. Because the brain is encased in the rigid skull and has a very complex cellular architecture, even minute changes in the volume of extracellular and intracellular compartments have a very strong impact on tissue excitability and function. The failure of cell volume control is a major feature of several neuropathologies, such as hyponatremia, stroke, epilepsy, hyperammonemia, and others. There is strong evidence that such dysregulation, especially uncontrolled cell swelling, plays a major role in adverse pathological outcomes. To protect themselves, brain cells utilize a variety of mechanisms to maintain their optimal volume, primarily by releasing or taking in ions and small organic molecules through diverse volume-sensitive ion channels and transporters. In principle, the mechanisms of cell volume regulation are not unique to the brain and share many commonalities with other tissues. However, because ions and some organic osmolytes (e.g., major amino acid neurotransmitters) have a strong impact on neuronal excitability, cell volume regulation in the brain is a surprisingly treacherous process, which may cause more harm than good. This topical review covers the established and emerging information in this rapidly developing area of physiology.
Collapse
Affiliation(s)
- Corinne S Wilson
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, United States
| | - Alexander A Mongin
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, United States; Department of Biophysics and Functional Diagnostics, Siberian State Medical University, Tomsk, Russian Federation
| |
Collapse
|
24
|
Orlov SN, Shiyan A, Boudreault F, Ponomarchuk O, Grygorczyk R. Search for Upstream Cell Volume Sensors: The Role of Plasma Membrane and Cytoplasmic Hydrogel. CURRENT TOPICS IN MEMBRANES 2018; 81:53-82. [PMID: 30243440 DOI: 10.1016/bs.ctm.2018.07.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
The plasma membrane plays a prominent role in the regulation of cell volume by mediating selective transport of extra- and intracellular osmolytes. Recent studies show that upstream sensors of cell volume changes are mainly located within the cytoplasm that displays properties of a hydrogel and not in the plasma membrane. Cell volume changes occurring in anisosmotic medium as well as in isosmotic environment affect properties of cytoplasmic hydrogel that, in turn, trigger rapid regulatory volume increase and decrease (RVI and RVD). The downstream signaling pathways include reorganization of 2D cytoskeleton and altered composition of polyphosphoinositides located on the inner surface of the plasma membrane. In addition to its action on physico-chemical properties of cytoplasmic hydrogel, cell volume changes in anisosmotic conditions affect the ionic strength of the cytoplasm and the [Na+]i/[K+]i ratio. Elevated intracellular ionic strength evoked by long term exposure of cells to hypertonic environment resulted in the activation of TonEBP and augmented expression of genes controlling intracellular organic osmolyte levels. The role of Na+i/K+i -sensitive, Ca2+i -mediated and Ca2+i-independent mechanisms of excitation-transcription coupling in cell volume-adjustment remains unknown.
Collapse
Affiliation(s)
- Sergei N Orlov
- Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia; Siberian State Medical University, Tomsk, Russia; National Research Tomsk State University, Tomsk, Russia
| | - Aleksandra Shiyan
- Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Francis Boudreault
- Centre de recherche, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Olga Ponomarchuk
- Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia; Centre de recherche, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Ryszard Grygorczyk
- Centre de recherche, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada; Department of Medicine, Faculty of Medicine, University of Montreal, Montreal, QC, Canada
| |
Collapse
|
25
|
Wilson CS, Mongin AA. The signaling role for chloride in the bidirectional communication between neurons and astrocytes. Neurosci Lett 2018; 689:33-44. [PMID: 29329909 DOI: 10.1016/j.neulet.2018.01.012] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 01/04/2018] [Accepted: 01/05/2018] [Indexed: 01/01/2023]
Abstract
It is well known that the electrical signaling in neuronal networks is modulated by chloride (Cl-) fluxes via the inhibitory GABAA and glycine receptors. Here, we discuss the putative contribution of Cl- fluxes and intracellular Cl- to other forms of information transfer in the CNS, namely the bidirectional communication between neurons and astrocytes. The manuscript (i) summarizes the generic functions of Cl- in cellular physiology, (ii) recaps molecular identities and properties of Cl- transporters and channels in neurons and astrocytes, and (iii) analyzes emerging studies implicating Cl- in the modulation of neuroglial communication. The existing literature suggests that neurons can alter astrocytic Cl- levels in a number of ways; via (a) the release of neurotransmitters and activation of glial transporters that have intrinsic Cl- conductance, (b) the metabotropic receptor-driven changes in activity of the electroneutral cation-Cl- cotransporter NKCC1, and (c) the transient, activity-dependent changes in glial cell volume which open the volume-regulated Cl-/anion channel VRAC. Reciprocally, astrocytes are thought to alter neuronal [Cl-]i through either (a) VRAC-mediated release of the inhibitory gliotransmitters, GABA and taurine, which open neuronal GABAA and glycine receptor/Cl- channels, or (b) the gliotransmitter-driven stimulation of NKCC1. The most important recent developments in this area are the identification of the molecular composition and functional heterogeneity of brain VRAC channels, and the discovery of a new cytosolic [Cl-] sensor - the Wnk family protein kinases. With new work in the field, our understanding of the role of Cl- in information processing within the CNS is expected to be significantly updated.
Collapse
Affiliation(s)
- Corinne S Wilson
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, United States
| | - Alexander A Mongin
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, United States; Department of Biophysics and Functional Diagnostics, Siberian State Medical University, Tomsk, Russian Federation.
| |
Collapse
|
26
|
Schober AL, Wilson CS, Mongin AA. Molecular composition and heterogeneity of the LRRC8-containing swelling-activated osmolyte channels in primary rat astrocytes. J Physiol 2017; 595:6939-6951. [PMID: 28833202 DOI: 10.1113/jp275053] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 08/14/2017] [Indexed: 01/27/2023] Open
Abstract
KEY POINTS The volume-regulated anion channel (VRAC) is a swelling-activated chloride channel that is permeable to inorganic anions and a variety of small organic molecules. VRAC is formed via heteromerization of LRRC8 proteins, among which LRRC8A is essential, while LRRC8B/C/D/E serve as exchangeable complementary partners. We used an RNAi approach and radiotracer assays to explore which LRRC8 isoforms contribute to swelling-activated release of diverse organic osmolytes in rat astrocytes. Efflux of uncharged osmolytes (myo-inositol and taurine) was suppressed by deletion of LRRC8A or LRRC8D, but not by deletion of LRRC8C+LRRC8E. Conversely, release of charged osmolytes (d-aspartate) was strongly reduced by deletion of LRRC8A or LRRC8C+LRRC8E, but largely unaffected by downregulation of LRRC8D. Our findings point to the existence of multiple heteromeric VRACs in the same cell type: LRRC8A/D-containing heteromers appear to dominate release of uncharged osmolytes, while LRRC8A/C/E, with the additional contribution of LRRC8D, creates a conduit for movement of charged molecules. ABSTRACT The volume-regulated anion channel (VRAC) is the ubiquitously expressed vertebrate Cl- /anion channel that is composed of proteins belonging to the LRRC8 family and activated by cell swelling. In the brain, VRAC contributes to physiological and pathological release of a variety of small organic molecules, including the amino acid neurotransmitters glutamate, aspartate and taurine. In the present work, we explored the role of all five LRRC8 family members in the release of organic osmolytes from primary rat astrocytes. Expression of LRRC8 proteins was modified using an RNAi approach, and amino acid fluxes via VRAC were quantified by radiotracer assays in cells challenged with hypoosmotic medium (30% reduction in osmolarity). Consistent with our prior work, knockdown of LRRC8A potently and equally suppressed the release of radiolabelled d-[14 C]aspartate and [3 H]taurine. Among other LRRC8 subunits, downregulation of LRRC8D strongly inhibited release of the uncharged osmolytes [3 H]taurine and myo-[3 H]inositol, without major impact on the simultaneously measured efflux of the charged d-[14 C]aspartate. In contrast, the release of d-[14 C]aspartate was preferentially sensitive to deletion of LRRC8C+LRRC8E, but unaffected by downregulation of LRRC8D. Finally, siRNA knockdown of LRRC8C+LRRC8D strongly inhibited the release of all osmolytes. Overall, our findings suggest the existence of at least two distinct heteromeric VRACs in astroglial cells. The LRRC8A/D-containing permeability pathway appears to dominate the release of uncharged osmolytes, while an alternative channel (or channels) is composed of LRRC8A/C/D/E and responsible for the loss of charged molecules.
Collapse
Affiliation(s)
- Alexandra L Schober
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, 12208, USA
| | - Corinne S Wilson
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, 12208, USA
| | - Alexander A Mongin
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, 12208, USA
| |
Collapse
|
27
|
Volume regulation of intestinal cells of echinoderms: Putative role of ion transporters (Na+/K+-ATPase and NKCC). Comp Biochem Physiol A Mol Integr Physiol 2016; 201:124-131. [DOI: 10.1016/j.cbpa.2016.07.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 07/01/2016] [Accepted: 07/08/2016] [Indexed: 11/24/2022]
|
28
|
Schwingshackl A. The role of stretch-activated ion channels in acute respiratory distress syndrome: finally a new target? Am J Physiol Lung Cell Mol Physiol 2016; 311:L639-52. [PMID: 27521425 DOI: 10.1152/ajplung.00458.2015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 08/05/2016] [Indexed: 02/06/2023] Open
Abstract
Mechanical ventilation (MV) and oxygen therapy (hyperoxia; HO) comprise the cornerstones of life-saving interventions for patients with acute respiratory distress syndrome (ARDS). Unfortunately, the side effects of MV and HO include exacerbation of lung injury by barotrauma, volutrauma, and propagation of lung inflammation. Despite significant improvements in ventilator technologies and a heightened awareness of oxygen toxicity, besides low tidal volume ventilation few if any medical interventions have improved ARDS outcomes over the past two decades. We are lacking a comprehensive understanding of mechanotransduction processes in the healthy lung and know little about the interactions between simultaneously activated stretch-, HO-, and cytokine-induced signaling cascades in ARDS. Nevertheless, as we are unraveling these mechanisms we are gathering increasing evidence for the importance of stretch-activated ion channels (SACs) in the activation of lung-resident and inflammatory cells. In addition to the discovery of new SAC families in the lung, e.g., two-pore domain potassium channels, we are increasingly assigning mechanosensing properties to already known Na(+), Ca(2+), K(+), and Cl(-) channels. Better insights into the mechanotransduction mechanisms of SACs will improve our understanding of the pathways leading to ventilator-induced lung injury and lead to much needed novel therapeutic approaches against ARDS by specifically targeting SACs. This review 1) summarizes the reasons why the time has come to seriously consider SACs as new therapeutic targets against ARDS, 2) critically analyzes the physiological and experimental factors that currently limit our knowledge about SACs, and 3) outlines the most important questions future research studies need to address.
Collapse
|
29
|
Mongin AA. Volume-regulated anion channel--a frenemy within the brain. Pflugers Arch 2015; 468:421-41. [PMID: 26620797 DOI: 10.1007/s00424-015-1765-6] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 11/16/2015] [Accepted: 11/20/2015] [Indexed: 10/22/2022]
Abstract
The volume-regulated anion channel (VRAC) is a ubiquitously expressed yet highly enigmatic member of the superfamily of chloride/anion channels. It is activated by cellular swelling and mediates regulatory cell volume decrease in a majority of vertebrate cells, including those in the central nervous system (CNS). In the brain, besides its crucial role in cellular volume regulation, VRAC is thought to play a part in cell proliferation, apoptosis, migration, and release of physiologically active molecules. Although these roles are not exclusive to the CNS, the relative significance of VRAC in the brain is amplified by several unique aspects of its physiology. One important example is the contribution of VRAC to the release of the excitatory amino acid neurotransmitters glutamate and aspartate. This latter process is thought to have impact on both normal brain functioning (such as astrocyte-neuron signaling) and neuropathology (via promoting the excitotoxic death of neuronal cells in stroke and traumatic brain injury). In spite of much work in the field, the molecular nature of VRAC remained unknown until less than 2 years ago. Two pioneer publications identified VRAC as the heterohexamer formed by the leucine-rich repeat-containing 8 (LRRC8) proteins. These findings galvanized the field and are likely to result in dramatic revisions to our understanding of the place and role of VRAC in the brain, as well as other organs and tissues. The present review briefly recapitulates critical findings in the CNS and focuses on anticipated impact on the LRRC8 discovery on further progress in neuroscience research.
Collapse
Affiliation(s)
- Alexander A Mongin
- Center for Neuropharmacology and Neuroscience, Albany Medical College, 47 New Scotland Ave., Albany, NY, 12208, USA.
| |
Collapse
|
30
|
Schober AL, Mongin AA. Intracellular levels of glutamate in swollen astrocytes are preserved via neurotransmitter reuptake and de novo synthesis: implications for hyponatremia. J Neurochem 2015; 135:176-85. [PMID: 26235094 DOI: 10.1111/jnc.13229] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 06/25/2015] [Accepted: 06/29/2015] [Indexed: 01/01/2023]
Abstract
Hyponatremia and several other CNS pathologies are associated with substantial astrocytic swelling. To counteract cell swelling, astrocytes lose intracellular osmolytes, including l-glutamate and taurine, through volume-regulated anion channel. In vitro, when swollen by exposure to hypo-osmotic medium, astrocytes lose endogenous taurine faster, paradoxically, than l-glutamate or l-aspartate. Here, we explored the mechanisms responsible for differences between the rates of osmolyte release in primary rat astrocyte cultures. In radiotracer assays, hypo-osmotic efflux of preloaded [(14) C]taurine was indistinguishable from d-[(3) H]aspartate and only 30-40% faster than l-[(3) H]glutamate. However, when we used HPLC to measure the endogenous intracellular amino acid content, hypo-osmotic loss of taurine was approximately fivefold greater than l-glutamate, and no loss of l-aspartate was detected. The dramatic difference between loss of endogenous taurine and glutamate was eliminated after inhibition of both glutamate reuptake [with 300 μM dl-threo-β-benzyloxyaspartic acid (TBOA)] and glutamate synthesis by aminotransferases [with 1 mM aminooxyacetic acid (AOA)]. Treatment with TBOA+AOA made reductions in the intracellular taurine and l-glutamate levels approximately equal. Taken together, these data suggest that swollen astrocytes actively conserve intracellular glutamate via reuptake and de novo synthesis. Our findings likely also explain why in animal models of acute hyponatremia, extracellular levels of taurine are dramatically elevated with minimal impact on extracellular l-glutamate. We identified mechanisms that allow astrocytes to conserve intracellular l-glutamate (Glu) upon exposure to hypo-osmotic environment. Cell swelling activates volume-regulated anion channel (VRAC) and triggers loss of Glu, taurine (Tau), and other cytosolic amino acids. Glu is conserved via reuptake by Na(+) -dependent transporters and de novo synthesis in the reactions of mitochondrial transamination (TA). These findings explain why, in acute hyponatremia, extracellular levels of Tau can be dramatically elevated with minimal changes in extracellular Glu.
Collapse
Affiliation(s)
- Alexandra L Schober
- Center for Neuropharmacology and Neuroscience, Albany Medical College, Albany, New York, USA
| | - Alexander A Mongin
- Center for Neuropharmacology and Neuroscience, Albany Medical College, Albany, New York, USA
| |
Collapse
|
31
|
Role of cytoskeleton network in anisosmotic volume changes of intact and permeabilized A549 cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1848:2337-43. [PMID: 26171817 DOI: 10.1016/j.bbamem.2015.07.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 07/06/2015] [Accepted: 07/10/2015] [Indexed: 11/20/2022]
Abstract
Recently we found that cytoplasm of permeabilized mammalian cells behaves as a hydrogel displaying intrinsic osmosensitivity. This study examined the role of microfilaments and microtubules in the regulation of hydrogel osmosensitivity, volume-sensitive ion transporters, and their contribution to volume modulation of intact cells. We found that intact and digitonin-permeabilized A549 cells displayed similar rate of shrinkage triggered by hyperosmotic medium. It was significantly slowed-down in both cell preparations after disruption of actin microfilaments by cytochalasin B, suggesting that rapid water release by intact cytoplasmic hydrogel contributes to hyperosmotic shrinkage. In hyposmotic swelling experiments, disruption of microtubules by vinblastine attenuated the maximal amplitude of swelling in intact cells and completely abolished it in permeabilized cells. The swelling of intact cells also triggered ~10-fold elevation of furosemide-resistant (86)Rb+ (K+) permeability and the regulatory volume decrease (RVD), both of which were abolished by Ba2+. Interestingly, RVD and K+ permeability remained unaffected in cytocholasin/vinblastine treated cells demonstrating that cytoskeleton disruption has no direct impact on Ba2+-sensitive K+-channels involved in RVD. Our results show, for the first time, that the cytoskeleton network contributes directly to passive cell volume adjustments in anisosmotic media via the modulation of the water retained by the cytoplasmic hydrogel.
Collapse
|
32
|
Orlov SN, Koltsova SV, Kapilevich LV, Gusakova SV, Dulin NO. NKCC1 and NKCC2: The pathogenetic role of cation-chloride cotransporters in hypertension. Genes Dis 2015; 2:186-196. [PMID: 26114157 PMCID: PMC4477834 DOI: 10.1016/j.gendis.2015.02.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 02/16/2015] [Indexed: 11/17/2022] Open
Abstract
This review summarizes the data on the functional significance of ubiquitous (NKCC1) and renal-specific (NKCC2) isoforms of electroneutral sodium, potassium and chloride cotransporters. These carriers contribute to the pathogenesis of hypertension via regulation of intracellular chloride concentration in vascular smooth muscle and neuronal cells and via sensing chloride concentration in the renal tubular fluid, respectively. Both NKCC1 and NKCC2 are inhibited by furosemide and other high-ceiling diuretics widely used for attenuation of extracellular fluid volume. However, the chronic usage of these compounds for the treatment of hypertension and other volume-expanded disorders may have diverse side-effects due to suppression of myogenic response in microcirculatory beds.
Collapse
Affiliation(s)
- Sergei N. Orlov
- Faculty of Biology, M.V. Lomonosov Moscow State University, Russia
- Тomsk State University, Russia
| | | | | | | | | |
Collapse
|
33
|
Salt and osmosensing: role of cytoplasmic hydrogel. Pflugers Arch 2015; 467:475-87. [DOI: 10.1007/s00424-014-1680-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 12/17/2014] [Accepted: 12/18/2014] [Indexed: 12/30/2022]
|
34
|
Orlov SN, Koltsova SV, Kapilevich LV, Dulin NO, Gusakova SV. Cation-chloride cotransporters: Regulation, physiological significance, and role in pathogenesis of arterial hypertension. BIOCHEMISTRY (MOSCOW) 2015; 79:1546-61. [DOI: 10.1134/s0006297914130070] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
35
|
Oenarto J, Görg B, Moos M, Bidmon HJ, Häussinger D. Expression of organic osmolyte transporters in cultured rat astrocytes and rat and human cerebral cortex. Arch Biochem Biophys 2014; 560:59-72. [DOI: 10.1016/j.abb.2014.06.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 06/18/2014] [Accepted: 06/21/2014] [Indexed: 01/21/2023]
|
36
|
Temperature-Induced Inactivation of Cytoplasmic Biogel Osmosensing Properties is Associated with Suppression of Regulatory Volume Decrease in A549 Cells. J Membr Biol 2014; 247:571-9. [DOI: 10.1007/s00232-014-9673-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 05/06/2014] [Indexed: 10/25/2022]
|
37
|
Parshina EY, Yusipovich AI, Platonova AA, Grygorczyk R, Maksimov GV, Orlov SN. Thermal inactivation of volume-sensitive K⁺,Cl⁻ cotransport and plasma membrane relief changes in human erythrocytes. Pflugers Arch 2013; 465:977-83. [PMID: 23377567 DOI: 10.1007/s00424-013-1221-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Revised: 01/17/2013] [Accepted: 01/18/2013] [Indexed: 10/27/2022]
Abstract
Previously, we reported that in mammalian erythrocytes irreversible annealing of spectrin heterodimers at 49-50 °C abolished cell volume-dependent regulation of ion carriers, thus suggesting an implication of a two-dimensional (2D) membrane carcass in volume sensing and/or signal transduction. To further examine this hypothesis, we employed atomic force microscopy. This method revealed folded membrane relief of fixed human erythrocytes with an average wave height of 3-5 nm covered by globular structures with a diameter of 40-50 nm and an average height of 1-2 nm. Erythrocyte swelling caused by reduction of medium osmolality decreased the height of membrane surface waves by 40 % and increased K(+),Cl(-) cotransport by approximately sixfold. Both volume-sensitive changes of membrane relief and activity of K(+),Cl(-) cotransporter were abolished by a 10-min preincubation at 50 °C. Our results strongly suggest that volume-dependent alterations of the human erythrocyte membrane relief are caused by reorganization of the 2D spectrin-actin network contributing to regulation of the activity of volume-sensitive ion transporters.
Collapse
Affiliation(s)
- E Yu Parshina
- Department of Biophysics and Laboratory of Physical Chemistry of Biomembranes, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | | | | | | | | | | |
Collapse
|
38
|
Cicuéndez M, Izquierdo-Barba I, Portolés MT, Vallet-Regí M. Biocompatibility and levofloxacin delivery of mesoporous materials. Eur J Pharm Biopharm 2013; 84:115-24. [DOI: 10.1016/j.ejpb.2012.11.029] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Revised: 11/20/2012] [Accepted: 11/26/2012] [Indexed: 01/07/2023]
|
39
|
Santos IA, Castellano GC, Freire CA. Direct relationship between osmotic and ionic conforming behavior and tissue water regulatory capacity in echinoids. Comp Biochem Physiol A Mol Integr Physiol 2013; 164:466-76. [DOI: 10.1016/j.cbpa.2012.12.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Revised: 12/06/2012] [Accepted: 12/10/2012] [Indexed: 10/27/2022]
|
40
|
Cuozzo F, Raciti M, Bertelli L, Parente R, Di Renzo L. Pro-death and pro-survival properties of ouabain in U937 lymphoma derived cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2012; 31:95. [PMID: 23153195 PMCID: PMC3541998 DOI: 10.1186/1756-9966-31-95] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Accepted: 11/12/2012] [Indexed: 12/17/2022]
Abstract
BACKGROUND Epidemiological studies revealed significantly lower mortality rates in cancer patients receiving cardiac glycosides, which turned on interest in the anticancer properties of these drugs. However, cardiac glycosides have also been shown to stimulate cell growth in several cell types. In the present investigation we analyzed the pro-death and pro-survival properties of ouabain in the human lymphoma derived cell line U937. METHODS ROS, intracellular Ca++, cell cycle were evaluated by loading the cells with fluorescent probes under cytofluorimetry. Cell counts and evaluation of trypan blue-excluding cells were performed under optic microscope. Protein detection was done by specific antibodies after protein separation from cellular lysates by SDS-PAGE and transfer blot. RESULTS High doses of ouabain cause ROS generation, elevation of [Ca++]i and death of lymphoma derived U937 cells. Lower doses of OUA activate a survival pathway in which plays a role the Na+/Ca++-exchanger (NCX), active in the Ca++ influx mode rather than in the Ca++ efflux mode. Also p38 MAPK plays a pro-survival role. However, the activation of this MAPK does not appear to depend on NCX. CONCLUSION This investigation shows that the cardiac glycoside OUA is cytotoxic also for the lymphoma derived cell line U937 and that can activate a survival pathway in which are involved NCX and p38 MAPK. These molecules can represent potential targets of combined therapy.
Collapse
Affiliation(s)
- Francesca Cuozzo
- Department of Experimental Medicine, University of Rome La Sapienza, Viale Regina Elena 324, Rome 00161, Italy
| | | | | | | | | |
Collapse
|
41
|
Bowens NH, Dohare P, Kuo YH, Mongin AA. DCPIB, the proposed selective blocker of volume-regulated anion channels, inhibits several glutamate transport pathways in glial cells. Mol Pharmacol 2012; 83:22-32. [PMID: 23012257 DOI: 10.1124/mol.112.080457] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
4-(2-Butyl-6,7-dichloro-2-cyclopentyl-indan-1-on-5-yl) oxobutyric acid (DCPIB) was identified as the selective blocker of volume-regulated anion channels (VRAC). VRAC are permeable to small inorganic and organic anions, including the excitatory neurotransmitter glutamate. In recent years DCPIB has been increasingly used for probing the physiologic and pathologic roles of VRAC and was found to potently suppress pathologic glutamate release in cerebral ischemia. Because ischemic glutamate release can be mediated by a plethora of mechanisms, in this study we explored the selectivity of DCPIB toward the majority of previously identified glutamate transporters and permeability pathways. l-[(3)H]glutamate, d-[(3)H]aspartate, and l-[(14)C]cystine were used to trace amino acid release and uptake. We found that in addition to its well-characterized effect on VRAC, DCPIB potently inhibited glutamate release via connexin hemichannels and glutamate uptake via the glutamate transporter GLT-1 in rat glial cells. In contrast, DCPIB had no direct effect on vesicular glutamate release from rat brain synaptosomes or the cystine/glutamate exchange in astrocytes. The compound did not affect the astrocytic glutamate transporter GLAST, nor did it block glutamate release via the P2X(7)/pannexin permeability pathway. The ability of DCPIB to directly block connexin hemichannels was confirmed using a gene-specific siRNA knockdown approach. Overall, our data demonstrate that DCPIB influences several glutamate transport pathways and that its effects on VRAC in vivo should be verified using additional pharmacological controls.
Collapse
Affiliation(s)
- Nicole H Bowens
- Center for Neuropharmacology and Neuroscience, Albany Medical College, 47 New Scotland Avenue, Albany, NY 12208, USA
| | | | | | | |
Collapse
|
42
|
Koltsova SV, Akimova OA, Kotelevtsev SV, Grygorczyk R, Orlov SN. Hyperosmotic and isosmotic shrinkage differentially affect protein phosphorylation and ion transport. Can J Physiol Pharmacol 2012; 90:209-17. [DOI: 10.1139/y11-119] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In the present work, we compared the outcome of hyperosmotic and isosmotic shrinkage on ion transport and protein phosphorylation in C11-MDCK cells resembling intercalated cells from collecting ducts and in vascular smooth muscle cells (VSMC) from the rat aorta. Hyperosmotic shrinkage was triggered by cell exposure to hypertonic medium, whereas isosmotic shrinkage was evoked by cell transfer from an hypoosmotic to an isosmotic environment. Despite a similar cell volume decrease of 40%–50%, the consequences of hyperosmotic and isosmotic shrinkage on cellular functions were sharply different. In C11-MDCK and VSMC, hyperosmotic shrinkage completely inhibited Na+,K+-ATPase and Na+,Pi cotransport. In contrast, in both types of cells isosmotic shrinkage slightly increased rather than suppressed Na+,K+-ATPase and did not change Na+,Pi cotransport. In C11-MDCK cells, phosphorylation of JNK1/2 and Erk1/2 mitogen-activated protein kinases was augmented in hyperosmotically shrunken cells by ∼7- and 2-fold, respectively, but was not affected in cells subjected to isosmotic shrinkage. These results demonstrate that the data obtained in cells subjected to hyperosmotic shrinkage cannot be considered as sufficient proof implicating cell volume perturbations in the regulation of cellular functions under isosmotic conditions.
Collapse
Affiliation(s)
- Svetlana V. Koltsova
- Research Centre, Centre hospitalier de l’Université de Montréal (CHUM) – Technopôle Angus, Montreal, QC H1W 4A4, Canada
| | - Olga A. Akimova
- Research Centre, Centre hospitalier de l’Université de Montréal (CHUM) – Technopôle Angus, Montreal, QC H1W 4A4, Canada
| | | | - Ryszard Grygorczyk
- Research Centre, Centre hospitalier de l’Université de Montréal (CHUM) – Technopôle Angus, Montreal, QC H1W 4A4, Canada
| | - Sergei N. Orlov
- Research Centre, Centre hospitalier de l’Université de Montréal (CHUM) – Technopôle Angus, Montreal, QC H1W 4A4, Canada
- Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
- Institute of General Pathology and Pathophysiology of the Russian Academy of Medical Sciences, Moscow, Russia
| |
Collapse
|
43
|
YUSIPOVICH AI, ZAGUBIZHENKO MV, LEVIN GG, PLATONOVA A, PARSHINA EY, GRYGORZCYK R, MAKSIMOV GV, RUBIN AB, ORLOV SN. Laser interference microscopy of amphibian erythrocytes: impact of cell volume and refractive index. J Microsc 2011; 244:223-9. [DOI: 10.1111/j.1365-2818.2011.03516.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
44
|
Hyzinski-García MC, Vincent MY, Haskew-Layton RE, Dohare P, Keller RW, Mongin AA. Hypo-osmotic swelling modifies glutamate-glutamine cycle in the cerebral cortex and in astrocyte cultures. J Neurochem 2011; 118:140-52. [PMID: 21517854 DOI: 10.1111/j.1471-4159.2011.07289.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In our previous work, we found that perfusion of the rat cerebral cortex with hypo-osmotic medium triggers massive release of the excitatory amino acid L-glutamate but decreases extracellular levels of L-glutamine (R. E. Haskew-Layton et al., PLoS ONE, 3: e3543). The release of glutamate was linked to activation of volume-regulated anion channels, whereas mechanism(s) responsible for alterations in extracellular glutamine remained unclear. When mannitol was added to the hypo-osmotic medium to reverse reductions in osmolarity, changes in microdialysate levels of glutamine were prevented, indicating an involvement of cellular swelling. As the main source of brain glutamine is astrocytic synthesis and export, we explored the impact of hypo-osmotic medium on glutamine synthesis and transport in rat primary astrocyte cultures. In astrocytes, a 40% reduction in medium osmolarity moderately stimulated the release of L-[(3) H]glutamine by ∼twofold and produced no changes in L-[(3) H]glutamine uptake. In comparison, hypo-osmotic medium stimulated the release of glutamate (traced with D-[(3) H]aspartate) by more than 20-fold. In whole-cell enzymatic assays, we discovered that hypo-osmotic medium caused a 20% inhibition of astrocytic conversion of L-[(3) H]glutamate into L-[(3) H]glutamine by glutamine synthetase. Using an HPLC assay, we further found a 35% reduction in intracellular levels of endogenous glutamine. Overall, our findings suggest that cellular swelling (i) inhibits astrocytic glutamine synthetase activity, and (ii) reduces substrate availability for this enzyme because of the activation of volume-regulated anion channels. These combined effects likely lead to reductions in astrocytic glutamine export in vivo and may partially explain occurrence of hyperexcitability and seizures in human hyponatremia.
Collapse
Affiliation(s)
- María C Hyzinski-García
- Center for Neuropharmacology and Neuroscience, Albany Medical College, Albany, New York, USA
| | | | | | | | | | | |
Collapse
|
45
|
Platonova A, Koltsova S, Maksimov GV, Grygorczyk R, Orlov SN. The death of ouabain-treated renal epithelial C11-MDCK cells is not mediated by swelling-induced plasma membrane rupture. J Membr Biol 2011; 241:145-54. [PMID: 21584679 DOI: 10.1007/s00232-011-9371-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Accepted: 05/05/2011] [Indexed: 10/18/2022]
Abstract
This study examined the role of cell volume modulation in plasma membrane rupture and death documented in ouabain-treated renal epithelial cells. Long-term exposure to ouabain caused massive death of C11-MDCK (Madin-Darby canine kidney) epithelial cells, documented by their detachment, chromatin cleavage and complete loss of lactate dehydrogenase (LDH), but did not affect the survival of vascular smooth muscle cells (VSMCs) from the rat aorta. Unlike the distinct impact on cell survival, 2-h exposure to ouabain led to sharp elevation of the [Na⁺](i)/[K⁺](i) ratio in both cell types. A similar increment of Na⁺(i) content was evoked by sustained inhibition of Na⁺,K⁺-ATPase in K⁺-free medium. However, in contrast to ouabain, C11-MDCK cells survived perfectly during 24-h exposure to K⁺-free medium. At 3 h, the volume of ouabain-treated C11-MDCK cells and VSMCs, measured by the recently developed dual-image surface reconstruction technique, was increased by 16 and 12%, respectively, whereas 5-10 min before the detachment of ouabain-treated C11-MDCK cells, their volume was augmented by ~30-40%. To examine the role of modest swelling in the plasma membrane rupture of ouabain-treated cells, we compared actions of hypotonic medium on volume and LDH release. We observed that LDH release from hypoosmotically swollen C11-MDCK cells was triggered when their volume was increased by approximately fivefold. Thus, our results showed that the rupture of plasma membranes in ouabain-treated C11-MDCK cells was not directly caused by cell volume modulation evoked by Na⁺,K⁺-ATPase inhibition and inversion of the [Na⁺](i)/[K⁺](i) ratio.
Collapse
Affiliation(s)
- Alexandra Platonova
- Research Centre, Centre Hospitalier de l'Université de Montréal (CRCHUM)-Technopôle Angus, 2901 Rachel Est, Montreal, QC H1W4A4, Canada
| | | | | | | | | |
Collapse
|
46
|
Koltsova SV, Platonova A, Maksimov GV, Mongin AA, Grygorczyk R, Orlov SN. Activation of P2Y receptors causes strong and persistent shrinkage of C11-MDCK renal epithelial cells. Am J Physiol Cell Physiol 2011; 301:C403-12. [PMID: 21562307 DOI: 10.1152/ajpcell.00018.2011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Purinergic receptors activate diverse signaling cascades and regulate the activity of cell volume-sensitive ion transporters. However, the effects of ATP and other agonists of P2 receptors on cell volume dynamics are only scarcely studied. In the present work, we used the recently developed dual-image surface reconstruction technique to explore the influence of purinergic agonists on cell volume in the C11-Madin-Darby canine kidney cell line resembling intercalated cells from kidney collecting ducts. Unexpectedly, we found that ATP and UTP triggered very robust (55-60%) cell shrinkage that lasted up to 2 h after agonist washout. Purinergic regulation of cell volume required increases in intracellular Ca(2+) and could be partially mimicked by the Ca(2+)-ionophore ionomycin or activation of protein kinase C by 4β-phorbol 12-myristate 13-acetate. Cell shrinkage was accompanied by strong reductions in intracellular K(+) and Cl(-) content measured using steady-state (86)Rb(+) and (36)Cl(-) distribution. Both shrinkage and ion efflux in ATP-treated cells were prevented by the anion channel blocker 5-nitro-2-(3-phenylpropylamino)benzoic acid (NPPB) and by the BK(Ca) channel inhibitors charybdotoxin, iberiotoxin, and paxilline. To evaluate the significance of cell-volume changes in purinergic signaling, we measured the impact of ATP on the expression of the immediate-early gene c-Fos. Thirty-minute treatment with ATP increased c-Fos immunoreactivity by approximately fivefold, an effect that was strongly inhibited by charybdotoxin and completely prevented by NPPB. Overall, our findings suggest that ATP-induced cell-volume changes are partially responsible for the physiological actions of purinergic agonists.
Collapse
Affiliation(s)
- Svetlana V Koltsova
- Research Centre, Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | | | | | | | | | | |
Collapse
|
47
|
Akimova OA, Lopina OD, Rubtsov AM, Hamet P, Orlov SN. Investigation of mechanism of p38 MAPK activation in renal epithelial cell from distal tubules triggered by cardiotonic steroids. BIOCHEMISTRY (MOSCOW) 2011; 75:971-8. [PMID: 21073417 DOI: 10.1134/s0006297910080043] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Ouabain and other cardiotonic steroids (CTS) kill renal epithelial cells from distal tubules (C7-MDCK) via interaction with Na,K-ATPase but independently of inhibition of Na,K-ATPase-mediated ion fluxes. Recently, we demonstrated that modest intracellular acidification and inhibition of p38 MAPK suppress death of C7-MDCK cells triggered by ouabain. In the present study we investigate the mechanism of p38 MAPK activation in renal epithelial cell from distal tubules evoked by cardiotonic steroids. Using Na+/K+ ionophores (monensin, nigericin) and media with different content of monovalent cations, we revealed that p38 MAPK phosphorylation in ouabain-treated renal epithelial cells is not caused by Na,K-ATPase inhibition and inversion of the [Na+](i)/[K+](i) ratio. We also demonstrated that attenuation of pH from 7.45 to 6.75 did not alter the level of p38 MAPK phosphorylation observed in ouabain-treated cells. Inhibitors of PKA, PKC, and PKG as well as protein phosphatases were unable to abolish p38 MAPK activation triggered by ouabain. Using phosphotyrosine antibodies we did not detect any effect of ouabain on activation of tyrosine kinases. Thus, our results show that activation of p38 MAPK and cytotoxic action of CTS are independent of intracellular Na+, K+, and H+ concentrations. The molecular origin of intermediates of death signaling induced by CTS via conformation changes of Na,K-ATPase with following activation of p38 MAPK should be examined further.
Collapse
Affiliation(s)
- O A Akimova
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia.
| | | | | | | | | |
Collapse
|
48
|
Erythrocyte morphological states, phases, transitions and trajectories. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2010; 1798:1767-78. [DOI: 10.1016/j.bbamem.2010.05.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2010] [Revised: 04/19/2010] [Accepted: 05/07/2010] [Indexed: 11/20/2022]
|
49
|
Rudkouskaya A, Chernoguz A, Haskew-Layton RE, Mongin AA. Two conventional protein kinase C isoforms, alpha and beta I, are involved in the ATP-induced activation of volume-regulated anion channel and glutamate release in cultured astrocytes. J Neurochem 2010; 105:2260-70. [PMID: 18315563 DOI: 10.1111/j.1471-4159.2008.05312.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Volume-regulated anion channels (VRACs) are activated by cell swelling and are permeable to inorganic and small organic anions, including the excitatory amino acids glutamate and aspartate. In astrocytes, ATP potently enhances VRAC activity and glutamate release via a P2Y receptor-dependent mechanism. Our previous pharmacological study identified protein kinase C (PKC) as a major signaling enzyme in VRAC regulation by ATP. However, conflicting results obtained with potent PKC blockers prompted us to re-evaluate the involvement of PKC in regulation of astrocytic VRACs by using small interfering RNA (siRNA) and pharmacological inhibitors that selectively target individual PKC isoforms. In primary rat astrocyte cultures, application of hypoosmotic medium (30% reduction in osmolarity) and 20 microM ATP synergistically increased the release of excitatory amino acids, measured with a non-metabolized analog of L-glutamate, D-[(3)H]aspartate. Both Go6976, the selective inhibitor of Ca(2+)-sensitive PKCalpha, betaI/II, and gamma, and MP-20-28, a cell permeable pseudosubstrate inhibitory peptide of PKCalpha and betaI/II, reduced the effects of ATP on D-[(3)H]aspartate release by approximately 45-55%. Similar results were obtained with a mixture of siRNAs targeting rat PKCalpha and betaI. Surprisingly, down-regulation of individual alpha and betaI PKC isozymes by siRNA was completely ineffective. These data suggest that ATP regulates VRAC activity and volume-sensitive excitatory amino acid release via cooperative activation of PKCalpha and betaI.
Collapse
Affiliation(s)
- Alena Rudkouskaya
- Center of Neuropharmacology and Neuroscience, Albany Medical College, Albany, NY 12208
| | - Artur Chernoguz
- Center of Neuropharmacology and Neuroscience, Albany Medical College, Albany, NY 12208
| | - Renée E Haskew-Layton
- Burke/Cornell Medical Research Institute of Cornell University, White Plains, NY 10605
| | - Alexander A Mongin
- Center of Neuropharmacology and Neuroscience, Albany Medical College, Albany, NY 12208
| |
Collapse
|
50
|
Chimote AA, Adragna NC, Lauf PK. Ion transport in a human lens epithelial cell line exposed to hyposmotic and apoptotic stress. J Cell Physiol 2010; 223:110-22. [PMID: 20049853 DOI: 10.1002/jcp.22015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Membrane transport changes in human lens epithelial (HLE-B3) cells under hyposmotic and apoptotic stress were compared. Cell potassium content, K(i), uptake of the K congener rubidium, Rb(i), and water content were measured after hyposmotic stress induced by hypotonicity, and apoptotic stress by the protein-kinase inhibitor staurosporine (STP). Cell water increased in hyposmotic (150 mOsm) as compared to isosmotic (300 mOsm) balanced salt solution (BSS) by >2-fold at 5 min and decreased within 15 min to baseline values accompanied by a 40% K(i) loss commensurate with cell swelling and subsequent cell shrinkage likely due to regulatory volume decrease (RVD). Loss of K(i), and accompanying water, and Rb(i) uptake in hyposmotic BSS were prevented by clotrimazole (CTZ) suggesting water shifts associated with K and Rb flux via intermediate conductance K (IK) channels, also detected at the mRNA and protein level. In contrast, 2 h after 2 microM STP exposure, the cells lost approximately 40% water and approximately 60% K(i), respectively, consistent with apoptotic volume decrease (AVD). Indeed, water and K(i) loss was at least fivefold greater after hyposmotic than after apoptotic stress. High extracellular K and 2 mM 4-aminopyridine (4-AP) but not CTZ significantly reduced apoptosis. Annexin labeling phosphatidylserine (PS) at 15 min suggested loss of lipid asymmetry. Quantitative PCR revealed significant IK channel expression during prolonged hyposmotic stress. Results suggest in HLE-B3 cells, IK channels likely partook in and were down regulated after RVD, whereas pro-apoptotic STP-activation of 4-AP-sensitive voltage-gated K channels preceded or accompanied PS externalization before subsequent apoptosis.
Collapse
Affiliation(s)
- Ameet A Chimote
- Cell Biophysics Group, Boonshoft School of Medicine, Wright State University, Dayton, Ohio 45435, USA
| | | | | |
Collapse
|